METHODS AND SYSTEMS TO PERFORM GENETICALLY VARIANT PROTEIN ANALYSIS, AND RELATED MARKER GENETIC PROTEIN VARIATIONS AND DATABASES

Information

  • Patent Application
  • 20210095348
  • Publication Number
    20210095348
  • Date Filed
    September 06, 2018
    5 years ago
  • Date Published
    April 01, 2021
    3 years ago
Abstract
Methods and systems to perform genetically variant protein analysis and related marker genetic protein variations and databases, which in several embodiments allow performing a reliable genetic variation protein analysis in biological samples of different types and conditions taking into account the features of the biological sample where the analysis is performed.
Description
FIELD

The present disclosure relates to analysis of genetic variations in individuals, and in particular to the preparation and analysis of biological samples for identification and/or detection of markers genetic information in biological material.


BACKGROUND

Use of biological material to answer questions pertaining to legal situations, including criminal and civil cases, has rapidly integrated traditional techniques of forensic science that depend on qualitative expert opinion.


In particular, DNA and protein analysis provide techniques which constitute evidence with a sound scientific footing.


Despite the progress made in this field, challenges remain to develop methods of genetic variation analysis resulting in reliable results from a broad spectrum of biological samples, and in particular to develop methods of genetic variation analysis which minimize false positive and/or false negative results due to the specific features of the biological sample where the investigation is performed.


SUMMARY

Provided herein are methods and systems to perform genetically variant protein analysis and related marker genetic protein variations and databases, which in several embodiments allow performing a reliable genetic variation protein analysis in biological samples of different types and conditions taking into account the features of the biological sample where the analysis is performed.


In particular, in several embodiments, the methods and systems and related marker genetic protein variations and databases herein described comprise and/or use marker genetic protein variations validated to be detectable in the biological sample where the genetic protein variation analysis is performed. In several embodiments, the methods and systems and related marker genetic protein variations and databases herein described use preparation methods which maximize recovery of processable protein from such biological sample.


According to a first aspect, a method to prepare a biological sample for proteomic analysis, is described. The method comprises applying to the biological sample an energy field to obtain a processed biological sample comprising solubilized proteins to be used in the proteomic analysis. In some preferred embodiments, applying to the biological sample an energy field is performed by sonication with an energy field ranging from 150 to 1,200 Watts and frequency ranging from 20 to 80 kHz. In another embodiment microwave energy of up to 1,200 Watts can be used to obtain a processed biological sample comprising solubilized proteins.


According to a second aspect, a method and system are described to provide a marker genetic protein variation for a biological organism and a marker genetic protein variation obtainable thereby. In the method and system, the provided marker genetic protein variation is validated to be detectable in a biological sample of an individual of the biological organism.


The method comprises: providing a marker exome sequence of the biological organism, the marker exome sequence comprising a marker genetic variation for the biological organism.


The method further comprises detecting peptide sequences in the biological sample of the individual of the biological organism by performing proteomic analysis of said biological sample to provide proteomically detected peptide sequences.


The method also comprises providing the marker genetic protein variation for the biological organism detectable in the sample of the biological organism by comparing the provided marker exome sequence with the proteomically detected peptide sequences to provide the marker genetic protein variation validated to be detectable in the biological sample of an individual of the biological organism.


The system comprises exome sequences databases and/or reagents to detect exome sequences in an individual of the biological organism, in combination with reagents to perform proteomic analysis of the biological sample for simultaneous combined or sequential use in the method to provide a marker genetic protein variation validated for a biological sample herein described.


According to a third aspect, a method and system to detect a marker genetic protein variation in a biological sample are described. In the method and system, the marker genetic protein variation validated to be detectable in the biological sample.


The method comprises providing a marker mass spectrum of a marker peptide comprising a marker genetic protein variation corresponding to the genetic protein variation; and performing mass spectrometry of a fractionated digested peptide of the biological sample to obtain a mass spectrum of each of the fractionated digested peptide.


The method further comprises comparing the mass spectrum of the fractionated digested peptide with the marker mass spectrum of a marker peptide comprising the marker genetic protein variation to detect the genetic protein variation in the biological sample.


The system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases for simultaneous combined or sequential use in the method to detect a marker genetic protein variation in a biological sample herein described.


According to a fourth aspect, a method and system to improve a marker genetic protein variation database system for a biological organism, and a database obtainable thereby, are described. In the method, system and database herein described, the marker genetic protein variation database system includes data for at least one biological organism and the improvement is the inclusion of one or more marker genetic protein validated to be detectable in a biological sample from an individual of the at least one biological organism.


The method comprises: producing a proteomic dataset from a biological sample from an individual of the at least one biological organism and comparing the proteomic dataset to a protein variant database to produce a set of proteomically detected proteins in the biological sample of the individual.


The method further comprises providing a set of represented genes proteomically detectable in the biological sample of the individual, the represented genes corresponding to the proteomically detected proteins in the biological sample of the individual.


The method also comprises: identifying a marker genetic protein variation validated for the biological sample of the individual, to be included in the marker genetic protein variation database system by providing a proteomically detectable genomic variation in the set of represented genes proteomically detectable in the biological sample of the individual, and providing the marker genetic protein variation validated for the biological sample by providing a proteomically detectable genetic protein variation corresponding to the detectable genomic variation in the biological sample of the individual.


The system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases for simultaneous combined or sequential use in the method to improve a marker genetic protein variation database system for a biological organism herein described.


According to a fifth aspect, a method and system to improve a pooled marker genetic protein variation database system and a pooled marker genetic protein variation database obtainable thereby. In the method and system and related database, the pooled marker genetic protein variation database system comprising marker genetic protein variations common to a plurality of individuals.


The method comprises: providing a number of proteomic datasets of individuals of the plurality of individuals, the number statistically significant for the plurality of individuals, identifying a protein common to the provided number of proteomic datasets; and selecting from the identified protein common to the provided proteomic datasets, a protein detectable in a biological sample of an individual of the plurality of individuals.


The method further comprises providing a number of exome datasets of the individuals of the plurality of individuals, the number statistically significant for the plurality of individuals; and identifying a genetic variation in the provided number of exome datasets.


The method also comprises selecting from the identified genetic variation, a genetic variation detectable in the biological sample; and comparing the selected proteins detectable in the biological sample with the selected genetic variations detectable in the biological sample, to provide a marker genetic protein variation common to a plurality of individuals of a biological organism type and validated to be detectable in the biological sample.


The system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases for simultaneous combined or sequential use in the method to improve a pooled marker genetic protein variation database system for a biological organism herein described.


According to a sixth aspect, a method and a system are described to detect a marker genetic variation for a biological organism validated to be detectable in a biological sample of an individual of the biological system.


The method comprises preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis; and fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample and a solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample.


The method further comprises detecting a genetic protein variation in the solubilized proteins from the sample by performing the proteomic analysis of the solubilized protein fraction; and detecting a genomic variation of the nuclear and/or mitochondrial genome by performing a genetic analysis of the solubilized DNA fraction.


The method also comprises comparing the detected genetic protein variation and/or the detected genomic variation with a marker genetic protein variation and/or of a marker genomic variation respectively from the marker genetic variation database system herein described.


The system comprises exome sequences databases and/or reagents to detect exome sequences in an individual of the biological organism, in combination with reagents to perform proteomic analysis of the biological sample for simultaneous combined or sequential use in the method to detect a marker genetic variation for a biological organism validated to be detectable in a biological sample of an individual of the biological system herein described.


According to a seventh aspect, a method to provide a marker genetic variation database system comprising marker genetic variation validated to be detectable in a biological sample, the method comprises preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis.


The method further comprises fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample and a solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample.


The method also comprises detecting a genetic protein variation in the solubilized proteins from the sample by performing the proteomic analysis of the solubilized protein fraction and detecting a genomic variation of the nuclear and/or mitochondrial genome by performing a genetic analysis of the solubilized DNA fraction.


The method additionally comprises combining the detected genetic protein variations and the detected genomic variation to provide the marker genetic variation database system comprising marker genetic variation validated to be detectable in a biological sample.


The system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases for simultaneous combined or sequential use in the method to provide the marker genetic variation database system comprising marker genetic variation validated to be detectable in a biological sample herein described.


According to an eight aspect, a method and system are described to perform genetic analysis of a sample of a biological organism.


The method comprises preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis, and fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample.


The method also comprises digesting the solubilized proteins from the sample with a site specific proteolytic enzyme to obtain digested solubilized proteins from the sample; fractionating the digested solubilized proteins to obtain fractionated digested peptides from the digested solubilized proteins from the biological sample and detecting a marker genetic variation of the fractionated digested peptides.


In the method, preparing the sample and/or detecting a genetic variation can be performed by any one of the methods according to any one of the first aspect to the seventh aspect of the instant disclosure. In particular, in methods according to the eighth aspect the preparing is performed by any one of the methods according to the first aspect herein described; and/or the detecting is performed by at least one of a first detecting method wherein the detecting is performed by any one of the methods according to the third aspect of the present disclosure; and a second detecting method wherein the detecting is performed by any one of the methods according to the sixth aspect of the present disclosure.


The system comprises exome sequences databases and/or reagents to detect exome sequences in an individual of the biological organism, in combination with reagents to perform proteomic analysis of the biological sample for simultaneous combined or sequential use in the method to perform genetic analysis of a sample of a biological organism herein described.


In preferred embodiments of the marker genetic protein variations, databases, methods and systems and related genetic protein variation analysis herein described, performing a proteomic analysis is carried out by performing mass spectrometry of a fractionated digested peptide of the biological sample to obtain a mass spectrum of each of the fractionated digested peptide.


In further preferred embodiments of the marker genetic protein variations, databases, methods and systems and related genetic protein variation analysis herein described, the sample is hair and/or skin.


The methods and systems and related marker genetic protein variations and databases herein described, allow in several embodiments performing a reliable genetic variation protein analysis in degraded samples, in samples from multiple contributors, in samples where genetic material is not present in detectable amounts, and/or in samples where the genetic material and/or protein material are present in low amounts, the reliable analysis performed.


In particular, the methods and systems and related marker genetic protein variations and databases herein described, allow in several embodiments to provide a sample for proteomic analysis with a reduced presence of fragments resulting from uncontrolled breaking of the protein, not due to the enzymatic digestion (e.g. through trypsin digestion).


Accordingly, the methods and systems and related marker genetic protein variations and databases herein described, allow in several embodiments performing proteolysis on samples including a small amount of processable material (e.g. single hair but also other kind of tissues possibly available in small amounts).


Additionally, the methods and systems and related marker genetic protein variations and databases herein described allow in several embodiments to provide a sample for proteomic analysis comprising a more representative/more complete detection of proteins present in the tissue sample per mass of tissue sample.


The methods and systems and related marker genetic protein variations and databases herein described, further allow, in several embodiments, to providing and/or using improved databases in view of inclusion of marker genetic protein variations validated for the biological sample where the genetic protein variation analysis is performed.


Accordingly, the methods and systems and related marker genetic protein variations and databases herein described, also allow, in several embodiments, to reduce false negatives present in databases built with a proteome-based discovery process.


Additionally, the methods and systems and related marker genetic protein variations and databases herein described which are based on marker genetic variation validated to be detectable in the biological sample of interest, also allow, in several embodiments, to provide and/or use a database customizable with validated markers genetically variant protein for an individual, a biological organisms or types of biological organism in accordance with the experimental design and particular query.


Furthermore, the methods and systems and related marker genetic protein variations and databases herein described, also allow, in several embodiments, to perform genetically variant protein analysis without the need of the “needle in a haystack” approach, in view of the ability to use proteomics to screen with validated marker genetic protein variation for an individual, alone or in combination with marker genomic variation (in nuclear and/or mitochondrial genomes), thus having a faster and reliable response to a specific query with respect to available methods to perform genetic variation analysis known to a skilled person.


Additionally, in view of the use of marker genetic protein variation validated for a biological sample analyzed, the methods and systems and related marker genetic protein variations and databases herein described, also allow, in several embodiments, to perform genetically variant protein analysis without the need to go through databases to obtain an output (even if such step could still be performed).


In view of the ability to perform combined analysis of genetic protein variation and nuclear and/or, preferably, mitochondrial genomic variation, the methods and systems and related marker genetic protein variations and databases herein described, also allow, in several embodiments, to provide a more accurate response to a query/increased ability to discriminate identity based on combined metrics from genetic protein variation and genomic variation following verification of proteomic as well as of genomic markers from a single biological sample (e.g. genomic mitochondrial markers herein also mtDNA markers).


In general, embodiments of the methods and systems and related marker genetic protein variations and databases herein described, which are based on at least one of the sample preparation methods herein described, the marker genetic protein variation validated for a specific sample herein described, and/or the combined analysis of genetic protein variation with nuclear and/or mitochondrial genomic variation herein described, provide a faster and/or more reliable genetic variation analysis for a specific biological sample with respect to methods, systems and databases available for a skilled person.


The methods and systems and related marker genetic protein variations and databases herein described, can be used in connection with various applications wherein an improved ability to perform genetic variation analysis of a biological sample is desired. For example, the methods and systems and related marker genetic protein variations and databases herein described can be used in several applications of forensic analysis, such as identification of individuals, biological organisms types and biological organism of interest from a biological sample, determining relatedness of individuals, paternity testing and additional forensic analysis applications identifiable by a skilled person. Additional exemplary applications include uses of the methods and systems in several fields wherein genetic variation analysis can be used including basic biology research, applied biology, bio-engineering, medical research, medical diagnostics, therapeutics, and in additional fields identifiable by a skilled person upon reading of the present disclosure.


The details of one or more embodiments of the disclosure are set forth in the accompanying drawings and the description below. Other features, objects, and advantages will be apparent from the description and drawings, and from the claims.





BRIEF DESCRIPTION OF THE DRAWINGS

The accompanying drawings, which are incorporated into and constitute a part of this specification, illustrate one or more embodiments of the present disclosure and, together with the detailed description and the examples, serve to explain the principles and implementations of the disclosure.



FIGS. 1A-1B show diagrams illustrating an exemplary individual identification using genetically variant protein analysis. FIG. 1A shows schematics illustrating the difference between a variant gasdermin (SEQ ID NOs: 1 and 2) with respect to a reference gasdermin wherein the gasdermin gene is GSDMA and the variant gasdermin is SNP=rs56030650. FIG. 1B (2 parts) illustrates an exemplary database including the SNP=rs56030650 and other variants in digested peptides (SEQ ID Nos: 3 to 85); together with the related frequency.



FIG. 2 shows a schematic overview of two exemplary methods for processing hair samples for proteomic analysis by tandem liquid chromatography mass-spectrometry (LC-MS/MS), for “Single hair” processing using an exemplary sample preparation method of the present disclosure or for “Bulk” hair processing performed with conventional preparation method, as will be understood by a skilled person. In the illustration of FIG. 2, method steps are separated by arrows.



FIG. 3 shows graphs reporting exemplary results of proteomic analysis metrics using samples processed using the exemplary sample preparation methods illustrated in FIG. 2. In particular, FIG. 3 shows a diagram illustrating a protein coverage heat maps (Panel A), protein coverage improvement in terms of number of amino acids detected (Panel B), number of protein identification (Panel C) and number of unique peptide identifications (Panel D) for sample preparations performed with conventional methods (indicated as “Bulk hair” or “Old Single Hair”) and with sample preparation methods herein described (indicated as “Single Hair” or “New Single Hair”).



FIG. 4 shows a schematic overview of an exemplary method for concomitant protein and mitochondrial DNA (mtDNA) recovery and evaluation in a single sample. In the schematics the methods are shown by arrows.



FIGS. 5A-5B show an exemplary mtDNA analysis performed according to embodiments herein described. FIG. 5A shows an exemplary mitochondrial genome (top) and exemplary primers for the related PCR/amplification (SEQ ID Nos. 136 to 143) (bottom). FIG. 5B shows photographs taken under ultraviolet light exposure of exemplary agarose gels stained with ethidium bromide showing DNA bands corresponding to amplicons of mtDNA haplogroup HV regions indicated in each lane of the gels, alongside a molecular size standard (indicated as “1 kb+Ladder”). In FIG. 5B, the mtDNA extract used for the amplification of the DNA bands shown was recovered from samples processed for both protein extraction and mtDNA extraction, as indicated in FIG. 4.



FIG. 6 shows DNA sequences of exemplary haplogroup HV mtDNA regions (SEQ ID NOS: 87 TO 90) using mtDNA extracts recovered from samples processed for both protein extraction and mtDNA extraction, as indicated in FIG. 4. In FIG. 6, the black boxes indicate exemplary SNPs identified in the sequences.



FIG. 7 shows a schematic illustration of the exome-driven (top-down) approaches according to the present disclosure in comparison with bottom-up approaches suitable to identify/detect genetic protein variations in a sample.



FIG. 8 shows a schematic representation of the steps of an exemplary “proteome-driven” GVP discovery and evaluation method.



FIG. 9 shows a schematic of an exemplary method for determination of an ‘Observed Gene Pool’ according to a top-down approach herein described.



FIG. 10 shows a schematic of an exemplary “exome-driven” GVP discovery method, showing integration of genetic and proteomic data according to embodiments herein described.



FIG. 11 shows a schematic of an exemplary application of an “exome-driven” validated GVP panel to operational samples.



FIGS. 12A-12B show a schematic approach for the construction of a common GVP identity Panel comprising validated marker genetic protein variations common to individuals of an exemplary biological organism types according to the disclosure (FIG. 12A) and an exemplary panel obtainable thereby (FIG. 12B).



FIG. 13 shows an exemplary graph reporting results of an exemplary approach to provide identity metrics to be used in methods and systems to detect/provide a validated genetic marker variation herein described as well as to build related databases.



FIG. 14 shows an exemplary graph reporting an approach to provide identity metrics to be used in methods and systems to detect/provide a validated genetic marker variation herein described as well as to build related databases.



FIG. 15 shows a schematic showing an exemplary application of rule calculation showing how linkage disequilibrium affects genotype match probabilities in methods and systems herein described.



FIG. 16 shows an exemplary validated GVP identity panel (SEQ ID NOS: 91 to 124) for bone samples obtainable with the top-down approach herein described.



FIG. 17 shows a schematic of an exemplary method to create a custom GVP identification profile for an individual.



FIG. 18 shows a schematic of an exemplary method of applying an Individual GVP panel to an operational sample.



FIG. 19 shows exemplary diagrams of DNA and protein chemical structures, showing sites of depurination (solid-black arrow), oxidation (shaded arrow), or hydrolysis (hollow arrow).



FIG. 20 shows a diagram of an exemplary overview of GVP identification and validation process.



FIG. 21 shows an exemplary electron microscope image of a cross-section of a single hair.



FIG. 22 shows a diagram of exemplary automated in-line sample processing.



FIG. 23 shows a graph reporting exemplary results of power of discrimination as a function of number of unique peptides identified. In particular, the arrow indicates an exemplary improvement in results from new instrumentation.



FIG. 24 shows a Venn diagram illustrating an exemplary incorporation of GVP profiles and DNA based measures of identity, wherein ‘STR’ refers to single tandem repeats, ‘GVP’ refers to genetically variant proteins and ‘mtDNA’ refers to mitochondrial DNA.



FIG. 25 shows a schematic showing exemplary use of GVP markers to predict biogeographic background.



FIG. 26 shows a pie chart reporting exemplary results of chemical markers detected in in hair samples.



FIG. 27 shows a schematic showing an exemplary GVP database design, wherein an entity relationship diagram shows types of data entities and the relationships between them. The exemplary design allows flexibility by storing additional characteristics as tag-value pairs.



FIG. 28 shows a schematic of an exemplary bone GVP analysis workflow.



FIG. 29 shows a schematic of an exemplary tooth sex-linked protein analysis workflow.



FIG. 30 shows a graph reporting exemplary results of protein coverage (number of amino acids covered) in ‘touch samples’ and ‘hair samples’.



FIGS. 31 to 39 illustrate exemplary steps of a method to perform genetic variation protein analysis for a sample tissues using databases (such as the panel of FIG. 34 SEQ ID NOS: 125 to 133), methods and systems herein described.





DETAILED DESCRIPTION

Provided herein are methods and systems to perform genetically variant protein analysis and related marker genetic protein variations and databases, which in several embodiments allow performing of a reliable genetic variation protein analysis in biological samples of different types and under different conditions, taking into account the features of the biological sample for which the analysis is performed.


The term “genetic variation” as used herein refers to diversity in gene frequencies and/or in gene sequences. In particular, genetic variation as used herein can refer to genes that are translated into corresponding proteins, which can result in diversity in corresponding protein frequency. Genetic variation in the sense of the disclosure can refer to differences between individuals or to differences between populations. Mutation is the ultimate source of genetic variation, but mechanisms such as sexual reproduction and genetic drift contribute to it as well.


Genetic variations in the sense of the disclosure comprise genomic variations (genetic variations in nuclear or mitochondrial DNA of individuals), and genetic protein variations (genetic variations within a genetically variant protein encoded by a non-synonymous variation in the protein coding region of the corresponding encoding gene).


Accordingly, the term “genetically variant protein”, or “GVP” as used herein refers to a protein encoded by a gene, wherein variants of the protein have a variation (e.g. a single amino acid polymorphisms (SAPs)) that is encoded by non-synonymous variation (e.g. a single nucleotide polymorphisms (nsSNPs)) in the protein-coding region of the gene (e.g., see FIGS. 1A-1B).


The term “single amino acid polymorphisms (SAPs))” refers to named amino acid variances derived from SNPs within coding regions. SAP can be quantitatively or qualitatively detected at the proteome level, with non-targeted or targeted proteomics as will be understood by a skilled person.


The term “single nucleotide polymorphism” or “SNP” refers to a variation in a single nucleotide that occurs at a specific position in the genome of an organism, where each variation occurs at a particular frequency within a population of the organism. For example, at a specific base position in the human genome, the base C appears in most individuals, but in a minority of individuals, the position is occupied by base A. There is a SNP at this specific base position, and the two possible nucleotide variations—C or A—are said to be alleles for this base position. SNPs can occur within protein-coding sequences of genes, non-coding regions of genes, or in the intergenic regions (regions between genes). The term “protein-coding” region, also referred to herein as the “coding region”, “coding DNA sequence” or “CDS” as used herein refers to the portion of a gene's DNA or RNA, composed of exons, that codes for protein. The region is bounded at the 5′ end by a start codon (typically ATG) and at the 3′ end with a stop codon (typically TAA, TAG, or TGA). The coding region in mRNA is bounded by the five prime untranslated region (5′-UTR) and the three prime untranslated region (3′-UTR), which are also parts of the exons. The CDS is the portion of an mRNA transcript that is translated by a ribosome.


As understood by those skilled in the art, SNPs within a protein-coding sequence do not necessarily change the amino acid sequence of the protein that is produced, due to degeneracy of the genetic code. SNPs in the coding region are of two types, synonymous and nonsynonymous SNPs. Synonymous SNPs do not alter the amino acid sequence of a protein while nonsynonymous SNPs change the amino acid sequence of a protein. The nonsynonymous SNPs are of two types: missense and nonsense. A missense mutation is a point mutation in which a SNP results in a codon that codes for a different amino acid. In contrast, a nonsense mutation is a point mutation in a sequence of DNA that results in a premature stop codon, also referred to as a nonsense codon, in the transcribed mRNA, and in a truncated, incomplete, and usually nonfunctional protein product.


The term “protein” as used herein indicates a polypeptide with a particular secondary and tertiary structure that can interact with another molecule and in particular, with other biomolecules including other proteins, polynucleotides such as DNA and RNA, lipids, metabolites, hormones, chemokines, and/or small molecules. The term “polypeptide” as used herein indicates an organic linear polymer composed of two or more amino acid monomers and/or analogs thereof. The term “polypeptide” includes amino acid polymers of any length including full-length proteins and peptides, as well as analogs and fragments thereof. A polypeptide of three or more amino acids is also called a protein oligomer, peptide, or oligopeptide. In particular, the terms “peptide” and “oligopeptide” usually indicate a polypeptide with less than 100 amino acid monomers. In particular, in a protein, the polypeptide provides the primary structure of the protein, wherein the term “primary structure” of a protein refers to the sequence of amino acids in the polypeptide chain covalently linked to form the polypeptide polymer. A protein “sequence” indicates the order of the amino acids that form the primary structure. Covalent bonds between amino acids within the primary structure can include peptide bonds or disulfide bonds, and additional bonds identifiable by a skilled person. Polypeptides in the sense of the present disclosure are usually composed of a linear chain of alpha-amino acid residues covalently linked by peptide bond or a synthetic covalent linkage. The two ends of the linear polypeptide chain encompassing the terminal residues and the adjacent segment are referred to as the carboxyl terminus (C-terminus) and the amino terminus (N-terminus) based on the nature of the free group on each extremity. Unless otherwise indicated, counting of residues in a polypeptide is performed from the N-terminal end (NH2-group), which is the end where the amino group is not involved in a peptide bond to the C-terminal end (—COOH group), which is the end where a COOH group is not involved in a peptide bond. Proteins and polypeptides can be identified by x-ray crystallography, direct sequencing, immunoprecipitation, and a variety of other methods as understood by a person skilled in the art. Proteins can be provided in vitro or in vivo by several methods identifiable by a skilled person. In some instances where the proteins are synthetic proteins, in at least a portion of the polymer two or more amino acid monomers and/or analogs thereof are joined through chemically-mediated condensation of an organic acid (—COOH) and an amine (—NH2) to form an amide bond or a “peptide” bond.


As used herein the term “amino acid”, “amino acid monomer”, or “amino acid residue” refers to organic compounds composed of amine and carboxylic acid functional groups, along with a side-chain specific to each amino acid. In particular, alpha- or α-amino acid refers to organic compounds composed of amine (—NH2) and carboxylic acid (—COOH), and a side-chain specific to each amino acid connected to an alpha carbon. Different amino acids have different side chains and have distinctive characteristics, such as charge, polarity, aromaticity, reduction potential, hydrophobicity, and pKa. Amino acids can be covalently linked to form a polymer through peptide bonds by reactions between the amine group of a first amino acid and the carboxylic acid group of a second amino acid. Amino acid in the sense of the disclosure refers to any of the twenty naturally occurring amino acids, non-natural amino acids, and includes both D and L optical isomers.


Methods and systems herein described and related marker genetic protein variations and databases herein described allow performance of genetic protein variation analysis of a sample of a biological organism taking into account the features of the biological sample where the analysis is performed as will be understood by a skilled person upon reading of the present disclosure.


The wording “biological organism” as used herein indicates an entity that exhibits the properties of life and that comprises a genome which is expressed and translated in a proteome. Exemplary biological organisms comprise multicellular animals, plants, and fungi; or unicellular microorganisms such as protists, bacteria, and archaea. In preferred embodiments the biological organism comprises animals and in particular higher animals and in particular vertebrates such as mammals and in particular human beings (Homo sapiens).


Genetic protein variation analysis typically comprises preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis.


Existing methods of sample preparation for proteomics generally comprise performing techniques of cell and tissue disruption, protein solubilization, removal of contaminants, and protein enrichment methods [1].


In particular methods of cell and tissue disruption typically comprise homogenization of the sample. Homogenization methods used for the proteomics purposes can be divided into five major categories: mechanical, ultrasonic, pressure, freeze—thaw, and osmotic/detergent lysis. Mechanical homogenization can be performed using rotor—stator homogenizers, open blade mills, or glass-glass milling, among others known to those skilled in the art. Ultrasonic homogenizers, also called as disintegrators, sonicators, or sonificators, are based on the piezoelectric effect and on the principle of cavitation while generating the high energy or ultrasonic wave, interacting with the sample. More specifically, ultrasonic homogenizers generate sound energy electronically; this energy is converted to mechanical energy, and these changes result in the formation and implosion of small bubbles in the sample. Energy, resolved after explosion/implosion of gas microbubbles, effectively destroys solid particles such as cells, causing cell rupture and successful cell lysis.. Ultrasonic devices are mainly used to homogenize small pieces of soft tissues (e.g., brain, blood, liver). Pressure homogenization typically uses a French press device, and is an effective method for homogenization of cells in suspension, but ineffective towards tissues or organs without previous preparation in another type of homogenizer. Freeze-thaw homogenization uses the effect of ice crystal formation in the tissue during freezing process. Osmotic and detergent lysis methods of disruption of cells utilize osmotic pressure or detergent interactions to destroy cells' walls and membranes. Osmotic lysis is often used to disrupt blood cells. Examples of commonly used detergents are Triton X-100, Tween 80, Nonidet P-40 (NP 40) and saponin.


In a genetic protein variation analysis, a homogenized sample is subjected to protein solubilization. Proteins in their native state are often insoluble. Breaking interactions involved in protein aggregation, e.g. disulfide/hydrogen bonds, van der Waals forces, ionic and hydrophobic interactions, allows disruption of proteins into a solution of individual polypeptides and thus promotes their solubilization. To avoid protein modifications, aggregation or precipitation resulting in the occurrence of artifacts and subsequent protein loss, sample solubilization process typically involves the use of chaotropes (e.g. urea and/or thiourea), detergents (e.g. 3-[(3-Cholamidopropyl)-dimethyl-ammonio]-1-propane sulfonate (CHAPS) or Triton X-100), reducing agents (dithiothreitol/dithioerythritol (DTT/DTE) or tributylphosphine (TBP)) and protease inhibitors in a sample buffer. Their proper use, together with the optimized cell disruption method, dissolution and concentration techniques determines effectiveness of solubilization. Chaotropes disrupt hydrogen bonds and hydrophilic interactions enabling proteins to unfold with all ionizable groups exposed to solution. Detergents and amphipathic molecules disrupt hydrophobic interactions, thus enabling protein extraction and solubilization. With respect to the ionic character of the hydrophilic group, they are classified into several groups: ionic (e.g. anionic sodium dodecyl sulfate (SDS)), non-ionic (uncharged, e.g. octyl glucoside, dodecyl maltoside and Triton X-100) or zwitterionic (having both positively and negatively charged groups with a net charge of zero, e.g. CHAPS, 3-[(3-Cholamidopropyl) dimethylammonio]-2-hydroxy-1-propanesulfonate (CHAPSO), tetradecanoylamidopropyl-dimethylammoniobutanesulfonate (ASB-14)). Reductants disrupt disulfide bonds between cysteine residues and thus promote unfolding of proteins. Typically, sulfhydryl reducing agents such as dithothreitol (DTT), dithioerythritol (DTE) are applied in the sample preparation protocol. To minimize uncontrolled enzymatic proteolysis by proteases present in samples, protein degradation can be minimized by quick and small scale tissue extraction, boiling the sample in SDS buffer with the high-pH Tris-base, or, on the contrary, lowering the pH and performing ice-cold precipitation in, e.g. 20% trichloroacetic acid. Alternatively, denaturation by boiling in water, focused microwave irradiation, and the use of organic solvents can be applied to inhibit proteases activity. Addition of protease inhibitors can be used to prevent uncontrolled enzymatic protein degradation in a sample. Addition of specific protease inhibitors (e.g. phenylmethylsulfonyl fluoride (PMSF), aminoethyl benzylsulfonyl fluoride (AEBSF), ethylene diamine tetraacetic acid (EDTA), pepstatin, benzamidine, leupeptin, aprotinin) or cocktails with a broader activity spectrum can be used.


In a genetic protein variation analysis, methods of homogenization and/or solubilization techniques for a particular sample type are identifiable by persons skilled in the art. Exemplary methods of homogenization comprise mechanical, ultrasonic, pressure, freeze-thaw, and osmotic/detergent lysis approaches as described herein. Exemplary method of solubilization comprise methods described herein that use reagents comprising one or more chaotropes, detergents, reducing agents and/or protease inhibitors in a sample buffer, as well as other materials and methods identifiable by skilled persons upon reading the present disclosure.


For example, exemplary methods to perform preparing a hair sample to obtain a processed hair sample comprising solubilized proteins to be used in a proteomic analysis comprise milling, denaturation, reduction, and alkylation. Some tissue types such as teeth and bones require additional steps to demineralize the sample material prior to homogenization and solubilization of proteins. There are several ways to extract peptide information from tissues such as teeth and bones, including using a hand-drill, crushing the sample material under liquid nitrogen and demineralization with EDTA or 1.2 M hydrochloric acid, and other methods identifiable by skilled persons.


Genetic protein variation analysis typically further comprises fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample.


In a genetic protein variation analysis, fractionating the processed sample typically comprises removing buffers, salts, and detergent from the processed sample. The pH and ionic strength of sample solutions considerably influence protein solubility. Therefore, buffers, salts and detergents are included in sample solutions and often tend to interfere with further protein separation steps, inhibit the digestion process, interfere with the mass spectrometry analysis, or complicate data analysis significantly, and thus need to be removed. Salts removal can be accomplished using methods such as dialysis (e.g. using spin columns), ultrafiltration, gel filtration, precipitation with TCA or organic solvents, and solid-phase extraction, some of which are used in commercially available clean-up kits identifiable by those skilled in the art. Typical detergent removal methods include dialysis, gel filtration chromatography, hydrophobic adsorption chromatography and protein precipitation. Detergents such as SDS can be removed with nanoscale hydrophilic phase chromatography or acetone precipitation. Commercially available kits, e.g., detergent precipitation reagents or gels effective for binding and removal milligram quantities of various detergents from protein solutions can be used (e.g. Extracti-Gel D Detergent Removing Gel, ReadyPrep 2-D Cleanup Kit, and the SDS-Out SDS Precipitation Reagent and Kit, Pierce). Hydrophobic adsorption employing the use of insoluble resin (e.g. CALBIOSORB, Calbiochem) can also be used to remove excess detergent.


In a genetic protein variation analysis, fractionating the processed sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample can further comprise removing abundant proteins from the processed sample. Protein concentration in biological samples can vary more than 10 orders of magnitude and thus proteomic analyses and detection of less abundant proteins can be hampered by those molecules present at higher concentration. In some cases, removal of abundant proteins can be performed to increase detection of other molecules present at low concentrations. Various techniques can be used for the removal of high-abundant proteins, such as those based on affinity chromatography employing dye-ligands, their derivatives, mimetic ligands, proteins A and G, and antibodies (immunoaffinity depletion), and specific kits (e.g., Proteome Purify Immunodepletion Kit) can be utilized. Numerous proteins are complexed with lipids, and this interaction reduces their solubility. Moreover, by forming complexes with detergents, lipids reduce protein enrichment/separation efficacy. The use of centrifugal filter devices and a sample buffer including CHAPS allows for efficient lipid and salt removal. In order to exclude polysaccharides from the sample, precipitation in TCA, acetone, ammonium sulfate or phenol/ammonium acetate, followed by centrifugation can be performed. In order to remove DNA and RNA, methods such as digestion with protease-free DNase and RNase, or alternatively, protein precipitation from the solution are typically performed.


In a genetic protein variation analysis, fractionating the processed sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample can comprise protein enrichment processes. Various protein enrichment methods can be used to reduce the complexity of the sample by its pre-fractionation, or to enrich it with proteins of interest. Pre-fractionation is performed to isolate a sample into distinguishable fractions containing restricted numbers of molecules. The sample can be fractionated using a variety of approaches including precipitation, centrifugation, liquid chromatography and electrophoresis-based methods, filtration, and velocity or equilibrium sedimentation, among others identifiable by skilled persons.


Fractionating the processed sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample can also comprise removing contaminants. Samples injected onto chromatographic columns cannot contain insoluble particles or dispersed molecules that may cause column clogging and malfunction. Such contaminants are typically removed by centrifugation and/or sample filtration using spin-filters (e.g., 45 μm pores). In addition, samples should not contain buffers affecting LC separation, e.g. samples injected onto column should not be dissolved in buffer with higher eluting strength than of mobile phase. High concentration of detergents should be avoided when using reverse phase separation whereas samples injected on the ion-exchange column should not contain high contraction of background salts and other ionic contaminants that might disturb ionic equilibrium. Volatile buffers such as ammonium acetate or ammonium bicarbonate, are typically used in this case.


In a genetic protein variation analysis, fractionating the processed sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample can comprise any materials and methods or combination of materials and methods for removal of contaminants such as salts, buffers and detergents from the sample, and methods of sample concentration, enrichment, fractionation, filtration, and other methods identifiable by skilled persons upon reading the present disclosure, as described herein or otherwise known in the art can be used to perform fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample.


Genetic protein variation analysis further comprises digesting the solubilized proteins from the sample to obtain digested solubilized proteins from the sample.


In a genetic protein variation analysis, digesting the solubilized proteins from the sample to obtain digested solubilized proteins from the sample can be performed non-enzymatically, e.g., with low pH or high temperatures, as well as enzymatically, e.g., by intra-molecular digestion or with a site specific proteolytic enzyme. In many methods, the digesting is performed with a site specific proteolytic enzyme.


In a genetic protein variation analysis, digesting the solubilized proteins from the sample with a site specific proteolytic enzyme to obtain digested solubilized proteins from the sample can be performed by any method identifiable to a skilled person. As understood by those skilled in the art, the terms “proteolytic enzyme”, “protease”, “peptidase”, and “proteinase” refers to any enzyme that performs proteolysis, wherein the term “proteolysis” as used herein refers to protein catabolism by hydrolysis of peptide bonds.


As understood by those skilled in the art, proteases can be classified into seven broad groups, comprising serine proteases, cysteine proteases, threonine proteases, aspartic proteases, glutamic proteases, metalloproteases, and asparagine peptide lyases.


As understood by those skilled in the art, proteolytic catalysis is achieved by one of two mechanisms, wherein aspartic, glutamic and metallo-proteases activate a water molecule which performs a nucleophilic attack on the peptide bond to hydrolyze it. In contrast, serine, threonine and cysteine proteases use a nucleophilic residue (usually in a catalytic triad). That residue performs a nucleophilic attack to covalently link the protease to the substrate protein, releasing the first half of the product. This covalent acyl-enzyme intermediate is then hydrolyzed by activated water to complete catalysis by releasing the second half of the product and regenerating the free enzyme.


The terms “site specific proteolytic enzyme”, “site specific protease”, “site specific peptidase”, and “site specific proteinase” refer to enzymes that perform proteolysis by cleavage of a protein substrate having a specific sequence. As understood by those skilled in the art, proteolysis can be highly promiscuous such that a wide range of protein substrates are hydrolyzed. This is the case for digestive enzymes such as trypsin which have to be able to cleave the array of proteins ingested into smaller peptide fragments. Promiscuous proteases typically bind to a single amino acid on the substrate and so only have specificity for that residue. For example, trypsin is specific for the sequences . . . KV\ . . . or . . . RV\. . . (‘\’=cleavage site). Conversely some proteases are highly specific and only cleave substrates with a certain sequence. Blood clotting (such as thrombin) and viral polyprotein processing (such as TEV protease) requires this level of specificity in order to achieve precise cleavage events. This is achieved by proteases having a long binding cleft or tunnel with several pockets along it which bind the specified residues. For example, TEV protease is specific for the sequence (SEQ ID No. 86) . . . ENLYFQ\S . . . (‘\’=cleavage site).


Materials and methods for digestion of proteins using various proteases are identifiable by those skilled in the art and described herein.


Genetic protein variation analysis also comprises fractionating the digested solubilized proteins to obtain fractionated digested peptides from the digested solubilized proteins from the biological sample.


Methods to perform fractionating the digested solubilized proteins to obtain fractionated digested peptides from the digested solubilized proteins from the biological sample comprise chromatographic methods. The term “chromatography” as used herein refers to a technique for the separation of a mixture. More specifically, the term “chromatography” is a physical method of separation that distributes components to separate between two phases, one stationary (stationary phase), the other (the mobile phase) moving in a definite direction.


In chromatography, a mixture is dissolved in a fluid called the mobile phase, which carries it through a structure holding another material called the stationary phase. The various constituents of the mixture travel at different speeds, causing them to separate. The separation is based on differential partitioning between the mobile and stationary phases. Subtle differences in a compound's partition coefficient result in differential retention on the stationary phase and thus affect the separation. Chromatography can be preparative or analytical. The purpose of preparative chromatography is to separate the components of a mixture for later use, and is thus a form of purification. Analytical chromatography is done normally with smaller amounts of material and is for establishing the presence or measuring the relative proportions of analytes in a mixture. The two are not mutually exclusive.


As understood by those skilled in the art, chromatography is based on the concept of partition coefficient, wherein any solute partitions between two immiscible solvents. The term “partition coefficient” as defined herein refer to the ratio of concentrations of a compound in a mixture of two immiscible phases at equilibrium, and represents a measure of the difference in solubility of the compound in these two phases. It is also referred to as “distribution coefficient”. When one solvent is made immobile (e.g., by adsorption on a solid support matrix) and another solvent is mobile it results in most common applications of chromatography. As understood by those skilled in the art, if the matrix support, or stationary phase, is polar (e.g. paper, silica etc.) it is referred to as “forward phase” or “normal phase” chromatography, and if it is non-polar (C-18) it is referred to as “reverse phase”.


Chromatography techniques can be categorized according to chromatographic bed shape, wherein “column chromatography” refers to a separation technique in which the stationary bed is within a tube, and “planar chromatography”, which refers to a separation technique in which the stationary phase is present as or on a plane, such as paper chromatography or thin layer chromatography. Accordingly, in some embodiments, any method using column chromatography or planar chromatography can be used to perform fractionating the digested solubilized proteins.


Chromatography techniques can also be categorized according to physical state of mobile phase. The term “gas chromatography” (GC), also sometimes known as “gas-liquid chromatography” (GLC), refers to a separation technique in which the mobile phase is a gas. The term “liquid chromatography” (LC) refers to a separation technique in which the mobile phase is a liquid. In particular, liquid chromatography that generally utilizes very small packing particles and a relatively high pressure is referred to as high performance liquid chromatography (HPLC). In HPLC the sample is forced by a liquid at high pressure (the mobile phase) through a column that is packed with a stationary phase composed of irregularly or spherically shaped particles, a porous monolithic layer, or a porous membrane. HPLC can be divided into two different sub-classes based on the polarity of the mobile and stationary phases. Methods in which the stationary phase is more polar than the mobile phase (e.g., toluene as the mobile phase, silica as the stationary phase) are termed “normal phase” or “forward phase” liquid chromatography, whereas the opposite (e.g., water-methanol mixture as the mobile phase and C18 (octadecylsilyl) as the stationary phase) is termed “reversed phase” liquid chromatography (RPLC).


Accordingly, gas chromatography or liquid chromatography can be used to perform fractionating the digested solubilized proteins in genetic protein variation analysis as will be understood by a skilled person.


Chromatography techniques can also be categorized according to separation mechanism. The term “ion exchange chromatography” refers to a technique that uses an ion exchange mechanism to separate analytes based on their respective charges. The term “size-exclusion chromatography” (SEC) also known as “gel permeation chromatography” (GPC) or “gel filtration chromatography” refers to a technique that separates molecules according to their size, or more accurately according to their hydrodynamic diameter or hydrodynamic volume. The term “expanded bed chromatographic adsorption” (EBA) refers to a biochemical separation process using a column that comprises a pressure equalization liquid distributor having a self-cleaning function below a porous blocking sieve plate at the bottom of the expanded bed, an upper part nozzle assembly having a backflush cleaning function at the top of the expanded bed, and a better distribution of the feedstock liquor added into the expanded bed ensuring that the fluid passed through the expanded bed layer displays a state of piston flow.


Accordingly, ion exchange chromatography, size-exclusion chromatography, or expanded bed chromatographic adsorption can be used to perform fractionating the digested solubilized proteins in genetic variation protein analysis of the instant disclosure. Other chromatography techniques can be used such as hydrophobic interaction chromatography, two-dimensional chromatography, simulated moving-bed chromatography, pyrolysis gas chromatography, fast protein liquid chromatography, countercurrent chromatography, periodic counter-current chromatography, aqueous normal-phase chromatography, or chiral chromatography, among others identifiable by persons skilled in the art can be used to perform fractionating the digested solubilized proteins.


In general, techniques identifiable by skilled persons that can be used to perform fractionating proteins or digested proteins of a biological sample comprise methods based on purification of peptides according to their isoelectric points (e.g., by running them through a pH graded gel or an ion exchange column), separation according to their size or molecular weight (e.g., via size exclusion chromatography or by SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) analysis), or separation by polarity/hydrophobicity (e.g., via high performance liquid chromatography or reversed-phase chromatography).


Additional methods for fractionating proteins or digested proteins of a biological sample that can be used in some embodiments described herein comprise affinity chromatography. The term “affinity chromatography” refers to a separation technique based upon molecular conformation, which frequently utilizes application specific resins. These resins have ligands attached to their surfaces which are specific for the compounds to be separated. For example, immunoaffinity chromatography uses the specific binding of an antibody-antigen to selectively purify the target protein. The procedure involves immobilizing a protein to a solid substrate (e.g. a porous bead or a membrane), which then selectively binds the target, while everything else flows through. The target protein can be eluted by changing the pH or the salinity. The immobilized ligand can be an antibody (such as Immunoglobulin G) or it can be a protein (such as Protein A), among others identifiable by those skilled in the art.


Genetic protein variation analysis also comprises detecting a marker genetic variation of the fractionated digested peptides.


Various techniques can be used to perform detecting a marker genetic variation of the fractionated digested peptides in a genetic variation protein analysis, such as mass spectrometry. Mass Spectrometry (MS) is an analytical technique that ionizes chemical species and sorts the ions based on their mass-to-charge ratio. In simpler terms, a mass spectrum measures the masses within a sample. Mass spectrometry is used in many different fields and is applied to pure samples as well as complex mixtures. A mass spectrum is a plot of the ion signal as a function of the mass-to-charge ratio. These spectra are used to determine the elemental or isotopic signature of a sample, the masses of particles and of molecules, and to elucidate the chemical structures of molecules, such as peptides and other chemical compounds.


The terms “liquid chromatography mass-spectrometry” or “LC-MS” as used herein refer to an analytical chemistry technique that combines the physical separation capabilities of liquid chromatography (LC, or high-performance liquid chromatography, HPLC, or ultra-high-performance liquid chromatography, UHPLC) with the mass analysis capabilities of mass spectrometry (MS). The terms “tandem mass spectrometry”, or “MS/MS” as used herein refers to a mass-spectrometry technique that involves more than one stage of mass spectrometry analysis, with a step form of fragmentation occurring in between the stages. In a tandem mass spectrometer, ions are formed in the ion source and separated by mass-to-charge ratio in the first stage of mass spectrometry (MS1). Ions of a particular mass-to-charge ratio (precursor ions) are selected and fragment ions (product ions) are created by collision-induced dissociation, ion-molecule reaction, photodissociation, or other processes. The resulting ions are then separated and detected in a second stage of mass spectrometry (MS2). Thus, the terms “tandem liquid chromatography mass-spectrometry” and “LC-MS/MS” as used herein refer to a technique that couples liquid chromatography and tandem mass-spectrometry.


Typically, for LC-MS/MS proteomic analysis, the stationary LC phase is a C18 reverse-phase column. The reverse-phase column uses the hydrophobicity of peptides for separation, utilizing a gradient from low to high organic-phase solvent. Acidified methanol and acetonitrile are commonly used as organic-phase, also known as “B” or “strong”, solvents because of their miscibility with aqueous solutions. Acidified water is most often the “weak” solvent, also known as “A”. Both buffers are acidified with the same acid, generally with formic acid or trifluoroacetic acid (TFA) at 0.1% or 0.01%, respectively.


Examples of tandem mass-spectrometry instruments used for LC-MS/MS proteomics analysis comprise sector instruments, time-of-flight instruments, quadrupole mass analyzers, ion traps, and orbitraps, among others identifiable by those skilled in the art.


In proteomic analysis using LC-MS/MS, following purification of proteins from tissue samples, the purified proteins are enzymatically digested by a protease, typically, trypsin, which cleaves the protein into smaller detectable peptides, with molecular weights of about 400 to 4000. The peptides are then resolved using very low flow rate liquid chromatography, such as reversed phase liquid chromatography, and are then ionized and vaporized using methods such as fast atom bombardment (FAB), chemical ionization (CI), atmospheric-pressure chemical ionization (APCI), electrospray ionization (ESI), and matrix-assisted laser desorption/ionization (MALDI). The charged peptide is then funneled using electric fields into the mass spectrometer where its mass is measured (MS1). The instrument then fragments individual peptide backbones using either collision-induced or electron transfer dissociation and the resulting fragment masses are also measured (MS2). Both of these fragmentation methods break the peptide backbone at regular points. This allows the amino acid sequence to be determined. The information from tandem liquid chromatography mass-spectrometry, therefore, has three dimensions: time of retention on reversed phase, peptide mass (MS1) and individual peptide fragmentation masses (MS2). Mass spectrometry has matured to the point where over 10,000 peptide fragmentations can be obtained per run. The mass accuracy of peptide and fragmentation masses is now 1 ppm in both MS and MS2, removing ambiguity from the analysis.


The fragmentation data can be resolved using the data within the sample, based on the intrinsic properties of the data related to the peptide fragmentation, to provide de novo sequence information through a de novo peptide identification algorithm for LC-MS/MS which infers peptide sequences without knowledge of genomic data. Examples of de novo sequencing algorithms comprise Cyclobranch, DeNovoX, DeNos, Lutefisk, Novor, PEAKS, and Supernovo, among others identifiable by those skilled in the art.


The fragmentation data can also be resolved through comparison with predicted sequences derived from genomic and protein databases such as GenBank and UniProt. This method provides a statistical measure of probability that any fragmentation dataset is the predicted amino acid sequence through a database search peptide identification algorithm for LC-MS/MS which takes place against a database containing all amino acid sequences assumed to be present in the analyzed sample. Examples of database search algorithms comprise Andromeda, Byonic, Comet, Tide, Greylag, InsPecT, Mascot, MassMatrix, MassWiz, MS Amanda, MS-GF+, MyriMatch, OMSSA, PEAKS DB, pFind, Phenyx, ProblD, ProteinPilot Software, Protein Prospector, RAId, SEQUEST, SIMS, Sim Tandem, SQID, and X!Tandem, among others identifiable by those skilled in the art.


The allelic frequencies associated with each nucleotide and amino acid polymorphism within the fragmentation data are a product of the reference populations used in the single nucleotide polymorphism (SNP) data bases. The term “allelic frequency” as defined herein refers to the relative frequency of an allele (variant of a gene) at a particular locus in a population, expressed as a fraction or percentage. Examples of databases of human SNPs and SAPs comprise dbSNP, which is a SNP database from the National Center for Biotechnology Information (NCBI), as well as the 1000 Genomes Project, UniProt, Protein Mutation Database, HPMD, MSIPI, MS-CanProVar, Ensembl, COSMIC, and dbSAP [2], among others identifiable by those skilled in the art.


Accordingly, in a genetic protein variation analysis, any method of mass-spectrometry identifiable by skilled persons can be used to perform detecting a marker genetic variation of the fractionated digested peptides, such as techniques that use time-of-flight instruments, quadrupole mass analyzers, ion traps, and orbitraps, among others identifiable by those skilled in the art, that use any ionization and vaporization methods such as fast atom bombardment (FAB), chemical ionization (CI), atmospheric-pressure chemical ionization (APCI), electrospray ionization (ESI), and matrix-assisted laser desorption/ionization (MALDI), among others identifiable by skilled persons. Additionally, any method of peptide fragmentation known in the art, such as collision-induced or electron transfer dissociation can be used to detect a marker genetic variation of the fractionated digested peptides, and any method of peptide fragmentation data deconvolution, such as de novo sequencing, or comparison of peptide fragmentation data with predicted sequences derived from genomic and protein databases such as GenBank and UniProt can be used to perform detecting a marker genetic variation of the fractionated digested peptides.


Additionally, in a genetic variation protein analysis any peptide identification algorithms that can be used in database searches, such as Andromeda, Byonic, Comet, Tide, Greylag, InsPecT, Mascot, MassMatrix, MassWiz, MS Amanda, MS-GF+, MyriMatch, OMSSA, PEAKS DB, pFind, Phenyx, ProblD, ProteinPilot Software, Protein Prospector, RAId, SEQUEST, SIMS, Sim Tandem, SQID, and X!Tandem, among others identifiable by those skilled in the art, or in de novo searches, such as Cyclobranch, DeNovoX, DeNos, Lutefisk, Novor, PEAKS, and Supernovo, among others identifiable by those skilled in the art, can be used to perform detecting a marker genetic variation of the fractionated digested peptides. Additionally, in some embodiments, any databases of human SNPs and SAPs such as dbSNP, 1000 Genomes Project, UniProt, Protein Mutation Database, HPMD, MSIPI, MS-CanProVar, Ensembl, COSMIC, and dbSAP [2], among others identifiable by those skilled in the art can be used to perform detecting a marker genetic variation of the fractionated digested peptides.


An exemplary genetic protein variation analysis including specific protocols for performance of the related steps is shown in the paper Parker et al 2016 [3] incorporated herein by reference in its entirety and supplementary information of Parker et al. (2016) incorporated herein by reference in its entirety.


In a genetic protein variation analysis performed with methods and systems in accordance with the present disclosure, preparing the sample and/or detecting a genetic variation can be performed by any one of the methods and/or using anyone of the systems and databases according to any one of the first aspect to the seventh aspect of the present disclosure.


Accordingly, in some embodiments, preparing a biological sample to obtain a processed biological sample comprising solubilized proteins to be used in proteomic analysis can be performed by the method to prepare a biological sample for proteomic analysis according to the first aspect of the present disclosure. The method comprises applying to the biological sample an energy field to obtain a processed biological sample comprising solubilized proteins to be used in the proteomic analysis.


In particular, the energy field applied in methods for preparing a biological sample according to the first aspect of the disclosure comprises electromagnetic fields applied with parameters selected to result in protein solubilization while reducing breakage of the intramolecular peptidic bonds of the proteins in the sample.


In a method for preparing a biological sample according to the first aspect of the disclosure, typically, energy is applied at the initial solubilization stage of sample processing. Sample solubilization process typically involves the use of chaotropes (e.g. urea and/or thiourea), detergents (e.g. 3-[(3-Cholamidopropyl)-dimethyl-ammonio]-1-propane sulfonate (CHAPS) or Triton X-100), reducing agents (dithiothreitol/dithioerythritol (DTT/DTE) or tributylphosphine (TBP)) and protease inhibitors in a sample buffer.


In some embodiments the sample buffer can comprise reducing agents such as DTT, Dodecyltrimethylammonium bromide (DTBA), Betamercatptoethanol (BME), tris(2-carboxyethyl)phosphine (TCEP), and DTE. In particular, the applying can be performed with detergent in concentration ranging from 0.001 M to 10 M; 0.05 M to 0.2 M more preferably; and most preferably 0.1 M. In preferred embodiments the detergent comprises DTT.


In some embodiments the sample buffer can comprise detergents such as SDD, SDS, CHAPS, a Triton X-100, Lithium Dodecyl Sulfate (LDS)Tergitol-type NP-40 (NP-40) which is nonyl phenoxypolyethoxylethanol, commercially available with CAS 9016-45-9. The detergent concentrations depend on temperature and ultrasonic treatment time as will be understood by a skilled person. Specifically, decreasing SDD concentration by 1% drastically increases time for solubilization (60 minutes to 24 hours), whereas decreasing ultrasonic treatment incubation temperature also increases time (every 5 degrees C. decreased requires two hours or more ultrasonic treatment time). Increasing detergent concentration past 2% does not result in significant decreased ultrasonic incubation time. In preferred embodiments, the detergent comprises SDD.


A skilled person will understand that the composition of the sample buffer can vary depending on the time and condition of applying to the biological sample an energy field and can be adjusted by a skilled person to optimize protein solubilization upon reading of the present disclosure.


