Miniaturized Proteomic Sample Preparation

Abstract
The disclosure provides methods of forming one or more single-cell proteomic samples, such as by: dispensing n droplets of lysis buffer onto a substantially planar solid surface, wherein n>2: dispensing a single cell into each of the n droplets of lysis buffer to produce n droplets with a lysed single cell: dispensing digestion buffer into each of the n droplets to digest proteins from each lysed single cell to produce n droplets comprising peptides: dispensing a chemical tag into at least a subset of the n droplets comprising the peptides to produce labeled peptides, thereby enabling the labeled peptides in a given droplet to be distinguishable from labeled peptides in at least one other droplet: and applying a fluid to merge at least a subset of the droplets into a combined droplet on the substantially planar surface, thereby combining the labeled peptides to form a single-cell proteomic sample.
Description
BACKGROUND

Single-cell measurements are essential for understanding biological systems composed of different cell types. Recent advances in single-cell RNA and protein methods have allowed analyzing single-cell heterogeneity at unprecedented scale and depth. These emerging single-cell methods have the potential to go beyond classifying cell types and to help characterize intrinsically single-cell processes, such as the cell division cycle (CDC) and its coordination with metabolism and cell growth. Crucial aspects of the CDC are regulated post-transcriptionally by protein synthesis and degradation and their characterization demands single-cell protein analysis. There is a need to improve single-cell proteomic sample preparation toward, for example, improved quantification of proteins and/or protein variabilities.


SUMMARY

Embodiments of the present invention include methods of single-cell proteomic sample preparation for analyzing peptides in samples with a low abundance of proteins.


In one aspect, the disclosure provides a method of forming a single-cell proteomic sample, said method comprising:

    • a) dispensing n droplets of lysis buffer onto a substantially planar solid surface, wherein n≥2;
    • b) dispensing a single cell into each of the n droplets of lysis buffer to produce n droplets, each comprising a lysed single cell;
    • c) dispensing digestion buffer into each of the n droplets to digest proteins from each lysed single cell to produce n droplets comprising peptides;
    • d) dispensing a chemical tag into each of the n droplets comprising the peptides to produce labeled peptides, wherein at least one droplet of the n droplets receives a different chemical tag from at least one other droplet of the n droplets, thereby enabling the labeled peptides in the at least one droplet to be distinguishable from the labeled peptides in the at least one other droplet; and
    • e) applying a fluid to merge at least a subset of the n droplets into a combined droplet on the substantially planar surface, thereby combining the labeled peptides to form a single-cell proteomic sample.


In some embodiments, each of the n droplets in step a), b), c), and/or d) has a volume of about 25 nanoliters (nl) or less. In particular embodiments, each of the n droplets in step a), b), c) and d) has a volume of about 25 nanoliters (nl) or less.


In some embodiments, the substantially planar solid surface is provided by a uniform glass slide. In certain embodiments, the substantially planar solid surface is etched with a geometric pattern. In particular embodiments, the substantially planar solid surface is fluorocarbon-coated.


In certain embodiments, n is ≥10.


In some embodiments, the lysis buffer comprises about 4-8 nanoliters of 90-100% dimethyl sulfoxide (DMSO).


In some embodiments, step b) comprises dispensing the single cell in a cell suspension buffer with a volume of about 100-1,000 picoliters. In particular embodiments, step b) comprises dispensing the single cell in a cell suspension buffer with a volume of about 300 picoliters.


In certain embodiments, the single cell is lysed in a total volume of about 4-10 nl for about 10-20 minutes.


In some embodiments, step c) comprises:

    • dispensing about 15-25 nl of about 120 ng/μl trypsin to each of the n droplets; and
    • digesting the proteins from each lysed single cell at about 1ºC above the dew point and a relative humidity of about 75% for about 4-5 hours.


In certain embodiments, the chemical tag comprises a “light” version of TMT label reagents dissolved in DMSO. In other embodiments, the chemical tag comprises a “heavy” version of TMT label reagents dissolved in DMSO.


In some embodiments, step d) comprises dispensing about 18-22 nl of a chemical tag into each of the n droplets comprising the peptides; and enabling the chemical tag to react with the peptides at room temperature and a relative humidity of about 75% for about 1 hour to produce the labeled peptides. In certain embodiments, each droplet of the n droplets receives a unique chemical tag, thereby enabling the labeled peptides in each droplet to be distinguishable from the labeled peptides in each other droplet.


In certain embodiments, the fluid is water. In particular embodiments, the fluid has a volume of about 1 μl.


In some embodiments, steps a) to e) are repeated at least once to form two or more single-cell proteomic samples on the substantially planar solid surface.


In certain embodiments, at least 100 droplets of lysis buffer are dispensed onto the substantially planar solid surface.


In some embodiments, at least 500-3,000 droplets of lysis buffer are dispensed onto the substantially planar solid surface.


In certain embodiments, the two or more single-cell proteomic samples comprises peptides from at least 100 cells. In particular embodiments, the two or more single-cell proteomic samples comprises peptides from about 100-10,000 cells.


In some embodiments, the disclosed methods further comprise performing at least one proteomic analysis on the single-cell proteomic sample. In particular embodiments, the at least one single-cell proteomic analysis enables identifying and/or quantifying protein covariation across the single cells.


In another aspect, the disclosure provides a method of performing a proteomic analysis comprising analyzing a single-cell proteomic sample formed by any of the methods described herein. In some embodiments, the analyzing comprises identifying and/or quantifying protein covariation across the single cells.


In another aspect, the disclosure provides a single-cell proteomic sample, for example, one formed by any one of the methods of single-cell proteomic sample formation described herein.


In another aspect, the disclosure provides kits and systems comprising reagents described herein (for example, one or more buffers) and/or an element that provides for a substantially planar surface and/or devices described herein.





BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.


The foregoing will be apparent from the following more particular description of example embodiments, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating embodiments.



FIGS. 1A-1D show parallel preparation of thousands of single cells by nPOP. FIG. 1A is schematic of a non-limiting example of nano-Proteomic-sample Preparation (nPOP) method illustrating the steps of cell lysis, protein digestion, peptide labeling with tandem mass tags (TMT), quenching of labeling reaction, and sample collection. These steps are performed for each single cell (corresponding to a single droplet). FIG. 1B shows that after barcoding, single-cell samples are automatically pooled into set samples, and the set samples are transferred into a 384-well plate, which is then placed into an autosampler for automated injection for LC-MS/MS. Any system that support 384-well plate injection (such as Dionex™ 3000) can implement this example workflow. FIG. 1C shows that the flat surface allows programming different droplet layouts, such as the 4 examples shown in the picture. FIG. 1D shows four slides with 2,016 single cells from an nPOP experiment using droplet configuration AL-01. Samples are surrounded by a perimeter of water for local humidity control. Slides are placed on a cooling surface to further prevent evaporation.



FIGS. 2A-2E depict proteome coverage and quality controls. FIG. 2A shows the number of proteins and peptides quantified per single cell from multiplexed samples prepared by nPOP and analyzed using 60 min active gradients on Q Exactive™ classic Mass Spectrometer. FIG. 2B shows the distributions of reporter ion (RI) intensities for all melanoma, monocyte, and negative controls. Intensities were mostly absent from negative control wells, which contained all reagents but not a single cell. FIG. 2C plots the average reporter ion intensity against the measured diameter of cells. A strong correlation between the two metrics shows that larger cells had increased protein contents. FIG. 2D shows that the mean quantitative variability per cell was tightly distributed, suggesting high consistency of sample preparation. The consistency of protein quantification was estimated as the coefficient of variation (CV) of the relative levels of peptides originated from the same protein. FIG. 2E Principal component analysis separates single-cell samples corresponding to melanoma cells or to U937 monocytes. 200 cell bulk samples were projected onto PCA to demonstrate agreement between bulk and single cell measurements.



FIGS. 3A-3C show protein correlations with joint distributions. The points represent the expression levels of two proteins in a single cell. FIG. 3A shows proteins that correlate in a similar manner within both cell types. FIG. 3B shows proteins that correlate with the opposite trend. FIG. 3C shows distributions of Euclidean distances of several complexes plotted along with the distribution for all proteins.



FIGS. 4A-4J identify functional protein groups that covary with cell division cycle (CDC)-markers. FIGS. 4A and 4F show proteins whose abundance varies with CDC phases identified using distributions of DNA content for Fluorescence-activated cell sorting (FACS) sorted cells. FIGS. 4B and 4G show correlations between CDC protein markers computed within the single cells from each type. FIGS. 4C and 4H depict Principal Component Analysis (PCA) of melanoma and monocyte cells in the space of CDC periodic genes. Cells in each PCA plot are colored by the mean abundance of proteins annotated to the marked phase. FIGS. 4D and 4I show boxplots display distributions for correlations between the CDC-phase markers and proteins from the proteins from the polyubiquitination gene ontology (GO) term. The difference between these distributions was evaluated by one-way ANOVA analysis to estimate statistical significance, FDR <5%. The distributions for other GO terms that covary in a similar way between the two cell lines are summarized with their medians plotted as a heatmap. FIGS. 4E and 4J show a similar analysis and display as in FIGS. 4D and 41 and are used to visualize GO terms whose covariation with the CDC is cell-type specific.



FIGS. 5A-5G show melanoma subpopulations. FIGS. 5A and 5F show PCA of melanoma cells which indicates two distinct clusters. Single cells were colored based on the protein abundances corresponding to transcripts previously identified as markers of primed cells (Emert et al., Nat Biotechnol. 39(7):865-76 (2021)). The single cells were also colored by the average abundance of protein sets exhibiting significant enrichment clusters A and B. FIG. 5B shows distributions of cells by CDC-phase for cells from cluster A and B. CDC-phases were determined from marker proteins from FIGS. 3A-3C. FIGS. 5C and 5G are protein sets showing distinct covariation in subpopulation A and B. The analysis and display are as in FIGS. 4E and 4J. FIG. 5D shows marginal distributions of protein abundances differentiating clusters A and B. FIG. 5E shows joint distributions of protein abundances differentiating clusters A and B.



FIGS. 6A-6E show functional protein covariation identified at the single-cell level by nPOP. Closely related pancreatic adenocarcinoma cell lines analyzed at the single-cell level by nPOP are easily clustered by time (FIG. 6A) and result in highly consistent protein quantification based on different peptides (FIG. 6B). The data allow identifying functional protein covariation (FIGS. 6C-6E).



FIGS. 7A-7D evaluate the efficiency of protein extraction by DMSO cell lysis. FIG. 7A Equal number of U-937 cells labeled with “Light” and “Heavy” isotopes via SILAC (stable isotope labeling by amino acids in cell culture) were lysed with urea or DMSO, diluted, and combined for digestion. FIG. 7B shows that the SILAC ratios for proteins from different cellular compartments show comparable protein recovery for DMSO and urea cell lysis. FIG. 7C Equal number of SILAC labeled “Light” Jurkat and “Heavy” U-937 cells were combined, and the mixed sample was then divided for cell lysis either by urea or by DMSO. FIG. 7C shows agreement between the SILAC ratios from the two methods which supports the use of DMSO lysis for quantitative protein analysis.



FIGS. 8A-8C depict another non-limiting example of workflow of nano-Proteomic sample Preparation (nPOP). FIG. 8A is a schematic of nPOP sample preparation method illustrating the steps of cell lysis, protein digestion, peptide labeling with isobaric chemical tags (TMT), and quenching with two additions of hydroxylamine. These steps are performed in parallel for all single cells and take place in small droplets. FIG. 8B is a representative field of droplets post trypsin addition. Droplets with single cells are clustered in groups of 13, and the number of cells are labeled and combined into one SCOPE2 sets using TMTpro. The single-cell droplets are surrounded by a perimeter of water droplets for maintaining high local humidity. FIG. 8C shows total ion current chromatograms from three runs demonstrating low contaminants and consistent chromatography.



FIGS. 9A-9D show reporter ion intensities in single cells and in negative controls. FIGS. 9A-9B show the reporter ion intensities for two representative Single Cell ProtEomics 2 (SCOPE2) sets prepared with nPOP. The panels show distributions of reporter ion intensities relative to the corresponding isobaric carrier for the set. RI intensities are mostly absent from negative control wells, which contains all reagents but not a single cell. FIG. 9C estimates the consistency of protein quantification using the coefficient of variation (CV) of the relative levels of peptides originating from the same protein. The median CVs per cells form a tight distribution, suggesting high consistency of sample preparation. FIG. 9D PCA separates samples corresponding to HeLa cells or to monocytes. The single cells cluster with bulk samples of 100 cells, indicating consistent relative protein quantitation.



FIGS. 10A-10H cluster cells based on cell type and cell cycle phase. PCA of HeLa cells in the space of proteins whose abundance is periodic with the cell cycle. Cells in each PCA plot are colored by the mean abundance of proteins annotated to the M/G1, G1/S, S, and G2 phases.





DETAILED DESCRIPTION

A description of example embodiments follows.


Traditionally, single-cell proteomic analyses have been performed by using fluorescent proteins or affinity reagents. While these approaches are powerful, mass spectrometry (MS) has the potential to increase the specificity and depth of single-cell protein quantification. For decades, MS has been a powerful tool for quantitative measurements of thousands of proteins in bulk samples consisting of thousands of cells or more.


Bulk samples are often prepared for liquid chromatography tandem MS analysis by using relatively large volumes (hundreds of microliters) and chemicals (detergents or chaotropic agents like urea) that are incompatible with MS analysis and require removal by cleanup procedures. The large volumes and cleanup procedures entail sample losses that may be prohibitive for small samples, such as single mammalian cells.


Definitions

Unless otherwise defined, all terms of art, notations and other scientific terms or terminology used herein are intended to have the meanings commonly understood by those of skill in the art to which this disclosure pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art. It will be further understood that terms, such as those defined in commonly-used dictionaries, should be interpreted as having a meaning that is consistent with their meaning in the context of the relevant art and/or as otherwise defined herein.


The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.


When introducing elements disclosed herein, the articles “a,” “an,” “the,” and “said” are intended to mean that there are one or more of the elements. Further, the one or more elements may be the same or different.


Throughout this specification and the claims which follow, unless the context requires otherwise, the word “comprise,” and variations such as “comprises” and “comprising”, will be understood to imply the inclusion of, e.g., a stated integer or step or group of integers or steps, but not the exclusion of any other integer or step or group of integer or step. When used herein, the term “comprising” can be substituted with the term “containing” or “including.”


As used herein, “consisting of” excludes any element, step, or ingredient not specified in the claim element. When used herein, “consisting essentially of” does not exclude materials or steps that do not materially affect the basic and novel characteristics of the claim. Any of the terms “comprising,” “containing,” “including,” and “having,” whenever used herein in the context of an aspect or embodiment of the disclosure, can in some embodiments, be replaced with the term “consisting of,” or “consisting essentially of” to vary scopes of the disclosure.


As used herein, the conjunctive term “and/or” between multiple recited elements is understood as encompassing both individual and combined options. For instance, where two elements are conjoined by “and/or,” a first option refers to the applicability of the first element without the second. A second option refers to the applicability of the second element without the first. A third option refers to the applicability of the first and second elements together. Any one of these options is understood to fall within the meaning, and, therefore, satisfy the requirement of the term “and/or” as used herein. Concurrent applicability of more than one of the options is also understood to fall within the meaning, and, therefore, satisfy the requirement of the term “and/or.”


It should be understood that for all numerical bounds describing some parameter in this application, such as “about,” “at least,” “less than,” and “more than,” the description also necessarily encompasses any range bounded by the recited values. Accordingly, for example, the description “at least 1, 2, 3, 4, or 5” also describes, inter alia, the ranges 1-2, 1-3, 1-4, 1-5, 2-3, 2-4, 2-5, 3-4, 3-5, and 4-5, et cetera.


Methods of the Disclosure

In various aspects, the disclosure provides methods of forming single-cell proteomic samples.


In one aspect, the disclosure provides a method of forming a single-cell proteomic sample, said method comprising:

    • a) dispensing n droplets of lysis buffer onto a substantially planar solid surface, wherein n≥2;
    • b) dispensing a single cell into each of the n droplets of lysis buffer to produce n droplets, each comprising a lysed single cell;
    • c) dispensing digestion buffer into each of the n droplets to digest proteins from each lysed single cell to produce n droplets comprising peptides;
    • d) dispensing a chemical tag into each of the n droplets comprising the peptides to produce labeled peptides, wherein at least one droplet of the n droplets receives a different chemical tag from at least one other droplet of the n droplets, thereby enabling the labeled peptides in the at least one droplet to be distinguishable from the labeled peptides in the at least one other droplet; and
    • e) applying a fluid to merge at least a subset of the n droplets into a combined droplet on the substantially planar surface, thereby combining the labeled peptides to form a single-cell proteomic sample.


In some embodiments, each of the n droplets in step a), b), c), and/or d) has a volume of less than 100 nanoliters (nl or nL), for example, less than 80, 60, 50, 40, 35, 30, 25, 22 or 20 nl. In certain embodiments, each of the n droplets in step a), b), c), and/or d) has a volume of about 100 nl or less, for example, about: 80, 60, 50, 40, 35, 30, 25, 22 or 20 nl or less. In particular embodiments, each of the n droplets in step a), b), c), and/or d) has a volume of about 25 nanoliters (nl) or less. In more particular embodiments, each of the n droplets in step a), b), c), and d) has a volume of about 25 nl or less.


In certain embodiments, the lysis buffer, the digestion buffer, the chemical tag, or a combination thereof is dispensed in a volume of about 1-20 nl per droplet, for example, about: 1-18, 1-16, 1-14, 1-12, 1-10, 1-8, I-6, I-4, 2-18, 2-16, 2-14, 2-12, 2-10, 2-8, 2-6, 2-4, 4-20, 4-18, 4-16, 4-14, 4-12, 4-10, 4-8, 4-6, 6-20, 6-18, 6-16, 6-14, 6-12, 6-10, 6-8, 8-20, 8-18, 8-16, 8-14, 8-12, 8-10, 10-20, 10-18, 10-16, 10-14, 10-12, 12-20, 12-18, 12-16, 12-14, 14-20, 14-18, 14-16, 16-20, 16-18 or 18-20 nl.


In some embodiments, the disclosure provides a method of forming at least two single-cell proteomic samples, wherein steps a) to e) are repeated at least once to form two or more single-cell proteomic samples. In certain embodiments, steps a) to e) are repeated at least 3 times, for example, at least 5, 10, 20, 30, 50, 80, 100, 120, 150, 180, 200, 250, 300, 350, 400, 500 or 1,000 times. In particular embodiments, steps a) to e) are repeated about 200 times.


Substantially Planar Solid Surfaces

As used herein, the term “substantially planar solid surface” refers to a surface that is substantially flat. In some embodiments, a substantially planar solid surface is a smooth surface. In certain embodiments, a substantially planar solid surface comprises etching, one or more (e.g., arrays of) very shallow dimples, or a combination thereof. A substantially planar solid surface enables small droplets (e.g., about 10-200 nl) of liquids to merge into a combined droplet when applying a fluid of a discrete volume (e.g., about 1 microliter (μl or μL)). A member (such as a multi-well plate or a microfuge tube) where its contents are closed off or surrounded, for example, by a wall, does not have a substantially planar solid surface. In particular embodiments, the substantially planar solid surface is provided by a slide, for example, a uniform glass slide.


In some embodiments, at least 90% of the points in the substantially planar surface are located on one of or between a pair of planes which are parallel and which are spaced from each other by a distance of not more than 5% of the largest dimension of the surface. In certain embodiments, the radius of curvature of the space is much greater than the cross-sectional dimensions, and the curvature does not substantially alter the function of the space. In particular embodiments, the substantially planar surface has a generally uniform thickness and having surface dimensions that are both much larger (e.g., ten to 100 times or more) than the thickness.


In certain embodiments, the substantially planar solid surface is etched, for example, with a laser. An “etched surface” refers to a surface that is made by etching.


In some embodiments, the substantially planar solid surface comprises etchings arranged in spaced relation to each other (e.g., into clusters of a discrete number of spots (see, e.g., FIGS. 1C and 8B)). In some embodiments, the substantially planar solid surface comprises etchings with a geometric pattern. The arrangement and/or geometric pattern may be programmable. For example, a geometric pattern may be designed by a person of ordinary skill in the art based on the goal of the proteomic analysis, sample multiplexing strategy, etc., or a combination thereof. Suitable geometric patterns may include about 1-120 clusters per substantially planar solid surface, for example, about: 18, 36, 54, 72, 90 or 108 clusters per surface; and each cluster may include about 1-20 spots, for example, about: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 spots. In some embodiments, each cluster has at least 14 spots. In certain embodiments, the geometric pattern includes 36 clusters with at least 14 spots per cluster. In particular embodiments, the geometric pattern includes 36 clusters with at least 16 spots per cluster.


In some embodiments, the substantially planar solid surface is unetched.


In some embodiments, the distance between two spots (e.g., two closest spots) within a cluster is about 0.1-10.0 mm, for example, about: 0.1, 0.15, 0.2, 0.25, 0.3, 0.35, 0.4, 0.45, 0.5, 0.55, 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5 or 10.0 mm. In certain embodiments, the distance between two spots (e.g., two closest spots) within a cluster is about: 0.1-9.5, 0.15-9.5, 0.15-9.0, 0.2-9.0, 0.2-8.5, 0.25-8.5, 0.25-8.0, 0.3-8.0, 0.3-7.5, 0.35-7.5, 0.35-7.0, 0.4-7.0, 0.4-6.5, 0.45-6.5, 0.45-6.0, 0.5-6.0, 0.5-5.5, 0.55-5.5, 0.55-5.0, 0.6-5.0, 0.6-4.5, 0.65-4.5, 0.65-4.0, 0.7-4.0, 0.7-3.5, 0.75-3.5, 0.75-3.0, 0.8-3.0, 0.8-2.5, 0.85-2.5, 0.85-2.0, 0.9-2.0, 0.9-1.5, 0.95-1.5 or 0.95-1.0. In particular embodiments, the distance between two spots (e.g., two closest spots) within a cluster is about 1.0 mm.


In some embodiments, the distance between the centers of two clusters (e.g., two neighboring clusters) is about 3.0-50 mm, for example, about: 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 8.0, 10, 15, 20, 30, 40 or 50 mm. In certain embodiments, the distance between the centers of two clusters (e.g., two neighboring clusters) is about: 3.0-40, 3.5-40, 3.5-30, 4.0-30, 4.0-20, 4.5-20, 4.5-15, 5.0-15, 5.0-10, 5.5-10, 5.5-8 or 6-8. In particular embodiments, the distance between the centers of two clusters (e.g., two neighboring clusters) is about 6 mm.


The distance between two spots (e.g., two closest spots) within a cluster and/or the distance between the centers of two clusters (e.g., two neighboring clusters) may be designed by a person of ordinary skill in the art based on the goal of the proteomic analysis, sample multiplexing strategy and/or desired throughput.


Methods disclosed herein can be compatible with many types of substantially planar solid surfaces with a wide range of sizes. In some embodiments, the length of the substantially planar solid surface is about 10 mm to 50 cm, for example, about: 20 mm to 50 cm, 20 mm to 25 cm, 40 mm to 25 cm, 40 mm to 12 cm, 50 mm to 12 cm, 50 mm to 10 cm, 100 mm to 10 cm, 100 mm to 5 cm, 200 mm to 5 cm, 200 mm to 2.5 cm, 500 mm to 2.5 cm or 500 mm to 1.0 cm.


In certain embodiments, the width of the substantially planar solid surface is about 5.0 mm to 30 cm, for example, about: 10 mm to 30 cm, 20 mm to 30 cm, 20 mm to 15 cm, 50 mm to 15 cm, 50 mm to 10 cm, 100 mm to 10 cm, 100 mm to 5.0 cm, 200 mm to 5.0 cm, 200 mm to 2.5 cm, 500 mm to 2.5 cm, 500 mm to 2.0 cm or 1.0 to 2.0 cm.


In particular embodiments, the substantially planar solid surface is provided by microscopic glass slides with dimensions of 75 mm by 25 mm (3″ by 1″) and about 1 mm thickness.


In certain embodiments, the substantially planar solid surface is coated with a compound (e.g., a compound that is neither hydrophobic nor hydrophilic) to stabilize the individual droplets. In particular embodiments, the substantially planar solid surface is fluorocarbon-coated. The term “fluorocarbon” refers to a compound formed by replacing one or more of the hydrogen atoms in a hydrocarbon with fluorine atoms.


In certain embodiments, movement of the substantially planar solid surface is minimized.


Lysing Single Cells

Lysing single cells comprises dispensing n droplets of lysis buffer onto the substantially planar solid surface (e.g., etched or unetched uniformed glass slide), wherein n≥2; and dispensing a single cell into each of the n droplets of lysis buffer to produce n droplets, each comprising a lysed single cell.


As used herein, the term “liquid droplet” refers to a very small drop of a liquid. In some embodiments, each individual droplet comprising the lysis buffer has a volume of about 1.0-10.0 nl, for example, about: 1.0, 2.0, 3.0, 4.0, 5.0, 6.0, 7.0, 8.0, 9.0, 10.0, 1.0-4.0, 1.0-6.0, 1.0-8.0, 2.0-4.0, 2.0-6.0, 2.0-8.0, 2.0-10.0, 4.0-6.0, 4.0-8.0, 4.0-10.0, 6.0-8.0, 6.0-10.0 or 8.0-10.0 nl. In certain embodiments, each individual droplet comprising the lysis buffer has a volume of about 10.0 nl or less, for example, about: 9.5, 9.0, 8.5, 8.0, 7.5, 7.0, 6.5, 6.0, 5.5, 5.0, 4.5 or 4.0 nl or less. In some embodiments, each individual droplet comprising the lysis buffer has a volume of about 4 nl. In particular embodiments, each individual droplet comprising the lysis buffer has a volume of about 8 nl.


In certain embodiments, the individual droplets of lysis buffer are dispensed using a first piezo dispensing capillary (PDC), for example, that of cellenONER (SCIENION GmbH, Berlin, Germany). In some embodiments, the first PDC is dedicated for handling organic solvents, protein solutions, or a combination thereof. In other embodiments, the individual droplets of lysis buffer are dispensed with MANTISR Liquid Handler (FORMULATRIXR, Bedford, MA) or HP D300e Digital Dispenser (Hewlett-Packard, Palo Alto, CA).


In some embodiments, n is >3, for example, >4, >5, >6, >7, >8, >9, >10, ≥11, ≥12, ≥13, >14, ≥15, ≥16, ≥17, >18, >19 or >20. In certain embodiments, n is about 2-20, for example, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20, or 2-18, 3-18, 3-16, 4-16, 4-14, 5-14, 5-12, 6-12 or 6-10. In particular embodiments, n is about 12-20. In more particular embodiments, n is about 14-18. In some embodiments, the n droplets are arranged in spaced relation to each other (e.g., into a cluster (see, e.g., FIGS. 1C and 8B)).


In some embodiments, the method comprises dispensing m times n droplets of lysis buffer onto a substantially planar solid surface, wherein n (corresponding to the number of droplets per subgroup/cluster)>2, and m (corresponding to the number of subgroups/clusters)≥2.


For example, a multiplexing format may be designed by a person of ordinary skill in the art based on the goal of the proteomic analysis, sample multiplexing strategy, etc., or a combination thereof. A suitable multiplexing format may include about 1-120 clusters per substantially planar solid surface, for example, about: 18, 36, 54, 72, 90 or 108 clusters per substantially planar solid surface; and each cluster may include about 1-20 droplets, for example, about: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 droplets. In certain embodiments, the multiplexing format comprises at least about 10 clusters, for example, at least about: 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 90 or 100 clusters.


In some embodiments, each cluster has at least about 6 droplets, for example, at least about: 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16 droplets. In particular embodiments, the multiplexing format includes about 14 droplets per cluster. In more particular embodiments, the multiplexing format includes about 16 droplets per cluster.


In certain embodiments, the multiplexing format includes at least 10 clusters, and each cluster comprising at least 10 droplets (e.g., 14-16 droplets). In particular embodiments, the multiplexing format includes 36 clusters with 14 droplets per cluster.


In some embodiments, a total of about 100-10,000 individual droplets comprising the lysis buffer are dispensed onto the substantially planar solid surface, for example, about: 100-9,000, 150-9,000, 150-8,000, 200-8,000, 200-6,000, 300-6,000, 300-5,000, 500-5,000, 500-4,000, 750-4,000, 750-3,000, 1,000-3,000, 1,500-3,000, 1,500-2,000 or 2,000-3,000 individual droplets. In certain embodiments, about 2,000 (e.g., 2016) individual droplets are dispensed onto the substantially planar solid surface.


In some embodiments, the lysis buffer is devoid of any compound incompatible with the proteomic analysis (e.g., mass spectrometry (MS)). In certain embodiments, the method is devoid of one or more steps for removing one or more incompatible compounds (“cleanup steps”).


In certain embodiments, the lysis buffer comprises a mass-spec compatible organic solvent and/or detergent, such as acetonitrile, n-Dodecyl-ß-D-maltopyranoside (DDM), n-Decyl-B-D-maltopyranoside (DM) and Rapigest.


In some embodiments, the lysis buffer comprises a compound compatible with the intended proteomic analysis (e.g., MS). In certain embodiments, the compound has a vapor pressure of about 0.500-0.700 mm Hg or less at 25° C. In particular embodiments, the compound has a vapor pressure of about 0.600 mm Hg at 25° C. In more particular embodiments, the compound is an organosulfur compound, for example, dimethyl sulfoxide (DMSO).


In some embodiments, the lysis buffer comprises 33-100% DMSO, for example, 40-100%, 50-100%, 60-100%, 70-100%, 80-100%, 90-100%, 92-100%, 94-100%, 95-100%, 96-100%, 97-100%, 98-100% or 99-100% DMSO. In certain embodiments, the lysis buffer comprises about 4.0-8.0 nl of 90-100% DMSO. In particular embodiments, the lysis buffer comprises (e.g., consists of) about 4.0 nl 90-100% DMSO. In more particular embodiments, the lysis buffer comprises (e.g., consists of) about 8.0 nl 90-100% DMSO.


In some embodiments, a perimeter of water (e.g., mass spectrometry grade water) droplets is dispensed in a perimeter surrounding each grid (see, e.g., FIGS. 1D and 8B) to provide local humidity and, thus, reaction volume control. In particular embodiments, the system is set to refresh the water droplet perimeter to control local humidity, e.g., periodically (e.g., every 40 minutes).


Single-Cell Dispensation

In some embodiments, the single cell is a prokaryotic cell. In certain embodiments, the single cell is a eukaryotic cell (e.g., an animal cell, a plant cell, a fungus cell, or a protist cell). Non-limiting examples of animals include humans, domestic animals, such as laboratory animals (e.g., cats, dogs, monkeys, pigs, rats, mice, etc.), household pets (e.g., cats, dogs, rabbits, etc.), livestock (e.g., pigs, cattle, sheep, goats, horses, etc.), and non-domestic animals. In particular embodiments, the single cell is a mammalian cell (e.g., a human cell).


In some embodiments, the single cell is a germ-line cell. In certain embodiments, the single cell is a somatic cell. Non-limiting examples of somatic cells include stem cells, red blood cells, white blood cells (e.g., neutrophils, eosinophils, basophils, or lymphocytes), platelets, nerve cells, neuroglial cells, muscle cells (e.g., skeletal muscle cells, cardiac muscle cells, or smooth muscle cells), cartilage cells, and skin cells. In certain embodiments, the individual cells comprise tumor cells (e.g., melanoma cells).


In some embodiments, the single cell has a diameter of less than 100 μm. In certain embodiments, the single cell has a diameter of about 10-20 μm. In particular embodiments, the single cell has a diameter of about 10-15 μm.


In some embodiments, the single-cell proteomic sample comprises peptides from at least two cells, for example, from at least about: 10, 15, 20, 30, 50, 80, 100, 150, 200, 250, 300, 500, 750, 1,000, 1,500, 2,000, 2,500, 3,000, 4,000, 5,000, 6,000, 7,000, 8,000 or 10,000 cells. In certain embodiments, the single-cell proteomic sample comprises peptides from about 10-10,000 cells, for example, about: 10-9,000, 15-9,000, 15-8,000, 30-8,000, 30-6,000, 50-6,000, 50-5,000, 100-5,000, 100-4,000, 150-4,000 or 150-3,000 cells. In some embodiments, the single-cell proteomic sample comprises peptides from at least 100 cells. In certain embodiments, the single-cell proteomic sample comprises peptides from at least 1,000 cells. In particular embodiments, the single-cell proteomic sample comprises peptides from at least 1,500 cells.


In some embodiments, the cells are a homogenous cell population (of the same cell type). In other embodiments, two or more cell types are dispensed into the n droplets of lysis buffer, for example, 3, 4, 5, 6, 7, 8, 9 or 10 or more cell types. Each cell type may comprise multiple subpopulations based on certain characteristics, for example, cell division cycle (CDC). In particular embodiments, the method further comprises enriching a subpopulation of cells, for example, with Fluorescence-activated cell sorting (FACS) (e.g., based on size, DNA content, cellular state, and/or surface marker), culture condition, reporter-based selection, or a combination thereof.


In some embodiments, (isolating and) dispensing the single cell uses a second piezo dispensing capillary (PDC), for example, that of cellenONER (SCIENION GmbH, Berlin, Germany). In some embodiments, the second PDC is dedicated to handling cell suspensions. In other embodiments, (isolating and) dispensing the single cell uses MANTISR Liquid Handler (FORMULATRIXR, Bedford, MA) or HP D300e Digital Dispenser (Hewlett-Packard, Palo Alto, CA).


In some embodiments, step b) comprises dispensing the single cell in a buffer (e.g., phosphate buffered saline (PBS)) with a measured volume. In certain embodiments, the measured volume is from about 30 picoliters to about 3,000 picoliters, for example, about: 30-2,400, 45-2,400, 45-1,800, 60-1,800, 60-1,200, 90-1,200, 90-900, 100-1,000, 120-900, 120-600, 150-600, 150-450, 200-450, 200-400, 200-300, 250-350, 260-340, 270-330, 280-320, 290-310 or 300-450 picoliters. In certain embodiments, the measured volume is less than 3,000 picoliters, for example, less than: 2,500, 2,400, 2,000, 1,800, 1,500, 1,200, 1,000, 800, 500, 450 or 400 picoliters. In particular embodiments, the measured volume is about 300 picoliters.


In certain embodiments, step b) comprises dispensing the single cell in a cell suspension buffer with a volume of about 100-1,000 picoliters. In particular embodiments, step b) comprises dispensing the single cell in a cell suspension buffer with a volume of about 300 picoliters.


In some embodiments, the method further comprises dispensing a cell suspension buffer devoid of any cell into one or more droplets of lysis buffer, for example, as a negative control for detecting background noise, contamination, etc., or a combination thereof.


Cell Lysis

In some embodiments, step b) enables lysing the single cell in a total volume of about 5.0-12.0 nl, for example, of about: 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.5, 11.0, 11.5, 12.0, 5.5-12.0, 5.5-11.5, 6.0-11.5, 6.0-11.0, 6.5-11.0, 6.5-10.5, 7.0-10.5, 7.0-10.0, 7.5-10.0, 7.5-9.5, 8.0-9.5 or 8.0-8.5 nl. In particular embodiments, step b) enables lysing the single cell in a total volume of about 7.5-8.5 nl. In particular embodiments, step b) enables lysing the single cell in a total volume of about 8.0-8.5 nl.


In certain embodiments, step b) enables lysing the single cell for about 10-20 minutes. In some particular embodiments, step b) enables lysing the single cell in a total volume of about 8-8.5 nl for about 10-20 minutes.


In some embodiments, 5.0-12.0 μl is the sum of the volume of the lysis buffer plus the volume of the single cell in its dispensing solution, for example, about: 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.5, 11.0, 11.5, 12.0, 5.5-12.0, 5.5-11.5, 6.0-11.5, 6.0-11.0, 6.5-11.0, 6.5-10.5, 7.0-10.5, 7.0-10.0, 7.5-10.0, 7.5-9.5, 8.0-9.5 or 8.0-8.5 nl. In particular embodiments, 4-10 μl is the sum of the volume of the lysis buffer plus the volume of the single cell in its dispensing solution.


Protein Digestion

In certain embodiments, the digestion buffer is a trypsin buffer, and dispensing digestion buffer into each of the n droplets produces a solution comprising about 100-150 ng/μl trypsin. In particular embodiments, dispensing digestion buffer into each of the n droplets produces a solution comprising about 120 ng/μl trypsin in about 5 mM HEPES buffer.


In certain embodiments, dispensing digestion buffer into each of the n droplets produces a solution with a volume of about 15-25 nl, for example, about: 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 15-20, 16-20, 16-19, 17-19 or 18-19 nl. In particular embodiments, dispensing digestion buffer into each of the n droplets produces a solution with a volume of about 18 nl.


In some embodiments, step c) comprises enabling the proteins from each lysed single cell to be digested at about 1ºC above the dew point, for example, about: 0.4-1.6° C., 0.5-1.5° C., 0.6-1.4° C., 0.7-1.3ºC, 0.8-1.2° C. or 0.9-1.1° C. above the dew point. In certain embodiments, step c) comprises enabling the proteins from each lysed single cell to be digested at a relative humidity of about 70-80%, for example, about: 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 71-79%, 72-78%, 73-77%, 74-76%, or 74.5-75.5%. As used herein, the term “relative humidity” refers to the amount of water vapor present in air expressed as a percentage of the amount needed for saturation at the same temperature. In particular embodiments, step c) comprises enabling the proteins from each lysed single cell to be digested at a relative humidity of about 75%. In more particular embodiments, step c) comprises enabling the proteins from each lysed single cell to be digested at about 1ºC above the dew point and at a relative humidity of about 75%. In some embodiments, the temperature, the relative humidity, or both are dynamically regulated.


In some embodiments, step c) comprises enabling the proteins from each lysed single cell to be digested for about 3-5 hours, for example, for about: 3, 3.5, 4, 4.5, 5, 3.1-4.9, 3.2-4.8, 3.3-4.7, 3.4-4.6, 3.5-4.5, 3.6-4.4, 3.7-4.3, 3.8-4.2 or 3.9-4.1 hours.


In some embodiments, step c) comprises:

    • dispensing about 15-25 nl of about 120 ng/μl trypsin into each of the n droplets; and
    • enabling the proteins from each lysed single cell to be digested at about 1° C. above the dew point and a relative humidity of about 75% for about 4-5 hours.


In certain embodiments, the digestion buffer is dispensed using the first piezo dispensing capillaries (PDC), for example, that of cellenONER (Lyon, France). In other embodiments, the digestion buffer is dispensed using MANTISR Liquid Handler (FORMULATRIXR, Bedford, MA) or HP D300e Digital Dispenser (Hewlett-Packard, Palo Alto, CA).


