Methods for reprograming non-hepatocyte cells into hepatocyte cells

Information

  • Patent Grant
  • 11613735
  • Patent Number
    11,613,735
  • Date Filed
    Wednesday, February 4, 2015
    9 years ago
  • Date Issued
    Tuesday, March 28, 2023
    a year ago
Abstract
A method for inducing reprogramming of a cell of a first type which is not a non-hepatocyte (non-hepatocyte cell), into a cell with functional hepatic drug metabolizing and transporting capabilities, is disclosed. The non-hepatocyte is induced to express or overexpress hepatic fate conversion and maturation factors, cultured in somatic cell culture medium, hepatocyte cell culture medium and hepatocyte maturation medium for a sufficient period of time to convert the non-hepatocyte cell into a cell with hepatocyte-like properties. The iHeps induced according to the methods disclosed herein are functional induced hepatocytes (iHeps) in that they express I and II drug-metabolizing enzymes and phase III drug transporters and show superior drug metabolizing activity compared to iHeps obtained by prior art methods. The iHeps thus provide a cell resource for pharmaceutical applications.
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application a U.S. National stage entry of PCT/CN2015/072232, filed Feb. 4, 2015, which claims benefit of Chinese Provisional Application No. 201410048337.X, filed Feb. 12, 2014, incorporated herein by reference.


REFERENCE TO SEQUENCE LISTING

The Sequence Listing submitted as a text file named “GHBCL_101_ST25_txt,” created on Feb. 2, 2015, and having a size of 41,627 bytes is hereby incorporated by reference.


FIELD OF THE INVENTION

The present invention generally relates to use of hepatocyte fate conversion and maturation factors for reprogramming eukaryotic cells into hepatocyte cells.


BACKGROUND OF THE INVENTION

Functional human cell types are in high demand in the field of regenerative medicine and drug development. They show great potential for repairing or replacing diseased and damaged tissues and can be valuable tools for pharmaceutical applications. However, the application of functional human cell types in these areas is limited due to a shortage of donors (Castell et al., Expert Opin. Drug Metab. Toxicol. 2:183-212 (2006)). To solve this dilemma, novel strategies for generating functionally mature cells are in high demand. Recently, lineage reprogramming has emerged as an effective method for changing the fate of somatic cells (Vierbuchen and Wernig, Mol. Cell, 47: 827-838 (2012)). In principle, one cell type can be converted directly to the final mature state of another cell type and can bypass its intermediate states during lineage reprogramming. Consequently, functionally mature cells may be obtained using this strategy and may potentially provide a promising source of functional human cells.


Functional human hepatocytes are the most significant in vitro model for evaluating drug metabolism and are potentially widely applicable in pharmaceutical development. Because unacceptable metabolic and toxicity effects on the liver are largely responsible for the failure of new chemical entities in drug discovery (Baranczewski et al., Pharmacol. Rep., 58:453-472 (2006)), it is essential to use human hepatocytes, which serve as the closest in vitro model of human liver in assays of absorption, distribution, metabolism, excretion, and toxicity (ADME/Tox), to identify compounds that display favorable pharmacokinetics (Sahi et al., Curr. Drug Discov. Technol., 7:188-198 (2010)). Currently, primary human hepatocytes that are derived from individuals with different genetic backgrounds are frequently used in drug development, but the resulting diversity of genetic backgrounds hinders the reproducibility of the results obtained from pharmaceutical studies using these cells. Additionally, the scarcity of human liver donors greatly limits the use of primary human hepatocytes (Castell et al., Expert Opin. Drug Metab. Toxicol. 2:183-212 (2006)) and, as a result, alternative resources for human hepatocytes with a high reproducibility are urgently required for use in drug discovery.


Different strategies to generate functional hepatocytes have been studied. Human hepatocytes have been derived from human pluripotent stem cells by directed differentiation (Cai et al., Hepatology, 45:1229-1239 (2007); Ogawa et al., Development, 140:3285-3296 (2013); Takebe et al., Nature, 499:481-484 (2013); Zhao et al., Cell Res., 23:157-161 (2013)). This strategy has progressed quickly in recent years, although the immature phenotype of the cells derived from pluripotent stem cells remains a technological obstacle. In principle, fully functional hepatocytes are relatively difficult to obtain using this method, as the whole process involves multiple key steps that affect the final stage of hepatocyte formation. In contrast, lineage reprogramming allows the lineage conversion of a somatic cell without passing through an intermediate state. Although mouse hepatocytes have been transdifferentiated from fibroblasts (Huang et al., Nature, 475:386-389 2011; Sekiya and Suzuki, Nature, 475:390-393 (2011)), these cells still express several hepatoblast markers such as α-fetoprotein (AFP) and lack the expression of several key cytochrome P450 enzymes (CYPs) that are responsible for drug metabolism, suggesting a functionally immature phenotype for these cells (Willenbring, Cell Stem Cell, 9:89-91 (2011)).


There is therefore a need for a method inducing non-hepatocyte cells into functional induced hepatocytes that show improved hepatocyte functional activity, when compared to known induced hepatocytes.


It is therefore an object of the present invention to provide a method of inducing conversion of a non-hepatocyte cell, into an induced hepatocyte cell (iHep) with metabolic function.


It is also an object of the present invention to provide induced hepatic cells with metabolic function.


It is still an object of the present invention to provide a method using induced hepatocytes for drug development, bioartificial liver system and in vivo and in-vitro hepatic applications.


It is further an object of the present invention to provide kits for reprogramming a non-hepatocyte into an iHep.


SUMMARY OF THE INVENTION

A method for inducing reprogramming of a cell of a first type which is not a hepatocyte (i.e., non-hepatocyte cells), into a hepatocyte-like cell, as indicated by functional hepatic drug metabolizing and transporting capabilities, is disclosed. These cells are denoted herein as induced hepatocytes (iHeps). The non-hepatocyte is treated to upregulate hepatic fate conversion and maturation factors (“collectively, “Hepatocyte inducing factors”), cultured in somatic cell culture medium (transformation phase), expanded in hepatocyte cell culture medium (expansion phase) and further cultured in hepatocyte maturation medium (maturation phase) for a sufficient period of time to convert the cell into a cell with hepatocyte-like properties.