The term “solubilize”, used herein with reference to solubilized proteins, refers to a transfer of proteins comprised within the biological sample to a solvent such as an aqueous solvent by disrupting the cells of the biological sample. Disruption of the cells of the biological sample can be performed by applying a force to the cell to alter the cell membrane continuity and integrity for a time and under condition to result in the lysis of the cell.


In some preferred embodiments, applying to the biological sample an energy field can be performed by sonication. The sonication process can be carried out using an ultrasonic processor operating at the ultrasound frequency of about 20-80 kHz and applying the sample the ultrasound for about 30-120 minutes. In some embodiments, the sonication process can be performed using an ultrasonic processor set to 1 to 100 kHz; preferably 5 to 50 kHz and more preferably 37 kHz.


In embodiments, wherein applying energy is performed by sonication, the power setting of the device can range from 1 to 100%; more preferably 50 to 100%; most preferably 100%.


In embodiments, wherein applying energy is performed by sonication, the applying can be performed by providing the energy with an ultrasonic mode selected from sweep, degas, and pulse. In preferred embodiments, applying energy can be performed by providing the energy with ultrasonic mode sweep.


In the preferred embodiments, wherein applying energy is performed by sonication, which includes any method for imparting acoustic energy to bring about cavitation of the sample including sonication baths, sonication probes/sonicators, or sonication flow-through systems are applicable. The biological sample can be subjected to sonication by placing a sample containing tube with a sonication bath or samples can be directly sonicated using a probe or by placing in a flow-through system directly.


As a person skilled in the art will understand, other mechanical cell disruption methods capable of creating high stress via pressure or abrasion with rapid agitation can also be used to mechanically disrupt the biological sample. Exemplary mechanical cell disruption methods include bead milling, cryomilling, microfluidizers, high pressure homogenizer, nitrogen cavitation, and others identifiable to a person skilled in the art.


In some other embodiments, applying to the biological sample an energy field through the application of microwaves can be performed by microwaving the biological sample using 500-1,200 Watt microwaves, wherein samples can be treated from 10 seconds to several minutes [4-7].


In some embodiments, applying energy can be performed with an incubation time ranging from 5 to 1,440 minutes; more preferably 20 to 90 minutes; most preferably 60 minutes.


In some embodiments, applying energy can be performed with temperature settings from 15 to 100° C.; more preferably 30 to 90° C.; most preferably 70° C.


The time and temperature of applying to the biological sample an energy field in accordance with the first aspect of the disclosure depend on the composition of the sample buffer as will be understood by a skilled person. For example, in embodiments where the applying is performed by sonication, presence and concentration of a detergent in the sample buffer depend on temperature and ultrasonic treatment time as will be understood by a skilled person. In particular, decreasing concentration of a detergent such as SDD, by 1% drastically increases time for solubilization (60 minutes to 24 hours). Whereas decreasing ultrasonic treatment incubation temperature also increases time (every 5 degrees C. decreased requires two hours or more ultrasonic treatment time). Increasing concentration of a detergent such as SDD in the sample buffer past 2% does not result in significant decreased ultrasonic incubation time. Additional adjustments and variations of the sample buffer compositions, time and temperature of applying to the biological sample an energy field in accordance with the first aspect of the disclosure are identifiable by a skilled person upon reading of the present disclosure.


In some embodiments the biological sample is a tissue sample. The term “tissue” as used herein refers to a cellular organizational level intermediate between cells and a complete organ or organism. A tissue is typically an ensemble of similar cells from the same origin that together carry out a specific function. Organs and organisms are then formed by the functional grouping together of multiple tissues. As used herein, the term tissue comprises ensembles of cells such as hair, skin, bone, teeth, blood and other body fluids, muscle, nerves, and other cellular material originating from one or more organisms, and also comprises artifacts originating from tissues such as fingerprints. In particular, as used herein, organisms from which tissues originate comprise mammals and in particular humans.


In some embodiments, the biological sample comprises hair. Hair is commonly found as trace evidence in crimes scene forensic investigations. Persistence of hair in the environment demonstrates the unique chemical stability that makes it an ideal biological material for analysis by forensic practices [8]. Largely, forensic analysis of hair evidence is performed by microscopic analysis of morphological characteristics and more recently mitochondrial DNA (mtDNA) sequencing. Both accepted techniques have intrinsic flaws (subjectivity and low discrimination, respectively) highlighting the essential need for development of new strategies to obtain information from hair evidence in the forensic communities [9, 10].


Specifically, proteomic analysis of hair has been shown to provide identification markers in the form of genetically variant peptides (GVPs) in human samples [3]. GVP detection targets mutations in protein amino acid sequences as a direct reflection of single-nucleotide polymorphisms (SNPs) found in DNA. The utility of this technique in forensic practice hinges on its ability to apply to practical sample sizes, for example a single hair.


In some embodiments, the biological sample can be a single hair. In some embodiments, the single-hair sample is about 0.1 to 20 cm in length, such as 2.5 cm, and 2-1630 μg in weight, such as 85 μg in some examples (see e.g. Example 2). Providing a single-hair sample can further comprise cutting the single-hair sample into pieces.


In some embodiments, the method of preparing the biological sample comprises providing a single-hair sample from an individual, dissolving the single-hair sample in a cell lysis solution, subjecting the cell lysis solution containing the single-hair sample to ultrasonication or thermolysis to provide a solubilized single-hair sample, and digesting the solubilized single-hair sample to obtain peptide samples. The obtained peptide samples are then subjected to proteomics analysis.


Exemplary methods to perform a proteomic tissue sample preparation using methods according to the first aspect and single hairs are described in Examples 2-4.


In some embodiments detecting a genetic variation can be performed with a method and system to provide a marker genetic protein variation for a biological organism and a marker genetic protein variation obtainable thereby according to the second aspect of the present disclosure. In these method and system, the provided marker genetic protein variation validated to be detectable and in particular proteomically detectable in a biological sample of an individual of the biological organism.


The method comprises: providing a marker exome sequence of the biological organism, the marker exome sequence comprising a marker genetic variation for the biological organism.


The method further comprises detecting peptide sequences in the biological sample of the individual of the biological organism by performing proteomic analysis of said biological sample to provide proteomically detected peptide sequences.


The method also comprises providing the marker genetic protein variation of the biological organism detectable in the sample of the biological organism by comparing the provided marker exome sequence with the proteomically detected peptide sequences to provide a marker genetic protein variation validated for the biological sample of an individual of the biological organism.


The system comprises exome sequence databases and/or reagents to detect exome sequences in an individual of the biological organism, in combination with reagents to perform proteomic analysis of the biological sample for simultaneous combined or sequential use in the method to provide a marker genetic protein variation validated for a biological sample herein described.


The term “exome” as used in the instant disclosure indicates the part of the genome of a biological organism composed of exons, the sequences which, when transcribed, remain within the mature RNA after introns are removed by RNA splicing and contribute to the final protein product encoded by that gene.


In some embodiments, providing at least one marker exome sequence from a genome each comprising a genetic variation of the genome comprises detecting exome sequences of the genome by sequencing exomes of the genome and detecting at least one marker exome sequence each comprising a genetic variation of the genome by comparing the detected exome sequences with a database of exome sequences of the biological organism.


The genome being sequenced for detecting exome sequences can be of the same individual of the biological organism where the biological sample is collected from for proteomic analysis, or a close relative of the individual who has a coefficient of relationship (r) of at least 0.5 with the individual. Herein, the coefficient of relationship is a measure of the degree of consanguinity or biological relationship between two individuals. For example, a parent and child pair have a value of r=0.5 and full siblings have a value of r=0.5.


Sequencing exomes of a genome can comprise collecting a sample from the individual and performing exome sequencing of the sample. In some instances, the sample is a blood sample or buccal sample. The type of sample collected from the individual for the exome sequencing can be the same or different from the type of sample collected for the proteomics analysis. For example, in some instances, the sample collected for the exome sequencing can be a blood sample while the biological sample collected for proteomic analysis can be a hair sample.


The exome sequencing can be performed by whole exome sequencing (WES or WXS). Whole exome sequencing typically comprises selecting the subset of DNA containing exons from the whole genome. Both array-based and in-solution capture techniques can be used to selectively capture the subset of DNA containing exons. The subset of DNA containing exons can then be sequenced using high-throughput DNA sequencing technology.


High-throughput DNA sequencing also referred to as next-generation sequencing (NGS) refers to a number of different modern nucleic acid sequencing technologies including Illumia™ sequencing, Roche 454™ sequencing, Ion torrent: Protein/PGM™ sequencing and SOLiD™ sequencing. Next-generation sequencing (NGS) generally refers to non-Sanger-based high-throughput DNA sequencing technologies. The NGS technologies can be based on immobilization of the nucleotide samples onto a solid support, cyclic sequencing reactions using automated fluidics devices and detection of molecular events by imaging. Cyclic array platforms achieve low costs by simultaneously decoding a two-dimensional array bearing millions or billions of distinct sequencing features, each containing one species of DNA physically immobilized on an array. In each cycle, an enzymatic process is applied to interrogate the identity of a single base position for all features in parallel. The enzymatic process is coupled to either the production of light or the incorporation of a fluorescent group. At the end of each cycle, data are acquired by imaging of the array. Subsequent cycles are typically performed interrogating different base position within the sequence. Detailed information about various next-generation sequencing approaches can be found in related literation and documents and will be understood by a person skilled in the art.


In some embodiments of the present disclosure exome sequencing can be performed by RNA exome sequencing e.g. with (e.g., with Illumina RNA Exome Capture Sequencing) as will be understood by a skilled person.


In particular, in certain tissue types (either coextracted in sample; e.g. skin or bone or from separate buccal swab) exome sequencing can be performed from RNA in the sample. In particular, in some embodiments the exome sequencing can be performed on the protein fraction of the sample wherein GVPs can be fractionated with their mRNA counterparts. In some embodiments exome sequencing can be performed following RNA extraction of samples (cell lysis, solubilization, purification) using a portion of a sample or a buccal swab and RNA-sequencing performed with technologies such as RNA-seq, RNA capture exome sequencing, and addition technologies identifiable by a skilled person RNA sequences can be translated into DNA subsequently and provide the presence/absence of missense SNPs that correspond to GVPs.


Detecting at least one marker exome sequence can be performed by comparing the detected exome sequences of the individual with a database of exome sequences of the biological organism. In general, the exome sequences generated from a sequencing procedure can be aligned to the sequence entries contained in the database of exome sequences of the biological organism using alignment/assembly tools identifiable by a person skilled in the art. Exemplary database of exome sequences of the biological organism includes the NHLBI Exome Sequencing Project (ESP) database.


In particular, the detected marker exome sequences are a set of exome sequences, each comprising one or more single nucleotide polymorphisms. Therefore, comparing the detected exome sequences of the individual with a database of exome sequences of the biological organism can identify one or more non-synonymous single nucleotide polymorphisms in the exome sequence of the individual.


The method further comprises detecting peptide sequences in the biological sample by performing proteomic analysis of the biological sample. The term “proteomic analysis” refers to the systematic identification and quantification of the complete set of proteins encoded in a biological system such as a cell, tissue, organ, biological fluid or organism. Proteomic analysis can be performed using mass spectrometry (MS) or liquid chromatography mass-spectrometry (LC-MS) as will be understood by a person skilled in the art. Performing proteomic analysis of the biological sample comprises fragmenting proteins in the biological sample into peptides, subjecting the fragmented sample to MS or LC-MS to obtain proteomic datasets, and analyzing the proteomic datasets to identify the peptide sequences of the biological sample. Analyzing the proteomic datasets can be performed using computational algorithm such as MASCOT, GPM or Petunia as will be understood by a person skilled in the art.


In certain embodiments, the proteomics analysis performed on the biological sample is shotgun proteomics analysis. Shotgun proteomic analysis refers to the use of bottom-up proteomics techniques in identifying proteins in complex mixtures using a combination of high performance liquid chromatography combined with mass spectrometry, and is an alternative to targeted proteomics and data-independent acquisition proteomics.


The method according to the second aspect of the instant disclosure, further comprises providing the marker genetic protein variation of the biological organism in the biological sample by comparing the detected marker exome sequence with the detected peptide sequences to provide a marker genetic protein variation validated for the biological organism.


The comparison can be performed by comparing each detected marker exome sequence comprising a generic variation of the genome such as SNPs with the detected peptide sequences stored in a database. The comparison can be carried out by any sequence comparison programs that compare a DNA sequence to a peptide sequence database, such as BLASTX. Such sequence comparison programs typically involve translating the DNA sequence in three frames and aligning the translated DNA sequence to each sequence in the peptide database, allowing gaps and frameshifts as will be understood by a person skilled in the art.


The detected marker exome sequence having a corresponding entry in the database containing the detected peptide sequences is then indicated as a marker genetic protein variation validated for the biological organism. The marker genetic protein variation validated for the biological organism can be further stored in a database which contains, for each data entry, a detected marker exome sequence comprising a genetic variation and a peptide sequence corresponding to the detected marker exome sequence. The data entry can further comprise an allele frequency for the genetic variation in the detected marker exome sequence.


In some embodiments, the biological organism is Homo sapiens. In some embodiments, the biological sample is a hair sample.


Exemplary validated marker exome sequences of Homo Sapiens are indicated in Examples 43 to 45 listing exemplary set of genes validated as being detectable in hair samples (Example 43, Table 8) bone samples (Example 44, Table 9) and skin samples (Example 45, Table 10) of a human being.


Exemplary validated marker genetic protein variations of Homo Sapiens are indicated in Examples 46 and Example 47 listing exemplary set of GVPs validated in hair samples (Example 46, Table 11) and skin samples (Example 47, Table 12) of a human being.


In some embodiments detecting a genetic variation can be performed with a method and system to detect a marker genetic protein variation in a biological sample according to a third aspect of the present disclosure. In the method and system, the marker genetic protein variation are validated to be detectable and in particular proteomically detectable in the biological sample.


The method comprises providing a marker mass spectrum of a marker peptide comprising a marker genetic protein variation corresponding to the genetic protein variation; and performing mass spectrometry of a fractionated digested peptide of the biological sample to obtain a mass spectrum of each of the fractionated digested peptide.


The method further comprises comparing the mass spectrum of the fractionated digested peptide with a marker mass spectrum of a marker peptide comprising the marker genetic protein variation to detect the genetic protein variation in the biological sample.


The system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases for simultaneous combined or sequential use in the method to detect a marker genetic protein variation in a biological sample herein described. In preferred embodiments, the reagents comprise one or more marker peptides in accordance with the present disclosure.


In the method according to the third aspect, any method of performing mass spectrometry of a fractionated digested peptide of the biological sample as described herein or otherwise identifiable by persons skilled in the art can be used to obtain a mass spectrum of each of the fractionated digested peptides.


As understood by skilled persons, mass-spectrometry of fractionated digested peptides of a biological sample can produce a large number of mass spectra. In embodiments described herein, the term “mass spec dataset” is used to refer to a plurality of mass spectra obtained for a plurality of fractionated digested peptides of a biological sample (e.g., see FIG. 9).


As understood by persons skilled in the art, mass spectrometry (MS) is an analytical technique that ionizes chemical species and sorts the ions based on their mass-to-charge ratio. In simpler terms, mass spectrometry measures the masses within a sample. Mass spectrometry is used in many different fields and is applied to pure samples as well as complex mixtures. The term “mass spectrum” as used herein refers to a plot reporting a signal of one or more ions as a function of mass-to-charge ratio of the ions. Accordingly, mass spectra can be used to determine the elemental or isotopic signature of a sample, the masses of particles and of molecules, and to elucidate the chemical structures of molecules, such as peptides and other chemical compounds.


The terms “tandem mass spectrometry”, or “MS/MS” as used herein refer to a mass-spectrometry technique that involves more than one stage of mass spectrometry analysis, with a step of fragmentation occurring in between the stages. In a tandem mass spectrometer, ions are formed in the ion source and separated by mass-to-charge ratio in the first stage of mass spectrometry (MS1). Ions of a particular mass-to-charge ratio (precursor ions) are selected and fragment ions (product ions) are created by collision-induced dissociation, ion-molecule reaction, photodissociation, or other processes. The resulting ions are then separated and detected in a second stage of mass spectrometry (MS2).


Accordingly, a mass spectrum of a peptide is a plot reporting a signal of one or more ions of a peptide as a function of mass-to-charge ratio of the ions. In particular, with reference to LC-MS/MS analysis of peptides (e.g. peptides produced by digesting proteins of a biological sample using a site-specific protease), a mass spectrum of a peptide can refer to a mass spectrum produced in the MS1 stage or the MS2 stage, wherein the mass spectrum produced in the MS1 stage refers to a mass spectrum of a peptide (e.g. a peptide produced by digesting a protein using a site specific protease) before fragmentation of the peptide occurs, and the mass spectrum produced in the MS2 stage refers to a mass spectrum produced after fragmentation of the peptide has occurred.


The term “marker peptide” as used herein refers to a peptide that comprises a genetic protein variation. In some embodiments, a marker peptide is a peptide produced by digesting a protein that comprises a genetic protein variation, wherein the marker peptide is the peptide produced by proteolytic digestion that comprises the genetic protein variation. In some embodiments, the genetic protein variation is encoded by a ‘rare’ non-synonymous single nucleotide polymorphism (nsSNP) having an allelic frequency lower than 0.5% or a ‘private’ nsSNP having an allelic frequency lower than 0.1% in a given population, wherein an allelic frequency is a product of the reference populations used in the single nucleotide polymorphism (SNP) data bases.


Accordingly, the terms “marker mass spectrum of a marker peptide” or “diagnostic LC-MS/MS spectrum” as used herein refer to a mass spectrum of a marker peptide. In some embodiments, the terms “marker mass spectrum of a marker peptide” or “diagnostic LC-MS/MS spectrum” as used herein refer to a mass spectrum of a marker peptide that is produced in the MS1 stage, or a mass spectrum of a marker peptide that is produced in the MS2 stage.


In some embodiments, the amino acid sequence of a marker peptide can be provided by first sequencing an exome of an individual, detecting a genetic variation comprised in a sequence of the exome of the individual, providing the corresponding encoded genetic protein variation by providing a translation of the exome sequence comprising the genetic variation, and providing the amino acid sequence of the peptide produced as a result of digesting the peptide using a site-specific protease (e.g. trypsin) (e.g., see FIG. 17). In other embodiments, an amino acid sequence of a marker peptide can be provided without reference to a specific individual exome sequence, but rather based on known marker peptide sequences, for example from a database such as dbSNP and others identifiable by skilled persons upon reading of the present disclosure.


In some embodiments, the amino acid sequence of a marker peptide for identification of an individual can be provided by sequencing the exomes of individuals related to the individual. In some embodiments, the individuals related to the individual can form a mother-father-child relationship.


Exemplary marker peptides comprising genetic protein variations are indicated in Examples 46 and Example 47 indicating exemplary set of GVPs and related mutated peptides validated in hair (Example 46, Table 11) and skin (Example 47, Table 12) samples. The marker peptides of Table 11 and Table 12 can be used in connection with method performed on biological samples from a human being.


In particular exemplary marker peptides that can be preferably used or comprise in the method and system according to the third aspect, comprise any combination of the peptides having sequence SEQ ID NO: 146 to SEQ ID NO: 748 (Example 46, Table 11) for detection in hair samples of human beings, and any combination of the peptides having sequence SEQ ID NO: 749 to SEQ ID NO: 829 (Example 47, Table 12) for detection in skin samples of human beings.


In some embodiments, a marker mass spectrum of a marker peptide can be provided by synthesizing a marker peptide and analyzing the marker peptide using LC-MS/MS. For example, peptides can be synthesized using biosynthetic methods, such as cell-based methods or cell-free methods known to those skilled in the art. Peptide biosynthesis can be performed by translation of DNA or RNA polynucleotides encoding the peptide. Thus, protein biosynthesis can be performed by providing cell-based or cell-free peptide translation systems with DNA or RNA polynucleotides encoding the peptide. Peptides can also be produced by liquid phase or solid-phase chemical peptide synthetic methods known to those skilled in the art. In other embodiments, a marker mass spectrum of a marker peptide can be provided by generating the mass spectrum in silico based on the predicted fragmentation products of the peptide as would be produced in the MS2 stage.


With regard to the method to detect a genetic protein variation in a biological sample according to the third aspect of the present disclosure, any method of performing mass spectrometry of a fractionated digested peptide of the biological sample as described herein or otherwise identifiable by persons skilled in the art can be used to obtain a mass spectrum of each of the fractionated digested peptides.


As understood by skilled persons, mass-spectrometry of fractionated digested peptides of a biological sample can produce a large number of mass spectra. In embodiments described herein, the term “mass spec dataset” is used to refer to a plurality of mass spectra obtained for a plurality of fractionated digested peptides of a biological sample (e.g., see FIG. 9).


In some embodiments, the step of comparing the mass spectrum of the fractionated digested peptides of the biological sample with a marker mass spectrum of a marker peptide as described herein can be performed without reference to a protein variant database.


In particular, in embodiments described herein, a mass spec data set produced from a set of fractionated digested peptides of a biological sample (e.g. an operational sample) can be spectrally searched directly with reference to a marker mass spectrum (e.g. see FIG. 17). The spectral searching with reference to the marker mass spectrum can be performed using commercially available or open source software such as MASCOT, PEAKS, and GPM, as well as others identifiable by those skilled in the art and described herein. Upon comparing the mass spec data set of the biological sample with a marker mass spectrum of a marker peptide, a detected identity between the marker mass spectrum of a marker peptide and a mass spectrum of a peptide of the biological sample indicates that the marker peptide is present in the biological sample (e.g., see FIG. 17).


In some embodiments, stable isotope labeled peptide standards can be used in the method to detect a genetic protein variation in a biological sample. For example, an internal standard of the marker peptide labeled with multiple stable isotopes (e.g., D replacing H residues in the peptide) can be added to the fractionated digested proteins of the biological sample analyzed by LC-MS/MS, so that the standard co-elutes with the native peptide to assist with identification, wherein the mass of the internal standard is shifted so that it doesn't interfere with the analysis. Stable isotopes of peptides can be obtained commercially (e.g., from Sigma Aldrich).


Accordingly, in some embodiments, a detected identity between the marker mass spectrum of a marker peptide and a mass spectrum of a peptide of the biological sample can be used to confirm the prior presence of an individual at a sample site (e.g., see FIG. 18).


In some embodiments, in the case of a detected identity between the marker mass spectrum of a marker peptide and a mass spectrum of a peptide of the biological sample, the spectral matching can be used to confirm the prior presence of an individual at a sample site when the biological sample comprises proteins from a plurality of individuals (e.g., see FIG. 18).


In some embodiments detecting a genetic variation can be performed with a database obtainable with methods and systems according to a fourth aspect of the present disclosure. According to the fourth aspect, a method and system to improve a marker genetic protein variation database system for a biological organism, and a database obtainable thereby, are described. In the method, system and database herein described, the marker genetic protein variation database system includes data for at least one biological organism and the improvement is inclusion of one or more marker genetic proteins validated to be detectable and in particular, proteomically detectable in the biological sample from an individual of the at least one biological organism.


In particular the methods and systems of the fourth aspect of the instant disclosure are based on a top-bottom exome-driven approach which begins with obtaining exome data, allowing identification of relevant SNPs, followed by proteomic validation of GVPs.


The method according to the fourth aspect comprises: producing a proteomic dataset from a biological sample from an individual of the at least one biological organism and comparing the proteomic dataset to a protein variant database to produce a set of proteomically detected proteins in the biological sample of the individual.


The method further comprises providing a set of represented genes proteomically detectable in the biological sample of the individual, the represented genes corresponding to the proteomically detected proteins in the biological sample of the individual.


The method also comprises: identifying a marker genetic protein variation validated for the biological sample of the individual, to be included in the marker genetic protein variation database system by providing a proteomically detectable genomic variation in the set of represented genes proteomically detectable in the biological sample of the individual, and providing the marker genetic protein variation validated genetic protein variation by providing a proteomically detectable genetic protein variation corresponding to the proteomically detectable genomic variation in the biological sample of the individual.


In some embodiments the proteomic data set is a mass spectrometry dataset.


The system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases for simultaneous combined or sequential use in the method to improve a marker genetic protein variation database system for a biological organism herein described.


Herein, “database” refers to an organized collection of information. “Database system” refers to a system that includes at least one computer for the creation and storage of a database in computer memory. The database system can be stand-alone, distributed (networked), cloud-based (i.e. networked in a cloud computing system), or any standard database configuration. The database system can be shared among applications or dedicated to a single application. The database system can be local or remote. The database can be navigational, relational, object model, document model, flat file, associative, array, multidimensional, semantic, or any other logical structure. “Protein variant database” refers to a database of variant proteins or protein isoforms that are members of a set of highly similar proteins that originate from a single gene or gene family and are the result of genetic differences.


Detected proteins from the biological sample are determined by a proteomic analysis of the mass spectra obtained from individual biological samples. That proteomic analysis involves one or more databases which contain the protein sequences and their accession numbers. The proteins identified in the sample are then related though their unique protein accession numbers to the genes that code for them (the represented genes). This permits linking the observed protein with the responsible gene and therefore the associated statistics for that gene (SNPs, frequencies, etc.).


The mass spectrometry dataset can be obtained by taking the biological sample, for example one prepared by as described herein by dissolving, ultrasonication, and digestion, and running it through a mass spectrometer to determine a mass spectrum of the sample. Mass spectrometry can include hard ionization, soft ionization, inductively coupled plasma, photoionization, glow discharge, or other techniques, which can be selected based on the type of sample provided and the data required. For example, tandem liquid chromatography mass spectrometry can be used for prepared hair samples.


The mass spectrometry dataset can be compared, using existing spectrometry data analysis tools, to existing or created libraries of known spectra of known proteins (e.g. RefSeq, UniProt, Protein Mutation Database, HPMD, MSIPI, MS-CanProVar, dbSNP, Ensembl, COSMIC, or a custom database containing all of the single amino acid polymorphisms above some threshold allelic frequency) to determine the protein content of the biological sample, a.k.a. the proteomically detected proteins.


The data can be formatted in a number of different well-known proteomic datafile formats: as examples, mzML, Mascot Generic Format (MGF), or any proprietary format.


The identified variations in the detected proteins provide markers for genetic information (e.g., identifying genetic information) which can be verified against the genomic variations detectable in the original biological sample. This, the validated genetic protein variation, can be produced by comparing the provided mass spectrometry dataset of the original biological sample with the proteomically detectable genetic protein variation.


Providing a proteomically detectable genomic variation in the set of represented genes proteomically detectable in the biological sample of the individual can be performed by providing exome sequence data of the individual and comparing the exome sequence data of the individual with sequences from the represented genes proteomically detectable in the biological sample of the individual to determine the proteomically detectable genomic variation in the biological sample of the individual. Providing the exome sequence data of the individual can, for example, be performed by the methods explained herein, or by other known methods. The exome data can be procured from the original biological sample, or from some other biological sample, even one of a different type (blood, hair, saliva, etc.) than the original. Additionally, the exome data can be procured from any genetically relevant source, such as a close family member of the individual. Additionally, the exome data can be procured from a database of already determined genetic data.


Furthermore, providing a proteomically detectable genetic protein variation corresponding to the proteomically detectable genomic variation in the biological sample of the individual, can be performed through single nucleotide polymorphism (SNP) annotation on the proteomically detectable genomic variation in the biological sample of the individual to produce a corresponding mutant/reference protein sequence; and providing the proteomically detectable genetic protein variation from the annotated proteomically detectable genomic variation in the biological sample of the individual.


“SNP annotation” (or “annotation”) as used herein refers to the process to predict the effect or function of an individual SNP by use of a tool (e.g., SNPeff, VEP, ANNOVAR, FATHMM, PhD-SNP, PolyPhen-2, SuSPect, F-SNP, AnnTools, SeattleSeq, SNPit, SCAN, Snap, SNPs&GO, LS-SNP, Snat, TREAT, TRAMS, Maviant, MutationTaster, SNPdat, Snpranker, NGS—SNP, SVA, VARIANT, SIFT, PhD-SNP and FAST-SNP). In annotation, biological information is extracted, collected, and displayed in a way that makes querying the data easier.


A genetic protein variation identity panel can be created by collecting the validated genetic protein variant proteomically detectable in the biological sample of the individual. This provides a genetic protein variation identity panel of the individual.


Exemplary represented genes and/or exome sequences of Homo Sapiens having a corresponding detected peptide sequence that can be used in the method and/or comprised in a database according to the fourth aspect are indicated in Examples 43 to 45 listing exemplary set of genes validated in hair samples (Example 43, Table 8) bone samples (Example 44, Table 9) and skin samples (Example 45, Table 10) of a human being.


Exemplary marker genetic protein variations validated in Homo Sapiens that can be used in the method and/or comprised in a database according to the fourth aspect if the instant disclosure, can comprise any one of the marker genetic protein variations indicated in Examples 46 and Example 47 listing exemplary set of GVPs validated in hair (Example 46, Table 11) and skin samples (Example 47, Table 12) of a human being.


In some embodiments, detecting a genetic variation can be performed with a pooled marker genetic variation database system obtainable with a method and system to improve a pooled marker genetic protein variation database system according to the fifth aspect of the present disclosure. In the method and system, the pooled marker genetic protein variation database system comprises marker genetic protein variations common to a plurality of individuals.


The method comprises: providing a number of proteomic datasets of individuals of the plurality of individuals, the number statistically significant for the plurality of individuals, identifying a protein common to the provided number of proteomic datasets; and selecting from the identified protein common to the provided proteomic datasets, a protein detectable in a biological sample of an individual of the plurality of individuals.


The method further comprises providing a number of exome datasets of the individuals of the plurality of individuals, the number statistically significant for the plurality of individuals; and identifying a genetic variation in the provided number of exome datasets.


The method also comprises selecting from the identified genetic variation, a genetic variation detectable in the biological sample; and comparing the selected proteins detectable in the biological sample with the selected genetic variations detectable in the biological sample, to provide a marker genetic protein variation common to a plurality of individuals of a biological organism type and validated to be detectable in the biological sample.


The system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases for simultaneous combined or sequential use in the method to improve a pooled marker genetic protein variation database system for a biological organism herein described.


The process for creating a marker genetic protein variation database system can be repeated for a plurality of individuals, preferably ones sharing the same genetic variant or variants to be cataloged in the database, to provide a database comprising validated genetic protein variations proteomically detectable in the biological sample of the plurality of individuals of that biological organism type.


This database can be formed by collecting the represented genes common to the individuals into a proteomically detectable gene pool, providing validated genetic protein variations proteomically detectable in the biological sample of the plurality of individuals of the biological organism from the collected common represented, and collecting the validated genetic protein variants proteomically detectable in the biological sample of the individuals, in a genetic protein variation panel comprising a genetic protein variation panel common to the individuals.


The proteomically detectable gene pool can contain data corresponding to proteins that are common to some or all the validated genetic protein variants proteomically detectable in the biological sample of a given individual. This can be set against a threshold limit, for example only proteins that are common in at least (or over) 50% of all individuals in the pool.


Providing validated genetic protein variations proteomically detectable in the biological sample of the plurality of individuals can be performed to only include genomic variation with a frequency greater than some threshold limit, for example 1%, in the plurality of the individuals into a proteomically detectable gene pool.


One aspect of a method to improve a marker genetic protein variation database system comprising marker genetic protein variations common to a plurality of individuals includes: providing a number of proteomic datasets of individuals of the plurality of individuals, the number statistically significant for the plurality of individuals, identifying one or more proteins common to the provided number of proteomic datasets; selecting from the identified proteins common to the provided proteomic datasets, a protein detectable in a biological sample (e.g., hair) of an individual of the plurality of individuals; providing a number of exome datasets of the individuals of the plurality of individuals, the number statistically significant for the plurality of individuals; identifying a genetic variation in the provided number of exome datasets; selecting from the identified genetic variation, a genetic variation detectable in the biological sample; and comparing the selected proteins detectable in the biological sample with the selected genetic variations detectable in the biological sample, to provide a marker genetic protein variation common to a plurality of individuals of a biological organism type and detectable in the biological sample.


The database system is realizable in a computer system, either as a single computer (processor, memory, etc.) or as a network of computers, including, as examples, cloud, intranet, internet, or parallel processing systems. The database system can be centralized and accessible by web-based searches, or stand-alone.


Once created, the database can be searched to create identity metrics for a questioned biological sample of the same type (hair, blood, saliva, etc.) by GVP matching.


The term “exome” as used herein refers to the part of the genome formed by exons, the sequences which when transcribed remain within the mature RNA after introns are removed by RNA splicing. It consists of all DNA that is transcribed into mature RNA in cells of any type as distinct from the transcriptome, which is the RNA that has been transcribed only in a specific cell population. For example, humans have about 180,000 exons, constituting about 1% of the human genome, or approximately 30 million base pairs.


Exome sequencing, also known as whole exome sequencing (WES or WXS), typically consists of two steps: the first step is to select only the subset of DNA consisting of exons. The second step is to sequence the exonic DNA using any high-throughput DNA sequencing technology. In the first step, target-enrichment methods allow the selective capture of genomic regions of interest from a DNA sample prior to sequencing. Both array-based and in-solution capture techniques can be used. In array-based capture, microarrays containing single-stranded oligonucleotides with sequences from a genome (e.g. human exome) tile the region of interest fixed to the surface. Genomic DNA is sheared to form double-stranded fragments. The fragments undergo end-repair to produce blunt ends and adaptors with universal priming sequences are added. These fragments are hybridized to oligos on the microarray. Unhybridized fragments are washed away and the desired fragments are eluted. The fragments can then be amplified using PCR. Next-generation sequencing techniques can also be used with array-based capture. For example, the Sequence Capture Human Exome 2.1M Array can be used to capture -180,000 coding exons. This method is both time-saving and cost-effective compared to PCR based methods. The Agilent Capture Array and the comparative genomic hybridization array are other methods that can be used for hybrid capture of target sequences. To capture genomic regions of interest using in-solution capture, a pool of custom oligonucleotides (probes) is synthesized and hybridized in solution to a fragmented genomic DNA sample. The probes (labeled with beads) selectively hybridize to the genomic regions of interest after which the beads (now including the DNA fragments of interest) can be pulled down and washed to clear excess material. The beads are then removed and the genomic fragments can be sequenced allowing for selective DNA sequencing of genomic regions (e.g., exons). In general, in the first step, any of a number of available exome enrichment platforms (e.g., Roche/NimbleGen's SeqCap EZ Human Exome Library, Illumina's Nextera Rapid Capture Exome, Agilent's SureSelect XT Human All Exon and Agilent's SureSelect QXT) can be used to allow the selective capture of genomic regions of interest from a DNA sample. In the second step, there are several sequencing platforms available in addition to classical Sanger sequencing. Other platforms include the Roche 454 sequencer, the Illumina Genome Analyzer II and the Life Technologies SOLiD & Ion Torrent, which can be used for exome sequencing. Any cellular material that contains genomic DNA can be used for exome sequencing, such as human blood samples, buccal sample and others identifiable by skilled persons.


Exome sequencing can also be performed by RNA exome sequencing (e.g., Illumina RNA Exome Capture Sequencing) according to approaches and techniques identifiable by a skilled person.


The term “exome-driven” as used herein refers to an approach of GVP discovery that begins with sequencing the exome of an individual, allowing identification of relevant SNPs, followed by proteomic validation of GVPs (see FIG. 7). Thus, the “exome-driven” approach features (1) obtaining exome sequence for each donor, (2) establishing a workflow to identify specific SNPs of interest, (3) targeted proteomic analysis allowing simplified identification of GVPs in raw MS data, and (4) allows a logic-driven GVP selection, identification, and validation process. In contrast, a “proteome-driven” discovery approach begins with proteomic analysis, followed by candidate peptide identification, and DNA validation of identified GVPs (see FIG. 7). Thus, the proteome-driven approach has limitations such as being a ‘needle in a haystack’ approach that is not compatible with targeted proteomic analysis and relies on manual MS interpretation to identify potential GVPs, wherein potential GVPs must then be validated by separate individual genotyping experiments.


In a typical “proteome-driven” GVP discovery approach that is used following existing methods and systems, a peptide mixture is obtained from a sample and is analyzed by LC-MS/MS. The resulting dataset is then analyzed with reference to a protein variant database using analysis software tools such as MASCOT, PEAKS, and GPM. Candidate GVPs in the observed proteins identified in the sample are screened using metrics such as match score, frequency, and qualitative assessment. The screened GVPs are then validated by confirming the GVPs comprise missense mutations genetically encoded by SNPs by genomic sequencing. The validated GVPs then are incorporated into a GVP database. FIG. 8 shows an exemplary schematic summarizing a typical proteome-driven GVP discovery approach (e.g. for hair samples).


The term “validated GVP” as used herein refers to a GVP that comprises a variation (e.g. a SAP) that has been confirmed to correspond to a variation (e.g., a nsSNP) in the exome of the same individual.


A schematic summarizing the “exome-driven” GVP discovery approach is shown in FIGS. 9 and 10. As shown in FIG. 9, for a given tissue type (e.g. hair), the proteins detected by LC-MS/MS for a given individual are referred to herein as “observed proteins” that are encoded by “represented genes”. Thus, the represented genes form the ‘Down-selected Target Genes’ of the ‘Observed Gene Pool’.


In some embodiments, the exome-driven GVP discovery approach described herein can be used to assemble a panel of validated GVPs for a population of individuals, referred to herein as a “Common GVP Panel” or “Pooled GVP Panel”. In particular, in the “Common GVP panel”, GVPs are down selected for common nsSNPs, and a consensus panel is assembled from a large cohort. As described herein, the term “common nsSNPs” refers to nsSNPs having a frequency >1% and having a worldwide distribution.


In some embodiments, the exome-driven GVP discovery approach described herein can be used to assemble a panel of validated GVPs for an individual, referred to herein as an “Individual GVP Panel”. In particular, for an ‘Individual GVP Panel’, GVPs can be down-selected based on low-frequency or ‘rare’ or ‘private’ nsSNPs and the GVP panel is unique to that individual (see FIG. 17). The term “down-select” as used herein refers to narrowing the field of choices based on specific conditions or characteristics. The term “rare SNPs” as used herein refers to nsSNPs having a frequency <0.05% in a given population.


An exemplary “exome-driven” GVP discovery method, showing integration of exomic and proteomic data for building a “Pooled GVP Panel” or an “Individual GVP Panel” is described in Example 14.


In some embodiments, exome-driven discovery of GVPs from a diverse cohort allows discovery of markers that are informative of biogeographic background.


The exome-driven GVP discovery methods and systems described herein can be used for discovery of validated GVPs for any tissue type. For example, an exemplary exome-driven method of building a panel of validated GVPs for hair samples is described in Example 15 and an exemplary panel of validated GVPs for bone is described in Example 21.


The exome-driven GVP discovery methods and systems described herein can be used in several embodiments in combination with samples from any tissue type prepared using any method.


In some embodiments, application of the product rule can be used to estimate the probability of a combination of individual nsSNPs (otherwise referred to herein as a “nsSNP profile”) in a population. The term “product rule” as used herein refers to the multiplication of frequencies of individual nsSNPs in a profile in a population to calculate the overall frequency of the combination of nsSNPs in a nsSNP profile in the population.


As understood by those skilled in the art, linkage disequilibrium (LD) can affect calculation of the overall frequency of the combination of nsSNPs in a nsSNP profile in the population, and thus can affect theoretical genotype match probabilities. The term “linkage disequilibrium” refers to non-random association of alleles at different loci in a given population. In general, DNA sequences that are close together on a chromosome have a tendency to be inherited together during the meiosis phase of sexual reproduction. Two loci that are physically near to each other are unlikely to be separated onto different chromatids during chromosomal crossover, and are therefore said to be more linked than markers that are far apart. Loci are said to be in linkage disequilibrium when the frequency of association of their different alleles is higher or lower than what would be expected if the loci were independent and associated randomly. Because nearby loci are often inherited together, in some embodiments the product rule doesn't directly apply. For example, many loci for exemplary validated GVPs shown in FIG. 13 are keratin genes, which are clustered on chromosomes 12 and 17. Thus, the loci encoding these GVPs may be linked though they are in different genes, and linked loci can be up to, for example, 220 kb apart. Therefore, in some embodiments, LD can be taken into account for calculation of the probability of an overall non-synonymous SNP profile in the population. LD can be factored into the calculation by computing LD between pairs of GVP loci located on the same chromosome, for example using data from the 1000 Genomes Project dataset. Next, clusters of linked loci can be grouped, by computation of joint genotype probabilities given LD for loci within each cluster and by multiplying cluster probabilities to get overall genotype likelihood.


In some embodiments, strategies for identification of candidate GVPs comprise studying a larger and more diverse cohort, increased proteomic detection through instrumentation, and bioinformatic data mining of previously collected datasets, among others identifiable by skilled persons upon reading of the present disclosure. In exemplary embodiments of the methods and systems described herein, sample sets comprise protein and DNA sample sets from cohorts comprising n=200-250 European Americans, n=30-50 African Americans, n=30-50 Hispanic, n=100 East Asian, and n=60 parent/offspring.


In some embodiments, the panel of validated GVPs is an Individual GVP panel.


In some embodiments, the panel of validated GVPs is a Pooled GVP panel.


A schematic of an exemplary method of how to apply an Individual or Pooled GVP panel to operational samples is shown in FIG. 11 and described in Example 16.


Exemplary represented validated genes and/or exome sequences of Homo Sapiens having a corresponding detected peptide sequence that can be used in the method and/or comprised in a database according to the fifth aspect of the instant disclosure are indicated in Examples 43 to 45 listing exemplary set of genes validated in hair samples (Example 43, Table 8) bone samples (Example 44, Table 8) and skin samples (Example 45, Table 10) of a human being.


Exemplary validated marker genetic protein variations that can be used in the method and/or comprised in a database according to the fifth aspect of the instant disclosure, can comprise any one of the marker genetic protein variations indicated in Examples 46 and Example 47 listing exemplary set of GVPs validated in hair (Example 46, Table 11) and skin (Example 47, Table 12) samples. The validated GVPs of Table 11, and Table 12 can preferably be used in connection with method performed on biological samples from a human being.


Further details concerning the methods and systems of the present disclosure will become more apparent hereinafter from the following detailed disclosure of examples by way of illustration only with reference to an experimental section.


In some embodiments detecting a genetic variation can be performed with a method and a system to detect a marker genetic variation for a biological organism validated to be detectable in a biological sample of an individual of the biological system, according to the sixth aspect of the present disclosure.


The method comprises preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis; and fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample and a solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample.


The method further comprises detecting a genetic protein variation in the solubilized proteins from the sample by performing the proteomic analysis of the solubilized protein fraction; and detecting a genomic variation of the nuclear and/or mitochondrial genome by performing a genetic analysis of the solubilized DNA fraction.


The method also comprises comparing the detected genetic protein variation and/or the detected genomic variation with a marker genetic protein variation and/or of a marker genomic variation respectively from the marker genetic variation database system herein described.


The system comprises exome sequences databases and/or reagents to detect exome sequences in an individual of the biological organism, in combination with reagents to perform proteomic analysis of the biological sample for simultaneous combined or sequential use in the method to detect a marker genetic variation for a biological organism validated to be detectable in a biological sample of an individual of the biological system herein described.


In embodiments of the method according to the sixth aspect, any method of preparing the biological sample identifiable by persons skilled in the art upon reading the present disclosure can be used in the method to detect a marker genetic variation in a biological sample of a biological organism.


. In embodiments of the method according to the sixth aspect, any method to perform fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample and a solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample can be used in the method to detect a marker genetic variation in a biological sample of a biological organism.


In some embodiments, the fractionating can be performed for example by several methods of DNA purification from a solution containing protein and DNA. In general, successful nucleic acid purification requires effective disruption of cells or tissue or organ material, denaturation of nucleoprotein complexes, inactivation of nucleases such as DNase, and absence of contamination.


For example, commonly used procedures for DNA purification from detergents, proteins, salts and reagents used in sample preparation comprise alcohol precipitation, phenol-chloroform extraction, and mini-column purification, among other techniques known in the art. Alcohol precipitation can be performed using e.g., using ice-cold ethanol or isopropanol. Since DNA is insoluble in these alcohols, it will aggregate together, giving a pellet upon centrifugation. Precipitation of DNA can be improved by increasing of ionic strength, for example by adding sodium acetate. Phenol—chloroform extraction can be performed in which phenol denatures proteins in the sample. After centrifugation of the sample, denatured proteins remain in the organic phase while aqueous phase containing nucleic acid is mixed with the chloroform that removes phenol residues from solution. Mini-column purification can be performed, in which nucleic acids bind (adsorb) to a solid phase (e.g., silica or other) depending on the pH and the salt concentration of the buffer. For example, an exemplary method of performing fractionation of a biological sample into a DNA fraction and a protein fraction using mini-column purification is described in Example 7.


In embodiments of the method and system of combined mtDNA and proteomic analysis from a single sample, any method of sample preparation identifiable by those skilled in the art that can provide an extract of purified protein suitable for proteomic analysis and a mtDNA extract and/or nuclear DNA extract suitable for mtDNA and/or nuclear DNA analysis from a single biological sample can be used, and is not limited to exemplary methods described herein.


The exemplary procedures described herein reveal that protein identification markers (GVPs) can be detected from one-inch hair samples using LC-MS/MS of peptides. In exemplary embodiments described herein, protein extraction by ultrasonication and harsh detergents can fully dissolve the hair matrix, maximizing the ability of enzyme proteolysis and subsequently peptide concentration in samples. Additionally, the exemplary protein extraction procedure described herein is compatible with mtDNA extraction, copy number determination, and hyper-variable region sequencing (Example 7). Thus, in some embodiments, GVP discovery and mtDNA sequencing in combination provide a substantial measure of human identity because of the vast variation in allelic frequencies of SNPs. These exemplary embodiments illustrate the potential proteomic analysis of hair evidence has for becoming a widely implemented forensic tool.


As understood by skilled persons, the term “genome” refers to the total heritable genetic material of an organism, comprising DNA (or RNA in RNA viruses), wherein a genome comprises a plurality of genes.


In particular, in eukaryotes, and in particular in animals, the genome comprises both a “nuclear genome” and a “mitochondrial genome”. In plants, the genome also comprises a “chloroplast genome”. Thus, in embodiments herein described, the term “genome” can be applied specifically to mean the genes that are stored on a complete set of nuclear DNA (also referred to herein as the “nuclear genome”, typically arranged on chromosomes in a eukaryotic cell's nucleus) and can also be applied to specifically refer to the genes that are within organelles that contain their own DNA, as with the “mitochondrial genome” or the “chloroplast genome”, as identifiable by persons skilled in the art upon reading of the present disclosure.


The mitochondrial genome is the entirety of hereditary information contained in mitochondria. Mitochondrial DNA (mtDNA) is not transmitted through nuclear DNA (nDNA).


While DNA is degraded as a function of biological processes, mitochondrial DNA has a higher template number than nuclear DNA and is more likely to survive apoptotic and subsequent environmental processes[11]. Accordingly, for some tissue sample types, recovery of both protein and mtDNA from tissue samples would allow incorporation of both proteomic and mtDNA haplotype analysis into a single measure of discrimination.


The terms “haplotype” or “haploid genotype” as used herein refers to a group of genes in an organism that are inherited together from a single parent and the term “haplogroup” refers to a group of similar haplotypes that share a common ancestor with a single-nucleotide polymorphism mutation. Accordingly, for example, a human mitochondrial DNA haplogroup is a haplogroup defined by differences in human mitochondrial DNA. The letter names of the haplogroups (not just mitochondrial DNA haplogroups) run from A to Z. The human mitochondrial genome is the entirety of hereditary information contained in human mitochondria. Mitochondrial DNA (mtDNA) is not transmitted through nuclear DNA (nDNA). In humans, as in most multicellular organisms, mitochondrial DNA is inherited only from the mother's ovum. In humans, mitochondrial DNA (mtDNA) forms closed circular molecules that contain 16,569 DNA base pairs, with each such molecule normally containing a full set of the mitochondrial genes. In humans, the 16,569 base pairs of mitochondrial DNA encode for 37 genes. Human mitochondrial DNA was the first significant part of the human genome to be sequenced.


For example, the current best practice to gain forensically informative genetic information from hair shafts is to obtain the mitochondrial DNA haplotype and determine the probability of occurrence in reference sample populations[12]. Incorporation of both proteomic and mtDNA haplotype analysis into a single measure of discrimination, would maximize the probative value of a biological sample such as hair shafts.


As understood by skilled persons, a genome (and in particular a nuclear genome) can comprise polynucleotides comprising repetitive DNA elements such as interspersed repeats, retrotransposons, long terminal repeats, non-long-terminal repeats, long-interspersed elements, short interspersed elements, DNA transposons, and tandem repeats, among others identifiable by skilled persons.


The term “interspersed repeat” refers to polynucleotide elements such as transposable elements (TEs), and in some embodiments can also refer to some protein coding gene families and pseudogenes. Transposable elements are able to integrate into the genome at another site within the cell. TEs can be classified into two categories, Class 1 (retrotransposons) and Class 2 (DNA transposons), as would be understood by skilled persons. Retrotransposons can be transcribed into RNA, which are then duplicated at another site into the genome. Retrotransposons can be divided into Long terminal repeats (LTRs) and Non-Long Terminal Repeats (Non-LTR). Long interspersed elements (LINEs) typically encode two Open Reading Frames (ORFs) to generate transcriptase and endonuclease, which are essential in retrotransposition. Short interspersed elements (SINEs) are typically less than 500 base pairs in length and require the LINEs machinery to function as nonautonomous retrotransposons. For example, the Alu element is the most common SINE found in primates, it has a length of about 350 base pairs and takes about 11% of the human genome with around 1,500,000 copies.


In particular, the term “tandem repeat” refers to a repeating pattern of one or more nucleotides in DNA wherein the repetitions are directly adjacent to each other. In particular, the term “minisatellite” refers to a tandem repeat having typically between 14 and 60 repeated nucleotides, whereas tandem repeats having fewer repeated nucleotides are typically referred to as “microsatellites” or “short tandem repeats” or “STR”.


In particular, an STR is type of microsatellite consisting of a unit of 2-13 or more base pairs repeated hundreds of times in a row on the DNA strand. A microsatellite is a tract of repetitive DNA in which certain DNA motifs (ranging in length from 2-13 base pairs) are repeated, typically 5-50 times. Microsatellites occur at thousands of locations within an organism's genome; additionally, they have a higher mutation rate than other areas of DNA leading to high genetic diversity. Microsatellites are often grouped according to the length of the unit of repeated base pairs. For example, the sequence TATATATATA (SEQ ID NO: 134) is a dinucleotide microsatellite, and GTCGTCGTCGTCGTC (SEQ ID NO: 135) is a trinucleotide microsatellite (with A being Adenine, G Guanine, C Cytosine, and T Thymine). Repeat units of four and five nucleotides are referred to as tetra- and pentanucleotide motifs, respectively. Most eukaryotes have microsatellites, with the notable exception of some yeast species, and these microsatellites are distributed throughout the genome. The human genome for example contains 50,000-100,000 dinucleotide microsatellites, and lesser numbers of tri-, tetra- and pentanucleotide microsatellites. Many are located in non-coding parts of the human genome and therefore do not produce proteins, but they can also be located in regulatory regions and coding regions. Microsatellites and minisatellites together are classified as VNTR (variable number of tandem repeats) DNA.


STRs are often used in forensics because although the repeating sequence of base pairs of a specific microsatellite does not change from person to person, the number of times the sequence repeats does change. This allows the number of repeats of a sequence to identify a person through his/her DNA if the number of sequence repeats matches the initial DNA basis used for comparison. STRs can also be used to eliminate a person from suspicion or reduce the suspicion of a person if he/she does not have the same number of sequence repeats as the comparate DNA. STRs are widely used for DNA profiling in kinship analysis (such as paternity testing) and in forensic identification. They are also used in genetic linkage analysis/marker assisted selection to locate a gene or a mutation responsible for a given trait or disease. Microsatellites are also used in population genetics to measure levels of relatedness between subspecies, groups and individuals.


In particular, STR analysis is a tool in forensic analysis that evaluates specific STR regions found on nuclear DNA. STR analysis measures the exact number of repeating units. This method differs from restriction fragment length polymorphism analysis (RFLP) since STR analysis does not cut the DNA with restriction enzymes. Instead, probes are attached to desired regions on the DNA, and a polymerase chain reaction (PCR) is employed to discover the lengths of the short tandem repeats. This method uses highly polymorphic regions that have short repeated sequences of DNA (the most common is 4 bases repeated, but there are other lengths in use, including 3 and 5 bases). Because unrelated individuals typically have different numbers of repeat units, STRs can be used to discriminate between unrelated individuals. These STR loci (locations on a chromosome) are targeted with sequence-specific primers and amplified using PCR. The DNA amplicons that result are then separated and detected using electrophoresis methods, such as capillary electrophoresis and gel electrophoresis.


Several STR-based DNA-profiling systems are in use, identifiable by those skilled in the art. For example, in North America, systems that amplify the “CODIS 13 core loci” are almost universal, whereas in the United Kingdom the “DNA-17” 17 loci system is in use. Whichever system is used, many of the STR regions used are the same. These DNA-profiling systems typically use multiplex PCR, whereby many STR regions are tested at the same time. For example, the 13 loci that are currently used for discrimination in CODIS are independently assorted (having a certain number of repeats at one locus does not change the likelihood of having any number of repeats at any other locus), and therefore the product rule for probabilities can be applied.


Accordingly, in embodiments of the method according to the sixth aspect described herein, any method of genetic analysis identifiable by skilled persons can be used for detecting a genomic variation of the nuclear and/or mitochondrial genome.


In embodiments of the method according to the sixth aspect described herein, any method of combining the detected genetic protein variations and the detected genomic variation can be used to provide the marker genetic variation database system of the biological sample, the detected genetic protein variations and the detected genomic variation to provide the marker genetic variation database system of the biological sample.


In embodiments of the method according to the sixth aspect described herein, comparing the detected genetic protein variation and/or the detected genomic variation with a marker genetic protein variation and/or of a marker genomic variation respectively from the marker genetic variation database system can be performed with any methods identifiable by a skilled person


In embodiments of the method and system of combined mtDNA and proteomic analysis from a single sample, any method of sample preparation identifiable by those skilled in the art that can provide an extract of purified protein suitable for proteomic analysis and a mtDNA extract suitable for mtDNA analysis from a single tissue sample can be used, and is not limited to exemplary methods described herein.


The system comprises equipment, reagents, and samples required to perform the method of the combined mtDNA and proteomic analysis from a single sample.


In some embodiments of a genetic variation analysis, detecting a genetic variation in a genetic variation analysis can be performed using a marker genetic variation database according to a seventh aspect herein described. The related method to provide the marker genetic variation database system comprising marker genetic variation validated to be detectable in a biological sample, comprises preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis.


The method further comprises fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample and a solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample.


The method also comprises detecting a genetic protein variation in the solubilized proteins from the sample by performing the proteomic analysis of the solubilized protein fraction and detecting a genomic variation of the nuclear and/or mitochondrial genome by performing a genetic analysis of the solubilized DNA fraction.


The method additionally comprises combining the detected genetic protein variations and the detected genomic variation to provide the marker genetic variation database system comprising marker genetic variation validated to be detectable in a biological sample.


The system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases for simultaneous combined or sequential use in the method to provide the marker genetic variation database system comprising marker genetic variation validated to be detectable in a biological sample herein described.


In some embodiments wherein preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis, is performed by the method according to the first aspect.