Single-Cell Proteomics

In some embodiments, the one or more single-cell proteomic samples are intended for tandem mass spectrometry. Tandem mass spectrometry, also referred to herein as MS/MS or MS2, involves multiple steps of mass spectrometry selection, with some 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 known to those skilled in the art. The resulting ions are then separated and detected in a second stage of mass spectrometry (MS2). A common use is for analysis of proteins and peptides.


In certain embodiments, the one or more single-cell proteomic samples are intended for quantitative proteomics. Quantitative proteomics can be used, for example, to determine the relative or absolute amount of proteins in a sample.


Several quantitative proteomics methods are based on MS/MS. One method commonly used for quantitative proteomics is isobaric tag labeling. Isobaric tag labeling enables simultaneous identification and quantification of proteins from multiple samples in a single analysis. To quantify proteins, peptides are labeled with chemical tags that have the same structure and nominal mass, but vary in the distribution of heavy isotopes in their structure. These tags, commonly referred to as tandem mass tags (TMT), are designed so that the mass tag is cleaved at a specific linker region upon higher-energy collisional-induced dissociation during tandem mass spectrometry, yielding reporter ions of different masses. Protein quantitation is accomplished by comparing the intensities of the reporter ions in the MS/MS spectra.


MS/MS can also be used for protein sequencing, as is understood by those skilled in the art. When intact proteins are introduced to a mass analyzer, it is termed “top-down proteomics,” and when proteins are digested into smaller peptides and subsequently introduced into the mass spectrometer, it is termed “bottom-up proteomics”. Shotgun proteomics is a variant of bottom-up proteomics in which proteins in a mixture are digested prior to separation and tandem mass spectrometry.


In some embodiments, the one or more single-cell proteomic samples are generated for cell classification, uncovering a regulatory process, associating a regulatory process with a functional outcome, or a combination thereof. In particular embodiments, the one or more single-cell proteomic samples are generated for understanding cell cycle regulation. In some embodiments, the one or more single-cell proteomic samples are generated for identifying proteins whose abundance differs in G1, S, and/or G2/M phase for two or more cell types.


In some embodiments, the one or more single-cell proteomic samples comprise 10 or more cells of the same cell type to minimize batch effects, background noise, or a combination thereof. In certain embodiments, the one or more single-cell proteomic samples comprise at least 10 cells of the same cell type, for example, at least: 15 cells, 20 cells, 30 cells, 50 cells, 80 cells, 100 cells, 150 cells, 200 cells, 250 cells, 300 cells, 500 cells, 750 cells, 1,000 cells, 1,500 cells, 2,000 cells, 2,500 cells or 3,000 cells of the same cell type. In certain embodiments, the one or more single-cell proteomic samples comprise about 10-10,000 cells of the same cell type, for example, about: 10-9,000 cells, 15-9,000 cells, 15-8,000 cells, 30-8,000 cells, 30-6,000 cells, 50-6,000 cells, 50-5,000 cells, 100-5,000 cells, 100-4,000 cells, 150-4,000 cells, 150-3,000, 500-3,000, 1,000-3,000, 1,000-2,500, 1,000-2,000, 1,500-3,000, 1,500-2,500 or 1,500-2,000 cells of the same cell type. In particular embodiments, the one or more single-cell proteomic samples comprise about 1,500-2,000 cells.


In some embodiments, the disclosed methods enable performing parallel sample preparation of multiple (e.g., hundreds or thousands of) single cells; obviating sample cleanup and associated losses; minimizing bias for cellular compartments; supporting accurate relative protein quantification, or a combination thereof.


In some embodiments, the disclosed methods further comprise performing at least one proteomic analysis on the single-cell proteomic sample. In particular embodiments, the at least one single-cell proteomic analysis enables identifying and/or quantifying protein covariation across the single cells.


In certain embodiments, the single-cell proteomic analysis is performed on a non-substantially planar solid surface, for example, in a multi-well plate or in a tube (such as a microfuge tube).


Peptide Labeling

Peptide labeling comprises dispensing a chemical tag into each of the n droplets comprising the peptides to produce labeled peptides, wherein at least one droplet of the n droplets receives a different chemical tag from at least one other droplet of the n droplets, thereby enabling the labeled peptides in the at least one droplet to be distinguishable from the labeled peptides in the at least one other droplet (e.g., to be distinguishable from labeled peptides in any other droplet within a cluster/subgroup).


In particular embodiments, each droplet of the n droplets receives a unique chemical tag, thereby enabling the labeled peptides in each droplet to be distinguishable from the labeled peptides in each other droplet.


In isobaric labeling for tandem mass spectrometry, proteins are extracted from cells, digested, and labeled with tags of the same mass. When fragmented during MS/MS, the reporter ions show the relative amount of the peptides in the samples.


In some embodiments, the chemical tag comprises (e.g., consists of) an isobaric tag. Two commercially available isobaric tags are iTRAQR and tandem mass tag (TMT) reagents. A TMT comprises four regions: mass reporter, cleavable linker, mass normalization, and protein reactive group. TMT reagents can be used to simultaneously analyze, e.g., 2-18 different peptide samples prepared from individual cells. TMT reagents include three types: (1) a reactive NHS ester functional group for labeling primary amines (e.g., TMTduplex™, TMTTMsixplex™, TMT10plex plus™, TMT11-131C™, TMTpro 16plex, TMTpro 18plex,), (2) a reactive iodoacetyl functional group for labeling free sulfhydryls (e.g., iodoTMT™) and (3) reactive alkoxyamine functional group for labeling of carbonyls (e.g., aminoxyTMT™).


In certain embodiments, the peptides are labeled by isobaric mass tags (e.g., TMT or TMTpro) for multiplexed analysis. In particular embodiments, the chemical tag comprises (e.g., consists of) TMTpro 16plex or TMTpro 18plex.


In certain embodiments, the chemical tag comprises (e.g., consists of) an isobaric tag for relative and absolute quantitation (iTRAQR). ITRAQR is a reagent for tandem mass spectrometry that is used to determine the amount of proteins from different sources in a single experiment. iTRAQ® uses stable isotope labeled molecules that can form a covalent bond with the N-terminus and side chain amines of proteins. The iTRAQR reagents are used to label peptides from different samples that are pooled and analyzed by liquid chromatography and tandem mass spectrometry. The fragmentation of the attached tag generates a low molecular mass reporter ion that can be used to relatively quantify the peptides and the proteins from which they originated.


This sample preparation methods described herein are also compatible with non-isobaric mass tags, for example, as demonstrated with mTRAQ (FIG. 6 of Derks et al., bioRxiv 467007 (doi.org/10.1101/2021.11.03.467007) (2021).


In some embodiments, the methods further comprise reducing the volumes of the individual droplets before labeling (e.g., by drying down the individual droplets). In certain embodiments, the volumes of the individual droplets are reduced to about 3-5 nl before dispensing the chemical tag into the corresponding droplet comprising the peptides, for example, about 3.0, 3.5, 4.0, 4.5, 5.0, 3.1-4.9, 3.2-4.8, 3.3-4.7, 3.4-4.6, 3.5-4.5, 3.6-4.4, 3.7-4.3, 3.8-4.2 or 3.9-4.1 nl. In particular embodiments, the volumes of the individual droplets are reduced to about 4 nl before dispensing the chemical tag into the corresponding droplet comprising the peptides.


In certain embodiments, step d) comprises dispensing a chemical tag in a volume of about 15-25 nl into each of the n droplets comprising the peptides, for example, the volume is about: 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 16-24, 17-23, 18-22, 19-21, 19.5-20.5, 19.6-20.4, 19.7-20.3, 19.8-20.2 or 19.9-20.1 nl. In some embodiments, step d) comprises dispensing a chemical tag in a volume of about 20 nl into each of the n droplets comprising the peptides. In particular embodiments, step d) comprises dispensing TMTpro™ (e.g., “light” version of TMTpro™ 14plex or TMTpro™ 16plex) in a volume of about 20 nl into each of the n droplets comprising the peptides.


In some embodiments, the chemical tag (e.g., TMT) is dissolved in DMSO. In certain embodiments, the chemical tag comprises TMT label reagents (such as of TMTpro™ 14plex or TMTpro™ 16plex) dissolved in DMSO. In particular embodiments, the chemical tag comprises a “light” version of TMT label reagents, also known as TMTO, dissolved in DMSO. In certain embodiments, the chemical tag comprises a “heavy” version of TMT label reagents, also known as TMT super heavy TMTsh, dissolved in DMSO.


In some embodiments, the concentration of the chemical label (e.g., TMTpro™ 14plex) is about 28 mM.


In some embodiments, step d) comprises enabling the chemical tag to react with the peptides at room temperature. In certain embodiments, step d) comprises enabling the chemical tag to react with the peptides at about 18-25° C., for example, at about: 18, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 22.5, 23, 23.5, 24, 24.5, 25, 18.5-25, 19-24.5, 19.5-24, 20-23.5, 20.5-23, 21-22.5 or 21.5-22° C. In particular embodiments, step d) comprises enabling the chemical tag to react with the peptides at about 20-23.5° C.


In certain embodiments, step d) comprises enabling the chemical tag to react with the peptides at in a total volume of about 18-30 nl, for example, about: 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 19-29, 20-28, 21-27, 22-26, 23-25, 23-24 or 24-25 nl. In particular embodiments, step d) comprises enabling the chemical tag to react with the peptides in a total volume of about 24 nl.


In certain embodiments, dispensing a chemical tag into each of the n droplets comprising the peptides uses the first piezo dispensing capillaries (PDC), for example, that of cellenONER (SCIENION GmbH, Berlin, Germany). In other embodiments, dispensing a chemical tag into each of the n droplets uses MANTIS® Liquid Handler (FORMULATRIX®, Bedford, MA) or HP D300e Digital Dispenser (Hewlett-Packard, Palo Alto, CA).


In certain embodiments, greater than 90.0% of all peptides are labeled with the (corresponding) chemical tag, for example, greater than: 92.5%, 95.0%, 96.0%, 97.0%, 98.0%, 99.0%, 99.5%, 99.8% or 99.9% of all peptides are labeled. In particular embodiments, greater than 99% of all peptides are labeled.


Quenching the Labeling Reactions

In some embodiments, the methods of the disclosure further comprise dispensing a quenching reagent into each of the n droplets to quench unconjugated chemical tag.


In certain embodiments, the quenching reagent comprises about 20-30 nl of 5% hydroxylamine.


In particular embodiments, step d) further comprises:

    • dispensing 20 nl of 5% hydroxylamine into each of the n droplets, and quenching unconjugated chemical tag for about 20 minutes; and
    • dispensing 30 nl of 5% hydroxylamine into each of the n droplets, and quenching unconjugated chemical tag for about 20 minutes.


In some embodiments, step d) further comprises enabling unconjugated chemical tag to be quenched at about 1ºC above the dew point, for example, about: 0.4-1.6° C., 0.5-1.5° C., 0.6-1.4ºC, 0.7-1.3ºC, 0.8-1.2° ° C. or 0.9-1.1ºC above the dew point. In certain embodiments, step d) further comprises enabling unconjugated chemical tag to be quenched at a relative humidity of about 70-80%, for example, about: 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 71-79%, 72-78%, 73-77%, 74-76%, or 74.5-75.5%. In particular embodiments, step d) further comprises enabling unconjugated chemical tag to be quenched at about 1ºC above the dew point and at a relative humidity of about 75%. In some embodiments, the temperature, the relative humidity, or both are dynamically regulated.


Pooling (Merging)

Pooling comprises applying a fluid to merge at least a subset the n droplets into a combined droplet on the substantially planar surface, thereby combining the labeled peptides to form a single-cell proteomic sample. In some embodiments, the fluid is water. In certain embodiments, the fluid has a volume of about 1 μl.


In some embodiments, the at least a subset the n droplets comprise n droplets. In certain embodiments, the at least a subset the n droplets comprise ≤n-1 droplets. In particular embodiments, the at least a subset the n droplets comprise ≤n-2 droplets.


In certain embodiments, step e) further comprises aspirating each combined droplet off the substantially planar solid surface in an acetonitrile solution. In some embodiments, the acetonitrile solution comprises about 100% acetonitrile, for example, about: 99.0-100%, 99.5-100%, 99.8-100% or 99.9-100% acetonitrile. In particular embodiments, the acetonitrile solution has a volume of about 5-15 μl, for example, about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 9.0-11.0, 9.1-10.9, 9.2-10.8, 9.3-10.7, 9.4-10.6, 9.5-10.5, 9.6-10.4, 9.7-10.3, 9.8-10.2 or 9.9-10.1 μl. In more particular embodiments, the total volume for aspirating each combined droplet is about 10 μl. Each single-cell proteomic sample can be transferred into a single well of a multi-well (e.g., a 384-well) plate.


In some embodiments, the combined droplet (comprising the labeled peptides) is transferred onto a non-substantially planar solid surface. In certain embodiments, the combined droplet (comprising the labeled peptides) is transferred into a container (e.g., a well within a multi-well plate, a tube such as a microfuge tube).


Drying

In some embodiments, the method further comprises drying the single-cell proteomic samples, for example in a speed-vacuum.


In some embodiments, the one or more single-cell proteomic sample are stored (for example, frozen at −80° C.) for future proteomic analysis. In certain embodiments, the one or more single-cell proteomic sample are reconstituted (for example, each in about 1.1 μl of 0.1% formic acid) for proteomic analysis (e.g., mass spectrometry analysis).


In another aspect, the disclosure provides a single-cell proteomic sample formed with any one of the methods described herein.


Many biological processes and regulatory dynamics, such as the cell division cycle, are reflected in protein covariation across single cells. Variabilities within a cell type are challenging to analyze with existing single-cell omics methods. In some embodiments, the sample preparation methods described herein enable quantifying and interpreting the covariations by single-cell proteomics with sufficiently high throughput and accuracy. As shown below, the sample preparation methods have been used to prepare 1,888 single cells and 128 negative controls in a single batch. Their analysis enabled quantifying the covariation among thousands of proteins and cell-cycle protein markers. The results demonstrate that protein covariation across single cells may reveal functionally concerted biological differences between closely related cell states.


A substantially planar solid surface enables parallel processing of a large number of multiplexed single-cell samples at a high density, thereby significantly increasing the throughput of single-cell proteomic analysis. Said surface also enables efficient merging of each multiplexed single-cell proteomic sample, thereby significantly reducing sample loss and sample processing time. A substantially planar solid surface also enables precise dispensing of very small volumes of single cells and reagents and keeping the droplets separated.


Single cells are isolated in very small volumes (e.g., about 300 picoliter), and all preparation steps, including cell lysing, protein digesting, and peptide labeling are performed in droplets of small volumes (e.g., below about 20 nl) on a substantially planar surface. Reduced volumes during sample preparation and increased throughput result in reductions in background signal, increased sample consistencies, and increased sensitivities.


EXAMPLES

Single-cell measurements are commonly used to identify different cell types from tissues composed of diverse cells (Regev et al., Elife 6:e27041 (2017) and Specht & Slavov, J Proteome Res. 17(8):2565-71 (2018)). This analysis is powering the construction of cell atlases, which can pinpoint cell types affected by various physiological processes. This cell classification requires analyzing a large number of cells and may tolerate measurement errors (Regev et al., Elife 6:e27041 (2017), Ziegenhain et al., Mol Cell 65(4):631-43 (2017), and Slavov, Science 367(6477):512-13 (2020)). In addition to classifying cells by type, single-cell measurements may reveal regulatory processes within a cell type and even associate them with different functional outcomes (Slavov, PLOS Biol. 20(1):e3001512 (2022), Shaffer et al., Nature. 546(7658):431-35 (2017) and Emert et al., Nat Biotechnol. 39(7):865-76 (2021)). For example, the covariation among proteins across single cells from the same type may reflect cell intrinsic dynamics, such as the cell division cycle (Slavov, PLOS Biol. 20(1):e3001512 (2022) and Mahdessian et al., Nature 590(7847):649-54 (2021)). Furthermore, protein covariation may reflect protein interactions within complexes or cellular states, such as senescence (Slavov, PLOS Biol. 20(1):e3001512 (2022)). However, estimating and interpreting protein covariation within a cell type requires high quantitative accuracy and high throughput (Slavov, PLOS Biol. 20(1):e3001512 (2022) and Slavov, Mol Cell Proteomics 21(1):100179 (2022)). Indeed, protein differences within a cell type are smaller than differences across cell types and can be easily swamped by batch effects and measurement noise. A goal is to minimize measurement noise to levels consistent with estimating and interpreting protein covariation across single cells from the same cell type. Towards this goal, an aim was to reduce batch effects and background noise, since these factors undermine the accuracy of single-cell proteomics by mass spectrometry (MS) (Slavov, Curr Opin Chem Biol. 60:1-9 (2021), Vanderaa & Gatto, Expert Rev Proteomics 18(10):835-43 (2021), Kelly, Mol Cell Proteomics 19(11): 1739-48 (2020), and Specht et al., Genome Biol. 22(1):50 (2021)). Specifically, an aim was to develop a widely accessible, robust, and automated sample preparation method that reduces volumes to a few nanoliters. A goal was to perform parallel sample preparation of thousands of single cells to increase the size of experimental batches and thus reduce batch effects (Vanderaa & Gatto, Expert Rev Proteomics 18(10):835-43 (2021), Klein et al., Cell 161(5):1187-201 (2015) and Macosko et al., Cell 161(5):1202-14 (2015)). To achieve high precision, an aim is to avoid any movement of the samples during the sample preparation stage, so that 1-10 nl volumes of reagents can be repeatedly dispensed to each droplet containing a single cell. The CellenONE cell sorting and liquid handling system was used to develop nano-Proteomic sample Preparation (nPOP), which allowed a 100-fold reduction of the sample volumes over the Minimal ProteOmic sample Preparation (mPOP) method (Specht et al., Genome Biol. 22(1):50 (2021), Harrison et al., bioRxiv 399774 (2018), Petelski et al., Nat Protoc. 16(12):5398-25 (2021) and Marx, Nat Methods 16(9):809-12 (2019)). nPOP enabled analysis of protein covariation within two cell lines, monocytes and melanoma. This enabled classifying cells by cell division cycle (CDC) phase and identifying a sub-population of melanoma cells. Comparative analysis between the cell lines identifies both similar and differential patterns of CDC associated protein covariation. Further, this analysis was applied within melanoma sub-populations, and differences in CDC associated protein covariation as well as a differential distribution of cells throughout phases of the CDC were identified.


Example 1. Methods
Cell Culture

U-937 and Jurkat cells were grown as suspension cultures in RPMI medium (HyClone 16777-145, Cytiva, Marlborough, MA) supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin (pen/strep) (15140122, ThermoFisher, Waltham, MA). Cells were passaged when a density of 106 cells/ml was reached, approximately every two days.


The melanoma cells (WM989-A6-G3, a gift from Arjun Raj, University of Pennsylvania) were grown as adherent cultures in TU2% media which is composed of 80% MCDB 153 (M7403, Sigma-Aldrich, St. Louis, MO), 10% Leibovitz L-15 (11415064, ThermoFisher, Waltham, MA), 2% fetal bovine serum, 0.5% penicillin-streptomycin and 1.68 mM Calcium Chloride (499609, Sigma-Aldrich, St. Louis, MO). Cells were passaged at 80% confluence (approximately every 3-4 days) in T75 flasks (Z707546, MilliporeSigma, Burlington, MA) using 0.25% Trypsin-EDTA (25200072, ThermoFisher, Waltham, MA) and re-plated at 30% confluence.


HPAF-II cells (CRL-1997™, ATCC, Manassas, VA) were cultured in EMEM (30-2003, ATCC, Manassas, VA), CFPAC-I cells (CRL-1918™, ATCC, Manassas, VA) were cultured in IMDM (30-2005), and BxPC-3 cells (CRL-1687™, ATCC, Manassas, VA) were cultured in RPMI 1640 (30-2001, ATCC, Manassas, VA). All media were supplemented with 10% fetal bovine serum (FBS) (F4135, MilliporeSigma, Burlington, MA) and 1% penicillin-streptomycin. Cells were passaged at 70% confluence.


Lysis Validation Experiment

Jurkat cells and U-937 cells cultured in heavy SILAC media (containing+10 Da Arg and +8 Da Lys) were washed and re-suspended in PBS at 20,000 cells per μl. Two solutions of equal cell count containing Jurkat and U-937 cells were made mixed in 1:1 ratios. One sample was lysed by diluting cells in 90% DMSO and the other was lysed in 6M urea. The DMSO cell lysate was diluted to a concentration of 33% DMSO and urea lysate was diluted to 0.5 M. Both solutions were digested in 15 ng/μl of trypsin for 12 hours. Each sample was then desalted using C18 stage tips and run using data dependent acquisition.


Carrier and Reference Channel Preparation in Bulk

The isobaric carrier consisting of a 1:1 mixture of melanoma and monocyte cells was prepared in bulk and aliquoted into carriers corresponding to 200 cells each. A single cell suspension of 22,000 cells was transferred to a 200 μl PCR tube (1402-3900, USA Scientific, Inc., Ocala, FL) and then processed via the mPOP sample preparation method (Harrison et al. bioRxiv 399774 (2018)). The reference channel was made from the same sample.


Bulk Melanoma and Monocyte Samples

Additional bulk samples of melanoma and monocyte cells were prepared for validating quantification of single cells. Cell pellets of 100,000 monocyte and melanoma cells were suspended in 50 μl of mass spectrometry grade water and lysed and digested via mPOP sample preparation (Harrison et al. bioRxiv 399774 (2018)). Samples were then labeled with TMT-16plex, combined, and diluted down to a concentration of 400 cells/μl for analysis by LC-MS.


Reagent Handling with CellenONE


The CellenONE (see, e.g., www.cellenion.com/technology/) was equipped with two piezo dispensing capillaries (PDC). One PDC was dedicated to handle cell suspensions. The other PDC was dedicated for all other reagent handling including organic solvents and protein solutions. Reagents were loaded into a 384-well plate in volumes of 30 μl. When aspirating protein solutions, 20 μl was aspirated to ensure the mixture was not diluted with system water. When dispensing DMSO, it was important to deactivate the humidifier. This allowed residual DMSO left on the tip of the PDC to evaporate quickly so dispensing was not affected. After each sample preparation, PDCs were washed with ethanol and cleaned under sonication to remove any built-up of material from inside of the PDC and ensure optimal performance.


Sample Preparation and Experimental Design

nPOP reactions were carried out on the surface of a fluorocarbon coated glass slide. The array layout was very flexible and adjustable to the experimental parameters. The droplets used for single-cell sample preparation were arranged in clusters, and the number of droplets per cluster equals the number of single cells per SCOPE2 (Single Cell ProtEomics 2) set. TMTpro 18plex and 14 droplets per cluster, corresponding to the 14 isobaric labels used for single cells, were used. The design allowed fitting 36 clusters per slide and 4 glass slides on the temperature controlled target holder, which enabled simultaneous processing of up to 14×36×4=2,016 single cells. Reducing the space between clusters can further increase the number of clusters per slide and thus the number of simultaneously prepared single cells. The array layout was optimized to keep droplets from the same set close in proximity but prevent reaction volumes from merging. Once an array layout was selected, 8 nl of DMSO was dispensed to each location of the array, forming the initial reaction volume for each single cell reaction. Lysis began when cells were dispensed inside a droplet of about 300 pl of PBS into these reaction volumes of DMSO. After lysis, 10 nl of solution containing trypsin and HEPES buffer was added to each reaction volume, for a final concentration of 120 ng/μl of trypsin and 5 mM HEPES and total volume of 18 nl.


The humidifier and cooling system were then turned on to prevent droplet evaporation. Relative humidity inside the CellenONE was set to 75%, and the chiller temperature was set to dynamically chase one degree above the dew point. Mass spectrometry grade water was dispensed in a perimeter surrounding each grid to provide further control for the local humidity of the reaction volumes. The system was set to refresh the water droplet perimeter to control local humidity every 40 minutes for 5 hours as proteins digest.


After proteins were digested for 5 hours, the humidity and cooling controls were turned off. 20 nl of TMT labels suspended in DMSO and concentrated at 28 mM were then dispensed to each reaction volume using the organic dispensing tip. When dispensing labels, humidifier was turned off to assist with dispensing. After single cells were left to label for 1 hour, 20 nl of 5% hydroxylamine solution was added to each reaction volume to quench labeling reaction. Humidity and cooling controls were returned to previous settings for quenching labeling reaction. After 20 minutes, another addition of 30 nl of 5% hydroxylamine was added.


After quenching proceeds for another 20 minutes, sample clusters were pooled by aspirating them off the slide surface in 10 μl of a 100% acetonitrile solution via CellenONE PDC and syringe pump controls. Pooled samples were then transferred into a 384-well plate (AB1384, ThermoFisher, Waltham, MA) and dried down to dryness in a speed-vacuum (Eppendorf, Germany) and either frozen at −80ºC for later analysis or immediately reconstituted in 1.1 μl of 0.1% formic acid (85178, ThermoFisher, Waltham, MA) for mass spectrometry analysis.


DNA Sorting for Bulk CDC Analysis

Melanoma and monocyte cells were incubated using Vy-17 brand Dye Cycle (V35003, ThermoFisher, Waltham, MA) following manufacturer's instructions. Cells were sorted via the Beckman CytoFLEX SRT (Beckman Coulter, Brea, CA). Post sorting, cells were pelleted and washed with Mass Spectrometry grade water and resuspended in water at a concentration of 2000 cells/μl. Cells were then frozen at −80ºC for 10 minutes and then heated to 90ºC for 10 minutes for lysis. Proteins were then digested overnight in a solution of 15 ng/μl of trypsin. Samples were analyzed via data independent acquisition.


LC-MS Platform

MS analysis was designed and performed according to the SCOPE2 guidelines and protocol (Specht et al., Genome Biol. 22(1):50 (2021), Petelski et al., Nat Protoc. 16(12):5398-425 (2021) and Specht & Slavov, J Proteome Res. 20(1):880-87 (2021)). Specifically, the single cells pooled into SCOPE2 sets were separated via online nlC on a Dionex UltiMate 3000 UHPLC; 1 μl out of 1.1 μl of sample was picked up out of a 384-well plate (AB1384, ThermoFisher, Waltham, MA) placed on an auto sampler height adjuster for PCR plates (6820.4089, ThermoFisher, Waltham, MA) and loaded onto a 25 cm×75 μl IonOpticks Aurora Series UHPLC column (AUR2-25075C18A). Buffer A was 0.1% formic acid in water and buffer B was 0.1% formic acid in 80 acetonitrile/20% water. A constant flow rate of 200 nl/min was used throughout sample loading and separation. Samples were loaded onto the column for 20 minutes at 1% B buffer, then ramped to 5 B buffer over two minutes. The active gradient then ramped from 5% B buffer to 25% B buffer over 53 minutes. The gradient was then ramped to 95% B buffer over 2 minutes and stayed at that level for 3 minutes. The gradient then dropped to 1% B buffer over 0.1 minutes and stayed at that level for 4.9 minutes. Loading and separating each sample took 95 minutes total. All samples were analyzed by a Thermo Scientific Q-Exactive mass spectrometer from minute 20 to 95 of the LC loading and separation process. Electrospray voltage was set to 1.8 V, applied at the end of the analytical column. To reduce atmospheric background ions and enhance the peptide signal-tonoise ratio, an Active Background Ion Reduction Device (ABIRD, ESI Source Solutons, LLC, Woburn, MA) was used at the nanospray interface. The temperature of ion transfer tube was 250° C. and the S-lens RF level was set to 80.


Single-Cell MS Data Acquisition

A prioritized analysis workflow (Huffman et al., bioRxiv 484655 (2022)) was used to increase consistency of identification and depth of coverage for the nPOP-prepared single-cell data shown in FIGS. 2A-2E, FIGS. 3A-3C, FIGS. 4A-4I, and FIGS. 5A-5G. A spectral library was built from two injections of a 10× concentrated aliquot of combined carrier and reference sample analyzed by DIA instrument methods 1 and 2, as well as an injection of a 5× concentrated aliquot of combined carrier and reference sample analyzed by DIA method 1. Both of these instrument methods are detailed in the methods section of Huffman, et al. (Huffman et al., bioRxiv 484655 (2022)). A subsequent injection of a 1× concentrated aliquot of carrier and reference sample was analyzed by DIA instrument method 1 to serve as a retention-time-calibration run. The results from this retention-time-calibration run were searched with Spectronaut to generate a prioritized inclusion list for subsequent scout runs and prioritized single-cell analyses. The prioritized inclusion lists were then imported into MaxQuant. Live (v. 2.0.3 with priority tiers) and used to analyze 1× concentrated carrier and reference samples or nPOP prepared single-cell samples, with settings detailed below.


LC-Settings for pSCOPE-Associated Experiments


Samples were analyzed using a 95-minute method with the following gradient characteristics: samples were loaded onto the column at 4% B; the gradient was then ramped to 8% at minute 12, 35% at minute 75, 95% at minute 77, 4% from minute 80.1 onward.


Inclusion-List Generation for Scout Experiments

Spectronaut search results of the retention-timecalibration run were filtered to EG.PEP≤ 0.02 and EG.Qvalue≤ 0.05. Additionally, precursors without TMTPro modifications (+304.2071 Da) on the peptide n-terminus or lysine residue were filtered out. The distribution of precursor intensities for the remaining precursors was then subset into tertiles for use in priority tier assignment. These precursors were then filtered such that a maximum of four peptides per protein were selected, with the most intense peptides per protein being selected. Filtered peptides with precursor intensities in the top intensity tertile were placed on the top priority tier, peptides with intensities in the middle intensity tertile were placed on the middle priority tier, and peptides with intensities in the bottom intensity tertile were placed on the bottom priority tier. All species matching the original EG.PEP and EG.Qvalue filtration characteristics that were not previously selected for a priority tier were assigned a priority below the previous bottom tier. These priority-tier-assigned peptides were then enabled for participation in MaxQuant.Live's realtime-retention-time-alignment algorithm, as well as MS2 upon detection. Any remaining PSMs outside of the original filtration criteria (EG.PEP≤ 0.02 and the EG.Qvalue≤ 0.05) were enabled for participation in MaxQuant.Live's realtime-retention-time-alignment algorithm, but not sent for MS2 upon detection.


Scout Experiment Instrument Method and Raw Data Analysis

1 μl injections of a 1× concentrated aliquot of mixed carrier-reference material were analyzed using the instrument method detailed in the prioritized acquisition parameters section and MaxQuant.Live parameters indicated in the associated table. The two raw files associated with these experiments were then searched using MaxQuant (v. 1.6.17.0) using a FASTA containing all entries from the human SwissProt database (swissprot_human_20211005. fasta, 20,386 proteins). TMTPro 16plex was enabled as a fixed modification on peptide n-termini and lysines via the reporter ion MS2 submenu. Methionine oxidation (+15.99492 Da) and protein n-terminal acetylation (+42.01056 Da) were enabled as variable modifications, and trypsin was selected for in silico digestion with enzyme mode set to specific. Up to 2 missed cleavages were allowed per peptide with a minimum length of 7 amino acids. Second peptide identifications were disabled, calculate peak properties was enabled, and msScans was enabled as an output file. PSM FDR and protein FDR were set to 1.


Pre-Prioritization Shotgun Experiment Instrument Method and Raw Data Analysis

One lul injection of a 1× concentrated aliquot of mixed carrier-reference material was analyzed using the LC settings indicated above. The following MS1 settings were used: 70k resolution, le6 AGC target, 100 ms maximum injection time, and a scan range of 450Th to 1600Th. MS2 scans were acquired with the following settings: 70k resolution, le6 AGC target, 300 ms maximum injection time, loop count (i.e., top-n) of 7, Isolation window of 0.7Th with a 0.3Th offset, fixed first mass of 100 m/z, NCE of 33, and a centroid spectrum data type. The minimum AGC target was 2e4, apex triggering was disabled, and charge exclusion was enabled for unassigned charge states, as well as charge states greater than 6. The peptide match setting was disabled, exclude isotopes was enabled, and dynamic exclusion was set to 30 seconds. Voltage was set to 0 for the first 25 minutes, sweep gas was applied from minute 24.6 to 25 to dislodge any accumulated droplets from the capillary tip. From minute 25 to 80, voltage was set to 1.7 kV, capillary temp to 250° C., and the S-lens RF level to 80. From minute 94.20 to 94.60, sweep gas was applied to dislodge any accumulated droplets from the capillary tip.


The raw file generated by this analysis was searched using the same maxquant settings as indicated in the Scout experiment instrument method and raw data analysis section.


Prioritized Inclusion List Generation

The PSMs generated from the scout runs using intensity dependent-tiers (wAL00191 and wAL00192) were partitioned into three categories: PSMs at PEP≤ 0.02 (set a), PSMs with 0.02<PEP≤ 0.05 (set B), and PSMs with PEPs >0.05 (set γ). Then the same set of PEP filters defined above for wAL00191 and wAL00192 were applied to the results of a DDA analysis conducted on an injection of a 1× concentrated aliquot of carrier and reference material to generate sets δ, ∈, and ξ. Furthermore, these last three precursor sets were assembled such that they each contained a unique set of precursors with respect to one another and the preceding set of precursors.


Sets α and δ were combined and filtered such that a maximum of 4 peptides per protein were selected, choosing those precursors with the highest precursor intensities, to form the top priority tier candidates. The excluded precursors from this filtration were then combined with sets β and ∈ to make up the middle priority tier candidates. Peptides from sets γ and ξ were then combined to form the bottom priority tier candidates.


The results from the retention-time-calibration experiment were then intersected with the priority tier sets, and the PSMs matching each set were given a corresponding priority index for use by MaxQuant. Live. Up to 8,600 of the most abundant remaining retention-time-calibrationexperiment-associated PSMs were then added to the bottom priority tier to provide additional identifiable precursors when higher priority precursors were not detected. All selected precursors were then enabled for participation in the MaxQuant.live real-time-retention-time-alignment algorithm, and for MS2 upon detection. All remaining PSMs that were not part of the priority tiers were then selected for participation in the MaxQuant.live real-time-retention-time-alignment algorithm, but not for MS2 upon detection.


Prioritized Acquisition Parameters, Scout Runs and Single-Cell Samples

All single-cell samples were resuspended in 1 μl of 0.1% formic acid (85178, Thermo Fisher, Waltham, MA) and injected from a 384-well plate (AB1384, Thermo Fisher, Waltham, MA). All 1× concentrated carrier and reference samples were resuspended in 1 μl of 0.1% formic acid (85178, Thermo Fisher, Waltham, MA) and injected from a glass HPLC insert (C4010-630, Thermo Fisher, Waltham, MA). LC settings indicated above were used in these analyses. Scan parameters were implemented following the MQ.live listening scan guidelines: Two Full MS-SIM scans were applied from minute 25 to 30 to trigger MaxQuant.live. Both MS-SIM scans had the following parameters in common: 70k resolution, le6 AGC target, and a 300 ms maximum injection time. The first MS-SIM scan covered 908 to 1070Th, since the acquisition started at minute 25 and ended at minute 95. The second MS-SIM scan covered the scan space from 909Th to the numeric MaxQuant.live method index to call. The total Xcalibur MS method time was 95 minutes. Tune files governing voltage and sweep gas were implemented as in the pre-prioritization shotgun method.


Limited FASTA File Generation for Raw Data Analysis Corresponding to Prioritized Samples

The swissprot_human_20211005. fasta was read into the R environment using the seqinr (Charif & Lobry, Biological and Medical Physics, Biomedical Engineering 207-32 (2007)) package, and only those proteins with peptides present on the inclusion list were retained to generate the AndrewsnPOP_FASTA_v2.fasta file, containing 3535 proteins, used to search the resulting prioritized single-cell experiments.


DDA MS Acquisition

After a precursor scan from 450 to 1600 m/z at 70,000 resolving power, the top 7 most intense precursor ions with charges 2 to 4 and above the AGC min threshold of 20,000 were isolated for MS2 analysis via a 0.7 Th isolation window with a 0.3 Th offset. These ions were accumulated for at most 300 ms before being fragmented via HCD at a normalized collision energy of 33 eV (normalized to m/z 500, z=1). The fragments were analyzed by an MS2 scan with 70,000 resolution. Dynamic exclusion was used with a duration of 30 seconds with a mass tolerance of 10 ppm.


DIA MS Acquisition of Bulk CDC Populations

Samples were run using the VI method from Derks et al. (Derks et al., bioRxiv 467007 (2021)). This method contains 140k resolution MS1 scans for improved MS1 level quantification.


Analysis of DDA MS Data

Raw data were searched by MaxQuant (Cox et al., Nat Biotechnol. 26(12): 1367-72 (2008) and Cox et al., J Proteome Res. 10(4):1794-805 (2011)). 1.6.17.0 against a protein sequence database including entries from the appropriate human SwissProt database (dow nloaded Jul. 30, 2018) and known contaminants such as human keratins and common lab contaminants. Fasta was limited to proteins which were included on prioritization list. MaxQuant searches were performed using the standard work flow (Tyanova et al., Nat Protoc. 11(12):2301-19 (2016)). Trypsin specificity was specified and up to two missed cleavages for peptides having from 5 to 26 amino acids were allowed. Methionine oxidation (+15.99492 Da) and protein N-terminal acetylation (+42.01056 Da) were set as variable modifications. Carbamidomethylation was disabled as a fixed modification. All peptide-spectrummatches (PSMs) and peptides found by MaxQuant were exported in the msms.txt and the evidence. txt files.


Analysis of Data Independent Acquisition MS Data

Data Independent Acquisition runs were searched with DIA-NN v1.8.0 (Demichev et al., Nat Methods. 17(1):41-44 (2020)) using an in silico fasta generated library enabled by deep learning.


SILAC Data Analysis

When comparing relative protein levels in Jurkat and U-937 cells, SILAC ratios for peptides were computed by taking dividing each channel by its median, and then taking the ratio of the light and heavy channels. When comparing absolute abundances between heavy and light U-937 cells to measure efficiency of extraction, label swap experiments were run so that both lysis conditions were measured with both heavy and light labels. The raw intensities for corresponding lysis methods were averaged and the ratio between different lysis methods was plotted.


Single-Cell Filtering and Normalization

The single-cell data were processed and normalized by the SCOPE2 pipeline (Specht et al., Genome Biol. 22(1):50 (2021) and Petelski et al., Nat Protoc. 16(12):5398-425 (2021)). This pipeline is also implemented by the scp Bioconductor package (Vanderaa & Gatto, Expert Rev Proteomics 18(10):835-43 (2021) and Vanderaa & Gatto, Bioconductor (2020)). Briefly, single cells with suboptimal quantification were removed prior to data normalization and analysis based on objective criteria: The internal consistency of protein quantification for each single cell was evaluated by calculating the coefficient of variation (CV) for proteins (leading razor proteins) identified with over 5 peptides for that cell. The coefficient of variation is defined as the standard deviation divided by the mean. The CVs were computed for the relative reporter ion intensities, i.e., the RI reporter ion intensities of each peptide were divided by their mean resulting in a vector of fold changes relative to the mean. Cells that fell outside the distribution were removed from analysis with a threshold of 0.41. Data was normalized as by procedure outlined by Specht et al. (Specht et al., Genome Biol. 22(1):50 (2021) and Specht et al., Genome Biol. 22(1):50 (2021)).