In a preferred embodiment, the non-hepatocyte cell is transformed to overexpress at least one of the following Hepatocyte inducing factors: Hepatocyte nuclear factor 1-alpha (HNF1A), Hepatocyte nuclear factor 4-alpha (HNF4A), and Hepatocyte nuclear factor 6-alpha (HNF6), Activating transcription factor 5 (ATF5), Prospero homeobox protein 1 (PROX1), and CCAAT/enhancer-binding protein alpha (CEBPA). In some embodiments the cell is transformed to express at least 2, at least 3, at least 4 or at least 5 of the hepatocyte inducing factors. In a preferred embodiment, the cell is transformed to overexpress all 6 Hepatocyte inducing factors. In some embodiments, the method further includes upregulating MYC, and/or downregulating p53 gene expression and/or protein activity. Non-hepatocytes (treated to upregulate hepatocyte inducing factors; and optionally upregulate MYC and optionally, downregulate p53) are then expanded in vitro to obtain iHeps. In one embodiment, transfected cells are cultured in somatic cell culture medium, for example, DMEM, for a period of at least 7 days, until about 80% confluence. The cells are then replated and expanded in hepatocyte cell culture medium (HCM) for about 15 to 30 days, preferably for about 18-30 days, and more preferably, for about 18 days, following which the cells are transferred into a hepatocyte maturation medium for about 5 days. Induced hepatocytes (iHeps) are obtained following this cell culture scheme.


The cells are identified as iheps, based on known structural and functional properties of hepatocytes.


Also disclosed are functional induced hepatocytes (iHeps). In a preferred embodiment, the induced hepatocytes are human induced hepatocytes (hiHeps). iHeps express at least one hepatocyte marker selected from the group consisting of albumin, Cytochrome P450 (Cyp)3A4, CYPB6, CYP1A2, CYP2C9, and CYP2C19. In a preferred embodiment, iHeps express at least two, three or four or five or six of CYPB6, CYP3A4, CYPB6, CYP1A2, CYP2C9, and CYP2C19.


Transplanted hiHeps repopulate up to 30% of the livers of Tet-uPA/Rag2−/−γc−/− mice and secrete more than 300 mg/ml human albumin in vivo. Thus human hepatocytes with drug metabolic function can be generated by lineage reprogramming, thus providing a cell resource for in vitro drug development and in vivo applications within the context of liver disease/failure.


Kits for inducing reprogramming of non-hepatocytes cells into iHeps are also disclosed. The kit includes factors which upregulate the Hepatocyte inducing factors disclosed herein, and optionally, factors which upregulate MYC and downregulate p53 gene expression and/or protein levels.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A is a bar graph showing gene expression analysis of ALB in F-HEPs, HEFs and 3H cells. n=2. FIG. 1B is a bar graph showing a quantitative comparison of the expression of hepatic transcription factors in 3H cells, fetal liver cells (FLCs), and F-HEPs. n=2. *p<0.05; **p<0.01; ***p<0.001. FIG. 1C is a bar graph showing gene expression analysis of liver-enriched transcription factors in 3H cells, FLCs and F-HEPs by qRT-PCR. n=2. FIG. 1D is a bar graph showing a quantitative analysis of the abundance of hepatic transcription factors in four individual F-HEPs. n=2. FIG. 1E is a schematic view of the hiHep reprogramming diagram. FIG. 1 F shows determination of the proliferation rate of the induced cells at different stages. Upper panel: MTT assay. Day 0 is set as the day when the induced cells were transferred to HCM (before p53 siRNA-GFP silence) or modified WEM (after p53 siRNA-GFP silence). Lower panel: Calculation of doubling time of the induced cells at the expansion stage (before p53 siRNA-GFP silence). Td, doubling time. FIG. 1G is a bar graph showing a quantitative analysis of ALBUMIN expression among hiHeps, HEFs, and F-HEPs. FIGS. 1H and 1I show reprogramming efficiency measured by flow cytometry analysis marked by ALB and AAT. n=3. APC, allophycocyanin. FIG. 1J is a bar graph showing a quantitative analysis of Albumin secretion among hiHeps, HEFs, and F-HEPs by ELISA. n=3. FIG. 1K shows the effect on the expression of hepatic functional genes after removal of individual factors detected by qRT-PCR. n=2. Data are presented as mean+/−s.d.



FIG. 2A shows endogenous gene expression analysis of hepatic transcription factors and fibroblast markers in hiHeps by RT-PCR. FIG. 2B shows the silence of exogenous genes detected by RT-PCR. Day 7, 7 days post infection. FIG. 2C shows relative expression of MYC during the hepatic conversion process measured by qRT-PCR. Day 7 and day 14, 7 and 14 days post infection. n=2.



FIGS. 3A-3C show a quantitative analysis of the expression of drug metabolic phase I (FIG. 3A) and phase II enzymes (FIG. 3B) and phase III transporters (FIG. 3C) in HEFs, HepG2 cells, ES-Heps, hiHeps, and F-HEPs. The relative expression of each gene was normalized to HEFs; if not detected, it was normalized to HepG2 cells. n=2. 1=HEFs; 2=HepG2 cells; 3=ES-Heps; 4=hiHeps; 5=F-Heps. FIG. 3D is a bar graph showing quantitative analysis of the expression of drug metabolic Phase II enzymes and Phase III transporters in HEFs, HepG2 cells, ES-Heps, hiHeps and F-HEPs. The relative expression for each gene was normalized to HEFs; if not detected, normalized to HepG2 cells. n=2. FIG. 3E is a bar graph showing quantitative comparison of phase I, phase II, phase III mRNA in hiHeps and HEFs to F-HEPs. FIG. 3F is a bar graph showing quantitative comparison of nuclear receptors mRNA in hiHeps to F-HEPs.



FIG. 4A shows the metabolic activities of CYP3A4 (3A4-T, testosterone; 3A4-M, midazolam), CYP1A2 (phenacetin), CYP2B6 (bupropion), CYP2C9 (diclofenac), and CYP2C19 [(S)-mephenytoin] in hiHeps, ES-Heps, F-HEPs1, F-HEPs2, HepG2 cells, and HEFs as determined by HPLC-MS. n=3. Two batches of freshly isolated primary human hepatocytes (F-HEPs1 and F-HEPs2) were applied as the positive control. The results are presented as pmol/min per million cells. Data are presented as mean±SD. FIG. 4B is a bar graph showing quantitative analysis of the fold-induction of the CYP3A4, CYP1A2 and CYP2B6 in hiHeps treated with different inducers. n=2. Rif, Rifampin; PB, Phenobarbital; ETOH, Ethanol; BNF, β-Naphthoflavone. FIG. 4C is a bar graph showing an analysis of the sensitivity of hiHeps to multiple model hepatotoxins. F-HEPs were used as the positive control. Data are presented as mean. n=3. FIG. 4D is a bar graph showing gene expression analysis of hepatic genes after hiHeps formation by qRT-PCR. The relative expression was normalized to that of day 0. Data are presented as mean+/−s.d.