In some embodiments detecting a genetic protein variation is performed by the method according to the sixth aspect.


Methods and systems and related marker genetic protein variations and databases herein described, can be used in several embodiments for proteomic information detection using liquid chromatography/mass spectrometry methods for forensic analysis of tissue samples to provide identity metrics of individuals. In several embodiments, the methods and systems described herein allow improved proteomic information recovery when genomic DNA is degraded or not available, and/or when there are multiple contributors to the sample.


In some embodiments of the instant disclosure a genetic analysis of a sample of a biological organism can be performed with methods and systems according to the eighth aspect of the disclosure. The method comprises


preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis;


fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample;


digesting the solubilized protein fraction from the sample to obtain digested peptides from the sample;


fractionating the digested peptides to obtain fractionated digested peptides from the digested solubilized proteins from the biological sample.


detecting a marker genetic variation of the fractionated digested peptides from the sample; in which


preparing the sample is performed according to any one of the methods according to the first aspect of the disclosure, comprising any one of the related sets of embodiments ; and/or


detecting a genetic variation is performed by at least one of


a first detecting method directed to detect a genetic protein variation by performing any one of the methods according to the third aspect, comprising any one of the related sets and subsets of claims; and


a second detecting method directed to detect a genetic variation by performing any one of the methods according to the sixth aspect of the disclosure comprising any one of the related sets of embodiments.


In the method of the eighth aspect the genetic analysis is directed to detect one or more genetic variations in the sample, and preferably comprises detection of at least one genetically variant protein, which more preferably has been validated in the sample where detection is performed. Therefore in preferred embodiments of the method of the eighth aspect of the disclosure the genetic analysis is a genetic protein variation analysis directed to detect in the sample one or more genetic variations validated in the analyzed sample.


In some embodiments of the method according to the eight aspect, the preparing can be performed with existing methods of sample preparation for proteomics. Typically, these methods comprise performing cell and tissue disruption and performing protein solubilization according to approaches identifiable by a skilled person upon reading of the present disclosure. Typically these methods can also comprise performing removal of contaminants and/or performing protein enrichment following performing protein solubilization, according to approaches identifiable by a skilled person upon reading of the present disclosure.


In preferred embodiments of the method according to the eight aspect however, the preparing is performed by any one of the embodiments the method according to the first aspect of the present disclosure as will be understood by a skilled person.


In more preferred embodiments of the method of the eight aspect wherein the preparing is performed according to the method of the first aspect, the applying is performed by sonication, with a related processor preferably set at 5 to 50 kHz and more preferably at 37 kHz with a power setting preferably set at 50 to 100%; most preferably at 100%. In more preferred embodiments the applying is performed with an ultrasonic mode sweep.


In more preferred embodiments of the method of the eight aspect wherein the preparing is performed according to the method of the first aspect, the applying can be performed with an incubation time from 20 to 90 minutes; most preferably 60 minutes


In more preferred embodiments of the method of the eight aspect wherein the preparing is performed according to the method of the first aspect, the applying can be performed with temperature settings from 30 to 90° C.; most preferably 70° C.


In any one of the embodiments of the method of the present disclosure according to the eighth aspect, the digesting can be performed with any methods identifiable by a skilled person upon reading of the present disclosure.


In preferred embodiments of method of the present disclosure according to the eighth aspect, the digesting is performed enzymatically with one or more proteolytic enzymes identifiable by a skilled person.


In more preferred embodiments of the method according to the eighth aspect, the digesting comprises digesting the solubilized proteins from the sample with a site specific proteolytic enzyme to obtain digested solubilized proteins from the sample.


In those more preferred embodiments the digesting can be performed in a sample buffer comprising an enzyme capable to perform site specific protease digestion such as trypsin, chymotrypsin, Lys-C, Arg-C, Asp-N, and Glu-C, non-specific; pepsin, and proteinase K.


In particular in those more preferred embodiments of the method according to the eighth aspect, the enzyme can be comprised in the sample buffer at concentrations for digest ranging from 0.0001 to 1 μg/μL; more preferably 0.01 to 0.001 μg/μL; most preferably 0.005 μg/μL.


In even more preferred embodiments of the method according to the eighth aspect, the proteolytic enzyme is trypsin.


In preferred embodiments of the method according to the eighth aspect of the present disclosure, the digesting is preceded by fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample. In those embodiments, the solubilized proteins are fractionated in a solubilized protein fraction and digesting the solubilized proteins is performed by digesting the solubilized protein fraction. In those embodiments fractionating the solubilized proteins can be performed by any one of the methods identifiable by a skilled person upon reading of the present disclosure typically comprises removing buffers, salts, and detergent from the processed sample. In more preferred embodiments fractionating the solubilized proteins can further comprise removing abundant proteins from the processed sample, protein enrichment processes and/or removing contaminants which can be performed with any one of the methods identifiable by a skilled person upon reading of the present disclosure.


In any one of the embodiments of the method according to the eighth aspect of the present disclosure, the genetic analysis also comprises detecting a marker genetic variation of the digested peptides.


In preferred embodiments of the method according to the eighth aspect of the present disclosure, the detecting is performed by mass spectrometry according to methods identifiable by a skilled person upon reading of the present disclosure. In those embodiments, the concentration of proteolytic enzyme in the sample buffer used during the digesting is set taking into account that increased concentrations can cause suppression of sample detection, decrease LC column capacity; and decrease ability to observe sample peptides by overcrowding mass a spectrometry detector as will be understood by a skilled person.


In those preferred embodiments of the method of the eighth aspect, wherein the proteomic analysis is performed by Mass Spectrometry, the digesting can be performed in a buffer comprising mass spectrometry compatible surfactant, such as for example, Invitrosol, ProteaseMax, Rapigest SF, and PPS Silent Surfactant), in concentration (percent w/v) ranges broadly from 0.0001 to 1.0%; more preferably 0.001 to 0.2%; and most preferably 0.01%. Increasing concentrations can cause issues with electrospray efficiency during MS data acquisition. In preferred embodiments, the surfactant comprise ProteaseMax.


In preferred embodiments of the method according to the eighth aspect, the detecting is preceded by fractionating the digested solubilized proteins to obtain fractionated digested peptides from the digested solubilized proteins from the biological sample. In those embodiments, the digested peptides are fractionated digested peptides and detecting a marker genetic variation of the digested peptides is performed by detecting a marker genetic variation of the fractionated digested peptides.


In those preferred embodiments of the method according to the eighth aspect, fractionating the digested solubilized proteins can be performed by any suitable method of fractionating proteins identifiable by a skilled person upon reading of the present disclosure. Preferably, fractionating the digested solubilized proteins can be performed by any chromatographic techniques identifiable by a skilled person upon reading of the present disclosure.


In more preferred embodiments of the method according to the eighth aspect, the fractionating is performed by liquid chromatography and the detecting is performed by mass spectrometry in an approach that combines the physical separation capabilities of liquid chromatography with the mass analysis capabilities of any mass spectrometry as will be understood by a skilled person upon reading of the present disclosure.


In even more preferred embodiments of the method according to the eighth of the present disclosure, the detecting is performed according to any one of the methods according to the third aspect or the sixth aspect of the instant disclosure and/or using any of the related databases.


In particular in some of the even more preferred embodiments of the method according to the eighth aspect, the detecting is performed according to the third aspect of the instant disclosure by


providing a marker mass spectrum of a marker peptide comprising a marker genetic protein variation corresponding to the genetic protein variation;


performing mass spectrometry of a fractionated digested peptide of the biological sample to obtain a mass spectrum of each of the fractionated digested peptide; and


comparing the mass spectrum of the fractionated digested peptide with a marker mass spectrum of a marker peptide comprising the marker genetic protein variation to detect the genetic protein variation in the biological sample.


In some embodiments of the even more preferred embodiments of the method according to the eighth aspect in which the detecting is performed by the method according to the third aspect of the present disclosure, the marker genetic protein variation is obtained by any one of the methods to provide a marker genetic protein variation for a biological organism according to the second aspect of the instant disclosure and/or is a marker genetic protein variation obtainable and/or obtained thereby.


In some embodiments of the even more preferred embodiments of the method according to the eighth aspect in which the detecting is performed by the method according to the third aspect of the present disclosure, the marker genetic protein variation comprises a marker genetic protein variation from the marker genetic protein variation database system according to the fourth aspect of the instant disclosure.


In some embodiments of the even more preferred embodiments of the method according to the eighth aspect in which the detecting is performed by the method according to the third aspect of the present disclosure, the marker genetic protein variation comprises a marker genetic protein variation from the marker genetic protein variation database system according to the fifth aspect of the instant disclosure.


In some embodiments of the even more preferred embodiments of the method according to the eighth aspect in which the detecting is performed by the method according to the third aspect of the present disclosure, the marker peptide comprises one or more of the marker peptides comprising a validate genetic protein variations indicated in Examples 46 and Example 47 indicating exemplary set of GVPs and related mutated peptides validated in hair samples (Example 46, Table 11) and skin samples (Example 47, Table 12) and in particular in hair and skin samples of human beings.


In particular exemplary marker peptides that can be preferably used or comprise in the method and system according to the eighth aspect, comprise any combination of the peptides having sequence SEQ ID NO: 150 to SEQ ID NO: 748 (Example 46, Table 11) for detection in hair samples, in particular for hair samples of human beings, and any combination of the peptides having sequence SEQ ID NO: 749 to SEQ ID NO: 829 (Example 47, Table 12) for detection in skin samples, in particular for skin samples of human beings.


In some of the even more preferred embodiments of the method according to the eighth aspect, the detecting is performed according to the sixth aspect of the instant disclosure by


preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis;


fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample and a solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample;


detecting a genetic protein variation in the solubilized proteins from the sample by performing the proteomic analysis of the solubilized protein fraction;


detecting a genomic variation of the nuclear and/or mitochondrial genome by performing a genetic analysis of the solubilized DNA fraction; and


comparing the detected genetic protein variation and/or the detected genomic variation with a marker genetic protein variation and/or of a marker genomic variation respectively from the marker genetic variation database system herein described.


In some embodiments of the even more preferred embodiments of the method according to the eighth aspect in which the detecting is performed by the method according to the sixth aspect of the present disclosure, detecting a genetic protein variation is performed by detecting one or more marker genetic protein variations obtained by any one of the methods to provide a marker genetic protein variation for a biological organism according to the second aspect of the instant disclosure and/or is a marker genetic protein variation obtainable and/or obtained thereby.


In some embodiments of the even more preferred embodiments of the method according to the eighth aspect in which the detecting is performed by the method according to the sixth aspect of the present disclosure, detecting a genetic protein variation is performed by detect a genetic protein variation in a biological sample according to any one of the methods according to the third aspect of the instant disclosure.


In some more preferred embodiments of the even more preferred embodiments of the method according to the eighth aspect in which the detecting is performed by the method according to the sixth aspect of the present disclosure, in which detecting a genetic protein variation is performed by detect a genetic protein variation in a biological sample according to any one of the methods according to the third aspect of the instant disclosure, the marker genetic protein variation comprises a marker genetic protein variation from the marker genetic protein variation database system according to the fourth aspect or the fifth aspect of the instant disclosure.


In some embodiments of the even more preferred embodiments of the method according to the eighth aspect in which the detecting is performed by the method according to the third aspect of the present disclosure or the sixth aspect of the present disclosure, the marker genetic protein variation are peptide sequences corresponding to (translated from at least a portion of) a marker exome sequences indicated in Examples 43 to 45 listing exemplary set of genes validated in hair (Example 43, Table 8) bone (Example 44, Table 9) and skin samples (Example 45, Table 10) of a human being.


Preferred validated marker genetic protein variations of Homo Sapiens are indicated in Examples 46 and Example 47 listing exemplary set of GVPs validated in hair sample (Example 46, Table 11) and skin sample (Example 47, Table 12) of a human being.


Additional preferred embodiments of the method according to the eighth aspect are identifiable by a skilled person upon reading of the instant disclosure.


Any one of the embodiments of the method according to the eight aspect of the instant disclosure can be performed with components of the system according to the eighth aspect of the instant disclosure.


In any one of the systems according to the eight aspect, the system comprises exome sequences databases and/or reagents to detect exome sequences in an individual of the biological organism, alone or in combination with reagents to perform proteomic analysis of the biological sample for simultaneous combined or sequential use in the method to perform genetic analysis of a sample of a biological organism herein described.


In embodiments of the system according to the eighth aspects configured to perform a method according to the eighth aspect of the disclosure wherein the preparing is performed by the method according to the first aspect of the present disclosure, the system comprises a sample buffer typically comprising chaotropes (e.g. urea and/or thiourea), detergents (e.g. 3-[(3-Cholamidopropyl)-dimethyl-ammonio]-1-propane sulfonate (CHAPS) or Triton X-100), reducing agents (dithiothreitol/dithioerythritol (DTT/DTE) or tributylphosphine (TBP)) and protease inhibitors. Preferred embodiments of the sample buffer are identifiable by a skilled person upon reading of the present disclosure


In embodiments of the system according to the eighth aspects configured to perform a method according to the eighth aspect of the disclosure wherein the detecting is performed according to any one of the methods according to the third aspect of the instant disclosure and/or using any of the related databases, the system comprises protein databases, and/or reagents to perform proteomic analysis of the biological sample in combination with exome sequence databases. In preferred embodiments, the reagents comprise a marker peptide in accordance with the present disclosure.


In embodiments of the system according to the eighth aspect, configured to perform a method according to the eighth aspect of the disclosure wherein the detecting is performed according to any one of the methods according to the sixth aspect of the instant disclosure and/or using any of the related databases, the system comprises exome sequences databases and/or reagents to detect exome sequences in an individual of the biological organism, in combination with reagents to perform proteomic analysis of the biological organism. In preferred embodiments, the reagents comprise a marker peptide in accordance with the present disclosure


In even more preferred embodiments of the system according to the eighth aspect in which the reagents in the system comprises a marker peptide, the marker peptide comprises one or more of the marker peptides comprising a genetic protein variations validated in Homo Sapiens indicated in Examples 46 and Example 47 indicating exemplary set of GVPs and related mutated peptides validated in hair (Example 46, Table 11) and skin (Example 47, Table 12) samples of human beings. In particular exemplary marker peptides that can be preferably used or comprise in the method and system according to the third aspect, comprise any combination of the peptides having sequence SEQ ID NO: 150 to SEQ ID NO: 748 (Example 46, Table 11) for detection in hair samples of human beings, and any combination of the peptides having sequence SEQ ID NO: 749 to SEQ ID NO: 829 (Example 47, Table 12) for detection in skin samples of human beings.


In view of the above exemplary systems of the instant disclosure according to the eight aspect of the instant disclosure, comprise:

    • one or more marker peptides which preferably can comprise
      • one or more of the peptides having sequence SEQ ID NO: 150 to SEQ ID NO: 748 for detection in hair samples of human beings; and/or
      • one or more of the peptides having sequence SEQ ID NO: 749 to SEQ ID NO: 829 for detection in skin samples of human beings;
    • reagents for dissolving and/or digesting the sample and/or for detecting a marker genetic protein variation comprising for example
      • a reducing agent such as DTT, DTBA, BME, TCEP, and DTE), with detergent concentration ranges broadly from 0.001 M to 10 M; more preferably 0.05 M to 0.2 M; and most preferably 0.1 M; even more preferably the reducing agents comprise DTT;
      • a surfactant such as Invitrosol, ProteaseMax, Rapigest SF, and PPS Silent Surfactant, in particular in embodiments where the proteomic analysis is then performed by mass spectrometry; with surfactant concentration (percent w/v) ranging from 0.0001 to 1.0%; more preferably 0.001 to 0.2%; and even more preferably 0.01%; preferably the surfactant comprise ProteaseMax;
      • a detergent such as SDD, SDS, CHAPS, Triton, NP-40, and LDS) with detergent concentration (percent w/v) ranging from 0.001% to 10%; more preferably 1 to 3%; even more preferably 2%; preferably the detergent comprises SDD;
      • an enzyme for protein digestion, in particular to cut the proteins in the sample in a site specific fashion, such as trypsin, chymotrypsin, Lys-C, Arg-C, Asp-N, and Glu-C, non-specific; pepsin, and proteinase K; with concentrations for digest ranging from .0001 to 1 μg/μL; more preferably 0.01 to 0.001 μg/μL; even more preferably 0.005 μg/μL; preferably the enzyme comprises trypsin;
      • a buffer such as ammonium bicarbonate (preferred), ammonium hydrogen bicarbonate, acetates, and formates; and/or
      • ammonium bicarbonate (ABC) in concentrations ranging from 0.001 to 1M; more preferably 0.01 to 0.1 M; even more preferably 0.05 M
    • to be combined in the system according to configurations identifiable by a skilled person upon reading of the present disclosure.


In some embodiments, the one or more marker peptide can be labeled.


The terms “label” and “labeled” as used herein refer to a molecule capable of detection, including but not limited to radioactive isotopes, fluorophores, chemiluminescent dyes, chromophores, enzymes, enzymes substrates, enzyme cofactors, enzyme inhibitors, dyes, metal ions, nanoparticles, metal sols, ligands (such as biotin, avidin, streptavidin or haptens) and the like. The term “fluorophore” refers to a substance or a portion thereof which is capable of exhibiting fluorescence in a detectable image. As a consequence, the wording “labeling signal” as used herein indicates the signal emitted from the label that allows detection of the label, including but not limited to radioactivity, fluorescence, chemoluminescence, production of a compound in outcome of an enzymatic reaction and the like.


Accordingly, in embodiments of the disclosure a labeled peptide is a peptide attaching a label making the peptide capable of detection.


The terms “detect” or “detection” as used herein indicates the determination of the existence, presence or fact of a target in a limited portion of space, including but not limited to a sample, a reaction mixture, a molecular complex and a substrate. The “detect” or “detection” as used herein can comprise determination of chemical and/or biological properties of the target, including but not limited to ability to interact, and in particular bind, other compounds, ability to activate another compound and additional properties identifiable by a skilled person upon reading of the present disclosure. The detection can be quantitative or qualitative. A detection is “quantitative” when it refers, relates to, or involves the measurement of quantity or amount of the target or signal (also referred as quantitation), which includes but is not limited to any analysis designed to determine the amounts or proportions of the target or signal. A detection is “qualitative” when it refers, relates to, or involves identification of a quality or kind of the target or signal in terms of relative abundance to another target or signal, which is not quantified.


In preferred embodiments of the disclosure, peptides comprised in one of any of the systems of the disclosure are isotopically labeled or chemically labeled.


In particular, in embodiments, wherein a peptide is isotopically labeled and the detecting is performed by MS, the peptide is preferably labeled at the C terminus amino acid if y-series fragments predominate the MSMS spectrum, and preferably labeled at the N terminus amino acid if b-series fragments predominate the MSMS spectrum.


In embodiments wherein the detecting is performed by mass spectrometry, the label can comprise tandem mass tags.


In embodiments of any systems of the disclosure, wherein one or more marker peptides are comprised in the system, reagents to similarly label the unknown sample can further be provided as component of the system as will be understood by a skilled person.


Additional components of the system according to any one of the systems herein described and in particular of the system according to the eight aspect of the disclosure can comprise:

    • a column and/or a filter and related reagents for separating the mitocontrial DNA fraction from the protein/peptide fraction;
    • reference material with known identity panel (e.g. a characterized hair sample);
    • a template of an instrument method preloaded with the MSMS transitions corresponding to the identity panel; and/or
    • a statistical tool to derive statistical measures (like random match probabilities and likelihood ratios) from the results of the detecting (e.g. LCMS results), for example statistical tools:
      • comprising population-specific frequencies for markers in an identity panel
      • accounting for linkage between markers if desired; and/or
      • providing algorithms for
    • individual identification;
    • paternity testing (or other familial relationship); and/or
    • ancestry determination,


      as well as possibly additional components in configurations selected to perform one or more methods herein described, the configurations identifiable by a skilled person upon reading of the present disclosure.


In preferred embodiments of the marker genetic protein variations, databases, methods and systems and related genetic protein variation analysis herein described, performing a proteomic analysis is carried out by performing mass spectrometry of a fractionated digested peptide of the biological sample to obtain a mass spectrum of each of the fractionated digested peptide.


In further preferred embodiments of the marker genetic protein variations, databases, methods and systems and related genetic protein variation analysis herein described, the sample is hair and/or skin.


Methods and systems and related marker genetic protein variations and databases herein described, also allow in several embodiments to provide more reliable results for a specific query (such as whether there is a match between a sample and a certain individual or groups of individuals linked together by common genetic features).


Methods and systems and related marker genetic protein variations and databases herein described, further allow in several embodiments to perform genetically variant protein analysis applicable to samples from all tissues and are therefore not limited to hair; also the targeted approaches can improve LC-MS/MS analysis of bulk sample as well as analysis of samples available in smaller amounts processable according to the first aspect with particular reference to forensics applications.


As used herein, the wordings “forensics”, “forensic science” or “forensic analysis” refers to the application of science to criminal and civil laws, and in particular with regard to criminal investigation, as governed by the legal standards of admissible evidence and criminal procedure. Additionally, as used herein, the wordings “forensics”, “forensic science” or “forensic analysis” also refer to the application of forensic techniques to other types of investigation, such as determination of relatedness of individuals, or bioarcheological research, among others identifiable by those skilled in the art upon reading of the present disclosure. Accordingly, forensics involves the collection, processing, and analysis of scientific evidence during the course of an investigation.


The systems herein disclosed can be provided in the form of kits of parts. In kit of parts for performing any one of the methods herein described, one or more marker peptide and/or other standards, and/or one or more databases can be included in the kit alone or in the presence of additional sequences, reagents such as labels, reducing agents, surfactants, detergents, enzymes, buffers, as well as additional components, such as columns, filters, templates, reference materials and/or statistical tools identifiable by a skilled person upon reading of the instant discloure.


In a kit of parts, the one or more marker peptide, standards, and/or databases and additional reagents identifiable by a skilled person are comprised in the kit independently possibly included in a composition together with suitable vehicle carrier or auxiliary agents. For example, one or more marker peptides can be included in one or more compositions together with reagents for detection also in one or more suitable compositions.


Additional components of kits of parts according to the disclosure are identifiable by a skilled person upon reading of the present disclosure.


In embodiments herein described, the components of the kit can be provided, with suitable instructions and other necessary reagents, in order to perform the methods here disclosed. The kit will normally contain the compositions in separate containers. Instructions, for example written or audio instructions, on paper or electronic support such as tapes, CD-ROMs, flash drives, or by indication of a Uniform Resource Locator (URL), which contains a pdf copy of the instructions for carrying out the assay, will usually be included in the kit. The kit can also contain, depending on the particular method used, other packaged reagents and materials (i.e. wash buffers and the like).


Further details concerning the identification of the suitable carrier agent or auxiliary agent of the compositions, and generally manufacturing and packaging of the kit, can be identified by the person skilled in the art upon reading of the present disclosure


EXAMPLES

The methods and systems herein described and related marker genetic protein variations and databases are further illustrated in the following examples, which are provided by way of illustration and are not intended to be limiting.


In particular, the following examples illustrate exemplary methods, systems and related marker genetic protein variations and databases described herein. A person skilled in the art will appreciate the applicability and the necessary modifications to adapt the features described in detail in the present section, to additional methods and systems according to embodiments of the present disclosure.


Example 1
Individual Identification Using Genetically Variant Protein Analysis


FIG. 1A shows a diagram of an exemplary genetically variant protein, gasdermin, encoded by the gene GSDMA, which is shown as a member of an exemplary panel of genetically variant proteins, shown as a list in FIG. 1B.


In particular FIG. 1A is a diagram showing partial sequences of an exemplary “Reference” gasdermin, showing a partial protein-coding DNA sequence GGTACCTGC (SEQ ID NO: 1) encoding the amino acid sequence Val Thr Leu, forming part of a peptide sequence GHEVTLEALPK (SEQ ID NO: 2). Shown below the “Reference” sequence diagram are exemplary frequencies of the “Reference” gasdermin peptide sequence in European (fEUR) and African (fAFR) populations.


Also in FIG. 1B is a diagram showing partial sequences of an exemplary “Variant” gasdermin, showing a partial protein-coding DNA sequence GGTAACTGC (SEQ ID NO: 2) (comprising a single nucleotide polymorphism (SNP) “A” indicated in a box labeled “SNP”) encoding the amino acid sequence Val Asn Leu within a genetically variant peptide (GVP) comprising a single amino acid polymorphism (SAP) “Asn” indicated in a box labeled “SAP”, forming part of a peptide sequence GHEVnLEALPK and GHEVTLEALPK (SEQ ID NO: 12 and 13). Shown below the “Variant” sequence diagram are exemplary frequencies of the “Reference” gasdermin peptide sequence in European (fEUR) and African (fAFR) populations. The exemplary SNP shown is identified as rs56030650, corresponding to an entry in the National Center for Biotechnology Information dbSNP database.


Example 2
Hair Sample Preparation for Proteomic Analysis

Single hair samples (1 inch; 25 mm) from three individuals were carefully measured and cut into four equal pieces. The cut hair was then placed into separate Protein LoBind Eppendorf tubes. 100 μL of extraction buffer containing 0.05 M ammonium bicarbonate (ABC), 0.1 M dithiothreitol (DTT), 2% sodium dodecanoate (SDD) was added to each tube. Samples were then incubated at 70° C. in an ultrasonic water bath (Elma) while being ultrasonicated at high energy and frequency settings for 60 minutes or until hair was completely dissolved into solution. SDD was removed by extraction with acidified ethyl acetate (pH 2-3, 0.75% trifluoroacetic acid). After addition of 100 uL acidified ethyl acetate to each tube, samples were quickly vortexed, incubated at room temperature for 5 min, and centrifuged for 5 min at max speed (20,000×g). The upper organic phase was removed, discarded to waste, and the extraction process was repeated once. The remaining lower aqueous phase was then readjusted to pH 8 with ABC [13]. Carbamidomethylation of free cysteines was performed by adding 6μL of iodoacetamide (1.0 M) and incubation for 60 min in the dark at 25° C. To further solubilize proteins, 0.01% protease max (3 μL of 1.0% w/v) was added to each sample. Prior to proteolysis, the solubilized protein solution was concentrated to 50uL using 10 kD molecular weight spin concentrators (Millipore). Trypsin (1 μL of 0.5 μg/μL) was then added to each protein sample. Protein digestion was performed at 25° C. for 20/22 hours while being continuously agitated by magnetic-bar stirring. Resulting peptide mixture is then filtered using 0.1 μm PTFE filter, and transferred into fresh vials for mass spectrometric analysis (stored at −4.0-20° C.). Additional step of speed vacuum (20 minutes at 60° C.) can be used to concentrate peptide fraction of samples.


Ultrasonic frequency of 37 kHz is used to maximize dissolving of hair as recommended for dissolving, mixing, dispersing in Elma Elmasonic P user manual. Lower frequency setting concentrates power throughout the water bath and results in better dissolving of hair than the higher option (80 kHz). Elevated temperature setting is used (70° C.) to achieve solubilization of hair matrix. Ultrasonic using sweep mode controls the sound pressure throughout the water bath. This setting applies a more homogeneous sounding of the cleaning bath by the continued displacement of the sound pressure maxima in the cleaning liquid, leading to a more uniform ultrasonic intensity throughout the ultrasonic tank and samples. Ultrasonic power setting of 100% is used for hair matrix solubilization to maximize the force applied. [Reference: www.imlab.be/imlab_n1/e1ma/Pdf/Elmasonic_P/Elmasonic_P_Operating_Instructions_ENG_Iml ab.pdf)


Lower temperature settings ranging from 50-65° C. increase the time needed for complete solubilization substantially (from average of 60 minutes to 12 hours), but can be used to dissolve hair. Time of ultrasonic treatment at 70° C. depends on each given sample. Average of 30 to 60 minutes is efficient for hair solubilization. Brief sonication (30 seconds to 5 minutes) at lower temperature 37° C. is commonly a technique used for protein extractions for various tissues [14-17]. Protein extraction procedure is implemented at atmospheric pressure however, increasing pressure could decrease the amount of time needed for extraction [18].


Adaptation of method to perform sample preparation for proteomic analysis herein described exemplified herein for single hair to bone, teeth, fingerprint and other sample types would be achieved in several ways. For bone and tooth samples, single-hair extraction buffer could be applied to samples prior to mechanical milling procedures. Acid etching could be performed using 1 M HCl. This would be amenable to SDD liquid-liquid extraction step in the single-hair method due to the need to acidify ethyl acetate for SDD removal [19, 20]. In this case, non-acidified ethyl acetate would be used to extract SDD from samples. For finger-print and other samples, the single-hair method can be implemented by decreasing ultrasonic incubation time and decreasing sonication temperature. Exemplary adaptation of the protocol described in the current example to bone and teeth are reported in the following Examples 3 and 4.


Example 3
Bone Sample Preparation for Proteomic Analysis

Associated soft tissue was resected from each rib and a 20 mg block of cortical bone, roughly 1×3×4 mm, resected using a dental drill (NSK NE-213G) equipped with a diamond tip blade at room temperature (25° C.). Each sample was transferred into milling tubes that contained 2.8 mm ceramic bead media (Omni-International, Kennesaw, Ga.). Acid etching was performed by milling for 3 min @ 6.00 m/s in the presence of 1.2 M HCl (200 μL), reducing by addition of 3 μmol DTT (1.0 M) and incubation at 56° C. for 60 min. The supernatant was neutralized to pH 7.5-8.0 with a threefold molar excess of ammonium bicarbonate. Carbamidomethylation was then conducted by adding 6 μmol iodoacetamide and incubating at 22° C. and for 60 min in the dark. The reaction was quenched by the addition of 6 μmol DTT for 5 min. Solubilized proteins were then digested with the addition of 0.5 μg trypsin (TPCK-treated, sequencing grade, Worthington Inc., Lakewood, N.J.), and 30μg ProteaseMAX™ (Promega Inc., Madison, Wis.). The protein digest was performed at 37° C. for 20 to 22 hr. After digestion, peptide samples were centrifuged (30 min, 16,300 g, 22° C.), the supernatant filtered using a centrifugal 0.1 μm PTFE filter (Millipore Inc., Billerica, Mass.), and transferred into autosampler vials for mass spectrometric analysis (stored at −4.0 to −20° C.).


Example 4
Teeth Sample Preparation for Proteomic Analysis

The protocol for tooth sample processing was adapted from the Porto et al. manuscript published in 2011. Wisdom tooth enamel samples from individuals (5 female, 5 male, and 1 archaeological) were stored at -20° C. until they were re-sectioned using a diamond tip blade at room temperature (25° C.). Enamel and enamel-dentine junction were carefully separated from the dentine, weighed, and -20 mg was transferred into milling tubes that also contain milling beads.


Prior to milling, 200 μL of 1.2 M HCl was added to each sample. Samples were milled in acid for 3 min @ 6.00 m/s and then centrifuged at max speed (5 min, 16,300 g, 22° C.). The supernatant were neutralized by measuring pH using paper and adjusting it to 7.5-8.0 pH by adding 2 M ammonium bicarbonate 90 μL. Soluble proteins were reduced by adding of 3 μL DTT (1 M) and incubating at 56° C. for 60 min. Alkylation was performed by adding 6 μL of iodoacetamide (1 M) at 25° C. and incubating for 60 min in the dark. Carbamidomethylation reaction was quenched by the addition of 6 μL DTT (1 M) and incubating at room temperature for 5 min. To further solubilize proteins, 0.01% protease max (3 μL of 1.0% w/v) was added to each sample. Trypsin (1 of 0.5 μg/μL) was then added to each protein sample, and then incubated at 37° C. for 20/22 hr. After digestion, peptide samples were centrifuged (30 min, 16,300 g, 22° C.) to remove particulates, filtered using 0.45 μm PTFE filters into fresh vials for mass spectrometric analysis (stored at −4.0-20° C.).


Reference is made to [19, 20], each incorporated herein by reference in its entirety.


Example 5
Proteolytic Cleavage of Prepared Samples

Various applicable methods can be used to perform proteolytic cleavage (and in particular trypsinization) of proteins as will be understood by a skilled person.


In particular, during protein solubilization reduction of cysteine disulfide bonds is achieved using 100 mM of reducing agent dithiothreitol (DTT). DTT concentrations can vary from 50 mM to 180 mM. Carbamidomethylation of free cysteines is performed by adding 6μL of iodoacetamide (1.0 M) and incubation for 60 min in the dark at 25° C. [21, 22]. Alkylation time can vary from 45-60 minutes, longer reaction times increase confidence in reaction completion.


To further solubilize proteins, 0.01% protease max (3 μL of 1.0% w/v) can be added to each sample. Prior to proteolysis, the solubilized protein solution was concentrated to 50 uL using 10 kD molecular weight spin concentrators (Millipore). Trypsin (1 μL of 0.5 μg/μL) is then added to each protein sample. Protein digestion is performed at 25° C. for 20/22 hours while being continuously agitated by magnetic-bar stirring.


Digestion time can range from 16-22 hours. Agitation can be achieved by other techniques including sample rotated, milling, and shaking [23].


Reference is also made to [1, 21-23], each of which is incorporated by reference in its entirety.


Example 6
Comparison of Methods for Sample Preparation for Proteomic Analysis

An exemplary method of single hair sample processing performed according to method to perform sample preparation herein described and subsequent proteomic analysis of GVPs is shown in the lower portion of the schematic of FIG. 2, which also shows an exemplary “Bulk” hair processing method wherein sample preparation is performed with conventional methods for comparison.


In an exemplary single hair processing method according to the schematics of FIG. 2, single hair samples (25 mm) from three individuals were carefully measured and cut into four equal pieces. The cut hair was then placed into separate Protein LoBind Eppendorf tubes. 100 of extraction buffer containing 0.05 M ammonium bicarbonate (ABC), 0.1 M dithiothreitol (DTT), 2% sodium dodecanoate (SDD) was added to each tube. Samples were then incubated at 70° C. in an ultrasonic water bath (Elma) while being ultrasonicated at high energy and frequency settings, (here 330 W and 37 kHz respectively) for 60 minutes or until hair was completely dissolved into solution. SDD was removed by extraction with acidified ethyl acetate (pH 2-3, 0.75% trifluoroacetic acid). After addition of 100 μL acidified ethyl acetate to each tube, samples were quickly vortexed, incubated at room temperature for 5 min, and centrifuged for 5 min at max speed (20,000 x g). The upper organic phase was removed, discarded to waste, and the extraction process was repeated once. The remaining lower aqueous phase was then readjusted to pH 8 with ABC [13]. Carbamidomethylation of free cysteines was performed by adding 6 μL of iodoacetamide (1.0 M) and incubation for 60 min in the dark at 25° C. To further solubilize proteins, 0.01% ProteaseMax reagent (Promega, 3μL of 1.0% w/v) was added to each sample. Prior to proteolysis, the solubilized protein solution was concentrated to 50 μL using 10 kD molecular weight spin concentrators (Millipore). Trypsin (1 μL of 0.5 μg/μL) was then added to each protein sample. Protein digestion was performed at 25° C. for 20-22 hours while being continuously agitated by magnetic-bar stirring. After digestion, peptide samples were centrifuged (30 min, 16,300 x g, 22° C.) to remove particulates, filtered using 0.1 μm PTFE filter, and transferred into fresh vials for mass spectrometric analysis (stored at −4.0-20° C.) .


For comparison, in an exemplary “Bulk” hair method (e.g., using 10 mg hair sample), performed with conventional sample preparation methods, the sample is initially denatured using dithiothreitol (DTT), ammonium bicarbonate (ABC), urea, and ProteaseMax reagent (Promega, P-max), followed by mechanical milling of the sample comprising multiple steps as described herein and identifiable by those skilled in the art together with cysteine protection. Following mechanical milling, the proteins present in the sample are proteolytically digested with trypsin in a reaction mixture together with DTT, ABC and P-max, followed by centrifugation and filtration before analysis by LC-MS/MS. In contrast, in the exemplary “Single hair” method (e.g., using 85 μg hair, 2.5 cm in length) the sample is initially dissolved using a reaction mixture comprising DTT, ABC and sodium dodecanoate (SDD) and sonication at 70° C.


After dissolving, the sample is separated into organic phase, which is discarded, and aqueous phase, which is retained and further processed for protection of free cysteines, and spin-filter concentration of solubilized proteins, prior to proteolytic digestion by trypsin and filtration, followed by proteomic analysis by LC-MS/MS.


Exemplary results of proteomic metrics for samples processed using the exemplary method to perform a proteomic tissue sample preparation using single hairs, compared to an exemplary “Bulk” hair processing method are shown in FIG. 3.


In particular, FIG. 3 shows exemplary results illustrating improvements in proteomic sample preparation performed with using methods for sample preparation herein described in comparison with convention sample preparation methods.


In particular FIG. 3 Panel A shows a diagram showing exemplary protein coverage heat maps for an exemplary conventional sample preparation method (indicated as ‘Bulk hair’) and an exemplary sample preparation method of the present disclosure (indicated as ‘Single hair’). In particular, the illustration of FIG. 3A show that the protein coverage from single hair provides detection of approx. 60% of amino acids relative to bulk method, wherein the 60% amino acids are observed with only ˜1% of the bulk sample amount. The illustration of FIG. 3B also shows a detection of ˜30% of known GVPs with the sample preparation method of the disclosure relative to convention methods (same subject).



FIG. 3 Panel B shows a graph reporting exemplary results of the number of amino acids observed (a measure of protein coverage) in samples processed using exemplary convention methods on bulk hair, and single hair' (indicated as “Bulk hair” and ‘Old Single hair’ respectively) or sample preparation according to the present disclosure (indicated as “New Single hair”). In particular, in the illustration of FIG. 3 Panel B, the graph shows an improvement in protein coverage (number of amino acids observed) using the sample preparation method of the disclosure which allow >80% increase in the number of amino acids observed and therefore allow proteomic results from 1″ single hairs to be on par with proteomic results obtainable on bulk hair prepared with conventional methods.



FIG. 3 Panel C and D show graphs reporting exemplary results of the number of protein identifications in each sample (Panel C) and unique peptide identifications in each sample (Panel D) in samples processed with convention methods and the sample preparation methods of the disclosure (indicated as “Bulk hair” and “Single hair” respectively). In particular FIG. 3 Panel C and D show an improvement in these additional proteomic metrics which indicates reliability of detection in a specific sample, in samples prepared with sample preparation methods of the disclosure vs conventional preparation methods. Such an improvement is observed despite having the sample preparation methods performed in a biological sample (single hair) with a lower amount of biological material (and in particular protein material available). Such an improvement is associated with an improved detection the genetically variant peptides identified in each sample as would be understood by a skilled person.


In particular, an optimization of the data illustrated in FIG. 3 Panel C and Panel D for GVP detection can include preparation of inclusion lists, Multiple Reaction Monitoring (MRM), Explore additional MS data acquisition strategies, peptide standards/SI labeled and use alternative proteases, as would be understood by a skilled person.


As also indicated in other sections of the present disclosure although in the exemplary illustration of FIG. 3, the sample preparation of the present disclosure is illustrated with respect to single hairs, the sample preparation is also applicable to bulk hair or other samples wherein protein material is available in larger quantity.


The GVPs detected using the sample preparation method herein described can be comprised in databases of validated marker genetic variation herein described to the extent such GVPs are marker for biological organisms, type of biological organisms or individual thereof. Accordingly, an operational scenario is expected to also utilize inclusion/exclusion lists wherein the exclusion lists can refer to validated GVPs which are not marker for a specific query of interest.


Example 7
Combined mtDNA and Proteomic Analysis in a Single Hair Sample

An exemplary method sample processing for subsequent proteomic analysis of GVPs combined with analysis of mtDNA from a same sample is shown in the schematics of FIG. 4.


In particular, in the schematic of FIG. 4 the exemplary method of protein and mtDNA extraction is performed following a sample preparation performed with the sample preparation method herein described followed by proteomic analysis of the protein fraction and the genomic analysis of the mtDNA fraction, comprising DNA amplification and sequencing of the mtDNA.


In particular single hair samples (25 mm) from three individuals were carefully measured and cut into four equal pieces. The cut hair was then placed into separate Protein LoBind Eppendorf tubes. 100 μL of extraction buffer containing 0.05 M ammonium bicarbonate (ABC), 0.1 M dithiothreitol (DTT), 2% sodium dodecanoate (SDD) was added to each tube. Samples were then incubated at 70° C. in an ultrasonic water bath (Elma) while being ultrasonicated at high energy and frequency settings, (here 330 W and 37 kHz respectively) for 60 minutes or until hair was completely dissolved into solution. SDD was removed by extraction with acidified ethyl acetate (pH 2-3, 0.75% trifluoroacetic acid).


After addition of 100 uL acidified ethyl acetate to each tube, samples were quickly vortexed, incubated at room temperature for 5 min, and centrifuged for 5 min at max speed (20,000×g). The upper organic phase was removed, discarded to waste, and the extraction process was repeated once.


The remaining lower aqueous phase was then readjusted to pH 8 with ABC [13]. Carbamidomethylation of free cysteines was performed by adding 6μL of iodoacetamide (1.0 M) and incubation for 60 min in the dark at 25° C. To further solubilize proteins, 0.01% ProteaseMax reagent (Promega, 3μL of 1.0% w/v) was added to each sample. Prior to proteolysis, the solubilized protein solution was concentrated to 50 μL using 10 kD molecular weight spin concentrators (Millipore). Trypsin (1 μL of 0.5 μg/μL) was then added to each protein sample. Protein digestion was performed at 25° C. for 20-22 hours while being continuously agitated by magnetic-bar stirring.


A protocol for isolation of DNA from tissues was provided by the Qiagen QlAamp DNA Micro Kit. The steps of the Qiagen QlAamp DNA Micro Kit manual were followed with exception that the lysis procedural steps that include adding proteinase K, addition of Qiagen proprietary buffer ‘ATL’, pulse-vortexing, overnight incubation at 56° C., and addition of Qiagen proprietary buffer ‘AL’ were omitted and the aforementioned trypsin incubation was substituted for these steps. Accordingly, ffollowing trypsin proteolysis, 100 μL of 100% ethanol was added to each sample as recommended by Qiagen QlAamp DNA Micro Kit instructions. Samples were then vortexed for 15 seconds, incubated at 25° C. for 5 minutes, then added into separate QIAmp miniElute columns. Columns were closed and centrifuged at 6000×g for one minute. Flow-through was collected as the peptide fraction of the extraction, filtered using a 0.1 μm PTFE filter, and transferred into fresh vials for mass spectrometric analysis (stored at +4.0 to −20° C., or +4 to −12). The bound DNA fraction was then washed according to Qiagen QlAamp DNA Micro Kit instructions and eluted twice into the same collection tube with 20 μL of warm (37° C.) water by centrifugation for one minute (20,000×g).


In the illustration of FIG. 4, the graph reports results of exemplary peptides identified by performing proteomic analysis of the protein fraction.


The genetic material recovered with the process outlined in FIG. 4, allows efficient DNA amplification/sequencing in view of the high-quality mtDNA recovered from proteomic extracts.


An exemplary illustration of DNA amplification/sequencing is illustrated in FIG. 5A wherein an exemplary mitochondrial genome and related primers are shown.


In particular the exemplary list of primers of FIG. 5A is for amplification and sequencing of amplicons of mtDNA haplogroup HV regions and is reported in Table 1 below.









TABLE 1







mtDNA gene primers for PCR and Sequencing:













SEQ





ID


Primer
Sequence
Usage
NO:





F15975
CTCCACCATTAGCACCCAAA
PCR and
136




Sequencing






F16524
AAGCCTAAATAGCCCACACG
PCR and
137




Sequencing






F015
CACCCTATTAACCACTCACG
PCR and
138




Sequencing






F403
TCTTTTGGCGGTATGCACTTT
PCR and
139




Sequencing






R16410m
GAGGATGGTGGTCAAGGGA
PCR and
140




Sequencing






R042
AGAGCTCCCGTGAGTGGTTA
PCR and
141




Sequencing






R389
CTGGTTAGGCTGGTGTTAGG
PCR and
142




Sequencing






R635
GATGTGAGCCCGTCTAAACA
PCR and
143




Sequencing









In a DNA amplification analysis of mtDNA, PCR was used for amplification of HV mtDNA regions. Amplicons were purified, quantified and sequenced using standard mtDNA protocols.


Exemplary results of PCR amplification of mtDNA recovered using the exemplary combined mtDNA and proteomic analysis sample processing protocol are shown in FIG. 5B.


The results of the above proteomic and genomic analysis can then be compared with databases to identify the validated marker GVPs to be detected and/or provided in databases herein described.



FIG. 6 shows an exemplary comparison of results of HV mtDNA region sequencing using mtDNA recovered using the exemplary combined mtDNA and proteomic analysis illustrated in the present example.


In particular in FIG. 6, an exemplary Clustal Omega alignment is shown of HV mtDNA regions of samples obtained from three independent subjects (indicated as U1.003b-A_HV1, SEQ ID NO: 88, L1.006a-A_HV1, SEQ ID NO: 89, and L1.046a+b-A_HV1, SEQ ID NO: 90) aligned with a reference mtDNA sequence (indicated as rCRS_HV1, SEQ ID NO: 87). The black boxes indicate exemplary SNPs identified in the sequences.


Example 8
Exome Sequence Analysis

Applicable methods to detect exome sequences of the sample of the biological organism are identifiable by a skilled person.


According to an exemplary protocol blood and buccal samples can be used to perform DNA collection from individuals. DNA is isolated from blood associated with each sample and was subsequently analyzed by Sanger sequencing (2016 Sorenson Genomics, LLC). Full exome sequencing of the extracted DNA was also obtained (10-0111_ACE Research Exome with Secondary Analysis; 8 Gb; Alignment, Variant Calling and Annotation; ©2016 Personalis Inc).


Comparison of detected exome sequences and a database of exome sequences of the biological organism can then be performed. Exemplary databases that can be used comprise protein and genome sequence databases such as Uniprot [24] (www.uniprot.org/), Exome Variant Server (evs.gs.washington.edu/EVS/) Swiss-Prot [25](www.ebi.ac.uk/swissprot/), Ensembl [26] (www.ensembl.org/index.html) can be used to identify genetically variant peptide sequences in proteins. Sequence alignment webservers including BLAST [27] (www.ncbi.nlm.nih.gov/BLAST/), Prowl [28]; (www.prowl.rockefeller.com), and Protein Information Resource [29, 30]; (pir.georgetown.edu/) can be used to determine if peptide sequences are unique to a single human gene.


References is also made to the following documents incorporated herein by reference in their entirety [25-30].


Example 9
Proteomic Analysis to Detect Peptide Sequences

Applicable methods to perform proteomic analysis to detect the peptide sequences are identifiable by a skilled person inclusive of any possible ways to perform a) LC separation of peptides orb) tandem MS analysis (to generate the ‘raw MS data’) c) analysis methods other than LC-MS/MS, e.g. protein quantification, antibody based assays, gel purification/isolation (2d and other),and additional methods.


In an exemplary approach, data acquisition was performed using Thermo Scientific Q Exactive Plus Hybrid Quadrupole-Orbitrap mass spectrometer fitted with Easy-nLC 1000 HPLC (Thermo Scientific, Asheville, N.C., USA). Various combinations of liquid-chromatography systems coupled to mass spectrometers, peptide fragmentation techniques, and ionization methods can be used to generate peptide sequence identifications [31, 32]. Peptides were separated by reversed-phase liquid chromatography using a mobile phase A (0.01% TFA in water) and mobile phase B (0.01% TFA in acetonitrile) in a 97 minute gradient. 2 μL of each sample were injected onto a C18 trap cartridge and preceded by an Easy-Spray™ nanoflow (1 mm×150 mm) column (Thermo Scientific, Asheville, N.C., USA) with a flow rate of 3 μL/min. Numerous reversed-phase columns are commercially produced and distributed that are applicable to perform proteomic analysis of peptide sequences [33-35]. Electrospray ionization was achieved in positive mode with a voltage of 2-4 kV. Dynamic exclusion data collection was implemented at a MS scan range of 180-1,800 m/z, top 10 precursor ions were chosen for subsequent MS/MS scans and excluded after 10 seconds.


Due to extremely small quantities of protein solubilized from extractions of a single hair, many conventional quantification assays have insufficient limits of detection for example Bradford assay and UV absorbance measurements at 280 nm [36, 37]. Peptide quantification via fluorometric assay (Pierce™) of small volumes using nano fluorospectrometer (NanoDrop™ 3300 Fluorospectrometer; Thermo Scientific™) is most applicable for the single-hair method [38].


References is also made to the following documents incorporated herein by reference in their entirety [31-38].


Example 10
Proteomic Analysis Performed by Liquid Chromatography and Mass Spectrometry

Liquid Chromatography and Mass Spectrometry data acquisition was performed using Thermo Scientific Q Exactive Plus Hybrid Quadrupole-Orbitrap mass spectrometer fitted with Easy-nLC 1000 HPLC (Thermo Scientific, Asheville, N.C., USA). Peptides were separated by reversed-phase liquid chromatography using a mobile phase A (0.01% TFA in water) and mobile phase B (0.01% TFA in acetonitrile) in a 97 minute gradient. 2 μL of each sample were injected onto a C18 trap cartridge and preceded by an Easy-Spray™ nanoflow (1 mm×150 mm) column (Thermo Scientific, Asheville, N.C., USA) with a flow rate of 3 μL/min. Electrospray ionization was achieved in positive mode with a voltage of 2-4 kV. Dynamic exclusion data collection was implemented at a MS scan range of 180-1,800 m/z, top 10 precursor ions were chosen for subsequent MS/MS scans and excluded after 24 seconds.


Data Analysis was performed using PEAKS 7.5 (Bioinformatics Solutions Inc., Waterloo, Ontario, Canada) protein identification software was used to search each RAW data file to determine the specific proteins that were identified in each sample. Search settings included partial posttranslational modifications including oxidation of methionine, deamidation of asparagine and glutamine, and hydroxyproline. Precursor mass error of 15 ppm using monoisotopic mass was used for parent ion identifications and a 0.05 Da for fragment ions masses. A decoy database was generated within the software using a protein library of all human protein sequences exported from UniProtKB/Swiss-Prot knowledgebase (The UniProt Consortium; www.uniprot.org/). The decoy database is used to determine the false determination rate (FDR) of protein identifications. Protein identifications (IDs) were filtered by a 1% FDR. Filtered protein IDs found in each individual data file was outputted and aligned using Scaffold proteomics software [39]. IDs were then additionally filtered by having two or more unique peptides detected.


Characterization of genetically variant peptides (GVPs) was performed using the Global Proteome Machine webserver (GPM; www.thegpm.org). Raw data was exported and converted into mgf format using MSconvertGUl (Proteowizard 2.1.×; proteowizard.sourceforge.net) and submitted to the Global Proteome Machine webserver (GPM; www.thegpm.org). Default search settings were used with the exception of the human male NCBI reference protein database, a 20 ppm error for the primary scan, inclusion of complete cysteine carbamidomethylation (C+57), and partial modifications of oxidized methionine (M+16), and deamidation (N+1, Q+1). Results from this search were filtered by single nucleotide polymorphism (SNPs) accessions (rs numbers) to obtain a list of previously characterized potential GVPs.


Genetically Variant Peptide Confirmation from Genetic Sequencing was performed as follows: DNA was isolated from blood associated with each sample and was subsequently analyzed by Sanger sequencing (2016 Sorenson Genomics, LLC). Full exome sequencing of the extracted DNA was also obtained (10-0111_ACE Research Exome with Secondary Analysis; 8 Gb; Alignment, Variant Calling and Annotation; ©2016 Personalis Inc). Genotypes obtained by exome that corresponded to missense variants were used to validate the observation of GVPs in proteomic data. Potential GVP identifications were filtered to cases where proteomic detection of a GVP was correlated to the correct SNP genotype determined in exome sequence data.


Exome validated genetically variant peptides (GVPs) observed in each sample were directly correlated to corresponding genotypes of missense single nucleotide polymorphism (SNP) at each locus. Using the 1000 genomes project database (1000 Genomes Project Consortium, Phase 3) population, random match probabilities (RMP) were calculated for each possible genotype (p=probability allele 1, q=probability allele 2) where both alleles p and q are defined by equation 1.










p





or





q

=


number





of





times





allele





observed


size





of





database






Eq
.





(
1
)








Genotype frequencies for each locus was calculated depending on heterozygosity of where heterozygous genotypes (2pq) and for minor allele homozygous (p2). Individual profile frequencies (P) were then calculated by implementation of the product rule on each set of observed genotypes and their calculated RMP values (al and for the first locus a2 for the second . . . ; Equation 2)






P(a1a2)=P(p1q1|p12P(p2q2|p22)   Eq. (2)


In cases where a heterozygous genotype was observed in the exome sequencing data and only one allele was detected in proteomic data, only the probability corresponding to the allele of the observed GVP was considered.


Example 11
Comparison of Detected Marker Exome Sequence with Detected Peptide Sequences to Provide a Validated Genetic Protein Variation

Applicable methods to perform comparing the detected marker exome sequence with the detected peptide sequences to provide a marker genetic protein variation validated for the same of the biological organism, are identifiable by a skilled person.


There are several approaches to validate detected genetically variant peptides. Exemplary methods comprise implementing different protein identification software algorithms, DNA sequencing techniques, and mass spectrometry peptide confirmation. Single-hair method implements program PEAKS 7.5 (Bioinformatics Solutions Inc., Waterloo, Ontario, Canada) for variant peptide detection.


A reference database created by translating polymorphisms (missense SNPs, insertions, deletions, and stops/gains) that influence protein sequences observed in exome results into mutated protein sequences are used for peptide identification within software parameters. Experimental conditions and instrumental capabilities inform parameters chosen for search. Search settings include partial posttranslational modifications including oxidation of methionine, deamidation of asparagine and glutamine, and carbamidomethylation of cysteine. Precursor mass error of 30 ppm using monoisotopic mass was used for parent ion identifications and a 0.05 Da for fragment ions masses.


Other parameter settings can be chosen depending on instrument dependent metrics including parents and fragment mass errors. Additionally, software program PEAKS 7.5 (Bioinformatics Solutions Inc., Waterloo, Ontario, Canada) protein identification software can be used to identify putative peptide variants using a specific capability called Spider [40] without using mutated reference databases. Another approach, outlined in [3] uses the Global Proteome Machine webserver (GPM; www.thegpm.org) to detect possible peptide variants. Genetic confirmation of detected peptide variants can be performed by Sanger sequencing [41], whole-exome DNA sequencing, or other DNA sequencing methods [42].


Alternatively, observed genetically variant peptides can be confirmed using synthetic peptide internal standards that can be isotopically labeled [43].


References is also made to the following documents incorporated herein by reference in their entirety [40-43].


Example 12
Exemplary Genetic Protein Variations

Any detectable genetic protein variations can be used in methods and systems herein described as will be understood by a skilled person. Exemplary GVP comprise not only SAPS but also insertions, deletions, and stops variation as will be understood by a skilled person


In particular, insertions, deletions, and stop mutations observed in exome sequencing results can be directly translated into reference mutated databases. Peptide masses reflecting these polymorphisms can also be predicted using in silico proteolysis analysis and targeted mass spectrometry techniques [44]. Targeted mass-spectrometry based techniques including parallel reaction monitoring, selected ion monitoring, or mass inclusion list methods during mass-spectrometry data acquisition can be used to confirm presence of variant peptides in samples [45-47].


References is also made to the following documents incorporated herein by reference in their entirety [44-47]


Example 13
Comparison of Top-Down Approaches and Bottom-Up Approaches for Identification/Detection of a Genetic Protein Variation

A schematic comparison of the steps used to perform a top-down approach of the disclosure versus the conventional approaches to identify genetic protein variations is shown in FIG. 7.