Principal Component Analysis for Single Cell Data Sets

From the protein x single cell matrix, all pairwise protein correlations (Pearson) were computed. Thus, for each protein, there was a computed vector of correlations with a length the same as the number of rows in the matrix (number of proteins). The dot product of this vector with itself was used to weight each protein prior to principal component analysis. The principal component analysis was performed on the correlation matrix of the weighted data.


Melanoma Sub Population Protein Set Enrichment Analysis

Protein set enrichment analysis was performed by t-test between Cluster A and B on the un-imputed data. It was required that a given gene set had at least 4 proteins measured in the single cells and that each population had at least 80% of cells with protein observations. The distribution of p-values was corrected for multiple hypothesis testing with the BH method. O nly GO terms were reported with Q value less than 0.0001 were reported.


Constructing CDC Phase Markers

Phase markers were constructed from proteins identify with differential abundant each CDC phase in both monocyte and melanoma cells. These proteins were first identified on the bulk level. To further narrow the list of proteins used to create phase markers, proteins that contained multiple, positively correlated peptides in the single cell samples were used. Phase markers were then constructed by averaging the abundances of all possible combinations of 2 or 3 proteins corresponding to each phase of the cell cycle. Groups of two markers for each CDC phase that were positively correlated were selected. This served as validation as it was expected that proteins that are highly abundant in same phase would positively covary. Groups of protein markers were then further filtered.


Markers were first constructed in the space of monocyte cells, and correlations between markers were validated in melanoma cells FIGS. 4B and 4G. Having validated the protein markers, protein markers within phase were averaged for downstream analysis.


Identifying Proteins that Covary with CDC Markers


To identify proteins that covary with the phase marker vectors, the phase marker vectors to the measured protein levels of each protein were correlated using Spearman correlation. The distribution of p-values obtained from the Spearman correlation test was adjusted using the BH method and the results were filtered at 1% FDR.


Cell Cycle Protein Set Enrichment Analysis

To identify functionally coherent sets of proteins that covary with the CDC phase markers, each protein was correlated to the median abundance of CDC proteins that showed similarity between melanoma and monocyte cells as plotted in FIGS. 4A and 4F. The resulting correlation vectors were analyzed by protein set enrichment analysis similar to previously reported analysis (Franks et al., PLOS Comput Biol. 13(5):e1005535 (2017)). In the case of cell-type specific co-variation, empirical bootstrapping was also used to estimate the Z-score corresponding to each correlation, and the distributions of Z-scores were then compared via ANOVA for estimating the statistical significance. O nly GO Terms having least 4 proteins were analyzed. ANOVA was used to estimate if the variance among the correlations of the proteins from the GO term, and the CDC phase markers can be explained by the CDC. The Benjamini-Hochberg method was then used to estimate the corresponding q values (FDR; false discovery rare) for each GO term. Among the set of GO terms within 5% FDR, the 20 GO terms whose correlations to the CDC phase markers was most similar or most different between the 2 cell lines were displayed in FIGS. 4A-4J.


Assigning Cells to CDC Phase

A greedy approach was taken to assign cells to a CDC phase. First, a vector comprised of length 3X the number of cells was created, where each value was the average abundance of G1, S, or G2 marker proteins. This vector was then sorted from highest to lowest. Subsequently iterated down the list and sorted cells into the G1, S, or G2 bin based off the phase of each value. 50% of cells were sorted into the G1 bin, 25% of cells were sorted into the S and G2 bins based off the distribution observed from the bulk FACS CDC sorting.


Protein Complex Analysis

A null distribution that consists of all pairwise Euclidean distances was computed for each protein. Euclidean distances were o nly calculated between observed values, and vectors were subsequently normalized to the number of pairwise observed values in each vector. Euclidean distances were then calculated in the same fashion from all proteins within complexes from the CORUM protein database (Giurgiu et al., Nucleic Acids Res. 47(D1):D559-D563 (2019)).









TABLE 1







MaxQuant.live settings for prioritized analysis










Scout exp.
nPOP samples













Global settings: Survey scan




ScanDataAsProfile
True
True


PositiveMode
True
True











MaxIT
100
ms
100
ms









Resolution
70,000    
70,000    


AgcTarget
1,000,000    
1,000,000    


MzRange
 (450, 1258)
 (450, 1258)


BoxCarScans
0
0


Global settings: TopN


NumOfMS2Scans
0
0


RealtimeCorrection


MzTolerances
(4.5, 5)  
(4.5, 5)  


RetentionTimeTolerances
(0.01, 2)  
(0.01, 2)  


SigmaScaleFactorRt
3
3


PeptideHistoryLength
2
2


MinUsedCorrectionPeptides
15 
15 


IntensityPeakRatioThreshold
   .01
   .01


PeptideDetectionIsoPeaks
2
2


IsotopeTolerance
9
9


Ms2DetectionNeeded
False
False


Ms2ExcludeDetectedPeptides
False
False


Ms2MinNormIntensity
  0.1
  0.1


Ms2MzTolerance
20 
20 


TargetedMs2


BatMode
False
False


AutoPriority
True
True


DefaultPriority
0
0


MaxNumOfScans
1
1


WindowAndOffsetInDalton
False
False


ScanDataAsProfile
False
False


WindowSize
  0.5
  0.5


MzOffset
0
0


LowerMzBound
100 
100 


CollisionEnergy
33 
33 











LifeTime
2,100
ms
2,100
ms









Resolution
70,000    
70,000    











MaxIT
300
ms
300
ms









AgcTarget
1,000,000    
1,000,000    


PositiveMode
True
True









Example 2. Sample Preparation

To reduce batch effects and background signal, the goal was to maximize the number of single cells prepared in parallel while minimizing the volumes of sample preparation. To this end, the idea of performing all sample preparation steps in droplets on the surface of a uniform glass slide was explored (FIGS. 1A-1D). This allows the freedom to arrange single cells in any geometry that best fits the experimental design (FIGS. 1A-1C). To facilitate this idea, clean reagents, compatible both with analysis by LC-MS and an open surface design, were needed. To this end, the use of 100% dimethyl sulfoxide (DMSO) was introduced as a reagent for cell lysis and protein extraction. Its low vapor pressure enables nanoliter droplets to persist on the surface of the open glass slide. Furthermore, its compatibility with MS analysis allows to obviate sample cleanup and associated losses and workflow complications. Control experiments indicate that DMSO efficiently delivers proteins to MS analysis without detectable bias for cellular compartments (FIG. 7B) and supports accurate relative protein quantification (FIG. 7D).


These data supported the use of DMSO for cell lysis performed by first dispensing an experimenter defined regular array of DMSO droplets, and subsequently adding a cell to each droplet for lysis (FIG. 1A). After lysis, proteins are digested for 4 hours by adding the protease trypsin dissolved in aqueous buffer. To control evaporation throughout the digestion step, slide temperature and internal humidity were controlled. Furthermore, a perimeter of water droplets was dispensed around the samples (FIG. 1D). See Example 1 for details.


For the next step, labeling peptides, it was found that the commonly used approach of dissolving labels in acetonitrile was unreliable due to low density and low surface tension of acetonitrile. To overcome this problem, DMSO dissolved labels were introduced, and robust performance of sub-nanoliter droplets over hundreds of samples were observed. This approach was validated by measuring labeling efficiency in pooled samples, and over 99% of all possible peptides were found to be TMT labeled. The final step of nPOP entailed collecting the samples and delivering them for LC-MS analysis. Clusters of labeled single cells were pooled into a single set, aspirated, and dispensed into a 384-well plate in a fully automated fashion for streamline sample injection (FIGS. 1A-1B).


The nPOP sample preparation was combined with prioritized quantification of proteins introduced by Huffman et al. (Huffman et al., bioRxiv 484655 (2022) and followed the guidelines of the SCOPE2 protocol (Specht et al., Genome Biol. 22(1):50 (2021) and Petelski et al., Nat Protoc. 16(12):5398-25 (2021)). The AL-01 sample layout design, which prepares 2,016 single cells in one day, was employed (FIG. 1D). Using this design, 1,556 single cells were successfully analyzed (FIG. 2A) as part of a single batch. This is lower than the 2,016 capacity due to: 1) including 128 negative controls, 2) having 175 single cells excluded from analysis (FIG. 2A) and 3) 15 sets lost because of LC malfunctions. To increase the depth and consistency of proteome coverage, the single-cell samples were analyzed by prioritized Single Cell ProtEomics (pSCOPE) introduced by Huffman et al. (Huffman et al., bioRxiv 484655 (2022), following the guidelines of the SCOPE2 protocol (Specht et al., Genome Biol. 22(1):50 (2021) and Petelski et al., Nat Protoc. 16(12):5398-25 (2021)).


Example 3. Single-Cell Data Quality Controls

To evaluate nPOP's ability to analyze protein covariation within and across cell types, two cell lines, WM989 melanoma and U-937 monocyte cells, were analyzed. The average number of proteins and peptides per single cell were 997 proteins and 2,630 peptides, with 2,844 proteins quantified across the 1,543 single cells prepared by nPOP (FIG. 2A). To quantify the extent of background noise in these measurements, the intensity of signal in negative controls was evaluated. The negative controls correspond to droplets that did not receive single cells, and their intensities reflect crosslabeling and nonspecific background noise (Specht et al., Genome Biol. 22(1):50 (2021) and Petelski et al., Nat Protoc. 16(12):5398-25 (2021)). The intensities in the negative controls, shown in FIG. 2B, were mostly absent or very low, indicating that background noise is low for samples prepared with nPOP. The intensities for single cells also show that peptides from melanoma cells were more abundant than peptides from monocyte cells, reflecting the different cell sizes (FIG. 2B). To further test the extent to which higher reporter ion signal in the melanoma cells reflects larger cell size, the measured diameter for single cells was plotted against the average reporter ion signal (FIG. 2C). Good agreement between diameter and average intensity, p=0.81, both between cell types and within cell types supports the differences in distributions for all melanoma and monocyte cells.


As an additional QC metric, the agreement in relative quantification derived from different peptides originating from the same protein was evaluated. The agreement was significantly higher in the single cells than the negative controls (FIG. 2D). Furthermore, the small spread of the distribution for the quantitative variability suggests high consistency of the automated sample preparation technique.


In addition to the increased throughput, nPOP reduced sample preparation batch effects that could introduce technical artifacts. Indeed, because all single cells were prepared on the same day, no sample preparation batch corrections needed to be applied to the data.


Next, principal component analysis (PCA) of the single-cell protein dataset was performed using all quantified proteins (FIG. 2E). The PCA indicates three distinct clusters of cells. The clusters correspond the cell types, with two sub populations of melanoma cells. The cell types separate along the first principal component (PC1), which accounts for 59% of the variance. To evaluate whether this separation reflects technical artifacts, such as differences in cell size or missing data, or biological differences between cell types, the proteomes of 200-cell samples of melanoma and monocyte cells analyzed by established bulk methods were projected (FIG. 2D).


The first step towards identifying within cell type protein covariation was to identify proteins that correlate significantly within monocyte and melanoma cells. Computing all pairwise correlations, 5,089 significant correlations were found in monocyte, and 4,679 correlations were found in melanoma cells at FDR <5%. 2,353 of these correlations were between the same pair of proteins. While most of these correlations shared the same trend, interestingly, 15 proteins showed opposite correlation trends. The joint distributions for proteins from these two cases were plotted in FIG. 3A and FIG. 3B, respectively.


A primary factor for observed protein covariation within a cell type may reflect proteins belonging to a complex. The goal was to identify whether observed protein covariation could be explained by proteins belonging to complexes. To this end, all pairwise Euclidean distances between proteins in know complexes from the CORUM database were computed (Giurgiu et al., Nucleic Acids Res. 47(D1):D559-D563 (2019)), and the distribution against all pairwise distances was tested. 96 protein complexes were identified in melanoma cells, and 89 were identified in monocytes at FDR <10%. Both cell types had similar agreement between Ribosomal proteins (FIG. 3C). A full list of differential protein complexes can be found in Table 4.


Example 4. Cell Cycle Analysis

A more challenging problem was quantifying CDC-related protein covariation within a cell type. As a first step towards this analysis, the potential to classify individual cells by their cell cycle phase was evaluated. To obtain a list of proteins whose abundance varies periodically with the cell division cycle, populations of each cell type were first sorted based off their DNA content (FIGS. 4A and 4F). The proteomes of the sorted cells were quantified, and proteins whose abundance differs in G1, S, and G2/M phase for both cell types were identified.


To construct robust markers for each phase, the abundances of groups of proteins corresponding to each phase of the cell cycle were averaged. For each CDC phase, two markers from non-overlapping sets of proteins were constructed. Positive correlation between markers from the same phase served as internal validation based on the expectation that proteins peaking in the same phase positively covary. Conversely, markers for different phases were expected to negatively correlate to each other (FIGS. 4B and 4G). Markers were first constructed in the space of monocyte cells, and correlations between markers were cross-validated in melanoma cells (FIGS. 4A and 4F). Having validated the protein markers, protein markers within phase were averaged for downstream analysis.


The proteomes of both melanoma and monocyte cells were then projected into a joint 2-dimensional space of the CDC marker proteins defined by principal component analysis (FIGS. 4B and 4G). Each cell was then color-coded based on the mean abundance of a given protein marker in the PCA plots for their respective phase (FIGS. 4C and 4H). The cells from both cell types cluster by CDC phase, which further suggests that the data capture CDC related protein dynamics.


To identify proteins that covary with the CDC periodic markers, the phase marker vectors were correlated to the measured protein abundances of all proteins quantified across many single cells. For 121 of these proteins in the melanoma and 113 in the monocyte, the correlations were statistically significant, FDR <0.01, suggesting that these proteins are CDC periodic. Specifically, NPM1 which facilitates ribosome biogenesis positively correlated with G1 phase in both melanoma and monocyte populations, p<10-15, p<10-8, respectively.


To increase the statistical power and identify functional covariation with the CDC, the next focus was the covariation of phase markers and proteins with similar functions as defined by the gene ontology (GO). The distributions of correlations between the 3 phase marker vectors and all quantified proteins from a GO term were compared (see the boxplots in FIGS. 4D and 41). For protein polyubiquitination, the distributions of correlations differed significantly between the CDC phases, and this phase-specific covariation was similar for the two cell types. Many other GO terms showed covariation to the phase markers that was similar for the two cell types. Instead of displaying the boxplot distributions for all of them, the distributions of correlations were summarized with their medians and displayed as a heatmap. Such functions with shared covariation included proteolysis in G2/M phase which implicate the role of protein degradation in cell cycle progression. Additionally, terms related to DNA repair and translation were correlated with G1 markers, confirming the role of cell growth and DNA repair post mitosis.


In addition to finding groups of proteins that showed similar cell cycle covariation between cell types, several GO terms also varied differential with CDC markers (FIGS. 4E and 4J). Such GO terms included terms related to cell signaling, metabolism and immune system related processes which may reflect the role of the monocyte as an immune cell. However, a larger majority of the 117 significant GO terms (Table 3) showed concerted trends between the two cell types highlighting the conservation CDC related processes.


Example 5. Melanoma Sub Population

Next, the two distinct clusters of melanoma cells observed in FIG. 2D were analyzed. Recent studies of these melanoma cells identified two populations with distinct transcriptomes (Emert et al., Nat Biotechnol. 39(7):865-76 (2021) and Fallahi-Sichani et al., Mol Syst Biol. 13(1):905 (2017)). The larger population is susceptible to treatment by the cancer drug vemurafenib, while the smaller one is primed to develop drug resistance (Emert et al., Nat Biotechnol. 39(7):865-76 (2021)).


To test if the clusters mapped to the same distinct cell states previously identified, the cells were color coded by the abundance of proteins whose transcripts were reported (Emert et al., Nat Biotechnol. 39(7):865-76 (2021)) to mark either the non-primed population (Cluster A) or the primed sub-population (Cluster B) (FIG. 5A). Primed markers were significantly more abundant in cluster B, p=2e-4, while non-primed had greater abundance in cluster A, p<le-15. Having established correspondence between the populations, additional protein differences between the two clusters were identified by performing PSEA. It resulted in 200 sets of functionally related proteins exhibiting differential abundance at FDR <1% (Table 5). Some of these sets were displayed by color coding the single cells from the PCA plot with the mean protein abundances for the set (FIG. 5A). Protein sets related to G2/M transition of mitosis, cyclin dependent kinase activity and protein degradation were more abundant in cluster A. In contrast, protein sets with increased abundance in cluster B related to senescence and cell cycle arrest. These results suggest that Cluster A cells are more proliferative than cluster B cells, consistent with prior report (Fallahi-Sichani et al., Mol Syst Biol. 13(1):905 (2017)).


To explore CDC differences further, the distribution of cells in each CDC phase across the two sub-populations were quantified. A substantially larger fraction of cells were found in cluster B in G1 phase, 78%, while only 4% of cells were found to be assigned to G2 phase (FIG. 5B). This result further bolsters the conclusion that cluster B cells divide slower than cluster A cells. The next goal was to identify additional groups of proteins that co-vary with CDC phase between the two populations. Upon repeating the analysis from FIGS. 4C and 4D on both melanoma populations, several sets of proteins were found to correlate significantly to the CDC markers. Many of these sets correlated deferentially to the markers within each cluster. Specifically, many terms for glucogenesis and signaling cascades displayed different correlation profiles to the CDC markers.


Lastly, 234 additional proteins were differential between cluster A and cluster B cells at FDR <1%. Some of these proteins were displayed in FIG. 5D, as distributions of abundances for individual proteins, and in FIG. 5E, as joint distributions for abundances of two proteins. Notably, increased abundance of the surface protein Transmembrane emp24 domain-containing protein 10, and decreased abundance of the transcription factor Hepatocyte nuclear factor 3-beta in cluster B were found (FIG. 5E). The remaining list of differential proteins can be found in Table 6.


Example 6. Surface protein analysis

NPOP was also applied to specifically study surface proteins in an additional experimental system, pancreatic ducal adenocarcinoma (PDAC) (FIGS. 6A and 6B). Identifying co-abundant surface proteins has valuable potential for therapeutics that utilize bi-specific antibodies or receptors (Dahlén et al., Ther Adv Vaccines Immunother. 6(1):3-17 (2018)). Thus, single cell proteomics may emerge as a useful tool for suggesting such pairs of proteins. To this end, the analysis of 34 different surface proteins including well known markers of PDAC, such as CEACAM5 and CEACAM6, was prioritized (Gebauer et al., PLOS One. 9(11):e113023 (2014)). Hierarchical clustering in the space of all pairwise protein protein correlations revealed co-abundant clusters of surface proteins (FIGS. 6C and 6E). Correlations were found between proteins such as CD44 and CEACAM6. Additionally, correlations between surface proteins and other intracellular proteins were computed, and 120 significant correlations at FDR <1% were found (FIG. 6D).


Existing single-cell omics methods excel at classifying cells by cell type. However, the regulatory dynamics resulting in cell to cell variability within a cell type are more challenging to analyze. To support such analysis, a highly parallel sample preparation that enables preparation of hundreds to thousands of single cells in a given experiment was introducee. It allows for reduced volumes and increased consistency of single-cell proteomic sample preparation. Furthermore, it can enable processing thousands of single cells in parallel and thus empower high-throughput, high-power biological analysis (Slavov, Nat Biotechnol. 39(7):809-10 (2021)).


To maximize access and flexibility, nPOP used only commercially available equipment and prepared single cells on an open surface that could be pragmatically reconfigured and adopted to different experimental designs. The open environment also obviated all sample movements and maximized the consistency and precision of the sample preparation. The open layout using a hydrophobic slide can be scaled up to simultaneously prepare thousands of single cells. Furthermore, nPOP is amenable to different coatings or hydrophobic surfaces which have the potential to further improve recovery.


NPOP allowed for deeper single cell proteomic analysis of the cell division cycle than the CDC analysis using the minimal sample preparation method (mPOP) (Specht et al., bioRxiv. 399774 (2018)). The data allowed identification of new proteins and functional groups of proteins associated with the cell cycle without the artifacts associated with synchronizing cell cultures (Cooper, FEBS J. 286(23):4650-56 (2019)). Furthermore, functional groups of proteins associated with the cell cycle were determined in an identified subpopulation of cells within the melanomas. These initial results demonstrate the feasibility of inferring co-regulation of biological processes from single-cell proteomics measurements.


Example 7. nPOP Workflow

A non-limiting example of an overall work flow for nPOP sample preparation includes cell isolation, cell lysis, protein digestion, peptide labeling, and pooling as illustrated in FIG. 8A. Sample preparation starts with dispensing droplets of 4 nl DMSO for cell lysis. The droplets are organized as regular grids (e.g., a cluster, see, e.g., FIG. 8B) to facilitate their automating deposition, regular additions during sample preparation and pooling at the end of the experiment. The second step of nPOP is the isolation and dispensing of single cells into the DMSO droplets. Each single cell is isolated in a 0.3 nl droplet and added to a DMSO droplet for lysis (FIG. 8A). After 20 minutes for cell lysis, a perimeter of 12 nl droplets of water (for maintaining high local humidity) is deposited around the perimeter of the four samples. The next nPOP step is the addition of trypsin with HEPES buffer for digesting the proteins into peptides. This step brings the total volume to 13.5 nl. Samples are digested at a 75 ng/μl of trypsin for 5 hours on slide. To further control evaporation, nPOP uses a humidifier to keep relative humidity inside the CellenONER at 75%. The temperature of the slide is set to dynamically adjust to one degree above the dew point inside the CellenONER and stays around 17ºC for digestion. After digestion, humidity is reduced, and the slide is brought to room temperature for labeling. The single cell droplets dry down on the slide to volumes of approximately 4 nl before labeling. TMT labels dissolved in DMSO are dispensed in volumes of 20 nl to the single cell droplets. Dissolving labels in DMSO is a distinctive and required aspect of nPOP that allows for easy handling of tiny droplets with TMT solution. The most commonly used solvent for TMT, acetonitrile, is difficult to handle with CellenONER. After samples are labeled for one hour at room temperature, labeling is quenched with the addition of 20 nl 5% hydroxylamine for 20 minutes. A second addition of hydroxylamine is then added and sample quenches for an additional 20 minutes.


To pool all single-cell samples into a set, 1 μl of water is pipetted by hand onto each array of labeled samples. Samples are then pipetted directly into glass inserts containing carrier and reference previously prepared using the mPOP protocol (Specht & Slavov, J Proteome Res. 17(8):2565-71 (2018)) for injection vials. To improve the recovery of labeled peptides, the footprint of each array can be washed by 4 μl of acetonitrile, which is collected and added to the corresponding combined set. This wash is optional and is used to maximize the recovery of labeled peptides from the slide.


Example 8. Single-Cell Protein Analysis with nPOP

nPOP is a general sample preparation method that can be used for either label-free MS analysis or multiplexed MS analysis as part of existing workflows reviewed by Slavov, Curr Opin Chem Biol. 60:1-9 (2021) and Kelly, Mol Cell Proteomics 19(11): 1739-48 (2020). Here, sample preparation by nPOP as part of the SCOPE2 protocol (Specht et al., Genome Biol. 22(1):50 (2021) and Petelski et al., Nat Protoc. 16(12):5398-25 (2021)) is demonstrated. Specifically, Minimal ProteOmic sample Preparation (mPOP) module (Specht & Slavov, J Proteome Res. 17(8):2565-71 (2018)) was replaced with nPOP and used all other modules of the SCOPE2 workflow, including an isobaric carrier (Specht & Slavov, J Proteome Res. 20(1):880-87 (2021)), Data-Driven Optimization of Mass Spectrometry (DO-MS) (Huffman et al., bioRxiv. 512152 (2019)), Data-driven Alignment of Retention Times for IDentification (DART-ID) (Chen et al., PLOS Comput Biol. 15(7):e1007082 (2019)), and the SCOPE2 data analysis pipeline (Specht et al., Genome Biol. 22(1):50 (2021) and Vanderaa et al., Bioconductor (2020)).


To evaluate the performance of nPOP for single-cell sample preparation, proteins in 176 single cells of two distinct cell types, Hela cells and U-937 monocytes, were measured. The sample preparation was done on two different days so that the data may reflect day-specific batch effects. The resulting SCOPE2 sets were run using less than 24 hours of instrument time. Samples were analyzed and data processed via the SCOPE pipeline (Specht et al., Genome Biol. 22(1):50 (2021)). To evaluate the single-cell data, the pipeline calculated the coefficient of variation (CV) of relative peptide levels belonging to the same protein. The relatively low CV values indicate that protein quantification from different peptides was internally consistent (FIG. 8C). Furthermore, the small spread of the distribution for the median CVs indicates that each cell is treated consistently by the automated sample preparation technique.


Next, principal component analysis (PCA) of the single-cell protein dataset was performed using all quantified proteins (FIG. 8D). The PCA indicates two distinct clusters of cells. The clusters corresponded with the cell type and separated along the first principal component (PC1), which accounted for 73% of the variance (FIG. 8D).


To validate further that the cell type separation was driven by accurate quantification of proteins (rather than by secondary factors such as cell size or missing data), bulk samples of Hela cells and monocytes were included in the PCA. Similar to previous analysis (Specht et al., Genome Biol. 22(1):50 (2021), Petelski et al., Nat Protoc. 16(12):5398-25 (2021) and Budnik et al., Genome Biol. 19(1):161 (2018)), the bulk samples clustered with the corresponding single cells. This clustering indicated that the single cell protein quantification was consistent with the proteomic measurements of established bulk methods.


Example 9. Cell Cycle Analysis

To test further the quantitative accuracy of the data, the heterogeneity within a cell type was studied. Differences in cell state were measured by analyzing the variation in known cell division cycle proteins. To do this, the data for CDC proteins were filtered and cells along the first two principal components were plotted. Each cell was then color coded based on the mean abundance of markers for M/G1 and G2/S phases in the cell. The color-coded cells clustered along the first and second principal component, indicating the feasibility of inferring cell cycle phase from the cells analyzed with nPOP.


A method that prepares single cells in 4-15 nanoliter volumes using only commercially available equipment is demonstrated. The current method prepares single cells in an open environment without a need to move samples in the process to maximize the consistency and precision of the sample preparation. The open layout using a glass slide is scalable to preparing hundreds of single cells at a time. Furthermore, the current method is amenable to different coatings or hydrophobic surfaces which have the potential to further improve recovery.









TABLE 2







Protein set enrichment analysis based on protein levels in cells isolated based on DNA content from FIGs. 4A-4J


















numberOf
fractionOfDB







GO_term
pVal
Matches
Observed
G1
S
G2
FDR



















32
regulation of cellular amino acid metabolic process
2.45E−29
43
0.843137
−0.16421
0.062426
0.083261
7.88E−26


34
cellular nitrogen compound metabolic process
4.58E−26
114
0.616216
−0.11926
0.064725
0.035002
7.37E−23


162
respiratory electron transport chain
7.39E−25
80
0.761905
0.005534
0.07522
−0.10121
7.93E−22


141
proteasome accessory complex
1.16E−24
17
0.62963
0.16543
0.036435
0.109147
9.35E−22


37
regulation of ubiquitin-protein ligase activity involved in mitotic cell cycle
3.75E−21
62
0.826667
−0.15151
0.044838
0.109049
2.41E−18


144
proteasome complex
9.28E−20
51
0.836066
−0.15017
0.032138
0.083261
4.98E−17


36
positive regulation of ubiquitin-protein ligase activity involved in mitotic cell
2.80E−19
58
0.816901
−0.15387
0.049323
0.107033
1.29E−16



cycle


223
negative regulation of ubiquitin-protein ligase activity involved in mitotic cell
4.44E−19
55
0.846154
−0.14787
0.047445
0.107033
1.78E−16



cycle


33
DNA damage response, signal transduction by p53 class mediator resulting in
5.96E−17
51
0.728571
−0.15151
0.06121
0.083893
2.13E−14



cell cycle arrest


192
ATP-dependent chromatin remodeling
2.79E−16
21
0.84
0.093528
−0.192
0.027195
8.98E−14


204
mitochondrial ribosome
6.82E−15
23
0.69697
−0.0765
0.163773
−0.0818
2.00E−12


14
protein polyubiquitination
3.50E−14
73
0.51773
−0.12806
0.042758
0.065777
9.39E−12


128
tricarboxylic acid cycle
1.35E−13
26
0.52
−0.04468
0.118258
−0.09065
3.35E−11


256
mitochondrial small ribosomal subunit
3.04E−13
18
1
−0.08978
0.187256
−0.07391
6.99E−11


165
G1/S transition of mitotic cell cycle
1.17E−12
102
0.563536
−0.15017
0.05532
0.084525
2.51E−10


71
melanosome
1.65E−12
84
0.823529
0.021763
0.045153
−0.08822
3.33E−10


246
DNA strand elongation involved in DNA replication
2.25E−12
27
0.870968
−0.1821
0.008833
0.10294
4.25E−10


49
nucleosomal DNA binding
9.79E−12
20
0.526316
0.093528
−0.17623
0.01104
1.68E−09


131
SWI/SNF complex
9.90E−12
12
0.545455
0.191848
−0.19741
0.023347
1.68E−09


30
viral infectious cycle
1.69E−11
100
0.847458
−0.06343
0.064388
−0.03382
2.72E−09


65
spindle
1.84E−11
76
0.520548
−0.09735
−0.0331
0.068958
2.82E−09


222
nucleobase-containing small molecule metabolic process
4.61E−11
56
0.717949
−0.12346
0.037102
0.053552
6.74E−09


84
mitochondrial respiratory chain complex I
9.69E−11
41
0.719298
0.009603
0.07337
−0.1029
1.36E−08


150
chromatin remodeling
1.02E−10
45
0.463918
0.131442
−0.16481
0.032692
1.37E−08


132
mitochondrial electron transport, NADH to ubiquinone
1.27E−10
39
0.78
0.010877
0.076719
−0.10643
1.64E−08


252
viral transcription
1.61E−10
74
0.902439
−0.06417
0.066076
−0.04127
1.99E−08


130
translational termination
2.99E−10
76
0.76
−0.06417
0.065467
−0.04096
3.57E−08


74
RNA polymerase II distal enhancer sequence-specific DNA binding
7.92E−10
25
0.555556
0.087479
−0.16763
0.041553
9.10E−08


123
translational elongation
9.35E−10
84
0.651163
−0.06146
0.056745
−0.04154
1.04E−07


15
integral to endoplasmic reticulum membrane
9.80E−10
32
0.340426
0.145693
0.0592
−0.11897
1.05E−07


24
proteasome core complex
1.15E−09
18
0.418605
−0.16359
0.067345
0.070964
1.20E−07


110
npBAF complex
1.43E−09
10
0.4
0.11687
−0.19075
−0.00053
1.44E−07


2
RNA polymerase II core promoter proximal region sequence-specific DNA
1.65E−09
47
0.315436
0.097175
−0.15762
0.027474
1.61E−07



binding


8
ion transport
1.74E−09
27
0.156977
−0.04792
0.140563
−0.1736
1.65E−07


219
nuclear-transcribed mRNA catabolic process, nonsense-mediated decay
3.07E−09
97
0.815126
−0.06446
0.041942
−0.01568
2.82E−07


120
NADH dehydrogenase (ubiquinone) activity
3.81E−09
39
0.58209
0.018774
0.067717
−0.10535
3.41E−07


179
SRP-dependent cotranslational protein targeting to membrane
5.56E−09
98
0.830508
−0.05122
0.061788
−0.0524
4.83E−07


97
nuclear inner membrane
6.17E−09
20
0.571429
0.118928
−0.16447
−0.00109
5.22E−07


100
regulation of translational initiation
8.04E−09
33
0.611111
−0.12036
0.036123
0.042677
6.63E−07


111
nBAF complex
9.58E−09
8
0.275862
0.168085
−0.21357
0.01725
7.70E−07


211
de novo' posttranslational protein folding
1.02E−08
30
0.769231
−0.15019
0.088715
0.007556
8.04E−07


195
chromatin organization
1.28E−08
68
0.561983
0.102038
−0.13131
0.032564
9.77E−07


243
catalytic step 2 spliceosome
1.36E−08
77
0.9625
0.011309
−0.03822
0.041113
1.01E−06


12
cellular lipid metabolic process
1.44E−08
87
0.524096
0.037441
0.029152
−0.08104
1.05E−06


94
pyruvate metabolic process
1.56E−08
19
0.542857
−0.04449
0.099324
−0.11296
1.12E−06


31
antigen processing and presentation of exogenous peptide antigen via MHC
2.06E−08
63
0.446809
−0.13681
0.063575
0.057411
1.44E−06



class I, TAP-dependent


5
mediator complex
5.46E−08
26
0.309524
0.143333
−0.16837
0.014175
3.74E−06


35
antigen processing and presentation of exogenous peptide antigen via MHC
5.64E−08
66
0.452055
−0.12806
0.063575
0.053209
3.78E−06



class I


88
electron carrier activity
6.05E−08
47
0.516484
−0.02651
0.077971
−0.07193
3.98E−06


63
translation initiation factor activity
6.93E−08
43
0.286667
−0.1238
0.02009
0.077118
4.46E−06


99
eukaryotic translation initiation factor 3 complex
9.76E−08
15
0.223881
−0.18804
0.079906
0.08019
6.16E−06


6
phagocytic vesicle membrane
1.31E−07
32
0.273504
0.04696
0.085917
−0.17535
8.13E−06


158
lipid particle
1.58E−07
29
0.402778
0.068452
−0.07888
−0.07974
9.58E−06


101
eukaryotic 43S preinitiation complex
2.06E−07
14
0.56
−0.19032
0.081462
0.08019
1.23E−05


182
snRNA processing
2.38E−07
11
0.37931
0.142306
−0.10625
−0.03667
1.37E−05


183
integrator complex
2.38E−07
11
0.366667
0.142306
−0.10625
−0.03667
1.37E−05


75
glucose metabolic process
2.80E−07
66
0.437086
−0.05355
0.054665
−0.02263
1.58E−05


53
purine base metabolic process
3.01E−07
24
0.666667
−0.11526
0.045347
0.02116
1.67E−05


98
formation of translation preinitiation complex
3.45E−07
13
0.52
−0.19032
0.081462
0.08019
1.85E−05


102
eukaryotic 48S preinitiation complex
3.45E−07
13
0.541667
−0.19032
0.081462
0.08019
1.85E−05


148
sperm protein complex
5.16E−07
8
0.363636
−0.13997
0.065658
0.060558
2.72E−05


127
protein disulfide isomerase activity
6.05E−07
16
0.516129
0.128265
−0.01208
−0.09251
3.13E−05


149
chaperonin-containing T-complex
6.13E−07
9
0.428571
−0.13997
0.065658
0.060558
3.13E−05


126
proteasome core complex, alpha-subunit complex
7.16E−07
7
0.269231
−0.18048
0.11554
0.058944
3.60E−05


118
ER-associated protein catabolic process
8.02E−07
24
0.571429
0.136369
−0.04677
−0.09942
3.97E−05


64
spindle pole
9.00E−07
43
0.307143
−0.05084
−0.03465
0.101614
4.39E−05


81
cytosolic large ribosomal subunit
9.38E−07
44
0.785714
−0.06298
0.066455
−0.04536
4.50E−05


216
ER membrane protein complex
1.00E−06
9
0.692308
0.163404
−0.06521
−0.14167
4.71E−05


161
chromosome segregation
1.01E−06
42
0.477273
−0.08845
−0.0467
0.094289
4.71E−05


106
DNA methylation
1.30E−06
12
0.387097
0.213756
−0.17843
−0.0265
5.98E−05


27
tRNA binding
1.33E−06
31
0.62
−0.09439
0.030347
0.052023
6.05E−05


232
positive regulation of innate immune response
1.40E−06
5
0.384615
−0.3905
0.144071
0.219182
6.25E−05


230
unsaturated fatty acid metabolic process
1.77E−06
9
0.818182
0.120954
0.034063
−0.18453
7.69E−05


231
alpha-linolenic acid metabolic process
1.77E−06
9
0.818182
0.120954
0.034063
−0.18453
7.69E−05


186
protein N-linked glycosylation via asparagine
1.83E−06
62
0.632653
0.093838
−0.04186
−0.07781
7.83E−05


11
triglyceride biosynthetic process
1.85E−06
21
0.396226
0.089435
0.024401
−0.16332
7.83E−05


226
regulation of proteasomal protein catabolic process
2.10E−06
7
0.583333
−0.16189
−0.01281
0.132206
8.76E−05


188
Ino80 complex
2.20E−06
13
0.448276
0.147167
−0.17969
0.014416
9.04E−05


124
tRNA aminoacylation for protein translation
2.24E−06
42
0.608696
−0.05202
0.050645
−0.01268
9.04E−05


136
mitochondrial membrane
2.25E−06
39
0.423913
−0.0135
0.061614
−0.085
9.04E−05


95
vitamin D receptor binding
2.34E−06
13
0.8125
0.147668
−0.15714
−0.01552
9.28E−05


155
dolichyl-diphosphooligosaccharide-protein glycotransferase activity
2.41E−06
7
0.5
0.0568
0.013482
−0.14572
9.44E−05


201
mitochondrial nucleoid
2.68E−06
37
0.902439
−0.00445
0.055354
−0.05293
0.000104


253
mitochondrial large ribosomal subunit
3.11E−06
14
0.933333
−0.04843
0.173239
−0.09096
0.000119


215
RNA polymerase II repressing transcription factor binding
3.28E−06
10
0.285714
0.090858
−0.12685
0.013096
0.000124


225
lysosomal lumen
3.39E−06
36
0.507042
0.077563
0.009343
−0.10295
0.000127


25
antigen processing and presentation of peptide antigen via MHC class I
4.14E−06
81
0.455056
−0.1132
0.0572
0.049277
0.000153


142
NuRD complex
4.57E−06
13
0.361111
0.0613
−0.16927
0.065695
0.000167


119
mitotic cell cycle spindle assembly checkpoint
5.02E−06
24
0.571429
−0.07541
−0.06166
0.166742
0.000182


206
proteasome binding
6.02E−06
8
0.727273
−0.1886
−0.04456
0.170854
0.000215


85
midbody
6.60E−06
69
0.534884
−0.04933
−0.00339
0.032295
0.000234


56
kinetochore
7.63E−06
52
0.530612
−0.03572
−0.04522
0.091451
0.000267


13
axonogenesis
7.80E−06
28
0.224
0.018824
0.087952
−0.16282
0.00027


22
antigen processing and presentation
8.00E−06
16
0.258065
0.04884
0.078843
−0.16837
0.000274