FIG. 5A is a line graph showing the level of human albumin in mouse serum was monitored by ELISA.



FIG. 5 B is a bar graph comparing human ALB secretion in mouse serum among ES-Heps (n=16), hiHeps (n=5), and F-HEPs (n=6).



FIG. 5C shows flow cytometry analysis of the engraftment efficiencies of hiHeps. Mouse 1 and mouse 2 secreted human ALB at 267 and 313 ug/ml, respectively. HN, human nuclei; PE, phycoerythrin.





DETAILED DESCRIPTION OF THE INVENTION
I. Definitions

As used herein a “culture” means a population of cells grown in a medium and optionally passaged. A cell culture may be a primary culture (e.g., a culture that has not been passaged) or may be a secondary or subsequent culture (e.g., a population of cells which have been subcultured or passaged one or more times).


As used herein, “downregulation” or “downregulate” refers to the process by which a cell decreases the quantity and/or activity of a cellular component, for example, DNA, RNA or protein, in response to an external variable.


As used herein, “embryonic stem cell (ESC)-derived hepatocytes (ES-Heps)” refer to induced hepatocytes derived according to the methods disclosed in Zhao, et al., Cell Res., 23(1):157-161 (2013).


As used herein, “functional induced hepatocytes (iHeps)” refers to induced hepatocytes which show the activity of at least one of CYP3A4, CYP2C9, or CYP2C19, at levels 50% higher than the activity of the same enzyme in ES-Heps obtained from the same organism. The activity of the enzyme can be 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100% or more, higher than the activity in ES-Heps.


As used herein, the term “host cell” refers to non-hepatocytes eukaryotic cells into which a recombinant nucleotide, such as a vector, can be introduced.


The term “induced hepatocytes” (iHeps) as used herein refers to cells which are not naturally occurring hepatocytes, and which are artificially derived from non-hepatocyte cells.


The term “isolated” or “purified” when referring to hiHEPS means chemically induced pluripotent stem cells at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% free of contaminating cell types such as non-hepatocyte cells. The isolated iheps may also be substantially free of soluble, naturally occurring molecules.


The terms “oligonucleotide” and “polynucleotide” generally refer to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. Thus, for instance, polynucleotides as used herein refers to, among others, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions. The term “nucleic acid” or “nucleic acid sequence” also encompasses a polynucleotide as defined above.


In addition, polynucleotide as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules. One of the molecules of a triple-helical region often is an oligonucleotide.


As used herein, the term polynucleotide includes DNAs or RNAs as described above that contain one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein. Moreover, DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein.


The term “percent (%) sequence identity” is defined as the percentage of nucleotides or amino acids in a candidate sequence that are identical with the nucleotides or amino acids in a reference nucleic acid sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared can be determined by known methods.


For purposes herein, the % sequence identity of a given nucleotides or amino acids sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given sequence C that has or comprises a certain % sequence identity to, with, or against a given sequence D) is calculated as follows:

100 times the fraction W/Z,

where W is the number of nucleotides or amino acids scored as identical matches by the sequence alignment program in that program's alignment of C and D, and where Z is the total number of nucleotides or amino acids in D. It will be appreciated that where the length of sequence C is not equal to the length of sequence D, the % sequence identity of C to D will not equal the % sequence identity of D to C


As used herein, “transformed” and “transfected” encompass the introduction of a nucleic acid (e.g. a vector) into a cell by a number of techniques known in the art.


As used herein, a “vector” is a replicon, such as a plasmid, phage, or cosmid, into which another DNA segment may be inserted so as to bring about the replication of the inserted segment. The vectors described herein can be expression vectors.


As used herein, an “expression vector” is a vector that includes one or more expression control sequences.


“Reprogramming” as used herein refers to the conversion of a one specific cell type to another. For example, a cell that is not a hepatocyte cab be reprogrammed into a cell that is morphologically and functionally like a hepatocyte.


As used herein “treating a cell/cells” refers to contacting the cell(s) with factors such as the nucleic acids disclosed herein to downregulate or upregulate the quantity and/or activity of a cellular component, for example, DNA, RNA or protein. This phrase also encompasses contacting the cell(s) with any factors including proteins and small molecules that can downregulate or upregulate the gene/protein of interest.


The term “upregulate expression of” means to affect expression of, for example to induce expression or activity, or induce increased/greater expression or activity relative to an untreated cell.


As used herein, “upregulation” or “upregulate” refers to the process by which a cell increases the quantity and/or activity of a cellular component, for example, DNA, RNA or protein, in response to an external variable.


“Variant” refers to a polypeptide or polynucleotide that differs from a reference polypeptide or polynucleotide, but retains essential properties. A typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical. A variant and reference polypeptide may differ in amino acid sequence by one or more modifications (e.g., substitutions, additions, and/or deletions). A substituted or inserted amino acid residue may or may not be one encoded by the genetic code. A variant of a polypeptide may be naturally occurring such as an allelic variant, or it may be a variant that is not known to occur naturally.


II. Compositions

A. Factors Inducing Non-Hepatocyte Cells into Hepatocyte-Like Properties


Obtaining fully functional cell types is a major challenge for drug discovery, bioartificial liver and regenerative medicine. Currently, a fundamental solution to this key problem is still lacking. Functional human induced hepatocytes (hiHeps) can be generated from fibroblasts by upregulating at least one factor selected from the group consisting of HNF1A, HNF4A, HNF6, ATF5, PROX1, and CEBPA, as well as MYC genes mRNA or protein levels. All known functional variants and isoforms of the hepatocyte inducing factors disclosed herein are contemplated. These known sequences are readily available in the National Center for Biotechnology Information Genebak database.


Preferably, p53 activity is additionally, downregulated as indicated by a downregulation of the p53 gene, mRNA and/or protein levels.