In particular, FIG. 7 shows a diagram indicating two different approaches to GVP discovery, one approach being “exome-driven” otherwise referred to herein as “top-down discovery” as shown in the top triangle (dark grey), and the other being “proteome-driven” otherwise referred to herein as “bottom-up discovery”, as shown in the bottom triangle (light grey).


As described herein, the proteome-based discovery approach begins with proteomic analysis, followed by candidate peptide identification, and DNA validation of identified GVPs.


Thus, the proteome-driven approach has limitations such as being a ‘needle in a haystack’ approach that is not compatible with targeted proteomic analysis and relies on manual MS interpretation to identify potential GVPs, wherein potential GVPs are then validated by separate individual genotyping experiments.


In contrast, the exome-driven approach begins with obtaining exome data, allowing identification of relevant SNPs, followed by proteomic validation of GVPs. Thus, the “exome-driven” approach features (1) obtaining exome sequence for each donor, (2) establishing a workflow to identify specific SNPs of interest, (3) targeted proteomic analysis allowing simplified identification of GVPs in raw MS data, and (4) allows a logic-driven GVP selection, identification, and validation process.


A more detailed exemplification of methods according to the bottom-up approach and the top-down approaches are illustrated in the following Examples 14 to 17.


Example 14
Identification of a Validated Common GVP Panel Following Bottom-Up Approach

An exemplary method to identify a pooled marker genetic variation database in accordance with embodiments herein described is illustrated in FIG. 8.


In particular, FIG. 8 shows a schematic of an exemplary “proteome-driven” GVP discovery and evaluation method. In the exemplary proteome-driven GVP discovery approach, a peptide mixture is obtained from a sample (e.g. from hair) and is analyzed by LC-MS/MS to provide a ‘Mass Spec Dataset’, which is then analyzed with reference to a protein variant database using analysis software tools such as MASCOT, PEAKS, and GPM. In the GVP discovery workflow, candidate GVPs in the observed proteins identified in the sample are screened using metrics such as match score, frequency, and qualitative assessment.


The screened GVPs are then validated by confirming the GVPs comprise missense mutations genetically encoded by SNPs by genomic sequencing to provide validated GVPs. The validated GVPs then are incorporated into a GVP database, which is used for analysis of operational samples, wherein matches to known GVPs provide identity metrics.


Example 15
Exome-Driven Identification of a Validated Common GVP Panel in a Sample

An exemplary top-down approach for identification of a panel of GVPs using an “exome-driven” discovery process are outlined in the schematic of FIG. 9 and FIG. 10, wherein the approach is exemplified for a hair sample.


In particular, FIG. 9 shows a schematic of an exemplary method wherein samples from a plurality of donors are used to build a database of the ‘Observed Gene Pool’ comprising the protein-coding genes that express proteins observed in a given sample type (e.g. hair). In the exemplary method, a peptide mixture is obtained from a sample (e.g. from hair) from a donor subject and is analyzed by LC-MS/MS to provide a ‘Mass Spec Dataset’, which is then analyzed with reference to a protein variant database using analysis software tools such as MASCOT, PEAKS, and GPM. The identified ‘Observed Proteins’ in the sample are thus encoded by ‘Represented Genes’ and form the ‘Down-selected Target Genes’ of the ‘Observed Gene Pool’. Accordingly, samples from a plurality of donors are used to build a database of the ‘Observed Gene Pool’ comprising the protein-coding genes that express proteins observed in a given sample type (e.g. hair).


The ‘Observed Gene Pool’ built according to the method exemplified in FIG. 9, can then be used in the ‘exome-driven’ discovery of GVPs exemplified in the schematics shown in FIG. 10.


In the exemplary method illustrated by the schematic of FIG. 10, a donor subject's exome is sequenced to provide ‘Individual Exome Data’. In particular, sequences of ‘Down-selected target Genes’ within the ‘Observed Gene Pool’ of a given tissue sample are analyzed to detect ‘Individualized SNPs in observable target genes’. The SNPs are then annotated with information regarding the particular encoded transcripts in which they are comprised, the minor allele frequency (MAF), the genomic codon in which they are comprised, and the corresponding location and change in the amino acid encoded by the missense mutation. Using this information, an ‘Individualized Protein Database’ is built for the donor, comprising the sequences of mutant and reference proteins. In addition, a peptide mixture is obtained from a sample of a particular tissue type (e.g. from hair) from the same donor subject and is analyzed by LC-MS/MS to provide an ‘Individual Mass Spec Dataset’, which is then analyzed with reference to the donor subject's ‘Individualized Protein Database’ using Troteomic Search Tools' such as Andromeda, Byonic, Comet, Tide, Greylag, InsPecT, Mascot, MassMatrix, MassWiz, MS Amanda, MS-GF+, MyriMatch, OMSSA, PEAKS DB, pFind, Phenyx, ProblD, ProteinPilot Software, Protein Prospector, RAId, SEQUEST, SIMS, Sim Tandem, SQID, and X!Tandem, among others identifiable by those skilled in the art. or de novo search such as Cyclobranch, DeNovoX, DeNos, Lutefisk, Novor, PEAKS, and Supernovo, among others identifiable by those skilled in the art to provide ‘Validated GVPs’ that can be used in an ‘Individual or Pooled GVP Panel’ . Thus, validated GVPs comprising proteins having SAPs present in the sample from the donor are identified by targeted selection based on the observed gene pool encoded by the exome sequence of the same donor. For a ‘Pooled GVP Panel’, the process is repeated for a plurality of donors.


Example 16
Application of an “Exome-Driven” Validated Common GVP Panel to Operational Samples

An exemplary application of a GVP panel of validated markers GVP identified and/or detected using methods and systems herein described is shown in FIG. 11.


According to the exemplified exome drive approach shown in FIG. 11, a peptide mixture is prepared and a ‘Mass Spec Dataset’ is obtained for an operational sample (e.g. a found sample from an unknown individual), such as a ‘Questioned Hair Sample’. Using ‘Targeted Search Tools, the ‘Mass Spec Dataset’ is analyzed with reference to a Pooled GVP Panel' (wherein the ‘Pooled GVP panel’ is also referred to herein as a ‘Common GVP panel’), thus providing ‘Identity Metrics’ for the operational sample.


In the ‘Common GVP panel’, GVPs are down selected for common nsSNPs, and a consensus panel is assembled from a large cohort. As described herein, the term “common nsSNPs” refers to nsSNPs having a frequency >1% having a worldwide distribution. A Pooled GVP panel can be provided from a population of individuals, which can then be used for analysis of an operational sample (e.g. a questioned hair sample found at a crime scene), for example in cases where a DNA sample from an individual of interest is not available; thus, identity metrics (such as biogeographic information) can be obtained for the operational sample based on the ‘Pooled GVP Panel’.


Example 17
Construction of a Common GVP Identity Panel

An exemplary method to provide a pooled marker genetic variation protein database is shown by FIGS. 12A-12B. In particular FIG. 12A shows a schematic showing exemplary construction of a validated pooled ‘common’ GVP identity panel and FIG. 12B shows an exemplary common GVP identity panel resulting from the approach of FIG. 12A.


In particular, the schematic of FIG. 12A shows an exemplary method for building a panel of validated common GVPs encoded by genes encoding proteins present in hair samples comprising 64 validated missense SNPs. In this exemplary “exome-driven” GVP discovery method, proteomic datasets and exome datasets are used together to validate a panel of common GVPs present in samples of a given tissue type (e.g. hair).


According to the illustration of FIG. 12A 72 proteomic datasets were provided, wherein 66 identified proteins were detected in at least 90% of individuals and 456 identified proteins were detected in at least 50% of individuals (FIG. 12 A top). Concurrently, exome sequences are obtained from donor individuals, in which 345 missense-encoding single nucleotide polymorphisms (msSNPs) were identified. Of these msSNPs, 285 had a frequency in the population of >1% (common msSNPs) (FIG. 12A bottom).


Of these common msSNPs, 64 encoded proteins that were also encoded by genes identified in the ‘Observable Gene Pool’. A list of the exemplary 64 GVPs identified by the approach of FIG. 12A is shown in FIG. 12B. In particular, FIG. 12B shows a list of an exemplary validated GVP identity panel for hair samples that were identified following the method summarized in the schematic shown in FIG. 12A. The abbreviated name of each of the 64 proteins identified is shown in the middle column (“Protein”), the entry number for the National Center for Biotechnology Information Single Nucleotide Polymorphism Database (“dbNSP”) missense mutation-encoding SNP is shown in the first column, and the allele frequency is shown in the third column (“Allele frequency”).


Example 18
Determination of Amounts of Proteins/GVP Detectable in a Hair Sample

Amount of proteins and number of GVP detectable in a hair sample can be provided with the approach exemplified in the schematics of FIG. 13.


According to the approach exemplified in FIG. 13, the amount/number can be provided by systematically looking at detectable proteins in individuals (e.g. up to 72 individuals) and then detecting the percentage of sample in which each protein is detected. In the Exemplary chart of FIG. 13, 4174 different proteins detected across cohort of 72 individuals 456 proteins detected in at least 50% of individuals and 66 proteins detected in at least 90% of individuals.


The related panel of proteins and GVPS is reported in Table 2 below












TABLE 2







Protein
Missense SNPs



















KRT86
245



KRT33A
141



KRT34
134



KRT36
216



KRT38
246



JUP
368



DSP
1162



LGALS3
114



SFN
83



LGALS7
10



KRT83
295



KRT85
245



SELENBP1
210



TRIM29
267










Example 19
Identity Metrics

Identity metrics provide the theoretical probability that any two randomly selected profiles with a given number of loci will match (where each locus encodes a validated GVP and the median match probability for these loci is shown on the y-axis), assuming independence of each locus.


For example, in the illustration of FIG. 14, each locus encodes a validated GVP in the exemplary panel shown in FIG. 12B and the median match probability for these loci is shown on the y-axis. If the number of loci sampled (shown on the x-axis) is 20, the probability is 5.5×10−7, or 1 in 1.8 million, and if the number of loci sampled is 30, the probability is 4.1×10−10, or 1 in 2.4 billion.


Accordingly, for a common panel of 64 validated GVPs, FIG. 14 shows a graph indicating the theoretical probability that any two randomly selected profiles with a given number of loci will match, assuming independence of each locus. As understood by those skilled in the art, linkage disequilibrium (LD) can affect theoretical genotype match probabilities such as those exemplified in FIG. 14.


Example 20
Linkage Disequilibrium Affects Genotype Match Probabilities


FIG. 15 shows an exemplary application of the product rule for calculation of the probability of an overall non-synonymous SNP profile in the population. However, nearby loci are often inherited together, therefore in some embodiments the product rule doesn't directly apply.


In the exemplary application of the product rule of FIG. 15, calculation of the probability of an overall non-synonymous SNP profile in the population (Pr(profile/population)) is estimated by determining the probability of detected nsSNP alleles, or allele combination in each gene, and then using the product rule to multiply these probabilities together (Pr(overall profile/population)). Shown are exemplary GVPs for three genes KRT35, KRT81, and TGM3, together with exemplary nsSNPs in these genes identified by their dbSNP entry IDs.


For example, many loci for exemplary validated GVPs shown in FIG. 12B are keratin genes, which are clustered on chromosomes 12 and 17. Thus, the loci encoding these GVPs may be linked though they are in different genes, and linked loci can be up to 220 kb apart]. Therefore, in some embodiments, LD can be taken into account for calculation of the probability of an overall non-synonymous SNP profile in the population. LD can be factored into the calculation by computing LD between pairs of GVP loci located on the same chromosome, for example using data from the 1000 Genomes Project dataset. Next, clusters of linked loci can be grouped, by computation of joint genotype probabilities given LD for loci within each cluster and by multiplying cluster probabilities to get overall genotype likelihood.


Example 21
Exome-Driven Identification of a Validated Common GVP Panel from Bone Samples

It is expected that GVP based identification can be expanded to additional tissue types, and that protein-based identification can be conducted with multiple forensically relevant protein sources, such as hair, bone, teeth, and fingerprint protein.



FIG. 16 shows a list of an exemplary validated GVP identity panel for bone samples, that were identified following the method similar to that indicated for hair samples as summarized in the schematic shown in FIGS. 12A-12B. The abbreviated name of each of the 17 exemplary bone-related genes identified is shown in the left column (“Gene name”), the identifier for the National Center for Biotechnology Information Single Nucleotide Polymorphism Database (dbNSP) mis sense mutation-encoding SNP is shown in the second column, together with the allele (“rs#_nuc”), the amino acid sequence of the encoded peptide comprising the SNP for each allele is shown in the third column (“Peptide”), the corresponding single amino acid polymorphism (“SAP”) is shown in the fourth column, and the allele frequency (“gf”) for European (“EUR”) and African (“AFR”) populations is shown in the last two columns.


Example 22
Exome-Driven Identification of a Validated Individual GVP Panel


FIG. 17 shows a schematic of an exemplary method to create a custom GVP identification profile for an individual.


In an exemplary method illustrated by the schematic of FIG. 17, a DNA sample is obtained from an individual (“Known DNA sample”) and the individual's exome is sequenced. One or more rare and/or private nsSNPs are then identified in the individual's exome, which can be used to create synthetic peptides encoded by the DNA sequences comprising the rare and/or private nsSNPs. Proteinaceous material (e.g. from a hair sample or other sample) is also collected from the same individual, which is processed and analyzed using LC-MS/MS. ‘Diagnostic’ LC-MS/MS spectra can then be generated for the synthetic peptides that can be used to identify a particular GVP from the individual in a complex LC-MS/MS dataset.


Accordingly. for an ‘Individual GVP Panel’, GVPs can be down-selected based on low-frequency or ‘rare’ or ‘private’ nsSNPs and the GVP panel is unique to that individual (see FIG. 17). The term “rare SNPs” as used herein refers to nsSNPs having a frequency <0.05% in a given population. For example, an ‘Individual GVP Panel’ can be provided when a DNA sample and optionally a protein sample is available from an individual of interest (e.g. a suspect of a crime in custody). The exome sequence of the individual is then obtained, rare nsSNPs identified, and ‘diagnostic’ LC-MS/MS spectra can then be generated for the synthetic peptides that can be used to identify a particular GVP particular to the individual.


Example 23
Application of an “Exome-Driven” Validated Individual GVP Panel to Operational Samples.


FIG. 18 shows a schematic of an exemplary method of applying an Individual GVP panel to an operational sample.


In the exemplary method, proteinaceous material (such as hair, house dust, fingerprint residue, urine/fecal matter, etc.) is collected (“Collection”) from a target location (e.g. a crime scene), wherein in some embodiments the proteinaceous material can comprise proteins originating from multiple contributors. Proteomic analysis of the proteinaceous material then provides a large number of highly complex fragmentation patterns. Spectral matching to a custom identification profile (“Unique synthetic peptide profile”, generated for a particular individual, e.g., following the exemplary method shown in FIG. 17) is performed, thus matching ‘diagnostic’ spectra for the individual to spectra present in the complex mixture in the LC-MS/MS data, thus confirming the prior presence of the individual at the target location. The exemplary method shown in the schematic is thus not dependent on identification of peptide sequences from databases, but instead uses a process of targeted spectral matching based on the individual GVP panel.


Accordingly, in the exemplary method illustrated by the schematics of FIG. 18, proteinaceous material (such as hair, house dust, fingerprint residue, urine/fecal matter, etc.) is collected from a target location (e.g. a crime scene), Spectral matching to a custom identification profile, is performed, thus matching ‘diagnostic’ spectra for the individual to spectra present in the complex mixture in the LC-MS/MS data, thus confirming the prior presence of the individual at the target location. The method is thus not dependent on identification of peptide sequences from databases, but instead uses targeted spectral matching based on the individual GVP profile. Thus, identity metrics can be obtained specific for the individual of interest and compared to the identity metrics of the operational sample. In particular, identification of rare nsSNPs in an individual allows in some embodiments the identification of a sample that originated from an individual in a complex sample that comprises samples from multiple contributors (see FIG. 18).


Example 24
Recovery of Trace DNA

Successful recovery of trace DNA was performed. In real-world data sets, there is 2% success rate at searchable profile from touch samples. 11% of rape kits result in successful prosecution. Table 3 shows examples of percentage of samples for which a profile is recovered [48].












TABLE 3







Recovered profile from samples
% of samples









None
44%



Unusable partial profile
21%



Mixture (usable)
22% (3%)



Usable partial profile
 6%



Full
 7%










Example 25
Value and Challenges of Protein-Based Approach

Exemplary advantages and challenges of a protein-based approach comprise those in Table 4 below.










TABLE 4





Advantages
Challenges







Genetic variation (nsSNPs) is
Lack of an equivalent to PCR for


retained in protein
amplification


Protein is considerably more stable
nsSNPs tend to be less


than DNA
discriminate than STR loci


Protein occurs at high levels in
Each protein source/tissue expresses


tissue
a subset of gene products


Extremely large pool of common
Technology limited until recently-


variants available
tools remain uncommon


New proteomic methodologies


allow attomole-level analysis









A large reservoir of genetic variation exists in the proteome: Up to 60 k common variants (>0.5%), an estimated >1700 in the hair proteome alone.



FIG. 19 shows exemplary diagrams of DNA and protein chemical structures, showing sites of depurination, oxidation, or hydrolysis.


Example 26
Overview of GVP Identification and Validation Process


FIG. 20 shows a diagram of an exemplary overview of GVP identification and validation process, showing a ‘proteome-driven’ GVP discovery approach.


Example 27
Automated In-Line Sample Processing


FIG. 22 shows a diagram of exemplary automated in-line sample processing


In particular, FIG. 22 describes an arrangement of fluidic components that enable automated in-line sample processing of proteinaceous samples such as hair. The microfluidics module including syringe pump, storage cell, associated valves (2-way and multiport valve 1) and reagent reservoirs allow for a controlled introduction of reagents to and from a digestion container, which contains the sample of interest. Each component can be software controlled to enable automation, precision and reproducibility. Flows leaving the digestion chamber are introduced to an additional multiport valve which can be controlled via software to allow automation. This valve will direct effluent to either a waste stream or a peptide capture column depending on the stage of the process that is occurring. The purpose of the peptide capture column is to concentrate the peptides resulting from the digestion process as well as to assist in removing reagents that may interfere with the analysis process. Finally, the second multiport valve allows for the introduction of an elution buffer that elutes the peptides from the peptide capture column and into a liquid chromatography/mass spectrometry system for proteomic analysis.


Example 28
Improved Data Acquisition Approaches Maximize Discovery

This example describes exemplary improved data acquisition approaches to maximize GVP discovery.


Improvements in instrumentation can maximize GVP discovery, for example, use of an advanced hybrid mass spectrometer such as the Q-Exactive Plus, which features nano-LC and nanoelectrospray, and advanced hybrid mass-spectrometry (quadrupole-orbitrap). FIG. 23 shows a graph reporting exemplary results of power of discrimination as a function of number of unique peptides identified. In particular, the arrow indicates an exemplary improvement in results from new instrumentation.


Other improved data acquisition approaches comprise use of exclusion lists, wherein data for peaks already collected in previous runs are not collected, and focusing on weaker peaks. Also, use of inclusion lists, wherein data is only collected on a specific list of GVPs that have been previously discovered in other samples, and/or predicted from genomic or proteomic databases. Also, use of improved reference databases, such as those that include all SAPs, wherein more GVPs allow greater power of discrimination.


Example 29
Incorporation of GVP Profiles and DNA Based Measures of Identity

Incorporation of GVP profiles and DNA based measures of identity can be performed by integrating single tandem repeat (STR) and mitochondrial DNA (mtDNA) genetic information with GVPs, (see FIG. 24) allowing an increase in the power of discrimination to reach levels of individuality (>1 in 7 billion). In some instances, this requires the elucidation of statistical dependence patterns between each method, as understood by those skilled in the art. In particular, DNA STR typing and mtDNA analysis can result in partial or null profiles.


Example 30
Use of GVP Markers to Predict Biogeographic Background

It is expected that analysis of a diverse cohort will reveal markers that are informative of biogeographic background.


An exemplary method is illustrated by the schematic of FIG. 25. In particular in the illustration of FIG. 25, the panel in top left shows an exemplary DNA data sequence, TTGTTATCCGCTCACAATTCCACACAAC (SEQ ID NO:144), and the panel in top right shows exemplary proteomic data showing a graph reporting exemplary likelihood ratio of European/African markers (EUR/AFR), which together can provide biostatistics useful for predicting biogeographic background. The graph on the bottom of FIG. 25 shows an exemplary predictive model reporting % European DNA in relation to likelihood ratio (L).


Inclusion of informative markers in likelihood ratio (L) and the biostatistical analytical model will enable prediction of biogeographic origin from proteomic data. The use of GVP markers will be validated to predict biogeographic background.


Example 31
Validation of GVP Application in Forensic Contexts

It is expected that comparison of MS data from two different protein samples from one individual will demonstrate the validity of the approaches described herein. For example, it is expected that GVP alleles will be consistent between physiological locations (e.g. hair from head versus body), and that GVP profiles will remain consistent with age, and/or chemical and/or environmental exposure.


In particular, in a study to identify chemical markers in hair that are indicative of exposures to hair dye, exemplary results indicate surfactants comprise the majority of chemicals in hair care products (see FIG. 26). Other hair care compounds comprise emulsifiers, moisturizers, and detergents, whereas hair dye compounds are not very abundant in the samples.


Example 32
GVP Database Design

GVP databases can be designed based on the indications provided in the present disclosure comprising marker GVPs for biological organism, a biological organism type or an individual thereof as will be understood by a skilled person.


An exemplary GVP database design is shown in FIG. 27. The Entity relationship (ER) diagram shows types of data entities and the relationships between them. The Scheme allows flexibility by storing additional characteristics as tag-value pairs as will be understood by a skilled person


The above schematics can be implemented by developing a central database resource for GVP and SNP genotyping, comprising web-based queries and data entry, bulk loading of sequencing and LC/MS data, streamlined data access for analysis tools, implemented using Django, a Python-based framework for web/database application development in accordance with the illustration of FIG. 27.


Example 33
GVP Analysis Workflow in Bones

An exemplary GVP analysis workflow is shown in FIG. 28.


Example 34
Tooth Sex-Linked Protein Analysis Workflow

An exemplary tooth sex-linked protein analysis workflow is shown in FIG. 29.


In this example, both amelogenin isoforms were identified from modern and archaeological teeth samples.


Example 35
Fingerprint/Touch Derived Samples

Touch samples were collected from multiple surfaces, such as those comprising DNA-incompatible materials. Samples were extracted with techniques identifiable by a skilled person. Samples were analyzed for protein coverage (see FIG. 30). As shown in FIG. 30, protein coverage from touch samples is similar to that achieved with hair samples


Example 36
Tissue Procurement

Cranial hair shafts and buffy coat DNA were collected from a cohort of 60 self-identifying unrelated European—Americans (EA1, Sorenson Forensics LLC, Salt Lake City). Genomic DNA from each subject was screened using the Investigative LEAD™ Ancestry DNA Test (Sorenson Forensics LLC, Salt Lake City, Utah) and genotype data was generated for 190 SNPs that are ‘Ancestry Informative Markers’, which span all 22 autosomal chromosomes[49]. Nine individuals had measurable non-European admixture and were excluded from the analysis. An additional collection was conducted using cranial hair shaft and nuclear DNA from another cohort of self-identified unrelated European—Americans (EA2, n=15). All material was collected using protocols, informed consents, and questionnaires that were approved by the Institutional Review Boards at Utah Valley University (IRB #00642) and Lawrence Livermore National Laboratory (IRB #11-007). Hair shaft material was also collected from a cohort of five African-American and five Kenyan subjects[50]. Cranial hair shafts were additionally collected from six individuals from two separate archaeological assemblages excavated in London and Kent: three individuals (S1-S3), dating from circa 1750-1850, and three individuals (S4-S6) from a cemetery in active use 1821-1853.


Example 37
Proteomic Data Acquisition and Identification of Single Amino Acid Polymorphism-Containing Peptides

Hair from subjects was processed physically and biochemically and data was acquired as described. Briefly, hair was ground or milled; treated in a solution of urea, DTT, and detergent; alkylated; and then proteolyzed with trypsin. Resulting peptide mixtures were analyzed using tandem liquid chromatography mass spectrometry. The resulting proteomic datasets were converted to the Mascot generic format and analyzed using three different approaches: Mascot (software version 2.2.03, Matrix Science, Inc., Boston, Mass.), X!Tandem, using the GPM manager software (www.thegpm.org, release SLEDGEHAMMER (2013.09.01)), or X!Tandem using the Petunia Graphic User Interface (TANDEM CYCLONE TPP, download=2011.12.01.1—LabKey, Insilicos, ISB). A custom protein reference database was used (51 Methods; zenodo.org/record/58223: DOI: 10.5281/zenodo.58223) to ensure the identification of genetically variant peptides by both Mascot and the Petunia GUI peptide spectra matching algorithms[51]. Resulting peptide lists were screened for the presence of genetically variant peptides and identifications were collated for each subject. Inferences made through the use of GPM manager or the use of the customized reference database, in either X!Tandem or MASCOT, were compared for redundancy 0. The mass spectrometry proteomics data that has been submitted to the Global Proteome Machine (www.thegpm.org,) can be publicly accessed[52].


Example 38
Validation of Identified Genetically Variant Peptides

Identified candidate genetically variant peptides were filtered to reduce false-positive assignment using the following criteria for exclusion: low-quality expectation scores (X!Tandem, log(e)<−2; Mascot, expectation score >0.05), if the corresponding nsSNPs were distributed at less than 0.8% in the sample population (minor allelic frequency <0.4%), the presence of masses in a MS/MS fragmentation spectrum from a GVP consistent with the alternative allele, the incorporation of biological post-translational modifications in the assigned sequence (such as phosphorylation), and high variance between theoretical and observed primary masses (>0.2 Da). Amino acid polymorphisms assigned due to likely chemical modification or conversion were also excluded from the analysis (www.unimod.org)[53-55]. Rejected single amino acid polymorphisms include methionine to phenylalanine, asparagine to aspartate, glutamine to glutamate and cysteine to serine[53, 55, 56]. Peptides that were potentially derived from paralogous sequences, or that were potentially expressed in more than one gene product, were removed from the analysis. Inferred nsSNP loci were directly validated by Sanger sequencing of the subjects' nuclear DNA.


Example 39
Statistical Treatment of Individual Inferred nsSNP Profiles

An estimation of the probability of a given inferred nsSNP allele profile being detected in a sample population was calculated using a frequentist estimation of allele frequency, or frequency of an allele combination, within the reading frame of a gene (Pr(inferred nsSNP allele gene combinationipopulation)), and a Bayesian application of the product-rule[57, 58]. The occurrence of alleles, or allele combinations, was counted in European (n=379) and African (n=246) sample populations (www.1000genomes.org; Phase 1)[59]. The 1000 Genome Project sample populations were selected as sample populations because the African population did not have European admixture. The final probability of an individual SNP, or SNP combination, occurring within a gene reading frame, was estimated as (x+½)/(n+1), where x is the number of individuals with a given SNP, or combination of SNPs, in a sample population of size n[60]. The above expression represents the Bayesian posterior mean of a binomial probability using the Jeffreys Beta (½, ½) prior, which has the advantage of giving a non-zero estimate of the population probability even for x=0[60, 61]. Full independence between genes was assumed.


The effect of observed allele variation on the overall profile probability was estimated by parametric bootstrap resampling from a binomial (n, (x +½)/(n+1)) distribution for each gene, multiplying the resulting probability estimates across genes, and taking the 5th and 95th percentiles of the resampling distribution (90% CI)[61]. A comparison of the inferred nsSNP profile probability in the sample European and African population was calculated as a likelihood (L) ratio (L=Pr(profilelEUR population)/Pr(profilelAFR population))[57].


Example 40
Same Sample Mitochondrial/Proteomics GV Detection and Database Building

An exemplary method is described to perform a same sample mitochondrial/proteomics genetic variation detection and database building according to the following steps of the instant disclosure.


Preparing the Biological Sample

Applicable method to perform preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis, are identifiably by a skilled person upon reading of the instant disclosure


In an exemplary approach using preparation methods of the instant disclosure, single hair samples (1 inch; 25 mm) from separate individuals were carefully measured and cut into four equal pieces. The cut hair was then placed into separate Protein LoBind Eppendorf tubes. 100 μL of extraction buffer containing 0.05 M ammonium bicarbonate (ABC), 0.1 M dithiothreitol (DTT), 2% sodium dodecanoate (SDD) was added to each tube. Samples were then incubated at 70° C. in an ultrasonic water bath (Elma) while being ultrasonicated at high energy and frequency settings for 60 minutes or until hair was completely dissolved into solution. SDD was removed by extraction with acidified ethyl acetate (pH 2-3, 0.75% trifluoroacetic acid). After addition of 100 uL acidified ethyl acetate to each tube, samples were quickly vortexed, incubated at room temperature for 5 min, and centrifuged for 5 min at max speed (20,000×g). The upper organic phase was removed, discarded to waste, and the extraction process was repeated once. The remaining lower aqueous phase was then readjusted to pH 8 with ABC [13]. Alternative step includes cold acetone precipitation overnight and resuspension of protein pellet into 0.05M ABC; 0.1M DTT; and 1% protease max. Carbamidomethylation of free cysteines was performed by adding 6 μL of iodoacetamide (1.0 M) and incubation for 60 min in the dark at 25° C. To further solubilize proteins, 0.01% protease max (3 μL of 1.0% w/v) was added to each sample. Prior to proteolysis, the solubilized protein solution was concentrated to 50uL using 10 kD molecular weight spin concentrators (Millipore). Trypsin (2 μL of 0.5 μg/μL) was then added to each protein sample. Protein digestion was performed at 25° C. for 20/22 hours while being continuously agitated by magnetic-bar stirring. Protocol for isolation of DNA from tissues was provided by the Qiagen Q1Aamp® DNA Micro Kit. Manual suggestions were following with exception to the lysis procedural steps that include adding proteinase K, additional of proprietary buffer ‘ATL’, pulse-vortexing, overnight incubation at 56° C., and addition of proprietary buffer ‘AL’. Previous trypsin incubation was substituted for these steps. Following trypsin proteolysis, 100 uL of 100% ethanol was added to each sample as recommended by Qiagen Q1Aamp® DNA Micro Kit instructions. Removing this set and not adding ethanol also yields amplifiable mtDNA from sample. Samples were then vortexed for 15 seconds, incubated at 25° C. for 5 minutes, then added into separate QIAmp miniElute columns. Columns were closed and centrifuged at 6000×g for one minute. Flow-through was collected as the peptide fraction of the extraction, filtered using 0.1 μm PTFE filter, and transferred into fresh vials for mass spectrometric analysis (stored at +4.0 -−20° C.). Additional step of speed vacuum (20 minutes at 60° C.) can be used to concentrate peptide fraction of samples. The bound mtDNA fraction was then washed according to Qiagen Q1Aamp® DNA Micro Kit instructions and eluted twice into the same collection tube with 25 uL of warm (37° C.) water by centrifugation for one minute (20,000×g).


Fractionating the Processed Biological Sample

Applicable method to perform fractionating the processed biological sample to obtain solubilized protein fraction and a solubilized DNA fraction can also be identified by a skilled person.


In particular a solubilized protein fraction comprising the solubilized proteins from the sample can be obtained by the following exemplary SDD extraction and protein concentration procedure step which includes cold acetone precipitation (−4° C.) overnight and resuspension of protein pellet into 0.05M ABC; 0.1M DTT; and 1% protease max. Additional step of speed vacuum (20 minutes at 60° C.) can be used to concentrate peptide fraction of samples subsequent to proteolysis step.


A solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample can be provided with the following exemplary method. Following trypsin proteolysis, 100 uL of 100% ethanol was added to each sample as recommended by Qiagen Q1Aamp® DNA Micro Kit instructions. Removing this set and not adding ethanol also yields amplifiable mtDNA from sample.


Detecting a Genetic Protein Variation in the Solubilized Protein Fraction

Applicable methods to perform detecting a genetic protein variation in the solubilized protein fraction from the sample by performing the proteomic analysis of the solubilized protein fraction are identifiable by a skilled person. in an exemplary method MS/MS data acquisition of peptide sequences was performed using Thermo Scientific Q Exactive Plus Hybrid Quadrupole-Orbitrap mass spectrometer fitted with Easy-nLC 1000 HPLC (Thermo Scientific, Asheville, N.C., USA). Peptides were separated by reversed-phase liquid chromatography using a mobile phase A (0.01% TFA in water) and mobile phase B (0.01% TFA in acetonitrile) in a 97 minute gradient. 2 of each sample were injected onto a C18 trap cartridge and preceded by an Easy-Spray™ nanoflow (1 mm×150 mm) column (Thermo Scientific, Asheville, N.C., USA) with a flow rate of 3 μL/min. Electrospray ionization was achieved in positive mode with a voltage of 2-4 kV. Dynamic exclusion data collection was implemented at a MS scan range of 180-1,800 m/z, top 10 precursor ions were chosen for subsequent MS/MS scans and excluded after 10 seconds.


Single-hair method implements program PEAKS 7.5 (Bioinformatics Solutions Inc., Waterloo, Ontario, Canada) for variant peptide detection. PEAKs software was used to search each RAW data file to determine the specific peptides that were identified in each sample. A reference database created by translating polymorphisms (missense SNPs, insertions, deletions, and stops/gains) that influence protein sequences observed in exome results into mutated protein sequences is used for peptide identification within software parameters. Experimental conditions and instrumental capabilities inform parameters chosen for search. Search settings include partial posttranslational modifications including oxidation of methionine, deamidation of asparagine and glutamine, and carbamidomethylation of cysteine. Precursor mass error of 30 ppm using monoisotopic mass was used for parent ion identifications and a 0.05 Da for fragment ions masses. A decoy database was generated within the software using a protein library of all human protein sequences exported from UniProtKB/Swiss-Prot knowledgebase (The UniProt Consortium; www.uniprot.org/). The decoy database is used to determine the false determination rate (FDR) of protein identifications. Protein identifications (IDs) were filtered by a 1% FDR. Data output from PEAKs searches including identified peptides, quality measures, and protein sequence position is then filtered for peptides containing predicted mutations using in-house text mining scripts.


Detecting a Genomic Variation in the DNA Fraction

Applicable method to perform detecting a genomic variation of the nuclear and/or mitochondrial genome by performing a genetic analysis of the solubilized DNA fraction; including methods to detect mitochondrial DNA variation or STR variation are identifiable by a skilled person, in an exemplary method to amplify mitochondrial control regions, PCR amplification was carried out with the following set of primers: F15975 and R16410m for HV1, F015 and R389 for HV2, F403 and R635 for HV3 in 50 ul reaction volumes with Q5 Hot Start High-Fidelity 2× Master Mix (New England Biolabs, Inc, Ipswich, Mass., USA), containing 0.2 uM each forward and reverse primers and 5 ul genomic DNA. Amplification was carried out on a PTC-200 DNA Engine (MJ Research, Waltham, MA, USA) under the following conditions: 98° C. for 2 min; 15 cycles of 98° C. for 10 s, 56° C. for 30 s, 72° C. for 30 s; 25 cycles of 98° C. for 20 s, 56° C. for 30 s, 72° C. for 30 s+10 s/cycle; and a final extension at 72° C. for 2 min. PCR amplicons were gel purified on a 2.0% agarose gel using QlAquick Gel Extraction Kit (Qiagen Inc, Germantown, Md., USA) according to the manufacturer's instructions with the exception the DNA was eluted with 35 ul EB Buffer. Purified PCR amplicons were visualized via gel electrophoresis on 2.0% agarose and quantified using QuBit 2.0 Fluorometer (Thermo Fisher Scientific, Waltham, Mass., USA). DNA sequencing was performed using a Big Dye Terminator v3.1 Cycle Sequencing Kit (Thermo Fisher Scientific, Waltham, Mass., USA) with the following cycling conditions: 96° C. for 1 min; 30 cycles of 96° C. for 10 s, 50° C. for 5 s, 60° C. for 2 min. Sequencing reactions were analyzed on an ABI 3500 Genetic Analyzer (Applied Biosystems). Primers used for sequencing were the appropriate primers used during amplification. The results were analyzed and de novo assembled using Geneious R9.1.8 (Biomatters Ltd, Auckland, NZ). To ensure sequence data quality, each genomic DNA was amplified and sequenced in duplicate.


mtDNA variants were detected by alignment using Clustal multiple sequence alignment tool [62, 63]. mtDNA mutation database MitoMaster [63] was used in addition to confirm prior record of the observed mutations.


Combining the Detected Genetic Protein Variations and the Detected Genomic Variation to Provide the Marker Genetic Variation

Applicable methods to perform combining the detected genetic protein variations and the detected genomic variation to provide the marker genetic variation database system of the biological sample, are identifiable by a skilled person. in an exemplary method Mutant genotypic frequencies available in mtDNA mutation database MitoMaster (Brandon 2009) and Ensembl [26] (www.ensembl.org/index.html)corresponding to the observed genetic variations in both peptides and mtDNA hyper-variable control regions were combined by calculating random match probabilities for each individual.


Comparing the Detected Genetic Protein Variation and/or the Detected Genomic Variation with a Marker Genetic Protein Variation and/or of a Marker Genomic Variation


Applicable methods to perform comparing the detected genetic protein variation and/or the detected genomic variation with a marker genetic protein variation and/or of a marker genomic variation respectively from the marker genetic variation database system are identifiable by a skilled person.


Exemplary methods include a range of possibilities from simply taking the two comparisons as independent verification of identity match or exclusion between samples or it could include a combined statistical model that taken into account the appropriate statistical metrics (e.g. random match probability) of both the proteomic marker(s) and the genetic marker(s) to give an overall greater statistical measure.


Example 41
GVP Analysis for a Sample Tissue

An example GVP analysis for a sample tissue can be broken down into the following parts, as shown in FIG. 31 and generally described as:

    • Part 0: Define a “tissue”—some set of genes to target
    • Part 1: Extract information of interest from Exome files and annotate under GRCh38
    • Part 2: Extract information of interest from annotated VCF, down select to preferred mutations, add supporting information
    • Part 3: Mutate protein sequences and create FASTA files suitable for use with PEAKs
    • Part 4: From PEAKs result, find “hits” peptides that carry programed-for mutations


Part 5: Analyze “hits” Process steps 1-3 describe the data analysis process that is used to extract relevant genetic information from exome data and relating those to detectable proteins, thereby identifying genetic markers for potential detectable GVPs. Those process steps can be used to provide a proteomically detectable genomic variation in a set of represented genes proteomically detectable in the biological sample of the individual.


Applicable methods to perform providing a set of represented genes proteomically detectable in the biological sample of the individual, are identifiable by a skilled person upon reading of the instant disclosure, wherein the represented genes correspond to the proteomically detected proteins in the biological sample of the individual.


In an exemplary approach, for a single-hair approach herein described implements program PEAKS 7.5 (Bioinformatics Solutions Inc., Waterloo, Ontario, Canada) for variant peptide detection. A reference database created by translating polymorphisms (missense SNPs, insertions, deletions, and stops/gains) that influence protein sequences observed in exome results into mutated protein sequences are used for peptide identification within software parameters. Search settings include partial posttranslational modifications including oxidation of methionine, deamidation of asparagine and glutamine, and carbamidomethylation of cysteine. Precursor mass error of 30 ppm using monoisotopic mass was used for parent ion identifications and a 0.05 Da for fragment ions masses. Additionally, software program PEAKS 7.5 (Bioinformatics Solutions Inc., Waterloo, Ontario, Canada) protein identification software can be used to identify putative peptide variants using a specific capability called Spider [40] without using mutated reference databases. Another approach, outlined in [3] uses the Global Proteome Machine webserver (GPM; www.thegpm.org) to detect possible peptide variants.


In particular, process step 2 described the process to extract information of interest from exome results, down select to preferred mutations, add supporting information. This particular step filters the exome data to down select for proteins that we know we can see proteomically. This step can be used to perform selecting from the identified genetic variation, a genetic variation detectable in the sample of the biological organism.


Process step 4 describes the process for identifying peptides in proteomic data output from raw MS datafile analysis (e.g. using PEAKS, GPM or other commercial proteomic search tool) that contain mutations predicted by the exome data analysis performed in steps 1-2 (iiid above). This step can be used to perform providing the marker genetic protein variation validated by providing a proteomically detectable genetic protein variation corresponding to the proteomically detectable genomic variation in the biological sample of the individual.


Process step 5 describes combining results of hits identified in step 4 above, applying filters (e.g. peptide is only coded for by the identified gene). Results in a summary file that provides a pooled set of GVPs for a plurality of individuals. This step can be used to perform providing a number of proteomic datasets of individuals of the plurality of individuals, the number statistically significant for the plurality of individuals, including how to determine a statistically significant number of datasets.


Process step 5 also describes combining results of hits identified in step 4 above, applying filters (e.g. peptide is only coded for by the identified gene). Results in a summary file that provides a pooled set of GVPs for a plurality of individuals and includes information on commonality, allele frequency and any additional genetic or statistical information required. This step can be used to identify identifying a protein common to the provided number of proteomic datasets; including threshold and ranges of percentage of commonality of observed proteins.


Process step 5 further describes combining results of hits identified in 4 above, applying filters (e.g. peptide is only coded for by the identified gene). Results in a summary file that provides a pooled set of GVPs for a plurality of individuals and includes information on commonality, allele frequency and any additional genetic or statistical information required. This step can also be used to perform selecting from the identified protein common to the provided proteomic datasets, a protein detectable in the sample of the individuals of the plurality of individuals


PART 0: Define a “Tissue”—Some Set of Genes to Target

The tissue file (e.g. Tissue.txt) can be created by picking genes that appear frequently in a set of MS files as taken for a range of samples of a given tissue type (e.g. skin300g.txt, hair691g.txt, skhr838g.txt).


An example tissue file content is shown in Table 5. The required fields in this example are the standard gene symbol and CHR (“standard gene symbol” has an entrezID number, as in hg19 or hg38).









TABLE 5







Example tissue.txt



















PA


PA
ENSG
Symbol
CHR
entrezID
Descr.
freq
















Q9Y277
ENSG00000078668
VDAC3
8
7419
Voltage-dependent
11







anion channel 3


P63167
ENSG00000088986
DYNLL1
12
8655
Dynein light chain
11







LC8-type 1


Q9P0M6
ENSG00000099284
H2AFY2
10
55506
H2A histone family
11







member Y2










PART 1: Extract Information of Interest from Exome Files and Annotate Under GRCh38


Read-in list of target genes—tissue.txt.


Read-in VCF file—fname.svindeLvar.vcfgz (gzipped version).


Read meta data to confirm genomic coordinates (expecting 37d.5): e.g. VCF file L2_0051 reference is: hs37d5.fa .


Create TAB IX if none exist—fname.svindeLvar.vcfgz.tbi .


Subset VCF to target genes.


Extract all mutations in the subset VCF, clean-up formatting and data types. Carry through exome quality metrics for each entry .


Remove entries with filter of LowQual or “.” (i.e. “to poor to call”). (See Table 6—note VQSR ranking).











TABLE 6






Freq
Freq


Filter
(L L2_51, hr691g)
(L4_01BUC, skhr838g)

















99.90 to 100.00
44
102


INDEL
37
33


INDEL, LowQual
27
40


LowQual
266
263


Pass
3178
5398









Drop cases where ALT is a coma-delimited list. (See FIG. 39).


“Lift over” genomic coordinates from GRCh37 to GRCh38 (This example uses GRCh38.10).


Error check to confirm all SNPs collected conform to GRCh38, drop any deviants.


Summarize: L2_0051 / hr691g—921 unique mutations processed.


Translate each surviving mutation into HGVS notation per varnomen.hgvs.org .


Write (no row names, no column names, one entry per row)—fname_tissue_hgvs.txt.


ZZ


18:g.46098362_46098363insACCCCC


18:g.63499047_63499048insTATATA


17:g.82081885_82081942de1


8:g.143729168C>G


2:g.131218699G>C


21:g.44627940C>T


Write companion file with linkage information for each mutation—fname_tissue_link.txt. The link file carries CHR, START, END, and rsID, which are not used beyond this point in the pipeline. (See FIG. 37).


Submit fname_tissue_hgvs.txt to ensembl Variant Effects Predictor (VEP) for GRCh38


PART 2: Extract Information of Interest from Annotated VCF, Down Select to Preferred Mutations, Add Supporting Information


For the mutations submitted as L2_0051_hr691g_hgvs.txt , ensembl VEP replies are as shown in FIG. 32. (See www.ensembl.org/Homo_sapiens/Tools/VEP).


Recover the annotation results from VEP fname_tissue_annt.txt. The VEP annotation might contain all available G1000 and ExAC. AFs, SIFT, and Polyphen scores can be added. Note: as of Aug. 24, 2017 G1000 remains, but ExAC is replaced by gnomAD.


Read-in annotations. An example is shown in FIG. 33. (See www.ensembLorg/info/genome/variation/predicted_data.html).


Down-select to:

    • 1) BIOTYPE=Protein_Coding
    • 2) Consequence=frameshift, stop_gained_frameshift, inframe_deletion, inframe_insertion, missense, synonymous, missense_splice, splice_synonymous, stop_gained, stop_gained, splice, start_lost, protein_altering.


From bioMart add: Swissprot PA number (if not, then trEMBL PA number), APPRIS rank, ensembl external_transcript_name. Where an rsID is not returned, use a shortened version of HGVS call as the mutation “name” under dbSNP. Carry through G1000, G1000_EUR, ExAC, ExAC_NFE (as of Aug. 24, 2017, carry through gnomAD_AF and gnomAD_NFE_AF).


Read in link file fname_tissue_link.txt, add-on related REF, ALT, GATK and exome quality metrics. (see FIG. 36).


Summarize:

    • 749 unique mutations by HGVS, 287 genes involved
    • 2063 total mutations in all transcripts
    • fraction 0.1177896 with GQ <max in L2_0051 (see Table 7)












TABLE 7







Effect
Freq



















Frameshift
13



inframe_deletion
17



inframe_insertion
15



Missense
711



missense, splice
18



splice, synonymous
24



start_lost
2



stop_gained
3



stop_gained, splice
2



synonymous
1257












    • out of 749 unique mutations:
      • 43 returned with no ExAC_NFE_MAF
      • 42 returned with no ExAC_MAF
      • 3 returned with ExAC_NFE_MAF=0
      • 0 returned with ExAC_MAF=0





Write mutations to target—fname_tissue_extract.txt. Note:*extract.txt created to support workflows where *extract.txt from different exomes are combined into a *predicted*extract.txt. For example: Combine L4_0001 (P1) and L4_0002 (P2) to predict the child (L4_0003) as L4_0003_T1_p12xc_tissue_extract.txt for Triad1 parents predict child where child's exome is L4_0003.


PART 3: Mutate Protein Sequences and Create FASTA Files Suitable for Use with PEAKs


Assumptions applied in GEN I mutations code:

    • Apply all mutations to one AA sequence (relaxed in GEN II code)
    • A frameshift is the end of useful information—treat the start of a frameshift as a stop-gained
    • Treat two or three SNPs per codon as happening in the same strand and use the combination mutation
    • Process all viable transcripts within a gene (use location and consequence per transcript), do not know which may be expressed.
    • “Base” protein sequence library in PEAKs is UniprotKB Swissprot (without isoforms)


PEAKs identifies a transcript by and passes-through the AccessionlEntry_Name portion of the FASTA header:

    • Uniprot SwissProt header for KRT86
      • >sp|O43790|KRT86_HUMAN Keratin, type II cuticular Hb6 OS=Homo sapiens









GN = KRT 86 PE = 1 SV = 1


(SEQ ID NO: 145)


MTCGSYCGGRAFSCISACGPRPGRCCITAAPYRGISCYRGLTGGFGSHSV


CGGFRAGSCGRSFGYRSGGVCGPSPPCITTV......










      • The PA is 043790 (Uniprot is distinguished by: PA without appended -nnn and the Entry_Name carries “_species”). Uniprot Entry_Name may not be a standard gene name (e.g. for KRTAP1-1 Uniprot uses Entry_Name=KTP11).



    • From the GEN I mutations code
      • Entry_Name=standard gene name with “m” appended to indicate a mutated entry
        • Accession=PA-ensembl_transcript_name (to differentiate between different transcripts. PA alone is not enough: a given transcript can have multiple PA. A given PA can refer to transcripts in multiple genes.)
        • Within a gene, do not include duplicated mutated transcripts.
        • For mutated: Replace “description” with a list of mutations applied in transcript reference coordinates, append “h” for heterozygous
        • ALL locations remain in transcripts reference coordinates, subsequent codes/analysis must unwind as needed

    • GEN I header for a first transcript of KRT86—ENST00000293525
      • >sp|Q43790-201|KRT86m A321V 5del152 H560Kh OS=Homo sapiens GN=KRT86 PE=1 SV=1
      • >sp|Q43790-201|KRT86 dummy comment OS=Homo sapiens GN=KRT86 PE=1 SV=1

    • GEN I header for a second transcript of KRT86—ENST00000423955
      • >sp|Q43790-202|KRT86m A319V 5de1152 H556Kh OS=Homo sapiens GN=KRT86 PE=1 SV=1
      • >sp|Q43790-2021KRT86 dummy comment OS=Homo sapiens GN=KRT86 PE=1 SV=1





Read in mutations to target—fname_tissue_extract.txt.


Convert all frameshifts to SNPs as X/* (“*” indicates a stop and “X” indicates a wild-card in the AA sequence) .


Detect multiple SNPs/codon events and compute change from combination, update mutations list.


Subset to genes that are mutated (i.e. drop genes that carry only synonymous mutations).


From bioMart upload AA sequences for all transcripts that may be called on. Within a gene: de-duplicate for transcripts that have identical AA sequences.Drop any transcript that carries an X (i.e. a wild-card AA).


Process the AA sequence for each transcript remaining. Apply stops (stop-gained and frameshifts) and trim AA sequence to length. Apply remaining SNPs that are in-range. Apply INDELs that are in range, process from tail-to-snout (as INDELs will accordion the sequence).


Generate FASTA headers for mutant and reference sequences.


Write: mutated AA sequences in FASTA format—fname_tissue_mutant_fasta.txt—and reference AA sequences in FASTA format—fname_tissue_ref_fasta.txt.


Submit for PEAKs analysis: use combination of “Base” protein list and mutant/ref FASTA.


PART 4: From PEAKs Result, Find Peptides that Carry Programed—For Mutations


Read-in PEAKs output (fname_tissueprotein-peptides.csv) and down-select to columns of interest (see FIG. 34).


Extract peptide sequence (remove PTMs and any lead/tail AA) e.g. R.TSC(+57.02)SSRPC(+57.02)V.P becomes TSCSSRPCV (SEQ ID NO. 127).


Separate PA and symbol, replace any UniprotKB Entry_Name with the standard gene name (e.g. replace KTP11 with KRTAP1-1).


Down select to those Protein Groups that carry a called-unique peptide assigned to a mutated transcript, and where the Peptide Group contains only the one mutated gene (may be a combination of mutated, reference and base transcripts from the one gene). Meaning of Unique in PEAKs output: The peptide (sans PTM) was detected uniquely within the present analysis. Such a called-unique peptide can be assigned to more than one transcript and/or gene. Each called-unique peptide is assigned to one Protein Group. There may be more than one called-unique peptide in a Protein Group. There may be more the one gene in a Protein Group. Filter by gene since Uniprot Entry_Name may not be a valid gene symbol for purposes of this example.


Read-in mutated FASTAfname_tissue_mutant_fasta.txt. Within each transcript (mutant FASTA entry) and for the selected Protein Groups:

    • Unwind mutations into transcript coordinates (snout-to-tail to account for action of any INDELs) and
    • Find those peptides that contain a programmed-for mutation.


Read-in fname_tissue_extract.txt . For “hits” (i.e. a programmed-for mutation found in a called-unique peptide) update entry with information about the mutation (e.g. dbSNP, AF' s, etc.).


Write out documented hits (group, peptide w/wo PTMs, MS meta data, mutation, AFs, GATK . . . )—fname_tissue_resu.txt.


PART 5: Hits Analysis

Read in hits results across a sample family—L4_0001_hair691g_resu.txt through L4_0063_hair691g_resu.txt (say)


Determine which peptides that carry the hits are unique within some test protein set:

    • Write a list of peptides (sans PTM) L4_peptide_summary.txt,
    • submit list of peptides to BLASTp or some other in-house- or web-tool to search for matches within the test protein set,
    • test protein set—UniprotKB(Swissprot+isoforms and trEMBL)_HUMAN (about 172,164 protein sequences), and
    • recover the results and indicate those peptides that are “no match” (i.e. the mutated peptide is not found in the test protein set).


Write—L4_resu_summary.txt (symbol, dbSNP, peptide sans PTM, no match, MS meta data, mutation meta data, AFs, GATK, file tag . . . )


Write—L4_resu_exec_summary.txt (symbol, dbSNP, no match in dnSNP, AFs, all file tags carrying this mutation) (see FIG. 35).


Supporting Tools

Create a tissue set

    • read in a collection of PEAKs files
    • convert Accession and Entry-Name to a proper symbol name
    • tabulate frequency of occurrence of symbols through the file set
    • Use bioMart to validate and add other information
    • Output to tissue_number_genes.txt : symbol, entrezID, ENSG, gene description and frequency of observation


Retention time analysis/prediction

    • read in a PEAKs file
    • Apply Gilar's peptide retention model, treat PTMs as different AAs, to provide
    • a multi-parameter linear regression model
    • an optimized multi-parameter SVM model
    • identify substantial outliers as possibly mis-identified in the MS/PEAKs analysis
    • test for applicability against other PEAKs files


Exclusion list

    • read in a collection of PEAKs files
    • through the whole set collect the M/Z for all called-unique entries
    • through the whole set collect the M/Z for all not-called-unique entries where the peak area is greater than some cut-off
    • Round-off all carried M/Z (as given to 4 places) to 2 places
    • Select those not-called-unique with M/Z that do not compete with the called-unique M/Z
    • Output a table with two columns: a name (e.g. X100000#) and the selected M/Z to 2 places.


Example 42
GVP Analysis for a Sample Tissue

An example GVP analysis for a sample tissue can also be broken down into the following parts, generally described as:

    • Part 0: Define a “tissue”—some set of genes to target
    • Part 1: Extract information of interest from Exome files, possibly phase the exome using a computational tool (e.g. WhatsHAP) or method (e.g. pedigree phasing) or combination thereof as is known in the arts, and annotate under GRCh38
    • Part 2: Extract information of interest from annotated VCF, down select to preferred mutations, add supporting information (e.g. allele and/or population frequencies from a data base such as gnomAD)
    • Part 3: Mutate protein sequences and create FASTA files suitable for use with PEAKs
    • Part 4: From PEAKs result, find “hits” peptides that carry programed-for mutations
    • Part 5: Analyze “hits” to determine if reference hits are unique (i.e. related to one genomic location within a defined set of transcripts associated with a species e.g. ensembl human) and if mutated hits are novel (i.e. not found within a defined set of transcripts associated with a species e.g. ensembl human).


Parts 1 to 5 of the present example can be performed with methods similar to the ones indicated in Example 41 modified in view of the indications provided in the present example as will be understood by a skilled person upon reading of the present disclosure.