217
histone H4-K16 acetylation
8.43E−06
14
0.608696
0.121237
−0.18519
0.069265
0.000285


50
nuclear pore
8.75E−06
51
0.451327
0.042184
−0.08528
0.011917
0.000293


67
ATP hydrolysis coupled proton transport
9.46E−06
14
0.157303
0.001607
0.078724
−0.087
0.000314


143
gluconeogenesis
1.02E−05
26
0.376812
−0.04908
0.072529
−0.03359
0.000335


167
long-chain fatty acid transport
1.09E−05
4
0.307692
0.360173
−0.21507
−0.19082
0.000355


20
cytokinesis after mitosis
1.16E−05
14
0.424242
−0.19888
−0.01315
0.094845
0.000371


77
G2/M transition of mitotic cell cycle
1.17E−05
81
0.536424
−0.07129
−0.00286
0.055686
0.000371


1
histone acetyltransferase complex
1.29E−05
12
0.363636
0.131663
−0.2084
0.051313
0.000407


154
long-chain fatty acid-CoA ligase activity
1.31E−05
6
0.222222
0.234518
−0.1022
−0.1933
0.000408


52
septin complex
1.39E−05
7
0.145833
0.083409
−0.11239
0.044319
0.000429


244
phagocytic vesicle
1.46E−05
23
0.793103
0.038123
0.024386
−0.11006
0.000446


151
regulation of acetyl-CoA biosynthetic process from pyruvate
1.49E−05
10
0.5
−0.0432
0.083934
−0.11644
0.000453


218
spindle microtubule
1.61E−05
22
0.578947
−0.14636
−0.105
0.169429
0.000486


23
threonine-type endopeptidase activity
1.64E−05
18
0.321429
−0.16359
0.087207
0.039216
0.000487


152
calcium ion-dependent exocytosis
1.68E−05
6
0.26087
−0.02642
0.184774
−0.25201
0.000495


79
condensed chromosome kinetochore
1.71E−05
52
0.742857
−0.00514
−0.06144
0.099829
0.000499


153
long-chain fatty acid metabolic process
1.73E−05
9
0.204545
0.205768
−0.08773
−0.18052
0.0005


60
protein polymerization
1.79E−05
10
0.212766
−0.17849
0.172902
0.022303
0.000515


193
oligosaccharyltransferase complex
1.81E−05
9
0.529412
0.113519
0.007354
−0.14441
0.000517


68
proton-transporting ATPase activity, rotational mechanism
1.96E−05
11
0.366667
0.001805
0.088199
−0.09178
0.000549


4
RNA polymerase II transcription cofactor activity
1.96E−05
25
0.342466
0.127641
−0.14458
0.014519
0.000549


117
proteasome activator complex
1.99E−05
3
0.1875
−0.19317
0.114138
0.053545
0.000551


220
proteasome regulatory particle
2.01E−05
8
0.571429
−0.15151
0.04543
0.064738
0.000554


17
positive regulation of interferon-beta production
2.08E−05
13
0.565217
−0.17264
0.106172
0.079535
0.000566


103
protein N-linked glycosylation
2.10E−05
12
0.363636
0.099357
0.036393
−0.20665
0.000567


228
nucleosome disassembly
2.32E−05
12
0.705882
0.110704
−0.16429
−0.01081
0.000621


28
kinase activity
2.37E−05
19
0.131034
−0.18302
−0.01083
0.092884
0.000631


163
fatty acid beta-oxidation
2.40E−05
25
0.568182
0.014322
0.059874
−0.08516
0.000634


59
microtubule-based process
2.42E−05
16
0.158416
−0.17205
0.090011
0.034098
0.000634


9
transmembrane transporter activity
2.49E−05
14
0.215385
0.005268
0.074119
−0.10636
0.000646


42
negative regulation of catalytic activity
2.54E−05
32
0.235294
−0.09192
0.029677
0.027552
0.000655


180
endoderm development
2.59E−05
9
0.225
0.264604
−0.24097
−0.08909
0.000662


115
chromatin modification
2.76E−05
59
0.5
0.076637
−0.11972
0.028932
0.000698


174
lamin binding
2.77E−05
7
0.466667
0.118928
−0.22315
−0.07637
0.000698


47
cytosolic small ribosomal subunit
2.96E−05
33
0.804878
−0.07318
0.069572
−0.03488
0.000739


187
structural constituent of nuclear pore
3.15E−05
8
0.615385
0.059971
−0.11294
−0.0033
0.00078


40
peroxisomal membrane
3.22E−05
34
0.53125
0.053474
0.034593
−0.10099
0.00079


194
purine ribonucleoside monophosphate biosynthetic process
3.55E−05
11
0.785714
−0.15026
0.102894
0.02011
0.000865


166
viral receptor activity
3.70E−05
10
0.285714
−0.01626
0.169814
−0.16554
0.000895


156
protein N-terminus binding
3.79E−05
57
0.491379
0.067428
−0.08954
−0.00992
0.00091


248
mitochondrial ATP synthesis coupled proton transport
4.03E−05
14
0.875
0.013494
0.062777
−0.08601
0.00096


10
hydrogen peroxide catabolic process
4.12E−05
8
0.4
−0.09879
0.076328
−0.00322
0.000975


245
transcription export complex
4.23E−05
13
1
0.100418
−0.11607
0.02015
0.000994


177
cytochrome-c oxidase activity
4.41E−05
14
0.264151
−0.03129
0.125649
−0.13295
0.001029


16
extrinsic to membrane
4.63E−05
18
0.264706
−0.03703
0.129824
−0.1151
0.001072


29
integral to nuclear inner membrane
4.74E−05
6
0.222222
0.233519
−0.2111
−0.09488
0.001091


172
pyrimidine base metabolic process
4.87E−05
15
0.517241
−0.13504
−0.02491
0.146294
0.001112


224
MLL1 complex
4.96E−05
25
0.925926
0.116412
−0.12195
0.01688
0.001125


210
RNA polymerase II carboxy-terminal domain kinase activity
5.17E−05
14
0.823529
0.093156
−0.15945
0.071303
0.001163


197
chaperone-mediated protein complex assembly
5.37E−05
8
0.5
−0.14572
0.069709
−0.00641
0.00119


146
L-methionine salvage from methylthioadenosine
5.38E−05
9
0.692308
−0.18348
0.031838
0.088304
0.00119


145
membrane protein ectodomain proteolysis
5.46E−05
15
0.6
0.054086
0.019713
−0.14771
0.00119


203
mRNA transcription from RNA polymerase II promoter
5.46E−05
3
0.214286
0.125953
−0.15307
−0.01308
0.00119


38
nucleosome
5.54E−05
12
0.144578
0.125919
−0.22708
−0.03249
0.00119


104
DNA-dependent ATPase activity
5.54E−05
25
0.396825
0.125031
−0.139
−0.05421
0.00119


237
termination of RNA polymerase I transcription
5.55E−05
15
0.625
0.091394
−0.11934
−0.03859
0.00119


208
intracellular steroid hormone receptor signaling pathway
5.86E−05
10
0.454545
0.143333
−0.15993
0.014863
0.001249


249
polyamine metabolic process
6.59E−05
8
0.5
−0.18348
0.104104
0.083016
0.001395


66
steroid biosynthetic process
6.98E−05
10
0.27027
0.122999
−0.06091
−0.17027
0.001468


19
prefoldin complex
7.32E−05
9
0.375
−0.12995
0.183968
−0.07403
0.00153


57
spliceosomal complex
7.64E−05
73
0.62931
0.008675
−0.04912
0.047342
0.001587


39
nuclear-transcribed mRNA catabolic process, deadenylation-dependent decay
7.98E−05
43
0.781818
−0.07308
0.005716
0.070863
0.001646


62
glycolysis
8.03E−05
26
0.19697
−0.06513
0.077414
−0.01372
0.001646


45
Rab GTPase activator activity
8.15E−05
20
0.151515
−0.14442
−0.0419
0.155899
0.00165


46
positive regulation of Rab GTPase activity
8.15E−05
20
0.151515
−0.14442
−0.0419
0.155899
0.00165


213
L-serine transmembrane transporter activity
8.28E−05
3
0.3
−0.20839
0.292347
−0.12203
0.001654


214
L-serine transport
8.28E−05
3
0.3
−0.20839
0.292347
−0.12203
0.001654


69
ribose phosphate diphosphokinase activity
8.92E−05
4
0.2
−0.19295
−0.19221
0.34583
0.00175


140
ribose phosphate diphosphokinase complex
8.92E−05
4
0.235294
−0.19295
−0.19221
0.34583
0.00175


239
histone H4-K5 acetylation
8.97E−05
11
0.733333
0.128957
−0.14589
0.069265
0.00175


240
histone H4-K8 acetylation
8.97E−05
11
0.733333
0.128957
−0.14589
0.069265
0.00175


108
MCM complex
0.000103
8
0.421053
−0.28921
0.076952
0.09496
0.001973


255
Nup107-160 complex
0.000103
10
1
0.046194
−0.10112
0.060993
0.001973


238
endoplasmic reticulum-Golgi intermediate compartment membrane
0.000103
22
0.758621
0.07661
0.051038
−0.13193
0.001973


114
R-SMAD binding
0.000119
9
0.346154
0.132935
−0.14438
−0.04764
0.002273


169
protein kinase C activity
0.000122
7
0.155556
−0.07321
−0.13457
0.187185
0.002301


48
aminopeptidase activity
0.000126
22
0.392857
−0.10693
0.020834
0.007109
0.002362


113
sphingolipid metabolic process
0.000133
32
0.380952
0.051022
−0.02366
−0.06775
0.00248


138
binding of sperm to zona pellucida
0.000133
11
0.211538
−0.16374
0.069709
−0.00641
0.002482


254
telomere maintenance via semi-conservative replication
0.000142
19
0.863636
−0.16631
−0.01194
0.141727
0.002612


105
regulation of glucose transport
0.000144
28
0.875
0.048286
−0.10247
0.049905
0.002612


229
S100 protein binding
0.000144
8
0.727273
0.008799
0.229014
−0.24767
0.002612


137
ATP-dependent helicase activity
0.000144
32
0.359551
−0.00537
−0.06924
0.055416
0.002612


87
kinesin complex
0.000144
19
0.118012
−0.13828
−0.05856
0.084166
0.002612


198
neutral amino acid transmembrane transporter activity
0.000145
3
0.230769
−0.15789
0.344988
−0.22054
0.002612


233
branched chain family amino acid catabolic process
0.000151
17
0.944444
−0.07479
0.088313
−0.05722
0.002701


82
hydrogen ion transmembrane transporter activity
0.000152
13
0.1625
−0.00818
0.08546
−0.07369
0.002701


41
CCR4-NOT complex
0.000157
10
0.588235
−0.23048
0.046281
0.106251
0.002785


90
protein homotetramerization
0.000158
36
0.461538
−0.05337
0.051517
−0.03821
0.002786


89
small ribosomal subunit
0.000169
16
0.347826
−0.07187
0.073501
−0.02462
0.002946


242
U12-type spliceosomal complex
0.000169
17
0.708333
0.02057
−0.04352
0.057469
0.002946


202
negative regulation of nuclear mRNA splicing, via spliceosome
0.000176
14
0.875
−0.00441
0.08028
−0.13313
0.003041


212
morphogenesis of embryonic epithelium
0.000181
2
0.111111
0.039417
0.168697
−0.19555
0.003113


122
ER to Golgi vesicle-mediated transport
0.000182
43
0.401869
0.06241
−0.07115
−0.01915
0.003113


221
Cajal body
0.000203
31
0.688889
−0.07604
−0.00639
0.052887
0.00346


196
peroxisome organization
0.00021
9
0.375
0.084063
−0.01852
−0.09929
0.003557


78
GPI anchor biosynthetic process
0.000214
5
0.083333
0.294805
−0.10397
−0.31349
0.003606


116
Ran GTPase binding
0.000217
22
0.333333
−0.10986
0.00611
0.111111
0.00363


86
centrosome organization
0.000228
16
0.290909
−0.23218
0.15245
0.011865
0.003806


93
protein targeting to mitochondrion
0.000231
44
0.709677
0.002085
0.066315
−0.04183
0.003834


107
negative regulation of DNA binding
0.000243
10
0.263158
0.161091
−0.1102
0.019468
0.003979


164
ferric iron binding
0.000243
6
0.176471
0.022437
0.098371
−0.16208
0.003979


3
core promoter binding
0.000244
13
0.25
0.092963
−0.15894
0.004264
0.003979


83
integrin complex
0.00025
13
0.175676
0.01912
0.061454
−0.10513
0.004067


73
amino acid transport
0.000263
8
0.205128
−0.0588
0.270568
−0.22054
0.004246


241
interaction with host
0.00027
17
0.5
−0.01271
0.078724
−0.04691
0.004347


235
1-acylglycerol-3-phosphate O-acyltransferase activity
0.000285
9
0.75
0.192441
−0.0867
−0.19068
0.004558


191
ceramide biosynthetic process
0.000301
12
0.363636
0.098225
−0.04928
−0.04302
0.004802


54
protein transporter activity
0.000318
44
0.289474
−0.07167
0.004818
0.032935
0.005045


168
positive regulation of erythrocyte differentiation
0.000321
7
0.269231
0.111565
−0.15136
0.038154
0.005065


257
exoribonuclease activity
0.000323
11
0.916667
−0.03415
−0.09216
0.086495
0.005065


112
removal of superoxide radicals
0.000324
6
0.25
−0.11763
0.149906
−0.0077
0.005065


139
cyclin binding
0.000331
9
0.264706
−0.04245
−0.19008
0.149559
0.005143


91
U1 snRNP
0.000334
12
0.375
−0.00596
−0.04122
0.078437
0.005163


18
sarcolemma
0.000338
22
0.151724
−0.07723
0.10522
−0.11605
0.005198


184
CDP-diacylglycerol biosynthetic process
0.000345
7
0.5
0.192441
−0.05254
−0.19068
0.005285


43
anchored to membrane
0.000348
12
0.095238
0.008488
0.170286
−0.26789
0.0053


72
cytoplasmic vesicle membrane
0.000352
52
0.436975
−0.04855
0.06129
−0.04708
0.00534


247
mitochondrial proton-transporting ATP synthase complex
0.000358
17
0.809524
0.009968
0.074119
−0.12894
0.005401


58
structural constituent of cytoskeleton
0.000363
43
0.277419
−0.07932
0.005999
0.003183
0.005455


250
NuA4 histone acetyltransferase complex
0.000367
10
0.526316
0.122766
−0.14403
0.028307
0.005473


251
histone H2A acetylation
0.000367
10
0.588235
0.122766
−0.14403
0.028307
0.005473


129
mitotic spindle
0.000373
19
0.575758
−0.12863
−0.01536
0.159479
0.005524


44
histone deacetylase activity
0.000386
8
0.170213
0.029522
−0.1256
0.118448
0.005693


7
chromosome, centromeric region
0.000391
35
0.507246
0.029928
−0.0804
0.077759
0.005745


159
RNA polymerase II transcription factor binding
0.000396
11
0.234043
0.141107
−0.13948
0.017762
0.005796


189
stress-activated MAPK cascade
0.000399
27
0.457627
−0.08651
−0.03594
0.120591
0.005805


207
antigen processing and presentation of exogenous peptide antigen via MHC
0.0004
52
0.490566
−0.01457
−0.01579
0.012593
0.005805



class II


157
2 iron, 2 sulfur cluster binding
0.000418
12
0.363636
0.016279
0.106304
−0.0999
0.006039


134
de novo' IMP biosynthetic process
0.000436
6
0.6
−0.14833
0.102894
0.02011
0.006257


190
protein deacetylation
0.000442
8
0.727273
−0.03055
0.1145
0.177156
0.006325


109
response to starvation
0.000469
10
0.175439
−0.03493
0.07223
−0.07056
0.006683


26
signalosome
0.00049
24
0.428571
−0.08648
−0.00991
0.094622
0.006951


227
nucleobase-containing small molecule interconversion
0.000496
16
0.888889
−0.12572
0.082345
0.089469
0.006997


61
somitogenesis
0.000501
12
0.162162
0.109622
−0.15469
0.028021
0.007037


133
thyroid hormone receptor binding
0.000505
16
0.551724
0.137006
−0.13891
0.030687
0.007062


160
peroxisomal matrix
0.00051
23
0.638889
0.013884
0.070204
−0.10498
0.007104


185
very long-chain fatty acid metabolic process
0.000519
6
0.315789
0.12026
−0.04046
−0.18221
0.007201


178
anaphase-promoting complex
0.000528
16
0.432432
−0.12885
−0.052
0.122231
0.007292


209
tRNA methylation
0.000539
9
0.409091
−0.18051
0.015363
0.117699
0.007414


205
blastocyst hatching
0.000555
3
0.166667
0.211324
−0.10701
−0.13018
0.007599


147
signal peptide processing
0.000568
6
0.26087
0.110347
0.00227
−0.19979
0.007749


135
small-subunit processome
0.000596
9
0.473684
0.116951
−0.11034
−0.00289
0.008086


80
protein dephosphorylation
0.000605
35
0.244755
−0.0257
−0.01538
0.095514
0.008156


199
cell adhesion molecule binding
0.000606
14
0.297872
−0.0367
0.114708
−0.09618
0.008156


21
collagen binding
0.000618
23
0.157534
0.113374
−0.03584
−0.09703
0.008287


170
neurogenesis
0.000633
7
0.194444
−0.20103
0.077332
0.086537
0.008431


173
stress fiber assembly
0.000634
3
0.214286
−0.06578
−0.1713
0.202157
0.008431


70
nucleotide biosynthetic process
0.000638
5
0.192308
−0.19295
−0.12374
0.251821
0.008447


200
natural killer cell mediated cytotoxicity
0.000647
6
0.315789
−0.1951
0.149906
0.022303
0.008534


171
cytoplasmic stress granule
0.000666
27
0.55102
−0.078
0.07114
−0.0299
0.008727


125
mitotic sister chromatid segregation
0.000669
16
0.666667
−0.12167
−0.08992
0.152312
0.008727


51
microtubule cytoskeleton
0.00067
61
0.317708
−0.02645
−0.01958
0.045348
0.008727


236
transcription elongation from RNA polymerase I promoter
0.000678
13
0.619048
0.089633
−0.11298
−0.04193
0.008802


175
grooming behavior
0.000723
4
0.166667
−0.07312
−0.10665
0.167383
0.009319


55
phospholipid biosynthetic process
0.000726
10
0.217391
0.084369
0.032999
−0.12047
0.009319


92
microtubule bundle formation
0.000727
11
0.34375
−0.2003
0.01471
0.117994
0.009319


96
microtubule associated complex
0.000751
17
0.515152
−0.05546
0.118891
−0.04635
0.009594


76
regulation of cell cycle
0.000757
45
0.269461
−0.01198
−0.0694
0.075213
0.009626


181
protein serine/threonine/tyrosine kinase activity
0.000768
13
0.351351
−0.14239
−0.07836
0.208618
0.009725


234
integral to mitochondrial inner membrane
0.000771
13
0.866667
0.041683
0.097702
−0.0773
0.009725


121
NAD-dependent histone deacetylase activity (H3-K9 specific)
0.000776
6
0.4
0.026897
−0.12567
0.11708
0.009749


176
clathrin binding
0.000781
9
0.243243
−0.01664
0.100249
−0.12557
0.009785
















TABLE 3







Protein set enrichment in the space of correlations between proteins and the CDC protein markers
















G1_M
S_M
G2_M
blank
G1_U
S_U
G2_U
diff



















regulation of acetyl-CoA biosynthetic process
−0.0173
0.1480
−0.0578
0
−0.0726
0.1684
−0.0726
0.1686


from pyruvate


purine base metabolic process
−0.0663
0.0265
0.0067
0
−0.0534
0.0523
0.0069
0.1840


cellular nitrogen compound metabolic process
−0.0308
0.0259
0.0081
0
−0.0510
0.0281
0.0163
0.1906


alternative nuclear mRNA splicing, via
0.1823
−0.0950
−0.0950
0
0.1478
−0.1259
−0.0289
0.1949


spliceosome


nucleosome
0.3259
−0.0833
−0.0793
0
0.1835
−0.0989
−0.0487
0.2301


retrograde vesicle-mediated transport, Golgi to ER
−0.0544
0.0513
−0.0160
0
−0.0795
0.0502
0.0239
0.2403


protein polyubiquitination
−0.0308
0.0259
0.0286
0
−0.0375
0.0221
0.0034
0.2411


nucleobase-containing small molecule metabolic
−0.0550
0.0286
0.0299
0
−0.0361
0.0268
0.0056
0.2467


process


DNA damage response, signal transduction by p53
−0.0321
0.0264
0.0240
0
−0.0488
0.0232
0.0043
0.2494


class mediator resulting in cell cycle arrest


neuromuscular process controlling balance
−0.0671
0.0683
−0.0536
0
−0.0821
0.0873
0.0131
0.2707


cerebral cortex development
−0.0900
0.0084
0.0429
0
−0.0509
0.0201
0.0212
0.3105


chromatin organization
0.1124
−0.0253
−0.0372
0
0.0583
−0.0559
−0.0242
0.3119


glycosphingolipid metabolic process
−0.0694
0.1001
−0.0406
0
−0.0317
0.1313
0.0103
0.3123


long-chain fatty-acyl-CoA biosynthetic process
−0.1165
0.0725
0.0822
0
−0.0753
0.0499
0.0060
0.3479


triglyceride biosynthetic process
−0.1165
0.0725
0.0822
0
−0.0753
0.0499
0.0060
0.3479


regulation of cellular amino acid metabolic process
−0.0253
0.0210
0.0286
0
−0.0483
0.0377
0.0053
0.3793


proteasome complex
−0.0294
0.0185
0.0286
0
−0.0483
0.0377
0.0034
0.3822


antioxidant activity
−0.0832
0.0735
0.0126
0
−0.0284
0.0584
−0.0300
0.3931


positive regulation of ubiquitin-protein ligase
−0.0273
0.0193
0.0180
0
−0.0470
0.0336
−0.0116
0.4053


activity involved in mitotic cell cycle


sphingolipid metabolic process
−0.0004
0.0974
−0.0447
0
−0.0778
0.1134
−0.0008
0.4104


organ regeneration
−0.0742
0.0793
0.0403
0
−0.0346
0.0556
−0.0494
0.4592


regulation of alternative nuclear mRNA splicing,
0.0798
−0.0692
−0.0266
0
0.0142
−0.0588
0.0231
0.4625


via spliceosome


chromatin DNA binding
0.1894
−0.0544
−0.0460
0
0.0384
−0.0232
−0.0427
0.4708


AU-rich element binding
0.1265
−0.0172
−0.0241
0
0.0471
−0.0167
−0.1082
0.4826


oxidative phosphorylation
0.0229
0.0636
−0.0853
0
0.0238
0.0762
0.0631
0.4834


extrinsic to plasma membrane
−0.0251
0.1114
−0.0378
0
−0.0302
0.0340
−0.1490
0.4999


respiratory chain
0.1530
0.0415
−0.0414
0
0.0531
0.0066
−0.1126
0.5045


actin filament binding
−0.0745
0.0536
−0.0167
0
−0.0227
0.0244
0.0102
0.5343


proteasome core complex, alpha-subunit complex
−0.0081
0.0604
−0.0239
0
−0.0596
0.0150
−0.0361
0.5367


nuclear chromatin
0.1070
−0.0256
−0.0186
0
0.0415
−0.0624
0.0375
0.5417


binding of sperm to zona pellucida
−0.0760
−0.0533
0.0545
0
0.0272
−0.0177
0.0382
0.5812


nucleosome assembly
0.2323
0.0107
−0.0460
0
0.0541
−0.0525
−0.0334
0.5920


nuclear euchromatin
0.1838
−0.0875
−0.0548
0
0.0585
−0.0336
0.0436
0.6011


cell body
−0.0816
0.0346
0.0302
0
−0.0031
0.0092
0.0236
0.6066


catalytic step 2 spliceosome
0.0534
−0.0448
−0.0107
0
0.0002
−0.0277
0.0050
0.6068


cytochrome-c oxidase activity
0.1105
0.0092
−0.0635
0
0.0132
0.0153
−0.0190
0.6407


phosphate ion binding
−0.0613
0.0001
0.0791
0
−0.0882
0.1059
−0.0216
0.6553


membrane organization
−0.0648
0.0390
0.0459
0
−0.0170
0.0083
0.0057
0.6574


cytosolic large ribosomal subunit
−0.0926
−0.1471
0.0440
0
0.0624
−0.0509
0.0204
0.6583


GDP binding
−0.0094
0.0540
0.0012
0
−0.0506
0.0165
−0.0248
0.6690


small ribosomal subunit
−0.1499
−0.0633
0.0859
0
0.0576
−0.0737
0.0071
0.6782


translation elongation factor activity
−0.1778
−0.1602
0.1270
0
0.0296
−0.0584
0.0303
0.6960


actin filament polymerization
−0.2064
0.0673
0.0156
0
0.0176
0.0591
−0.0257
0.6981


Z disc
−0.0552
−0.0183
0.0394
0
−0.0196
0.0035
0.0001
0.7103


endoplasmic reticulum unfolded protein response
0.0098
0.0267
−0.0083
0
−0.0295
0.0125
0.0067
0.7328


translational elongation
−0.1143
−0.1466
0.0610
0
0.0598
−0.0375
0.0168
0.7511


rRNA binding
−0.1135
−0.1256
0.0460
0
0.0597
−0.0239
0.0241
0.7556


ruffle
−0.0776
−0.0450
0.0215
0
−0.0157
0.0169
0.0063
0.7595


viral transcription
−0.1080
−0.1466
0.0607
0
0.0598
−0.0337
0.0168
0.7630


chaperonin-containing T-complex
−0.0810
−0.0653
0.0428
0
0.0229
−0.0061
0.0190
0.7884


sperm protein complex
−0.0810
−0.0653
0.0428
0
0.0229
−0.0061
0.0190
0.7884


SRP-dependent cotranslational protein targeting to
−0.0926
−0.1141
0.0447
0
0.0544
−0.0221
0.0126
0.7962


membrane


translational termination
−0.1075
−0.1465
0.0552
0
0.0590
−0.0264
0.0141
0.8017


nuclear-transcribed mRNA catabolic process,
−0.1017
−0.1255
0.0489
0
0.0536
−0.0221
0.0141
0.8020


nonsense-mediated decay


natural killer cell mediated cytotoxicity
−0.1795
−0.1473
0.1505
0
0.1150
−0.0569
0.0053
0.8098


viral infectious cycle
−0.1058
−0.1421
0.0552
0
0.0587
−0.0221
0.0141
0.8180


ribosomal small subunit biogenesis
−0.1175
0.1554
0.1020
0
0.0401
−0.0075
0.0324
0.8247


de novo' posttranslational protein folding
−0.0722
0.0462
0.0672
0
0.0095
−0.0191
0.0198
0.8310


respiratory electron transport chain
0.0598
0.0192
−0.0362
0
0.0061
−0.0161
−0.0058
0.8347


platelet degranulation
−0.0640
0.0689
0.0388
0
0.0205
0.0161
−0.0165
0.8570


blood microparticle
−0.1046
−0.0898
0.0689
0
0.0150
−0.0190
−0.0076
0.8754


protein disulfide isomerase activity
0.0917
0.0573
−0.1152
0
−0.0138
0.0157
−0.0005
0.8895


glycolysis
−0.1081
−0.0693
0.0546
0
−0.0146
0.0070
−0.0566
0.9062


positive regulation of protein insertion into
−0.0695
0.0411
0.0880
0
0.0210
−0.0036
0.0101
0.9134


mitochondrial membrane involved in apoptotic


signaling pathway


MHC class II protein complex binding
−0.1600
−0.0124
0.1197
0
0.0905
−0.0065
0.0106
0.9143


cytosolic small ribosomal subunit
−0.1230
−0.1266
0.0849
0
0.0690
−0.0075
−0.0010
0.9634


protein polymerization
−0.1994
0.0203
0.1495
0
0.0768
0.0050
−0.0002
0.9776


microtubule-based process
−0.1498
0.0291
0.0908
0
0.0238
−0.0308
−0.0002
1


cellular component movement
−0.0188
0.0209
0.0220
0
0.0293
−0.0229
−0.0067
1


male gonad development
−0.1006
0.0477
0.0322
0
0.0125
−0.0015
−0.0080
1


negative regulation of protein kinase activity
−0.1137
0.0293
0.0910
0
0.0457
0.0081
−0.0852
1