1. Nucleic Acids Encoding Hepatocyte Inducing Factors


i. HNF1A


HNF1A (also known as TCF1) is a tumor suppressor gene involved in liver tumorigenesis. It is located on the long arm of chromosome 12, encoded by 10 exons, spanning 23 kilobases, and is expressed in various tissues, including liver, kidney, pancreas, and digestive tract. It encodes a transcription factor HNF1, which, in the liver, is implicated in hepatocyte differentiation and is required for expression of certain liver-specific genes, including albumin, β-fibrinogen, and α1-antitrypsin. Courtois, et al., Science, 30(4827:688-692 (1987). The HNF1A gene is conserved in chimpanzee, Rhesus monkey, dog, cow, mouse, rat, chicken, zebrafish, and frog.


In a preferred embodiment, a nucleotide encoding HNF1A is represented below by SEQ ID NO:1.











(SEQ ID NO: 1)



atggtttcta aactgagcca gctgcagacg gagctcctgg







cggccctgct cgagtcaggg ctgagcaaag aggcactgat







ccaggcactg ggtgagccgg ggccctacct cctggctgga 







gaaggccccc tggacaaggg ggagtcctgc ggcggcggtc







gaggggagct ggctgagctg cccaatgggc tgggggagac







tcggggctcc gaggacgaga cggacgacga tggggaagac 







ttcacgccac ccatcctcaa agagctggag aacctcagcc







ctgaggaggc ggcccaccag aaagccgtgg tggagaccct







tctgcaggag gacccgtggc gtgtggcgaa gatggtcaag 







tcctacctgc agcagcacaa catcccacag cgggaggtgg







tcgataccac tggcctcaac cagtcccacc tgtcccaaca







cctcaacaag ggcactccca tgaagacgca gaagcgggcc 







gccctgtaca cctggtacgt ccgcaagcag cgagaggtgg







cgcagcagtt cacccatgca gggcagggag ggctgattga







agagcccaca ggtgatgagc taccaaccaa gaaggggcgg 







aggaaccgtt tcaagtgggg cccagcatcc cagcagatcc







tgttccaggc ctatgagagg cagaagaacc ctagcaagga







ggagcgagag acgctagtgg aggagtgcaa tagggcggaa 







tgcatccaga gaggggtgtc cccatcacag gcacaggggc







tgggctccaa cctcgtcacg gaggtgcgtg tctacaactg







gtttgccaac cggcgcaaag aagaagcctt ccggcacaag 







ctggccatgg acacgtacag cgggcccccc ccagggccag







gcccgggacc tgcgctgccc gctcacagct cccctggcct







gcctccacct gccctctccc ccagtaaggt ccacggtgtg 







cgctatggac agcctgcgac cagtgagact gcagaagtac







cctcaagcag cggcggtccc ttagtgacag tgtctacacc







cctccaccaa gtgtccccca cgggcctgga gcccagccac 







agcctgctga gtacagaagc caagctggtc tcagcagctg







ggggccccct cccccctgtc agcaccctga cagcactgca







cagcttggag cagacatccc caggcctcaa ccagcagccc 







cagaacctca tcatggcctc acttcctggg gtcatgacca







tcgggcctgg tgagcctgcc tccctgggtc ctacgttcac







caacacaggt gcctccaccc tggtcatcgg cctggcctcc 







acgcaggcac agagtgtgcc ggtcatcaac agcatgggca







gcagcctgac caccctgcag cccgtccagt tctcccagcc







gctgcacccc tcctaccagc agccgctcat gccacctgtg 







cagagccatg tgacccagag ccccttcatg gccaccatgg







ctcagctgca gagcccccac gccctctaca gccacaagcc







cgaggtggcc cagtacaccc acacgggcct gctcccgcag 







actatgctca tcaccgacac caccaacctg agcgccctgg







ccagcctcac gcccaccaag caggtcttca cctcagacac







tgaggcctcc agtgagtccg ggcttcacac gccggcatct 







caggccacca ccctccacgt ccccagccag gaccctgccg







gcatccagca cctgcagccg gcccaccggc tcagcgccag







ccccacagtg tcctccagca gcctggtgct gtaccagagc 







tcagactcca gcaatggcca gagccacctg ctgccatcca







accacagcgt catcgagacc ttcatctcca cccagatggc







ctcttcctcc cag 






A nucleic acid encoding HNF1A can include a sequence having at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to SEQ ID NO:1 or a functional fragment or variant of SEQ ID NO:1.


A number of naturally occurring variants of nucleic acids encoding HNF1A and their activities are known in the art, and include, but are not limited to, the transcript variant for HNF1A as represented by GenBank Accession No: XM_005253931.1.


ii. HNF6


HNF6 was originally characterized as a transcriptional activator of the liver promoter of the 6-phosphofructo-2-kinase (pfk-2) gene, is expressed in liver, brain, spleen, pancreas, and testis. Lannoy, et al., J. Biol. Chem., 273:13552-13562 (1998). Alternative splicing results in multiple transcript variants.


In one embodiment, HNF6 is represented by SEQ ID NO:2.











(SEQ ID NO: 2)