Example 43
Exemplary Genes Comprising Marker Exome Sequences Validated in Hair Type Samples

An exemplary set of genes that can be used in methods and systems herein described as well as in related databases is reported herein. In particular, the exemplary set of genes comprises genes validated as proteomically detectable in hair samples of Homo Sapiens which can be used in methods and systems to detect a genetic variation and/or perform a genetic variation analysis where the biological organism is a human being, as well as in related databases, in accordance with the various aspects of the present disclosure.


Specifically, Table 8 shows a list of exemplary genes that appear in MS files taken for samples of a hair of a human being. The fields in this example indicate the preference (X=more preferred), the standard gene symbol (gene symbol), the chromosome where the gene is located (chr), a description of the gene (gene description) and the gene identifier in the database Ensembl at the date of filing of the instant disclosure (Ensembl Gene Identifier).


The exemplary genes of Table 8 can therefore be used in methods and systems of the disclosure wherein the sample comprises an hair sample from human beings,









TABLE 8







Exemplary genes identified in mass spectrometric analysis from hair type samples











X = more



Ensembl gene


preferable
gene symbol
chr
gene description
identifier















VDAC3
8
voltage dependent anion channel 3
ENSG00000078668



DYNLL1
12
dynein light chain LC8-type 1
ENSG00000088986



H2AFY2
10
H2A histone family member Y2
ENSG00000099284



SNU13
22
SNU13 homolog, small nuclear
ENSG00000100138





ribonucleoprotein (U4/U6.U5)



AHCY
20
adenosylhomocysteinase
ENSG00000101444



FBL
19
fibrillarin
ENSG00000105202



MYL12B
18
myosin light chain 12B
ENSG00000118680



EPHX2
8
epoxide hydrolase 2
ENSG00000120915



RPS10
6
ribosomal protein S10
ENSG00000124614



BMP2
20
bone morphogenetic protein 2
ENSG00000125845



SNRPN
15
small nuclear ribonucleoprotein polypeptide N
ENSG00000128739



AFDN
6
afadin, adherens junction formation factor
ENSG00000130396



PRPH
12
peripherin
ENSG00000135406



COX5B
2
cytochrome c oxidase subunit 5B
ENSG00000135940



ACTR2
2
ARP2 actin related protein 2 homolog
ENSG00000138071



CSTB
21
cystatin B
ENSG00000160213



HIST1H2AA
6
histone cluster 1 H2A family member a
ENSG00000164508



KLK6
19
kallikrein related peptidase 6
ENSG00000167755



DYNLRB2
16
dynein light chain roadblock-type 2
ENSG00000168589



RAB1B
11
RAB1B, member RAS oncogene family
ENSG00000174903



GBA
1
glucosylceramidase beta
ENSG00000177628



RCC1
1
regulator of chromosome condensation 1
ENSG00000180198



RUVBL2
19
RuvB like AAA ATPase 2
ENSG00000183207



TMED9
5
transmembrane p24 trafficking protein 9
ENSG00000184840



KRT77
12
keratin 77
ENSG00000189182



ANXA4
2
annexin A4
ENSG00000196975



FAM49A
2
family with sequence similarity 49 member A
ENSG00000197872



KRTAP4-1
17
keratin associated protein 4-1
ENSG00000198443



PRR9
1
proline rich 9
ENSG00000203783



FIS1
7
fission, mitochondrial 1
ENSG00000214253



KRTAP10-9
21
keratin associated protein 10-9
ENSG00000221837



KRTAP10-10
21
keratin associated protein 10-10
ENSG00000221859



ARPC4
3
actin related protein 2/3 complex subunit 4
ENSG00000241553



EIF6
20
eukaryotic translation initiation factor 6
ENSG00000242372



EIF5AL1
10
eukaryotic translation initiation factor 5A-like 1
ENSG00000253626



RNASET2
6
ribonuclease T2
ENSG00000026297



ALDH3A2
17
aldehyde dehydrogenase 3 family member A2
ENSG00000072210



EIF3I
1
eukaryotic translation initiation factor 3 subunit
ENSG00000084623





I



HNRNPC
14
heterogeneous nuclear ribonucleoprotein C
ENSG00000092199





(C1/C2)



CRAT
9
carnitine O-acetyltransferase
ENSG00000095321



NUTF2
16
nuclear transport factor 2
ENSG00000102898



ECH1
19
enoyl-CoA hydratase 1
ENSG00000104823



ENDOU
12
endonuclease, poly(U) specific
ENSG00000111405



KHDRBS1
1
KH RNA binding domain containing, signal
ENSG00000121774





transduction associated 1



DYNLRB1
20
dynein light chain roadblock-type 1
ENSG00000125971



NDUFA2
5
NADH:ubiquinone oxidoreductase subunit A2
ENSG00000131495



EDEM1
3
ER degradation enhancing alpha-mannosidase
ENSG00000134109





like protein 1



NARS
18
asparaginyl-tRNA synthetase
ENSG00000134440



RPS6
9
ribosomal protein S6
ENSG00000137154



HNRNPA1L2
13
heterogeneous nuclear ribonucleoprotein A1-
ENSG00000139675





like 2



PKLR
1
pyruvate kinase, liver and RBC
ENSG00000143627



ARL8A
1
ADP ribosylation factor like GTPase 8A
ENSG00000143862



ZNF462
9
zinc finger protein 462
ENSG00000148143



PRSS53
16
protease, serine 53
ENSG00000151006



CXADR
21
coxsackie virus and adenovirus receptor
ENSG00000154639



CBR1
21
carbonyl reductase 1
ENSG00000159228



PSMB4
1
proteasome subunit beta 4
ENSG00000159377



C21orf33
21
chromosome 21 open reading frame 33
ENSG00000160221



PGAM2
7
phosphoglycerate mutase 2
ENSG00000164708



LMAN2
5
lectin, mannose binding 2
ENSG00000169223



GNB2
7
G protein subunit beta 2
ENSG00000172354



MYL6B
12
myosin light chain 6B
ENSG00000196465



PSAP
10
prosaposin
ENSG00000197746



DDX39B
6
DExD-box helicase 39B
ENSG00000198563



RACK1
5
receptor for activated C kinase 1
ENSG00000204628



TUBB8
10
tubulin beta 8 class VIII
ENSG00000261456



RPS10-NUDT3
6
RPS10-NUDT3 readthrough
ENSG00000270800



PRSS3
9
protease, serine 3
ENSG00000010438



SARS
1
seryl-tRNA synthetase
ENSG00000031698



PSMC5
17
proteasome 26S subunit, ATPase 5
ENSG00000087191



HNRNPM
19
heterogeneous nuclear ribonucleoprotein M
ENSG00000099783



PABPC1L
20
poly(A) binding protein cytoplasmic 1 like
ENSG00000101104



PGRMC1
X
progesterone receptor membrane component 1
ENSG00000101856



NUP93
16
nucleoporin 93
ENSG00000102900



GPRC5D
12
G protein-coupled receptor class C group 5
ENSG00000111291





member D



PTK7
6
protein tyrosine kinase 7 (inactive)
ENSG00000112655



GLO1
6
glyoxalase I
ENSG00000124767



RPL23
17
ribosomal protein L23
ENSG00000125691



TUBB2B
6
tubulin beta 2B class IIb
ENSG00000137285



PPP2R1B
11
protein phosphatase 2 scaffold subunit Abeta
ENSG00000137713



SLC40A1
2
solute carrier family 40 member 1
ENSG00000138449



ARHGDIA
17
Rho GDP dissociation inhibitor alpha
ENSG00000141522



RPS11
19
ribosomal protein S11
ENSG00000142534



RPL7A
9
ribosomal protein L7a
ENSG00000148303



RPS3
11
ribosomal protein S3
ENSG00000149273



DBI
2
diazepam binding inhibitor, acyl-CoA binding
ENSG00000155368





protein



PDCD6IP
3
programmed cell death 6 interacting protein
ENSG00000170248



YOD1
1
YOD1 deubiquitinase
ENSG00000180667



SHMT2
12
serine hydroxymethyltransferase 2
ENSG00000182199



NDUFA13
19
NADH:ubiquinone oxidoreductase subunit A13
ENSG00000186010



HIST1H1T
6
histone cluster 1 H1 family member t
ENSG00000187475



PCBP2
12
poly(rC) binding protein 2
ENSG00000197111



SIRPA
20
signal regulatory protein alpha
ENSG00000198053



RNF39
6
ring finger protein 39
ENSG00000204618



CTC-260F20.3
19

ENSG00000258674



KRTAP10-7
21
keratin associated protein 10-7
ENSG00000272804



CH507-9B2.4
21

ENSG00000276612



CH507-9B2.3
21

ENSG00000280071



ARSF
X
arylsulfatase F
ENSG00000062096



GNB1
1
G protein subunit beta 1
ENSG00000078369



KHSRP
19
KH-type splicing regulatory protein
ENSG00000088247



RPLP0
12
ribosomal protein lateral stalk subunit P0
ENSG00000089157



PABPC4
1
poly(A) binding protein cytoplasmic 4
ENSG00000090621



EZR
6
ezrin
ENSG00000092820



AP1B1
22
adaptor related protein complex 1 beta 1
ENSG00000100280





subunit



PSMC6
14
proteasome 26S subunit, ATPase 6
ENSG00000100519



PSMD7
16
proteasome 26S subunit, non-ATPase 7
ENSGOOOOO1O3O35



MYH14
19
myosin heavy chain 14
ENSG00000105357



PSMA1
11
proteasome subunit alpha 1
ENSG00000129084



FBP2
9
fructose-bisphosphatase 2
ENSG00000130957



TPT1
13
tumor protein, translationally-controlled 1
ENSGOOOOO133112



ATIC
2
5-aminoimidazole-4-carboxamide
ENSG00000138363





ribonucleotide formyltransferase/IMP





cyclohydrolase



RPS2
16
ribosomal protein S2
ENSG00000140988



CSNK1D
17
casein kinase 1 delta
ENSG00000141551



SH3BGRL3
1
SH3 domain binding glutamate rich protein like
ENSG00000142669





3



SPINT1
15
serine peptidase inhibitor, Kunitz type 1
ENSG00000166145



PGK2
6
phosphoglycerate kinase 2
ENSG00000170950



KRT27
17
keratin 27
ENSG00000171446



EIF2S3L
12
Putative eukaryotic translation initiation factor
ENSG00000180574





2 subunit 3-like protein



CAPN12
19
calpain 12
ENSG00000182472



KRT73
12
keratin 73
ENSG00000186049



PTRH1
9
peptidyl-tRNA hydrolase 1 homolog
ENSG00000187024



KRTAP10-6
21
keratin associated protein 10-6
ENSG00000188155



XRCC6
22
X-ray repair cross complementing 6
ENSG00000196419



DYNC1H1
14
dynein cytoplasmic 1 heavy chain 1
ENSG00000197102



SERPINB13
18
serpin family B member 13
ENSG00000197641



RPL10A
6
ribosomal protein L10a
ENSG00000198755



ASPRV1
2
aspartic peptidase, retroviral-like 1
ENSG00000244617



RP1-5O6.7
22
Casein kinase I isoform epsilon
ENSG00000283900



CAPG
2
capping actin protein, gelsolin like
ENSG00000042493



TUBA3D
2
tubulin alpha 3d
ENSG00000075886



BCORL1
X
BCL6 corepressor-like 1
ENSG00000085185



FH
1
fumarate hydratase
ENSG00000091483



ACOT7
1
acyl-CoA thioesterase 7
ENSG00000097021



SRSF3
6
serine and arginine rich splicing factor 3
ENSG00000112081



TRIM25
17
tripartite motif containing 25
ENSG00000121060



PSMF1
20
proteasome inhibitor subunit 1
ENSG00000125818



ASS1
9
argininosuccinate synthase 1
ENSG00000130707



EIF5A
17
eukaryotic translation initiation factor 5A
ENSG00000132507



EPRS
1
glutamyl-prolyl-tRNA synthetase
ENSG00000136628



GRHPR
9
glyoxylate and hydroxypyruvate reductase
ENSG00000137106



WARS
14
tryptophanyl-tRNA synthetase
ENSG00000140105



UQCRC2
16
ubiquinol-cytochrome c reductase core protein
ENSG00000140740





II



RPL11
1
ribosomal protein L11
ENSG00000142676



PSMA5
1
proteasome subunit alpha 5
ENSG00000143106



RPS3A
4
ribosomal protein S3A
ENSG00000145425



RPS14
5
ribosomal protein S14
ENSG00000164587



TPSAB1
16
tryptase alpha/beta 1
ENSG00000172236



DES
2
desmin
ENSG00000175084



IDH2
15
isocitrate dehydrogenase (NADP(+)) 2,
ENSG00000182054





mitochondrial



TPSB2
16
tryptase beta 2 (gene/pseudogene)
ENSG00000197253



TUBA3C
13
tubulin alpha 3c
ENSG00000198033



UBA52
19
ubiquitin A-52 residue ribosomal protein fusion
ENSG00000221983





product 1



TOLLIP
11
toll interacting protein
ENSG00000078902



ERMP1
9
endoplasmic reticulum metallopeptidase 1
ENSG00000099219



ABCD1
X
ATP binding cassette subfamily D member 1
ENSG00000101986



PPP2CB
8
protein phosphatase 2 catalytic subunit beta
ENSG00000104695



MTCH2
11
mitochondrial carrier 2
ENSG00000109919



PPP2CA
5
protein phosphatase 2 catalytic subunit alpha
ENSG00000113575



STX12
1
syntaxin 12
ENSG00000117758



LAMTOR5
1
late endosomal/lysosomal adaptor, MAPK and
ENSG00000134248





MTOR activator 5



CKAP4
12
cytoskeleton associated protein 4
ENSG00000136026



RPS8
1
ribosomal protein S8
ENSG00000142937



COX6C
8
cytochrome c oxidase subunit 6C
ENSG00000164919



TPP1
11
tripeptidyl peptidase 1
ENSG00000166340



RPS21
20
ribosomal protein S21
ENSG00000171858



HECTD4
12
HECT domain E3 ubiquitin protein ligase 4
ENSG00000173064



PSMD2
3
proteasome 26S subunit, non-ATPase 2
ENSG00000175166



TALDO1
11
transaldolase 1
ENSG00000177156



PDE4DIP
1
phosphodiesterase 4D interacting protein
ENSG00000178104



TUBA8
22
tubulin alpha 8
ENSG00000183785



HIST2H2AB
1
histone cluster 2 H2A family member b
ENSG00000184270



TACSTD2
1
tumor-associated calcium signal transducer 2
ENSG00000184292



EIF3CL
16
eukaryotic translation initiation factor 3 subunit
ENSG00000205609





C-like



RP11-295K3.1
11

ENSG00000250644



ATP6V0A1
17
ATPase H+ transporting V0 subunit a1
ENSG00000033627



RPL18
19
ribosomal protein L18
ENSG00000063177



WNT3
17
Wnt family member 3
ENSG00000108379



PRDX4
X
peroxiredoxin 4
ENSG00000123131



KIAA0368
9
KIAA0368
ENSG00000136813



ATP6V1G1
9
ATPase H+ transporting V1 subunit G1
ENSG00000136888



KRT71
12
keratin 71
ENSG00000139648



EIF4A3
17
eukaryotic translation initiation factor 4A3
ENSG00000141543



RBMX
X
RNA binding motif protein, X-linked
ENSG00000147274



H2AFZ
4
H2A histone family member Z
ENSG00000164032



CTSB
8
cathepsin B
ENSG00000164733



PDHB
3
pyruvate dehydrogenase (lipoamide) beta
ENSG00000168291



GLTPD2
17
glycolipid transfer protein domain containing 2
ENSG00000182327



KRTAP9-8
17
keratin associated protein 9-8
ENSG00000187272



APRT
16
adenine phosphoribosyltransferase
ENSG00000198931



RPS18
6
ribosomal protein S18
ENSG00000231500



HAGH
16
hydroxyacylglutathione hydrolase
ENSG00000063854



ME1
6
malic enzyme 1
ENSG00000065833



TUBB4A
19
tubulin beta 4A class IVa
ENSG00000104833



GAPDHS
19
glyceraldehyde-3-phosphate dehydrogenase,
ENSG00000105679





spermatogenic



HIP1R
12
huntingtin interacting protein 1 related
ENSG00000130787



RPL8
8
ribosomal protein L8
ENSG00000161016



DCD
12
dermcidin
ENSG00000161634



HSP90B1
12
heat shock protein 90 beta family member 1
ENSG00000166598



PA2G4
12
proliferation-associated 2G4
ENSG00000170515



IMPDH2
3
inosine monophosphate dehydrogenase 2
ENSG00000178035



FAHD1
16
fumarylacetoacetate hydrolase domain
ENSG00000180185





containing 1



EIF3C
16
eukaryotic translation initiation factor 3 subunit
ENSG00000184110





C



H2AFX
11
H2A histone family member X
ENSG00000188486



AP2A1
19
adaptor related protein complex 2 alpha 1
ENSG00000196961





subunit



KRT25
17
keratin 25
ENSG00000204897



NAV3
12
neuron navigator 3
ENSG00000067798



RTCB
22
RNA 2′,3′-cyclic phosphate and 5′-OH ligase
ENSG00000100220



H2AFV
7
H2A histone family member V
ENSG00000105968



EIF3A
10
eukaryotic translation initiation factor 3 subunit
ENSG00000107581





A



METAP2
12
methionyl aminopeptidase 2
ENSG00000111142



RTN4
2
reticulon 4
ENSG00000115310



EFHD1
2
EF-hand domain family member D1
ENSG00000115468



ATP6V1B1
2
ATPase H+ transporting V1 subunit B1
ENSG00000116039



YPEL5
2
yippee like 5
ENSG00000119801



PCMT1
6
protein-L-isoaspartate (D-aspartate) O-
ENSG00000120265





methyltransferase



ACLY
17
ATP citrate lyase
ENSG00000131473



RAN
12
RAN, member RAS oncogene family
ENSG00000132341



HNRNPD
4
heterogeneous nuclear ribonucleoprotein D
ENSG00000138668



PSMB6
17
proteasome subunit beta 6
ENSG00000142507



RPL7
8
ribosomal protein L7
ENSG00000147604



KRT24
17
keratin 24
ENSG00000167916



CHTF8
16
chromosome transmission fidelity factor 8
ENSG00000168802



CAPZA2
7
capping actin protein of muscle Z-line alpha
ENSG00000198898





subunit 2



AK2
1
adenylate kinase 2
ENSG00000004455



RPS20
8
ribosomal protein S20
ENSG00000008988



PITHD1
1
PITH domain containing 1
ENSG00000057757



RPL6
12
ribosomal protein L6
ENSG00000089009



MLF2
12
myeloid leukemia factor 2
ENSG00000089693



DNAJB6
7
DnaJ heat shock protein family (Hsp40)
ENSG00000105993





member B6



AJUBA
14
ajuba LIM protein
ENSG00000129474



ATP6V1E1
22
ATPase H+ transporting V1 subunit E1
ENSG00000131100



COX4I1
16
cytochrome c oxidase subunit 411
ENSG00000131143



TXN
9
thioredoxin
ENSG00000136810



NONO
X
non-POU domain containing, octamer-binding
ENSG00000147140



ATP5H
17
ATP synthase, H+ transporting, mitochondrial
ENSG00000167863





Fo complex subunit D



HIST3H3
1
histone cluster 3 H3
ENSG00000168148



ATP5I
4
ATP synthase, H+ transporting, mitochondrial
ENSG00000169020





Fo complex subunit E



KRT9
17
keratin 9
ENSG00000171403



NCCRP1
19
non-specific cytotoxic cell receptor protein 1
ENSG00000188505





homolog (zebrafish)



POTEJ
2
POTE ankyrin domain family member J
ENSG00000222038



AP000304.12
21

ENSG00000249209



SRI
7
sorcin
ENSG00000075142



ETFB
19
electron transfer flavoprotein beta subunit
ENSG00000105379



ACTA2
10
actin, alpha 2, smooth muscle, aorta
ENSG00000107796



DLST
14
dihydrolipoamide S-succinyltransferase
ENSG00000119689



RTN3
11
reticulon 3
ENSGOOOOO133318



SPINK5
5
serine peptidase inhibitor, Kazal type 5
ENSG00000133710



RAC1
7
ras-related C3 botulinum toxin substrate 1 (rho
ENSG00000136238





family, small GTP binding protein Rac1)



ACTG2
2
actin, gamma 2, smooth muscle, enteric
ENSG00000163017



RPN1
3
ribophorin I
ENSG00000163902



CFL1
11
cofilin 1
ENSG00000172757



GDI1
X
GDP dissociation inhibitor 1
ENSG00000203879



KRTAP10-11
21
keratin associated protein 10-11
ENSG00000243489



HSP90AB1
6
heat shock protein 90 alpha family class B
ENSG00000096384





member 1



ENO2
12
enolase 2
ENSG00000111674



LYPLA1
8
lysophospholipase I
ENSG00000120992



ECHS1
10
enoyl-CoA hydratase, short chain 1
ENSG00000127884



CHAC1
15
ChaC glutathione specific gamma-
ENSG00000128965





glutamylcyclotransferase 1



IL1F10
2
interleukin 1 family member 10 (theta)
ENSG00000136697



PADI1
1
peptidyl arginine deiminase 1
ENSG00000142623



CALM2
2
calmodulin 2
ENSG00000143933



CALM3
19
calmodulin 3
ENSG00000160014



S100A9
1
S100 calcium binding protein A9
ENSG00000163220



TUBB6
18
tubulin beta 6 class V
ENSG00000176014



CALM1
14
calmodulin 1
ENSG00000198668



RPS16
19
ribosomal protein S16
ENSG00000105193



TYRP1
9
tyrosinase related protein 1
ENSG00000107165



CAPZA1
1
capping actin protein of muscle Z-line alpha
ENSG00000116489





subunit 1



RPL13
16
ribosomal protein L13
ENSG00000167526



HINT1
5
histidine triad nucleotide binding protein 1
ENSG00000169567



SDR16C5
8
short chain dehydrogenase/reductase family
ENSG00000170786





16C member 5



S100A16
1
S100 calcium binding protein A16
ENSG00000188643



PHB2
12
prohibitin 2
ENSG00000215021



ACTN1
14
actinin alpha 1
ENSG00000072110



FSCN1
7
fascin actin-bundling protein 1
ENSG00000075618



MYL6
12
myosin light chain 6
ENSG00000092841



PFN1
17
profilin 1
ENSG00000108518



CPEB4
5
cytoplasmic poly adenylation element binding
ENSG00000113742





protein 4



ACTN4
19
actinin alpha 4
ENSG00000130402



EIF2S3
X
eukaryotic translation initiation factor 2 subunit
ENSG00000130741





gamma



NECTIN4
1
nectin cell adhesion molecule 4
ENSG00000143217



ACAA2
18
acetyl-CoA acyltransferase 2
ENSG00000167315



SEC24C
10
SEC24 homolog C, COPII coat complex
ENSG00000176986





component



FCHSD1
5
FCH and double SH3 domains 1
ENSG00000197948



S100A6
1
S100 calcium binding protein A6
ENSG00000197956



CTNND1
11
catenin delta 1
ENSG00000198561



CTNNA2
2
catenin alpha 2
ENSG00000066032



ENO3
17
enolase 3
ENSG00000108515



IMMT
2
inner membrane mitochondrial protein
ENSG00000132305



EIF2S1
14
eukaryotic translation initiation factor 2 subunit
ENSG00000134001





alpha



PABPC3
13
poly(A) binding protein cytoplasmic 3
ENSG00000151846



G6PD
X
glucose-6-phosphate dehydrogenase
ENSG00000160211



KRT4
12
keratin 4
ENSG00000170477



RPL12
9
ribosomal protein L12
ENSG00000197958



PRSS1
7
protease, serine 1
ENSG00000204983



EPPK1
8
epiplakin 1
ENSG00000261150



ATP2B4
1
ATPase plasma membrane Ca2+ transporting 4
ENSG00000058668



CDC42
1
cell division cycle 42
ENSG00000070831



CAPZB
1
capping actin protein of muscle Z-line beta
ENSG00000077549





subunit



CSNK1A1
5
casein kinase 1 alpha 1
ENSG00000113712



GOT1
10
glutamic-oxaloacetic transaminase 1
ENSG00000120053



PLB1
2
phospholipase B1
ENSG00000163803



METAP1
4
methionyl aminopeptidase 1
ENSG00000164024



SLC3A2
11
solute carrier family 3 member 2
ENSG00000168003



CSNK1E
22
casein kinase 1 epsilon
ENSG00000213923



PEBP1
12
phosphatidylethanolamine binding protein 1
ENSG00000089220



EEF1A2
20
eukaryotic translation elongation factor 1 alpha
ENSG00000101210





2



ILVBL
19
ilvB acetolactate synthase like
ENSG00000105135



KPNB1
17
karyopherin subunit beta 1
ENSG00000108424



PPIB
15
peptidylprolyl isomerase B
ENSG00000166794



KRT28
17
keratin 28
ENSG00000173908



KRTAP6-1
21
keratin associated protein 6-1
ENSG00000184724



RPS4X
X
ribosomal protein S4, X-linked
ENSG00000198034



MT-CO2
MT
mitochondrially encoded cytochrome c oxidase
ENSG00000198712





II



VCL
10
vinculin
ENSG00000035403



DLD
7
dihydrolipoamide dehydrogenase
ENSG00000091140



DDTL
22
D-dopachrome tautomerase-like
ENSG00000099974



TUBB1
20
tubulin beta 1 class VI
ENSG00000101162



CPT1A
11
carnitine palmitoyltransferase 1A
ENSG00000110090



PGLS
19
6-phosphogluconolactonase
ENSG00000130313



HADHB
2
hydroxyacyl-CoA dehydrogenase/3-ketoacyl-
ENSG00000138029





CoA thiolase/enoyl-CoA hydratase





(trifunctional protein), beta subunit



PPA2
4
pyrophosphatase (inorganic) 2
ENSG00000138777



TMED10
14
transmembrane p24 trafficking protein 10
ENSG00000170348



KRT72
12
keratin 72
ENSG00000170486



HIST1H2BL
6
histone cluster 1 H2B family member 1
ENSG00000185130



KRTAP10-3
21
keratin associated protein 10-3
ENSG00000212935



PPP1CB
2
protein phosphatase 1 catalytic subunit beta
ENSG00000213639



ACPP
3
acid phosphatase, prostate
ENSG00000014257



RNH1
11
ribonuclease/angiogenin inhibitor 1
ENSG00000023191



SUN2
22
Sad1 and UNC84 domain containing 2
ENSG00000100242



CEP250
20
centrosomal protein 250
ENSG00000126001



DSG3
18
desmoglein 3
ENSG00000134757



HIST1H2BA
6
histone cluster 1 H2B family member a
ENSG00000146047



GJA1
6
gap junction protein alpha 1
ENSG00000152661



ATP5O
21
ATP synthase, H+ transporting, mitochondrial
ENSG00000241837





F1 complex, O subunit



DDT
22
D-dopachrome tautomerase
ENSG00000099977



TARS
5
threonyl-tRNA synthetase
ENSG00000113407



CLTC
17
clathrin heavy chain
ENSG00000141367



ACOX1
17
acyl-CoA oxidase 1
ENSG00000161533



KRT6C
12
keratin 6C
ENSG00000170465



NIPSNAP1
22
nipsnap homolog 1
ENSG00000184117



POTEI
2
POTE ankyrin domain family member I
ENSG00000196834



RP4-777O23.3
7

ENSG00000281039



SLC25A5
X
solute carrier family 25 member 5
ENSG00000005022



PABPC1
8
poly(A) binding protein cytoplasmic 1
ENSG00000070756



CELSR1
22
cadherin EGF LAG seven-pass G-type receptor
ENSG00000075275





1



HNRNPH2
X
heterogeneous nuclear ribonucleoprotein H2
ENSG00000126945



CSRP1
1
cysteine and glycine rich protein 1
ENSG00000159176



FBP1
9
fructose-bisphosphatase 1
ENSG00000165140



UQCRFS1
19
ubiquinol-cytochrome c reductase, Rieske iron-
ENSG00000169021





sulfur polypeptide 1



HIST2H2AC
1
histone cluster 2 H2A family member c
ENSG00000184260



P4HB
17
prolyl 4-hydroxylase subunit beta
ENSG00000185624



HIST1H2AD
6
histone cluster 1 H2A family member d
ENSG00000196866



VDAC1
5
voltage dependent anion channel 1
ENSG00000213585



NME1
17
NME/NM23 nucleoside diphosphate kinase 1
ENSG00000239672



HSPE1-MOB4
2
HSPE1-MOB4 readthrough
ENSG00000270757



ACADVL
17
acyl-CoA dehydrogenase, very long chain
ENSG00000072778



PROCR
20
protein C receptor
ENSG00000101000



C1QBP
17
complement C1q binding protein
ENSG00000108561



CTSD
11
cathepsin D
ENSG00000117984



LDHA
11
lactate dehydrogenase A
ENSG00000134333



EIF4A2
3
eukaryotic translation initiation factor 4A2
ENSG00000156976



ENGASE
17
endo-beta-N-acetylglucosaminidase
ENSG00000167280



KRT19
17
keratin 19
ENSG00000171345



TUFM
16
Tu translation elongation factor, mitochondrial
ENSG00000178952



HIST3H2A
1
histone cluster 3 H2A
ENSG00000181218



KRTAP4-16
17
keratin associated protein 4-16
ENSG00000241241



TUBB3
16
tubulin beta 3 class III
ENSG00000258947



COMT
22
catechol-O-methyltransferase
ENSG00000093010



ATP5D
19
ATP synthase, H+ transporting, mitochondrial
ENSG00000099624





F1 complex, delta subunit



KRT17
17
keratin 17
ENSG00000128422



RPS27A
2
ribosomal protein S27a
ENSG00000143947



PDIA3
15
protein disulfide isomerase family A member 3
ENSG00000167004



HSPA6
1
heat shock protein family A (Hsp70) member 6
ENSG00000173110



ALYREF
17
Aly/REF export factor
ENSG00000183684



HIST1H2AE
6
histone cluster 1 H2A family member e
ENSG00000277075



HIST1H2AB
6
histone cluster 1 H2A family member b
ENSG00000278463



ATOX1
5
antioxidant 1 copper chaperone
ENSG00000177556



GGCT
7
gamma-glutamylcyclotransferase
ENSG00000006625



RAB7A
3
RAB7A, member RAS oncogene family
ENSG00000075785



CUX2
12
cut like homeobox 2
ENSG00000111249



CAT
11
catalase
ENSG00000121691



LMNB2
19
lamin B2
ENSG00000176619



HIST3H2BB
1
histone cluster 3 H2B family member b
ENSG00000196890



KRTAP26-1
21
keratin associated protein 26-1
ENSG00000197683



NME2
17
NME/NM23 nucleoside diphosphate kinase 2
ENSG00000243678



GPI
19
glucose-6-phosphate isomerase
ENSG00000105220



GIPC1
19
GIPC PDZ domain containing family member 1
ENSG00000123159



MAP7
6
microtubule associated protein 7
ENSG00000135525



ACTA1
1
actin, alpha 1, skeletal muscle
ENSG00000143632



HK1
10
hexokinase 1
ENSG00000156515



ACTC1
15
actin, alpha, cardiac muscle 1
ENSG00000159251



TUBA1C
12
tubulin alpha 1c
ENSG00000167553



HNRNPH1
5
heterogeneous nuclear ribonucleoprotein H1
ENSG00000169045



HSPA1L
6
heat shock protein family A (Hsp70) member 1
ENSG00000204390





like


X
SLC25A3
12
solute carrier family 25 member 3
ENSG00000075415


X
HSP90AA1
14
heat shock protein 90 alpha family class A
ENSG00000080824





member 1


X
GARS
7
glycyl-tRNA synthetase
ENSG00000106105


X
KRT18
12
keratin 18
ENSG00000111057


X
TAGLN2
1
transgelin 2
ENSG00000158710


X
PCBP1
2
poly(rC) binding protein 1
ENSG00000169564


X
CYCS
7
cytochrome c, somatic
ENSG00000172115


X
KRTAP19-5
21
keratin associated protein 19-5
ENSG00000186977


X
CDH1
16
cadherin 1
ENSG00000039068


X
PARK7
1
Parkinsonism associated deglycase
ENSG00000116288


X
HNRNPA3
2
heterogeneous nuclear ribonucleoprotein A3
ENSG00000170144


X
SERPINB5
18
serpin family B member 5
ENSG00000206075


X
H2AFJ
12
H2A histone family member J
ENSG00000246705


X
UQCRC1
3
ubiquinol-cytochrome c reductase core protein I
ENSG00000010256


X
PHGDH
1
phosphoglycerate dehydrogenase
ENSG00000092621


X
ECHDC1
6
ethylmalonyl-CoA decarboxylase 1
ENSG00000093144


X
PRDX1
1
peroxiredoxin 1
ENSG00000117450


X
GOT2
16
glutamic-oxaloacetic transaminase 2
ENSG00000125166


X
TKT
3
transketolase
ENSG00000163931


X
TUBA1A
12
tubulin alpha 1a
ENSG00000167552


X
KRT15
17
keratin 15
ENSG00000171346


X
UQCRH
1
ubiquinol-cytochrome c reductase hinge protein
ENSG00000173660


X
RPLP2
11
ribosomal protein lateral stalk subunit P2
ENSG00000177600


X
KRT76
12
keratin 76
ENSG00000185069


X
KRT3
12
keratin 3
ENSG00000186442


X
NME1-NME2
17
NME1-NME2 readthrough
ENSG00000011052


X
GRN
17
granulin precursor
ENSG00000030582


X
SSBP1
7
single stranded DNA binding protein 1
ENSG00000106028


X
HNRNPA2B1
7
heterogeneous nuclear ribonucleoprotein A2/B1
ENSG00000122566


X
ENDOD1
11
endonuclease domain containing 1
ENSG00000149218


X
ALDOA
16
aldolase, fructose-bisphosphate A
ENSG00000149925


X
GSDMA
17
gasdermin A
ENSG00000167914


X
KRT2
12
keratin 2
ENSG00000172867


X
HIST2H3PS2
1
histone cluster 2 H3 pseudogene 2
ENSG00000203818


X
AHNAK
11
AHNAK nucleoprotein
ENSG00000124942


X
ARL8B
3
ADP ribosylation factor like GTPase 8B
ENSG00000134108


X
ATP6V1B2
8
ATPase H+ transporting V1 subunit B2
ENSG00000147416


X
TCHH
1
trichohyalin
ENSG00000159450


X
HIST1H2AJ
6
histone cluster 1 H2A family member j
ENSG00000276368


X
GDI2
10
GDP dissociation inhibitor 2
ENSG00000057608


X
HIST1H2BJ
6
histone cluster 1 H2B family member j
ENSG00000124635


X
GFAP
17
glial fibrillary acidic protein
ENSG00000131095


X
PMEL
12
premelanosome protein
ENSG00000185664


X
KRTAP10-12
21
keratin associated protein 10-12
ENSG00000189169


X
S100A14
1
S100 calcium binding protein A14
ENSG00000189334


X
KRTAP4-3
17
keratin associated protein 4-3
ENSG00000196156


X
YWHAH
22
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000128245





monooxygenase activation protein eta


X
PDIA6
2
protein disulfide isomerase family A member 6
ENSG00000143870


X
FABP5
8
fatty acid binding protein 5
ENSG00000164687


X
HEPHL1
11
hephaestin like 1
ENSGOOOOO181333


X
CRIP2
14
cysteine rich protein 2
ENSG00000182809


X
KRT14
17
keratin 14
ENSG00000186847


X
APOD
3
apolipoprotein D
ENSG00000189058


X
H1F0
22
H1 histone family member 0
ENSG00000189060


X
HSPA1B
6
heat shock protein family A (Hsp70) member
ENSG00000204388





1B


X
HSPA1A
6
heat shock protein family A (Hsp70) member
ENSG00000204389





1A


X
RBM14
11
RNA binding motif protein 14
ENSG00000239306


X
KRTAP7-1
21
keratin associated protein 7-1
ENSG00000274749





(gene/pseudogene)


X
VIM
10
vimentin
ENSG00000026025


X
CTNNA1
5
catenin alpha 1
ENSG00000044115


X
SFPQ
1
splicing factor proline and glutamine rich
ENSG00000116560


X
COX5A
15
cytochrome c oxidase subunit 5A
ENSG00000178741


X
RP11-566K11.2
16

ENSG00000198211


X
HSPA9
5
heat shock protein family A (Hsp70) member 9
ENSG00000113013


X
HSPE1
2
heat shock protein family E (Hsp10) member 1
ENSG00000115541


X
ANXA1
9
annexin A1
ENSG00000135046


X
MEMO1
2
mediator of cell motility 1
ENSG00000162959


X
KRT78
12
keratin 78
ENSG00000170423


X
CALML5
10
calmodulin like 5
ENSG00000178372


X
KRT6B
12
keratin 6B
ENSG00000185479


X
BLMH
17
bleomycin hydrolase
ENSG00000108578


X
HIST1H3J
6
histone cluster 1 H3 family member j
ENSG00000197153


X
HIST1H3D
6
histone cluster 1 H3 family member d
ENSG00000197409


X
HIST2H2BF
1
histone cluster 2 H2B family member f
ENSG00000203814


X
HIST1H3G
6
histone cluster 1 H3 family member g
ENSG00000273983


X
HIST1H3B
6
histone cluster 1 H3 family member b
ENSG00000274267


X
HIST1H3E
6
histone cluster 1 H3 family member e
ENSG00000274750


X
HIST1H3I
6
histone cluster 1 H3 family member i
ENSG00000275379


X
HIST1H3A
6
histone cluster 1 H3 family member a
ENSG00000275714


X
HIST1H3F
6
histone cluster 1 H3 family member f
ENSG00000277775


X
HIST1H3C
6
histone cluster 1 H3 family member c
ENSG00000278272


X
HIST1H3H
6
histone cluster 1 H3 family member h
ENSG00000278828


X
HIST1H1D
6
histone cluster 1 H1 family member d
ENSG00000124575


X
KRT16
17
keratin 16
ENSG00000186832


X
TUBA4A
2
tubulin alpha 4a
ENSG00000127824


X
RIDA
8
reactive intermediate imine deaminase A
ENSG00000132541





homolog


X
HSD17B4
5
hydroxysteroid 17-beta dehydrogenase 4
ENSG00000133835


X
DSG1
18
desmoglein 1
ENSG00000134760


X
CLIC3
9
chloride intracellular channel 3
ENSG00000169583


X
FAM83H
8
family with sequence similarity 83 member H
ENSG00000180921


X
HIST2H3D
1
histone cluster 2 H3 family member d
ENSG00000183598


X
TUBB
6
tubulin beta class I
ENSG00000196230


X
KRTAP4-6
17
keratin associated protein 4-6
ENSG00000198090


X
TXNRD1
12
thioredoxin reductase 1
ENSG00000198431


X
HIST2H3C
1
histone cluster 2 H3 family member c
ENSG00000203811


X
HIST2H3A
1
histone cluster 2 H3 family member a
ENSG00000203852


X
EEF1G
11
eukaryotic translation elongation factor 1
ENSG00000254772





gamma


X
LGALS1
22
galectin 1
ENSG00000100097


X
ACTBL2
5
actin, beta like 2
ENSG00000169067


X
FABP4
8
fatty acid binding protein 4
ENSG00000170323


X
PGAM1
10
phosphoglycerate mutase 1
ENSG00000171314


X
POTEE
2
POTE ankyrin domain family member E
ENSG00000188219


X
KRT6A
12
keratin 6A
ENSG00000205420


X
KRTAP4-12
17
keratin associated protein 4-12
ENSG00000213416


X
HIST1H2BB
6
histone cluster 1 H2B family member b
ENSG00000276410


X
HEXB
5
hexosaminidase subunit beta
ENSG00000049860


X
PLD3
19
phospholipase D family member 3
ENSG00000105223


X
ALDH2
12
aldehyde dehydrogenase 2 family
ENSG00000111275





(mitochondrial)


X
LMNB1
5
lamin B1
ENSG00000113368


X
HNRNPA1
12
heterogeneous nuclear ribonucleoprotein A1
ENSG00000135486


X
VCP
9
valosin containing protein
ENSG00000165280


X
PRDX2
19
peroxiredoxin 2
ENSG00000167815


X
FASN
17
fatty acid synthase
ENSG00000169710


X
KRT10
17
keratin 10
ENSG00000186395


X
HIST1H2BK
6
histone cluster 1 H2B family member k
ENSG00000197903


X
KRTAP4-5
17
keratin associated protein 4-5
ENSG00000198271


X
TGM1
14
transglutaminase 1
ENSG00000092295


X
AIM1
6
absent in melanoma 1
ENSG00000112297


X
H2AFY
5
H2A histone family member Y
ENSG00000113648


X
HIST1H1C
6
histone cluster 1 H1 family member c
ENSG00000187837


X
KRTAP2-2
17
keratin associated protein 2-2
ENSG00000214518


X
PKP1
1
plakophilin 1
ENSG00000081277


X
PGK1
X
phosphoglycerate kinase 1
ENSG00000102144


X
KRT20
17
keratin 20
ENSG00000171431


X
KRT79
12
keratin 79
ENSG00000185640


X
HIST1H2BH
6
histone cluster 1 H2B family member h
ENSG00000275713


X
TTBK2
15
tau tubulin kinase 2
ENSG00000128881


X
SOD1
21
superoxide dismutase 1
ENSG00000142168


X
HIST1H2BD
6
histone cluster 1 H2B family member d
ENSG00000158373


X
YWHAG
7
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000170027





monooxygenase activation protein gamma


X
PLEC
8
plectin
ENSG00000178209


X
ATG9B
7
autophagy related 9B
ENSG00000181652


X
LAMP1
13
lysosomal associated membrane protein 1
ENSG00000185896


X
HIST2H2AA3
1
histone cluster 2 H2A family member a3
ENSG00000203812


X
KRTAP4-11
17
keratin associated protein 4-11
ENSG00000212721


X
HIST2H2AA4
1
histone cluster 2 H2A family member a4
ENSG00000272196


X
HADHA
2
hydroxyacyl-CoA dehydrogenase/3-ketoacyl-
ENSG00000084754





CoA thiolase/enoyl-CoA hydratase





(trifunctional protein), alpha subunit


X
CRYAB
11
crystallin alpha B
ENSG00000109846


X
KRT8
12
keratin 8
ENSG00000170421


X
KRTAP16-1
17
keratin associated protein 16-1
ENSG00000212657


X
HIST1H2BN
6
histone cluster 1 H2B family member n
ENSG00000233822


X
HIST1H2BO
6
histone cluster 1 H2B family member o
ENSG00000274641


X
CS
12
citrate synthase
ENSG00000062485


X
ATP6V1A
3
ATPase H+ transporting V1 subunit A
ENSG00000114573


X
TUBA1B
12
tubulin alpha 1b
ENSG00000123416


X
YWHAQ
2
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000134308





monooxygenase activation protein theta


X
EIF4A1
17
eukaryotic translation initiation factor 4A1
ENSG00000161960


X
PHB
17
prohibitin
ENSG00000167085


X
HIST1H2BC
6
histone cluster 1 H2B family member c
ENSG00000180596


X
KRTAP4-9
17
keratin associated protein 4-9
ENSG00000212722


X
HIST1H2BM
6
histone cluster 1 H2B family member m
ENSG00000273703


X
HIST1H2BG
6
histone cluster 1 H2B family member g
ENSG00000273802


X
HIST1H2BE
6
histone cluster 1 H2B family member e
ENSG00000274290


X
HIST1H2BF
6
histone cluster 1 H2B family member f
ENSG00000277224


X
HIST1H2BI
6
histone cluster 1 H2B family member i
ENSG00000278588


X
HSPA5
9
heat shock protein family A (Hsp70) member 5
ENSG00000044574


X
ACAA1
3
acetyl-CoA acyltransferase 1
ENSG00000060971


X
KRT23
17
keratin 23
ENSG00000108244


X
PRDX6
1
peroxiredoxin 6
ENSG00000117592


X
HSPD1
2
heat shock protein family D (Hsp60) member 1
ENSG00000144381


X
RPSA
3
ribosomal protein SA
ENSG00000168028


X
LYG2
2
lysozyme g2
ENSG00000185674


X
PLCD1
3
phospholipase C delta 1
ENSG00000187091


X
KRTAP9-9
17
keratin associated protein 9-9
ENSG00000198083


X
KRTAP4-8
17
keratin associated protein 4-8
ENSG00000204880


X
GSTP1
11
glutathione S-transferase pi 1
ENSG00000084207


X
LDHB
12
lactate dehydrogenase B
ENSG00000111716


X
GPNMB
7
glycoprotein nmb
ENSG00000136235


X
YWHAB
20
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000166913





monooxygenase activation protein beta


X
TUBB4B
9
tubulin beta 4B class IVb
ENSG00000188229


X
HSD17B10
X
hydroxysteroid 17-beta dehydrogenase 10
ENSG00000072506


X
KRT1
12
keratin 1
ENSG00000167768


X
KRTAP4-4
17
keratin associated protein 4-4
ENSG00000171396


X
LRRC15
3
leucine rich repeat containing 15
ENSG00000172061


X
HIST2H2BE
1
histone cluster 2 H2B family member e
ENSG00000184678


X
KRT5
12
keratin 5
ENSG00000186081


X
POTEF
2
POTE ankyrin domain family member F
ENSG00000196604


X
KRTAP9-6
17
keratin associated protein 9-6
ENSG00000212659


X
KRTAP2-1
17
keratin associated protein 2-1
ENSG00000212725


X
KRTAP4-2
17
keratin associated protein 4-2
ENSG00000244537


X
HIST1H2AH
6
histone cluster 1 H2A family member h
ENSG00000274997


X
H3F3B
17
H3 histone family member 3B
ENSG00000132475


X
H3F3A
1
H3 histone family member 3A
ENSG00000163041


X
S100A3
1
S100 calcium binding protein A3
ENSGOOOOO188O15


X
PPIA
7
peptidylprolyl isomerase A
ENSG00000196262


X
HIST1H2AI
6
histone cluster 1 H2A family member i
ENSG00000196747


X
HIST1H2AG
6
histone cluster 1 H2A family member g
ENSG00000196787


X
KRTAP2-3
17
keratin associated protein 2-3
ENSG00000212724


X
KRTAP2-4
17
keratin associated protein 2-4
ENSG00000213417


X
KRTAP9-4
17
keratin associated protein 9-4
ENSG00000241595


X
LY6G6D
6
lymphocyte antigen 6 family member G6D
ENSG00000244355


X
HIST1H2AK
6
histone cluster 1 H2A family member k
ENSG00000275221


X
HIST1H2AL
6
histone cluster 1 H2A family member l
ENSG00000276903


X
HIST1H2AM
6
histone cluster 1 H2A family member m
ENSG00000278677


X
YWHAE
17
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000108953





monooxygenase activation protein epsilon


X
PADI3
1
peptidyl arginine deiminase 3
ENSG00000142619


X
HIST1H1E
6
histone cluster 1 H1 family member e
ENSG00000168298


X
KRTAP9-1
17
keratin associated protein 9-1
ENSG00000240542


X
DUSP14
17
dual specificity phosphatase 14
ENSG00000276023


X
NEU2
2
neuraminidase 2
ENSG00000115488


X
DSC3
18
desmocollin 3
ENSG00000134762


X
LMNA
1
lamin A/C
ENSG00000160789


X
YWHAZ
8
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000164924





monooxygenase activation protein zeta


X
KRTAP9-7
17
keratin associated protein 9-7
ENSG00000180386


X
HIST1H2AC
6
histone cluster 1 H2A family member c
ENSG00000180573


X
ANXA2
15
annexin A2
ENSG00000182718


X
KRTAP9-2
17
keratin associated protein 9-2
ENSG00000239886


X
ACTB
7
actin beta
ENSG00000075624


X
KRT7
12
keratin 7
ENSG00000135480


X
CTNNB1
3
catenin beta 1
ENSG00000168036


X
HIST1H1B
6
histone cluster 1 H1 family member b
ENSG00000184357


X
KRTAP13-1
21
keratin associated protein 13-1
ENSG00000198390


X
ENO1
1
enolase 1
ENSG00000074800


X
HSPA8
11
heat shock protein family A (Hsp70) member 8
ENSG00000109971


X
TUBB2A
6
tubulin beta 2A class IIa
ENSG00000137267


X
EEF1A1
6
eukaryotic translation elongation factor 1 alpha
ENSG00000156508





1


X
KRT80
12
keratin 80
ENSG00000167767


X
GDPD3
16
glycerophosphodiester phosphodiesterase
ENSG00000102886





domain containing 3


X
TPI1
12
triosephosphate isomerase 1
ENSG00000111669


X
PPL
16
periplakin
ENSG00000118898


X
FAM26D
6
family with sequence similarity 26 member D
ENSG00000164451


X
VDAC2
10
voltage dependent anion channel 2
ENSG00000165637


X
KRT75
12
keratin 75
ENSG00000170454


X
PKM
15
pyruvate kinase, muscle
ENSG00000067225


X
KRT37
17
keratin 37
ENSG00000108417


X
KRTAP1-1
17
keratin associated protein 1-1
ENSG00000188581


X
KRTAP9-3
17
keratin associated protein 9-3
ENSG00000204873


X
CKMT1A
15
creatine kinase, mitochondrial 1A
ENSG00000223572


X
CKMT1B
15
creatine kinase, mitochondrial 1B
ENSG00000237289


X
UBC
12
ubiquitin C
ENSG00000150991


X
UBB
17
ubiquitin B
ENSG00000170315


X
KRT13
17
keratin 13
ENSG00000171401


X
ATP5B
12
ATP synthase, H+ transporting, mitochondrial
ENSG00000110955





F1 complex, beta polypeptide


X
HSPA2
14
heat shock protein family A (Hsp70) member 2
ENSG00000126803


X
EEF2
19
eukaryotic translation elongation factor 2
ENSG00000167658


X
ACTG1
17
actin gamma 1
ENSG00000184009


X
KRTAP1-3
17
keratin associated protein 1-3
ENSG00000221880


X
KRTAP4-7
17
keratin associated protein 4-7
ENSG00000240871


X
HIST1H4H
6
histone cluster 1 H4 family member h
ENSG00000158406


X
C1orf204
1
chromosome 1 open reading frame 204
ENSG00000188004


X
KRTAP24-1
21
keratin associated protein 24-1
ENSG00000188694


X
HIST1H4C
6
histone cluster 1 H4 family member c
ENSG00000197061


X
HIST1H4J
6
histone cluster 1 H4 family member j
ENSG00000197238


X
HIST4H4
12
histone cluster 4 H4
ENSG00000197837


X
VSIG8
1
V-set and immunoglobulin domain containing 8
ENSG00000243284


X
HIST2H4B
1
histone cluster 2 H4 family member b
ENSG00000270276


X
HIST2H4A
1
histone cluster 2 H4 family member a
ENSG00000270882


X
HIST1H4K
6
histone cluster 1 H4 family member k
ENSG00000273542


X
HIST1H4F
6
histone cluster 1 H4 family member f
ENSG00000274618


X
HIST1H4L
6
histone cluster 1 H4 family member l
ENSG00000275126


X
HIST1H4I
6
histone cluster 1 H4 family member i
ENSG00000276180


X
HIST1H4E
6
histone cluster 1 H4 family member e
ENSG00000276966


X
HIST1H4D
6
histone cluster 1 H4 family member d
ENSG00000277157


X
HIST1H4A
6
histone cluster 1 H4 family member a
ENSG00000278637


X
HIST1H4B
6
histone cluster 1 H4 family member b
ENSG00000278705


X
MDH2
7
malate dehydrogenase 2
ENSG00000146701


X
CALML3
10
calmodulin like 3
ENSG00000178363


X
KRTAP13-2
21
keratin associated protein 13-2
ENSG00000182816


X
MIF
22
macrophage migration inhibitory factor
ENSG00000240972





(glycosylation-inhibiting factor)


X
LAP3
4
leucine aminopeptidase 3
ENSG00000002549


X
HSPB1
7
heat shock protein family B (small) member 1
ENSG00000106211


X
KRT32
17
keratin 32
ENSG00000108759


X
GAPDH
12
glyceraldehyde-3-phosphate dehydrogenase
ENSG00000111640


X
TGM3
20
transglutaminase 3
ENSG00000125780


X
ATP5A1
18
ATP synthase, H+ transporting, mitochondrial
ENSG00000152234





F1 complex, alpha subunit 1, cardiac muscle


X
KRTAP11-1
21
keratin associated protein 11-1
ENSG00000182591


X
PKP3
11
plakophilin 3
ENSG00000184363


X
KRT40
17
keratin 40
ENSG00000204889


X
KRT81
12
keratin 81
ENSG00000205426


X
KRTAP3-3
17
keratin associated protein 3-3
ENSG00000212899


X
KRTAP3-2
17
keratin associated protein 3-2
ENSG00000212900


X
KRTAP3-1
17
keratin associated protein 3-1
ENSG00000212901


X
KRT33A
17
keratin 33A
ENSG00000006059


X
KRT31
17
keratin 31
ENSG00000094796


X
DSP
6
desmoplakin
ENSG00000096696


X
KRT36
17
keratin 36
ENSG00000126337


X
KRT34
17
keratin 34
ENSG00000131737


X
KRT33B
17
keratin 33B
ENSG00000131738


X
LGALS3
14
galectin 3
ENSG00000131981


X
KRT85
12
keratin 85
ENSG00000135443


X
TRIM29
11
tripartite motif containing 29
ENSG00000137699


X
SELENBP1
1
selenium binding protein 1
ENSG00000143416


X
KRT84
12
keratin 84
ENSG00000161849


X
KRT82
12
keratin 82
ENSG00000161850


X
KRT86
12
keratin 86
ENSG00000170442


X
KRT83
12
keratin 83
ENSG00000170523


X
KRT38
17
keratin 38
ENSG00000171360


X
JUP
17
junction plakoglobin
ENSG00000173801


X
DSG4
18
desmoglein 4
ENSG00000175065


X
SFN
1
stratifin
ENSG00000175793


X
LGALS7B
19
galectin 7B
ENSG00000178934


X
KRT39
17
keratin 39
ENSG00000196859


X
KRT35
17
keratin 35
ENSG00000197079


X
LGALS7
19
galectin 7
ENSG00000205076


X
KRTAP1-5
17
keratin associated protein 1-5
ENSG00000221852









Example 44
Exemplary Genes Comprising Marker Exome Sequences Validated in Bone Type Samples

An exemplary set of genes that can be used in methods and systems herein described as well as in related databases is reported herein. In particular, the exemplary set of genes comprises genes validated as proteomically detectable in bone samples of a Homo Sapiens which can be used in methods and systems to detect a genetic variation and/or perform a genetic variation analysis wherein the biological organism is a human being, as well as in related databases, in accordance with the various aspects of the present disclosure.


Specifically, Table 9 shows a list of exemplary genes that appear in MS files taken for samples of a bone of human beings. The fields in this example are the preference (X=more preferred), the standard gene symbol (gene symbol), the chromosome where the gene is located (chr), a description of the gene (gene description) and the gene identifier in the database Ensembl at the date of filing of the instant disclosure (Ensembl Gene Identifier).


The exemplary genes of Table 9 can be therefore used in particular in methods and systems of the disclosure wherein the sample comprises a bone sample from human beings.