prefoldin complex
0.0078
−0.0066
0.1214
0
−0.0901
0.0197
−0.0170
1


somitogenesis
0.2031
−0.0837
−0.0527
0
−0.0755
0.0226
0.0089
1
















TABLE 4







Additional proteins that correlate significantly to the CDC protein markers














prot
Phase
pval
cor
qval
celltype

















1
O00483
G1
0.002768
0.169348
0.037656
Melanoma


2
O14949
G1
0.002344
0.147051
0.033665
Melanoma


3
O14949
S
0.000517
0.204846
0.011391
Melanoma


4
O14979
G1
5.59E−07
0.215654
4.49E−05
Melanoma


5
O15143
S
0.002802
0.098584
0.037903
Melanoma


6
O43390
G1
9.54E−06
0.172743
0.000516
Melanoma


7
O43684
G1
0.002057
0.165257
0.030478
Melanoma


8
O43809
G1
2.46E−05
0.178244
0.001021
Melanoma


9
O60313
S
0.001803
0.163247
0.028151
Melanoma


10
O60637
G1
0.000187
0.211688
0.00515
Melanoma


11
O60869
G2
7.04E−08
0.238294
7.28E−06
Melanoma


12
O60925
G2
0.003253
0.131627
0.041701
Melanoma


13
O75153
G2
7.64E−24
0.544395
2.09E−21
Melanoma


14
O75367
G1
0.001815
0.138599
0.028238
Melanoma


15
O75390
G1
0.002781
0.07631
0.037731
Melanoma


16
O75494
G1
0.000787
0.226767
0.01602
Melanoma


17
O75494
S
0.000949
0.175789
0.018477
Melanoma


18
O94776
S
1.53E−05
0.161673
0.000741
Melanoma


19
O95433
S
0.002129
0.138191
0.031154
Melanoma


20
O95881
S
0.003864
0.13557
0.046574
Melanoma


21
P00403
G1
0.00131
0.16342
0.022751
Melanoma


22
P00441
S
9.92E−59
0.600106
6.59E−56
Melanoma


23
P00505
G1
0.004013
0.137244
0.047995
Melanoma


24
P02545
G1
2.02E−11
0.329407
3.25E−09
Melanoma


25
P04406
G2
0.000387
0.168334
0.009187
Melanoma


26
P04792
G2
0.000401
0.133196
0.009432
Melanoma


27
P06748
G1
6.66E−09
0.342976
7.94E−07
Melanoma


28
P07195
G2
0.001065
0.13238
0.019987
Melanoma


29
P07305
G1
2.17E−09
0.299325
2.80E−07
Melanoma


30
P07437
G2
0.001428
0.158986
0.024079
Melanoma


31
P07910
G1
0.002543
0.195706
0.035751
Melanoma


32
P08311
S
8.90E−05
0.123551
0.002857
Melanoma


33
P08567
G2
0.00035
0.214853
0.008699
Melanoma


34
P08670
G1
0.000915
0.197406
0.017885
Melanoma


35
P09012
S
0.00289
0.092693
0.038308
Melanoma


36
P09651
G1
0.001176
0.148675
0.021288
Melanoma


37
P09669
G1
8.03E−09
0.213605
9.34E−07
Melanoma


38
P10412
G1
2.78E−27
0.388801
8.62E−25
Melanoma


39
P10809
G1
0.002616
0.099564
0.036551
Melanoma


40
P11310
S
1.44E−29
0.429362
4.79E−27
Melanoma


41
P11387
G1
0.002001
0.114461
0.030142
Melanoma


42
P11940
G2
1.83E−05
0.167822
0.000811
Melanoma


43
P12236
G1
0.002655
0.06312
0.036771
Melanoma


44
P12956
G1
2.07E−05
0.218699
0.000907
Melanoma


45
P13010
G1
8.90E−05
0.272558
0.002857
Melanoma


46
P13473
S
0.001462
0.140278
0.024566
Melanoma


47
P13667
G1
0.001112
0.148656
0.020607
Melanoma


48
P14174
G2
0.000326
0.162901
0.008246
Melanoma


49
P14324
G2
1.40E−54
0.597771
8.15E−52
Melanoma


50
P14618
G2
0.001114
0.149989
0.020607
Melanoma


51
P14854
G1
0.003012
0.075339
0.039477
Melanoma


52
P14866
G1
0.003711
0.123486
0.045559
Melanoma


53
P14866
S
0.003516
0.069344
0.044212
Melanoma


54
P14927
G1
0.001216
0.164223
0.021908
Melanoma


55
P15559
G2
0.00043
0.161882
0.010054
Melanoma


56
P16104
G1
5.27E−40
0.546449
2.45E−37
Melanoma


57
P16150
S
0.001531
0.189542
0.02535
Melanoma


58
P16401
G1
 3.12E−213
0.883314
 1.45E−209
Melanoma


59
P16402
G1
6.70E−14
0.295448
1.42E−11
Melanoma


60
P16403
G1
 1.75E−102
0.729705
2.71E−99
Melanoma


61
P16949
G2
0.002363
0.118805
0.033828
Melanoma


62
P17096
G1
0.000472
0.112873
0.01076
Melanoma


63
P17844
G1
0.001129
0.173822
0.020607
Melanoma


64
P17931
G2
0.0017
0.140174
0.027269
Melanoma


65
P19338
G1
0.00288
0.213242
0.03829
Melanoma


66
P19838
S
0.003787
0.292409
0.046019
Melanoma


67
P19878
G1
0.000867
0.317421
0.017094
Melanoma


68
P20290
G2
0.00304
0.150959
0.039737
Melanoma


69
P20671
G1
9.48E−14
0.500856
1.92E−11
Melanoma


70
P20700
G1
3.28E−06
0.161075
0.000206
Melanoma


71
P20962
G2
0.00033
0.139458
0.008247
Melanoma


72
P21333
G2
0.003579
0.09646
0.044644
Melanoma


73
P22087
G1
2.02E−11
0.317622
3.25E−09
Melanoma


74
P22307
S
0.002059
0.135241
0.030478
Melanoma


75
P22626
G1
3.41E−09
0.242429
4.29E−07
Melanoma


76
P23246
G1
8.62E−07
0.287383
6.37E−05
Melanoma


77
P24158
S
0.00052
0.223539
0.011414
Melanoma


78
P24534
G2
0.002112
0.156845
0.031004
Melanoma


79
P24539
G1
0.000123
0.14206
0.003755
Melanoma


80
P25705
G1
3.43E−06
0.137582
0.000213
Melanoma


81
P25787
S
6.11E−06
0.232148
0.000351
Melanoma


82
P26599
G1
6.91E−06
0.212563
0.000392
Melanoma


83
P27348
G2
0.004145
0.112018
0.049308
Melanoma


84
P27824
S
0.001765
0.112216
0.027844
Melanoma


85
P28072
S
0.002875
0.111916
0.03829
Melanoma


86
P30084
S
0.003094
0.144295
0.040217
Melanoma


87
P30101
G1
0.001226
0.141828
0.021945
Melanoma


88
P31040
G1
0.002479
0.112175
0.035167
Melanoma


89
P31040
S
0.003773
0.106521
0.046019
Melanoma


90
P33991
G1
0.004082
0.080676
0.048706
Melanoma


91
P35232
G1
1.56E−08
0.180357
1.69E−06
Melanoma


92
P35613
S
9.12E−38
0.472115
3.86E−35
Melanoma


93
P36957
G1
0.0002
0.105997
0.005429
Melanoma


94
P37108
G1
0.003654
0.141164
0.045217
Melanoma


95
P38159
G1
8.92E−10
0.269653
1.24E−07
Melanoma


96
P38646
G1
0.000359
0.09239
0.008844
Melanoma


97
P40926
G1
5.37E−06
0.187222
0.000312
Melanoma


98
P43243
G1
4.00E−11
0.289435
6.20E−09
Melanoma


99
P43307
S
0.002922
0.145585
0.038623
Melanoma


100
P47985
G1
4.25E−05
0.184489
0.001585
Melanoma


101
P48637
S
5.85E−30
0.45043
2.09E−27
Melanoma


102
P49327
G2
0.000834
0.156784
0.016799
Melanoma


103
P49411
G1
0.001028
0.128178
0.019522
Melanoma


104
P50402
G1
1.18E−05
0.139485
0.000603
Melanoma


105
P50502
G2
0.001378
0.147762
0.023458
Melanoma


106
P51991
G1
7.98E−06
0.224153
0.000447
Melanoma


107
P52272
G1
3.16E−06
0.15495
0.000202
Melanoma


108
P52434
G2
0.001978
0.147077
0.029984
Melanoma


109
P56381
G1
9.74E−07
0.199585
7.08E−05
Melanoma


110
P56545
G1
0.00163
0.185884
0.026524
Melanoma


111
P61006
S
0.00238
0.199354
0.033969
Melanoma


112
P61026
G1
0.000757
0.10029
0.015576
Melanoma


113
P61289
G2
5.75E−63
0.597737
4.46E−60
Melanoma


114
P61604
S
0.002164
0.110594
0.031562
Melanoma


115
P62306
S
0.000839
0.217185
0.016835
Melanoma


116
P62314
G1
0.000358
0.142013
0.008844
Melanoma


117
P62805
G1
 2.08E−179
0.870036
 4.84E−176
Melanoma


118
P62807
G1
7.90E−84
0.693619
9.19E−81
Melanoma


119
P62826
G2
0.002741
0.171895
0.037622
Melanoma


120
P63162
S
3.91E−05
0.18419
0.001516
Melanoma


121
P68371
G2
7.45E−05
0.153233
0.002494
Melanoma


122
P78527
G1
1.97E−14
0.257906
4.37E−12
Melanoma


123
P84103
G1
3.80E−07
0.228718
3.34E−05
Melanoma


124
P99999
G1
5.63E−05
0.194457
0.002016
Melanoma


125
Q00325
G1
6.44E−07
0.152629
4.99E−05
Melanoma


126
Q00839
G1
3.45E−15
0.379258
8.45E−13
Melanoma


127
Q01130
G1
0.001927
0.047233
0.029495
Melanoma


128
Q03252
G1
0.001716
0.104034
0.027345
Melanoma


129
Q07666
G1
9.47E−08
0.248984
9.58E−06
Melanoma


130
Q07955
G1
5.43E−05
0.113668
0.00196
Melanoma


131
Q08211
G1
0.000115
0.198862
0.003583
Melanoma


132
Q13151
G1
4.16E−05
0.170652
0.001574
Melanoma


133
Q13243
G2
4.04E−05
0.397253
0.001555
Melanoma


134
Q13247
G1
0.000722
0.192208
0.014991
Melanoma


135
Q13257
G2
1.54E−30
0.668233
5.99E−28
Melanoma


136
Q13561
G1
0.002575
0.152915
0.036091
Melanoma


137
Q13595
S
0.001903
0.238034
0.029228
Melanoma


138
Q14247
G2
0.000234
0.181435
0.006153
Melanoma


139
Q14677
G2
0.002081
0.100842
0.030644
Melanoma


140
Q14978
G1
2.16E−05
0.214911
0.000931
Melanoma


141
Q15233
G1
0.000189
0.218454
0.005168
Melanoma


142
Q15365
S
0.001026
0.147359
0.019522
Melanoma


143
Q15370
G2
0.003911
0.123677
0.047028
Melanoma


144
Q15392
G1
0.001652
0.112106
0.026777
Melanoma


145
Q15424
G1
0.001564
0.141457
0.025619
Melanoma


146
Q15907
G1
0.000561
0.1549
0.012129
Melanoma


147
Q16778
G1
1.80E−40
0.522673
9.32E−38
Melanoma


148
Q16836
G1
8.07E−05
0.123684
0.002662
Melanoma


149
Q16891
G1
0.001888
0.1483
0.029228
Melanoma


150
Q5XPI4
G1
0.003822
0.167665
0.04619
Melanoma


151
Q71DI3
G1
6.16E−66
0.598935
5.73E−63
Melanoma


152
Q7Z434
S
1.62E−14
0.450597
3.77E−12
Melanoma


153
Q86UE4
S
7.37E−05
0.12934
0.002485
Melanoma


154
Q8TCJ2
G1
0.002008
0.156284
0.030142
Melanoma


155
Q8TER0
S
0.003351
0.344735
0.042724
Melanoma


156
Q92522
G1
1.54E−06
0.174952
0.000103
Melanoma


157
Q92616
S
0.000648
0.131248
0.013753
Melanoma


158
Q92945
G1
0.000773
0.084775
0.015846
Melanoma


159
Q92947
G1
0.000232
0.247869
0.006131
Melanoma


160
Q96AE4
G1
0.002507
0.09384
0.03546
Melanoma


161
Q96SU4
S
0.002725
0.121777
0.03751
Melanoma


162
Q99623
G1
8.36E−10
0.209161
1.22E−07
Melanoma


163
Q99729
G1
6.47E−11
0.298252
9.72E−09
Melanoma


164
Q99848
G1
5.01E−07
0.268132
4.17E−05
Melanoma


165
Q99878
G1
9.61E−25
0.371213
2.80E−22
Melanoma


166
Q99880
G1
6.92E−18
0.413637
1.79E−15
Melanoma


167
Q9BQE3
G2
0.00013
0.209228
0.003891
Melanoma


168
Q9BVC6
G1
5.35E−05
0.133219
0.001944
Melanoma


169
Q9BZH6
S
0.000477
0.139022
0.01082
Melanoma


170
Q9H773
S
0.000181
0.21258
0.005022
Melanoma


171
Q9NVP1
G1
0.000162
0.133282
0.004673
Melanoma


172
Q9NX63
G1
0.001629
0.129154
0.026524
Melanoma


173
Q9NX63
S
0.00086
0.204211
0.017094
Melanoma


174
Q9UBM7
G1
1.13E−08
0.22183
1.25E−06
Melanoma


175
Q9UKM9
G1
1.53E−09
0.269349
2.04E−07
Melanoma


176
Q9UMS4
G1
1.44E−07
0.228575
1.42E−05
Melanoma


177
Q9Y277
S
0.00175
0.144972
0.027758
Melanoma


178
O14979
G1
0.000277
0.125208
0.020463
Monocyte


179
P00441
S
 3.46E−101
0.73174
5.37E−98
Monocyte


180
P00558
G1
0.000114
0.176898
0.010231
Monocyte


181
P04075
G1
5.76E−08
0.216304
1.58E−05
Monocyte


182
P05204
G1
0.00078
0.151994
0.043739
Monocyte


183
P06748
G1
3.58E−05
0.185525
0.003964
Monocyte


184
P07437
G1
0.000189
0.189549
0.014438
Monocyte


185
P09429
G1
4.87E−08
0.202385
1.41E−05
Monocyte


186
P09651
G1
9.44E−05
0.137829
0.008872
Monocyte


187
P10412
G1
2.25E−08
0.165507
7.47E−06
Monocyte


188
P11142
G1
0.000732
0.148992
0.041553
Monocyte


189
P11310
S
2.86E−30
0.531988
1.33E−27
Monocyte


190
P12236
G1
2.92E−05
0.14846
0.003393
Monocyte


191
P16104
G1
5.66E−12
0.243373
2.19E−09
Monocyte


192
P16401
G1
 1.66E−150
0.7938
 7.72E−147
Monocyte


193
P16403
G1
2.22E−38
0.46972
1.48E−35
Monocyte


194
P17844
G1
9.53E−05
0.16121
0.008872
Monocyte


195
P18124
G1
8.55E−06
0.175857
0.001326
Monocyte


196
P19338
G1
3.20E−07
0.210507
7.84E−05
Monocyte


197
P22087
G1
6.36E−06
0.172042
0.001138
Monocyte


198
P22626
G1
7.75E−06
0.167487
0.001265
Monocyte


199
P23141
S
0.000433
0.134933
0.028954
Monocyte


200
P23297
S
2.65E−05
0.146244
0.00316
Monocyte


201
P24534
G1
7.15E−05
0.142242
0.007078
Monocyte


202
P26373
G1
2.09E−05
0.155062
0.002669
Monocyte


203
P29401
G1
9.24E−06
0.19487
0.001344
Monocyte


204
P30084
S
0.000672
0.125445
0.039092
Monocyte


205
P31350
G1
8.41E−05
0.168671
0.008152
Monocyte


206
P31949
G1
0.000419
0.077157
0.028698
Monocyte


207
P35613
S
3.64E−54
0.551083
3.39E−51
Monocyte


208
P39023
G1
3.48E−05
0.144492
0.003954
Monocyte


209
P46776
G1
9.11E−06
0.175346
0.001344
Monocyte


210
P46781
G1
0.000458
0.120098
0.029581
Monocyte


211
P48637
S
5.13E−35
0.501312
2.65E−32
Monocyte


212
P49207
G1
1.35E−05
0.168547
0.001798
Monocyte


213
P49588
S
0.000842
0.177025
0.045668
Monocyte


214
P51452
S
0.000106
0.326714
0.009684
Monocyte


215
P52566
G1
1.39E−07
0.203099
3.60E−05
Monocyte


216
P60709
G1
5.03E−06
0.187335
0.000971
Monocyte


217
P61254
G1
5.22E−06
0.177382
0.000971
Monocyte


218
P62249
G1
0.000307
0.138409
0.021943
Monocyte


219
P62328
G1
3.00E−06
0.200186
0.000635
Monocyte


220
P62701
G1
0.000484
0.116107
0.030857
Monocyte


221
P62805
G1
 4.45E−115
0.702605
 1.04E−111
Monocyte


222
P62807
G1
4.62E−46
0.446036
3.59E−43
Monocyte


223
P62888
G1
4.50E−05
0.180981
0.004873
Monocyte


224
P62899
G1
0.000668
0.117687
0.039092
Monocyte


225
P84090
G1
2.24E−05
0.190342
0.002747
Monocyte


226
P84103
G1
0.000231
0.154852
0.017342
Monocyte


227
Q00839
G1
0.000129
0.146843
0.011145
Monocyte


228
Q01130
G1
1.12E−05
0.162499
0.001545
Monocyte


229
Q04941
S
0.00062
0.212157
0.037298
Monocyte


230
Q13257
G2
5.61E−64
0.902197
6.53E−61
Monocyte


231
Q14247
S
0.000583
0.126623
0.035707
Monocyte


232
Q16778
G1
1.86E−22
0.320098
7.85E−20
Monocyte


233
Q71DI3
G1
1.24E−37
0.443394
7.20E−35
Monocyte


234
Q7Z434
S
0.000144
0.339479
0.011545
Monocyte


235
Q8IY50
G1
0.000574
0.16569
0.035637
Monocyte


236
Q8TER0
S
2.96E−06
0.474726
0.000635
Monocyte


237
Q96I99
S
0.000987
0.128073
0.049918
Monocyte


238
Q96KB5
S
0.000497
0.289828
0.031244
Monocyte


239
Q99878
G1
3.81E−11
0.223383
1.36E−08
Monocyte


240
Q99880
G1
2.93E−08
0.258416
9.08E−06
Monocyte


241
Q9BQE3
G1
0.000147
0.120295
0.011626
Monocyte


242
Q9UIG0
G1
0.000181
0.12043
0.014074
Monocyte


243
Q9UMS4
G1
7.88E−06
0.16617
0.001265
Monocyte
















TABLE 5







Differentially abundant proteins


between melanoma sub-populations













pvals
prot
FC
qval
Condition
















1
3.12E−14
E9PAV3
−0.82568
2.83E−13
Cluster A


2
0.000425
O00193
−0.61361
0.001112
Cluster A


3
1.86E−05
O00232
0.355623
6.46E−05
Cluster B


4
2.29E−16
O00244
−0.63274
2.46E−15
Cluster A


5
9.88E−09
O00299
−0.45768
4.96E−08
Cluster A


6
1.49E−07
O00483
0.644639
6.53E−07
Cluster B


7
0.002487
O00541
0.200772
0.00563
Cluster B


8
2.64E−06
O00625
−0.65861
1.03E−05
Cluster A


9
1.52E−08
O14561
−0.67198
7.45E−08
Cluster A


10
8.35E−05
O14737
−0.26512
0.000248
Cluster A


11
1.27E−05
O14818
0.201452
4.56E−05
Cluster B


12
0.000841
O14880
0.506075
0.002113
Cluster B


13
1.65E−06
O14949
0.570996
6.51E−06
Cluster B


14
3.50E−07
O14979
0.409702
1.49E−06
Cluster B


15
0.001232
O15160
0.453257
0.003
Cluster B


16
0.000466
O15258
0.258014
0.001213
Cluster B


17
2.00E−08
O15427
0.452887
9.75E−08
Cluster B


18
0.001684
O43143
0.151812
0.003953
Cluster B


19
6.33E−09
O43149
−0.8572
3.27E−08
Cluster A


20
6.21E−23
O43175
−0.7895
1.13E−21
Cluster A


21
2.09E−11
O43390
0.478258
1.39E−10
Cluster B


22
0.000266
O43615
0.263402
0.000723
Cluster B


23
0.000105
O43660
0.532088
0.00031
Cluster B


24
6.30E−10
O43707
−0.35768
3.61E−09
Cluster A


25
6.35E−05
O43719
0.669254
0.000196
Cluster B


26
2.23E−07
O43776
−0.70144
9.66E−07
Cluster A


27
0.000503
O43809
0.312914
0.001301
Cluster B


28
9.07E−07
O43852
0.2741
3.69E−06
Cluster B


29
6.31E−05
O60506
0.12646
0.000195
Cluster B


30
7.20E−07
O60637
0.673247
2.96E−06
Cluster B


31
0.001285
O60701
−0.37834
0.003113
Cluster A


32
5.28E−05
O60762
0.487165
0.000168
Cluster B


33
3.36E−08
O60869
−0.5431
1.58E−07
Cluster A


34
1.34E−05
O75083
−0.26055
4.80E−05
Cluster A


35
2.08E−30
O75347
−1.54061
8.86E−29
Cluster A


36
7.55E−05
O75367
0.281095
0.000228
Cluster B


37
0.004071
O75368
−0.35157
0.008915
Cluster A


38
0.000553
O75390
0.182302
0.00142
Cluster B


39
5.39E−05
O75396
0.284217
0.00017
Cluster B


40
3.19E−05
O75475
0.517588
0.000106
Cluster B


41
1.17E−08
O75494
0.382192
5.85E−08
Cluster B


42
0.000237
O75533
0.246317
0.000646
Cluster B


43
7.48E−06
O75822
−0.4246
2.78E−05
Cluster A


44
4.88E−19
O75874
−0.61024
6.57E−18
Cluster A


45
5.63E−09
O75937
−0.59283
2.92E−08
Cluster A


46
0.001174
O75947
0.244923
0.002865
Cluster B


47
0.000212
O75952
−0.40123
0.000586
Cluster A


48
4.09E−05
O76021
0.292958
0.000133
Cluster B


49
0.000103
O94776
0.184518
0.000305
Cluster B


50
0.004086
O94905
0.207211
0.008934
Cluster B


51
5.88E−10
O95831
0.52995
3.39E−09
Cluster B


52
0.001268
O96000
0.423476
0.003082
Cluster B


53
2.11E−32
P00338
−1.0044
1.35E−30
Cluster A


54
0.000141
P00390
0.31313
0.000409
Cluster B


55
1.12E−11
P00403
0.549323
7.59E−11
Cluster B


56
4.74E−05
P00491
0.558853
0.000152
Cluster B


57
1.60E−13
P00505
0.416121
1.31E−12
Cluster B


58
3.74E−30
P00558
−0.73015
1.49E−28
Cluster A


59
1.37E−13
P02545
0.349926
1.13E−12
Cluster B


60
0.002469
P02746
−0.41888
0.0056
Cluster A


61
0.000781
P02786
0.312129
0.001973
Cluster B


62
6.88E−37
P04075
−1.07509
1.76E−34
Cluster A


63
3.12E−13
P04080
−0.55725
2.42E−12
Cluster A


64
2.18E−34
P04406
−1.10982
2.32E−32
Cluster A


65
1.21E−15
P04792
−0.49493
1.26E−14
Cluster A


66
3.10E−11
P04843
0.354144
2.00E−10
Cluster B


67
4.50E−13
P05023
0.486066
3.40E−12
Cluster B


68
7.75E−07
P05141
0.31598
3.17E−06
Cluster B


69
0.000468
P05198
−0.24527
0.001218
Cluster A


70
0.003529
P05387
0.325099
0.007811
Cluster B


71
1.16E−19
P06396
−1.06764
1.62E−18
Cluster A


72
2.53E−14
P06454
−0.93455
2.33E−13
Cluster A


73
0.000809
P06576
0.228252
0.002036
Cluster B


74
3.62E−30
P06703
−1.28798
1.49E−28
Cluster A


75
1.53E−37
P06733
−1.36521
7.99E−35
Cluster A


76
0.001015
P06737
−0.22981
0.00251
Cluster A


77
7.10E−20
P06744
−0.605
1.03E−18
Cluster A


78
1.53E−05
P06748
0.381867
5.39E−05
Cluster B


79
1.59E−05
P06753
−0.42719
5.61E−05
Cluster A


80
2.11E−29
P07195
−0.89401
7.51E−28
Cluster A


81
5.10E−14
P07237
0.354456
4.53E−13
Cluster B


82
1.40E−07
P07305
0.567936
6.19E−07
Cluster B


83
4.66E−10
P07339
0.348277
2.77E−09
Cluster B


84
2.01E−08
P07355
−0.27484
9.75E−08
Cluster A


85
1.50E−29
P07437
−1.38465
5.49E−28
Cluster A


86
0.00053
P07686
0.466031
0.001363
Cluster B


87
1.65E−18
P07737
−0.68596
2.15E−17
Cluster A


88
2.29E−18
P07814
−0.55223
2.92E−17
Cluster A


89
2.99E−37
P07900
−0.76978
9.56E−35
Cluster A


90
8.00E−14
P07910
0.290417
6.87E−13
Cluster B


91
0.00023
P07954
0.229223
0.000629
Cluster B


92
0.000404
P08195
0.525502
0.001063
Cluster B


93
2.45E−31
P08238
−0.86512
1.20E−29
Cluster A


94
0.001004
P08559
0.609111
0.002493
Cluster B


95
2.86E−05
P08574
0.714068
9.55E−05
Cluster B


96
5.53E−05
P08579
0.264134
0.000173
Cluster B


97
3.59E−05
P08621
0.209994
0.000118
Cluster B


98
0.000951
P08670
0.24358
0.002371
Cluster B


99
4.22E−07
P08708
−0.42354
1.76E−06
Cluster A


100
1.65E−05
P08758
−0.30339
5.78E−05
Cluster A


101
9.71E−08
P09012
0.339135
4.37E−07
Cluster B


102
1.75E−05
P09104
−0.35185
6.13E−05
Cluster A


103
2.04E−18
P09429
−0.9781
2.64E−17
Cluster A


104
2.95E−13
P09496
−0.42384
2.32E−12
Cluster A


105
0.001377
P09525
0.640199
0.003297
Cluster B


106
5.36E−10
P09651
0.34817
3.15E−09
Cluster B


107
0.001055
P09661
0.521305
0.002606
Cluster B


108
5.33E−13
P09669
0.623897
3.99E−12
Cluster B


109
0.000377
P0DJD0
−0.64078
0.001002
Cluster A


110
3.06E−13
P0DP25
−0.56427
2.39E−12
Cluster A


111
0.000314
P10412
0.278471
0.000844
Cluster B


112
7.55E−05
P10599
−0.28802
0.000228
Cluster A


113
5.57E−10
P10768
−0.5146
3.26E−09
Cluster A


114
6.40E−14
P10809
0.384987
5.57E−13
Cluster B


115
2.75E−13
P11021
0.353903
2.17E−12
Cluster B


116
3.45E−34
P11142
−0.5486
3.40E−32
Cluster A


117
3.40E−06
P11177
0.374213
1.30E−05
Cluster B


118
0.003608
P11310
0.272773
0.00797
Cluster B


119
0.001949
P11387
0.177075
0.004515
Cluster B


120
1.10E−23
P11413
−0.88257
2.07E−22
Cluster A


121
6.61E−08
P11586
−0.3257
3.02E−07
Cluster A


122
1.50E−09
P11766
−0.51913
8.32E−09
Cluster A


123
4.24E−27
P11940
−0.59068
1.21E−25
Cluster A


124
1.35E−21
P12236
0.483392
2.21E−20
Cluster B


125
7.03E−15
P12268
−0.49227
6.87E−14
Cluster A


126
0.000156
P12955
−0.3736
0.000442
Cluster A


127
0.000284
P12956
0.27326
0.000767
Cluster B


128
0.001533
P13010
0.129831
0.003644
Cluster B


129
6.19E−07
P13073
0.409011
2.55E−06
Cluster B


130
0.001366
P13473
0.43576
0.003283
Cluster B


131
5.60E−14
P13489
−0.58986
4.94E−13
Cluster A


132
1.53E−36
P13639
−0.86102
2.80E−34
Cluster A


133
8.30E−15
P13667
0.443775
7.98E−14
Cluster B


134
2.12E−05
P13797
−0.46211
7.26E−05
Cluster A


135
1.23E−05
P14174
−0.8638
4.44E−05
Cluster A


136
0.000179
P14314
0.226635
0.000501
Cluster B


137
1.29E−19
P14324
−0.72374
1.77E−18
Cluster A


138
2.22E−07
P14550
−0.43144
9.66E−07
Cluster A


139
3.52E−36
P14618
−1.03235
5.62E−34
Cluster A


140
7.86E−19
P14625
0.436893
1.04E−17
Cluster B


141
0.000145
P14854
0.370584
0.000418
Cluster B


142
1.37E−13
P14866
0.351631
1.13E−12
Cluster B


143
0.000417
P14868
−0.20749
0.001092
Cluster A


144
6.46E−12
P14927
0.919169
4.44E−11
Cluster B


145
2.76E−25
P15559
−0.89132
6.61E−24
Cluster A


146
6.51E−28
P15880
−0.86918
2.03E−26
Cluster A


147
4.15E−10
P16104
0.430715
2.48E−09
Cluster B


148
6.71E−09
P16401
0.359916
3.45E−08
Cluster B


149
1.29E−06
P16402
0.440066
5.19E−06
Cluster B


150
2.54E−16
P16403
0.483059
2.71E−15
Cluster B


151
3.17E−08
P16615
0.452749
1.50E−07
Cluster B


152
2.36E−09
P16949
−0.33546
1.27E−08
Cluster A


153
2.68E−05
P17813
−0.95997
8.99E−05
Cluster A


154
0.001607
P17844
0.112873
0.003791
Cluster B


155
5.70E−18
P17931
−0.60021
6.94E−17
Cluster A


156
0.002284
P17987
−0.14194
0.005217
Cluster A


157
8.91E−27
P18077
−0.74137
2.37E−25
Cluster A


158
3.07E−33
P18124
−0.68313
2.31E−31
Cluster A


159
6.40E−10
P18621
−0.46983
3.65E−09
Cluster A


160
2.43E−15
P18669
−0.71882
2.47E−14
Cluster A


161
6.23E−06
P18859
0.582091
2.33E−05
Cluster B


162
2.17E−16
P19105
−0.98987
2.35E−15
Cluster A


163
8.96E−05
P19623
−0.32276
0.000265
Cluster A


164
5.22E−10
P20042
−0.55471
3.08E−09
Cluster A


165
5.82E−12
P20290
−0.66705
4.02E−11
Cluster A


166
0.001081
P20340
0.288453
0.00266
Cluster B


167
8.78E−11
P20674
0.331045
5.40E−10
Cluster B


168
4.25E−08
P20700
0.425694
1.97E−07
Cluster B


169
4.53E−12
P20962
−0.66494
3.17E−11
Cluster A


170
0.000227
P21281
−0.27116
0.000624
Cluster A


171
0.001157
P21333
−0.0888
0.002829
Cluster A


172
5.54E−11
P22061
−0.83965
3.50E−10
Cluster A


173
3.64E−09
P22087
0.419214
1.92E−08
Cluster B


174
9.74E−09
P22102
−0.40844
4.90E−08
Cluster A


175
6.00E−17
P22234
−0.87472
6.92E−16
Cluster A


176
0.004433
P22307
0.391144
0.00961
Cluster B


177
2.60E−05
P22314
−0.31986
8.76E−05
Cluster A


178
1.79E−15
P22392
−0.75493
1.85E−14
Cluster A


179
8.40E−11
P22626
0.264726
5.19E−10
Cluster B


180
0.00022
P22695
0.274443
0.000607
Cluster B


181
0.000281
P23193
−0.34296
0.000759
Cluster A


182
1.08E−19
P23246
0.453158
1.52E−18
Cluster B


183
1.06E−06
P23284
0.235905
4.28E−06
Cluster B


184
8.46E−09
P23297
−0.54046
4.28E−08
Cluster A


185
9.02E−23
P23396
−0.59555
1.63E−21
Cluster A


186
1.34E−13
P23526
−0.41283
1.11E−12
Cluster A


187
5.06E−36
P23528
−1.37736
7.20E−34
Cluster A


188
0.000206
P23588
−0.23232
0.000573
Cluster A


189
3.10E−31
P24534
−1.09398
1.47E−29
Cluster A


190
2.42E−18
P24539
0.727454
3.06E−17
Cluster B


191
2.65E−11
P24752
0.382243
1.76E−10
Cluster B


192
4.12E−06
P24941
−0.30691
1.57E−05
Cluster A


193
1.57E−06
P25398
−0.50855
6.23E−06
Cluster A


194
9.47E−23
P25705
0.57104
1.68E−21
Cluster B


195
2.50E−25
P26038
−0.5567
6.15E−24
Cluster A


196
0.000155
P26368
0.502116
0.000442
Cluster B


197
1.67E−30
P26373
−0.72597
7.38E−29
Cluster A


198
0.004603
P26447
0.174892
0.009962
Cluster B


199
0.00353
P26583
−0.25724
0.007811
Cluster A


200
8.52E−06
P26639
−0.24982
3.12E−05
Cluster A


201
2.88E−26
P26641
−0.75723
7.36E−25
Cluster A


202
9.34E−14
P27635
−0.45446
7.87E−13
Cluster A


203
4.22E−13
P27797
0.382461
3.22E−12
Cluster B


204
3.36E−18
P27824
0.554246
4.21E−17
Cluster B


205
3.71E−14
P29401
−0.44459
3.35E−13
Cluster A


206
4.57E−11
P29692
−0.51322
2.91E−10
Cluster A


207
1.69E−09
P30040
0.385868
9.30E−09
Cluster B


208
6.97E−09
P30041
−0.51789
3.55E−08
Cluster A


209
3.63E−05
P30048
0.418534
0.000119
Cluster B


210
1.10E−22
P30050
−0.60264
1.89E−21
Cluster A


211
6.81E−09
P30084
0.542131
3.48E−08
Cluster B


212
0.003624
P30086
−0.22016
0.007987
Cluster A


213
7.07E−11
P30101
0.338817
4.45E−10
Cluster B


214
2.01E−09
P30533
0.429159
1.09E−08
Cluster B


215
0.000109
P30626
−0.26781
0.000319
Cluster A


216
8.40E−13
P31040
0.920718
6.21E−12
Cluster B


217
2.20E−05
P31939
−0.40174
7.49E−05
Cluster A


218
3.94E−06
P31942
0.227399
1.51E−05
Cluster B


219
3.10E−06
P31943
0.330107
1.20E−05
Cluster B


220
2.00E−21
P31946
−0.69249
3.24E−20
Cluster A


221
2.06E−24
P31947
−1.56597
4.31E−23
Cluster A


222
3.24E−24
P31949
−0.79482
6.48E−23
Cluster A


223
1.24E−21
P32119
−0.79627
2.06E−20
Cluster A


224
3.62E−08
P32969
−0.3943
1.69E−07
Cluster A


225
1.38E−05
P33991
0.241691
4.91E−05
Cluster B


226
2.16E−14
P34897
0.429394
2.00E−13
Cluster B


227
1.07E−06
P34932
−0.23008
4.33E−06
Cluster A


228
6.22E−28
P35232
0.587405
1.99E−26
Cluster B


229
0.000458
P35241
−0.35663
0.001196
Cluster A


230
1.31E−07
P35268
−0.31236
5.85E−07
Cluster A


231
0.001121
P35580
−0.22232
0.002748
Cluster A


232
5.55E−05
P35610
0.530629
0.000174
Cluster B


233
5.75E−07
P35613
0.403417
2.39E−06
Cluster B


234
5.63E−14
P35637
−0.71123
4.94E−13
Cluster A


235
0.002035
P35659
0.246953
0.004682
Cluster B


236
3.16E−10
P36542
0.398698
1.89E−09
Cluster B


237
0.001967
P36551
0.312979
0.004541
Cluster B


238
2.22E−20
P36578
−0.5004
3.37E−19
Cluster A


239
7.20E−05
P36776
0.347853
0.000219
Cluster B


240
2.23E−13
P36957
0.57224
1.78E−12
Cluster B


241
3.31E−05
P37108
0.324531
0.000109
Cluster B


242
2.46E−06
P37802
−0.38857
9.60E−06
Cluster A


243
1.50E−05
P37837
−0.28877
5.33E−05
Cluster A


244
1.99E−10
P38159
0.289836
1.21E−09
Cluster B


245
2.98E−08
P38606
−0.2953
1.43E−07
Cluster A


246
2.16E−12
P38646
0.23637
1.53E−11
Cluster B


247
0.000357
P38919
−0.19906
0.000951
Cluster A


248
3.57E−27
P39019
−0.80608
1.04E−25
Cluster A


249
4.57E−27
P39023
−0.65706
1.27E−25
Cluster A


250
5.33E−08
P39656
0.348306
2.46E−07
Cluster B


251
2.05E−31
P40121
−1.2558
1.09E−29
Cluster A


252
2.99E−06
P40227
−0.38208
1.16E−05
Cluster A


253
5.88E−18
P40429
−0.53509
7.10E−17
Cluster A


254
3.75E−07
P40925
−0.29951
1.58E−06
Cluster A


255
4.12E−22
P40926
0.480023
7.02E−21
Cluster B


256
2.65E−13
P40939
0.421507
2.11E−12
Cluster B


257
0.00032
P41091
−0.44608
0.000858
Cluster A


258
2.63E−05
P41250
−0.25185
8.84E−05
Cluster A


259
3.34E−09
P42677
−0.55661
1.77E−08
Cluster A


260
4.95E−14
P42704
0.392109
4.43E−13
Cluster B


261
0.000342
P42765
0.359394
0.000914
Cluster B


262
2.33E−24
P42766
−0.88902
4.73E−23
Cluster A


263
1.56E−12
P43243
0.402815
1.12E−11
Cluster B


264
0.003746
P43246
0.446104
0.008233
Cluster B


265
3.86E−08
P43304
0.566857
1.80E−07
Cluster B


266
1.51E−05
P43307
0.488912
5.34E−05
Cluster B


267
8.24E−11
P43487
−0.88924
5.12E−10
Cluster A


268
0.000224
P46013
0.234344
0.000617
Cluster B


269
0.001727
P46060
0.431159
0.004038
Cluster B


270
0.000167
P46087
0.257009
0.000472
Cluster B


271
4.32E−26
P46776
−0.67854
1.08E−24
Cluster A


272
1.04E−16
P46777
−0.54192
1.16E−15
Cluster A


273
2.92E−11
P46778
−0.47598
1.90E−10
Cluster A


274
8.21E−21
P46779
−0.51359
1.30E−19
Cluster A


275
4.30E−32
P46781
−0.9441
2.62E−30
Cluster A


276
6.49E−17
P46782
−0.8983
7.41E−16
Cluster A


277
2.49E−07
P46977
0.592938
1.08E−06
Cluster B


278
9.13E−14
P47813
−0.61921
7.78E−13
Cluster A


279
1.43E−24
P47914
−0.98786
3.09E−23
Cluster A


280
0.000178
P47985
0.401999
0.0005
Cluster B


281
0.000145
P48643
−0.20277
0.000418
Cluster A


282
0.000406
P49189
−0.29647
0.001066
Cluster A


283
6.18E−31
P49207
−0.75898
2.82E−29
Cluster A


284
0.000147
P49321
−0.24422
0.000421
Cluster A


285
1.56E−37
P49327
−0.97639
7.99E−35
Cluster A


286
8.69E−06
P49368
−0.22257
3.18E−05
Cluster A


287
1.96E−15
P49411
0.405932
2.00E−14
Cluster B


288
0.002496
P49588
−0.17698
0.005641
Cluster A


289
0.0007
P49736
0.233924
0.00178
Cluster B


290
8.36E−15
P49755
0.921753
7.98E−14
Cluster B


291
3.58E−16
P49773
−0.78305
3.78E−15
Cluster A


292
3.34E−05
P50213
0.295064
0.00011
Cluster B


293
4.81E−13
P50395
−0.48282
3.62E−12
Cluster A


294
7.08E−09
P50402
0.35498
3.59E−08
Cluster B


295
0.001295
P50454
0.377681
0.003132
Cluster B


296
1.75E−16
P50502
−0.67741
1.92E−15
Cluster A


297
0.002327
P50552
0.392631
0.005306
Cluster B


298
2.84E−07
P50990
−0.31324
1.22E−06
Cluster A


299
3.73E−05
P50991
−0.30816
0.000121
Cluster A


300
0.000605
P51148
0.40263
0.001551
Cluster B


301
7.50E−17
P51149
0.519755
8.41E−16
Cluster B


302
0.000621
P51159
0.356667
0.001588
Cluster B


303
2.75E−09
P51572
0.454622
1.47E−08
Cluster B


304
0.00015
P51610
0.301292
0.000429
Cluster B


305
0.000183
P51659
0.512309
0.000512
Cluster B


306
1.79E−09
P51991
0.456262
9.80E−09
Cluster B


307
2.90E−08
P52209
−0.34196
1.40E−07
Cluster A


308
5.64E−12
P52272
0.289145
3.92E−11
Cluster B


309
7.29E−05
P52565
−0.60092
0.000221
Cluster A


310
0.00094
P52566
−0.22612
0.002348
Cluster A


311
1.15E−07
P52907
−0.39994
5.12E−07
Cluster A


312
1.22E−09
P53396
−0.33979
6.81E−09
Cluster A


313
8.31E−05
P53801
0.631089
0.000248
Cluster B


314
7.81E−05
P54136
−0.27424
0.000234
Cluster A


315
8.88E−06
P54819
0.374041
3.23E−05
Cluster B


316
1.83E−12
P55060
−0.44626
1.30E−11
Cluster A


317
0.000366
P55072
−0.17802
0.000973
Cluster A


318
6.50E−15
P55084
0.557281
6.39E−14
Cluster B


319
0.001008
P55265
0.392475
0.002498
Cluster B


320
5.65E−05
P55327
−0.50409
0.000176
Cluster A


321
3.51E−05
P55809
0.491307
0.000115
Cluster B


322
5.47E−09
P55854
−0.42826
2.84E−08
Cluster A


323
3.06E−14
P56381
0.582334
2.80E−13
Cluster B


324
0.001718
P57103
−0.63399
0.004025
Cluster A


325
4.61E−24
P58546
−1.60773
9.07E−23
Cluster A


326
0.000166
P59998
−0.23955
0.00047
Cluster A


327
1.02E−09
P60174
−0.32861
5.74E−09
Cluster A


328
0.001307
P60228
−0.47963
0.003154
Cluster A


329
8.44E−20
P60660
−0.70355
1.21E−18
Cluster A


330
5.21E−09
P60709
−0.2329
2.72E−08
Cluster A


331
1.41E−19
P60842
−0.61683
1.92E−18
Cluster A


332
1.19E−12
P60866
−0.69578
8.72E−12
Cluster A


333
1.45E−06
P61009
0.429563
5.74E−06
Cluster B


334
1.21E−11
P61026
0.425535
8.13E−11
Cluster B


335
1.88E−05
P61081
−0.27211
6.50E−05
Cluster A


336
1.08E−05
P61088
−0.82012
3.90E−05
Cluster A


337
2.51E−28
P61247
−0.65198
8.23E−27
Cluster A


338
1.33E−34
P61254
−0.78173
1.55E−32
Cluster A


339
1.20E−17
P61313
−0.5487
1.42E−16
Cluster A


340
2.25E−21
P61353
−0.57239
3.60E−20
Cluster A


341
9.83E−15
P61604
0.417969
9.32E−14
Cluster B


342
4.87E−15
P61927
−0.61789
4.83E−14
Cluster A


343