atgaacgcgc agctgaccat ggaagcgatc ggcgagctgc







acggggtgag ccatgagccg gtgcccgccc ctgccgacct







gctgggcggc agcccccacg cgcgcagctc cgtggcgcac 







cgcggcagcc acctgccccc cgcgcacccg cgctccatgg







gcatggcgtc cctgctggac ggcggcagcg gcggcggaga







ttaccaccac caccaccggg cccctgagca cagcctggcc 







ggccccctgc atcccaccat gaccatggcc tgcgagactc







ccccaggtat gagcatgccc accacctaca ccaccttgac







ccctctgcag ccgctgcctc ccatctccac agtctcggac 







aagttccccc accatcacca ccaccaccat caccaccacc







acccgcacca ccaccagcgc ctggcgggca acgtgagcgg







tagcttcacg ctcatgcggg atgagcgcgg gctggcctcc 







atgaataacc tctatacccc ctaccacaag gacgtggccg







gcatgggcca gagcctctcg cccctctcca gctccggtct







gggcagcatc cacaactccc agcaagggct cccccactat 







gcccacccgg gggccgccat gcccaccgac aagatgctca







cccccaacgg cttcgaagcc caccacccgg ccatgctcgg







ccgccacggg gagcagcacc tcacgcccac ctcggccggc 







atggtgccca tcaacggcct tcctccgcac catccccacg







cccacctgaa cgcccagggc cacgggcaac tcctgggcac







agcccgggag cccaaccctt cggtgaccgg cgcgcaggtc 







agcaatggaa gtaattcagg gcagatggaa gagatcaata







ccaaagaggt ggcgcagcgt atcaccaccg agctcaagcg







ctacagcatc ccacaggcca tcttcgcgca gagggtgctc 







tgccgctccc aggggaccct ctcggacctg ctgcgcaacc







ccaaaccctg gagcaaactc aaatccggcc gggagacctt







ccggaggatg tggaagtggc tgcaggagcc ggagttccag 







cgcatgtccg cgctccgctt agcagcatgc aaaaggaaag







aacaagaaca tgggaaggat agaggcaaca cacccaaaaa







gcccaggttg gtcttcacag atgtccagcg tcgaactcta 







catgcaatat tcaaggaaaa taagcgtcca tccaaagaat







tgcaaatcac catttcccag cagctggggt tggagctgag







cactgtcagc aacttcttca tgaacgcaag aaggaggagt 







ctggacaagt ggcaggacga gggcagctcc aattcaggca







actcatcttc ttcatcaagc acttgtacca aagca






A nucleic acid encoding HNF6 can include a sequence having at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to SEQ ID NO:2 or a functional fragment or variant of SEQ ID NO:2.


A number of naturally occurring variants of nucleic acids encoding HNF6 and their activities are known in the art. A human hepatocyte nuclear factor 6 (HNF6) gene is described under NCBI GenBank Accession No. AF035581. A Homo sapiens transcript variant mRNA is disclosed under Genbank Accession No. NM_004498.2.


iii. HNF4A


Hepatocyte nuclear factor 4 alpha (HNF4alpha, NR2A1, gene symbol HNF4A) is a highly conserved member of the nuclear receptor (NR) superfamily of ligand-dependent transcription factors (Sladeck, et al., Genes Dev., 4(12B): 2353-65 (1990). HNF4A1 is expressed in liver (hepatocytes), kidney, small intestine, etc. HNF4A2 is the most predominant isoform in the liver. HNF4A regulates most if not all of the apolipoprotein genes in the liver and regulates the expression of many cytochrome P450 genes (e.g., CYP3A4, CYP2D6) and Phase II enzymes and hence may play a role in drug metabolism (Gonzalez, et al., Drug Metab. Pharmacokinet., 23(1):2-7 (2008).


In one embodiment, HNF4 is represented by SEQ ID NO:3.











(SEQ ID NO: 3)



atgcgactct ccaaaaccct cgtcgacatg gacatggccg







actacagtgc tgcactggac ccagcctaca ccaccctgga







atttgagaat gtgcaggtgt tgacgatggg caatgacacg 







tccccatcag aaggcaccaa cctcaacgcg cccaacagcc







tgggtgtcag cgccctgtgt gccatctgcg gggaccgggc







cacgggcaaa cactacggtg cctcgagctg tgacggctgc 







aagggcttct tccggaggag cgtgcggaag aaccacatgt







actcctgcag atttagccgg cagtgcgtgg tggacaaaga







caagaggaac cagtgccgct actgcaggct caagaaatgc 







ttccgggctg gcatgaagaa ggaagccgtc cagaatgagc







gggaccggat cagcactcga aggtcaagct atgaggacag







cagcctgccc tccatcaatg cgctcctgca ggcggaggtc 







ctgtcccgac agatcacctc ccccgtctcc gggatcaacg







gcgacattcg ggcgaagaag attgccagca tcgcagatgt







gtgtgagtcc atgaaggagc agctgctggt tctcgttgag 







tgggccaagt acatcccagc tttctgcgag ctccccctgg







acgaccaggt ggccctgctc agagcccatg ctggcgagca







cctgctgctc ggagccacca agagatccat ggtgttcaag 







gacgtgctgc tcctaggcaa tgactacatt gtccctcggc







actgcccgga gctggcggag atgagccggg tgtccatacg







catccttgac gagctggtgc tgcccttcca ggagctgcag 







atcgatgaca atgagtatgc ctacctcaaa gccatcatct







tctttgaccc agatgccaag gggctgagcg atccagggaa







gatcaagcgg ctgcgttccc aggtgcaggt gagcttggag 







gactacatca acgaccgcca gtatgactcg cgtggccgct







ttggagagct gctgctgctg ctgcccacct tgcagagcat







cacctggcag atgatcgagc agatccagtt catcaagctc 







ttcggcatgg ccaagattga caacctgttg caggagatgc







tgctgggagg gtcccccagc gatgcacccc atgcccacca







ccccctgcac cctcacctga tgcaggaaca tatgggaacc 







aacgtcatcg ttgccaacac aatgcccact cacctcagca







acggacagat gtccacccct gagaccccac agccctcacc







gccaggtggc tcagggtctg agccctataa gctcctgccg 







ggagccgtcg ccacaatcgt caagcccctc tctgccatcc







cccagccgac catcaccaag caggaagtta tc 






A nucleic acid encoding HNF4 can include a sequence having at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to SEQ ID NO:3 or a functional fragment or variant of SEQ ID NO:3.


A number of naturally occurring variants of nucleic acids encoding HNF4 and their activities are known in the art. A human hepatocyte nuclear factor 4 gene is described under NCBI GenBank Accession No. BC137539.1.


iv. ATF5


ATF5 encodes activating transcription factor 5. ATF5 transcripts and protein are expressed in a wide variety of tissues, in particular, high expression of transcripts in liver.


In one embodiment, ATF5 is represented by SEQ ID NO:4.