TABLE 9







Exemplary genes identified in mass spectrometric analysis of bone type samples











X = more



Ensembl gene


preferred
gene symbol
chr
gene description
identifier















TUBB8
10
tubulin beta 8 class VIII
ENSG00000261456



TTR
18
transthyretin
ENSG00000118271



FBN2
5
fibrillin 2
ENSG00000138829



COL4A6
X
collagen type IV alpha 6 chain
ENSG00000197565



COL15A1
9
collagen type XV alpha 1 chain
ENSG00000204291



ACAN
15
aggrecan
ENSG00000157766



CNN2
19
calponin 2
ENSG00000064666



CDK5RAP2
9
CDK5 regulatory subunit associated protein 2
ENSG00000136861



TPSAB1
16
tryptase alpha/beta 1
ENSG00000172236



MATR3
5
matrin 3
ENSG00000280987



RP1L1
8
RP1 like 1
ENSG00000183638



IGFBP3
7
insulin like growth factor binding protein 3
ENSG00000146674



FBLN1
22
fibulin 1
ENSG00000077942



CAPZB
1
capping actin protein of muscle Z-line beta
ENSG00000077549





subunit



POSTN
13
periostin
ENSG00000133110



ELN
7
elastin
ENSG00000049540



MFAP5
12
microfibrillar associated protein 5
ENSG00000197614



UBB
17
ubiquitin B
ENSG00000170315



DDT
22
D-dopachrome tautomerase
ENSG00000099977



VIT
2
vitrin
ENSG00000205221



CYCS
7
cytochrome c, somatic
ENSG00000172115



CTSD
11
cathepsin D
ENSG00000117984



TRH
3
thyrotropin releasing hormone
ENSG00000170893



COL13A1
10
collagen type XIII alpha 1 chain
ENSG00000197467



ATP11A
13
ATPase phospholipid transporting 11A
ENSG00000068650



RPL27A
11
ribosomal protein L27a
ENSG00000166441



UBC
12
ubiquitin C
ENSG00000150991



MFGE8
15
milk fat globule-EGF factor 8 protein
ENSG00000140545



RPS10
6
ribosomal protein S10
ENSG00000124614



RPS20
8
ribosomal protein S20
ENSG00000008988



TGFBI
5
transforming growth factor beta induced
ENSG00000120708



SRP14
15
signal recognition particle 14
ENSG00000140319



RPL19
17
ribosomal protein L19
ENSG00000108298



KMT2D
12
lysine methyltransferase 2D
ENSG00000167548



TPP1
11
tripeptidyl peptidase 1
ENSG00000166340



GRIN2D
19
glutamate ionotropic receptor NMDA type
ENSG00000105464





subunit 2D



ANGPTL7
1
angiopoietin like 7
ENSG00000171819



CA2
8
carbonic anhydrase 2
ENSG00000104267



HBE1
11
hemoglobin subunit epsilon 1
ENSG00000213931



AMBP
9
alpha-1-microglobulin/bikunin precursor
ENSG00000106927



ORM1
9
orosomucoid 1
ENSG00000229314



PF4
4
platelet factor 4
ENSG00000163737



CYBB
X
cytochrome b-245 beta chain
ENSG00000165168



C2
6
complement C2
ENSG00000166278



C4A
6
complement C4A (Rodgers blood group)
ENSG00000244731



HSPA1B
6
heat shock protein family A (Hsp70) member
ENSG00000204388





1B



PF4V1
4
platelet factor 4 variant 1
ENSG00000109272



HSPA5
9
heat shock protein family A (Hsp70) member 5
ENSG00000044574



ACTN1
14
actinin alpha 1
ENSG00000072110



LCP1
13
lymphocyte cytosolic protein 1
ENSG00000136167



PLA2G2A
1
phospholipase A2 group IIA
ENSG00000188257



HIST1H1T
6
histone cluster 1 H1 family member t
ENSG00000187475



PPIB
15
peptidylprolyl isomerase B
ENSG00000166794



RPL12
9
ribosomal protein L12
ENSG00000197958



PEBP1
12
phosphatidylethanolamine binding protein 1
ENSG00000089220



RDX
11
radixin
ENSG00000137710



MYH9
22
myosin heavy chain 9
ENSG00000100345



NPTX2
7
neuronal pentraxin 2
ENSG00000106236



CXCL12
10
C-X-C motif chemokine ligand 12
ENSG00000107562



H2BFS
21
H2B histone family member S
ENSG00000234289



SNRPD3
22
small nuclear ribonucleoprotein D3 polypeptide
ENSG00000100028



RPL7A
9
ribosomal protein L7a
ENSG00000148303



RPS4X
X
ribosomal protein S4, X-linked
ENSG00000198034



RPS26
12
ribosomal protein S26
ENSG00000197728



RPL39
X
ribosomal protein L39
ENSG00000198918



RPS21
20
ribosomal protein S21
ENSG00000171858



CAP1
1
adenylate cyclase associated protein 1
ENSG00000131236



DPT
1
dermatopontin
ENSG00000143196



KHDRBS1
1
KH RNA binding domain containing, signal
ENSG00000121774





transduction associated 1



GAS6
13
growth arrest specific 6
ENSG00000183087



PDIA6
2
protein disulfide isomerase family A member 6
ENSG00000143870



HIST3H3
1
histone cluster 3 H3
ENSG00000168148



TMEM119
12
transmembrane protein 119
ENSG00000183160



TMPRSS6
22
transmembrane protease, serine 6
ENSG00000187045



AEBP1
7
AE binding protein 1
ENSG00000106624



COL27A1
9
collagen type XXVII alpha 1 chain
ENSG00000196739



PGLYRP2
19
peptidoglycan recognition protein 2
ENSG00000161031



TUBB1
20
tubulin beta 1 class VI
ENSG00000101162



COL17A1
10
collagen type XVII alpha 1 chain
ENSG00000065618



PRSS56
2
protease, serine 56
ENSG00000237412



GLIPR2
9
GLI pathogenesis related 2
ENSG00000122694



APP
21
amyloid beta precursor protein
ENSG00000142192



CPNE1
20
copine 1
ENSG00000214078



RAN
12
RAN, member RAS oncogene family
ENSG00000132341



HSPE1
2
heat shock protein family E (Hsp10) member 1
ENSG00000115541



MATR3
5
matrin 3
ENSG00000015479



HINT1
5
histidine triad nucleotide binding protein 1
ENSG00000169567



RPS23
5
ribosomal protein S23
ENSG00000186468



CLU
8
clusterin
ENSG00000120885



EZR
6
ezrin
ENSG00000092820



HSPA8
11
heat shock protein family A (Hsp70) member 8
ENSG00000109971



RPL8
8
ribosomal protein L8
ENSG00000161016



ACAT1
11
acetyl-CoA acetyltransferase 1
ENSG00000075239



C4B
6
complement C4B (Chido blood group)
ENSG00000224389



HMBS
11
hydroxymethylbilane synthase
ENSG00000256269



APOA1
11
apolipoprotein A1
ENSG00000118137



FTH1
11
ferritin heavy chain 1
ENSG00000167996



COMP
19
cartilage oligomeric matrix protein
ENSG00000105664



RPS27A
2
ribosomal protein S27a
ENSG00000143947



CLEC11A
19
C-type lectin domain containing 11A
ENSG00000105472



APOA2
1
apolipoprotein A2
ENSG00000158874



APCS
1
amyloid P component, serum
ENSG00000132703



FN1
2
fibronectin 1
ENSG00000115414



C8A
1
complement C8 alpha chain
ENSG00000157131



TUBB
6
tubulin beta class I
ENSG00000196230



LPA
6
lipoprotein(a)
ENSG00000198670



CFH
1
complement factor H
ENSG00000000971



HIST1H2AG
6
histone cluster 1 H2A family member g
ENSG00000196787



HIST1H2AI
6
histone cluster 1 H2A family member i
ENSG00000196747



HIST1H2AK
6
histone cluster 1 H2A family member k
ENSG00000275221



HIST1H2AM
6
histone cluster 1 H2A family member m
ENSG00000278677



HIST1H2AL
6
histone cluster 1 H2A family member l
ENSG00000276903



POTEI
2
POTE ankyrin domain family member I
ENSG00000196834



HSPA1A
6
heat shock protein family A (Hsp70) member
ENSG00000204389





1A



HIST1H2AD
6
histone cluster 1 H2A family member d
ENSG00000196866



CMA1
14
chymase 1
ENSG00000092009



LOX
5
lysyl oxidase
ENSG00000113083



THBS2
6
thrombospondin 2
ENSG00000186340



CDC42
1
cell division cycle 42
ENSG00000070831



RPS25
11
ribosomal protein S25
ENSG00000118181



TUBB4B
9
tubulin beta 4B class IVb
ENSG00000188229



DMP1
4
dentin matrix acidic phosphoprotein 1
ENSG00000152592



TUBB2A
6
tubulin beta 2A class IIa
ENSG00000137267



PLEC
8
plectin
ENSG00000178209



PGAM4
X
phosphoglycerate mutase family member 4
ENSG00000226784



HIST3H2BB
1
histone cluster 3 H2B family member b
ENSG00000196890



LRRC59
17
leucine rich repeat containing 59
ENSG00000108829



HIST1H2AH
6
histone cluster 1 H2A family member h
ENSG00000274997



HIST1H2AJ
6
histone cluster 1 H2A family member j
ENSG00000276368



MYOC
1
myocilin
ENSG00000034971



H2AFJ
12
H2A histone family member J
ENSG00000246705



TUBB2B
6
tubulin beta 2B class IIb
ENSG00000137285



TNMD
X
tenomodulin
ENSG00000000005



RPS10-NUDT3
6
RPS10-NUDT3 readthrough
ENSG00000270800



COL14A1
8
collagen type XIV alpha 1 chain
ENSG00000187955



PCMT1
6
protein-L-isoaspartate (D-aspartate) O-
ENSG00000120265





methyltransferase



IGHG1
14
immunoglobulin heavy constant gamma 1
ENSG00000211896





(G1m marker)



IGLL5
22
immunoglobulin lambda like polypeptide 5
ENSG00000254709



HIST1H3D
6
histone cluster 1 H3 family member d
ENSG00000282988



GSTP1
11
glutathione S-transferase pi 1
ENSG00000084207



HP1BP3
1
heterochromatin protein 1 binding protein 3
ENSG00000127483



YWHAE
17
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000108953





monooxygenase activation protein epsilon



RPL3
22
ribosomal protein L3
ENSG00000100316



RPL31
2
ribosomal protein L31
ENSG00000071082



RARRES2
7
retinoic acid receptor responder 2
ENSG00000106538



CA1
8
carbonic anhydrase 1
ENSG00000133742



RPL26L1
5
ribosomal protein L26 like 1
ENSG00000037241



RPL15
3
ribosomal protein L15
ENSG00000174748



RPL6
12
ribosomal protein L6
ENSG00000089009



CRIP2
14
cysteine rich protein 2
ENSG00000182809



RPL26
17
ribosomal protein L26
ENSG00000161970



APOH
17
apolipoprotein H
ENSG00000091583



RPL27
17
ribosomal protein L27
ENSG00000131469



A2M
12
alpha-2-macroglobulin
ENSG00000175899



IGHG4
14
immunoglobulin heavy constant gamma 4
ENSG00000211892





(G4m marker)



HPX
11
hemopexin
ENSG00000110169



FTL
19
ferritin light chain
ENSG00000087086



HIST1H2BJ
6
histone cluster 1 H2B family member j
ENSG00000124635



MIF
22
macrophage migration inhibitory factor
ENSG00000240972





(glycosylation-inhibiting factor)



HIST1H1D
6
histone cluster 1 H1 family member d
ENSG00000124575



COL9A1
6
collagen type IX alpha 1 chain
ENSG00000112280



PRDX6
1
peroxiredoxin 6
ENSG00000117592



SFN
1
stratifin
ENSG00000175793



MDH2
7
malate dehydrogenase 2
ENSG00000146701



CRIP1
14
cysteine rich protein 1
ENSG00000213145



COL4A4
2
collagen type IV alpha 4 chain
ENSG00000081052



HNRNPK
9
heterogeneous nuclear ribonucleoprotein K
ENSG00000165119



COL24A1
1
collagen type XXIV alpha 1 chain
ENSG00000171502



CAVIN1
17
caveolae associated protein 1
ENSG00000177469



HIST1H2BA
6
histone cluster 1 H2B family member a
ENSG00000146047


X
ADH1C
4
alcohol dehydrogenase 1C (class I), gamma
ENSG00000248144





polypeptide


X
YWHAH
22
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000128245





monooxygenase activation protein eta


X
RPS7
2
ribosomal protein S7
ENSG00000171863


X
MYL6
12
myosin light chain 6
ENSG00000092841


X
FGG
4
fibrinogen gamma chain
ENSG00000171557


X
RPL23
17
ribosomal protein L23
ENSG00000125691


X
APOD
3
apolipoprotein D
ENSG00000189058


X
CLEC3B
3
C-type lectin domain family 3 member B
ENSG00000163815


X
ENO2
12
enolase 2
ENSG00000111674


X
RPL18
19
ribosomal protein L18
ENSG00000063177


X
HSPB1
7
heat shock protein family B (small) member 1
ENSG00000106211


X
ANXA2
15
annexin A2
ENSG00000182718


X
RPS19
19
ribosomal protein S19
ENSG00000105372


X
A1BG
19
alpha-1-B glycoprotein
ENSG00000121410


X
BLVRB
19
biliverdin reductase B
ENSG00000090013


X
HMGN4
6
high mobility group nucleosomal binding
ENSG00000182952





domain 4


X
HIST1H2BK
6
histone cluster 1 H2B family member k
ENSG00000197903


X
CILP
15
cartilage intermediate layer protein
ENSG00000138615


X
PGK1
X
phosphoglycerate kinase 1
ENSG00000102144


X
IGHA2
14
immunoglobulin heavy constant alpha 2 (A2m
ENSG00000211890





marker)


X
C1QA
1
complement C1q A chain
ENSG00000173372


X
C1QC
1
complement C1q C chain
ENSG00000159189


X
C9
5
complement C9
ENSG00000113600


X
ANXA1
9
annexin A1
ENSG00000135046


X
SPARC
5
secreted protein acidic and cysteine rich
ENSG00000113140


X
RNASE2
14
ribonuclease A family member 2
ENSG00000169385


X
COL8A1
3
collagen type VIII alpha 1 chain
ENSG00000144810


X
COL4A5
X
collagen type IV alpha 5 chain
ENSG00000188153


X
ACTBL2
5
actin, beta like 2
ENSG00000169067


X
EMILIN1
2
elastin microfibril interfacer 1
ENSG00000138080


X
YWHAB
20
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000166913





monooxygenase activation protein beta


X
POTEF
2
POTE ankyrin domain family member F
ENSG00000196604


X
GC
4
GC, vitamin D binding protein
ENSG00000145321


X
H2AFY
5
H2A histone family member Y
ENSG00000113648


X
VCAN
5
versican
ENSG00000038427


X
YWHAZ
8
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000164924





monooxygenase activation protein zeta


X
NPM1
5
nucleophosmin
ENSG00000181163


X
PROC
2
protein C, inactivator of coagulation factors Va
ENSG00000115718





and VIIIa


X
TNC
9
tenascin C
ENSG00000041982


X
YWHAQ
2
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000134308





monooxygenase activation protein theta


X
COL8A2
1
collagen type VIII alpha 2 chain
ENSG00000171812


X
SERPINA10
14
serpin family A member 10
ENSG00000140093


X
CD44
11
CD44 molecule (Indian blood group)
ENSG00000026508


X
AK1
9
adenylate kinase 1
ENSG00000106992


X
PARK7
1
Parkinsonism associated deglycase
ENSG00000116288


X
CP
3
ceruloplasmin
ENSG00000047457


X
IGHA1
14
immunoglobulin heavy constant alpha 1
ENSG00000211895


X
LMNA
1
lamin A/C
ENSG00000160789


X
S100A8
1
S100 calcium binding protein A8
ENSG00000143546


X
COL4A2
13
collagen type IV alpha 2 chain
ENSG00000134871


X
HMGB1
13
high mobility group box 1
ENSG00000189403


X
PGAM1
10
phosphoglycerate mutase 1
ENSG00000171314


X
PRDX5
11
peroxiredoxin 5
ENSG00000126432


X
CORO1A
16
coronin 1A
ENSG00000102879


X
PRDX2
19
peroxiredoxin 2
ENSG00000167815


X
GGT5
22
gamma-glutamyltransferase 5
ENSG00000099998


X
YWHAG
7
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000170027





monooxygenase activation protein gamma


X
COL28A1
7
collagen type XXVIII alpha 1 chain
ENSG00000215018


X
POTEE
2
POTE ankyrin domain family member E
ENSG00000188219


X
COL26A1
7
collagen type XXVI alpha 1 chain
ENSG00000160963


X
SOST
17
sclerostin
ENSG00000167941


X
EEF1D
8
eukaryotic translation elongation factor 1 delta
ENSG00000104529


X
VCL
10
vinculin
ENSG00000035403


X
GSN
9
gelsolin
ENSG00000148180


X
TKT
3
transketolase
ENSG00000163931


X
HP
16
haptoglobin
ENSG00000257017


X
FHL1
X
four and a half LIM domains 1
ENSG00000022267


X
ACTA1
1
actin, alpha 1, skeletal muscle
ENSG00000143632


X
SPP2
2
secreted phosphoprotein 2
ENSG00000072080


X
SPP1
4
secreted phosphoprotein 1
ENSG00000118785


X
FGB
4
fibrinogen beta chain
ENSG00000171564


X
ENO3
17
enolase 3
ENSG00000108515


X
CFL1
11
cofilin 1
ENSG00000172757


X
COL21A1
6
collagen type XXI alpha 1 chain
ENSG00000124749


X
ALDOA
16
aldolase, fructose-bisphosphate A
ENSG00000149925


X
PKM
15
pyruvate kinase, muscle
ENSG00000067225


X
RPL13
16
ribosomal protein L13
ENSG00000167526


X
CILP2
19
cartilage intermediate layer protein 2
ENSG00000160161


X
PLG
6
plasminogen
ENSG00000122194


X
HMGN2
1
high mobility group nucleosomal binding
ENSG00000198830





domain 2


X
PROS1
3
protein S (alpha)
ENSG00000184500


X
SOD3
4
superoxide dismutase 3
ENSG00000109610


X
EPX
17
eosinophil peroxidase
ENSG00000121053


X
RNASE3
14
ribonuclease A family member 3
ENSG00000169397


X
HIST1H1C
6
histone cluster 1 H1 family member c
ENSG00000187837


X
ITIH2
10
inter-alpha-trypsin inhibitor heavy chain 2
ENSG00000151655


X
DEFA1
8
defensin alpha 1
ENSG00000206047


X
DEFA1B
8
defensin alpha 1B
ENSG00000240247


X
ACTC1
15
actin, alpha, cardiac muscle 1
ENSG00000159251


X
FMOD
1
fibromodulin
ENSG00000122176


X
HIST2H3D
1
histone cluster 2 H3 family member d
ENSG00000183598


X
HIST2H3C
1
histone cluster 2 H3 family member c
ENSG00000203811


X
HIST2H3A
1
histone cluster 2 H3 family member a
ENSG00000203852


X
FLNA
X
filamin A
ENSG00000196924


X
PRDX1
1
peroxiredoxin 1
ENSG00000117450


X
GPI
19
glucose-6-phosphate isomerase
ENSG00000105220


X
COL11A2
6
collagen type XI alpha 2 chain
ENSG00000204248


X
OLFML3
1
olfactomedin like 3
ENSG00000116774


X
HSPD1
2
heat shock protein family D (Hsp60) member 1
ENSG00000144381


X
AHSG
3
alpha 2-HS glycoprotein
ENSG00000145192


X
COL6A3
2
collagen type VI alpha 3 chain
ENSG00000163359


X
LYZ
12
lysozyme
ENSG00000090382


X
SOD1
21
superoxide dismutase 1
ENSG00000142168


X
ACTG1
17
actin gamma 1
ENSG00000184009


X
SERPINC1
1
serpin family C member 1
ENSG00000117601


X
C3
19
complement C3
ENSG00000125730


X
FGA
4
fibrinogen alpha chain
ENSG00000171560


X
ANG
14
angiogenin
ENSG00000214274


X
CAT
11
catalase
ENSG00000121691


X
IGF1
12
insulin like growth factor 1
ENSG00000017427


X
ENO1
1
enolase 1
ENSG00000074800


X
H1F0
22
H1 histone family member 0
ENSG00000189060


X
CA3
8
carbonic anhydrase 3
ENSG00000164879


X
ELANE
19
elastase, neutrophil expressed
ENSG00000197561


X
LGALS1
22
galectin 1
ENSG00000100097


X
EEF2
19
eukaryotic translation elongation factor 2
ENSG00000167658


X
PGAM2
7
phosphoglycerate mutase 2
ENSG00000164708


X
HIST1H1B
6
histone cluster 1 H1 family member b
ENSG00000184357


X
OGN
9
osteoglycin
ENSG00000106809


X
PDIA3
15
protein disulfide isomerase family A member 3
ENSG00000167004


X
COL10A1
6
collagen type X alpha 1 chain
ENSG00000123500


X
COL16A1
1
collagen type XVI alpha 1 chain
ENSG00000084636


X
PCOLCE
7
procollagen C-endopeptidase enhancer
ENSG00000106333


X
OLFML1
11
olfactomedin like 1
ENSG00000183801


X
HIST2H2AB
1
histone cluster 2 H2A family member b
ENSG00000184270


X
COL22A1
8
collagen type XXII alpha 1 chain
ENSG00000169436


X
HTRA1
10
HtrA serine peptidase 1
ENSG00000166033


X
OMD
9
osteomodulin
ENSG00000127083


X
TLN1
9
talin 1
ENSG00000137076


X
COL1A2
7
collagen type I alpha 2 chain
ENSG00000164692


X
EEF1A1
6
eukaryotic translation elongation factor 1 alpha
ENSG00000156508





1


X
COL5A1
9
collagen type V alpha 1 chain
ENSG00000130635


X
COL6A1
21
collagen type VI alpha 1 chain
ENSG00000142156


X
C1QB
1
complement C1q B chain
ENSG00000173369


X
LTF
3
lactotransferrin
ENSG00000012223


X
MEPE
4
matrix extracellular phosphoglycoprotein
ENSG00000152595


X
COL12A1
6
collagen type XII alpha 1 chain
ENSG00000111799


X
FBN1
15
fibrillin 1
ENSG00000166147


X
PFN1
17
profilin 1
ENSG00000108518


X
KNG1
3
kininogen 1
ENSG00000113889


X
IGF2
11
insulin like growth factor 2
ENSG00000167244


X
MPO
17
myeloperoxidase
ENSG00000005381


X
THBS1
15
thrombospondin 1
ENSG00000137801


X
MGP
12
matrix Gla protein
ENSG00000111341


X
COL6A2
21
collagen type VI alpha 2 chain
ENSG00000142173


X
AZU1
19
azurocidin 1
ENSG00000172232


X
HIST1H2BO
6
histone cluster 1 H2B family member o
ENSG00000274641


X
HIST1H2BB
6
histone cluster 1 H2B family member b
ENSG00000276410


X
DEFA3
8
defensin alpha 3
ENSG00000239839


X
TPI1
12
triosephosphate isomerase 1
ENSG00000111669


X
HIST1H3H
6
histone cluster 1 H3 family member h
ENSG00000278828


X
HIST1H3I
6
histone cluster 1 H3 family member i
ENSG00000275379


X
HIST1H3J
6
histone cluster 1 H3 family member j
ENSG00000197153


X
HIST1H3A
6
histone cluster 1 H3 family member a
ENSG00000275714


X
HIST1H3B
6
histone cluster 1 H3 family member b
ENSG00000274267


X
HIST1H3C
6
histone cluster 1 H3 family member c
ENSG00000278272


X
HIST1H3D
6
histone cluster 1 H3 family member d
ENSG00000197409


X
HIST1H3E
6
histone cluster 1 H3 family member e
ENSG00000274750


X
HIST1H3F
6
histone cluster 1 H3 family member f
ENSG00000277775


X
HIST1H3G
6
histone cluster 1 H3 family member g
ENSG00000273983


X
H3F3A
1
H3 histone family member 3A
ENSG00000163041


X
HSPG2
1
heparan sulfate proteoglycan 2
ENSG00000142798


X
COL7A1
3
collagen type VII alpha 1 chain
ENSG00000114270


X
AHNAK
11
AHNAK nucleoprotein
ENSG00000124942


X
HIST2H2BE
1
histone cluster 2 H2B family member e
ENSG00000184678


X
ASPN
9
asporin
ENSG00000106819


X
HIST3H2A
1
histone cluster 3 H2A
ENSG00000181218


X
HIST1H2AC
6
histone cluster 1 H2A family member c
ENSG00000180573


X
COL5A2
2
collagen type V alpha 2 chain
ENSG00000204262


X
HBB
11
hemoglobin subunit beta
ENSG00000244734


X
COL11A1
1
collagen type XI alpha 1 chain
ENSG00000060718


X
MB
22
myoglobin
ENSG00000198125


X
VIM
10
vimentin
ENSG00000026025


X
HIST1H2BC
6
histone cluster 1 H2B family member c
ENSG00000180596


X
HIST1H2BF
6
histone cluster 1 H2B family member f
ENSG00000277224


X
HIST1H2BE
6
histone cluster 1 H2B family member e
ENSG00000274290


X
HIST1H2BG
6
histone cluster 1 H2B family member g
ENSG00000273802


X
HIST1H2BI
6
histone cluster 1 H2B family member i
ENSG00000278588


X
H2AFV
7
H2A histone family member V
ENSG00000105968


X
PPIA
7
peptidylprolyl isomerase A
ENSG00000196262


X
BGN
X
biglycan
ENSG00000182492


X
ACTB
7
actin beta
ENSG00000075624


X
IGFBP5
2
insulin like growth factor binding protein 5
ENSG00000115461


X
GAPDH
12
glyceraldehyde-3-phosphate dehydrogenase
ENSG00000111640


X
ALB
4
albumin
ENSG00000163631


X
COL3A1
2
collagen type III alpha 1 chain
ENSG00000168542


X
SERPINF1
17
serpin family F member 1
ENSG00000132386


X
H3F3B
17
H3 histone family member 3B
ENSG00000132475


X
CHAD
17
chondroadherin
ENSG00000136457


X
F2
11
coagulation factor II, thrombin
ENSG00000180210


X
F9
X
coagulation factor IX
ENSG00000101981


X
F10
13
coagulation factor X
ENSG00000126218


X
SERPINA1
14
serpin family A member 1
ENSG00000197249


X
IGHG2
14
immunoglobulin heavy constant gamma 2
ENSG00000211893





(G2m marker)


X
HBD
11
hemoglobin subunit delta
ENSG00000223609


X
COL1A1
17
collagen type I alpha 1 chain
ENSG00000108821


X
COL2A1
12
collagen type II alpha 1 chain
ENSG00000139219


X
TF
3
transferrin
ENSG00000091513


X
BGLAP
1
bone gamma-carboxyglutamate protein
ENSG00000242252


X
VTN
17
vitronectin
ENSG00000109072


X
HIST1H2AB
6
histone cluster 1 H2A family member b
ENSG00000278463


X
HIST1H2AE
6
histone cluster 1 H2A family member e
ENSG00000277075


X
S100A9
1
SI00 calcium binding protein A9
ENSG00000163220


X
CKM
19
creatine kinase, M-type
ENSG00000104879


X
DCN
12
decorin
ENSG00000011465


X
CTSG
14
cathepsin G
ENSG00000100448


X
H2AFZ
4
H2A histone family member Z
ENSG00000164032


X
HIST1H1E
6
histone cluster 1 H1 family member e
ENSG00000168298


X
H2AFX
11
H2A histone family member X
ENSG00000188486


X
IBSP
4
integrin binding sialoprotein
ENSG00000029559


X
PRTN3
19
proteinase 3
ENSG00000196415


X
COL5A3
19
collagen type V alpha 3 chain
ENSG00000080573


X
LUM
12
lumican
ENSG00000139329


X
PRELP
1
proline and arginine rich end leucine rich repeat
ENSG00000188783





protein


X
HIST1H2BD
6
histone cluster 1 H2B family member d
ENSG00000158373


X
HIST1H4I
6
histone cluster 1 H4 family member i
ENSG00000276180


X
HIST1H4K
6
histone cluster 1 H4 family member k
ENSG00000273542


X
HIST1H4J
6
histone cluster 1 H4 family member j
ENSG00000197238


X
HIST1H4L
6
histone cluster 1 H4 family member l
ENSG00000275126


X
HIST2H4A
1
histone cluster 2 H4 family member a
ENSG00000270882


X
HIST2H4B
1
histone cluster 2 H4 family member b
ENSG00000270276


X
HIST1H4A
6
histone cluster 1 H4 family member a
ENSG00000278637


X
HIST1H4B
6
histone cluster 1 H4 family member b
ENSG00000278705


X
HIST1H4C
6
histone cluster 1 H4 family member c
ENSG00000197061


X
HIST1H4D
6
histone cluster 1 H4 family member d
ENSG00000277157


X
HIST1H4E
6
histone cluster 1 H4 family member e
ENSG00000276966


X
HIST1H4F
6
histone cluster 1 H4 family member f
ENSG00000274618


X
HIST1H4H
6
histone cluster 1 H4 family member h
ENSG00000158406


X
HIST4H4
12
histone cluster 4 H4
ENSG00000197837


X
HBA2
16
hemoglobin subunit alpha 2
ENSG00000188536


X
HBA1
16
hemoglobin subunit alpha 1
ENSG00000206172


X
HIST2H2AC
1
histone cluster 2 H2A family member c
ENSG00000184260


X
HIST2H2BF
1
histone cluster 2 H2B family member f
ENSG00000203814


X
HIST2H2AA3
1
histone cluster 2 H2A family member a3
ENSG00000203812


X
HIST2H2AA4
1
histone cluster 2 H2A family member a4
ENSG00000272196


X
HIST1H2BH
6
histone cluster 1 H2B family member h
ENSG00000275713


X
HIST1H2BN
6
histone cluster 1 H2B family member n
ENSG00000233822


X
HIST1H2BM
6
histone cluster 1 H2B family member m
ENSG00000273703


X
HIST1H2BL
6
histone cluster 1 H2B family member l
ENSG00000185130









Example 45
Exemplary Genes Comprising Marker Exome Sequences Validated in Skin Samples

An exemplary set of genes that can be used in methods and systems herein described as well as in related databases is reported herein. In particular, the exemplary set of genes comprises genes validated as proteomically detectable in skin samples of Homo Sapiens which can be used in methods and systems to detect a genetic variation and/or perform a genetic variation analysis, as well as in related databases, in accordance with the various aspects of the present disclosure.


Specifically, Table 10 shows a list of exemplary genes that appear in MS files taken for skin samples of human beings. The fields in this example are the preference (X=more preferable), the standard gene symbol (gene symbol), the chromosome wherein the gene is located (chr), a description of the gene (gene description) and an identifier in the database Ensembl at the date of filing of the instant disclosure (Ensembl Gene Identifier).


The exemplary genes of Table 10 can be used in particular in methods and system of the disclosure wherein the sample comprises a skin sample from human beings.









TABLE 10







Exemplary genes identified in mass spectrometric analysis of skin samples











X = more



Ensembl gene


preferable
gene symbol
chr
gene description
identifier















TULP1
6
tubby like protein 1
ENSG00000112041



ACTN4
19
actinin alpha 4
ENSG00000130402



PLXNC1
12
plexin C1
ENSG00000136040



KRT33A
17
keratin 33A
ENSG00000006059



LDHA
11
lactate dehydrogenase A
ENSG00000134333



PIGR
1
polymeric immunoglobulin receptor
ENSG00000162896



LTF
3
lactotransferrin
ENSG00000012223



SERPINB2
18
serpin family B member 2
ENSG00000197632



GSN
9
gelsolin
ENSG00000148180



TUBB
6
tubulin beta class I
ENSG00000196230



IVL
1
involucrin
ENSG00000163207



LCT
2
lactase
ENSG00000115850



NEFH
22
neurofilament heavy
ENSG00000100285



APEH
3
acylaminoacyl-peptide hydrolase
ENSG00000164062



IDE
10
insulin degrading enzyme
ENSG00000119912



ARF4
3
ADP ribosylation factor 4
ENSG00000168374



VCL
10
vinculin
ENSG00000035403



AMPD1
1
adenosine monophosphate deaminase 1
ENSG00000116748



PSMA2
7
proteasome subunit alpha 2
ENSG00000106588



PEBP1
12
phosphatidylethanolamine binding
ENSG00000089220





protein 1



KIF5B
10
kinesin family member 5B
ENSG00000170759



TALDO1
11
transaldolase 1
ENSG00000177156



ME1
6
malic enzyme 1
ENSG00000065833



CENPF
1
centromere protein F
ENSG00000117724



SSR4
X
signal sequence receptor subunit 4
ENSG00000180879



VAMP7
X
vesicle associated membrane protein 7
ENSG00000124333



S100A10
1
S100 calcium binding protein A10
ENSG00000197747



ARF3
12
ADP ribosylation factor 3
ENSG00000134287



TPM4
19
tropomyosin 4
ENSG00000167460



TUBA4A
2
tubulin alpha 4a
ENSG00000127824



TUBB4B
9
tubulin beta 4B class IVb
ENSG00000188229



ARF5
7
ADP ribosylation factor 5
ENSG00000004059



MAP3K10
19
mitogen-activated protein kinase kinase
ENSG00000130758





kinase 10



AKAP13
15
A-kinase anchoring protein 13
ENSG00000170776



TUBB3
16
tubulin beta 3 class III
ENSG00000258947



RAB39A
11
RAB39A, member RAS oncogene
ENSG00000179331





family



FAM208B
10
family with sequence similarity 208
ENSG00000108021





member B



RAB12
18
RAB12, member RAS oncogene family
ENSG00000206418



ANO7
2
anoctamin 7
ENSG00000146205



TUBA3E
2
tubulin alpha 3e
ENSG00000152086



S100A7A
1
S100 calcium binding protein A7A
ENSG00000184330



RAB43
3
RAB43, member RAS oncogene family
ENSG00000172780



MAP7D3
X
MAP7 domain containing 3
ENSG00000129680



RASEF
9
RAS and EF-hand domain containing
ENSG00000165105



HIST3H2BB
1
histone cluster 3 H2B family member b
ENSG00000196890



SPATA5
4
spermatogenesis associated 5
ENSG00000145375



SYNE1
6
spectrin repeat containing nuclear
ENSG00000131018





envelope protein 1



RB1CC1
8
RB1 inducible coiled-coil 1
ENSG00000023287



TTC28
22
tetratricopeptide repeat domain 28
ENSG00000100154



RAB39B
X
RAB39B, member RAS oncogene
ENSG00000155961





family



IL12RB2
1
interleukin 12 receptor subunit beta 2
ENSG00000081985



TUBB2B
6
tubulin beta 2B class IIb
ENSG00000137285



RAB34
17
RAB34, member RAS oncogene family
ENSG00000109113



LACRT
12
lacritin
ENSG00000135413



RAB33B
4
RAB33B, member RAS oncogene
ENSG00000172007





family



RAB6B
3
RAB6B, member RAS oncogene family
ENSG00000154917



COG5
7
component of oligomeric golgi complex
ENSG00000164597





5



NOSIP
19
nitric oxide synthase interacting protein
ENSG00000142546



WNK2
9
WNK lysine deficient protein kinase 2
ENSG00000165238



RAB27B
18
RAB27B, member RAS oncogene
ENSG00000041353





family



PPL
16
periplakin
ENSG00000118898



KRT34
17
keratin 34
ENSG00000131737



PNP
14
purine nucleoside phosphorylase
ENSG00000198805



CST4
20
cystatin S
ENSG00000101441



CST1
20
cystatin SN
ENSG00000170373



ANXA1
9
annexin A1
ENSG00000135046



SEMG1
20
semenogelin I
ENSG00000124233



CAPN1
11
calpain 1
ENSG00000014216



PRSS1
7
protease, serine 1
ENSG00000204983



HSP90AA1
14
heat shock protein 90 alpha family class
ENSG00000080824





A member 1



GSTP1
11
glutathione S-transferase pi 1
ENSG00000084207



HARS
5
histidyl-tRNA synthetase
ENSG00000170445



DES
2
desmin
ENSG00000175084



GM2A
5
GM2 ganglioside activator
ENSG00000196743



RAB3B
1
RAB3B, member RAS oncogene family
ENSG00000169213



RAB4A
1
RAB4A, member RAS oncogene family
ENSG00000168118



PSMA1
11
proteasome subunit alpha 1
ENSG00000129084



CAPZB
1
capping actin protein of muscle Z-line
ENSG00000077549





beta subunit



ALDH9A1
1
aldehyde dehydrogenase 9 family
ENSG00000143149





member A1



PSMB3
17
proteasome subunit beta 3
ENSG00000277791



SERPINB8
18
serpin family B member 8
ENSG00000166401



RAB13
1
RAB13, member RAS oncogene family
ENSG00000143545



HIST1H4I
6
histone cluster 1 H4 family member i
ENSG00000276180



HIST1H4K
6
histone cluster 1 H4 family member k
ENSG00000273542



HIST1H4J
6
histone cluster 1 H4 family member j
ENSG00000197238



HIST1H4L
6
histone cluster 1 H4 family member l
ENSG00000275126



HIST2H4A
1
histone cluster 2 H4 family member a
ENSG00000270882



HIST2H4B
1
histone cluster 2 H4 family member b
ENSG00000270276



HIST1H4A
6
histone cluster 1 H4 family member a
ENSG00000278637



HIST1H4B
6
histone cluster 1 H4 family member b
ENSG00000278705



HIST1H4C
6
histone cluster 1 H4 family member c
ENSG00000197061



HIST1H4D
6
histone cluster 1 H4 family member d
ENSG00000277157



HIST1H4E
6
histone cluster 1 H4 family member e
ENSG00000276966



HIST1H4F
6
histone cluster 1 H4 family member f
ENSG00000274618



HIST1H4H
6
histone cluster 1 H4 family member h
ENSG00000158406



HIST4H4
12
histone cluster 4 H4
ENSG00000197837



SEMG2
20
semenogelin II
ENSG00000124157



MAP2K5
15
mitogen-activated protein kinase kinase
ENSG00000137764





5



TUBA3D
2
tubulin alpha 3d
ENSG00000075886



TUBA3C
13
tubulin alpha 3c
ENSG00000198033



CCDC40
17
coiled-coil domain containing 40
ENSG00000141519



KRT40
17
keratin 40
ENSG00000204889



SDR9C7
12
short chain dehydrogenase/reductase
ENSG00000170426





family 9C member 7



SHROOM3
4
shroom family member 3
ENSG00000138771



RAB3C
5
RAB3C, member RAS oncogene family
ENSG00000152932



S100A16
1
S100 calcium binding protein A16
ENSG00000188643



SPEF2
5
sperm flagellar 2
ENSG00000152582



KIF13B
8
kinesin family member 13B
ENSG00000197892



TUBA8
22
tubulin alpha 8
ENSG00000183785



TGM5
15
transglutaminase 5
ENSG00000104055



CREG1
1
cellular repressor of El A stimulated
ENSG00000143162





genes 1



PGK1
X
phosphoglycerate kinase 1
ENSG00000102144



RAB3A
19
RAB3A, member RAS oncogene family
ENSG00000105649



RAB6A
11
RAB6A, member RAS oncogene family
ENSG00000175582



CALML3
10
calmodulin like 3
ENSG00000178363



PSMB6
17
proteasome subunit beta 6
ENSG00000142507



KDM5A
12
lysine demethylase 5A
ENSG00000073614



HSPA9
5
heat shock protein family A (Hsp70)
ENSG00000113013





member 9



GDI2
10
GDP dissociation inhibitor 2
ENSG00000057608



SCAP
3
SREBF chaperone
ENSG00000114650



RAB11B
19
RAB11B, member RAS oncogene
ENSG00000185236





family



UGP2
2
UDP-glucose pyrophosphorylase 2
ENSG00000169764



RAB41
X
RAB41, member RAS oncogene family
ENSG00000147127



ZFYVE27
10
zinc finger FYVE-type containing 27
ENSG00000155256



REEP3
10
receptor accessory protein 3
ENSG00000165476



PLBD1
12
phospholipase B domain containing 1
ENSG00000121316



HIST2H2AB
1
histone cluster 2 H2A family member b
ENSG00000184270



H2AFZ
4
H2A histone family member Z
ENSG00000164032



POTEI
2
POTE ankyrin domain family member I
ENSG00000196834



EEF2
19
eukaryotic translation elongation factor 2
ENSG00000167658



PSMA3
14
proteasome subunit alpha 3
ENSG00000100567



S100A11
1
S100 calcium binding protein A11
ENSG00000163191



MYH9
22
myosin heavy chain 9
ENSG00000100345



RAB11A
15
RAB11A, member RAS oncogene
ENSG00000103769





family



ACTA2
10
actin, alpha 2, smooth muscle, aorta
ENSG00000107796



KRT33B
17
keratin 33B
ENSG00000131738



LGALSL
2
galectin like
ENSG00000119862



ACTBL2
5
actin, beta like 2
ENSG00000169067



H2AFV
7
H2A histone family member V
ENSG00000105968



DLG5
10
discs large MAGUK scaffold protein 5
ENSG00000151208



MUCL1
12
mucin like 1
ENSG00000172551



ALOXE3
17
arachidonate lipoxygenase 3
ENSG00000179148



RNASE7
14
ribonuclease A family member 7
ENSG00000165799



KRT37
17
keratin 37
ENSG00000108417



FMNL1
17
formin like 1
ENSG00000184922



RAB3D
19
RAB3D, member RAS oncogene family
ENSG00000105514



TPM3
1
tropomyosin 3
ENSG00000143549



HIST1H2AG
6
histone cluster 1 H2A family member g
ENSG00000196787



HIST1H2AI
6
histone cluster 1 H2A family member i
ENSG00000196747



HIST1H2AK
6
histone cluster 1 H2A family member k
ENSG00000275221



HIST1H2AM
6
histone cluster 1 H2A family member m
ENSG00000278677



HIST1H2AL
6
histone cluster 1 H2A family member l
ENSG00000276903



H2AFX
11
H2A histone family member X
ENSG00000188486



HIST1H2AD
6
histone cluster 1 H2A family member d
ENSG00000196866



SERPINB4
18
serpin family B member 4
ENSG00000206073



EIF3E
8
eukaryotic translation initiation factor 3
ENSG00000104408





subunit E



RAN
12
RAN, member RAS oncogene family
ENSG00000132341



ACTG2
2
actin, gamma 2, smooth muscle, enteric
ENSG00000163017



HIST2H2AC
1
histone cluster 2 H2A family member c
ENSG00000184260



HIST2H2AA3
1
histone cluster 2 H2A family member a3
ENSG00000203812



HIST2H2AA4
1
histone cluster 2 H2A family member a4
ENSG00000272196



RAB44
6
RAB44, member RAS oncogene family
ENSG00000255587



HIST1H2BA
6
histone cluster 1 H2B family member a
ENSG00000146047



HIST1H2AH
6
histone cluster 1 H2A family member h
ENSG00000274997



HIST1H2AA
6
histone cluster 1 H2A family member a
ENSG00000164508



HIST1H2AJ
6
histone cluster 1 H2A family member j
ENSG00000276368



KRT82
12
keratin 82
ENSG00000161850



HIST1H2BK
6
histone cluster 1 H2B family member k
ENSG00000197903



CSTA
3
cystatin A
ENSG00000121552



HIST1H2AB
6
histone cluster 1 H2A family member b
ENSG00000278463



HIST1H2AE
6
histone cluster 1 H2A family member e
ENSG00000277075



HIST1H2BJ
6
histone cluster 1 H2B family member j
ENSG00000124635



HIST1H2BO
6
histone cluster 1 H2B family member o
ENSG00000274641



HIST1H2BB
6
histone cluster 1 H2B family member b
ENSG00000276410



VCP
9
valosin containing protein
ENSG00000165280



H2BFS
21
H2B histone family member S
ENSG00000234289



HIST1H2BD
6
histone cluster 1 H2B family member d
ENSG00000158373



PSMA6
14
proteasome subunit alpha 6
ENSG00000100902



YWHAG
7
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000170027





monooxygenase activation protein





gamma



HIST1H2BC
6
histone cluster 1 H2B family member c
ENSG00000180596



HIST1H2BF
6
histone cluster 1 H2B family member f
ENSG00000277224



HIST1H2BE
6
histone cluster 1 H2B family member e
ENSG00000274290



HIST1H2BG
6
histone cluster 1 H2B family member g
ENSG00000273802



HIST1H2BI
6
histone cluster 1 H2B family member i
ENSG00000278588



ACTC1
15
actin, alpha, cardiac muscle 1
ENSG00000159251



ACTA1
1
actin, alpha 1, skeletal muscle
ENSG00000143632



TUBA1B
12
tubulin alpha lb
ENSG00000123416



PLEC
8
plectin
ENSG00000178209



HIST2H2BE
1
histone cluster 2 H2B family member e
ENSG00000184678



HIST2H2BF
1
histone cluster 2 H2B family member f
ENSG00000203814



PPRC1
10
peroxisome proliferator-activated
ENSG00000148840





receptor gamma, coactivator-related 1



SBSN
19
suprabasin
ENSG00000189001



TUBA1A
12
tubulin alpha 1a
ENSG00000167552



HIST3H2A
1
histone cluster 3 H2A
ENSG00000181218



HIST1H2AC
6
histone cluster 1 H2A family member c
ENSG00000180573



HIST1H2BH
6
histone cluster 1 H2B family member h
ENSG00000275713



HIST1H2BN
6
histone cluster 1 H2B family member n
ENSG00000233822



HIST1H2BM
6
histone cluster 1 H2B family member m
ENSG00000273703



HIST1H2BL
6
histone cluster 1 H2B family member l
ENSG00000185130



TUBA1C
12
tubulin alpha 1c
ENSG00000167553



THRA
17
thyroid hormone receptor, alpha
ENSG00000126351



GLRX
5
glutaredoxin
ENSG00000173221



AHNAK
11
AHNAK nucleoprotein
ENSG00000124942



SYPL1
7
synaptophysin like 1
ENSG00000008282



RRBP1
20
ribosome binding protein 1
ENSG00000125844



PSMD14
2
proteasome 26S subunit, non-ATPase 14
ENSG00000115233



ALDOA
16
aldolase, fructose-bisphosphate A
ENSG00000149925



THRB
3
thyroid hormone receptor beta
ENSG00000151090



KRT32
17
keratin 32
ENSG00000108759



TADA2B
4
transcriptional adaptor 2B
ENSG00000173011



HSPA1A
6
heat shock protein family A (Hsp70)
ENSG00000204389





member 1A



HSPA1B
6
heat shock protein family A (Hsp70)
ENSG00000204388





member 1B



YWHAQ
2
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000134308





monooxygenase activation protein theta



PSMA5
1
proteasome subunit alpha 5
ENSG00000143106



LCN1
9
lipocalin 1
ENSG00000160349



KRT31
17
keratin 31
ENSG00000094796



C1orf68
1
chromosome 1 open reading frame 68
ENSG00000198854



DBF4B
17
DBF4 zinc finger B
ENSG00000161692



PSMA8
18
proteasome subunit alpha 8
ENSG00000154611



A2ML1
12
alpha-2-macroglobulin like 1
ENSG00000166535



PSMA7
20
proteasome subunit alpha 7
ENSG00000101182



KRT38
17
keratin 38
ENSG00000171360



LMNA
1
lamin A/C
ENSG00000160789



TXN
9
thioredoxin
ENSG00000136810



CTSA
20
cathepsin A
ENSG00000064601



HSPA6
1
heat shock protein family A (Hsp70)
ENSG00000173110





member 6



YWHAB
20
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000166913





monooxygenase activation protein beta



RAB2A
8
RAB2A, member RAS oncogene family
ENSG00000104388



ECM1
1
extracellular matrix protein 1
ENSG00000143369



ASPRV1
2
aspartic peptidase, retroviral-like 1
ENSG00000244617



NCCRP1
19
non-specific cytotoxic cell receptor
ENSG00000188505





protein 1 homolog (zebrafish)



KRT222
17
keratin 222
ENSG00000213424



S100A14
1
S100 calcium binding protein A14
ENSG00000189334



ALOX12B
17
arachidonate 12-lipoxygenase, 12R type
ENSG00000179477



RAB2B
14
RAB2B, member RAS oncogene family
ENSG00000129472



CPA4
7
carboxypeptidase A4
ENSG00000128510



KRT83
12
keratin 83
ENSG00000170523



YWHAH
22
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000128245





monooxygenase activation protein eta



RAB35
12
RAB35, member RAS oncogene family
ENSG00000111737



LOR
1
loricrin
ENSG00000203782



RAB8A
19
RAB8A, member RAS oncogene family
ENSG00000167461



RAB10
2
RAB10, member RAS oncogene family
ENSG00000084733



KRT81
12
keratin 81
ENSG00000205426



KRT35
17
keratin 35
ENSG00000197079



KRT86
12
keratin 86
ENSG00000170442



ALB
4
albumin
ENSG00000163631



AZGP1
7
alpha-2-glycoprotein 1, zinc-binding
ENSG00000160862



SFN
1
stratifin
ENSG00000175793



YWHAZ
8
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000164924





monooxygenase activation protein zeta



KRT85
12
keratin 85
ENSG00000135443



POTEE
2
POTE ankyrin domain family member E
ENSG00000188219



KRT26
17
keratin 26
ENSG00000186393



RAB8B
15
RAB8B, member RAS oncogene family
ENSG00000166128



ENO2
12
enolase 2
ENSG00000111674



UBC
12
ubiquitin C
ENSG00000150991



FLG
1
filaggrin
ENSG00000143631



CTNNB1
3
catenin beta 1
ENSG00000168036



KRT20
17
keratin 20
ENSG00000171431



PRPH
12
peripherin
ENSG00000135406



YWHAE
17
tyrosine 3-monooxygenase/tryptophan 5-
ENSG00000108953





monooxygenase activation protein





epsilon



POTEF
2
POTE ankyrin domain family member F
ENSG00000196604



ENO3
17
enolase 3
ENSG00000108515



HSP90B1
12
heat shock protein 90 beta family
ENSG00000166598





member 1



RAB15
14
RAB15, member RAS oncogene family
ENSG00000139998



RPS27A
2
ribosomal protein S27a
ENSG00000143947



FABP5
8
fatty acid binding protein 5
ENSG00000164687



PKP1
1
plakophilin 1
ENSG00000081277



KRT74
12
keratin 74
ENSG00000170484



GSDMA
17
gasdermin A
ENSG00000167914



S100A8
1
S100 calcium binding protein A8
ENSG00000143546



HSP90AB1
6
heat shock protein 90 alpha family class
ENSG00000096384





B member 1



UBB
17
ubiquitin B
ENSG00000170315



BLMH
17
bleomycin hydrolase
ENSG00000108578



GGCT
7
gamma-glutamylcyclotransferase
ENSG00000006625



HSPA2
14
heat shock protein family A (Hsp70)
ENSG00000126803





member 2



RAB1B
11
RAB1B, member RAS oncogene family
ENSG00000174903



CAT
11
catalase
ENSG00000121691



CTSD
11
cathepsin D
ENSG00000117984



SERPINB3
18
serpin family B member 3
ENSG00000057149



UBA52
19
ubiquitin A-52 residue ribosomal protein
ENSG00000221983





fusion product 1



EEF1A2
20
eukaryotic translation elongation factor 1
ENSG00000101210





alpha 2



DSC1
18
desmocollin 1
ENSG00000134765



KRT25
17
keratin 25
ENSG00000204897



POF1B
X
premature ovarian failure, 1B
ENSG00000124429



KRT12
17
keratin 12
ENSG00000187242



KRT36
17
keratin 36
ENSG00000126337



S100A9
1
S100 calcium binding protein A9
ENSG00000163220



PKM
15
pyruvate kinase, muscle
ENSG00000067225



S100A7
1
S100 calcium binding protein A7
ENSG00000143556



HAL
12
histidine ammonia-lyase
ENSG00000084110



CALML5
10
calmodulin like 5
ENSG00000178372



PIP
7
prolactin induced protein
ENSG00000159763



LGALS7
19
galectin 7
ENSG00000205076



LGALS7B
19
galectin 7B
ENSG00000178934



HSPB1
7
heat shock protein family B (small)
ENSG00000106211





member 1



RAB1A
2
RAB1A, member RAS oncogene family
ENSG00000138069



GAPDHS
19
glyceraldehyde-3-phosphate
ENSG00000105679





dehydrogenase, spermatogenic


X
ANXA2
15
annexin A2
ENSG00000182718


X
VIM
10
vimentin
ENSG00000026025


X
KPRP
1
keratinocyte proline rich protein
ENSG00000203786


X
KRT84
12
keratin 84
ENSG00000161849


X
GFAP
17
glial fibrillary acidic protein
ENSG00000131095


X
EIF4A2
3
eukaryotic translation initiation factor
ENSG00000156976





4A2


X
SERPINB12
18
serpin family B member 12
ENSG00000166634


X
HSPA5
9
heat shock protein family A (Hsp70)
ENSG00000044574





member 5


X
KRT28
17
keratin 28
ENSG00000173908


X
KRT73
12
keratin 73
ENSG00000186049


X
KRT19
17
keratin 19
ENSG00000171345


X
CASP14
19
caspase 14
ENSG00000105141


X
EIF4A1
17
eukaryotic translation initiation factor
ENSG00000161960





4A1


X
DSC3
18
desmocollin 3
ENSG00000134762


X
KRT72
12
keratin 72
ENSG00000170486


X
KRT24
17
keratin 24
ENSG00000167916


X
KRT23
17
keratin 23
ENSG00000108244


X
ARG1
6
arginase 1
ENSG00000118520


X
TGM3
20
transglutaminase 3
ENSG00000125780


X
KRT71
12
keratin 71
ENSG00000139648


X
ENO1
1
enolase 1
ENSG00000074800


X
KRT18
12
keratin 18
ENSG00000111057


X
LYZ
12
lysozyme
ENSG00000090382


X
TGM1
14
transglutaminase 1
ENSG00000092295


X
DCD
12
dermcidin
ENSG00000161634


X
PRDX1
1
peroxiredoxin 1
ENSG00000117450


X
EEF1A1
6
eukaryotic translation elongation factor 1
ENSG00000156508





alpha 1


X
GAPDH
12
glyceraldehyde-3-phosphate
ENSG00000111640





dehydrogenase


X
JUP
17
junction plakoglobin
ENSG00000173801


X
PRDX2
19
peroxiredoxin 2
ENSG00000167815


X
KRT27
17
keratin 27
ENSG00000171446


X
KRT7
12
keratin 7
ENSG00000135480


X
KRT15
17
keratin 15
ENSG00000171346


X
FLG2
1
filaggrin family member 2
ENSG00000143520


X
KRT80
12
keratin 80
ENSG00000167767


X
KRT75
12
keratin 75
ENSG00000170454


X
HSPA1L
6
heat shock protein family A (Hsp70)
ENSG00000204390





member 1 like


X
KRT6A
12
keratin 6A
ENSG00000205420


X
HRNR
1
hornerin
ENSG00000197915


X
HSPA8
11
heat shock protein family A (Hsp70)
ENSG00000109971





member 8


X
DSP
6
desmoplakin
ENSG00000096696


X
KRT76
12
keratin 76
ENSG00000185069


X
KRT13
17
keratin 13
ENSG00000171401


X
DSG1
18
desmoglein 1
ENSG00000134760


X
KRT79
12
keratin 79
ENSG00000185640


X
ACTB
7
actin beta
ENSG00000075624


X
ACTG1
17
actin gamma 1
ENSG00000184009


X
KRT17
17
keratin 17
ENSG00000128422


X
KRT78
12
keratin 78
ENSG00000170423


X
KRT8
12
keratin 8
ENSG00000170421


X
KRT3
12
keratin 3
ENSG00000186442


X
KRT4
12
keratin 4
ENSG00000170477


X
KRT6C
12
keratin 6C
ENSG00000170465


X
KRT16
17
keratin 16
ENSG00000186832


X
KRT77
12
keratin 77
ENSG00000189182


X
KRT5
12
keratin 5
ENSG00000186081


X
KRT14
17
keratin 14
ENSG00000186847


X
KRT6B
12
keratin 6B
ENSG00000185479


X
KRT9
17
keratin 9
ENSG00000171403


X
KRT2
12
keratin 2
ENSG00000172867


X
KRT1
12
keratin 1
ENSG00000167768


X
KRT10
17
keratin 10
ENSG00000186395









Example 46
Exemplary GVP Detectable in Hair Samples

An exemplary set of GVPs that can be used in methods and systems herein described as well as in related databases is reported herein. In particular, the exemplary set of GVP comprises genes validated as proteomically detectable in hair samples of a Homo Sapiens which can be used in methods and systems to detect a genetic variation and/or perform a genetic variation analysis, as well as in related databases, in accordance with the various aspects of the present disclosure.