1.06E−17
P61956
−0.93005
1.26E−16
Cluster A


344
3.30E−13
P61970
−0.81314
2.54E−12
Cluster A


345
2.24E−33
P61981
−0.89703
1.79E−31
Cluster A


346
7.20E−30
P62081
−0.84394
2.71E−28
Cluster A


347
2.35E−31
P62241
−0.87734
1.20E−29
Cluster A


348
3.18E−23
P62244
−0.92185
5.90E−22
Cluster A


349
3.97E−30
P62249
−1.05765
1.54E−28
Cluster A


350
2.13E−27
P62258
−1.24916
6.34E−26
Cluster A


351
4.17E−08
P62263
−0.38402
1.94E−07
Cluster A


352
1.00E−11
P62266
−0.59355
6.81E−11
Cluster A


353
7.44E−12
P62269
−0.88374
5.09E−11
Cluster A


354
1.32E−06
P62273
−1.55548
5.29E−06
Cluster A


355
2.84E−25
P62277
−0.57524
6.61E−24
Cluster A


356
9.86E−33
P62280
−1.1753
7.01E−31
Cluster A


357
1.61E−20
P62424
−0.48845
2.51E−19
Cluster A


358
6.15E−22
P62701
−0.53683
1.04E−20
Cluster A


359
2.06E−12
P62750
−0.30544
1.46E−11
Cluster A


360
2.11E−32
P62753
−1.22349
1.35E−30
Cluster A


361
2.24E−10
P62805
0.32429
1.35E−09
Cluster B


362
8.13E−11
P62807
0.421736
5.08E−10
Cluster B


363
0.000296
P62820
0.139936
0.000796
Cluster B


364
2.20E−33
P62826
−1.2103
1.79E−31
Cluster A


365
2.84E−25
P62829
−0.70939
6.61E−24
Cluster A


366
3.08E−08
P62834
0.572888
1.46E−07
Cluster B


367
3.57E−18
P62847
−0.92265
4.44E−17
Cluster A


368
2.61E−20
P62851
−0.74325
3.93E−19
Cluster A


369
4.31E−18
P62854
−0.97447
5.31E−17
Cluster A


370
1.71E−20
P62857
−0.81644
2.64E−19
Cluster A


371
0.001284
P62861
−0.34391
0.003113
Cluster A


372
1.45E−08
P62888
−0.36215
7.21E−08
Cluster A


373
8.51E−29
P62899
−0.74303
2.86E−27
Cluster A


374
1.36E−24
P62906
−0.62127
2.99E−23
Cluster A


375
1.79E−09
P62910
−0.37741
9.80E−09
Cluster A


376
6.18E−05
P62913
−0.31267
0.000192
Cluster A


377
1.18E−26
P62917
−0.5499
3.08E−25
Cluster A


378
1.92E−27
P62937
−1.14452
5.86E−26
Cluster A


379
0.001416
P62942
−0.25551
0.003379
Cluster A


380
5.74E−25
P62987
−0.66072
1.31E−23
Cluster A


381
0.004039
P62995
0.244584
0.008862
Cluster B


382
5.78E−32
P63104
−1.00792
3.36E−30
Cluster A


383
2.54E−06
P63162
0.338502
9.90E−06
Cluster B


384
2.67E−08
P63173
−0.50742
1.29E−07
Cluster A


385
1.02E−23
P63220
−2.4649
1.95E−22
Cluster A


386
7.01E−36
P63241
−1.71532
8.96E−34
Cluster A


387
4.34E−06
P63244
−0.3341
1.65E−05
Cluster A


388
0.00262
P67809
−0.20563
0.005889
Cluster A


389
3.64E−07
P67812
0.585149
1.54E−06
Cluster B


390
6.75E−25
P68036
−0.72305
1.52E−23
Cluster A


391
5.48E−08
P68363
−1.7886
2.52E−07
Cluster A


392
6.97E−27
P68371
−1.04751
1.90E−25
Cluster A


393
0.000403
P68400
0.252293
0.001063
Cluster B


394
0.001061
P69849
0.426735
0.002614
Cluster B


395
8.35E−15
P78371
−0.58565
7.98E−14
Cluster A


396
0.00137
P78417
−0.13674
0.003287
Cluster A


397
5.34E−17
P78527
0.284545
6.20E−16
Cluster B


398
0.000511
P80723
0.728923
0.001321
Cluster B


399
9.99E−16
P83731
−0.59037
1.05E−14
Cluster A


400
3.61E−11
P83881
−0.39767
2.32E−10
Cluster A


401
2.04E−24
P84098
−1.28032
4.31E−23
Cluster A


402
6.49E−08
P99999
0.336349
2.97E−07
Cluster B


403
0.000267
Q00059
0.300231
0.000726
Cluster B


404
8.24E−06
Q00325
0.320047
3.04E−05
Cluster B


405
2.30E−08
Q00610
−0.26091
1.11E−07
Cluster A


406
4.15E−07
Q00688
−0.66944
1.74E−06
Cluster A


407
2.87E−15
Q00839
0.453312
2.86E−14
Cluster B


408
0.002542
Q01130
−0.24878
0.005724
Cluster A


409
2.60E−15
Q01469
−0.69946
2.62E−14
Cluster A


410
1.31E−12
Q01518
−0.63043
9.46E−12
Cluster A


411
0.001352
Q01650
0.336574
0.003256
Cluster B


412
0.000124
Q01813
−0.34486
0.000361
Cluster A


413
0.001632
Q01844
0.432603
0.003845
Cluster B


414
0.001832
Q02218
0.322724
0.00426
Cluster B


415
3.55E−20
Q02543
−0.44844
5.27E−19
Cluster A


416
8.71E−05
Q02790
−0.23755
0.000259
Cluster A


417
2.95E−29
Q02878
−0.64358
1.02E−27
Cluster A


418
0.002913
Q02978
0.280501
0.00649
Cluster B


419
2.43E−05
Q03252
0.262842
8.27E−05
Cluster B


420
3.17E−05
Q04837
0.284435
0.000105
Cluster B


421
1.15E−36
Q06830
−1.96378
2.44E−34
Cluster A


422
9.35E−14
Q07020
−0.40355
7.87E−13
Cluster A


423
0.000203
Q07021
0.910802
0.000563
Cluster B


424
4.39E−06
Q07666
0.429952
1.66E−05
Cluster B


425
5.50E−05
Q07955
0.241926
0.000173
Cluster B


426
1.53E−14
Q08211
0.345416
1.43E−13
Cluster B


427
0.001605
Q08380
0.341092
0.003791
Cluster B


428
0.002762
Q08722
0.479918
0.006187
Cluster B


429
0.002531
Q08945
0.316821
0.00571
Cluster B


430
0.00156
Q09028
0.203954
0.003701
Cluster B


431
1.40E−06
Q09666
−0.3841
5.59E−06
Cluster A


432
3.00E−08
Q12906
0.301276
1.43E−07
Cluster B


433
3.08E−05
Q12931
0.151581
0.000103
Cluster B


434
4.82E−05
Q13011
0.290689
0.000155
Cluster B


435
1.93E−05
Q13151
0.287588
6.64E−05
Cluster B


436
0.00417
Q13242
0.478419
0.009054
Cluster B


437
3.61E−07
Q13247
0.298732
1.53E−06
Cluster B


438
1.95E−06
Q13263
0.285788
7.68E−06
Cluster B


439
0.001934
Q13347
−0.26185
0.004489
Cluster A


440
2.62E−09
Q13404
−0.46985
1.41E−08
Cluster A


441
0.000194
Q13409
−0.25841
0.000539
Cluster A


442
7.22E−10
Q13428
0.433707
4.08E−09
Cluster B


443
8.33E−05
Q13435
0.215308
0.000248
Cluster B


444
2.96E−07
Q13442
−0.52039
1.27E−06
Cluster A


445
0.000937
Q13451
0.191705
0.002344
Cluster B


446
1.85E−07
Q13637
0.616847
8.09E−07
Cluster B


447
2.11E−05
Q13838
−0.35218
7.25E−05
Cluster A


448
0.000645
Q13951
−0.27726
0.001642
Cluster A


449
9.62E−14
Q14019
−0.68323
8.04E−13
Cluster A


450
8.44E−34
Q14247
−1.06257
7.71E−32
Cluster A


451
2.80E−05
Q14376
−0.33939
9.38E−05
Cluster A


452
6.52E−13
Q14444
−0.44996
4.85E−12
Cluster A


453
6.41E−05
Q14566
0.312227
0.000197
Cluster B


454
1.40E−07
Q14697
0.22257
6.19E−07
Cluster B


455
4.26E−07
Q14956
0.629553
1.78E−06
Cluster B


456
1.28E−11
Q14978
0.360392
8.59E−11
Cluster B


457
9.27E−06
Q14980
0.252161
3.37E−05
Cluster B


458
0.00076
Q15008
−0.17083
0.001929
Cluster A


459
6.09E−20
Q15056
−0.6192
8.96E−19
Cluster A


460
0.004127
Q15061
0.394754
0.008993
Cluster B


461
6.75E−17
Q15084
0.446317
7.64E−16
Cluster B


462
0.000172
Q15149
0.212718
0.000486
Cluster B


463
7.77E−05
Q15181
−0.37909
0.000234
Cluster A


464
2.78E−11
Q15233
0.35504
1.83E−10
Cluster B


465
1.64E−13
Q15293
0.466244
1.33E−12
Cluster B


466
5.44E−05
Q15363
0.582913
0.000171
Cluster B


467
9.02E−08
Q15366
−0.57619
4.08E−07
Cluster A


468
3.17E−09
Q15370
−0.39154
1.69E−08
Cluster A


469
1.83E−05
Q15382
−0.72495
6.34E−05
Cluster A


470
1.48E−08
Q15392
0.457466
7.29E−08
Cluster B


471
3.15E−06
Q15393
0.297394
1.21E−05
Cluster B


472
0.000522
Q15417
−0.51015
0.001345
Cluster A


473
3.63E−13
Q15424
0.400138
2.78E−12
Cluster B


474
7.97E−06
Q15459
0.279085
2.95E−05
Cluster B


475
5.57E−19
Q15691
−0.73942
7.43E−18
Cluster A


476
1.17E−12
Q15696
−0.82614
8.59E−12
Cluster A


477
6.81E−05
Q15717
0.227067
0.000208
Cluster B


478
0.000338
Q15758
0.290283
0.000906
Cluster B


479
1.71E−13
Q15907
0.34981
1.38E−12
Cluster B


480
6.50E−05
Q16181
−0.31381
0.000199
Cluster A


481
0.002789
Q16543
−0.28875
0.006235
Cluster A


482
0.002045
Q16629
0.321038
0.004695
Cluster B


483
4.03E−09
Q16778
0.376063
2.11E−08
Cluster B


484
4.66E−06
Q16836
0.320968
1.76E−05
Cluster B


485
5.78E−10
Q16891
0.382721
3.35E−09
Cluster B


486
2.04E−06
Q1KMD3
0.457295
8.02E−06
Cluster B


487
0.000145
Q2TAY7
0.49664
0.000418
Cluster B


488
4.39E−06
Q3ZCQ8
0.417587
1.66E−05
Cluster B


489
0.002826
Q5SSJ5
0.234401
0.006307
Cluster B


490
0.000773
Q5T3I0
0.455183
0.001958
Cluster B


491
1.87E−37
Q5VTE0
−1.81353
7.99E−35
Cluster A


492
0.000138
Q5XKP0
0.461118
0.000401
Cluster B


493
2.23E−09
Q71DI3
0.369699
1.21E−08
Cluster B


494
2.95E−17
Q7KZF4
−0.44767
3.46E−16
Cluster A


495
0.001585
Q86UE4
0.260247
0.003753
Cluster B


496
0.001963
Q86XP3
0.43685
0.004541
Cluster B


497
1.20E−05
Q8N5M9
0.625843
4.33E−05
Cluster B


498
2.52E−05
Q8N7Z2
0.75849
8.53E−05
Cluster B


499
1.03E−09
Q8NBS9
0.452789
5.74E−09
Cluster B


500
5.31E−05
Q8NBX0
0.509982
0.000169
Cluster B


501
0.001807
Q8NC51
−0.23208
0.00421
Cluster A


502
7.14E−10
Q8TCJ2
0.682638
4.06E−09
Cluster B


503
1.46E−07
Q8TCT9
0.587904
6.42E−07
Cluster B


504
0.000107
Q8TDN6
0.625323
0.000315
Cluster B


505
0.000176
Q8WU90
−0.56266
0.000494
Cluster A


506
3.68E−05
Q8WW12
−0.404
0.00012
Cluster A


507
1.31E−05
Q8WXH0
0.557275
4.68E−05
Cluster B


508
0.000219
Q8WYA6
0.496705
0.000604
Cluster B


509
1.78E−05
Q8WYQ5
−1.25789
6.20E−05
Cluster A


510
0.000172
Q92522
0.291841
0.000486
Cluster B


511
6.09E−07
Q92598
−0.41817
2.52E−06
Cluster A


512
2.79E−11
Q92734
−0.56871
1.83E−10
Cluster A


513
5.11E−05
Q96AG4
0.242719
0.000163
Cluster B


514
3.69E−09
Q96C19
−0.3718
1.94E−08
Cluster A


515
5.71E−06
Q96I99
0.388862
2.15E−05
Cluster B


516
0.000228
Q96IX5
0.299308
0.000625
Cluster B


517
0.00238
Q96SB4
0.162596
0.005416
Cluster B


518
1.08E−22
Q99497
−0.62536
1.89E−21
Cluster A


519
7.05E−14
Q99536
−0.57329
6.10E−13
Cluster A


520
6.20E−05
Q99613
−0.37232
0.000192
Cluster A


521
9.36E−20
Q99623
0.434747
1.33E−18
Cluster B


522
5.92E−06
Q99714
0.318625
2.22E−05
Cluster B


523
7.43E−08
Q99729
0.415402
3.38E−07
Cluster B


524
0.004616
Q99757
0.313102
0.009973
Cluster B


525
0.000393
Q99829
−0.19002
0.001038
Cluster A


526
0.004116
Q99832
−0.11634
0.008983
Cluster A


527
1.75E−05
Q99848
0.624443
6.12E−05
Cluster B


528
8.48E−06
Q99873
−0.34667
3.12E−05
Cluster A


529
0.000866
Q99878
0.206655
0.002171
Cluster B


530
0.000379
Q99986
1.57746
0.001005
Cluster B


531
1.47E−31
Q9BQE3
−1.13268
8.19E−30
Cluster A


532
1.22E−08
Q9BQG0
0.426151
6.07E−08
Cluster B


533
2.84E−11
Q9BVC6
0.455681
1.85E−10
Cluster B


534
0.002663
Q9BVP2
0.59797
0.005975
Cluster B


535
3.14E−08
Q9BXS5
−0.46504
1.49E−07
Cluster A


536
0.00225
Q9BY44
0.589738
0.005149
Cluster B


537
0.000135
Q9BZH6
0.44511
0.000392
Cluster B


538
0.001396
Q9C0B1
−0.25084
0.003336
Cluster A


539
0.000108
Q9GZT3
0.474864
0.000316
Cluster B


540
3.41E−07
Q9GZZ1
−0.45864
1.46E−06
Cluster A


541
0.001676
Q9H0A0
0.628824
0.003941
Cluster B


542
2.17E−05
Q9H1E3
−0.56292
7.41E−05
Cluster A


543
2.10E−24
Q9H299
−1.08255
4.34E−23
Cluster A


544
6.84E−05
Q9H2W6
0.66679
0.000208
Cluster B


545
1.65E−10
Q9H3K6
−0.40339
1.01E−09
Cluster A


546
0.000966
Q9H3N1
0.398583
0.002404
Cluster B


547
0.000486
Q9H444
0.407015
0.00126
Cluster B


548
4.36E−05
Q9H9B4
0.57824
0.000141
Cluster B


549
9.96E−07
Q9HAV0
0.434474
4.04E−06
Cluster B


550
0.000147
Q9HAV7
0.238317
0.000422
Cluster B


551
1.43E−07
Q9HB71
−0.58267
6.29E−07
Cluster A


552
0.00027
Q9HC38
0.239842
0.000732
Cluster B


553
0.002439
Q9NQP4
0.676201
0.00554
Cluster B


554
1.31E−12
Q9NR30
0.333618
9.46E−12
Cluster B


555
1.96E−08
Q9NR45
−1.9634
9.59E−08
Cluster A


556
0.001092
Q9NRV9
−0.15657
0.002681
Cluster A


557
0.001773
Q9NRX4
−0.60578
0.004138
Cluster A


558
9.88E−08
Q9NSD9
−0.46788
4.44E−07
Cluster A


559
8.40E−10
Q9NTK5
−0.32783
4.73E−09
Cluster A


560
5.35E−05
Q9NVI7
0.230967
0.000169
Cluster B


561
6.91E−06
Q9NVP1
0.472615
2.58E−05
Cluster B


562
3.80E−11
Q9NX63
0.41873
2.43E−10
Cluster B


563
0.001504
Q9NYF8
0.343114
0.003582
Cluster B


564
0.003628
Q9NZR2
0.382122
0.007987
Cluster B


565
5.23E−10
Q9UBM7
0.386689
3.08E−09
Cluster B


566
7.50E−11
Q9UHD8
−0.55872
4.70E−10
Cluster A


567
8.96E−08
Q9UII2
0.334059
4.06E−07
Cluster B


568
7.92E−05
Q9UJ41
−0.57763
0.000237
Cluster A


569
3.25E−07
Q9UJZ1
0.388886
1.39E−06
Cluster B


570
2.50E−05
Q9UK45
−0.94633
8.47E−05
Cluster A


571
6.04E−05
Q9UK76
−0.6992
0.000188
Cluster A


572
1.05E−14
Q9UKM9
0.444614
9.83E−14
Cluster B


573
0.004138
Q9UKV3
0.331258
0.009002
Cluster B


574
1.30E−06
Q9UKY7
−0.59835
5.22E−06
Cluster A


575
0.000638
Q9ULV4
−0.28439
0.001628
Cluster A


576
0.001995
Q9ULZ3
0.260849
0.004596
Cluster B


577
6.83E−05
Q9UM00
0.35684
0.000208
Cluster B


578
4.45E−05
Q9UMS4
0.374463
0.000144
Cluster B


579
0.002089
Q9UMX5
0.332133
0.004789
Cluster B


580
0.003481
Q9UQ35
0.211948
0.007729
Cluster B


581
7.16E−24
Q9UQ80
−0.86166
1.39E−22
Cluster A


582
7.24E−07
Q9Y237
−0.74523
2.97E−06
Cluster A


583
5.74E−10
Q9Y261
−2.39187
3.34E−09
Cluster A


584
1.48E−16
Q9Y266
−0.62088
1.63E−15
Cluster A


585
2.23E−10
Q9Y277
0.559255
1.35E−09
Cluster B


586
3.87E−12
Q9Y2S6
−0.76764
2.72E−11
Cluster A


587
4.71E−05
Q9Y2W1
−0.27429
0.000152
Cluster A


588
0.003295
Q9Y2X3
0.259104
0.007329
Cluster B


589
0.000108
Q9Y3B4
0.616503
0.000317
Cluster B


590
4.99E−05
Q9Y4G6
−1.13097
0.00016
Cluster A


591
7.81E−06
Q9Y4L1
0.249345
2.90E−05
Cluster B


592
0.000787
Q9Y5B9
0.185761
0.001986
Cluster B
















TABLE 6







Protein set enrichment analysis between melanoma sub-populations


















numberOf
fractionOfDB
Cond1med
Cond2med





GO_term
pVal
Matches
Observed
int
int
qVal
dif



















1
symporter activity
2.76E−09
3
0.0625
0.0705
0.3146
8.14E−09
−0.2441


2
primary cilium
1.34E−09
5
0.1190
0.0025
0.4329
4.04E−09
−0.4303


3
core promoter binding
9.06E−15
7
0.1346
−0.0026
0.2972
3.97E−14
0.2998


4
placenta development
3.49E−07
5
0.0820
−0.1133
−0.4332
8.48E−07
0.3199


5
extrinsic to internal side of plasma membrane
7.00E−17
8
0.1600
0.1004
−0.2319
3.39E−16
0.3324


6
positive regulation of neuron differentiation
5.39E−09
9
0.0600
0.0397
−0.2022
1.57E−08
0.2418


7
MAPK cascade
2.62E−11
12
0.0845
0.1048
−0.1794
8.99E−11
0.2842


8
activation of MAPK activity
7.62E−06
13
0.1040
0.0473
−0.2264
1.65E−05
0.2737


9
positive regulation of epithelial cell proliferation
5.10E−08
6
0.0714
0.3063
−0.1582
1.34E−07
0.4645


10
Rho GTPase activator activity
1.33E−45
7
0.2333
−0.0459
−1.1085
1.87E−44
1.0626


11
cytoskeleton organization
2.16E−17
20
0.1515
0.0852
−0.3638
1.07E−16
0.4491


12
positive regulation of Rho GTPase activity
6.73E−34
7
0.1321
0.0019
−1.0512
6.72E−33
1.0531


13
phagocytic vesicle membrane
2.28E−16
13
0.1111
−0.0679
0.2775
1.07E−15
−0.3454


14
intermediate filament
1.85E−09
7
0.0446
0.3323
0.6053
5.55E−09
−0.2730


15
chloride transport
7.93E−09
5
0.0735
0.0165
−0.3134
2.26E−08
0.3299


16
transmembrane transporter activity
4.13E−63
12
0.1846
0.0942
0.4025
8.79E−62
0.3083


17
odontogenesis of dentin-containing tooth
4.94E−06
7
0.0588
0.0441
0.2580
1.08E−05
0.2139


18
endonuclease activity
1.32E−16
10
0.1786
0.0616
−0.2292
6.26E−16
0.2908


19
peroxidase activity
1.18E−17
8
0.2000
0.2018
−0.0631
6.02E−17
0.2649


20
hydrogen peroxide catabolic process
4.21E−25
9
0.4500
0.1843
−0.1840
3.20E−24
0.3683


21
acid-amino acid ligase activity
2.91E−36
7
0.0560
0.1301
−0.5181
3.14E−35
0.6482


22
triglyceride biosynthetic process
6.55E−35
6
0.1132
0.2813
−0.2853
6.66E−34
0.5666


23
long-chain fatty-acyl-CoA biosynthetic process
6.55E−35
6
0.3529
0.2813
−0.2853
6.66E−34
0.5666


24
cellular lipid metabolic process
2.27E−12
34
0.2048
0.1012
0.2368
8.51E−12
−0.1356


25
skeletal system development
1.87E−20
14
0.0915
0.3169
−0.0447
1.09E−19
0.3617


26
extracellular matrix disassembly
4.74E−05
22
0.1930
−0.0140
0.1576
9.46E−05
−0.1715


27
adipose tissue development
3.63E−05
6
0.1622
0.0206
0.1627
7.34E−05
−0.1420


28
negative regulation of cell growth
2.60E−05
16
0.0994
0.0812
−0.0636
5.32E−05
0.1447


29
axonogenesis
1.41E−10
17
0.1360
0.0515
−0.0893
4.57E−10
0.1409


30
negative regulation of microtubule polymerization
2.02E−29
6
0.4286
0.0403
−0.2787
1.78E−28
0.3190


31
positive regulation of cellular component movement
3.26E−77
4
0.2667
0.0403
0.4593
9.88E−76
0.4996


32
glutathione metabolic process
5.29E−09
10
0.1449
−0.0257
−0.3265
1.54E−08
0.3007


33
pyridoxal phosphate binding
1.63E−06
11
0.0679
0.1065
0.1870
3.77E−06
−0.0805


34
ubiquitin-ubiquitin ligase activity
2.77E−05
4
0.3333
0.0054
0.2534
5.65E−05
−0.2481


35
integral to endoplasmic reticulum membrane
2.34E−05
10
0.1064
−0.0036
0.2176
4.80E−05
0.2212


36
response to oxidative stress
5.26E−10
20
0.1136
0.1467
−0.0623
1.63E−09
0.2090


37
learning or memory
2.92E−24
5
0.0694
−0.0350
−0.5563
2.16E−23
0.5213


38
ATP-dependent protein binding
3.55E−06
3
0.2727
0.0082
−0.1710
7.92E−06
0.1793


39
negative regulation of sequence-specific DNA binding transcription
3.44E−16
8
0.1270
0.0093
−0.2079
1.59E−15
0.2172



factor activity


40
negative regulation of T cell receptor signaling pathway
1.93E−06
4
0.1905
0.2498
−0.1641
4.44E−06
0.4139


41
cytoskeletal protein binding
2.09E−07
15
0.1485
−0.0332
−0.3650
5.22E−07
0.3318


42
extrinsic to membrane
2.92E−13
13
0.1912
−0.0156
−0.2922
1.18E−12
0.2766


43
double-stranded RNA binding
6.74E−12
24
0.3000
0.0125
0.0008
2.43E−11
0.0117


44
single-stranded RNA binding
4.12E−15
5
0.1190
0.0031
0.2632
1.83E−14
−0.2601


45
helicase activity
7.06E−11
15
0.1095
0.0705
−0.3055
2.32E−10
0.3760


46
response to virus
1.17E−82
31
0.2039
0.0444
−0.2977
4.11E−81
0.3421


47
actin filament organization
2.04E−07
8
0.0899
−0.1308
−0.8892
5.13E−07
0.7584


48
multicellular organism growth
1.18E−11
6
0.0462
0.1847
0.6057
4.19E−11
−0.4210


49
cilium
1.04E−14
23
0.1494
0.1024
0.2633
4.49E−14
0.3657


50
aspartic-type endopeptidase activity
2.04E−08
4
0.0488
0.0474
0.3875
5.62E−08
−0.3400


51
intermediate filament cytoskeleton
3.32E−05
13
0.1238
0.0687
0.2199
6.73E−05
−0.1512


52
glucose homeostasis
2.25E−77
6
0.0451
0.1080
−0.7183
6.94E−76
0.8263


53
anion transport
1.84E−08
3
0.0714
−0.0281
0.1523
5.09E−08
0.1804


54
voltage-gated anion channel activity
1.84E−08
3
0.2000
−0.0281
0.1523
5.09E−08
−0.1804


55
PML body
3.61E−14
13
0.1215
0.1276
0.1615
1.51E−13
0.2891


56
apoptotic signaling pathway
2.39E−28
19
0.1557
0.1409
−0.4552
2.04E−27
0.5961


57
positive regulation of apoptotic signaling pathway
2.51E−14
4
0.0784
0.1245
−0.2836
1.06E−13
0.4081


58
protein export from nucleus
3.48E−64
14
0.3111
0.0809
−0.2336
7.50E−63
0.3146


59
embryo implantation
2.87E−06
4
0.0435
0.0101
−0.1593
6.49E−06
0.1694


60
ATP-dependent DNA helicase activity
3.91E−14
14
0.2692
−0.0052
0.1367
1.63E−13
−0.1419


61
DNA duplex unwinding
3.27E−07
18
0.2169
0.0299
0.1222
7.98E−07
−0.0923


62
cellular protein modification process
3.25E−19
20
0.1274
0.0780
−0.0953
1.80E−18
0.1734


63
antigen processing and presentation
2.30E−20
7
0.1129
0.0029
0.4295
1.33E−19
−0.4265


64
proteolysis involved in cellular protein catabolic process
8.16E−06
5
0.1190
−0.1262
0.0621
1.76E−05
0.1883


65
antigen processing and presentation of peptide antigen via MHC class I
3.02E−08
57
0.3202
−0.0286
0.0329
8.12E−08
0.0615


66
peptide antigen binding
2.20E−08
5
0.0420
−0.0958
0.1983
5.99E−08
0.2941


67
protein peptidyl-prolyl isomerization
1.67E−20
16
0.1720
0.0082
0.2224
9.90E−20
0.2306


68
peptidyl-prolyl cis-trans isomerase activity
1.67E−20
16
0.1720
0.0082
−0.2224
9.90E−20
0.2306


69
nucleoside diphosphate kinase activity
3.61E−13
5
0.1111
0.2514
−0.2527
1.44E−12
0.5041


70
nucleoside diphosphate phosphorylation
3.61E−13
5
0.1136
0.2514
−0.2527
1.44E−12
0.5041


71
GTP biosynthetic process
3.52E−14
3
0.0833
0.2785
0.2801
1.47E−13
0.5585


72
UTP biosynthetic process
6.81E−11
4
0.1081
0.1666
−0.2017
2.25E−10
0.3682


73
CTP biosynthetic process
1.41E−12
4
0.1081
0.1841
−0.2017
5.42E−12
0.3858


74
tRNA binding
2.68E−07
15
0.3000
−0.0021
−0.1247
6.63E−07
0.1227


75
endosome to lysosome transport
6.22E−12
5
0.1471
0.1312
0.4981
2.25E−11
−0.3669


76
skeletal muscle cell differentiation
2.30E−06
4
0.0625
0.1282
0.4153
5.21E−06
0.2871


77
cellular response to organic cyclic compound
2.24E−08
5
0.0746
−0.0754
−0.3551
6.08E−08
0.2796


78
spindle assembly
1.88E−31
4
0.0548
0.1006
−0.9763
1.72E−30
1.0769


79
regulation of heart rate by cardiac conduction
6.04E−40
4
0.1667
0.3252
−0.6161
7.21E−39
0.9413


80
negative regulation of retinoic acid receptor signaling pathway
5.52E−10
3
0.0652
−0.2428
0.1134
1.71E−09
−0.3563


81
response to antibiotic
2.32E−05
3
0.0612
0.0337
0.1976
4.77E−05
−0.1640


82
hippo signaling cascade
2.57E−64
6
0.1667
0.2426
−0.5479
5.70E−63
0.7905


83
retina homeostasis
2.21E−20
9
0.2250
0.0474
−0.2762
1.28E−19
0.3236


84
cell cortex
1.90E−12
26
0.1436
0.1600
−0.0661
7.17E−12
0.2262


85
blood microparticle
4.94E−52
22
0.1358
0.0237
−0.3317
8.11E−51
0.3553


86
lipid particle organization
1.40E−71
4
0.3333
0.2428
−0.8938
3.77E−70
1.1365


87
viral infectious cycle
0
89
0.7542
0.0755
−0.5257
0
0.6012


88
membrane organization
1.16E−51
58
0.3946
0.0756
−0.1968
1.88E−50
0.2724


89
ruffle
1.86E−62
32
0.2270
0.0847
−0.2427
3.90E−61
0.3273


90
neuron differentiation
1.09E−05
9
0.0938
0.1505
0.0565
2.30E−05
0.0940


91
nucleosome
1.45E−68
18
0.2169
0.0272
0.3488
3.64E−67
−0.3216


92
nucleosome assembly
5.01E−45
33
0.1823
0.0206
0.2396
6.75E−44
−0.2190


93
nuclear-transcribed mRNA catabolic process, deadenylation-dependent
7.76E−24
20
0.3636
0.0383
−0.2885
5.66E−23
0.3268



decay


94
nuclear-transcribed mRNA poly(A) tail shortening
2.24E−35
9
0.3000
0.0401
−0.3240
2.35E−34
0.3641


95
regulation of translation
6.57E−53
19
0.2568
0.0870
−0.3151
1.11E−51
0.4020


96
estrogen receptor binding
1.15E−14
8
0.2759
0.0776
0.3261
4.96E−14
−0.2486


97
gene silencing by RNA
2.66E−10
9
0.2727
0.1104
−0.2671
8.35E−10
0.3775


98
negative regulation of intracellular estrogen receptor signaling pathway
3.15E−20
4
0.3333
0.1520
0.5860
1.81E−19
−0.4340


99
negative regulation of catalytic activity
1.78E−14
19
0.1397
0.0388
−0.1225
7.60E−14
0.1613


100
DNA binding, bending
2.22E−08
12
0.2034
−0.0230
−0.1468
6.05E−08
0.1238


101
regulation of cell proliferation
2.17E−06
21
0.1214
0.0683
0.2382
4.96E−06
−0.1699


102
autophagic vacuole assembly
9.32E−07
5
0.0877
0.0031
0.2057
2.19E−06
−0.2026


103
synaptic vesicle
8.40E−07
9
0.0581
0.0120
0.2800
1.98E−06
−0.2681


104
calcium-dependent protein binding
5.47E−12
19
0.2235
−0.0247
−0.1651
1.99E−11
0.1404


105
establishment of cell polarity
 2.46E−101
4
0.0784
0.3553
−0.9167
1.38E−99
1.2720


106
single fertilization
2.02E−15
5
0.0962
0.1055
−0.3123
9.03E−15
0.4178


107
NADP binding
1.94E−36
13
0.1625
0.1179
−0.3299
2.12E−35
0.4477


108
histone deacetylase activity
6.89E−06
5
0.1064
−0.0406
0.1368
1.49E−05
−0.1774


109
cytoplasmic microtubule
5.79E−82
11
0.1310
0.1127
−0.6804
1.98E−80
0.7931


110
histone deacetylation
1.29E−25
7
0.1186
0.0518
0.3953
9.95E−25
−0.3436


111
ubiquitin binding
8.62E−07
13
0.2500
0.0313
−0.2052
2.03E−06
0.2365


112
beta-tubulin binding
1.57E−10
9
0.1800
0.0572
0.3093
5.04E−10
−0.2521


113
Rab GTPase activator activity
1.46E−19
4
0.0303
−0.0263
−0.4140
8.21E−19
0.3877


114
positive regulation of Rab GTPase activity
1.46E−19
4
0.0303
−0.0263
−0.4140
8.21E−19
0.3877


115
methyltransferase activity
3.45E−19
12
0.0645
0.0690
−0.2851
1.91E−18
0.3541


116
regulation of growth
3.09E−07
12
0.2308
0.0290
−0.1444
7.61E−07
0.1734


117
negative regulation of viral genome replication
3.83E−08
9
0.2143
0.0102
0.2127
1.02E−07
−0.2025


118
cytosolic small ribosomal subunit
0
33
0.8049
0.0993
−0.6353
0
0.7347


119
chromatin
1.24E−06
24
0.2182
0.0045
−0.0739
2.88E−06
0.0784


120
cysteine-type endopeptidase activity
1.99E−06
10
0.0926
−0.0747
0.1546
4.57E−06
−0.2293


121
regulation of translational fidelity
2.64E−08
6
0.2727
0.0441
−0.1194
7.13E−08
0.1635


122
nuclear pore
1.33E−32
30
0.2655
0.1103
−0.2307
1.26E−31
0.3410


123
mRNA transport
2.18E−06
21
0.3000
0.0471
0.1606
4.96E−06
0.1134


124
microtubule cytoskeleton
1.54E−24
30
0.1563
0.1194
−0.1106
1.16E−23
0.2300


125
negative regulation of Wnt receptor signaling pathway
1.77E−07
4
0.0656
0.0312
−0.2562
4.49E−07
0.2875


126
post-embryonic development
1.95E−10
12
0.0774
0.1640
0.4913
6.23E−10
−0.3273


127
regulation of alternative nuclear mRNA splicing, via spliceosome
3.71E−08
12
0.4000
0.0220
0.1497
9.86E−08
0.1278


128
antioxidant activity
1.03E−12
4
0.1905
0.1147
−0.2135
4.01E−12
0.3282


129
mRNA binding
7.48E−56
36
0.3333
0.0451
−0.1553
1.36E−54
0.2004


130
sequence-specific DNA binding RNA polymerase II transcription factor
9.11E−34
3
0.0288
0.2147
0.8005
8.97E−33
−0.5858



activity


131
purine base metabolic process
4.56E−11
17
0.4722
0.0051
−0.1525
1.52E−10
0.1576


132
sodium:potassium-exchanging ATPase complex
3.24E−11
3
0.1154
0.0680
0.5054
1.10E−10
−0.4373


133
apical part of cell
2.53E−09
14
0.0946
0.0169
−0.1250
7.49E−09
0.1420


134
microtubule organizing center
3.38E−07
27
0.1765
0.0101
−0.0960
8.24E−07
0.1062


135
negative regulation of NF-kappaB transcription factor activity
6.85E−06
8
0.1039
−0.0904
0.0420
1.49E−05
−0.1324


136
tumor necrosis factor-mediated signaling pathway
4.27E−05
3
0.0667
−0.0463
0.1898
8.56E−05
−0.2362


137
spliceosomal complex
4.85E−26
54
0.4655
0.0109
0.1803
3.78E−25
−0.1694


138
structural constituent of cytoskeleton
1.17E−70
30
0.1935
0.0011
−0.3164
3.01E−69
0.3175


139
microtubule-based process
 1.19E−115
12
0.1188
0.0609
−0.8660
 7.48E−114
0.9269


140
protein polymerization
 1.20E−139
8
0.1702
0.0704
−1.0025
 9.42E−138
1.0729


141
rRNA processing
 4.27E−184
45
0.3846
0.0652
−0.4179
 4.48E−182
0.4830


142
ribonucleoprotein complex
 2.02E−156
66
0.4314
0.0591
−0.1540
 1.99E−154
0.2132


143
dendritic spine
1.92E−08
15
0.1200
0.1852
−0.0085
5.29E−08
0.1937


144
somitogenesis
1.43E−08
5
0.0676
0.0916
0.3582
4.01E−08
−0.2666


145
glycolysis
 1.13E−306
22
0.1667
0.1840
−0.4889
 1.62E−304
0.6729


146
response to ischemia
4.89E−05
3
0.0882
−0.0610
0.2024
9.73E−05
−0.2634


147
RNA processing
7.99E−24
30
0.2290
−0.0006
0.1877
5.80E−23
−0.1883


148
regulation of cell morphogenesis
1.27E−52
5
0.2632
0.2686
−0.7196
2.10E−51
0.9882


149
transcription regulatory region sequence-specific DNA binding
9.40E−09
4
0.0482
0.0375
0.3156
2.67E−08
−0.2780


150
translation initiation factor activity
8.02E−41
34
0.2267
−0.0037
0.2885
9.80E−40
0.2848


151
immune system process
2.17E−08
9
0.2093
0.1911
−0.1314
5.97E−08
0.3224


152
negative regulation of type I interferon production
1.22E−23
5
0.1429
0.0887
−0.4462
8.81E−23
0.5349


153
cellular response to interferon-gamma
2.00E−71
4
0.1379
0.1112
−0.3624
5.26E−70
0.4737


154
cellular response to interleukin-1
3.45E−09
4
0.0909
−0.0024
0.2461
1.01E−08
−0.2485


155
negative regulation of protein serine/threonine kinase activity
3.37E−55
6
0.1935
0.0660
−0.3827
5.96E−54
0.4487


156
spindle pole
9.18E−07
26
0.1857
0.0174
0.2370
2.16E−06
−0.2196


157
cerebral cortex development
3.88E−32
11
0.1375
0.1873
−0.3623
3.60E−31
0.5496


158
spindle
3.96E−09
35
0.2397
0.0818
−0.0456
1.16E−08
0.1274


159
proton-transporting V-type ATPase, V1 domain
1.62E−12
4
0.2500
0.1335
−0.1283
6.17E−12
0.2617


160
hydrogen ion transporting ATP synthase activity, rotational mechanism
3.79E−65
8
0.3077
0.1019
0.4455
8.53E−64
−0.3436


161
proton-transporting ATPase activity, rotational mechanism
7.99E−07
12
0.4000
0.1049
0.2277
1.89E−06
−0.1228


162
Rho GTPase binding
3.68E−08
11
0.2075
−0.0131
0.1994
9.81E−08
0.1863


163
DNA-directed DNA polymerase activity
4.94E−05
6
0.0674
−0.0117
0.3337
9.81E−05
−0.3454


164
DNA-dependent DNA replication
8.35E−08
8
0.0870
0.0605
0.3330
2.16E−07
−0.2725


165
melanosome
9.96E−10
73
0.7157
0.0963
0.0679
3.02E−09
0.0283


166
bone mineralization
1.57E−07
3
0.0652
−0.0597
0.3057
3.98E−07
−0.3654


167
cytoplasmic vesicle membrane
1.17E−92
30
0.2521
0.1624
−0.3644
5.76E−91
0.5268


168
amino acid transport
4.65E−08
5
0.1282
−0.1159
0.1735
1.23E−07
−0.2894


169
regulation of release of sequestered calcium ion into cytosol by
5.57E−07
3
0.2143
0.0933
−0.0929
1.33E−06
0.1862



sarcoplasmic reticulum


170
sarcoplasmic reticulum membrane
4.37E−11
7
0.2333
0.0937
0.3509
1.46E−10
−0.2572


171
liver development
5.72E−13
13
0.0813
0.0131
0.2189
2.25E−12
−0.2059


172
RNA polymerase II distal enhancer sequence-specific DNA binding
4.38E−06
14
0.3111
−0.0408
0.0626
9.67E−06
−0.1034


173
neural crest cell migration
1.77E−92
4
0.0702
0.3660
−0.8998
8.44E−91
1.2658


174
developmental growth
1.98E−12
4
0.0769
0.0295
−0.4418
7.45E−12
0.4713


175
ruffle membrane
1.09E−27
23
0.2473
0.0407
−0.3348
9.12E−27
0.3755


176
protein self-association
1.12E−14
9
0.1800
0.1385
0.5579
4.82E−14
−0.4194


177
regulation of cell shape
3.37E−13
23
0.1250
0.0178
−0.2560
1.36E−12
0.2738


178
DNA catabolic process, endonucleolytic
5.09E−13
9
0.1011
0.0426
−0.1945
2.00E−12
0.2370


179
glucose metabolic process
 3.84E−115
50
0.3311
0.1273
−0.2688
 2.33E−113
0.3961


180
postsynaptic density
3.50E−29
13
0.0677
0.0640
−0.3200
3.05E−28
0.3841


181
p53 binding
1.91E−10
8
0.1290
−0.0494
0.2327
6.11E−10
−0.2822


182
cellular response to hydrogen peroxide
6.94E−08
8
0.1250
0.0778
−0.1775
1.81E−07
0.2553


183
recycling endosome
4.27E−16
12
0.1463
0.0088
0.3471
1.96E−15
−0.3382


184
response to toxin
2.05E−07
10
0.0901
0.0741
−0.0994
5.15E−07
0.1734


185
response to cytokine stimulus
3.45E−08
5
0.0556
0.0000
0.2740
9.22E−08
−0.2740


186
chloride channel activity
2.42E−10
6
0.0968
0.0219
−0.3337
7.64E−10
0.3556


187
ATPase activity, coupled
4.34E−14
7
0.2500
0.0624
−0.3040
1.79E−13
0.3664


188
chaperone mediated protein folding requiring cofactor
1.70E−38
4
0.1212
0.0926
−0.4196
1.94E−37
0.5122


189
G2/M transition of mitotic cell cycle
2.23E−99
48
0.3179
0.0944
−0.2645
1.21E−97
0.3589


190
regulation of ion transmembrane transport
1.86E−05
11
0.0618
0.0070
−0.1937
3.87E−05
0.2007


191
chloride channel complex
1.10E−08
5
0.0862
0.0071
−0.3284
3.09E−08
0.3355


192
protein complex assembly
7.43E−19
27
0.1971
0.0411
−0.1599
3.98E−18
0.2009


193
cortical cytoskeleton
1.01E−10
10
0.2941
−0.0269
−0.1778
3.30E−10
0.1509


194
JNK cascade
6.36E−17
4
0.0667
0.0967
−0.4574
3.09E−16
0.5540


195
telomere maintenance
1.80E−11
24
0.3038
0.0521
0.2155
6.25E−11
−0.1634


196
protein sumoylation
2.10E−06
9
0.2813
0.1265
−0.0576
4.79E−06
0.1841


197
M band
5.72E−91
10
0.3571
0.1855
−0.5734
2.51E−89
0.7589


198
I band
3.67E−48
4
0.1600
0.1431
−0.7377
5.61E−47
0.8808


199
cytosolic large ribosomal subunit
0
46
0.8214
0.0575
−0.4983
0
0.5558


200
protein heterooligomerization
6.72E−15
15
0.1049
0.1137
−0.1556
2.96E−14
0.2693