(SEQ ID NO: 4)



atgtcactcc tggcgaccct ggggctggag ctggacaggg







ccctgctccc agctagtggg ctgggatggc tcgtagacta







tgggaaactc cccccggccc ctgcccccct ggctccctat 







gaggtccttg ggggagccct ggagggcggg cttccagtgg







ggggagagcc cctggcaggt gatggcttct ctgactggat







gactgagcga gttgatttca cagctctcct ccctctggag 







cctcccttac cccccggcac cctcccccaa ccttccccaa







ccccacctga cctggaagct atggcctccc tcctcaagaa







ggagctggaa cagatggaag acttcttcct agatgccccg 







cccctcccac caccctcccc gccgccacta ccaccaccac







cactaccacc agccccctcc ctccccctgt ccctcccctc







ctttgacctc ccccagcccc ctgtcttgga tactctggac 







ttgctggcca tctactgccg caacgaggcc gggcaggagg







aagtggggat gccgcctctg cccccgccac agcagccccc







tcctccttct ccacctcaac cttctcgcct ggccccctac 







ccacatcctg ccaccacccg aggggaccgc aagcaaaaga







agagagacca gaacaagtcg gcggctctga ggtaccgcca







gcggaagcgg gcagagggtg aggccctgga gggcgagtgc 







caggggctgg aggcacggaa tcgcgagctg aaggaacggg







cagagtccgt ggagcgcgag atccagtacg tcaaggacct







gctcatcgag gtttacaagg cccggagcca gaggacccgt 







agctgc 






A nucleic acid encoding ATF5 can include a sequence having at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to SEQ ID NO:4 or a functional fragment or variant of SEQ ID NO:4. A number of naturally occurring variants of nucleic acids encoding ATF5 and their activities are known in the art. A human ATF5 transcript variant 3 (mRNA) is described under Genbank Accession No. NM_001290746.1 (Abe, et al., J. Biol. Chem., 289(7):3888-3900 (2014)).


v. PROX1


In one embodiment, PROX1 is represented by SEQ ID NO:5.











(SEQ ID NO: 5)



atgcctgacc atgacagcac agccctctta agccggcaaa







ccaagaggag aagagttgac attggagtga aaaggacggt







agggacagca tctgcatttt ttgctaaggc aagagcaacg 







ttttttagtg ccatgaatcc ccaaggttct gagcaggatg







ttgagtattc agtggtgcag catgcagatg gggaaaagtc







aaatgtactc cgcaagctgc tgaagagggc gaactcgtat 







gaagatgcca tgatgccttt tccaggagca accataattt







cccagctgtt gaaaaataac atgaacaaaa atggtggcac







ggagcccagt ttccaagcca gcggtctctc tagtacaggc 







tccgaagtac atcaggagga tatatgcagc aactcttcaa







gagacagccc cccagagtgt ctttcccctt ttggcaggcc







tactatgagc cagtttgata tggatcgctt atgtgatgag 







cacctgagag caaagcgcgc ccgggttgag aatataattc







ggggtatgag ccattccccc agtgtggcat taaggggcaa







tgaaaatgaa agagagatgg ccccgcagtc tgtgagtccc 







cgagaaagtt acagagaaaa caaacgcaag caaaagcttc







cccagcagca gcaacagagt ttccagcagc tggtttcagc







ccgaaaagaa cagaagcgag aggagcgccg acagctgaaa 







cagcagctgg aggacatgca gaaacagctg cgccagctgc







aggaaaagtt ctaccaaatc tatgacagca ctgattcgga







aaatgatgaa gatggtaacc tgtctgaaga cagcatgcgc 







tcggagatcc tggatgccag ggcccaggac tctgtcggaa







ggtcagataa tgagatgtgc gagctagacc caggacagtt







tattgaccga gctcgagccc tgatcagaga gcaggaaatg 







gctgaaaaca agccgaagcg agaaggcaac aacaaagaaa







gagaccatgg gccaaactcc ttacaaccgg aaggcaaaca







tttggctgag accttgaaac aggaactgaa cactgccatg 







tcgcaagttg tggacactgt ggtcaaagtc ttttcggcca







agccctcccg ccaggttcct caggtcttcc cacctctcca







gatcccccag gccagatttg cagtcaatgg ggaaaaccac 







aatttccaca ccgccaacca gcgcctgcag tgctttggcg







acgtcatcat tccgaacccc ctggacacct ttggcaatgt







gcagatggcc agttccactg accagacaga agcactgccc 







ctggttgtcc gcaaaaactc ctctgaccag tctgcctccg







gccctgccgc tggcggccac caccagcccc tgcaccagtc







gcctctctct gccaccacgg gcttcaccac gtccaccttc 







cgccacccct tcccccttcc cttgatggcc tatccatttc







agagcccatt aggtgctccc tccggctcct tctctggaaa







agacagagcc tctcctgaat ccttagactt aactagggat 







accacgagtc tgaggaccaa gatgtcatct caccacctga







gccaccaccc ttgttcacca gcacacccgc ccagcaccgc







cgaagggctc tccttgtcgc tcataaagtc cgagtgcggc 







gatcttcaag atatgtctga aatatcacct tattcgggaa







gtgcaatgca ggaaggattg tcacccaatc acttgaaaaa







agcaaagctc atgttttttt atacccgtta tcccagctcc 







aatatgctga agacctactt ctccgacgta aagttcaaca







gatgcattac ctctcagctc atcaagtggt ttagcaattt







ccgtgagttt tactacattc agatggagaa gtacgcacgt 







caagccatca acgatggggt caccagtact gaagagctgt







ctataaccag agactgtgag ctgtacaggg ctctgaacat







gcactacaat aaagcaaatg actttgaggt tccagagaga 







ttcctggaag ttgctcagat cacattacgg gagtttttca







atgccattat cgcaggcaaa gatgttgatc cttcctggaa







gaaggccata tacaaggtca tctgcaagct ggatagtgaa 







gtccctgaga ttttcaaatc cccgaactgc ctacaagagc







tgcttcatga g 






A nucleic acid encoding PROX1 can include a sequence having at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to SEQ ID NO:5 or a functional fragment or variant of SEQ ID NO:5. A number of naturally occurring variants of nucleic acids encoding PROX1 and their activities are known in the art.


vi. CEBPA


CEBPA encodes a basic leucine zipper (bZIP) transcription factor which can bind as a homodimer to certain promoters and enhancers.


In one embodiment, CEBPA is represented by SEQ ID NO:6.