Specifically, Table 11 shows a list of exemplary GVP detectable in hair samples of human beings. The fields in Table 11 are the chromosome where the gene is located (CHR), the gene name (gene name), mutation identifier (mutation ID), the sequence of the corresponding mutated peptide (mutated peptide (GVP)), the related sequence identifier in the sequence listing of the instant disclosure (SEQ ID NO), and the subpopulations including all populations (ALL), Non-Finnish European subpopulation (NFE), African subpopulation (AFR), East Asian subpopulation (EAS), South Asian subpopulation (SAS), and Latino subpopulation (AMR).


The exemplary GVPs of Table 11 can be therefore be used in methods and systems of the disclosure wherein the sample comprises hair samples from human beings.









TABLE 11







Exemplary GYP detectable in hair samples


















gene
mutation

SEQ








CHR
name
ID
mutated peptide (GYP)
ID NO
ALL
NFE
AFR
EAS
SAS
AMR





17
KRT33
rs617416
AAPAVDLNR
146


X






B
63













 8
RIDA
rs146537
AAYQVAVLPK
147










203













17
KRTAP
rs149188
ACCQTSFCGFR
148



X

X



1-1
249













21
KRTAP
rs713213
ACQPTCYQR
149
X
X
X

X
X



11-1
55













21
KRTAP
rs713213
ACQPTCYQRTSCVSNPCQ
150
X
X
X

X
X



11-1
55
VTCSR












17
KRT32
rs207156
ADLEAQVEYLKEELMCL
151


X







1
K












17
KRT32
rs207156
ADLEAQVEYLKEELMCL
152


X







1
KK












12
KRT82
rs377470
ADLETNTEALVQEIDFLK
153










048













17
KRT32
rs260495
AELERQNQEYQVLLDVR
154
X
X








6













17
KRT32
rs260495
AELERQNQEYQVLLDVR
155
X
X








6
AR












12
KRT81
rs798978
AFRCISACGPRPGR
156


X
X






79













12
KRT81
rs202205
AFSCISACGPQPGR
157










489













12
KRT81
rs202205
AFSCISACGPQPGRC
158










489













 2
POTEF
rs762202
AGFASDDAPR
159










335













12
KRT6B
rs144860
AGGSYGFGGAR
160
X
X
X
X
X
X




693













12
KRT85
rs616300
AGSCGHSF
161




X





04













12
KRT85
rs616300
AGSCGHSFGYR
162




X





04













12
KRT6A
rs115403
AIGGGLSSVGGGSSTIKY
163


X


X




01
STTSSSSR












 1
S100A3
rs360227
AKPLEQAVAAIVCTFQEY
164
X
X
X

X
X




42
AGR












 6
HIST1
rs757147
ALAVAGYDVEKNNSR
165









H1E
711













17
GSDM 
rs721293
ALETLQER
166





X



A
8













19
MYH14
rs680446
ALRAELEALLSSKDDIGK
167











SVHELER












12
KRT81
rs207158
APYRGISCYRGLTGGFGS
168
X
X
X
X
X
X




8
HSVCR












17
KRT32
rs110789
AQMQCMITNVEAQLAEI
169
X

X
X
X
X




93
QADLERQNQEYQVLLDV












R












17
KRT32
rs260495
AQMQCMITNVEAQLAEI
170
X
X








6
RAELERQNQEYQVLLDV












17
KRT40
rs806473
ARLEGEINMYR
171
X
X

X
X
X




3













17
KRT40
rs116498
ARLEGEINMYR
172
X
X

X
X
X




34













17
KRT32
rs207156
ARLEGEINMYR
173
X
X
X
X
X
X




3













17
KRT32
rs260495
ARYSSQLAQMQCMITNV
174
X
X








6
EAQLAEIRAELERQNQEY












QVLLDVR












17
KRT34
rs617406
ARYSSQLSQVQSLITNVE
175










68
SQLAEIRCDLEWQNQEY












QVLLDVR












17
KRT34
rs207159
ARYSSQLSQVQSLITNVE
176
X
X
X
X
X
X




9
SQLAEIRCDLEWQNQEY












QVLLDVR












17
KRT37
rs991672
ASAASMCLLANVAHANR
177
X

X
X

X




4













17
KRT33
rs129375
ATQTEELNKQVVSSSEQL
178
X
X
X
X
X
X



A
19
QSYQVEIIELRR












12
KRT81
rs476178
ATVIRHGETLCR
179










6













13
TUBA3
rs362150
AVFVDLEPTVLDEVR
180









C
77













13
TUBA3
rs362150
AVFVDLEPTVLDEVRTGT
181









C
77
YR












21
KRTAP
rs713213
CCEPTACQPTCYQRTSCV
182
X
X
X

X
X



11-1
55
SNPCQVTCSR












12
KRT82
rs617305
CCQINIEPIFEGYISALRR
183










90













17
KRTAP
rs129386
CCQNTCCRTTCCQPTCVT
184
X
X
X
X
X




9-6
92
SCCQPSCCSTPCCQPICCG












SSCCGQTSCGSSCGQSSS












CAPVYCRR












17
KRTAP
rs238824
CCQPCCHPTCYQTTCFRT
185









9-1

TCCQPTCCQPTCCR












17
KRTAP
rs389784
CCQPTCCRPSCGQTTCCR
186









4-2














17
KRTAP
rs720768
CCQPTCYRPSCCVSSCCR
187



X





4-9
5
PQCCQPVCCQPTCCR












17
KRTAP
rs739831
CCRSSCCPSCCQTTCCR
188


X






4-6
72













17
KRT34
rs199674
CDLERQNQEYQVLLDVC
189










249
AR












17
KRT34
rs617406
CDLEWQNQEYQVLLDVR
190










68













12
KRT83
rs285766
CECCQSNLEPLFAGYIET
191
X
X
X
X
X
X




3
LRR












17
KRT40
rs178430
CEDGVSTSNEKETMQFL
192
X
X
X
X
X
X




15
NDR












17
KRT39
rs112557
CEPSPWTFCK
193


X







906













21
KRTAP
rs713213
CEPTACQPTCYQR
194
X
X
X

X
X



11-1
55













17
KRTAP
rs626233
CETSCYQPR
195


X
X

X



1-5
75













17
KRTAP
rs389784
CFRPQCCQSVCCQPTCCR
196









4-2

PSCGQTTCCR












17
KRTAP
rs116553
CGQVLCQETCCRPSCCQT
197



X





4-7
10
TCCR












17
KRTAP
rs383835
CGSVCSDQGCSQVLCQE
198



X





4-7

TCCRPSCCQTTCCR












17
KRT35
rs189378
CHYETLVENNRR
199










138













12
KRT83
rs285767
CKPCGQLNTTCGGGSCG
200










1
QGRY












17
KRT33
rs754250
CQLGDHLNVEVDAAPTV
201









A
148
DLNQVLNETR












17
KRT33
rs617416
CQLGDRLNVEVDAAPAV
202


X






B
63
DLNR












17
KRT33
rs617416
CQLGDRLNVEVDAAPAV
203


X






B
63
DLNRVLNETR












17
KRT34
rs139103
CQLGDRLNVEVDTAPTV
204










580
DLNQVLNETR












12
KRT83
rs140635
CQNSKLEAAVAQSEQQS
205










030
EAALSDAR












17
KRTAP
rs129386
CQNTCCRTTCCQPTCVTS
206
X
X
X
X
X




9-6
92
CCQPSCCSTPCCQPICCGS












SCCGQTSCGSSCGQSSSC












APVYCR












17
KRTAP
rs129438
CQPSCCETSCCQPSCCET
207









1-5
24
SCCQPSCWQISSCGTGCG












IGGGISYGQEGSSGAVST












R












17
KRTAP
rs626233
CQPSCCETSCYQPR
208


X
X

X



1-5
75













17
KRTAP
rs389784
CQSVCCQPTCCRPSCGQT
209









4-2

TCCR












17
KRTAP
rs149188
CQTSFCGFR
210



X

X



1-1
249













17
KRTAP
rs620672
CQTTCCRTTCCRPSCCVS
211

X







4-2
92
SCCRPQCCQSVCCQPSCC












SPSCCQTTCCR












17
KRTAP
rs116504
CQTTCCRTTCYRPSCCVS
212



X





4-7
84
SCCRPQCCQSVCCQPTCC












RPSCCETTCCHPR












17
KRT32
rs728300
CQYEAMVEANHR
213
X
X
X
X
X
X



46














17
KRT40
rs721957
CQYETVLANNRR
214











17
KRTAP
rs745728
CRPQCCQTICCR
215



X





4-4
64













17
KRTAP
rs626228
CRTGCGIGGGIGYGQEGS
216
X

X
X

X



1-3
49
SGAVSTR












17
KRTAP
rs116553
CSDQGCGQVLCQETCCR
217



X





4-7
10
PSCCQTTCCR












17
KRT38
rs138667
CTVNALEVK
218










284













17
KRT38
rs138667
CTVNALEVKR
219










284













17
KRT40
rs178430
DGVSTSNEKETMQFLND
220
X
X
X
X
X
X




15
RLASYLEKVR












12
KRT81
rs141587
DLNMDCIIDEIK
221










304













12
KRT81
rs141587
DLNMDCIIDEIKAQYDDI
222










304
VTR












12
KRT83
rs285246
DLNMDCMVAEIK
223
X
X
X
X
X
X




4













12
KRT83
rs285246
DLNMDCMVAEIKAQYD
224
X
X
X
X
X
X




4
DIATR












 2
NEU2
rs223339
DLTDAAIGPAYREWSTFA
225
X

X

X
X




1
VGPGHCLQLNDR












 2
NEU2
rs223339
DLTDTAIGPAYR
226










0













12
KRT84
rs951773
DMARQLREYQELMNAK
227











LGLDIEIATYR












 1
SFN
rs149812
DMPPTNPIR
228










347













17
KRT33
rs124506
DNAELKNLIR
229

X


X




B
21













17
KRT33
rs124506
DNAELKNLIRER
230

X


X




B
21













17
KRT31
rs650362
DNVELENLIR
231
X
X








7













17
KRT31
rs650362
DNVELENLIRER
232
X
X








7













17
KRT32
rs169669
DSLENMLTESEAR
233










29













17
KRT34
rs148645
DSLENTLTESEAHYSSQL
234










199
SQMQSLITNVESQLAEIR












CDLER












17
KRT34
rs148645
DSLENTLTESEAHYSSQL
235










199
SQMQSLITNVESQLAEIR












CDLERQNQEYQVLLDVR












17
KRT34
rs617406
DSLENTLTESEAHYSSQL
236










68
SQVQSLITNVESQLAEIRC












DLEW












17
KRT32
rs110789
DSLENTLTESEARYSSQL
237
X

X
X
X
X




93
AQMQCMITNVEAQLAEI












QADLER












17
KRT32
rs110789
AQMQCMITNVEAQLAEI
238
X

X
X
X
X




93
DSLENTLTESEARYSSQL












QADLERQNQEYQVLLDV












R












17
KRT32
rs260495
DSLENTLTESEARYSSQL
239
X
X








6
AQMQCMITNVEAQLAEI












RAELERQNQEYQVLLDV












R












17
KRT39
rs178430
DSQECILMETEAR
240
X
X
X
X
X
X




21













12
KRT82
rs377470
DVDTAFLMKADLETNTE
241










048
ALVQEIDFLK












12
KRT85
rs112554
EAECVEANSGR
242










450













12
KRT85
rs112554
EAECVEANSGRLAS
248243










450













12
KRT85
rs112554
EAECVEANSGRLASELN
244










450
HVQEVLEGYKK












12
KRT85
rs112554
EAECVEANSGRLASELN
245










450
HVQEVLEGYKKK












17
KRT32
rs110789
EAQLAEIQADLERQNQE
246
X

X
X
X
X




93
YQVLLDVR












12
KRT86
rs139895
EEINELNCMIQR
247










699













20
TGM3
rs604806
EEYVQEDAGILFVGSTNR
248


X







6













17
KRT39
rs721325
EHCSACGPLSQILVK
249
X
X


X
X




6













17
KRT32
rs117304
EIMQFLNDR
250










287













17
KRT32
rs117304
EIMQFLNDRLASYLTR
251










287













17
KRT32
rs260495
EIRAELERQNQEYQVLLD
252
X
X








6
VR












12
KRT82
rs143454
ELDVDGIIAEIKAQYDDIT
253










001
SR












12
KRT82
rs617305
ELDVDSIIAEIK
254










89













12
KRT82
rs617305
ELDVDSIIAEIKAQYDDIA
255










89
SR












 1
SFN
rs777552
EMPPSNPIR
256










55













16
PPL
rs203791
ENLQLETR
257


X

X





2













21
KRTAP
rs713213
EPTACQPTCYQR
258
X
X
X
X
X




11-1
55













17
KRT31
rs650362
ERDNVELENLIR
259
X
X








7













17
KRT31
rs650362
ERDNVELENLIRER
260
X
X








7













17
KRT33
rs347718
ESQLAEIHSDLERQNQEY
261









B
86
QVLLDVR












21
KRTAP
rs713213
ETCCEPTACQPTCYQR
262
X
X
X

X
X



11-1
55













17
KRTAP
rs149483
ETCCHPSCCETTCCR
263


X






4-9
591













17
KRTAP
rs113376
ETCCHPSCCETTCCR
264
X

X
X

X



4-11
601













17
KRT33
rs140696
ETMQFLNDCLASYLEK
265









A
036













17
KRT33
rs140696
ETMQFLNDCLASYLEKV
266









A
036
R












17
KRT33
rs140696
ETMQFLNDCLASYLEKV
267









A
036
RQLERDNAELENLIR












17
KRT34
rs112570
EVEQWFATQTEELNKQV
268










296
VSSSEQLQSCQVEIIELR












17
KRT34
rs112570
EVEQWFATQTEELNKQV
269










296
VSSSEQLQSCQVEIIELRR












17
KRT33
rs129375
EVEQWFATQTEELNKQV
270
X
X
X
X
X
X



A
19
VSSSEQLQSYQVEIIE












17
KRT33
rs129375
EVEQWFATQTEELNKQV
271
X
X
X
X
X
X



A
19
VSSSEQLQSYQVEIIELR












17
KRT33
rs129375
EVEQWFATQTEELNKQV
272
X
X
X
X
X
X



A
19
VSSSEQLQSYQVEIIELRR












17
KRT34
rs777791
EVEQWFATQTEK
273










92













17
KRT34
rs777791
EVEQWFATQTEKLNK
274










92













17
KRT34
rs777791
EVEQWFATQTEKLNKQV
275










92
VSSSEQLQSCQAEIIELRR












20
TGM3
rs149720
FDILPSQSGTK
276










612













12
KRT86
rs587172
FLEQQNKLLETKLPFYQN
277
X
X

X
X
X




66
R












12
KRT83
rs285766
FLEQQNKLLETKLQFYQ
278
X
X
X
X
X
X




3
NCECCQSNLEPLFAGYIE












TLRR












12
KRT82
rs377470
FLMKADLETNTEALVQEI
279










048
DFLKSLYEEEICLLQSQIS












ETSVIVK












17
KRTAP
rs626228
FPSFSTSGTCSSSCCQPSC
280
X

X
X

X



1-3
49
CETSCCQPSCCQTSSCRT












GCGIGGGIGYGQEGSSGA












VSTR












12
KRT81
rs798978
FRCISACGPRPGR
281


X
X






79













12
KRT81
rs798978
FRCISACGPRPGRCCITAA
282


X
X






79
PYR












17
KRT39
rs142154
FSLDDCNWYGEGINSNE
283










718
KETMQILNER












17
KRT39
rs778437
FSLDDCSR
284










878













17
KRTAP
rs350240
FSTGGTCDSSCCQPSCCE
285


X






1-1
33
TSCCQPSCYQTSSYGTGC












GIGGGIGYGQEGSSGAVS












TR












12
KRT82
rs173226
GAFLYDPCGVSTPVLSTG
286


X

X





3
VLR












12
KRT82
rs265865
GAFLYEPCGVSMPVLSTG
287
X

X
X
X
X




8
VLR












17
KRTAP
rs142863
GCGTGGGIGYGQEGSSG
288









1-3
014
AVSTR












11
TRIM2
rs116041
GCPSLMR
289



X

X



9
69













21
KRTAP
rs380401
GCQEICWEPTSCQTSYVE
290
X
X
X
X
X
X



13-2
0
SRPCQTSCYRPR












21
KRTAP
rs380401
GCQEICWEPTSCQTSYVE
291
X
X
X
X
X
X



13-2
0
SRPCQTSCYRPRT












21
KRTAP
rs117415
GCRPSCYGGYGFSGFY
292









19-5
039













12
KRT81
rs207158
GFGSHSVCR
293
X
X
X
X
X
X




8













17
KRTAP
rs626228
GFPSFSTSGTCSSSCCQPS
294
X

X
X

X



1-3
49
CCETSCCQPSCCQTSSCR












TGCGIGGGIGYGQEGSSG












AVSTR












21
KRTAP
rs617483
GFSYPSNLVYSTDLCSPSI
295









13-2
17
CQLGSSLYR












12
KRT81
rs207158
GGFGSHSVCR
296
X
X
X
X
X
X




8













12
KRT2
rs263404
GGGFGGGSGFGGGSGFS
297
X
X
X
X

X




1
GGGFGGGGFGGGR












12
KRT2
rs764122
GGGFGGGSSFGGGSGFSG
298










02
GGFSGGGFGGGR












12
KRT84
rs795397
GGPDFGYR
299










00













 1
SELEN
rs727101
GGPVQVLEDK
300









BP1
12













 1
SELEN
rs727101
GGPVQVLEDKELK
301









BP1
12













17
KRTAP
rs349771
GGVSCHTTCYRPTCVISS
302


X
X

X



4-11

CPRPLC












17
KRTAP
rs349771
GGVSCHTTCYRPTCVISS
303


X
X

X



4-11

CPRPLCCASSC












17
KRTAP
rs349771
GGVSCHTTCYRPTCVISS
304


X
X

X



4-11

CPRPLCCASSCC












 5
HEXB
rs108058
GILVDTSR
305
X
X

X
X
X




90













12
KRT83
rs285767
GLCKPCGQLNTTCGGGS
306










1
CGQGRY












 1
PKP1
rs142096
GLPQIAHLLQSGNSDVVR
307










411













12
KRT82
rs201747
GLQALGCLGSR
308










652













12
KRT81
rs207158
GLTGGFGSHSVCR
309
X
X
X
X
X
X




8













12
KRT81
rs207158
GLTGGFGSHSVCRG
310
X
X
X
X
X
X




8













12
KRT81
rs207158
GLTGGFGSHSVCRGFR
311
X
X
X
X
X
X




8










12
KRT81
rs207158
GLTGGFGSHSVCRGFRA
312
X
X
X
X
X
X







8










12
KRT6B
rs285383
GPGFPVCPPGGIQEVTVN
313
X
X
X
X
X
X




43
QNLLTPLNLQIDPAIQR












17
KRTAP
rs145881
GQEGSSGAVSTCIR
314









1-5
217













12
KRT83
rs285767
GQLNTTCGGGSCGQGRY
315










1













 6
DSP
rs692906
GQSEADSDKNATILELR
316
X
X
X
X
X
X




9













17
KRTAP
rs116553
GQVLCQETCCRPSCCQTT
317



X





4-7
10
CCR












17
KRTAP
rs140898
GRVSCHTTCYRPTCVISS
318


X
X

X



4-11
464
CPRPVCCASSCC












12
KRT86
rs572429
GSCGRSFGYHSGGVCGPS
319










51
PPCITTVSVNESLLTPLNL












EIDPNAQCVKQEEK












12
KRT81
rs207158
GSHSVCR
320
X
X
X
X
X
X




8













17
KRTAP
rs116553
GSVCSDQGCGQDLCQET
321



X





4-7
10
CCRPSCCQTTCCR












17
KRTAP
rs116553
GSVCSDQGCGQVLCQET
322



X





4-7
10
CCRPSCCQTTCCR












18
SERPI
rs145555
GVALSNVVHK
323
X

X
X
X
X



NB5
5













18
SERPI
rs145555
GVALSNVVHKVCLEITED
324
X

X
X
X
X



NB5
5
GGDSIEVPGAR












12
KRT86
rs566778
GVDCAYLR
325










56













11
PKP3
rs200371
GVGGAVPGAVLEPVAPA
326

X








913
PSVR












17
KRTAP
rs349771
GVSCHTTCYRPTCVISSC
327


X
X

X



4-11














17
KRTAP
rs349771
GVSCHTTCYRPTCVISSC
328


X
X

X



4-11

PR












17
KRTAP
rs349771
GVSCHTTCYRPTCVISSC
329


X
X

X



4-11

PRPL












17
KRTAP
rs349771
GVSCHTTCYRPTCVISSC
330


X
X

X



4-11

PRPLCC












17
KRTAP
rs349771
GVSCHTTCYRPTCVISSC
331


X
X

X




4-11
PRPLCCA












17
KRTAP
rs349771
GVSCHTTCYRPTCVISSC
332


X
X

X



4-11

PRPLCCASS












17
KRTAP
rs349771
GVSCHTTCYRPTCVISSC
333


X
X

X



4-11

PRPLCCASSCC












12
KRT82
rs265865
GVSMPVLSTGVLR
334
X

X
X
X
X




8













11
HEPHL
rs194578
HFCTDPDSVDKK
335









1
3













11
HEPHL
rs194578
HFCTDPDSVDKKDAVFQ
336









1
3
R












 7
ATG9B
rs780489
HFSELPHELR
337
X
X
X
X

X




3













12
KRT81
rs476178
HGETLCR
338










6













12
KRT83
rs200128
HGETLCR
339










355













12
KRT83
rs285246
HISDTSVVVKLDNSRDLN
340
X
X
X
X
X
X




4
MDCMVAEIKAQYDDIAT












R












17
KRT33
rs148752
HNAELENLIR
341









A
041













17
KRT33
rs148752
HNAELENLIRER
342









A
041













 6
DSP
rs140965
HQNQNTIQELLQNCSDYL
343










835
MR












12
KRT85
rs616300
HSFGYR
344




X





04













12
KRT86
rs572429
HSGGVCGPSPPCITTVSV
345










51
NESLLTPLNLEIDPNAQC












VK












12
KRT86
rs572429
HSGGVCGPSPPCITTVSV
346










51
NESLLTPLNLEIDPNAQC












VKQEEKEQIK












17
KRT32
rs144111
HTVNTLEIELQAQHSLR
347










267













17
KRT32
rs144111
HTVNTLEIELQAQHSLRD
348










267
SLENTLTESEAR












17
BLMH
rs105056
HVPEEVLAVLEQEPIVLP
349
X
X
X
X
X
X




5
AWDPMGALA












12
KRT85
rs616300
IAVGGFRAGSCGHSFGYR
350




X





04













12
KRT85
rs139493
IAVGGSRAGSCGR
351










548













 2
IL1F10
rs676127
ICTLPNR
352



X






6













20
TGM3
rs114998
IDVPTLEPK
353










364













20
TGM3
rs214830
IDVPTLGPKER
354











14
LGALS
rs11125
IHVLVEPDHFK
355
X
X


X




3














17
KRT40
rs990830
ILCMKAENSR
356
X
X

X
X
X




4













17
KRT32
rs207156
ILDDLTLCKADLEAQVEY
357


X







1
LKEELMCLK












17
KRT32
rs207156
ILDDLTLCKADLEAQVEY
358


X







1
LKEELMCLKK












17
KRT34
rs566233
ILNELTLCK
359










643













17
KRT34
rs566233
ILNELTLCKSDLESQVESL











643
REELICLK
360











17
KRT34
rs566233
ILNELTLCKSDLESQVESL
361










643
REELICLKK












12
KRT81
rs202205
ISACGPQPGR
362










489













12
KRT83
rs285246
ISDTSVVVKLDNSRDLN
363
X
X
X
X
X
X




4
MDCMVAEIKAQYDDIAT












R












21
KRTAP
rs963684
ISNPCSTTYSRPLTFVSSG
364
X
X

X
X
X



11-1
5
SQPLGGISSVCQPVGGIST












VCQPVGGVSTVCQPACG












VSR












 6
DSP
rs749679
ITNLTQQLEQAPIVK
365










496













 6
DSP
rs749679
ITNLTQQLEQAPIVKK
366










496













 6
HIST1
rs757147
KALAVAGYDVEKNNSR
367









HIE
711













 6
HIST1
rs200744
KATGAAIPK
368









HIE
473













12
KRT83
rs766508
KKYEEEVALQATAENEF
369










559
VALKK












12
KRT83
rs285246
KLDNSRDLNMDCMVAEI
370
X
X
X
X
X
X




4
KAQYDDIATR












17
KRT35
rs761727
KNHEEEVNSLHCQLGDR
371










354













12
KRT83
rs285767
KPCGQLNTTCGGGSCGQ
372










1
GRY












12
KRT81
rs751670
KSDLEANVDALIQEIDFL
373










289
R












12
KRT81
rs751670
KSDLEANVDALIQEIDFL
374










289
RR












12
KRT86
rs111429
KSDLEANVEALIQEIDFL
375










470
RWLYEEEIRVLQSHISDT












SVVVK












12
KRT84
rs161393
KVQFLEQQNKLLETK
376
X
X

X
X
X




1













12
KRT82
rs179163
KYEEELSLRPCVQNEFVA
377










4
LKK












12
KRT83
rs766508
KYEEEVALQATAENEFV
378










559
ALKK












 5
HEXB
rs774999
LAPGTVVEVWKDSAYPE
379










35
ELSR












21
KRTAP
rs617459
LASCGSLLYRPTCSR
380
X

X

X




10-12
11













17
KRT34
rs201477
LASDDFRSKYQMEQSLR
381










948













17
KRT34
rs372070
LASDNFR
382










920













17
KRT34
rs372070
LASDNFRSKYQTEQSLR
383










920













17
KRT40
rs140634
LASYLEKVH
384










473













17
KRT13
rs989136
LAVDDFR
385


X







1













 1
SEN
rs787079
LAYQEAMDISK
386










84













 1
SEN
rs787079
LAYQEAMDISKK
387










84













12
KRT83
rs285767
LCKPCGQLNTTCGGGSC
388










1
GQGRY












12
TXNR
rs713419
LCLSPPASDSR
389
X
X
X

X
X



D1
3













12
KRT3
rs388795
LDLDSIIAEVGA
390


X
X
X
X




4













14
LGALS
rs101483
LDNNWGKEER
391


X






3
71













12
KRT81
rs141587
LDNSRDLNMDCIIDEIKA
392
X
X
X
X
X
X




304
QYDDIVTR












12
KRT83
rs285246
LDNSRDLNMDCMVAEIK
393
X
X
X
X
X
X




4













12
KRT83
rs285246
LDNSRDLNMDCMVAEIK
394










4
AQYDDIATR












12
KRT83
rs140635
LEAAVAQSEQQSEAALS
395










030
DAR












12
KRT83
rs140635
LEAAVAQSEQQSEAALS
396










030
DARCK












17
KRT32
rs207156
LEGEINMYR
397
X
X
X
X
X
X




3













17
KRT31
rs650362
LERDNVELENLIR
398
X
X








7













17
KRT39
rs112120
LESEITTYR
399










285













 1
VSIG8
rs626244
LGCPYILDPEDYGPNGLD
400


X







68
IEWMQVNSDPAHHR












17
KRT33
rs347718
LITNVESQLAEIHSDLER
401









B
86













17
KRT37
rs169668
LLDDVTLAK
402
X
X
X
X
X
X




11













17
KRT37
rs169668
LLDDVTLAKADLEAQQE
403
X
X
X
X
X
X




11
SLKEEQLSLKSNHEQEVK












12
KRT86
rs587172
LLETKLPFYQNR
404
X
X

X
X
X




66













12
KRT86
rs587172
LLETKLPFYQNRECCQSN
405
X
X

X
X
X




66
LEPLFEGYIETLRR












17
KRT32
rs146792
LNIEVDTAPPVDLTR
406










525













12
KRT81
rs141587
LNMDCIIDEIKAQYDDIV
407










304
TR












12
KRT83
rs285767
LNTTCGGGSCGQGRY
408










1













17
KRT33
rs617416
LNVEVDAAPAVDLNR
409


X






B
63













17
KRT31
rs112544
LNVEVDAAPTVDLNRVL
410










857
NETRSQYEVLVETNRR












17
KRT36
rs757906
LNVEVDGAPPVDLNKILE
411



X

X




52
DMR












12
KRT86
rs587172
LPFYQNR
412
X
X

X
X
X




66













12
KRT86
rs587172
LPFYQNRECCQSNLEPLF
413
X
X

X
X
X




66
EGYIETLRR












17
KRT32
rs374478
LPTTFRPASCLSKTYLSSS
414
X
X
X
X
X
X




6
CRAASGISGSMGPGSWY












SEGAFNGNEKETMQFLN












DR












12
KRT83
rs285766
LQFYQNCECCQSNLEPLF
415
X
X
X
X
X
X




3
AGYIETLR












12
KRT83
rs285766
LQFYQNCECCQSNLEPLF
416
X
X
X
X
X
X




3
AGYIETLRR












16
PPL
rs203791
LQLERENLQLETR
417


X

X





2













 6
DSP
rs207629
LQRVQCDLQK
418
X

X
X
X
X




9













17
KRT33
rs129375
LQSYQVEIIELRRTVNAL
419
X
X
X
X
X
X



A
19
EIELQAQHNLR












17
KRTAP
rs349771
LRPVCGGVSCHTT
420


X
X

X



4-11














12
TXNR
rs713419
LSPPASDSR
421
X
X
X

X
X



D1
3













12
KRT85
rs771843
LSSRSSLSHTQDVDCAYL
422










00
RKSDLEANVEALVEESSF












LR












12
KRT83
rs140635
LTAEVENAKCQNSKLEA
423










030
AVAQSEQQSEAALSDAR












12
KRT81
rs207158
LTGGFGSHSVCR
424
X
X
X
X
X
X




8













12
KRT81
rs207158
LTGGFGSHSVCRGFR
425
X
X
X
X
X
X




8













 1
SELEN
rs727101
LTGQLFLGGSIVKGGPVQ
426









BP1
12
VLEDKELK












 6
DSP
rs413028
LTVNSAIAR
427










85













19
PGLS
rs183992
LVPFNHAESTYGLYR
428










141













17
KRT34
rs201477
LVVNIDNAKLASDDFRSK
429










948
YQMEQSLR












17
KRT34
rs372070
LVVNIDNAKLASDNFR
430










920













17
KRT34
rs372070
LVVNIDNAKLASDNFRSK
431










920













17
KRT34
rs372070
LVVNIDNAKLASDNFRSK
432










920
YQTEQSLR












17
KRT33
rs145389
LVVRIDNAK
433









A
769













17
KRT33
rs145389
LVVRIDNAKLASDDFR
434









A
769













17
KRT33
rs145389
LVVRIDNAKLASDDFRTK
435









A
769













12
KRT83
rs285767
LVVSTGLCKPCGQLNTTC
436










1
GGGSCGQGRY












 1
S100A3
rs360227
MAKPLEQAVAAIVCTFQ
437
X
X
X

X
X




42
EYAGR












12
KRT83
rs285246
MDCMVAEIK
438
X
X
X
X
X
X




4













12
KRT83
rs285246
MDCMVAEIKAQYDDIAT
439
X
X
X
X
X
X




4
R












17
KRT39
rs178430
MRDSQECILMETEAR
440
X
X
X
X
X
X




21













12
KRT83
rs285246
MVAEIKAQYDDIATR
441
X
X
X
X
X
X




4













22
COMT
rs4680
MVDFAGMKDKVTLVVG
442
X

X
X
X
X





ASQDIIPQLK












17
KRT36
rs230135
MVNALEIELQAQHSMR
443


X
X






4













17
KRTAP
rs149483
MVSSCCGSVCSDQGCGQ
444


X






4-9
591
DLCQETCCHPSCCETTCC












R












17
KRTAP
rs116553
MVSSCCGSVCSDQGCGQ
445



X





4-7
10
DLCQETCCRPSCCQTTCC












R












17
KRTAP
rs383835
MVSSCCGSVCSDQGCSQ
446



X





4-7

VLCQETCCRPSCCQTTCC












RTTCYRPSCCVSS












17
KRTAP
rs749779
MVSSCCGSVSSEQSCGLE
447


X
X





4-5
892
NCCCPSCCQTTCCR












17
KRT32
rs207156
MVVNTDNAK
448



X

X




0













17
KRT32
rs207156
MVVNTDNAKLAADDFR
449



X

X




0













17
KRTAP
rs749779
NCCCPSCCQTTCCR
450


X
X





4-5
892













17
KRT40
rs151006
NEKETMQFLNDRLANYL
451
X
X

X
X
X




8
EKVR












17
KRT40
rs201002
NHEEEVNLLHEQLGDR
452
X
X

X
X
X




7













17
KRT35
rs761727
NHEEEVNSLHCQLGDR
453










354













17
KRT35
rs761727
NHEEEVNSLHCQLGDRL
454










354
NVEVDAAPPVDLNRVLE












EMR












12
KRT7
rs658087
NKYEDEINR
455










0













 1
PKP1
rs569372
NLSSADAGHQTMR
456










122













12
KRT83
rs285767
NLVVSTGLCKPCGQLNT
457










1
TCGGGSCGQGRY












11
TRIM2
rs116041
NNPGCPSLMR
458



X

X



9
69













14
HSPA2
rs140108
NQVAVNPTNTIFDAKR
459










798













17
KRT31
rs650362
NVELENLIR
460
X
X








7













17
KRT37
rs991672
NVFVSPIDVGCQPVAEAS
461
X

X
X

X




4
AASMCLLANVAHANR












20
TGM3
rs214814
NWNGSVEILK
462
X
X
X
X
X
X





20
TGM3
rs214814
NWNGSVEILKNWKK
463
X
X
X
X
X
X





17
KRTAP
rs989425
PACYETTCCR
464


X






9-9
8













12
KRT83
rs285767
PCGQLNTTCGGGSCGQG
465










1
RY












21
KRTAP
rs963684
PCSTTYSRPLTFVSSGSQP
466
X
X

X
X
X



11-1
5
LGGISSVCQPVGGISTVC












QPVGGVSTVCQPACGVS












R












17
KRTAP
rs238824
PICGSSCCQPCCHPTCYQ
467









9-1

TTCFRTTCCQPTCCQPTC












CRNTSCQPT












17
KRTAP
rs353820
PLCCQTTCRPR
468


X






4-1
39













21
KRTAP
rs963684
PLTFVSSGSQPLGGISSVC
469
X
X

X
X
X



11-1
5
QPVGGISTVCQPVGGVST












VCQPACGVSR












17
KRTAP
rs720768
PQCCQPVCCQPTCCRPR
470



X





4-9
5













17
KRTAP
rs238830
PQCCQSVCYQPTCCHPSC
471

X

X





4-5

CISSCCHPYCCESSCCRPC












CCRPSCCQTTCCR












17
KRTAP
rs745728
PQCCQTICCR
472



X





4-4
64













 1
PKP1
rs142096
PQIAHLLQSGNSDVVR
473










411













17
KRTAP
rs626228
PSCCQTSSCR
474
X

X
X

X



1-3
49













17
KRTAP
rs620672
PSCCSPSCCQTTCCR
475

X







4-2
92













17
KRTAP
rs116504
PSCCVSSCCRPQCCQSVC
476



X





4-7
84
CQPTCCRPSCCETTCCHP












RCCI












17
KRTAP
rs739831
PSCCVSSCCRPQCCQSVC
477


X






4-6
72
CQPTCCRSSCCPSCCQTT












CCR












21
KRTAP
rs481894
PSSCQPTCCTSSPCQQAC
478
X
X
X
X
X
X



10-10
9
CVPVCSKSVCYMPVCSG












ASTSCCQQSSCQPACCTA












SCCR












21
KRTAP
rs481895
PSSCQPTCCTSSPCQQAC
479


X






10-10
0
CVPVCSKSVCYMPVCSG












ASTSCCQQSSCQPACCTA












SCCR












17
KRTAP
rs382959
PTGPATTICSSDKSCCCG
480
X
X

X
X
X



3-2
8













17
KRTAP
rs349771
PVCGGVSCHTTCYRPTC
481


X
X

X



4-11

VISSCPRPLCCASSCC












 1
VSIG8
rs412648
PVVPMCWTEGHMTYGN
482










27
DVVLK












17
KRT32
rs110789
QCMITNVEAQLAEIQADL
483
X

X
X
X
X




93
ERQNQEYQVLLDVR












12
KRT84
rs161393
QFLEQQNKLLETK
484
X
X

X
X
X




1













17
KRT33
rs124506
QLERDNAELK
485

X


X




B
21













17
KRT33
rs124506
QLERDNAELKNLIR
486

X


X




B
21













17
KRT33
rs124506
QLERDNAELKNLIRER
487

X


X




B
21













17
KRT31
rs650362
QLERDNVELENLIR
488
X
X








7













17
KRT31
rs650362
QLERDNVELENLIRER
489
X
X








7













17
KRT36
rs808268
QLERENVELESR
490


X







3













17
KRT33
rs148752
QLERHNAELENLIR
491









A
041













17
KRT33
rs148752
QLERHNAELENLIRER
492









A
041













16
PPL
rs806372
QLLAGLDKVASDLDQQE
493










7
K












20
TGM3
rs146717
QLLVDFSCNKFPAIK
494










993













12
KRT75
rs199744
QLQTQVGDTSVVLSMDN
495










850
NCNLDLDSIIAEVK












12
KRT84
rs951773
QLREYQELMNAKLGLDI
496











EIATYRR












17
KRT39
rs178430
QNQEYEILMDVK
497


X
X






23













17
KRT34
rs199674
QNQEYQVLLDVCAR
498










249













17
KRT34
rs199674
QNQEYQVLLDVCARLEC
499










249
EINTYR












17
KRT40
rs806473
QNQEYQVLLDVKARLEG
500
X
X

X
X
X




3
EINTYR












17
KRTAP
rs129386
QNTCCRTTCCQPTCVTSC
501
X
X
X
X
X




9-6
92
CQPSCCSTPCCQPICCGSS












CCGQTSCGSSCGQSSSCA












PVYCR












17
KRTAP
rs374150
QPCCHPTCCQNTCCRTTC
502









9-3
255
CQPICVTSCCQPSCCSTPC












CQPTRCGSSCGQSSSCAP












VYCR












17
KRTAP
rs626228
QPSCCQTSSCR
503
X

X
X

X



1-3
49













17
KRTAP
rs181901
QPVCCGSSCCGQTSCGSS
504









9-6
202
CGQSSSCAPVYCR












17
KRTAP
rs720768
QPVCCQPTCCRPRCCISS
505



X





4-9
5
CCRPSCCVSSCCKPQCCQ












SVCCQPNCCRPS












12
KRT83
rs285246
QSHISDTSVVVKLDNSRD
506
X
X
X
X
X
X




4
LNMDCMVAEIKAQYDDI












ATR












17
KRT27
rs116593
QSVEADLNGLR
507










021













17
KRT27
rs116593
QSVEADLNGLRR
508










021













14
LGALS
rs11125
QSVFPFESGKPFKIHVLVE
509
X
X


X




3

PDHFK












17
KRTAP
rs149188
QTSFCGFR
510



X

X



1-1
249













21
KRTAP
rs380401
QTSYVESRPCQTSCYRPR
511
X
X
X
X
X
X



13-2
0













21
KRTAP
rs963684
QTTCISNPCSTTYSRPLTF
512
X
X

X
X
X



11-1
5
VSSGSQPLGGISSVCQPV












GGISTVCQPVGGVSTVCQ












PACGVSR












17
KRT33
rs129375
QVEIIELR
513
X
X
X
X
X
X



A
19













17
KRT33
rs129375
QVEIIELRR
514
X
X
X
X
X
X



A
19













17
KRT34
rs112570
QVVSSSEQLQSCQVEIIEL
515










296
R












17
KRT34
rs112570
QVVSSSEQLQSCQVEIIEL
516










296
RR












17
KRT33
rs129375
QVVSSSEQLQSYQVEIIEL
517
X
X
X
X
X
X



A
19
R












17
KRT33
rs129375
QVVSSSEQLQSYQVEIIEL
518
X
X
X
X
X
X



A
19
RR












17
KRT33
rs129375
QVVSSSEQLQSYQVEIIEL
519
X
X
X
X
X
X



A
19
RRTVNALEIELQAQHNLR












17
KRTAP
rs374150
RCGSSCGQSSSCAPVYCR
520









9-3
255













12
KRT83
rs285246
RDLNMDCMVAEIKAQY
521
X
X
X
X
X
X




4
DDIATR












12
KRT85
rs112554
REAECVEANSGR
522










450













12
KRT85
rs112554
REAECVEANSGRLASELN
523










450
HVQEVLEGYK












12
KRT85
rs112554
REAECVEANSGRLASELN
524










450
HVQEVLEGYKK












17
KRT33
rs129375
REVEQWFATQTEELNKQ
525
X
X
X
X
X
X



A
19
VVSSSEQLQSYQVEIIELR












R












17
KRT34
rs777791
REVEQWFATQTEK
526










92













17
KRT34
rs777791
REVEQWFATQTEKLNK
527










92













12
KRT84
rs951773
REYQELMNAKLGLDIEIA
528











TYR












12
KRT81
rs207158
RGLTGGFGSHSVCR
529
X
X
X
X
X
X




8













 6
DSP
rs692906
RGQSEADSDKNATILELR
530
X
X
X
X
X
X




9













17
KRT32
rs207156
RILDDLTLCKADLEAQVE
531


X







1
YLKEELMCLK












17
KRT34
rs566233
RILNELTLCK
532










643













17
KRT36
rs230135
RMVNALEIELQAQHSMR
533


X
X






4













17
KRTAP
rs137947
RPCCCRPSCCQTTCCR
534


X






4-5
981













17
KRTAP
rs777211
RPSCCIPCCCRPTCVISTC
535
X
X


X




4-7
664
PRPLCC












17
KRT31
rs650362
RQLERDNVELENLIR
536
X
X








7













12
KRT84
rs951773
RQLREYQELMNAKLGLD
537











IEIATYR












21
KRTAP
rs963684
RQTTCISNPCSTTYSRPLT
538
X
X

X
X
X



11-1
5
FVSSGSQPLGGISSVCQPV












GGISTVCQPVGGVSTVCQ












PACGVSR












12
KRT86
rs572429
RSFGYHSGGVCGPSPPCI
539










51
TTVSVNESLLTPLNLEIDP












NAQCVK












12
KRT86
rs572429
RSFGYHSGGVCGPSPPCI
540










51
TTVSVNESLLTPLNLEIDP












NAQCVKQEEKEQIK












17
KRTAP
rs739831
RSSCCPSCCQTTCCR
541


X






4-6
72













17
KRT40
rs806491
RTASALEIELQAQQSLTE
542










0
SLECTVAETEAQYSSQLA












QIQRLIDNLENQLAEIR












17
KRTAP
rs199605
RTCYHPTTVCLPGCLNQS
543









9-4
390
CGSSCCQPCCR












17
KRTAP
rs199605
RTCYHPTTVCLPGCLNQS
544









9-4
390
CGSSCCQPCCRPACCETT












CFQPTCVY












17
KRTAP
rs219137
RTCYHPTTVCLPGCLNQS
545









9-4
9
CGSSCCQPCCRPACCETT












CFQPTCVY












17
KRTAP
rs199605
RTCYHPTTVCLPGCLNQS
546









9-4
390
CGSSCCQPCCRPACCETT












CFQPTCVYS












17
KRTAP
rs219137
RTCYHPTTVCLPGCLNQS
547









9-4
9
CGSSCCQPCCRPACCETT












CFQPTCVYS












17
KRTAP
rs219137
RTCYYPTTVCLPGCLNQS
548









9-4
9
CGSNCCQPCCRPACCETT












CFQPTCVYS












17
KRTAP
rs219137
RTCYYPTTVCLPGCLNQS
549









9-4
9
CGSNCCQPCCRPACCETT












CFQPTCVYSCCQPFCC












17
KRTAP
rs626228
RTGCGIGGGIGYGQEGSS
550
X

X
X

X



1-3
49
GAVSTR












17
KRTAP
rs142863
RTGCGTGGGIGYGQEGSS
551









1-3
014
GAVSTR












17
KRTAP
rs626228
RTGCGTGGGIGYGQEGSS
552
X

X
X

X



1-3
49
GAVSTR












12
KRT86
rs139895
RTKEEINELNCMIQR
553










699













17
KRT31
rs151023
RTVNSLEIELQAQHNLR
554










228













17
KRT31
rs151023
RTVNSLEIELQAQHNLRD
555










228
SLENTLTESEAR












17
KRT32
rs169669
RTVNTLEIELQAQHSLRD
556










29
SLENMLTESEAR












14
LGALS
rs101483
RVIVCNTKLDNNWGKEE
557


X






3
71
R












21
KRTAP
rs343029
RVPVPSCCVPTSSCQPSCS
558
X
X

X
X
X



10-12
39
R












21
KRTAP
rs343029
RVPVPSCCVPTSSCQPSCS
559
X
X

X
X
X



10-12
39
RL












17
KRT35
rs743686
RVSAMYSSSPCKLPSLSP
560



X







VARSFSACSVGLGR












12
KRT86
rs749337
RVSSDPSNSNVVVGTTN
561










520
ACAPSAR












17
KRT32
rs110789
RYSSQLAQMQCMITNVE
562
X

X
X
X
X




93
AQLAEIQADLERQNQEY












QVLLDVR












19
GIPC1
rs454588
SAGGRPGSGPQLGSGR
563
X
X
X

X
X




94













17
JUP
rs412834
SAIVHLINYQDDAELATH
564

X








25
ALPELTK












17
JUP
rs412834
SAIVHLINYQDDAELATH
565

X








25
ALPELTKLLNDEDPVVVT












K












17
JUP
rs150245
SAIVHLINYQDDAK
566










906













17
JUP
rs150245
SAIVHLINYQDDAKLATR
567










906













17
KRT35
rs207160
SARPICVPCPGGRF
568



X






1













 1
SFN
rs149812
SAYQEAMDISKKDMPPT
569










347
NPIR












17
KRTAP
rs116553
SCCGSVCSDQGCGQVLC
570




X




4-7
10
QETCCRPSCCQTTCCR












17
KRTAP
rs777211
SCCISSCCRRPTCVISTCP
571
X
X


X




4-7
664
R












17
KRTAP
rs777211
SCCISSCCRRPTCVISTCP
572
X
X


X




4-7
664
RPL












17
KRTAP
rs142863
SCCQPSCCQTSSCGTGCG
573









1-3
014
TGGGIGYGQEGSSGAVST












R












17
KRTAP
rs149188
SCCQTSFCGFR
574



X

X



1-1
249













17
KRTAP
rs626228
SCCQTSSCRTGCGIGGGI
575
X

X
X

X



1-3
49
GYGQEGSSGAVSTR












17
KRTAP
rs389784
SCCQTTCCRTTCCRPSCC
576









4-2

VSSCFRPQCCQSVCCQPT












CCRPSCGQTTCCR












17
KRTAP
rs389784
SCCVSSCFRPQCCQSVCC
577









4-2

QPTCCRPSCGQTTCCRT












12
KRT85
rs616300
SCGHSFGYR
578




X





04













12
KRT86
rs572429
SCGRSFGYHSGGVCGPSP
579










51
PCITTVSVNESLLTPLNLE












IDPNAQCVKQEEKEQIK












17
KRTAP
rs626228
SCRTGCGIGGGIGYGQEG
580
X

X
X

X



1-3
49
SSGAVSTR












17
KRTAP
rs626233
SCYQPR
581


X
X

X



1-5
75













12
KRT81
rs751670
SDLEANVDALIQEIDFLR
582










289
R












17
KRTAP
rs116553
SDQGCGQDLCQETCCRP
583



X





4-7
10
SCCQTTCCR












 1
PKP1
rs347049
SEPDLYYDPR
584


X







38













12
KRT86
rs572429
SFGYHSGGVCGPSPPCITT
585










51
VSVNESLLTPLNLEIDPN












AQCVK












12
KRT86
rs572429
SFGYHSGGVCGPSPPCITT
586










51
VSVNESLLTPLNLEIDPN












AQCVKQEEK












12
KRT86
rs572429
SFGYHSGGVCGPSPPCITT
587










51
VSVNESLLTPLNLEIDPN












AQCVKQEEKEQIK












12
KRT86
rs572429
SFGYHSGGVCGPSPPCITT
588










51
VSVNESLLTPLNLEIDPN












AQCVKQEEKEQIKSLNSR












17
KRTAP
rs626228
SFSTSGTCSSSCCQPSCCE
589
X

X
X

X



1-3
49
TSCCQPSCCQTSSCRTGC












GIGGGIGYGQEGSSGAVS












TR












17
KRT39
rs721325
SGAIESTAPACTSSSPCSL
590
X
X

X
X





6
KEHCSACGPLSQILVK












17
KRT39
rs721325
SGAIESTAPACTSSSPCSL
591
X
X

X
X





6
KEHCSACGPLSQILVKI












12
KRT81
rs476178
SKCEEMKATVIRHGETLC
592










6
R












17
KRT37
rs200713
SKCHESTVCPNYQSYFR
593










258













17
KRT34
rs201477
SKYQMEQSLR
594










948













12
KRT85
rs139493
SLCNLGSCGPRIAVGGSR
595










548
A












17
KRT40
rs200400
SLGETNAELESR
596










895













21
KRTAP
rs151147
SLGYGGCGFPSLGYGVG
597









13-1
550
FCHPTYLASR












17
KRT37
rs169668
SLHQLVEADKCGTQKLL
598
X
X
X
X
X
X




11
DDVTLAK












17
KRT37
rs149061
SLHQLVEVDKCGTQK
599










216













17
KRT39
rs721325
SLKEHCSACGPLSQILVK
600
X
X


X
X




6













17
KRT33
rs140430
SLLESEDCKLPSNPCATT
601









A
944
NACDKSTGPCISKPCGLR












AR












17
KRT24
rs114431
SLNDRLANYLDKVR
602










517













11
PKP3
rs777522
SLSLSLADSGHLPDLHGF
603










15
NSYGSHR












11
PKP3
rs148364
SLTSLIR
604










325













12
KRT82
rs265865
SMPVLSTGVLR
605
X

X
X
X
X




8













17
KRT35
rs743686
SPCKLPSLSPVAR
606



X







21
KRTAP
rs113360
SPCQTSCYHPR
607









13-2
916













 9
CRAT
rs311863
SPMVPLPMPK
608










5













17
KRT32
rs110789
SQLAQMQCMITNVEAQL
609
X

X
X
X
X




93
AEIQADLERQNQEYQVL












LDVR












17
KRT32
rs260495
SQLAQMQCMITNVEAQL
610
X
X








6
AEIRAELERQNQEYQVLL












DVR












17
KRT34
rs150738
SQLGDCLNVEVDTAPTV
611










879
DLNQVLNETR












17
KRT34
rs223971
SQLGDCLNVEVDTAPTV
612










0
DLNQVLNETRSQYEALV












ETNRR












17
KRT34
rs150738
SQLGDCLNVEVDTAPTV
613










879
DLNQVLNETRSQYEALV












ETNRR












17
KRT34
rs140296
SQLGDRLNLEVDTAPTV
614










098
DLNQVLNETR












17
KRT31
rs112544
SQYEVLVETNR
615










857













17
KRT31
rs112544
SQYEVLVETNRR
616










857













17
KRT31
rs112544
SQYEVLVETNRREVEQW
617










857
FTTQTEELNKQVVSSSEQ












LQSYQAEIIELR












11
PKP3
rs200371
SRGVGGAVPGAVLEPVA
618

X








913
PAPSVR












21
KRTAP
rs963684
SRPLTFVSSGSQPLGGISS
619
X
X

X
X
X



11-1
5
VCQPVGGISTVCQPVGG












VSTVCQPACGVSR












21
KRTAP
rs963684
SRQTTCISNPCSTTYSRPL
620
X
X

X
X
X



11-1
5
TFVSSGSQPLGGISSVCQP












VGGISTVCQPVGGVSTVC












QPACGVSR












17
KRTAP
rs739831
SSCCPSCCQTTCCRTTCC
621


X






4-6
72
R












17
KRTAP
rs749779
SSEQSCGLENCCCPSCCQ
622


X
X





4-5
892
TTCCR












17
KRTAP
rs145881
SSGAVSTCIR
623









1-5
217













12
KRT1
rs14024
SSGGSSSVR
624
X
X
X

X
X





21
KRTAP
rs113360
SSPCQTSCYHPR
625









13-2
916













17
KRT33
rs129375
SSSEQLQSYQVEIIELRRT
626
X
X
X
X
X
X



A
19
VNALEIELQAQHNLRDSL












ENTLTESEAR












17
KRT35
rs743686
SSSPCKLPSLSPVAR
627



X







18
DSG4
rs617348
SSTMGALRDYADADINM
628


X







47
AFLDSYFSEK












17
KRTAP
rs145585
STCCQPSCVIR
629









9-1
952













17
KRT33
rs140430
STGPCISKPCG
630









A
944













17
KRT33
rs140430
STGPCISKPCGL
631









A
944













17
KRT33
rs140430
STGPCISKPCGLR
632









A
944













17
KRTAP
rs129386
STPCCQPICCGSSCCGQTS
633
X
X
X
X
X




9-6
92
CGSSCGQSSSCAPVYCR












21
KRTAP
rs372198
STSCRPLSYLSR
634









24-1
438













17
KRTAP
rs626228
STSGTCSSSCCQPSCCETS
635
X

X
X

X



1-3
49
CCQPSCCQTSSCRTGCGI












GGGIGYGQEGSSGAVSTR












17
KRTAP
rs142863
STSGTCSSSCCQPSCCETS
636









1-3
014
CCQPSCCQTSSCRTGCGT












GGGIGYGQEGSSGAVSTR












17
KRTAP
rs626228
STSGTCSSSCCQPSCCETS
637
X

X
X

X



1-3
49
CCQPSCCQTSSCRTGCGT












GGGIGYGQEGSSGAVSTR












21
KRTAP
rs963684
STTYSRPLTFVSSGSQPLG
638
X
X

X
X
X



11-1
5
GISSVCQPVGGISTVCQP












VGGVSTVCQPACGVSR












17
KRT37
rs144652
STVNALEVER
639










431













17
KRTAP
rs350240
SYGTGCGIGGGIGYGQEG
640


X






1-1
33
SSGAVSTR












 6
DSP
6:g.7568
SYKPIILR
641










542A > T













21
KRTAP
rs201732
SYVSSPCCR
642


X
X





10-6
843













21
KRTAP
rs713213
TACQPTCYQR
643
X
X
X

X
X



11-1
55













17
KRT40
rs806491
TASALEIELQAQQSLTESL
644










0
ECTVAETEAQYSSQLAQI












QR












12
KRT76
rs111702
TATENEFVGLKK
645
X
X
X
X
X
X




71













17
KRTAP
rs199605
TCYHPTTVCLPGCLNQSC
646









9-4
390
GSSCCQPCCRPACCETTC











FQPTCVY













17
KRTAP
rs219137
TCYHPTTVCLPGCLNQSC
647









9-4
9
GSSCCQPCCRPACCETTC












FQPTCVY












17
KRTAP
rs142863
TGCGTGGGIGYGQEGSS
648









1-3
014
GAVSTR












17
KRTAP
rs626228
TGCGTGGGIGYGQEGSS
649
X

X
X

X



1-3
49
GAVSTR












12
KRT81
rs207158
TGGFGSHSVCR
650
X
X
X
X
X
X




8













12
KRT81
rs207158
TGGFGSHSVCRGFRA
651
X
X
X
X
X
X




8













17
KRT40
rs178430
TGSCNSPCLVGNCAWCE
652
X
X
X
X
X
X




15
DGVSTSNEKETMQFLND












RLASYLEKVR












18
DSG4
rs722925
TICIDSPSVLISVNEHSYG
653


X







2
SPFTFCVVDEPPGTADM












WDVR












12
KRT86
rs139895
TKEEINELNCMIQR
654










699













17
KRT35
rs207160
TNCSARPICVPCPGGR
655



X






1













17
KRT35
rs207160
TNCSARPICVPCPGGRF
656



X






1













17
KRT35
rs124516
TNYSPRPICVPCPGGR
657
X
X
X
X
X
X




52













17
KRT35
rs124516
TNYSPRPICVPCPGGRF
658
X
X
X
X
X
X




52













17
KRTAP
rs626233
TSCYQPR
659


X
X

X



1-5
75













17
KRTAP
rs149188
TSFCGFR
660



X

X



1-1
249













18
ATP5A
rs779587
TSIAVDTIINQKR
661









1
05













12
KRT83
rs285246
TSVVVKLDNSRDLNMDC
662
X
X
X
X
X
X




4
MVAEIKAQYDDIATR












17
KRTAP
rs129386
TTCCQPTCVTSCCQPSCC
663
X
X
X
X
X




9-6
92
STPCCQPICCGSSCCGQTS












CGSSCGQSSSCAPVYCR












17
KRTAP
rs752970
TTCCRPSCCG
664









4-1
851













17
KRTAP
rs752970
TTCCRPSCCGS
665









4-1
851













17
KRTAP
rs752970
TTCCRPSCCGSS
666









4-1
851













17
KRTAP
rs752970
TTCCRPSCCGSSC
667









4-1
851













17
KRTAP
rs750304
TTCCRPSCCRPR
668









4-4
09













17
KRTAP
rs389784
TTCCRPSCCVSSCFRPQC
669









4-2

CQSVCCQPTCC












17
KRTAP
rs389784
TTCCRTTCCRPSCCVSSC
670









4-2

FRPQCCQSVCCQPTCCR












17
KRTAP
rs389784
TTCCRTTCCRPSCCVSSC
671









4-2

FRPQCCQSVCCQPTCCRP












SCGQTTCCR












17
KRTAP
rs144403
TTCFQPTCVSSSCQPSCC
672









9-9
228













17
KRTAP
rs219137
TTCFQPTCVYSCCQPFCC
673









9-4
9













12
KRT83
rs285767
TTCGGGSCGQGRY
674










1













17
KRTAP
rs112082
TTCWKPTTVTTCSSTPCC
675
X
X
X
X
X
X



9-3
369
QPSCCVSSCCQPCCHPTC












CQNTCCRTTCCQPI












17
KRTAP
rs577716
TTCWKPTTVTTCSSTS
676


X






9-7
67













17
KRTAP
rs577716
TTCWKPTTVTTCSSTSC
677


X






9-7
67













17
KRTAP
rs577716
TTCWKPTTVTTCSSTSCC
678


X






9-7
67
QPSCCVSSCCQPCCHPTC












CQNTCCRTTCCQPTC












17
KRTAP
rs444509
TTSCRPSCCVS
679


X






4-4














17
KRTAP
rs444509
TTSCRPSCCVSS
680


X






4-4














 1
TCHH
rs251566
TVDLILELLDR
681










3













17
KRT32
rs147160
TVGTPCSPCPQGRY
682










974













17
KRT31
rs151023
TVNSLEIELQAQHNLR
683










228













17
KRT31
rs151023
TVNSLEIELQAQHNLRDS
684










228
LENTLTESEAR












17
KRT31
rs151023
TVNSLEIELQAQHNLRDS
685










228
LENTLTESEARYSSQLSQ












VQSLITNVESQLAEIR












17
KRT32
rs169669
TVNTLEIELQAQHSLRDS
686










29
LENMLTESEAR












17
KRT32
rs374478
TYLSSSCR
687
X
X
X
X
X
X




6













17
KRTAP
rs389784
VCCQPTCCRPSCGQTTCC
688









4-2

R












17
KRTAP
rs116553
VCSDQGCGQVLCQETCC
689



X





4-7
10
RPSCCQTTCCR












17
KRT31
rs650362
VELENLIR
690
X
X








7













17
KRT40
rs140634
VHSLEETNAELESR
691










473













14
LGALS
rs101483
VIVCNTKLDNNWGKEER
692


X






3
71













12
KRT83
rs285246
VKLDNSRDLNMDCMVA
693
X
X
X
X
X
X




4
EIKAQYDDIATR












17
KRT32
rs728300
VLEEMRCQYEAMVEAN
694
X
X
X
X
X
X




46
HR












18
DSC3
rs276937
VLNDGTVYTAR
695
X
X
X


X





17
KRT31
rs112544
VLNETRSQYEVLVETNR
696










857













17
KRT31
rs112544
VLNETRSQYEVLVETNR
697










857
R












 8
FAM83
rs996960
VNLHHVDFLR
698









H
0













 6
DSP
rs207629
VQCDLQKANSSATETINK
699
X

X
X
X
X




9
LKVQEQELTR












 6
DSP
rs287639
VQEQELTCLR
700










67













20
TGM3
rs149720
VRFDILPSQSGTK
701










612













12
KRT86
rs587172
VRFLEQQNKLLETKLPFY
702
X
X

X
X
X




66
QNR












17
KRT33
rs124506
VRQLERDNAELK
703

X


X




B
21













17
KRT33
rs124506
VRQLERDNAELKNLIR
704

X


X




B
21













17
KRT31
rs650362
VRQLERDNVELENLIR
705
X
X








7













17
KRT31
rs650362
VRQLERDNVELENLIRER
706
X
X








7













17
KRT33
rs148752
VRQLERHNAELENLIR
707









A
041













17
KRT33
rs148752
VRQLERHNAELENLIRER
708









A
041













17
KRTAP
rs626228
VRWCRPDCR
709
X
X
X
X
X
X



1-3
47













17
KRT35
rs743686
VSAMYSSSPCK
710



X







17
KRT35
rs743686
VSAMYSSSPCKLPSLSPV
711



X







AR












17
KRTAP
rs140898
VSCHTTCYRPTCVISSCPR
712


X
X

X



4-11
464
PVC












17
KRTAP
rs140898
VSCHTTCYRPTCVISSCPR
713


X
X

X



4-11
464
PVCCA












17
KRT34
rs116116
VSGNSCGPCGTSQK
714










504













12
KRT86
rs749337
VSSDPSNSNVVVGTTNA
715










520













12
KRT86
rs749337
VSSDPSNSNVVVGTTNA
716










520
CAPSAR












21
KRTAP
rs963684
VSSGSQPLGGISSVCQPV
717
X
X

X
X
X



11-1
5
GGISTVCQPVGGVSTVCQ












PACGVSR












17
KRT33
rs129375
VSSSEQLQSYQVEIIELR
718
X
X
X
X
X
X



A
19













17
JUP
rs112682
VSVELTNSLFKHDPAAW
719



X

X




1
EAAQSMIPINEPYGDDLD












ATYRPMYSSDVPLDPLE












M












12
KRT83
rs285246
VVKLDNSRDLNMDCMV
720
X
X
X
X
X
X




4
AEIKAQYDDIATR












12
KRT83
rs285246
VVVKLDNSRDLNMDCM
721
X
X
X
X
X
X




4
VAEIKAQYDDIATR












12
KRT2
rs638043
WELLQQMNVDTRPINLE
722
X
X


X
X





PIFQGYIDSLKR












12
KRT86
rs111429
WLYEEEIR
723










470













12
KRT86
rs111429
WLYEEEIRVLQSHISDTS
724










470
VVVK












17
KRTAP
rs444509
YCQTTCCRTTSCRPSCCV
725


X






4-4

SSCCRPQCCQTTCCR












12
KRT83
rs766508
YEEEVALQATAENEFVA
726










559
LKK












17
KRT31
rs112544
YEVLVETNRR
727










857













17
KRT34
rs201477
YQMEQSLR
728










948













17
KRT33
rs347718
YSLENTLTESEARYSSQL
729









B
86
SQVQSLITNVESQLAEIHS












DLERQNQEYQVLLDVR












17
KRT40
rs806491
YSSQLAQIQRLIDNLENQ
730










0
LAEIR












17
KRT36
rs116573
YSSQLAQMQCLISTVEAQ
731
X
X
X

X
X




23
LSEIR












17
KRT36
rs116573
YSSQLAQMQCLISTVEAQ
732
X
X
X

X
X




23
LSEIRCDLER












17
KRT36
rs116573
YSSQLAQMQCLISTVEAQ
733
X
X
X

X
X




23
LSEIRCDLERQNQEYQVL












LDVK












17
KRT32
rs110789
YSSQLAQMQCMITNVEA
734
X

X
X
X
X




93
QLAEIQADLER












17
KRT32
rs110789
YSSQLAQMQCMITNVEA
735
X

X
X
X
X




93
QLAEIQADLERQNQEYQ












VLLDVR












17
KRT32
rs260495
YSSQLAQMQCMITNVEA
736
X
X








6
QLAEIQAELERQNQEYQ












VLLDVR












17
KRT32
rs110789
YSSQLAQMQCMITNVEA
737
X

X
X
X
X




93
QLAEIQAELERQNQEYQ












VLLDVR












17
KRT32
rs260495
YSSQLAQMQCMITNVEA
738
X
X








6
QLAEIRAELER












17
KRT32
rs260495
YSSQLAQMQCMITNVEA
739
X
X








6
QLAEIRAELERQNQEYQV












LLDVR












17
KRT34
rs148645
YSSQLSQMQSLITNVESQ
740










199
LAEIR












17
KRT33
rs347718
YSSQLSQVQSLITNVESQ
741









B
86
LAEIHSDLER












17
KRT33
rs347718
YSSQLSQVQSLITNVESQ
742









B
86
LAEIHSDLERQNQEYQVL












LDVR












17
KRT34
rs199674
YSSQLSQVQSLITNVESQ
743










249
LAEIRCDLERQNQEYQVL












LDVC












17
KRT34
rs617406
YSSQLSQVQSLITNVESQ
744










68
LAEIRCDLEWQNQEYQV












LLDVR












17
KRT35
rs743686
YSSSPCKLPSLSPVAR
745



X







11
GSTP1
rs1695
YVSLIYTNYEAGKDDYV
746
X
X
X
X
X






K












11
GSTP1
rs1695
YVSLIYTNYEVGKDDYV
747
X
X
X
X
X






K












11
GSTP1
rs11382
YVSLIYTNYEVGKDDYV
748
X
X


X





2
K





X = more preferable for sub-population






Example 47
Exemplary GVP Detectable in Skin Samples

An exemplary set of GVPs that can be used in methods and systems herein described as well as in related databases is reported herein. In particular, the exemplary set of GVPs comprises genes validated as proteomically detectable in skin samples of a Homo Sapiens which can be used in methods and systems to detect a genetic variation and/or perform a genetic variation analysis, as well as in related databases, in accordance with the various aspects of the present disclosure.


Specifically, Table 12 shows a list of exemplary GVP detectable in skin samples. The fields in Table 12 are the name of the gene (gene name), mutation identifier (mutation ID), sequence of the mutated peptide (mutated peptide (GVP)), sequence identifier in the sequence listing of the instant disclosure (SEQ ID NO), and the subpopulations including all populations (ALL), Non-Finnish European subpopulation (NFE), African subpopulation (AFR), East Asian subpopulation (EAS), South Asian subpopulation (SAS), and Latino subpopulation (AMR).


The exemplary GVPs of Table 12 can be used in method and systems of the instant disclosure wherein the sample comprises a skin sample from human beings.









TABLE 12