201
mitochondrial proton-transporting ATP synthase complex, coupling
1.11E−27
5
0.4545
0.1134
0.5065
9.30E−27
−0.3931



factor F(o)


202
protein localization
5.79E−07
12
0.1379
0.1527
0.4441
1.38E−06
−0.2915


203
T cell activation
1.08E−07
6
0.0938
0.0074
0.1496
2.77E−07
−0.1422


204
mitochondrial respiratory chain complex I
3.97E−06
9
0.1579
0.1040
0.4186
8.78E−06
−0.3146


205
NAD metabolic process
4.83E−21
3
0.1765
0.2828
−0.2346
2.99E−20
0.5174


206
cellular carbohydrate metabolic process
9.11E−40
7
0.1373
0.2311
−0.1412
1.06E−38
0.3723


207
NAD binding
9.83E−21
21
0.2414
0.1693
−0.0252
6.03E−20
0.1945


208
kinesin complex
7.66E−22
15
0.0932
0.2500
−0.3960
5.08E−21
0.6461


209
protein serine/threonine phosphatase activity
7.16E−10
17
0.2615
0.0799
0.2206
2.20E−09
−0.1407


210
positive regulation of DNA binding
4.81E−43
7
0.2414
−0.0171
−0.4702
6.21E−42
0.4530


211
positive regulation of protein phosphorylation
8.74E−07
17
0.1012
0.0585
−0.1902
2.06E−06
0.2487


212
protein disulfide oxidoreductase activity
6.18E−13
7
0.1795
−0.0757
−0.2602
2.42E−12
0.1845


213
cell redox homeostasis
1.59E−12
20
0.1342
−0.0338
0.1726
6.07E−12
−0.2065


214
small ribosomal subunit
0
13
0.2826
0.1058
−0.7565
0
0.8623


215
rRNA binding
 9.03E−240
12
0.3000
0.0760
−0.5321
 1.09E−237
0.6081


216
protein methylation
6.24E−14
5
0.0847
0.0592
−0.2891
2.56E−13
0.3483


217
fatty acid biosynthetic process
4.48E−12
7
0.0875
0.4321
−0.1686
1.64E−11
0.6007


218
U1 snRNP
5.40E−17
12
0.3750
0.0210
0.1874
2.63E−16
−0.2084


219
mRNA splice site selection
1.67E−17
8
0.1818
0.0310
0.3085
8.45E−17
−0.2775


220
RS domain binding
2.91E−07
5
0.3846
−0.0064
0.2875
7.18E−07
−0.2939


221
nuclear periphery
8.78E−06
5
0.4167
−0.1023
0.3065
1.88E−05
−0.4087


222
protein import into nucleus
5.08E−09
11
0.1774
0.0532
−0.0212
1.48E−08
0.0744


223
cell projection assembly
2.82E−24
3
0.2000
0.2165
−0.4015
2.11E−23
0.6180


224
response to insulin stimulus
4.21E−17
6
0.0583
0.0756
0.3782
2.07E−16
−0.3026


225
positive regulation of protein binding
2.21E−78
7
0.1296
0.0653
−0.6790
6.96E−77
0.7443


226
protein targeting to mitochondrion
2.07E−10
25
0.4032
0.0849
0.2611
6.58E−10
−0.1762


227
cell leading edge
8.20E−06
10
0.1515
0.1216
−0.0921
1.76E−05
0.2137


228
mast cell granule
1.73E−76
4
0.1333
0.3477
−0.6160
5.14E−75
0.9637


229
positive regulation of insulin secretion
3.97E−05
6
0.0984
0.1055
0.4181
7.98E−05
−0.3126


230
myosin complex
1.30E−11
12
0.1111
0.1370
−0.1418
4.59E−11
0.2788


231
regulation of gene expression
1.96E−05
8
0.0860
0.1581
−0.1596
4.06E−05
0.3177


232
positive regulation of proteasomal ubiquitin-dependent protein catabolic
1.00E−21
9
0.1607
0.0718
−0.2466
6.49E−21
0.3184



process


233
condensed nuclear chromosome
4.75E−13
11
0.3056
0.0478
0.3658
1.88E−12
−0.3180


234
nuclear inner membrane
2.88E−40
10
0.2857
0.0419
0.3551
3.46E−39
−0.3132


235
formation of translation preinitiation complex
6.07E−09
13
0.5200
−0.0183
−0.2185
1.75E−08
0.2002


236
eukaryotic translation initiation factor 3 complex
1.80E−08
14
0.2090
−0.0189
−0.1858
5.02E−08
0.1669


237
regulation of translational initiation
1.34E−21
27
0.5000
0.0030
−0.2245
8.64E−21
0.2275


238
eukaryotic 43S preinitiation complex
6.07E−09
13
0.5200
−0.0183
−0.2185
1.75E−08
0.2002


239
eukaryotic 48S preinitiation complex
6.07E−09
13
0.5417
−0.0183
−0.2185
1.75E−08
0.2002


240
protein N-linked glycosylation
3.23E−07
3
0.0909
0.0314
0.3826
7.93E−07
−0.3513


241
Rho protein signal transduction
9.20E−44
13
0.2167
0.0497
−0.8038
1.20E−42
0.8536


242
DNA metabolic process
8.58E−12
5
0.1000
0.0711
−0.2524
3.07E−11
0.3235


243
DNA-dependent ATPase activity
3.04E−05
5
0.0794
−0.1703
0.0236
6.18E−05
−0.1939


244
regulation of actin cytoskeleton organization
4.17E−21
10
0.1370
−0.0419
−0.6609
2.59E−20
0.6190


245
response to hormone stimulus
1.37E−35
10
0.1724
0.0156
0.3757
1.46E−34
−0.3601


246
lipoprotein metabolic process
1.87E−16
3
0.0417
−0.0719
0.2379
8.83E−16
−0.3097


247
negative regulation of protein kinase activity
2.11E−88
12
0.1290
0.0908
−0.5506
8.31E−87
0.6414


248
fatty acid metabolic process
2.22E−13
6
0.0952
0.2441
−0.1549
9.01E−13
0.3989


249
muscle cell homeostasis
8.67E−19
4
0.1250
0.0726
−0.5939
4.63E−18
0.6664


250
ESC/E(Z) complex
3.65E−06
4
0.1429
−0.1091
0.1234
8.10E−06
−0.2325


251
establishment of mitotic spindle orientation
3.77E−05
3
0.0882
0.0530
0.3136
7.61E−05
−0.2606


252
inner cell mass cell proliferation
2.67E−05
3
0.1250
0.0106
0.2412
5.47E−05
−0.2307


253
retrograde vesicle-mediated transport, Golgi to ER
4.27E−09
15
0.4545
0.0158
0.1946
1.25E−08
−0.1788


254
regulation of cell adhesion
1.15E−06
12
0.2182
0.0974
−0.0750
2.68E−06
0.1724


255
bone resorption
2.43E−11
4
0.1081
0.0514
0.3704
8.36E−11
−0.3190


256
uropod
3.71E−14
4
0.2857
−0.0786
−0.2817
1.55E−13
0.2031


257
cellular component movement
7.40E−44
35
0.3241
−0.0436
−0.3191
9.72E−43
0.2756


258
myosin II complex
1.88E−21
3
0.2727
0.1398
−0.1386
1.20E−20
0.2783


259
regulation of blood pressure
2.04E−05
9
0.1200
−0.0237
0.2003
4.22E−05
−0.2240


260
DNA damage response, signal transduction resulting in induction of
9.15E−11
7
0.0769
0.0628
−0.0451
2.98E−10
0.1079



apoptosis


261
DNA-dependent DNA replication initiation
1.26E−10
8
0.2222
−0.1097
0.0908
4.08E−10
−0.2005


262
MCM complex
4.30E−11
7
0.3684
−0.1105
0.0900
1.44E−10
−0.2005


263
cell division
1.33E−50
21
0.1963
0.0561
−0.2698
2.12E−49
0.3259


264
damaged DNA binding
3.44E−06
20
0.1587
0.0462
−0.0431
7.69E−06
0.0893


265
double-strand break repair via nonhomologous end joining
5.49E−09
7
0.2500
0.1137
0.3252
1.59E−08
−0.2115


266
ADP binding
1.06E−05
14
0.3590
0.0403
0.1046
2.24E−05
−0.0643


267
flavin adenine dinucleotide binding
9.40E−29
16
0.0851
0.0363
0.3354
8.14E−28
−0.2992


268
keratinocyte differentiation
9.44E−22
8
0.1212
0.1356
−0.4606
6.17E−21
0.5962


269
fatty-acyl-CoA binding
2.57E−34
7
0.2414
0.0747
0.5318
2.58E−33
−0.4571


270
embryo development
2.37E−05
13
0.0935
0.0505
0.1017
4.87E−05
−0.0512


271
response to starvation
8.38E−12
5
0.0877
0.0057
0.3514
3.00E−11
−0.3457


272
hippocampus development
4.08E−08
7
0.1045
0.2322
−0.1195
1.08E−07
0.3518


273
ion channel binding
1.29E−29
16
0.1600
0.0685
−0.1936
1.14E−28
0.2622


274
brush border
4.95E−05
5
0.1250
0.0005
−0.0916
9.82E−05
0.0922


275
F-actin capping protein complex
2.75E−61
5
0.3333
0.3471
−0.4389
5.63E−60
0.7860


276
ribosome binding
5.66E−10
19
0.3654
0.0026
−0.1109
1.74E−09
0.1135


277
response to amino acid stimulus
6.97E−53
4
0.0800
0.2121
−0.4568
1.17E−51
0.6689


278
positive regulation of translation
4.31E−22
19
0.3115
0.0426
−0.2001
2.90E−21
0.2428


279
positive regulation of stress fiber assembly
6.98E−24
5
0.1020
0.0674
−0.5145
5.12E−23
0.5818


280
positive regulation of protein kinase B signaling cascade
5.37E−06
7
0.0761
0.0084
0.2691
1.17E−05
−0.2608


281
cell body
1.64E−78
20
0.2299
0.0095
−0.2873
5.27E−77
0.2969


282
ATP biosynthetic process
2.99E−06
9
0.3750
0.0703
0.2771
6.74E−06
0.2068


283
response to activity
4.85E−08
10
0.1923
0.0652
0.2565
1.28E−07
−0.1912


284
positive regulation of neuron apoptotic process
1.61E−24
8
0.1194
0.1132
−0.3507
1.21E−23
0.4639


285
positive regulation of blood pressure
2.91E−10
3
0.1304
0.0913
−0.2747
9.15E−10
0.3660


286
Rac GTPase binding
8.81E−08
11
0.2558
−0.0332
−0.0232
2.28E−07
−0.0100


287
regulation of the force of heart contraction
1.75E−17
3
0.1111
−0.0033
0.4506
8.80E−17
0.4539


288
positive regulation of cell growth
1.81E−17
18
0.2045
0.1953
0.0290
9.07E−17
0.1663


289
removal of superoxide radicals
1.28E−54
5
0.2083
0.3803
−0.3708
2.24E−53
0.7511


290
internal side of plasma membrane
1.88E−07
8
0.1333
0.0899
−0.3034
4.73E−07
0.3933


291
positive regulation of NF-kappaB transcription factor activity
4.12E−14
23
0.1533
0.1065
−0.0644
1.70E−13
0.1709


292
fructose 6-phosphate metabolic process
1.46E−10
5
0.1923
−0.0239
−0.3041
4.69E−10
0.2802


293
anterior/posterior pattern specification
2.11E−05
6
0.0462
0.0220
0.3121
4.37E−05
−0.2901


294
Z disc
5.83E−18
23
0.1456
0.0807
−0.1475
3.02E−17
0.2282


295
epithelial cell differentiation
8.91E−06
22
0.2750
−0.0205
−0.1203
1.91E−05
0.0998


296
toll-like receptor signaling pathway
5.27E−06
18
0.1565
0.1001
−0.0696
1.15E−05
0.1698


297
transcriptional repressor complex
1.90E−26
12
0.1538
0.0582
−0.2754
1.52E−25
0.3336


298
nuclear chromatin
4.09E−23
29
0.1667
0.0082
0.1644
2.86E−22
−0.1562


299
iron-sulfur cluster binding
5.24E−08
3
0.0811
0.0714
−0.3253
1.37E−07
0.3967


300
secretory granule membrane
9.28E−13
3
0.0833
−0.1127
0.3309
3.61E−12
−0.4436


301
maternal placenta development
1.83E−09
3
0.1667
−0.0974
0.4196
5.50E−09
−0.5170


302
nuclear envelope lumen
9.16E−81
3
0.2500
0.0464
−0.8702
3.01E−79
0.9166


303
regulation of insulin secretion
3.83E−16
18
0.2000
0.0009
0.2089
1.77E−15
−0.2080


304
microtubule cytoskeleton organization
2.19E−27
14
0.0859
0.1694
−0.1871
1.80E−26
0.3565


305
Ran GTPase binding
1.13E−07
13
0.1970
0.0276
−0.1731
2.89E−07
0.2007


306
repressing transcription factor binding
1.36E−23
6
0.1500
0.0962
−0.3316
9.75E−23
0.4278


307
ER-associated protein catabolic process
1.85E−20
8
0.1905
−0.0070
0.2512
1.09E−19
−0.2582


308
regulation of neuron apoptotic process
4.08E−58
4
0.1538
0.1807
−0.5285
7.75E−57
0.7091


309
pseudopodium
3.64E−05
7
0.2917
0.0529
−0.1218
7.34E−05
0.1747


310
CenH3-containing nucleosome assembly at centromere
1.46E−06
9
0.3600
0.0744
0.2157
3.40E−06
−0.1413


311
NADH dehydrogenase (ubiquinone) activity
9.29E−06
8
0.1194
0.1258
0.4901
1.98E−05
−0.3642


312
replication fork
3.80E−11
4
0.1000
0.0559
0.4037
1.28E−10
−0.3478


313
chromatin DNA binding
8.30E−26
9
0.1304
0.0410
0.3417
6.41E−25
−0.3007


314
cellular response to heat
1.55E−11
7
0.1591
−0.0221
0.2861
5.43E−11
−0.3082


315
histone H3 deacetylation
9.53E−15
6
0.2400
0.0265
0.3381
4.16E−14
−0.3116


316
ER to Golgi vesicle-mediated transport
4.95E−13
16
0.1495
0.0244
0.1880
1.96E−12
−0.1635


317
protein secretion
5.77E−36
6
0.2222
0.0689
0.4599
6.18E−35
−0.3911


318
protein maturation
3.42E−15
3
0.1154
−0.0181
0.3479
1.52E−14
−0.3660


319
double-strand break repair
7.32E−14
20
0.2500
0.0539
0.2068
2.99E−13
−0.1529


320
response to ionizing radiation
1.42E−07
4
0.0667
0.0726
0.4400
3.62E−07
−0.3674


321
translation elongation factor activity
 1.05E−275
10
0.1961
0.1009
−0.7616
 1.38E−273
0.8625


322
translational elongation
0
84
0.6512
0.0734
−0.5621
0
0.6355


323
cell chemotaxis
5.91E−06
6
0.0845
−0.1442
−0.4587
1.28E−05
0.3145


324
activating transcription factor binding
9.12E−06
4
0.1538
−0.0098
0.2681
1.95E−05
−0.2779


325
substantia nigra development
4.81E−15
20
0.4348
0.1008
−0.0618
2.13E−14
0.1626


326
positive regulation of phagocytosis
1.11E−16
5
0.1563
−0.1615
0.2038
5.31E−16
−0.3653


327
brush border membrane
7.79E−38
4
0.0588
−0.1205
−0.8762
8.71E−37
0.7556


328
tRNA aminoacylation for protein translation
5.63E−22
28
0.4058
0.0034
−0.1856
3.74E−21
0.1890


329
actomyosin structure organization
2.20E−06
6
0.1935
0.0949
−0.1093
4.99E−06
0.2043


330
proteasome core complex, alpha-subunit complex
3.44E−07
7
0.2692
−0.0375
0.0570
8.36E−07
0.0946


331
protein disulfide isomerase activity
2.95E−70
10
0.3226
−0.0087
0.3645
7.50E−69
−0.3732


332
COPI-coated vesicle
1.88E−18
3
0.2727
0.0380
0.7951
9.89E−18
−0.7571


333
syntaxin binding
5.26E−11
5
0.0962
−0.0756
0.2779
1.75E−10
−0.3535


334
DNA damage checkpoint
2.71E−11
4
0.0678
0.1067
0.5327
9.29E−11
−0.4259


335
negative regulation of DNA replication
1.01E−14
5
0.1667
−0.1111
−0.7643
4.39E−14
0.6532


336
tricarboxylic acid cycle
4.85E−35
21
0.4200
0.0544
0.2481
5.00E−34
−0.1937


337
isocitrate metabolic process
2.15E−09
3
0.2727
0.0465
−0.3325
6.37E−09
0.3791


338
positive regulation of nitric oxide biosynthetic process
6.44E−86
4
0.1000
0.0667
−0.6094
2.42E−84
0.6761


339
negative regulation of ryanodine-sensitive calcium-release channel
1.05E−08
3
0.2727
0.1002
−0.0999
2.98E−08
0.2001



activity


340
microvillus
2.33E−14
12
0.2308
0.0411
−0.1549
9.86E−14
0.1960


341
adult locomotory behavior
3.46E−19
11
0.1264
0.1305
−0.1355
1.91E−18
0.2660


342
transcription from mitochondrial promoter
9.32E−06
3
0.2727
0.1532
0.4923
1.99E−05
−0.3391


343
aerobic respiration
1.56E−11
7
0.2800
−0.0184
0.2265
5.45E−11
−0.2449


344
cytokine binding
3.84E−21
5
0.2000
0.0944
−0.4401
2.40E−20
0.5345


345
negative regulation of proteolysis
6.15E−10
5
0.1163
0.2571
−0.1700
1.89E−09
0.4270


346
translational termination
0
78
0.7800
0.0735
−0.5473
0
0.6207


347
T cell differentiation in thymus
1.03E−09
5
0.0893
0.0537
0.3345
3.13E−09
−0.2808


348
regulation of cyclin-dependent protein kinase activity
1.83E−33
6
0.0674
0.0772
−0.6234
1.76E−32
0.7006


349
mitochondrial electron transport, NADH to ubiquinone
8.49E−06
9
0.1800
0.0644
0.2295
1.82E−05
−0.1651


350
neuron apoptotic process
1.43E−07
9
0.1429
0.2412
−0.1299
3.63E−07
0.3711


351
intrinsic apoptotic signaling pathway
1.81E−81
19
0.3167
0.1540
−0.3102
6.09E−80
0.4642


352
isomerase activity
5.45E−08
6
0.2069
−0.0341
0.2653
1.43E−07
−0.2994


353
de novo' IMP biosynthetic process
2.14E−26
6
0.6000
0.0453
−0.3111
1.69E−25
0.3564


354
cerebellum development
1.07E−08
4
0.0784
0.0427
−0.2844
3.02E−08
0.3271


355
tetrahydrofolate biosynthetic process
1.55E−14
3
0.2500
0.0551
−0.3561
6.65E−14
0.4112


356
small-subunit processome
7.06E−43
4
0.2105
0.1071
−0.6545
8.97E−42
0.7616


357
cellular response to oxidative stress
1.23E−40
9
0.2045
0.1776
−0.2669
1.49E−39
0.4445


358
glycogen biosynthetic process
3.17E−07
6
0.1538
0.0696
−0.1749
7.79E−07
0.2445


359
mitochondrial transport
2.58E−54
3
0.1034
0.2583
−0.4483
4.47E−53
0.7065


360
acyl-CoA dehydrogenase activity
4.47E−06
3
0.0857
0.0335
0.3614
9.84E−06
−0.3279


361
DNA topoisomerase (ATP-hydrolyzing) activity
2.49E−05
3
0.2727
−0.1182
0.0638
5.11E−05
−0.1819


362
DNA topological change
8.13E−06
5
0.1136
−0.0845
−0.1469
1.75E−05
0.0624


363
transcription cofactor activity
4.19E−08
10
0.1163
0.1380
−0.1365
1.11E−07
0.2745


364
RNA helicase activity
2.35E−07
8
0.6667
0.0344
0.1927
5.84E−07
−0.1584


365
binding of sperm to zona pellucida
2.82E−47
12
0.2308
0.0028
−0.2384
4.20E−46
0.2413


366
ossification
1.83E−08
7
0.0745
0.0846
−0.2926
5.08E−08
0.3772


367
cyclin-dependent protein kinase activity
4.72E−05
3
0.0698
0.1909
−0.0634
9.43E−05
0.2543


368
circadian rhythm
1.66E−11
9
0.1154
0.0715
0.2641
5.79E−11
−0.1926


369
GDP binding
1.53E−28
21
0.3818
−0.0091
0.3117
1.32E−27
−0.3208


370
skeletal muscle tissue development
3.26E−21
4
0.0533
0.0849
−0.4791
2.06E−20
0.5640


371
pentose-phosphate shunt
1.41E−71
7
0.2258
0.0774
−0.3483
3.77E−70
0.4257


372
blood vessel development
1.96E−12
5
0.0685
0.3305
0.7654
7.39E−12
−0.4349


373
NuRD complex
1.62E−10
7
0.1944
−0.0583
0.1386
5.20E−10
−0.1969


374
mRNA catabolic process
1.43E−12
3
0.1765
0.0213
−0.3699
5.46E−12
0.3911


375
gluconeogenesis
1.97E−91
22
0.3188
0.1681
−0.2531
8.88E−90
0.4212


376
adenyl nucleotide binding
5.55E−12
3
0.0698
0.0760
−0.3502
2.01E−11
0.4262


377
clathrin coat of trans-Golgi network vesicle
4.68E−22
4
0.2222
0.1513
−0.1391
3.14E−21
0.2903


378
clathrin coat of coated pit
9.81E−22
5
0.2381
0.1484
−0.1391
6.39E−21
0.2874


379
receptor tyrosine kinase binding
3.62E−24
8
0.2105
0.0284
−0.3162
2.67E−23
0.3446


380
positive regulation of peptidyl-serine phosphorylation
4.29E−05
9
0.1552
0.1396
−0.0602
8.60E−05
0.1997


381
cell periphery
2.98E−08
6
0.1579
0.1888
−0.2996
8.02E−08
0.4884


382
DNA ligation involved in DNA repair
4.84E−30
3
0.3000
−0.0353
−0.6013
4.33E−29
0.5660


383
copper ion transport
1.17E−20
3
0.1071
0.1457
−0.2765
7.11E−20
0.4222


384
membrane protein ectodomain proteolysis
2.08E−10
5
0.2000
0.0071
0.1713
6.61E−10
−0.1642


385
signal peptide processing
3.72E−07
3
0.1304
−0.0387
0.3418
9.00E−07
0.3805


386
serine-type peptidase activity
6.33E−12
9
0.0938
−0.0842
0.4508
2.28E−11
0.5350


387
sperm protein complex
1.77E−61
9
0.4091
0.0030
−0.2561
3.68E−60
0.2591


388
chaperonin-containing T-complex
1.60E−59
9
0.4286
0.0019
−0.2561
3.16E−58
0.2580


389
positive regulation of ATPase activity
3.97E−07
6
0.2000
0.0197
−0.1975
9.59E−07
0.2171


390
single-stranded DNA binding
6.04E−07
29
0.2900
−0.0186
0.1155
1.44E−06
−0.1341


391
nucleocytoplasmic transport
2.10E−16
7
0.1346
0.1384
0.0175
9.89E−16
0.1209


392
oxaloacetate metabolic process
1.76E−14
6
0.3158
0.1843
0.4418
7.52E−14
−0.2575


393
apoptotic cell clearance
1.89E−27
3
0.1111
−0.0353
0.4007
1.56E−26
−0.4360


394
Golgi organization
3.45E−11
13
0.2167
0.0217
0.3457
1.17E−10
−0.3241


395
barbed-end actin filament capping
1.63E−20
5
0.3333
0.3442
−0.2589
9.71E−20
0.6031


396
oxidoreductase activity, acting on NADH or NADPH
4.51E−11
3
0.1304
0.0556
0.4292
1.51E−10
0.3735


397
hemidesmosome
6.25E−06
5
0.2500
−0.0297
0.1078
1.36E−05
−0.1375


398
positive regulation of dendrite morphogenesis
3.96E−05
3
0.0811
−0.0059
−0.2831
7.96E−05
0.2772


399
negative regulation of endothelial cell proliferation
1.24E−19
6
0.1818
0.0610
0.4194
7.05E−19
−0.3583


400
malate metabolic process
2.09E−07
3
0.1364
0.1559
0.3094
5.22E−07
−0.1535


401
protein tetramerization
1.87E−07
9
0.2432
−0.0171
−0.2124
4.71E−07
0.1953


402
regulation of circadian rhythm
3.64E−06
6
0.2000
0.0341
0.2330
8.09E−06
−0.1989


403
spindle assembly involved in mitosis
1.14E−05
4
0.3636
−0.0301
−0.1648
2.40E−05
0.1347


404
positive regulation of fibroblast proliferation
1.52E−66
6
0.0769
0.2888
−0.4795
3.52E−65
0.7683


405
protein kinase C binding
3.74E−19
17
0.3036
0.0308
−0.1962
2.05E−18
0.2270


406
cotranslational protein targeting to membrane
1.10E−08
4
0.3333
−0.0479
0.3067
3.09E−08
−0.3546


407
glycoprotein binding
7.60E−27
13
0.1494
−0.0545
0.2138
6.15E−26
−0.2683


408
dendrite cytoplasm
3.59E−07
4
0.1739
−0.0001
0.2307
8.70E−07
−0.2309


409
apolipoprotein binding
2.81E−18
3
0.1364
0.0455
0.5615
1.48E−17
−0.5160


410
chaperone-mediated protein folding
7.74E−13
16
0.3902
−0.0193
0.1316
3.02E−12
−0.1509


411
cofactor binding
7.50E−11
4
0.1250
0.0767
−0.2231
2.46E−10
0.2998


412
response to heat
5.06E−07
10
0.1695
−0.1153
0.1835
1.21E−06
−0.2988


413
positive regulation of protein import into nucleus, translocation
1.16E−24
5
0.3125
0.0410
−0.5842
8.80E−24
0.5432


414
nuclear chromosome
1.29E−07
11
0.3667
0.0063
0.1873
3.30E−07
−0.1810


415
chromatin remodeling
9.33E−06
19
0.1959
0.0114
0.1211
1.99E−05
0.1097


416
cellular response to interleukin-4
2.88E−21
7
0.2000
−0.0347
−0.4376
1.82E−20
0.4030


417
neuron projection development
1.80E−36
12
0.0795
0.1699
−0.2523
1.98E−35
0.4222


418
lung development
1.26E−06
11
0.0840
0.1057
0.3106
2.92E−06
−0.2048


419
translation initiation factor binding
2.84E−17
5
0.1724
−0.0021
0.2707
1.41E−16
0.2686


420
cytoplasmic membrane-bounded vesicle
1.04E−05
23
0.1783
−0.0414
−0.2561
2.21E−05
0.2147


421
ATP metabolic process
1.14E−12
3
0.0909
0.1667
−0.1389
4.39E−12
0.3055


422
response to ethanol
6.04E−09
15
0.0932
0.1773
−0.0286
1.75E−08
0.2058


423
regulation of acetyl-CoA biosynthetic process from pyruvate
5.32E−11
8
0.4000
−0.0338
0.2211
1.77E−10
0.2549


424
Ras GTPase binding
3.44E−06
4
0.2000
−0.0695
0.4286
7.69E−06
−0.4981


425
fatty acid transport
1.99E−14
3
0.1429
0.2694
0.6519
8.49E−14
−0.3825


426
positive regulation of protein serine/threonine kinase activity
3.01E−18
6
0.2308
−0.0602
−0.5967
1.58E−17
0.5364


427
dolichyl-diphosphooligosaccharide-protein glycotransferase activity
2.91E−17
6
0.4286
0.0258
0.3661
1.43E−16
−0.3403


428
regulation of sodium ion transport
2.14E−05
3
0.1667
0.0245
0.3275
4.42E−05
−0.3030


429
peptide binding
2.03E−09
11
0.2200
0.0219
−0.0439
6.04E−09
0.0658


430
androgen receptor signaling pathway
1.97E−35
10
0.2083
0.0951
−0.4147
2.08E−34
0.5098


431
protein neddylation
1.62E−08
3
0.2727
0.0431
−0.2579
4.53E−08
0.3010


432
tubulin binding
1.21E−71
5
0.0980
0.0539
−0.6659
3.34E−70
0.7198


433
protein N-terminus binding
3.10E−45
21
0.1810
0.0777
−0.1761
4.28E−44
0.2538


434
osteoblast differentiation
6.53E−40
31
0.2366
0.0419
0.1884
7.68E−39
−0.1464


435
striated muscle cell differentiation
4.02E−35
4
0.2000
0.4791
−0.6384
4.16E−34
1.1175


436
negative regulation of release of cytochrome c from mitochondria
4.84E−07
5
0.2778
0.2515
0.5898
1.17E−06
−0.3383


437
tropomyosin binding
1.91E−38
4
0.1667
0.4163
−0.5945
2.17E−37
1.0108


438
ubiquitin ligase complex
4.10E−14
9
0.1184
0.0643
−0.2040
1.70E−13
0.2683


439
male germ cell nucleus
7.53E−07
3
0.0938
0.0681
0.2913
1.79E−06
−0.2232


440
regulation of synaptic plasticity
1.47E−22
7
0.0959
0.1049
−0.4518
9.99E−22
0.5567


441
2 iron, 2 sulfur cluster binding
5.41E−06
5
0.1515
0.1634
0.5588
1.18E−05
0.3954


442
sodium:potassium-exchanging ATPase activity
3.24E−11
3
0.2500
0.0680
0.5054
1.10E−10
−0.4373


443
response to gamma radiation
2.29E−05
5
0.1220
0.0846
0.3212
4.72E−05
−0.2367


444
RNA polymerase II transcription factor binding
2.35E−11
6
0.1277
−0.0183
0.1702
8.10E−11
0.1884


445
peroxisomal matrix
1.17E−12
10
0.2778
0.1076
−0.1400
4.50E−12
0.2477


446
cellular senescence
2.02E−08
5
0.2381
−0.1977
0.1491
5.56E−08
−0.3468


447
positive regulation of extrinsic apoptotic signaling pathway
2.64E−08
5
0.1190
−0.0587
0.2038
7.13E−08
0.2624


448
cellular calcium ion homeostasis
6.91E−16
15
0.1327
−0.0501
0.2832
3.14E−15
0.3332


449
response to salt stress
1.48E−23
3
0.1200
−0.1103
−0.8640
1.06E−22
0.7538


450
defense response to Gram-positive bacterium
1.65E−06
7
0.0959
0.0195
0.2875
3.81E−06
−0.2679


451
chromosome segregation
1.09E−05
17
0.1932
−0.0449
0.0671
2.30E−05
0.1120


452
nuclear heterochromatin
1.22E−11
8
0.2105
0.0910
0.2700
4.30E−11
0.1790


453
protein localization to nucleus
1.54E−06
5
0.1111
0.0195
0.2002
3.56E−06
−0.1807


454
late endosome
2.39E−10
16
0.1345
0.0090
0.2374
7.56E−10
−0.2284


455
polysome
1.50E−17
11
0.2973
−0.0253
−0.2169
7.62E−17
0.1915


456
negative regulation of translation
2.40E−32
18
0.2903
0.0404
−0.1847
2.23E−31
0.2250


457
DNA damage response, signal transduction by p53 class mediator
3.41E−13
7
0.1842
0.0781
0.3064
1.37E−12
−0.2283



resulting in induction of apoptosis


458
regulation of angiogenesis
1.52E−17
3
0.1000
−0.0264
−0.5445
7.72E−17
0.5182


459
activation of cysteine-type endopeptidase activity involved in apoptotic
5.25E−18
16
0.1702
−0.0254
0.1415
2.73E−17
−0.1668



process


460
endocytic vesicle
7.50E−06
10
0.1515
−0.0763
0.2451
1.62E−05
−0.3214


461
respiratory electron transport chain
 1.86E−147
44
0.4190
0.1104
0.4538
 1.72E−145
−0.3435


462
regulation of signal transduction
5.61E−45
3
0.0968
0.1135
−0.6958
7.49E−44
0.8093


463
DNA unwinding involved in replication
4.26E−12
7
0.4667
−0.1421
0.0555
1.56E−11
−0.1976


464
histone binding
3.66E−10
27
0.3375
0.0399
0.1738
1.14E−09
0.1340


465
cellular response to glucose starvation
1.63E−18
5
0.2083
0.0077
0.3064
8.67E−18
−0.2987


466
fatty acid beta-oxidation
9.00E−48
12
0.2727
0.0709
0.4462
1.35E−46
0.3753


467
positive regulation of interferon-alpha production
3.61E−11
3
0.1429
−0.0389
0.2760
1.22E−10
−0.3149


468
positive regulation of T cell activation
2.16E−19
4
0.1667
−0.0518
0.2760
1.20E−18
−0.3278


469
endoplasmic reticulum unfolded protein response
1.01E−23
26
0.2796
0.0182
0.2047
7.28E−23
−0.1865


470
positive regulation of protein secretion
4.62E−28
6
0.1200
0.2172
−0.4498
3.93E−27
0.6669


471
microtubule plus-end binding
8.72E−20
5
0.2273
0.1441
−0.3441
4.98E−19
0.4882


472
neutrophil chemotaxis
1.07E−05
4
0.0755
0.1694
−0.1674
2.26E−05
0.3368


473
membrane protein intracellular domain proteolysis
9.41E−09
3
0.1500
0.1662
0.5931
2.67E−08
−0.4269


474
TOR signaling cascade
1.34E−95
4
0.2353
0.1135
−1.0843
7.06E−94
1.1978


475
negative regulation of cell adhesion
3.58E−06
9
0.2195
−0.1855
0.6847
7.98E−06
0.4992


476
alternative nuclear mRNA splicing, via spliceosome
3.13E−39
5
0.3125
0.0372
0.3437
3.60E−38
−0.3065


477
nuclear speck
3.07E−25
69
0.3651
0.0382
0.1823
2.35E−24
−0.1442


478
cortical actin cytoskeleton
1.02E−20
10
0.2222
−0.0259
0.7477
6.24E−20
0.7218


479
glucose binding
4.66E−28
5
0.2273
0.1165
−0.4140
3.95E−27
0.5305


480
response to steroid hormone stimulus
1.03E−17
3
0.1200
−0.0259
−0.4611
5.28E−17
0.4352


481
response to endoplasmic reticulum stress
5.97E−49
13
0.2321
−0.0108
0.3529
9.31E−48
−0.3637


482
cellular response to reactive oxygen species
4.76E−05
6
0.3529
−0.0968
0.1602
9.48E−05
0.2571


483
reactive oxygen species metabolic process
1.41E−06
7
0.2258
−0.0740
0.1367
3.27E−06
−0.2107


484
NADH metabolic process
1.20E−08
4
0.3636
0.1700
0.4043
3.37E−08
−0.2343


485
succinate metabolic process
1.65E−15
5
0.4167
0.0385
0.4135
7.41E−15
0.3750


486
galactose catabolic process
1.23E−05
4
0.4000
0.0839
−0.1210
2.56E−05
0.2049


487
positive regulation of interleukin-6 production
1.74E−16
3
0.0526
−0.0463
0.2715
8.24E−16
−0.3178


488
negative regulation of actin filament polymerization
2.82E−14
5
0.2632
0.0237
−0.4420
1.19E−13
0.4657


489
oxidative phosphorylation
6.47E−28
8
0.4706
0.0053
0.2990
5.46E−27
−0.2936


490
androgen receptor binding
9.79E−31
10
0.2564
0.0745
−0.1461
8.87E−30
0.2206


491
mitochondrial intermembrane space
8.59E−37
19
0.2468
0.0463
0.3382
9.53E−36
−0.2919


492
semaphorin receptor binding
1.72E−27
4
0.1905
−0.0150
−1.0965
1.42E−26
1.0815


493
melanosome transport
5.35E−10
4
0.1481
−0.0625
0.2929
1.66E−09
−0.3554


494
B cell proliferation
3.64E−12
3
0.0769
−0.0416
0.3223
1.34E−11
−0.3639


495
ion transmembrane transporter activity
5.94E−16
5
0.4167
0.2073
−0.4874
2.71E−15
0.6947


496
receptor internalization
4.92E−06
6
0.1538
0.0608
−0.0820
1.08E−05
0.1429


497
positive regulation of type I interferon production
3.09E−06
18
0.2609
0.0849
0.2182
6.95E−06
−0.1333


498
Hsp70 protein binding
5.95E−07
10
0.4167
0.1214
−0.1565
1.42E−06
0.2779


499
cytoplasmic stress granule
1.92E−17
18
0.3673
0.0417
−0.1715
9.59E−17
0.2132


500
enoyl-CoA hydratase activity
4.15E−44
4
0.5000
0.0995
0.6085
5.49E−43
−0.5090


501
long-chain-enoyl-CoA hydratase activity
6.35E−40
3
0.5000
0.0729
0.5234
7.52E−39
−0.4505


502
cleavage furrow
2.25E−11
14
0.2979
0.0795
−0.1091
7.79E−11
0.1887


503
stress fiber
2.36E−14
20
0.3390
0.0405
−0.1974
9.97E−14
0.2380


504
respiratory chain
1.58E−20
3
0.2727
0.1732
0.6190
9.45E−20
−0.4458


505
I-kappaB kinase/NF-kappaB cascade
1.26E−20
4
0.0833
0.1142
−0.4574
7.60E−20
0.5715


506
regulation of heart contraction
7.41E−12
5
0.1087
0.1342
0.2101
2.66E−11
0.3443


507
protein targeting
1.59E−55
13
0.2203
0.1083
−0.3452
2.85E−54
0.4535


508
myosin V binding
1.10E−11
3
0.2727
−0.0476
0.3582
3.93E−11
−0.4058


509
microtubule plus end
6.41E−11
6
0.3333
0.1223
−0.2645
2.12E−10
0.3867


510
spindle organization
1.46E−08
5
0.1923
0.1745
0.3029
4.08E−08
0.4773


511
neural tube development
2.19E−32
4
0.1429
0.3628
−0.3263
2.05E−31
0.6892


512
membrane depolarization
1.21E−33
3
0.1304
0.1498
−0.4710
1.19E−32
0.6207


513
cholesterol binding
8.05E−07
6
0.1463
−0.0711
0.0877
1.91E−06
−0.1588


514
mitochondrial fusion
2.75E−11
4
0.2353
0.1136
0.5389
9.41E−11
−0.4252


515
germ cell programmed cell death
1.04E−10
3
0.2308
0.0994
0.3617
3.38E−10
−0.2623