(SEQ ID NO: 6)



atggagtcgg ccgacttcta cgaggcggag ccgcggcccc







cgatgagcag ccacctgcag agccccccgc acgcgcccag







cagcgccgcc ttcggctttc cccggggcgc gggccccgcg 







cagcctcccg ccccacctgc cgccccggag ccgctgggcg







gcatctgcga gcacgagacg tccatcgaca tcagcgccta







catcgacccg gccgccttca acgacgagtt cctggccgac 







ctgttccagc acagccggca gcaggagaag gccaaggcgg







ccgtgggccc cacgggcggc ggcggcggcg gcgactttga







ctacccgggc gcgcccgcgg gccccggcgg cgccgtcatg 







cccgggggag cgcacgggcc cccgcccggc tacggctgcg







cggccgccgg ctacctggac ggcaggctgg agcccctgta







cgagcgcgtc ggggcgccgg cgctgcggcc gctggtgatc 







aagcaggagc cccgcgagga ggatgaagcc aagcagctgg







cgctggccgg cctcttccct taccagccgc cgccgccgcc







gccgccctcg cacccgcacc cgcacccgcc gcccgcgcac 







ctggccgccc cgcacctgca gttccagatc gcgcactgcg







gccagaccac catgcacctg cagcccggtc accccacgcc







gccgcccacg cccgtgccca gcccgcaccc cgcgcccgcg 







ctcggtgccg ccggcctgcc gggccctggc agcgcgctca







aggggctggg cgccgcgcac cccgacctcc gcgcgagtgg







cggcagcggc gcgggcaagg ccaagaagtc ggtggacaag 







aacagcaacg agtaccgggt gcggcgcgag cgcaacaaca







tcgcggtgcg caagagccgc gacaaggcca agcagcgcaa







cgtggagacg cagcagaagg tgctggagct gaccagtgac 







aatgaccgcc tgcgcaagcg ggtggaacag ctgagccgcg







aactggacac gctgcggggc atcttccgcc agctgccaga







gagctccttg gtcaaggcca tgggcaactg cgcg 






A nucleic acid encoding CEBPA can include a sequence having at least 80%, 85%, 90%, 95%, 99%, or 100% sequence identity to SEQ ID NO:6 or a functional fragment or variant of SEQ ID NO:6. A number of naturally occurring variants of nucleic acids encoding CEBPA and their activities are known in the art.


vii. MYC


Myc (c-Myc) is a regulator gene that codes for a transcription factor, which is multifunctional, nuclear phosphoprotein that plays a role in cell cycle progression, apoptosis and cellular transformation.


In one embodiment, MYC is represented by SEQ ID NO:7.











(SEQ ID NO: 7)



ctggattttt ttcgggtagt ggaaaaccag cagcctcccg







cgacgatgcc cctcaacgtt agcttcacca acaggaacta







tgacctcgac tacgactcgg tgcagccgta tttctactgc 







gacgaggagg agaacttcta ccagcagcag cagcagagcg







agctgcagcc cccggcgccc agcgaggata tctggaagaa







attcgagctg ctgcccaccc cgcccctgtc ccctagccgc 







cgctccgggc tctgctcgcc ctcctacgtt gcggtcacac







ccttctccct tcggggagac aacgacggcg gtggcgggag







cttctccacg gccgaccagc tggagatggt gaccgagctg 







ctgggaggag acatggtgaa ccagagtttc atctgcgacc







cggacgacga gaccttcatc aaaaacatca tcatccagga







ctgtatgtgg agcggcttct cggccgccgc caagctcgtc 







tcagagaagc tggcctccta ccaggctgcg cgcaaagaca







gcggcagccc gaaccccgcc cgcggccaca gcgtctgctc







cacctccagc ttgtacctgc aggatctgag cgccgccgcc 







tcagagtgca tcgacccctc ggtggtcttc ccctaccctc







tcaacgacag cagctcgccc aagtcctgcg cctcgcaaga







ctccagcgcc ttctctccgt cctcggattc tctgctctcc 







tcgacggagt cctccccgca gggcagcccc gagcccctgg







tgctccatga ggagacaccg cccaccacca gcagcgactc







tgaggaggaa caagaagatg aggaagaaat cgatgttgtt 







tctgtggaaa agaggcaggc tcctggcaaa aggtcagagt







ctggatcacc ttctgctgga ggccacagca aacctcctca







cagcccactg gtcctcaaga ggtgccacgt ctccacacat 







cagcacaact acgcagcgcc tccctccact cggaaggact







atcctgctgc caagagggtc aagttggaca gtgtcagagt







cctgagacag atcagcaaca accgaaaatg caccagcccc 







aggtcctcgg acaccgagga gaatgtcaag aggcgaacac







acaacgtctt ggagcgccag aggaggaacg agctaaaacg







gagctttttt gccctgcgtg accagatccc ggagttggaa 







aacaatgaaa aggcccccaa ggtagttatc cttaaaaaag







ccacagcata catcctgtcc gtccaagcag aggagcaaaa







gctcatttct gaagaggact t ttgcggaa acgacgagaa 







cagttgaaac acaaacttga acagctacgg aactcttgtg







cg 






2. Vectors Encoding Hepatocyte Inducing Factors


The Hepatocyte inducing factors are introduced into a host cell using suitable transformation vectors. Nucleic acids, such as those described above, can be inserted into vectors for expression in cells. As used herein, a “vector” is a replicon, such as a plasmid, phage, virus or cosmid, into which another DNA segment may be inserted so as to bring about the replication of the inserted segment. Vectors can be expression vectors. An “expression vector” is a vector that includes one or more expression control sequences, and an “expression control sequence” is a DNA sequence that controls and regulates the transcription and/or translation of another DNA sequence.


Nucleic acids in vectors can be operably linked to one or more expression control sequences. For example, the control sequence can be incorporated into a genetic construct so that expression control sequences effectively control expression of a coding sequence of interest. Examples of expression control sequences include promoters, enhancers, and transcription terminating regions. A promoter is an expression control sequence composed of a region of a DNA molecule, typically within 100 nucleotides upstream of the point at which transcription starts (generally near the initiation site for RNA polymerase II). To bring a coding sequence under the control of a promoter, it is necessary to position the translation initiation site of the translational reading frame of the polypeptide between one and about fifty nucleotides downstream of the promoter. Enhancers provide expression specificity in terms of time, location, and level. Unlike promoters, enhancers can function when located at various distances from the transcription site. An enhancer also can be located downstream from the transcription initiation site. A coding sequence is “operably linked” and “under the control” of expression control sequences in a cell when RNA polymerase is able to transcribe the coding sequence into mRNA, which then can be translated into the protein encoded by the coding sequence.


Suitable expression vectors include, without limitation, plasmids and viral vectors derived from, for example, bacteriophage, baculoviruses, tobacco mosaic virus, herpes viruses, cytomegalo virus, retroviruses, vaccinia viruses, adenoviruses, lentiviruses and adeno-associated viruses. Numerous vectors and expression systems are commercially available from such corporations as Novagen (Madison, Wis.), Clontech (Palo Alto, Calif.), Stratagene (La Jolla, Calif.), and Invitrogen Life Technologies (Carlsbad, Calif.).