 Exemplary GYP detectable in skin samples
















gene
mutation

SEQ ID








name
ID
mutated peptide (GVP)
NO
All
NFE
AFR
EAS
SAS
AMR





DSC1
rs17800159
AASSQTPTMCTTTVTIK
749
X
X


X
X





KRT78
rs61764062
ALALALYQIK
750
X


X
X
X





KRT6B
rs144860693
AGGSYGFGGAR
751
X
X
X
X
X
X





ECM1
rs13294
APYPNYDRD1LTID1SR
752
X
X
X
X
X
X





ECM1
rs13294
DILTIDISR
753
X
X
X
X
X
X





POF1B
rs363774
EELGHLQNDLTSLENDK
754











POF1B
rs363774
EELGHLONDLTSLENDKMR
755











FLG2
rs3818831
EIHPVLK
756
X
X
X

X
X





FLG2
rs3818831
EFHPVLKNPDDPDTVDVIMH
757
X
X
X

X
X





FLG2
rs3818831
EFHPVLKNPDDPDTVDVIMHMLDR
758
X
X
X

X
X





ECM1
rs3737240
EGMPAPFGDQSHPEPESWNAAQHCQQDR
759
X
X
X
X
X
X





FLG2
rs3818831
ELLEKEFHPVLK
760
X
X
X

X
X





KRT6A
rs144401677
EQGTKTVRQNMEPLFEQYINNLR
761











KRT78
rs2013335
FGEWSGGPGLSLCPPGGIQEVTINQNPL
762


X







TPLK












KRT2
rs638043
FLEQQNQVLQ1KWELLQQMNVDTRPINL
763
X
X


X
X




EPIFQGYIDSLKR












KRT14
rsl1551758
FSSGGAYGLGGGYGGGF
764


X








KRT14
rs6503640
FSSGGAYGLGGGYGGGF
765











KRT14
rs3826550
FSSGGAYGLGGGYGGGFSSSSSSFGSGF
766
X
X
X
X
X
X




GGGYGGGLGTGLGGGFGGGFAGGDGLLV











GSEK












FLG2
rs3818831
GELKELLEKEFHPVLK
767
X
X
X

X
X





HAL
rs7297245
GETISGGNIHGEYPAK
768











KRT2
rs2634041
GGGFGGGSGFGGGSGF
769
X
X
X
X

X





KRT2
rs2634041
GGGFGGGSGFGGGSGFSGGGF
770
X
X
X
X

X





KRT2
rs2634041
GGGFGGGSGFGGGSGFSGGGFGGGGFGG
771
X
X
X
X

X




GR












KRT10
rs747151268
GGGSFGGGFGGGFGGDGGLLSGNEK
772
X
X
X
X
X
X





KRT10
rs17855579
GGGSFGGGYGGGSSGGGSSGGGY
773











KRT10
rs17855579
GGGSFGGGYGGGSSGGGSSGGGYGGGH
774











KRTI0
rs17855579
GGGSFGGGYGGGSSGGGSSGGGYGGGHG
775










G












KRT10
rs17855579
GGGSFGGGYGGGSSGGGSSGGGYGGGHG
776










GSSGGGY












KRT10
rs17855579
GGGSFGGGYGGGSSGGGSSGGGYGGGHG
777










GSSGGGYGGGSSGGGY












KRT77
rs636127
GGSGGGYGSGCGGGGGSYGGSGR
778











KPRP
rs16834461
GHPAVCQPQGR
779
X
X
X
X
X
X





Clorf68
rs1332500
GSGLGAGQGTNGASVK
780
X
X

X
X
X





KRT1
rs14024
GSSSGGVKSSGGSSSVR
781
X
X
X

X
X





KRT10
rs4261597
GSYGSSSFGGSYGGSFGGGSFGGGSFGG
782










GSFGGGGFGGGGFGGGFGGGFGGDGGLL











SGNEK












FLG
rs7512857
HAGIGHGQASSAVR
783
X
X
X

X
X





JUP
rs1126821
HDPAAWEAAQSMIP1NEPYGDDLDATYR
784



X

X




PM












JUP
rs1126821
HDPAAWEAAQSMIPINEPYGDDLDATYR
785



X

X




PMYSSDV












JUP
rs1126821
HDPAAWEAAQSMIPINEPYGDDLDATYR
786



X

X




PMYSSDVPLDPLEMH












DSC1
rs28620831
HGLVATHTLTVR
787

X









S100A7
rs3014837
IDKPSLLTMMK
788











JUP
rs41283425
INYQDDAELATHALPELTK
789

X









KRT14
rs59780231
LEQEITTYR
790



X

X





JUP
rs41283425
LINYQDDAELATHALPELTK
791

X









KPRP
rs17612167
LPLHQC
792
X
X

X
X
X





KPRP
rs4329520
LRPEPS1SLEPR
793



X
X






KRT5
rs11549950
LSGEGVGPVNISVVTSSVSSGYGSGSGY
794
X
X
X

X
X




GGGLGGGLGGGLGGGLAGGGSGS












POF1B
rs363774
LVLSTFSNIREELGHLQNDLTSLENDK
795











KRT2
rs638043
MNVDTRPINLEPIFQGYIDSLKR
796
X
X


X
X





JUP
rs199826380
NLSDVATKOEGLENVLK
797











DSP
rs17604693
NTNFAQK
798











KRT2
rs638043
NVDTRPINLEPIFQGYIDSLK
799
X
X


X
X





KRT2
rs638043
NVDTRPINLEPIFQGYIDSLKR
800
X
X


X
X





TGM3
rs214814
NWNGSVEILK
801
X
X
X
X
X
X





DSG1
rs3752095
PILDPLGYGNVTVTESFrrSDTLKPSVH
802
X
X
X
X
X
X




VHDNRPASXVVVTER












JUP
rs199826380
QEGLENVLK
803











KRT6B
rs11170126
QNLELLFEQYINNLR
804











KRT6A
rs144401677
QNMEPLFEQYINNLR
805











ECM1
rs13294
RAPYPNYDRDILTIDISR
806
X
X
X
X
X
X





S100A7
rs3014837
RDDKIDKPSLLTMMK
807











JUP
rs41283425
SAIVHLINYQDDALLATHALPELTK
808

X









ANXA2
rs17845226
SALSGHLETL1LGLLK
809
X
X



X





KRT5
rs11549949
SGGLSVGGSGFSASSGR
810
X
X
X

X
X





FLG2
rs16842865
SGHSSYGQHGFGSSQSSGYGQHGSSSGQ
811










TSGFGQHK












KRT78
rs2253798
SLNSFGR
812
X
X
X
X
X
X





KRT1
rs14024
SSGGSSSVR
813
X
X
X

X
X





KRT14
rs3826550
SSSSSSFGSGFGGGYGGGLGTGLGGGFG
814
X
X
X
X
X
X




GGFAGGDGLLVGSEK












Clorf68
rs41268474
STSYCYLAPR
815
X
X
X
X
X
X





KRT14
rs59780231
TRLEQEITTY
816



X

X





KRT14
rs59780231
TRLEQEITTYR
817



X

X





LOR
rs6661601
TSGGGGGGGGGGGGGCGFFGGGGSGGGS
818
X
X
X
X
X
X




SGSGCGY












DSC1
rs17800159
TTTVTIK
819
X
X


X
X





KR12
rs638043
VDTRPINLEPIFQGYIDSLK
820
X
X


X
X





KRT2
rs638043
VDTRP1NLEP1F0GY1DSLKR
821
X
X


X
X





DSC3
rs35630063
VEDENDSHPVFrEAIYNFEVLESSR
822











DSG1
rs139922779
VVSPISGADLHGMLEMPDLR
823











DSG1
rs139922779
VVSPISGADLHGMLEMPDLRDGSNVIVT
824










ER












KRT2
rs638043
WELLQQMNVDTR
825
X
X


X
X





KRT2
rs638043
WELLOOMNVDPRPINLEPIFOGY
826
X
X


X
X





KRT2
rs638043
WELLQQMNVDTRPINLEPIFQGYIDSLK
827
X
X


X
X





KRT2
rs638043
WELLQQMNVDTRP1NLEPIFQGYIDSLK
828
X
X


X
X




R












KRT36
rs11657323
YSSQLAQMQCLISTVEAQLSEIR
829
X
X
X

X
X





X = more preferable for sub-population






In summary according to the first aspect, a method is described to prepare a biological sample for proteomic analysis, the method comprising applying to the biological sample an energy field resulting in an increased thermodynamic or total energy of the sample to obtain a processed biological sample comprising solubilized proteins to be used in the proteomic analysis.


In a first set of embodiments of the method of the first aspect, applying to the biological sample an energy field is performed by sonication and in particular by sonication baths, sonication probes, or flow-through sonication systems. In a second set of embodiments of the method of the first aspect which can comprise the method of the first aspect performed according to the first set of embodiments, the biological sample is hair and/or skin. In a third set of embodiments of the method of the first aspect which can comprise the method of the first aspect performed according to the first set of embodiments of the method of the first aspect, the biological sample can be bone or teeth.


In summary according to the second aspect, a method is described to provide a marker genetic protein variation of a biological organism in a biological sample of the biological organism.

  • The method comprises:


detecting exome sequences of the sample of the biological organism by sequencing exomes of a genome from the sample of the biological organism;


detecting a marker exome sequence comprising a genetic variation of the genome of the biological organism by comparing the detected exome sequences with a database of exome sequences of the biological organism;


detecting peptide sequences of the sample of the biological organism by performing proteomic analysis of the sample of the biological organism; and providing the marker genetic protein variation of the biological organism in the sample of the biological organism by comparing the detected marker exome sequence with the detected peptide sequences to provide a marker genetic protein variation validated for the same of the biological organism.


In a first set of embodiments of the method of the second aspect, the biological organism is Homo sapiens. In a second set of embodiments of the method of the second aspect which can comprise the method of the second aspect performed according to the first set of embodiments, the biological sample is hair.


According to the second aspect of the disclosure, a marker genetic protein variation of a biological organism is also described. The marker genetic protein variation of the second aspect is validated for a sample of the biological organism, and is obtainable and obtained by any one of the method according to the second aspect.


In summary according to the third aspect, a method is described to improve a marker genetic protein variation database system including data for at least one biological organism. The method comprises


producing a mass spectrometry dataset from a biological sample from an individual of the at least one biological organism;


comparing the mass spectrometry dataset to a protein variant database to produce a set of proteomically detected proteins in the biological sample of the individual;


providing a set of represented genes proteomically detectable in the biological sample of the individual, the represented genes corresponding to the proteomically detected proteins in the biological sample of the individual; and


identifying a marker genetic protein variation validated for the biological sample of the individual, to be included in the marker genetic protein variation database system by


providing a proteomically detectable genomic variation in the set of represented genes proteomically detectable in the biological sample of the individual, and


providing the marker genetic protein variation validated genetic protein variation by providing a proteomically detectable genetic protein variation corresponding to the proteomically detectable genomic variation in the biological sample of the individual.


In a first set of embodiments of the method of the third aspect, providing the marker validated genetic protein variation, further comprises: providing a mass spectrometry dataset from the biological sample of the individual; and comparing the provided mass spectrometry dataset with the proteomically detectable genetic protein variation to provide the validated genetic protein variation.


In a second set of embodiments of the method of the third aspect which can comprise the method of the third aspect performed according to the first set of embodiments, providing a proteomically detectable genomic variation in the set of represented genes proteomically detectable in the biological sample of the individual is performed by providing exome sequence data of the individual; and comparing the exome sequence data of the individual with sequences from the represented genes proteomically detectable in the biological sample of the individual to determine the proteomically detectable genomic variation in the biological sample of the individual.


In a third set of embodiments of the method of the third aspect which can comprise the method of the third aspect performed according to the first set of embodiments or the second set of embodiments, providing a proteomically detectable genetic protein variation corresponding to the proteomically detectable genomic variation in the biological sample of the individual, is performed by: performing annotation on the proteomically detectable genomic variation in the biological sample of the individual to produce a corresponding mutant/reference protein sequence; and providing the proteomically detectable genetic protein variation from the annotated proteomically detectable genomic variation in the biological sample of the individual.


In a fourth set of embodiments of the method of the third aspect, which can comprise the method of the third aspect performed according to the first set of embodiments, the second set of embodiments or the third set of embodiments, the method further comprises creating a genetic protein variation identity panel by collecting the validated genetic protein variant proteomically detectable in the biological sample of the individual to provide a genetic protein variation identity panel of the individual.


In a fifth set of embodiments of the method of the third aspect, which can comprise the method of the third aspect performed according to the first set of embodiments, the second set of embodiments, the third set of embodiments or the fourth set of embodiments, the steps are repeated for a plurality of individuals of the at least one biological organism, to provide a database comprising validated genetic protein variations proteomically detectable in the biological sample of the plurality of individuals of the biological organism type.


In a first subset of embodiments of the fifth set of embodiments of the method according to the third aspect, the method further comprises: collecting the represented genes common to the plurality of the individuals into a proteomically detectable gene pool; providing validated genetic protein variations proteomically detectable in the biological sample of the plurality of individuals of the at least one biological organism from the collected common represented; and collecting the validated genetic protein variant proteomically detectable in the biological sample of the plurality of individuals, in the genetic protein variation panel is a genetic protein variation panel common to the plurality of individuals.


In a second subset of embodiments of the fifth set of embodiments of the method according the third aspect, the proteomically detectable gene pool contains data corresponding to proteins that are common to over 50% of all the validated genetic protein variant proteomically detectable in the biological sample of the individual.


In some embodiments of the first subset of embodiments or the second subset of embodiments of the fifth set of embodiments of the method according to the third aspect, the providing validated genetic protein variations proteomically detectable in the biological sample of the plurality of individuals is performed to only include genomic variation with a frequency greater than 1% in the plurality of the individuals into a proteomically detectable gene pool.


In a sixth set of embodiments of the method of the third aspect, which can comprise the method of the third aspect performed according to the first set of embodiments, the second set of embodiments, the third set of embodiments, the fourth set of embodiments or the fifth set of embodiments comprising any related subsets of embodiments, the at least one biological organism is Homo sapiens.


In a seventh set of embodiments of the method of the third aspect, which can comprise the method of the third aspect performed according to the first set of embodiments, the second set of embodiments, the third set of embodiments, the fourth set of embodiments, the fifth set of embodiments comprising any related subsets of embodiments, or the sixth set of embodiments, the biological sample is hair or skin.


According to the third aspect, a marker genetic protein variation database system is also described obtainable and/or obtained by the methods according to third aspect, which comprises the method of the third aspect performed according to any one of the related sets or subsets of embodiments.


In summary according to the fourth aspect, a method is described to improve a marker genetic protein variation database system comprising marker genetic protein variations common to a plurality of individuals. The method comprises


providing a number of proteomic datasets of individuals of the plurality of individuals, the number statistically significant for the plurality of individuals;


identifying a protein common to the provided number of proteomic datasets;


selecting from the identified protein common to the provided proteomic datasets, a protein detectable in a biological sample of an individual of the plurality of individuals;


providing a number of exome datasets of the individuals of the plurality of individuals, the number statistically significant for the plurality of individuals;


identifying a genetic variation in the provided number of exome datasets;


selecting from the identified genetic variation, a genetic variation detectable in the biological sample; and


comparing the selected proteins detectable in the biological sample with the selected genetic variations detectable in the biological sample,


to provide a marker genetic protein variation common to a plurality of individuals of a biological organism type and detectable in the biological sample.


In a first set of embodiments of the method of the fourth aspect, the individual is a Homo sapiens.


In a second set of embodiments of the method of the fourth aspect which can comprise the method of the fourth aspect performed according to the first set of embodiments, the biological sample is hair.


According to the fourth aspect, a marker genetic protein variation database system is also described, comprising genetic protein variations common to a plurality of individuals. The genetic protein variation database system is obtainable by the method according to sixth aspect, which comprises the method of the fourth aspect performed according to any one of the related sets of embodiments.


In summary, according to the fifth aspect a method is described to detect a genetic protein variation in a biological sample. The method comprises


providing a marker mass spectrum of a marker peptide comprising a marker genetic protein variation corresponding to the genetic protein variation;


performing mass spectrometry of a fractionated digested peptide of the biological sample to obtain a mass spectrum of each of the fractionated digested peptide; and


comparing the mass spectrum of the fractionated digested peptide with a marker mass spectrum of a marker peptide comprising the marker genetic protein variation to detect the genetic protein variation in the biological sample.


In a first set of embodiments of the method according to the fifth aspect, the fractionated digested peptides are obtained by preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in the protein analysis, fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample, digesting the solubilized proteins from the sample with a site specific proteolytic enzyme to obtain digested solubilized proteins from the sample, and fractionating the digested solubilized proteins to obtain fractionated digested peptides from the digested solubilized proteins from the biological sample.


In a first subset of embodiments of the first set of embodiments of the method of the fifth aspect preparing the biological sample is performed according to the method of the first aspect of the disclosure comprising any one of the related sets of embodiments.


In a second set of embodiments of the method of the fifth aspect which can comprise the method of the fifth aspect performed according to the first set of embodiments, the marker peptide comprises a plurality of marker peptides each comprising a marker genetic protein variation.


In a third set of embodiments of the method of the fifth aspect which can comprise the method of the fifth aspect performed according to the first set of embodiments or the second set of embodiments, the marker genetic protein variation comprises a marker genetic protein variation according to the second aspect of the disclosure.


In a fourth set of embodiments of the method of the fifth aspect which can comprise the method of the fifth aspect performed according to the first set of embodiments, the second set of embodiments or the third set of embodiments, the marker genetic protein variation comprises a marker genetic protein variation from a marker genetic protein variation database system according to the third aspect of the disclosure comprising any one of the related sets of embodiments.


In a fifth set of embodiments of the method of the fifth aspect which can comprise the method of the fifth aspect performed according to the first set of embodiments, the second set of embodiments, the third set of embodiments or the fourth set of embodiments, the marker genetic protein variation comprises a marker genetic protein variation from a marker genetic protein variation database system according to the fourth aspect of the disclosure comprising any one of the related sets of embodiments.


In summary according to the sixth aspect, a method is described to provide a marker genetic variation database system for a biological sample. The method comprises:


preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis.


fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample and a solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample;


detecting a genetic protein variation in the solubilized proteins from the sample by performing the proteomic analysis of the solubilized protein fraction;


detecting a genomic variation of the nuclear and/or mitochondrial genome by performing a genetic analysis of the solubilized DNA fraction; and combining the detected genetic protein variations and the detected genomic variation to provide the marker genetic variation database system of the biological sample.


In a first set of embodiments of the method according to the sixth aspect, preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis, is performed by the method of the first aspect, comprising any one of the related sets of embodiments.


In a second set of embodiments of the method of the sixth aspect which can comprise the method of the sixth aspect performed according to the first set of embodiments, detecting a genetic protein variation is performed by the method according to the fifth aspect comprising any one of the related sets and subsets of embodiments.


In a third set of embodiments of the method of the sixth aspect which can comprise the method of the sixth aspect performed according to the first set of embodiments or second sets of embodiments, the genetic protein variation is a single amino acid polymorphism (SAP), an amino acid deletion and/or an amino acid insertion.


In a fourth set of embodiments of the method of the sixth aspect which can comprise the method of the sixth aspect performed according to the first set of embodiments, second sets of embodiments or third sets of embodiments, the genomic variation is a single nucleotide polymorphism (SNP), a nucleotide deletion or a nucleotide insertion.


In a fifth set of embodiments of the method of the sixth aspect which can comprise the method of the sixth aspect performed according to the first set of embodiments, the second set of embodiments, the third set of embodiments or the fourth set of embodiments, the genomic variation is within the short tandem repeat (STR) regions of the genome.


In a sixth set of embodiments of the method of the sixth aspect which can comprise the method of the sixth aspect performed according to the first set of embodiments, the second set of embodiments, the third set of embodiments the fourth set of embodiments or the fifth set of embodiments, the genomic variation is within the mitochondrial DNA.


According to the sixth aspect, a marker genetic variation database system is also described obtainable by the method according to the sixth aspect of the disclosure, comprising any one of the related sets of embodiments.


In summary according to the seventh aspect, a method is described to detect a marker genetic variation in a biological sample of a biological organism. The method comprises preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis;


fractionating the processed biological sample to obtain


a solubilized protein fraction comprising the solubilized proteins from the sample and


a solubilized DNA fraction comprising solubilized nuclear and/or mitochondrial genome from the sample;


detecting a genetic protein variation in the solubilized proteins from the sample by performing the proteomic analysis of the solubilized protein fraction;


detecting a genomic variation of the nuclear and/or mitochondrial genome by performing a genetic analysis of the solubilized DNA fraction; and


comparing the detected genetic protein variation and/or the detected genomic variation with a marker genetic protein variation and/or of a marker genomic variation respectively from the marker genetic variation database system of the sixth aspect of the disclosure.


In a first set of embodiments of the method according to the seventh aspect, preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis, is performed by the method according to the first aspect of the disclosure comprising any one of the related sets of embodiments.


In a second set of embodiments of the method of the seventh aspect which can comprise the method of the seventh aspect performed according to the first set of embodiments, detecting a genetic protein variation is performed by the method according to the fifth aspect of the disclosure comprising any one of the related sets and subsets of embodiments.


In a third set of embodiments of the method of the seventh aspect which can comprise the method of the seventh aspect performed according to the first set of embodiments or second sets of embodiments, the genetic protein variation is a single amino acid polymorphism (SAP), an amino acid deletion and/or an amino acid insertion.


In a fourth set of embodiments of the method of the seventh aspect which can comprise the method of the seventh aspect performed according to the first set of embodiments, second sets of embodiments or third sets of embodiments, the genomic variation is a single nucleotide polymorphism (SNP), a nucleotide deletion or a nucleotide insertion.


In a fifth set of embodiments of the method of the seventh aspect which can comprise the method of the seventh aspect performed according to the first set of embodiments, the second set of embodiments, the third set of embodiments or the fourth set of embodiments, the genomic variation is within the short tandem repeat (STR) regions of the genome.


In a sixth set of embodiments of the method of the seventh aspect which can comprise the method of the seventh aspect performed according to the first set of embodiments, the second set of embodiments, the third set of embodiments or the fourth set of embodiments, the genomic variation is within the mitochondrial DNA.


In summary according to the eight aspect of the disclosure, a method is described to perform genetic analysis of a sample of a biological organism. The method comprises preparing the biological sample to obtain a processed biological sample comprising solubilized proteins to be used in a proteomic analysis;


fractionating the processed biological sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample;


digesting the solubilized protein fraction from the sample to obtain digested peptides from the sample;


fractionating the digested peptides to obtain fractionated digested peptides from the digested solubilized proteins from the biological sample.


detecting a marker genetic variation of the fractionated digested peptides from the sample; in which


preparing the sample is performed according to any one of the methods according to the first aspect of the disclosure, comprising any one of the related sets of embodiments ; and/or


detecting a genetic variation is performed by at least one of


the method to detect a genetic protein variation of any one of the methods according to the fifth aspect, comprising any one of the related sets and subsets of claims; and


the method to detect a genetic variation of any one of the methods according to the seventh aspect of the disclosure comprising any one of the related sets of embodiments.


Preferably in any one of the embodiments of the method to perform genetic analysis of a sample of a biological organism of the eight aspect the preparing is performed according to any one of the methods according to the first aspect of the disclosure, comprising any one of the related sets of embodiments and the detecting is performed at least one of the method to detect a genetic protein variation of any one of the methods according to the fifth aspect, comprising any one of the related sets and subsets of claims; and the method to detect a genetic variation of any one of the methods according to the seventh aspect of the disclosure comprising any one of the related sets of embodiments.


In view of the above, in summary described herein are methods and systems to perform genetically variant protein analysis and related marker genetic protein variations and databases, which in several embodiments allow performing a reliable genetic variation protein analysis in biological samples of different types and conditions taking into account the features of the biological sample where the analysis is performed. The examples set forth above are provided to give those of ordinary skill in the art a complete disclosure and description of how to perform the embodiments of the methods and systems of the disclosure, and are not intended to limit the scope of what the inventors regard as their disclosure. Those skilled in the art will recognize how to adapt the features of the exemplified methods and systems herein disclosed to additional methods and systems according to various embodiments and scope of the claims.


All patents and publications mentioned in the specification are indicative of the levels of skill of those skilled in the art to which the disclosure pertains.


The entire disclosure of each document cited (including patents, patent applications, journal articles, abstracts, laboratory manuals, books, or other disclosures) in the Background, Summary, Detailed Description, and Examples is hereby incorporated herein by reference. All references cited in this disclosure are incorporated by reference to the same extent as if each reference had been incorporated by reference in its entirety individually. However, if any inconsistency arises between a cited reference and the present disclosure, the present disclosure takes precedence. Further, the computer readable form of the sequence listing of the ASCII text file IL-13212-Sequence-Listing_ST25 is incorporated herein by reference in its entirety.


The terms and expressions which have been employed herein are used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the disclosure claimed. Thus, it should be understood that although the disclosure has been specifically disclosed by embodiments, exemplary embodiments and optional features, modification and variation of the concepts herein disclosed can be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this disclosure as defined by the appended claims.


It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the content clearly dictates otherwise. The term “plurality” includes two or more referents unless the content clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosure pertains.


When a Markush group or other grouping is used herein, all individual members of the group and all combinations and possible sub-combinations of the group are intended to be individually included in the disclosure. Every combination of components or materials described or exemplified herein can be used to practice the disclosure, unless otherwise stated. One of ordinary skill in the art will appreciate that methods, system elements, and materials other than those specifically exemplified may be employed in the practice of the disclosure without resort to undue experimentation. All art-known functional equivalents, of any such methods, device elements, and materials are intended to be included in this disclosure. Whenever a range is given in the specification, for example, a temperature range, a frequency range, a time range, or a composition range, all intermediate ranges and all subranges, as well as, all individual values included in the ranges given are intended to be included in the disclosure. Any one or more individual members of a range or group disclosed herein may be excluded from a claim of this disclosure. The disclosure illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein.


A number of embodiments of the disclosure have been described. The specific embodiments provided herein are examples of useful embodiments of the disclosure and it will be apparent to one skilled in the art that the disclosure can be carried out using a large number of variations of the genetic circuits, genetic molecular components, and methods steps set forth in the present description. As will be obvious to one of skill in the art, methods and systems useful for the present methods and systems may include a large number of optional composition and processing elements and steps.


In particular, it will be understood that various modifications may be made without departing from the spirit and scope of the present disclosure. Accordingly, other embodiments are within the scope of the following claims.


REFERENCES



  • 1. Bodzon-Kulakowska, A., et al., Methods for samples preparation in proteomic research. Journal of Chromatography B, 2007. 849(1): p. 1-31.

  • 2. Cao, R., et al., dbSAP: single amino-acid polymorphism database for protein variation detection. Nucleic acids research, 2016. 45(D1): p. D827-D832.

  • 3. Parker, G. J., et al., Demonstration of protein-based human identification using the hair shaft proteome. PloS one, 2016. 11(9): p. e0160653.

  • 4. Ochoa-Rivas, A., et al., Microwave and Ultrasound to Enhance Protein Extraction from Peanut Flour under Alkaline Conditions: Effects in Yield and Functional Properties of Protein Isolates. Food and Bioprocess Technology, 2017. 10(3): p. 543-555.

  • 5. Phongthai, S., S.-T. Lim, and S. Rawdkuen, Optimization of microwave-assisted extraction of rice bran protein and its hydrolysates properties. Journal of Cereal Science, 2016. 70: p. 146-154.

  • 6. Sun, W., et al., Microwave-assisted protein preparation and enzymatic digestion in proteomics. Molecular & Cellular Proteomics, 2006. 5(4): p. 769-776.

  • 7. Ye, X. and L. Li, Microwave-assisted protein solubilization for mass spectrometry-based shotgun proteome analysis. Analytical chemistry, 2012. 84(14): p. 6181-6191.

  • 8. Lubec, G., et al., Structural stability of hair over three thousand years. Journal of archaeological science, 1987. 14(2): p. 113-120.

  • 9. Kaye, D. H., Ultracrepidarianism in Forensic Science: The Hair Evidence Debacle. 2015.

  • 10. Robertson, J., Managing the forensic examination of human hairs in contemporary forensic practice. Australian Journal of Forensic Sciences, 2017. 49(3): p. 239-260.

  • 11. McNevin, D., et al., Short tandem repeat (STR) genotyping of keratinised hair. Part 1. Review of current status and knowledge gaps. Forensic Sci Int, 2005. 153(2-3): p. 237-46.

  • 12. Melton, T., et al., Forensic mitochondrial DNA analysis of 691 casework hairs. J Forensic Sci, 2005. 50(1): p. 73-80.

  • 13. Rice, R. H., G. E. Means, and W. D. Brown, Stabilization of bovine trypsin by reductive methylation. Biochimica et Biophysica Acta (BBA)-Protein Structure, 1977. 492(2): p. 316-321.

  • 14. Cox, B. and A. Emili, Tissue subcellular fractionation and protein extraction for use in mass-spectrometry-based proteomics. Nature protocols, 2006. 1(4): p. 1872.

  • 15. Fic, E., et al., Comparison of protein precipitation methods for various rat brain structures prior to proteomic analysis. Electrophoresis, 2010. 31(21): p. 3573-3579.

  • 16. Gupta, N., et al., Quantitative proteomic analysis of B cell lipid rafts reveals that ezrin regulates antigen receptor-mediated lipid raft dynamics. Nature immunology, 2006. 7(6): p. 625.

  • 17. Harder, A., et al., Comparison of yeast cell protein solubilization procedures for two-dimensional electrophoresis. Electrophoresis, 1999. 20(4-5): p. 826-829.

  • 18. Shao, S., et al., Reproducible tissue homogenization and protein extraction for quantitative proteomics using MicroPestle-assisted pressure-cycling technology. Journal of proteome research, 2016. 15(6): p. 1821-1829.

  • 19. Rice, R. H., Proteomic analysis of hair shaft and nail plate. J Cosmet Sci, 2011. 62(2): p. 229-36.

  • 20. Wu, P. W., et al., Proteomic analysis of hair shafts from monozygotic twins: Expression profiles and genetically variant peptides. Proteomics, 2017.

  • 21. Canas, B., et al., Trends in sample preparation for classical and second generation proteomics. Journal of Chromatography A, 2007. 1153(1): p. 235-258.

  • 22. Gundry, R. L., et al., Preparation of proteins and peptides for mass spectrometry analysis in a bottom-up proteomics workflow. Current protocols in molecular biology, 2009: p. 10.25. 1-10.25. 23.

  • 23. Feist, P. and A. B. Hummon, Proteomic challenges: sample preparation techniques for microgram-quantity protein analysis from biological samples. International journal of molecular sciences, 2015. 16(2): p. 3537-3563.

  • 24. Consortium, U., UniProt: a hub for protein information. Nucleic acids research, 2014: p. gku989.

  • 25. Boeckmann, B., et al., The SWISS-PROT protein knowledgebase and its supplement TrEMBL in 2003. Nucleic acids research, 2003. 31(1): p. 365-370.

  • 26. Hubbard, T., et al., The Ensembl genome database project. Nucleic acids research, 2002. 30(1): p. 38-41.

  • 27. Johnson, M., et al., NCBI BLAST: a better web interface. Nucleic acids research, 2008. 36(suppl_2): p. W5-W9.

  • 28. Vihinen, M., Bioinformatics in proteomics. Biomolecular engineering, 2001. 18(5): p. 241-248.

  • 29. Barker, W. C., et al., The protein information resource (PIR). Nucleic acids research, 2000. 28(1): p. 41-44.

  • 30. Wu, C. H., et al., The protein information resource. Nucleic acids research, 2003. 31(1): p. 345-347.

  • 31. Bantscheff, M., et al., Quantitative mass spectrometry in proteomics: a critical review.



Analytical and bioanalytical chemistry, 2007. 389(4): p. 1017-1031.

  • 32. Domon, B. and R. Aebersold, Mass spectrometry and protein analysis. science, 2006. 312(5771): p. 212-217.
  • 33. Gobom, J., et al., Sample purification and preparation technique based on nano-scale reversed-phase columns for the sensitive analysis of complex peptide mixtures by matrix-assisted laser desorption/ionization mass spectrometry. Journal of Mass Spectrometry, 1999. 34(2): p. 105-116.
  • 34. Guillarme, D., et al., New trends in fast and high-resolution liquid chromatography: a critical comparison of existing approaches. Analytical and bioanalytical chemistry, 2010. 397(3): p. 1069-1082.
  • 35. Ŝtulík, K., et al., Stationary phases for peptide analysis by high performance liquid chromatography: a review. Analytica chimica acta, 1997. 352(1-3): p. 1-19.
  • 36. Noble, J. E. and M. J. Bailey, Quantitation of protein. Methods in enzymology, 2009. 463: p. 73-95.
  • 37. Sapan, C. V., R. L. Lundblad, and N. C. Price, Colorimetric protein assay techniques. Biotechnology and applied Biochemistry, 1999. 29(2): p. 99-108.
  • 38. Nahnsen, S., et al., Tools for label-free peptide quantification. Molecular & Cellular Proteomics, 2013. 12(3): p. 549-556.
  • 39. Searle, B. C., Scaffold: a bioinformatic tool for validating MS/MS-based proteomic studies. Proteomics, 2010. 10(6): p. 1265-1269.
  • 40. Han, Y., B. Ma, and K. Zhang, SPIDER: software for protein identification from sequence tags with de novo sequencing error. Journal of bioinformatics and computational biology, 2005. 3(03): p. 697-716.
  • 41. Metzker, M. L., Sequencing technologies—the next generation. Nature reviews. Genetics, 2010. 11(1): p. 31.
  • 42. Ng, S. B., et al., Targeted capture and massively parallel sequencing of twelve human exomes. Nature, 2009. 461(7261): p. 272.
  • 43. Brun, V., et al., Isotope-labeled protein standards toward absolute quantitative proteomics. Molecular & Cellular Proteomics, 2007. 6(12): p. 2139-2149.
  • 44. Fusaro, V. A., et al., Prediction of high-responding peptides for targeted protein assays by mass spectrometry. Nature biotechnology, 2009. 27(2): p. 190-198.
  • 45. Gallien, S., et al., Selectivity of LC-MS/MS analysis: implication for proteomics experiments. Journal of proteomics, 2013. 81: p. 148-158.
  • 46. Jaffe, J. D., et al., Accurate Inclusion Mass Screening A bridge from unbiased discovery to targeted assay development for biomarker verification. Molecular & Cellular Proteomics, 2008. 7(10): p. 1952-1962.
  • 47. Wu, A. H., et al., Role of liquid chromatography-high-resolution mass spectrometry (LC-HR/MS) in clinical toxicology. Clinical Toxicology, 2012. 50(8): p. 733-742.
  • 48. Raymond, J. J., et al., Trace DNA success rates relating to volume crime offences. Forensic Science International: Genetics Supplement Series, 2009. 2(1): p. 136-137.
  • 49. Cann, H. M., et al., A human genome diversity cell line panel. Science, 2002. 296(5566): p. 261-2.
  • 50. Laatsch, C. N., et al. Human hair shaft proteomic profiling: individual differences, site specificity and cuticle analysis. PeerJ, 2014. 2, DOI: 10.7717/peerj.506.
  • 51. Bunger, M. K., et al., Detection and validation of non-synonymous coding SNPs from orthogonal analysis of shotgun proteomics data. J Proteome Res, 2007. 6(6): p. 2331-40.
  • 52. Fenyo, D., J. Eriksson, and R. Beavis, Mass spectrometric protein identification using the global proteome machine. Methods Mol Biol, 2010. 673: p. 189-202.
  • 53. Jeong, J., et al., Novel oxidative modifications in redox-active cysteine residues. Mol Cell Proteomics, 2011. 10(3): p. M110 000513.
  • 54. Solazzo, C., et al., Modeling deamidation in sheep alpha-keratin peptides and application to archeological wool textiles. Anal Chem, 2014. 86(1): p. 567-75.
  • 55. Ghesquiere, B. and K. Gevaert, Proteomics methods to study methionine oxidation. Mass Spectrom Rev, 2014. 33(2): p. 147-56.
  • 56. Robinson, N. E., Protein deamidation. Proc Natl Acad Sci U S A, 2002. 99(8): p. 5283-8.
  • 57. Evert, I. W. and B. S. Weir, Interpreting DNA Evidence: Statistical Genetics for Forensic Scientists. 1st ed. 1998: Sinauer Associates.
  • 58. Butler, J. M., Fundamentals of Forensic DNA Typing. 2010: Academic Press.
  • 59. Durbin, R. M., et al., A map of human genome variation from population-scale sequencing. Nature, 2010. 467(7319): p. 1061-73.
  • 60. Jeffreys, H., An Invariant Form for the Prior Probability in Estimation Problems. Proceedings of the Royal Society of London. Series A, Mathematical and Physical Sciences, 1946. 186(1007): p. 453-461.
  • 61. Gelman, A., et al., Bayesian Data Analysis. Second Edition ed. CRC Texts in Statistical Science. Vol. Book 106. 2003: Chapman & Hall.
  • 62. Thompson, J. D., D. G. Higgins, and T. J. Gibson, CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic acids research, 1994. 22(22): p. 4673-4680.
  • 63. Brandon, M. C., et al., MITOMASTER: a bioinformatics tool for the analysis of mitochondrial DNA sequences. Human mutation, 2009. 30(1): p. 1-6.

Claims
  • 1. A method to perform genetic analysis of a sample of a biological organism, the method comprising preparing the sample to obtain a processed sample comprising solubilized proteins;fractionating the processed sample to obtain a solubilized protein fraction comprising the solubilized proteins from the sample;digesting the solubilized protein fraction from the sample to obtain digested peptides from the sample;fractionating the digested peptides to obtain fractionated digested peptides from the digested solubilized proteins from the sample; anddetecting a marker genetic variation of the fractionated digested peptides from the sample through proteomic analysis;
  • 2. The method of claim 1, wherein the preparing the sample comprises performing cell and tissue disruption and performing protein solubilization.
  • 3. The method of claim 2, wherein preparing the sample comprises: performing removal of contaminants and/or performing protein enrichment following performing protein solubilization.
  • 4. The method of claim 1, wherein the applying is performed by sonication.
  • 5-9. (canceled)
  • 10. The method of claim 1, wherein the fractionating the processed sample and/or the fractionating the digested peptides is performed by a chromatography technique.
  • 11. The method of claim 1, wherein the digesting is performed enzymatically with one or more site specific proteolytic enzymes.
  • 12. The method of claim 11, wherein the one or more site specific proteolytic enzymes comprise trypsin, chymotrypsin, Lys-C, Arg-C, Asp-N, and Glu-C, non-specific; pepsin, and proteinase K.
  • 13. (canceled)
  • 14. The method of claim 1, wherein the detecting a marker genetic variation of the digested peptides from the sample is performed by mass spectrometry.
  • 15. (canceled)
  • 16. The method of claim 1, wherein providing a marker mass spectrum of a marker peptide comprising a marker genetic protein variation corresponding to the marker genetic protein variation, is performed by synthesizing a marker peptide and analyzing the marker peptide by performing mass spectrometry.
  • 17. The method of claim 1, wherein performing mass spectrometry of a digested peptide of the sample to obtain a mass spectrum of each of the digested peptide is performed by tandem mass spectrometry.
  • 18. The method of claim 1, wherein the marker peptide comprises a plurality of marker peptides each comprising a marker genetic protein variation.
  • 19. The method of claim 1, wherein comparing the mass spectrum of the fractionated digested peptides of the sample with a marker mass spectrum is performed by comparing the mass spectrum of the fractionated digested peptides with a mass spectrum of a protein variant database.
  • 20. The method of claim 19, wherein the protein variant database comprises a marker genetic protein variation validated to be detectable in the sample.
  • 21. The method of claim 1, wherein the genetic protein variation is a single amino acid polymorphism (SAP), an amino acid deletion and/or an amino acid insertion.
  • 22. The method of claim 1, wherein the genomic variation is a single nucleotide polymorphism (SNP), a nucleotide deletion and/or a nucleotide insertion.
  • 23. The method of claim 1, wherein the genomic variation is within the short tandem repeat (STR) regions of the genome or within the mitochondrial DNA.
  • 24. (canceled)
  • 25. The method of claim 1, wherein the genetic protein variation in the second detecting method is a marker genetic protein variation and detecting a genetic protein variation in the second detecting method is performed by the first detecting method.
  • 26. The method of claim 1 any one of claims 1 to 25, wherein the marker genetic protein variation comprises a marker genetic protein variation validated to be detectable in the sample.
  • 27-29. (canceled)
  • 30. The method of claim 1, wherein the sample is a single-hair sample.
  • 31. The method of claim 1, wherein the sample is hair, and wherein the marker peptide comprises a validated genetic protein variation of a gene listed in Table 8 of the specification.
  • 32. The method of claim 1, wherein the sample is hair, and wherein the marker genetic protein variation comprises one or more of the genetic protein variations listed in Table 11 of the specification.
  • 33-39. (canceled)
  • 40. A system to perform genetic analysis of a sample of a biological organism, the system comprising a reagent for preparing the sample by applying to the sample an energy field to obtain a processed sample comprising solubilized proteins;a marker peptide comprising a genetic protein variation validated to be detectable in the sample and/ora database validated to be detectable in the sample;
  • 41. The system of claim 40, wherein the database validated to be detectable in the sample comprises genetic protein variations common to a plurality of individuals of the biological organism.
  • 42. (canceled)
  • 43. The system of claim 40, wherein the sample is a single-hair sample.
  • 44. The system of claim 40, wherein the sample is hair, and wherein the marker peptide comprises a validated genetic protein variation of a gene listed in Table 8 of the specification.
  • 45. The system of claim 40, wherein the sample is hair, and wherein the marker genetic protein variation comprises one or more of the genetic protein variations listed in Table 11 of the specification.
  • 46. The system of claim 40, wherein the sample is hair, and wherein the marker peptide comprises one or more peptides having sequence SEQ ID NO: 151 to SEQ ID NO: 721.
  • 47-50. (canceled)
CROSS REFERENCE TO RELATED APPLICATIONS

The present application claims priority to U.S. Provisional Application No. 62/555,001, entitled “Methods and Systems to Perform Genetically Variant Protein Analysis, and Related Marker Genetic Protein Variations and Databases” filed on Sep. 6, 2017 with docket number IL-13212, the content of which is incorporated herein by reference in its entirety.

STATEMENT OF INTEREST

The invention was made with Government support under Contract No. DE-AC52-07NA27344 between the U.S. Department of Energy and Lawrence Livermore National Security, LLC, for the operation of Lawrence Livermore National Security. The Government may have certain rights to the invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2018/049775 9/6/2018 WO 00
Provisional Applications (1)
Number Date Country
62555001 Sep 2017 US