516
response to radiation
1.00E−07
5
0.1786
−0.0510
−0.3620
2.59E−07
0.3109


517
rough endoplasmic reticulum
5.22E−11
8
0.1739
0.0781
0.3728
1.74E−10
−0.2947


518
protein oligomerization
5.07E−10
12
0.2667
0.1420
0.3705
1.58E−09
0.2285


519
phosphatidylinositol-4,5-bisphosphate binding
1.51E−18
8
0.1270
0.0498
−0.2914
8.02E−18
0.3411


520
release of cytochrome c from mitochondria
5.53E−07
8
0.2105
0.1631
0.0397
1.32E−06
0.1234


521
B cell activation
1.01E−07
6
0.2069
−0.0367
0.2404
2.59E−07
−0.2771


522
negative regulation of gene expression
1.54E−05
9
0.0783
0.0684
−0.1719
3.22E−05
0.2402


523
negative regulation of extrinsic apoptotic signaling pathway
9.13E−10
8
0.1538
0.1847
−0.0916
2.79E−09
0.2763


524
MyD88-dependent toll-like receptor signaling pathway
4.06E−10
17
0.1932
0.0747
−0.1016
1.27E−09
0.1763


525
nitric oxide metabolic process
5.43E−27
4
0.1818
0.1779
−0.3707
4.41E−26
0.5486


526
cytochrome-c oxidase activity
2.42E−53
9
0.1698
0.1961
0.6031
4.14E−52
−0.4070


527
mitochondrial respiratory chain
3.20E−11
4
0.1600
0.1569
0.5555
1.09E−10
−0.3986


528
nuclear outer membrane
1.60E−33
9
0.3000
0.1319
0.4995
1.55E−32
−0.3675


529
mitochondrion transport along microtubule
2.11E−14
4
0.2667
0.0147
0.3741
8.97E−14
−0.3594


530
MyD88-independent toll-like receptor signaling pathway
2.19E−08
13
0.1646
0.0883
−0.1830
5.97E−08
0.2713


531
toll-like receptor 3 signaling pathway
2.19E−08
13
0.1605
0.0883
−0.1830
5.97E−08
0.2713


532
potassium channel regulator activity
3.52E−33
5
0.1020
0.3392
−0.6883
3.36E−32
1.0274


533
SRP-dependent cotranslational protein targeting to membrane
0
95
0.8051
0.0670
−0.4935
0
0.5605


534
coated pit
1.88E−12
18
0.3000
−0.0156
0.3177
7.10E−12
−0.3332


535
fibroblast growth factor binding
5.26E−48
4
0.1481
0.1213
−0.5611
7.97E−47
0.6824


536
ribosomal large subunit biogenesis
4.95E−89
11
0.6875
0.0282
−0.4485
2.00E−87
0.4767


537
phosphoprotein binding
5.13E−86
9
0.2093
0.1394
−0.3112
1.97E−84
0.4506


538
voltage-gated chloride channel activity
7.84E−09
4
0.1429
0.0072
−0.3284
2.24E−08
0.3356


539
establishment of protein localization
3.49E−06
5
0.1852
−0.0355
−0.1767
7.80E−06
0.1412


540
trans-Golgi network membrane
2.56E−16
10
0.1923
0.1430
−0.1841
1.19E−15
0.3271


541
positive regulation of epithelial cell migration
1.66E−19
5
0.2000
0.0793
−0.5088
9.28E−19
0.5880


542
chemoattractant activity
2.38E−57
5
0.2174
0.1378
−0.4689
4.47E−56
0.6067


543
positive chemotaxis
8.98E−18
8
0.2424
0.0035
−0.5299
4.61E−17
0.5334


544
toll-like receptor 10 signaling pathway
5.75E−23
15
0.2273
0.1128
−0.2384
3.96E−22
0.3512


545
T cell receptor signaling pathway
1.10E−06
15
0.1163
0.0726
−0.1098
2.57E−06
0.1825


546
early endosome to late endosome transport
1.73E−11
4
0.2353
0.0218
0.1553
6.04E−11
−0.1335


547
clathrin coat
2.12E−19
4
0.2857
0.1426
−0.1375
1.18E−18
0.2801


548
platelet degranulation
2.11E−36
23
0.2805
0.0247
−0.2830
2.29E−35
0.3077


549
calcium-mediated signaling using intracellular calcium source
1.84E−07
3
0.2143
0.2178
0.6084
4.66E−07
−0.3906


550
negative regulation of DNA damage response, signal transduction by p53
1.03E−12
4
0.2857
0.2809
−0.0904
3.99E−12
0.3713



class mediator


551
regulation of phosphoprotein phosphatase activity
7.57E−11
4
0.2500
0.1542
0.5613
2.47E−10
−0.4071


552
positive regulation of calcium ion import
8.18E−10
3
0.1875
0.3561
−0.1168
2.50E−09
0.4729


553
intrinsic apoptotic signaling pathway in response to endoplasmic
2.60E−07
5
0.1136
0.0024
0.3731
6.45E−07
−0.3706



reticulum stress


554
protein N-linked glycosylation via asparagine
1.18E−39
26
0.2653
0.0140
0.2756
1.37E−38
−0.2616


555
protein destabilization
1.92E−06
3
0.1200
0.1061
0.3400
4.43E−06
−0.2339


556
glucose 6-phosphate metabolic process
2.44E−57
4
0.2500
0.0972
−0.5205
4.52E−56
0.6177


557
regulation of nitric-oxide synthase activity
3.14E−26
5
0.2381
0.1766
−0.3690
2.46E−25
0.5456


558
3-hydroxyacyl-CoA dehydrogenase activity
2.64E−42
5
0.5000
0.0480
0.4523
3.30E−41
−0.4042


559
stress-activated MAPK cascade
2.74E−12
12
0.2034
0.0882
−0.2384
1.02E−11
0.3266


560
cytokine production
3.81E−45
3
0.0698
0.2413
−0.5954
5.18E−44
0.8367


561
positive regulation of heart rate
1.01E−07
3
0.1765
−0.0014
0.4307
2.60E−07
−0.4321


562
endoplasmic reticulum-Golgi intermediate compartment
1.26E−42
24
0.4068
−0.0246
0.3250
1.58E−41
−0.3496


563
COPII vesicle coating
8.84E−18
6
0.2857
0.0762
0.7531
4.55E−17
−0.6769


564
innate immune response in mucosa
3.36E−06
5
0.2500
0.0207
0.2668
7.52E−06
−0.2460


565
mRNA 3′-end processing
7.51E−08
27
0.6279
0.0236
0.1923
1.95E−07
−0.1687


566
positive regulation of actin filament depolymerization
5.82E−68
4
0.3333
0.3196
−0.8686
1.43E−66
1.1882


567
enzyme inhibitor activity
3.13E−06
5
0.1351
−0.0511
0.1614
7.05E−06
−0.2125


568
superoxide dismutase activity
7.26E−10
3
0.1875
0.1272
−0.2807
2.22E−09
0.4079


569
V(D)J recombination
1.13E−16
4
0.3077
0.0288
−0.2561
5.42E−16
0.2849


570
ankyrin binding
7.80E−08
8
0.3478
0.0428
0.2823
2.02E−07
−0.2395


571
ATP-dependent chromatin remodeling
4.46E−06
14
0.5600
−0.0526
0.0623
9.84E−06
−0.1149


572
oligosaccharyltransferase complex
2.91E−17
6
0.3529
0.0258
0.3661
1.43E−16
−0.3403


573
purine ribonucleoside monophosphate biosynthetic process
5.68E−30
10
0.7143
0.0336
−0.2795
5.06E−29
0.3131


574
chromatin organization
4.28E−13
25
0.2066
0.0119
0.1804
1.70E−12
−0.1685


575
activation of cysteine-type endopeptidase activity involved in apoptotic
3.16E−09
3
0.2500
0.2076
0.4005
9.30E−09
−0.1929



process by cytochrome c


576
mannose binding
4.57E−12
6
0.2500
0.0070
0.3630
1.67E−11
−0.3560


577
chaperone-mediated protein complex assembly
3.97E−13
8
0.5000
0.0984
−0.1251
1.58E−12
0.2235


578
neutral amino acid transmembrane transporter activity
2.61E−08
3
0.2308
0.1580
0.1654
7.08E−08
−0.3234


579
intracellular estrogen receptor signaling pathway
2.51E−10
5
0.2500
−0.0274
0.3119
7.91E−10
−0.3394


580
cell adhesion molecule binding
4.85E−07
6
0.1277
−0.0786
−0.3765
1.17E−06
0.2978


581
natural killer cell mediated cytotoxicity
8.13E−83
5
0.2632
0.2181
−0.8636
2.91E−81
1.0816


582
inclusion body
9.03E−11
4
0.2105
−0.0725
−0.3216
2.95E−10
0.2491


583
mitochondrial nucleoid
5.92E−94
23
0.5610
0.0572
0.3425
3.01E−92
0.2853


584
negative regulation of nuclear mRNA splicing, via spliceosome
7.05E−32
12
0.7500
0.0118
0.3004
6.50E−31
−0.2885


585
MHC class I protein binding
1.53E−46
4
0.2667
0.0942
−0.1965
2.25E−45
0.2907


586
fatty acid binding
1.33E−31
4
0.2222
0.0805
−0.4035
1.22E−30
0.4841


587
negative regulation of epidermal growth factor receptor signaling
4.37E−23
13
0.2889
0.0799
−0.2671
3.05E−22
0.3471



pathway


588
negative regulation of proteasomal ubiquitin-dependent protein catabolic
1.70E−58
5
0.2778
−0.1116
−0.9291
3.31E−57
0.8174



process


589
RNA-induced silencing complex
9.07E−07
4
0.2222
0.1843
−0.1633
2.13E−06
0.3477


590
insulin-like growth factor receptor binding
1.96E−26
3
0.1579
0.1049
−0.6143
1.56E−25
0.7192


591
FK506 binding
7.64E−07
7
0.3889
−0.0333
−0.2172
1.81E−06
0.1839


592
AU-rich element binding
1.07E−09
6
0.3750
−0.0208
0.2254
3.24E−09
−0.2462


593
toll-like receptor 4 signaling pathway
1.84E−15
17
0.1753
0.0750
−0.2384
8.26E−15
0.3134


594
de novo' posttranslational protein folding
 7.88E−127
27
0.6923
0.0090
−0.3270
 5.40E−125
0.3360


595
glycerophospholipid biosynthetic process
1.33E−22
11
0.1209
0.0842
0.5178
9.04E−22
−0.4336


596
cellular respiration
3.63E−08
3
0.1667
0.1375
0.3671
9.67E−08
−0.2297


597
monosaccharide binding
7.64E−34
3
0.2000
−0.0313
−0.4969
7.57E−33
0.4657


598
negative regulation of androgen receptor signaling pathway
2.89E−24
3
0.2143
0.1146
0.5411
2.15E−23
−0.4265


599
toll-like receptor 9 signaling pathway
5.75E−23
15
0.2055
0.1128
−0.2384
3.96E−22
0.3512


600
positive regulation of binding
1.10E−06
3
0.1304
0.0814
−0.2157
2.57E−06
0.2970


601
protein refolding
3.60E−21
9
0.6000
0.0992
−0.2652
2.26E−20
0.3644


602
GTP-dependent protein binding
3.17E−09
8
0.4211
−0.0394
0.4241
9.34E−09
−0.4635


603
DNA-(apurinic or apyrimidinic site) lyase activity
4.59E−17
3
0.1579
0.0633
−0.2605
2.25E−16
0.3237


604
proline-rich region binding
1.37E−16
5
0.2174
0.0403
−0.4542
6.53E−16
0.4945


605
potassium ion binding
5.09E−16
3
0.1579
0.1615
−0.3096
2.33E−15
0.4712


606
RNA polymerase II repressing transcription factor binding
2.71E−05
5
0.1429
−0.0406
0.1277
5.54E−05
−0.1683


607
protein K63-linked ubiquitination
7.18E−19
4
0.1333
0.1562
−0.3582
3.86E−18
0.5144


608
mitochondrial inner membrane presequence translocase complex
8.99E−22
3
0.1304
0.1451
0.5932
5.90E−21
−0.4481


609
ameboidal cell migration
5.03E−19
3
0.1875
−0.0162
−0.6165
2.71E−18
0.6003


610
mRNA stabilization
1.95E−09
4
0.3077
0.0127
0.2751
5.83E−09
0.2878


611
nuclear-transcribed mRNA catabolic process, nonsense-mediated decay
0
95
0.7983
0.0689
−0.5303
0
0.5993


612
actin-dependent ATPase activity
1.89E−20
6
0.5000
0.1090
−0.1451
1.11E−19
0.2541


613
snRNA binding
9.69E−10
3
0.1579
−0.0265
0.2451
2.95E−09
−0.2717


614
small nuclear ribonucleoprotein complex
1.77E−18
11
0.5500
−0.0379
0.2145
9.34E−18
−0.2524


615
Golgi to plasma membrane protein transport
4.93E−06
5
0.2000
0.1017
0.3734
1.08E−05
−0.2716


616
positive regulation of cell cycle arrest
1.01E−06
3
0.1579
−0.0066
0.3840
2.36E−06
0.3906


617
RAGE receptor binding
4.09E−08
5
0.3846
−0.2398
0.5446
1.08E−07
0.3048


618
Cajal body
2.11E−13
19
0.4222
0.0507
0.2010
8.58E−13
0.1503


619
transferrin transport
2.70E−05
13
0.3939
0.0785
−0.0243
5.50E−05
0.1027


620
regulated secretory pathway
4.42E−33
3
0.2308
0.0000
0.4514
4.20E−32
−0.4514


621
mitotic spindle organization
1.01E−22
11
0.6471
0.0613
−0.3467
6.91E−22
0.4080


622
nucleobase-containing small molecule metabolic process
9.97E−12
37
0.4744
−0.0085
−0.1280
3.56E−11
0.1195


623
toll-like receptor 2 signaling pathway
3.97E−23
16
0.2192
0.1129
−0.2384
2.79E−22
0.3513


624
TRIF-dependent toll-like receptor signaling pathway
2.19E−08
13
0.1711
0.0883
−0.1830
5.97E−08
0.2713


625
toll-like receptor TLR1:TLR2 signaling pathway
3.97E−23
16
0.2254
0.1129
−0.2384
2.79E−22
0.3513


626
toll-like receptor TLR6:TLR2 signaling pathway
3.97E−23
16
0.2254
0.1129
−0.2384
2.79E−22
0.3513


627
platelet alpha granule lumen
1.65E−10
6
0.1250
0.0331
−0.4666
5.28E−10
0.4997


628
anatomical structure morphogenesis
2.58E−06
12
0.1304
0.1693
−0.1574
5.82E−06
0.3267


629
endocytic vesicle membrane
3.37E−12
9
0.1343
0.0869
−0.3210
1.25E−11
0.4079


630
establishment of endothelial barrier
7.09E−08
4
0.2667
−0.0493
−0.1888
1.85E−07
0.1395


631
translation factor activity, nucleic acid binding
2.73E−66
12
0.5000
0.0201
−0.4813
6.24E−65
0.5014


632
lysosomal lumen
2.26E−12
16
0.2254
−0.0233
0.2468
8.47E−12
−0.2701


633
regulation of neuron differentiation
1.39E−11
4
0.2105
0.0224
−0.4443
4.90E−11
0.4666


634
RNA splicing, via transesterification reactions
1.99E−09
15
0.6000
−0.0025
0.1758
5.93E−09
−0.1783


635
nucleobase-containing small molecule interconversion
1.05E−05
13
0.7222
0.0100
−0.0867
2.24E−05
0.0967


636
protein K11-linked ubiquitination
2.23E−30
4
0.1538
0.1179
−0.5181
2.01E−29
0.6360


637
vitamin metabolic process
7.29E−15
17
0.1977
0.1952
−0.1572
3.20E−14
0.3524


638
water-soluble vitamin metabolic process
7.29E−15
17
0.2152
0.1952
−0.1572
3.20E−14
0.3524


639
generation of precursor metabolites and energy
6.60E−43
13
0.2500
0.0888
0.4378
8.45E−42
−0.3491


640
regulation of interferon-gamma-mediated signaling pathway
2.34E−16
7
0.4375
−0.0797
−0.5310
1.09E−15
0.4513


641
regulation of type I interferon-mediated signaling pathway
1.89E−17
5
0.1852
−0.0785
−0.5369
9.44E−17
0.4584


642
COPI coating of Golgi vesicle
1.85E−07
12
0.9231
0.0015
0.1628
4.66E−07
−0.1613


643
activation of signaling protein activity involved in unfolded protein
7.12E−35
21
0.3231
0.0183
0.2687
7.19E−34
−0.2503



response


644
S100 protein binding
2.31E−46
8
0.7273
0.1083
−0.3835
3.37E−45
0.4918


645
response to unfolded protein
2.22E−26
19
0.3800
0.0547
−0.1920
1.75E−25
0.2467


646
toll-like receptor 5 signaling pathway
5.75E−23
15
0.2308
0.1128
−0.2384
3.96E−22
0.3512


647
nuclear telomere cap complex
5.87E−06
3
0.2500
0.1305
0.3069
1.28E−05
−0.1764


648
monocyte chemotaxis
3.08E−58
3
0.1765
0.2268
−0.3659
5.92E−57
0.5927


649
nucleotide-binding domain, leucine rich repeat containing receptor
2.63E−56
9
0.2143
0.0098
−0.4285
4.81E−55
0.4383



signaling pathway


650
regulation of transcription from RNA polymerase II promoter in response
5.22E−18
5
0.1923
0.1116
−0.2351
2.72E−17
0.3467



to hypoxia


651
branched chain family amino acid catabolic process
1.16E−12
9
0.5000
−0.0376
0.2211
4.47E−12
−0.2587


652
IkappaB kinase complex
2.15E−11
4
0.3636
−0.0952
0.2555
7.44E−11
−0.3507


653
nucleotide-binding oligomerization domain containing signaling pathway
1.72E−45
4
0.1600
0.1392
−0.4623
2.40E−44
0.6016


654
protein transmembrane transport
2.22E−06
5
0.4167
0.0503
0.3942
5.04E−06
−0.3439


655
striated muscle contraction
9.16E−51
3
0.2000
0.1439
−0.8200
1.47E−49
0.9639


656
actin filament capping
4.49E−05
6
0.3750
0.0840
−0.1424
8.97E−05
0.2264


657
Set1C/COMPASS complex
1.23E−05
3
0.3333
−0.1002
0.2421
2.57E−05
−0.3423


658
endoplasmic reticulum-Golgi intermediate compartment membrane
1.41E−26
10
0.3448
0.0715
0.4562
1.14E−25
−0.3847


659
muscle filament sliding
1.12E−05
8
0.2105
0.1088
−0.0931
2.37E−05
0.2019


660
interaction with host
9.25E−09
10
0.2941
0.1047
−0.0943
2.63E−08
0.1990


661
mitotic nuclear envelope disassembly
4.71E−16
19
0.5135
0.1425
0.4297
2.16E−15
−0.2872


662
U12-type spliceosomal complex
1.38E−05
16
0.6667
−0.0146
0.1053
2.88E−05
−0.1199


663
site of double-strand break
2.36E−06
5
0.3125
0.0194
0.1003
5.33E−06
−0.0809


664
catalytic step 2 spliceosome
3.96E−67
55
0.6875
0.0000
0.2034
9.45E−66
−0.2034


665
cellular aldehyde metabolic process
7.45E−09
4
0.3333
0.1663
−0.1128
2.14E−08
0.2790


666
positive regulation of protein insertion into mitochondrial membrane
 6.37E−146
11
0.4074
0.1797
−0.5360
 5.57E−144
0.7157



involved in apoptotic signaling pathway


667
termination of RNA polymerase II transcription
7.51E−08
27
0.5870
0.0236
0.1923
1.95E−07
−0.1687


668
low-density lipoprotein particle receptor binding
2.50E−21
3
0.2308
0.0657
0.4366
1.59E−20
−0.3709


669
cytoskeletal anchoring at plasma membrane
1.87E−05
5
0.3571
−0.0337
−0.1178
3.89E−05
0.0841


670
antibacterial humoral response
4.13E−06
6
0.2609
0.0225
0.2650
9.14E−06
−0.2425


671
phagocytic vesicle
2.18E−10
9
0.3103
−0.0844
0.2957
6.92E−10
−0.3801


672
Sin3 complex
2.56E−07
4
0.3333
−0.0578
0.1446
6.34E−07
−0.2025


673
DNA strand elongation involved in DNA replication
6.80E−09
19
0.6129
−0.0807
0.0730
1.95E−08
−0.1537


674
mitotic nuclear envelope reassembly
4.97E−14
7
0.7000
0.1580
0.4892
2.04E−13
−0.3312


675
NADPH binding
3.13E−07
5
0.5000
0.2438
−0.1276
7.70E−07
0.3714


676
mitochondrial proton-transporting ATP synthase complex
6.76E−72
16
0.7619
0.0830
0.3891
1.94E−70
−0.3061


677
mitochondrial ATP synthesis coupled proton transport
4.83E−67
14
0.8750
0.0893
0.3971
1.14E−65
−0.3079


678
clathrin-coated endocytic vesicle membrane
2.64E−13
7
0.1628
0.1477
−0.1274
1.07E−12
0.2751


679
virus-host interaction
3.75E−19
4
0.3333
0.0776
0.4858
2.05E−18
−0.4083


680
pantothenate metabolic process
5.91E−68
3
0.2500
0.5473
−0.4194
1.43E−66
0.9668


681
lamellipodium membrane
2.40E−22
4
0.2500
0.2027
−0.4494
1.62E−21
0.6521


682
glutamate metabolic process
9.23E−10
3
0.2500
0.1404
0.4504
2.81E−09
−0.3099


683
positive regulation of blood vessel endothelial cell migration
5.55E−21
3
0.1765
0.1120
−0.4174
3.41E−20
0.5293


684
MHC class II protein complex binding
 2.90E−231
7
0.4118
0.1567
−0.6323
 3.26E−229
0.7891


685
viral transcription
0
76
0.9268
0.0748
−0.5495
0
0.6243


686
3′-UTR-mediated mRNA stabilization
5.32E−14
7
0.5833
−0.0525
0.2277
2.18E−13
−0.2802


687
ribosomal small subunit biogenesis
0
12
0.9231
0.1003
−0.8253
0
0.9256


688
RNA polymerase II transcription regulatory region sequence-specific
1.44E−09
4
0.4444
0.1225
0.5360
4.33E−09
−0.4135



DNA binding transcription factor activity involved in negative regulation



of transcription









REFERENCES



  • 1. Regev A, Teichmann S A, Lander E S, Amit I, Benoist C, Birney E, Bodenmiller B, Campbell P, Carninci P, Clatworthy M, Clevers H, Deplancke B, Dunham I, Eberwine J, Eils R, Enard W, Farmer A, Fugger L, Gottgens B, Hacohen N, Haniffa M, Hemberg M, Kim S, Klenerman P, Kriegstein A, Lein E, Linnarsson S, Lundberg E, Lundeberg J, Majumder P, Marioni J C, Merad M, Mhlanga M, Nawijn M, Netea M, Nolan G, Pe′er D, Phillipakis A, Ponting C P, Quake S, Reik W, Rozenblatt-Rosen O, Sanes J, Satija R, Schumacher T N, Shalek A, Shapiro E, Sharma P, Shin J W, Stegle O, Stratton M, Stubbington M J T, Theis F J, Uhlen M, van Oudenaarden A, Wagner A, Watt F, Weissman J, Wold B, Xavier R, Yosef N; Human Cell Atlas Meeting Participants. The Human Cell Atlas. Elife. 2017 Dec. 5; 6:e27041.

  • 2. Specht H, Slavov N. Transformative Opportunities for Single-Cell Proteomics. J Proteome Res. 2018 Aug. 3; 17(8):2565-2571.

  • 3. Ziegenhain C, Vieth B, Parekh S, Reinius B, Guillaumet-Adkins A, Smets M, Leonhardt H, Heyn H, Hellmann I, Enard W. Comparative Analysis of Single-Cell RNA Sequencing Methods. Mol Cell. 2017 Feb. 16; 65(4):631-643.e4.

  • 4. Slavov N. Unpicking the proteome in single cells. Science. 2020 Jan. 31; 367(6477):512-513.

  • 5 Slavov N. Learning from natural variation across the proteomes of single cells. PLOS Biol. 2022 Jan. 5; 20(1):e3001512.

  • 6. Shaffer S M, Dunagin M C, Torborg S R, Torre E A, Emert B, Krepler C, Beqiri M, Sproesser K, Brafford P A, Xiao M, Eggan E, Anastopoulos I N, Vargas-Garcia C A, Singh A, Nathanson K L, Herlyn M, Raj A. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature. 2017 Jun. 15; 546(7658):431-435.

  • 7. Emert B L, Cote C J, Torre E A, Dardani I P, Jiang C L, Jain N, Shaffer S M, Raj A. Variability within rare cell states enables multiple paths toward drug resistance. Nat Biotechnol. 2021 July;39(7):865-876.

  • 8. Mahdessian D, Cesnik A J, Gnann C, Danielsson F, Stenström L, Arif M, Zhang C, Le T, Johansson F, Shutten R, Bäckström A, Axelsson U, Thul P, Cho N H, Carja O, Uhlén M, Mardinoglu A, Stadler C, Lindskog C, Ayoglu B, Leonetti M D, Pontén F, Sullivan D P, Lundberg E. Spatiotemporal dissection of the cell cycle with single-cell proteogenomics. Nature. 2021 February;590(7847):649-654.

  • 9. Slavov N. Scaling Up Single-Cell Proteomics. Mol Cell Proteomics. 2022 Jan;21(1): 100179.

  • 10. Slavov N. Single-cell protein analysis by mass spectrometry. Curr Opin Chem Biol. 2021 Feb;60:1-9.

  • 11. Vanderaa C, Gatto L. Replication of single-cell proteomics data reveals important computational challenges. Expert Rev Proteomics. 2021 October; 18(10):835-843.

  • 12. Kelly R T. Single-cell Proteomics: Progress and Prospects. Mol Cell Proteomics. 2020 Nov; 19(11):1739-1748.

  • 13. Specht H, Emmott E, Petelski A A, Huffman R G, Perlman D H, Serra M, Kharchenko P, Koller A, Slavov N. Single-cell proteomic and transcriptomic analysis of macrophage heterogeneity using SCOPE2. Genome Biol. 2021 Jan. 27; 22(1):50.

  • 14. Klein A M, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L, Weitz D A, Kirschner M W. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell. 2015 May 21;161(5):1187-1201.

  • 15. Macosko E Z, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas A R, Kamitaki N, Martersteck E M, Trombetta J J, Weitz D A, Sanes J R, Shalek A K, Regev A, McCarroll S A. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell. 2015 May 21; 161(5): 1202-1214.

  • 16. Harrison Specht, Guillaume Harmange, David H. Perlman, Edward Emmott, Zachary Niziolek, Bogdan Budnik, Nikolai Slavov. Automated sample preparation for high-throughput single-cell proteomics. bioRxiv. Aug. 25, 2018. 399774 (doi.org/10.1101/399774)

  • 17. Petelski A A, Emmott E, Leduc A, Huffman R G, Specht H, Perlman D H, Slavov N. Multiplexed single-cell proteomics using SCOPE2. Nat Protoc. 2021 December; 16(12):5398-5425.

  • 18. Marx V. A dream of single-cell proteomics. Nat Methods. 2019 September; 16(9):809-812.

  • 19. R Gray Huffman, Andrew Leduc, Christoph Wichmann, Marco di Gioia, Francesco Borriello, Harrison Specht, Jason Derks, Saad Khan, Edward Emmott, Aleksandra A. Petelski, David H Perlman, Jürgen Cox, Ivan Zanoni, Nikolai Slavov. Prioritized single-cell proteomics reveals molecular and functional polarization across primary macrophages, bioRxiv Mar. 16, 2022. 484655 (doi.org/10.1101/2022.03.16.484655).

  • 20. Giurgiu M, Reinhard J, Brauner B, Dunger-Kaltenbach I, Fobo G, Frishman G, Montrone C, Ruepp A. CORUM: the comprehensive resource of mammalian protein complexes-2019. Nucleic Acids Res. 2019 Jan. 8; 47(D1):D559-D563.

  • 21. Fallahi-Sichani M, Becker V, Izar B, Baker G J, Lin J R, Boswell S A, Shah P, Rotem A, Garraway L A, Sorger P K. Adaptive resistance of melanoma cells to RAF inhibition via reversible induction of a slowly dividing de-differentiated state. Mol Syst Biol. 2017 Jan. 9; 13(1):905.

  • 22. Dahlén E, Veitonmäki N, Norlén P. Bispecific antibodies in cancer immunotherapy. Ther Adv Vaccines Immunother. 2018 February;6(1):3-17.

  • 23. Gebauer F, Wicklein D, Horst J, Sundermann P, Maar H, Streichert T, Tachezy M, Izbicki J R, Bockhorn M, Schumacher U. Carcinoembryonic antigen-related cell adhesion molecules (CEACAM) 1, 5 and 6 as biomarkers in pancreatic cancer. PLOS One. 2014 Nov. 19; 9(11):e113023.

  • 24. Slavov N. Increasing proteomics throughput. Nat Biotechnol. 2021 July;39(7):809-810.

  • 25. Hughes C S, Foehr S, Garfield D A, Furlong E E, Steinmetz L M, Krijgsveld J. Ultrasensitive proteome analysis using paramagnetic bead technology. Mol Syst Biol. 2014 Oct. 30; 10(10):757.

  • 26. Hughes C S, Moggridge S, Müller T, Sorensen P H, Morin G B, Krijgsveld J. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments. Nat Protoc. 2019 Jan; 14(1):68-85.

  • 27. Kulak N A, Pichler G, Paron I, Nagaraj N, Mann M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat Methods. 2014 Mar; 11(3):319-24.

  • 28. Cooper S. The synchronization manifesto: a critique of whole-culture synchronization. FEBS J. 2019 December; 286(23):4650-4656.

  • 29. Specht H, Slavov N. Optimizing Accuracy and Depth of Protein Quantification in Experiments Using Isobaric Carriers. J Proteome Res. 2021 Jan. 1; 20(1):880-887.

  • 30. Charif, D. & Lobry, J. SeqinR 1.0-2: a contributed package to the R project for statistical computing devoted to biological sequences retrieval and analysis. Biological and Medical Physics, Biomedical Engineering (eds Bastolla, U., Porto, M., Roman, H. & Vendruscolo, M.) ISBN: 978-3-540-35305-8, 207-232 (2007).

  • 31. Jason Derks, Andrew Leduc, R. Gray Huffman, Harrison Specht, Markus Ralser, Vadim Demichev, Nikolai Slavov. Increasing the throughput of sensitive proteomics by plexDIA. bioRxiv Nov. 3, 2021. 467007 (doi.org/10.1101/2021.11.03.467007).

  • 32. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol. 2008 Dec;26(12):1367-72.

  • 33. Cox J, Neuhauser N, Michalski A, Scheltema R A, Olsen J V, Mann M. Andromeda: a peptide search engine integrated into the MaxQuant environment. J Proteome Res. 2011 Apr. 1; 10(4):1794-805.

  • 34. Tyanova S, Temu T, Cox J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat Protoc. 2016 December; 11(12):2301-2319.

  • 35. Demichev V, Messner C B, Vernardis S I, Lilley K S, Ralser M. DIA-NN: neural networks and interference correction enable deep proteome coverage in high throughput. Nat Methods. 2020 January; 17(1): 41-44.

  • 36. Vanderaa C, Gatto L. Replication of single-cell proteomics data reveals important computational challenges. Expert Rev Proteomics. 2021 October; 18(10):835-843. (Previously: Christophe Vanderaa, Laurent Gatto. Utilizing Scp for the analysis and replication of single-cell proteomics data. bioRxiv. Apr. 12, 2021. 439408 (doi.org/10.1101/2021.04.12.439408).

  • 37. Vanderaa, C. & Gatto, L. Mass Spectrometry-Based Single-Cell Proteomics Data Analysis. Bioconductor, 10.18129/B9.bioc.scp (2020).

  • 38. Specht H, Emmott E, Petelski A A, Huffman R G, Perlman D H, Serra M, Kharchenko P, Koller A, Slavov N. Single-cell proteomic and transcriptomic analysis of macrophage heterogeneity using SCOPE2. Genome Biol. 2021 Jan. 27; 22(1):50.

  • 39. Franks A, Airoldi E, Slavov N. Post-transcriptional regulation across human tissues. PLOS Comput Biol. 2017 May 8; 13(5):e1005535.

  • 40. Gray Huffman, Harrison Specht, Albert Chen, Nikolai Slavov. DO-MS: Data-Driven Optimization of Mass Spectrometry Methods bioRxiv. Jan. 6, 2019. 512152 (doi.org/10.1101/512152).

  • 41. Chen A T, Franks A, Slavov N. DART-ID increases single-cell proteome coverage. PLOS Comput Biol. 2019 Jul. 1; 15(7):e1007082 (previously bioRxiv. Aug. 23, 2018. 399121 (https://doi.org/10.1101/399121)).

  • 42. Budnik B, Levy E, Harmange G, Slavov N. SCOPE-MS: mass spectrometry of single mammalian cells quantifies proteome heterogeneity during cell differentiation. Genome Biol. 2018 Oct. 22; 19(1): 161. (previously bioRxiv. Jan. 24, 2017. 102681 (doi.org/10.1101/102681)).



The teachings of all patents, published applications and references cited herein are incorporated by reference in their entirety.


While example embodiments have been particularly shown and described, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the embodiments encompassed by the appended claims.

Claims
  • 1. A method of forming a single-cell proteomic sample, said method comprising: a) dispensing n droplets of lysis buffer onto a substantially planar solid surface, wherein n≥2;b) dispensing a single cell into each of the n droplets of lysis buffer to produce n droplets, each comprising a lysed single cell;c) dispensing digestion buffer into each of the n droplets to digest proteins from each lysed single cell to produce n droplets comprising peptides;d) dispensing a chemical tag into each of the n droplets comprising the peptides to produce labeled peptides, wherein at least one droplet of the n droplets receives a different chemical tag from at least one other droplet of the n droplets, thereby enabling the labeled peptides in the at least one droplet to be distinguishable from the labeled peptides in the at least one other droplet; ande) applying a fluid to merge at least a subset of the n droplets into a combined droplet on the substantially planar surface, thereby combining the labeled peptides to form a single-cell proteomic sample.
  • 2. The method of claim 1, wherein each of the n droplets in step a), b), c), and/or d) has a volume of about 25 nanoliters (nl) or less.
  • 3. The method of claim 1, wherein each of the n droplets in step a), b), c) and d) has a volume of about 25 nanoliters (nl) or less.
  • 4. The method of claim 1, wherein the substantially planar solid surface is provided by a uniform glass slide.
  • 5. The method of claim 1, wherein the substantially planar solid surface is etched with a geometric pattern.
  • 6. The method of claim 1, wherein the substantially planar solid surface is fluorocarbon-coated.
  • 7. The method of claim 1, wherein n is ≥10.
  • 8. The method of claim 1, wherein the lysis buffer comprises about 4-8 nanoliters of 90-100% dimethyl sulfoxide (DMSO).
  • 9. The method of claim 1, wherein step b) comprises dispensing the single cell in a cell suspension buffer with a volume of about 100-1,000 picoliters.
  • 10. The method of claim 9, wherein step b) comprises dispensing the single cell in a cell suspension buffer with a volume of about 300 picoliters.
  • 11. The method of claim 1, wherein the single cell is lysed in a total volume of about 4-10 nl for about 10-20 minutes.
  • 12. The method of claim 1, wherein step c) comprises: dispensing about 15-25 nl of about 120 ng/μl trypsin to each of the n droplets; anddigesting the proteins from each lysed single cell at about 1ºC above the dew point and a relative humidity of about 75% for about 4-5 hours.
  • 13. The method of claim 1, wherein the chemical tag comprises a “light” version of TMT label reagents dissolved in DMSO.
  • 14. The method of claim 1, wherein the chemical tag comprises a “heavy” version of TMT label reagents dissolved in DMSO.
  • 15. The method of claim 1, wherein step d) comprises: dispensing about 18-22 nl of a chemical tag into each of the n droplets comprising the peptides; and enabling the chemical tag to react with the peptides at room temperature and a relative humidity of about 75% for about 1 hour to produce the labeled peptides.
  • 16. The method of claim 1, wherein the fluid is water.
  • 17. The method of claim 1, wherein the fluid has a volume of about 1 μl.
  • 18. The method of claim 1, wherein steps a) to e) are repeated at least once to form two or more single-cell proteomic samples on the substantially planar solid surface.
  • 19. The method of claim 18, wherein at least 100 droplets of lysis buffer are dispensed onto the substantially planar solid surface.
  • 20. The method of claim 19, wherein at least 500-3,000 droplets of lysis buffer are dispensed onto the substantially planar solid surface.
  • 21. The method of claim 18, wherein the two or more single-cell proteomic samples comprises peptides from at least 100 cells.
  • 22. The method of claim 21, wherein the two or more single-cell proteomic samples comprises peptides from about 100-10,000 cells.
  • 23. The method of claim 1, wherein each droplet of the n droplets receives a unique chemical tag, thereby enabling the labeled peptides in each droplet to be distinguishable from the labeled peptides in each other droplet.
  • 24. A method of performing a proteomic analysis comprising analyzing a single-cell proteomic sample formed by the method of claim 1.
  • 25. The method of claim 24, wherein the analyzing comprises identifying and/or quantifying protein covariation across the single cells.
  • 26. A single-cell proteomic sample formed by the method of claim 1.
RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 63/179,035, filed on Apr. 23, 2021 and U.S. Provisional Application No. 63/179,184, filed on Apr. 23, 2021. The entire teachings of the above applications are incorporated herein by reference.

GOVERNMENT SUPPORT

This invention was made with government support under Grant No. GM123497 awarded by the National Institutes of Health. The government has certain rights in the invention. The subject matter disclosed in this application was developed, and the claimed invention was made by, or on behalf of, one or more parties to a joint Research Agreement that was in effect on or before the effective filing date of the claimed invention. The parties to the Joint Research Agreement are as follows Northeastern University and SCIENION GmbH.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2022/071883 4/22/2022 WO
Provisional Applications (2)
Number Date Country
63179035 Apr 2021 US
63179184 Apr 2021 US