B. Cells to be Induced


Cells that can be reprogrammed include embryonic stem cells (ESC), induced pluripotent stem cells (iPSC), fibroblast cells, adipose-derived stem cells (ADSC), neural derived stem cells, blood keratinocytes, intestinal epithelial cells and other non-hepatocyte somatic cells. In a preferred embodiment, the non-hepatocyte cell is a fibroblast cell, for example an embryonic fibroblasts (HEFs) or foreskin fibroblasts. The cells are preferably obtained from a mammal, for example, rat, mice, monkeys, dogs, cats, cows, rabbits, horses, pigs Preferably, the cells are obtained from a human subject.


C. Induced Hepatocyte Cells


iHeps are disclosed, which are obtained for example, by a method which includes treating non-hepatocyte cells to overexpress the hepatic fate conversion factors HNF1A, HNF4A, and HNF6 along with the maturation factors ATF5, PROX1, and CEBPA. The non-hepatocyte is treated to overexpress at least one hepatocyte inducing factor selected from the group consisting of HNF1A, HNF4A, HNF6, ATF5, PROX1, and CEBPA. In some embodiments the non-hepatocyte is treated to overexpress or transformed to express at least 2, at least 3, at least 4 or at least 5 of the hepatocyte inducing factors. In a preferred embodiment, the cell is transformed to overexpress all 6 Hepatocyte inducing factors.


iHeps show typical and functional characteristics of hepatocytes in the organisms from which the cell induced was obtained. For example, iHeps show the typical morphology for primary human hepatocytes. iHeps express at least one hepatic marker selected from the group consisting of albumin, Cytochrome P450 (Cyp)3A4 and CypB6. Like primary human hepatocytes, hiHeps express an additional spectrum of phase I and II drug-metabolizing enzymes and phase III drug transporters and albumin. The metabolic activities of at least one of CYP3A4, CYPB6, CYP1A2, CYP2C9, and CYP2C19 are comparable between hiHeps and freshly isolated primary human hepatocytes. Preferably, the iHeps are functional as determined by the metabolic activity of these enzymes being at least 50% higher than the activity of the same enzyme in ES-Heps obtained from the same organism. The activity of the enzyme can be 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100% or more, higher than the activity in ES-Heps. Most preferably, the activities of all these CYP enzymes in hiHeps are at least 100-fold higher than that of ES-Heps.


In some embodiments, MYC expression levels in iHeps are lower than the levels found in normal hepatocytes in the corresponding organism as measured for example, by quantitative reverse transcriptase polymerase chain reaction (RT-qPCR), i.e., if the donor organism for the non-hepatocyte cell to be induced is a human subject, the levels are compared to normal hepatocytes found in humans.


Functional hiHeps may also express at least one drug metabolic phase II enzyme or phase II transporter selected from the group consisting of UDP glucuronosyltransferase (UGT)1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, GSTA1, UGT2B7, UGT2515, Microsomal glutathione-S-transferase 1 (MGST1), nicotinamide N-methyltransferase (NNMT), NTCP, organic anion-transporting polypeptide 1B3 (OATP1B3), Multidrug resistance protein(MRP)6, MRP2, Flavin-containing monooxygenase 5 (FMO5), Monoamine oxidase (MAO)A, MAOB, and epoxide hydrolase 1 (EPHX1). Preferably, endogenous expression of Forkhead box (FOX)A1, FOXA2, FOXA3 and Liver receptor homolog 1 (LRH1) is activated in hiHeps.


In some embodiment where the cell being induced is not an epithelial cell, hiHeps additionally express at least one epithelial cell marker, for example, E-cadherin, and where the cell being induced is a fibroblast, the hiHeps obtained following induction of fibroblasts using the methods disclosed herein, do not express the fibroblast marker genes such as COLIA1, PDGFRB, THY1 and α-fetoprotein as measured for example by RT-PCR.


With respect to functional characteristics associated with mature hepatocytes, hiHeps possess at least one characteristic selected from the group consisting of: albumin secretion, LDL uptake, indocyanine green (ICG) incorporation from cell culture medium and exclusion of the absorbed ICG after withdrawal, glycogen synthesis and storage, and fatty droplet accumulation.


III. Method of Making

Huang, et al., Nature, 475:386-389 (2011) disclose the direct induction of hepatocyte-like cells from mouse tail-tip fibroblasts by transduction of Gata4, Hnf1α and Foxa3, and inactivation of p19(Arf). Induced cells show typical epithelial morphology. Sekiya and Suzuki, Nature, 475:390-393 (2011)), identified three specific combinations of two transcription factors, Hnf4α plus Foxa1, Foxa2 or Foxa3, that can convert mouse embryonic and adult fibroblasts into cells that resemble hepatocytes in vitro. Cai, et al., Hepatology, 45(5):1229-39 (2007) disclose a three-stage method to direct the differentiation of human embryonic stem cells (hESCs) into hepatic cells in serum-free medium. Human ESCs were first differentiated into definitive endoderm cells by 3 days of Activin A treatment. Next, the presence of fibroblast growth factor-4 and bone morphogenetic protein-2 in the culture medium for 5 days induced efficient hepatic differentiation from definitive endoderm cells, followed by 10 days of further in vitro maturation. Zhao, et al., Cell Res., 23(1):157-161 (2013) disclose a method of promoting the maturation of hESCs into cells with hepatocyte-like properties by inducing expression of PROX1 and HNF6.


In the methods disclosed herein, the non-hepatocyte is reprogrammed into an iHep by upregulating Hepatocyte inducing factors in the cell, optionally in combination with upregulating MYC and downregulating p53 and culturing the cells for a sufficient period of time as disclosed herein to convert the cell into a cell with hepatocyte-like properties. The non-hepatocyte cells to be induced are obtained from the donor animal using methods known in the art. The cells are placed in culture and cultured using methods that are known in the art.


The reprogramming method includes the following steps: (a) treat the cells to upregulate hepatocyte inducing factors and culture the cells in cell culture medium (transformation phase); (b) replate and culture the cells in HCM (expansion phase), and (c) a maturation phase, where cells are cultured in a hepatocyte maturation medium. A schematic for the disclosed method is shown in FIG. 1E. At the transformation phase, the cells are treated to upregulate at least one hepatocyte inducing factor selected from the group consisting of HNF1A, HNF4A, HNF6, ATF5, PROX1, and CEBPA. Preferably, the cells are additionally treated to upregulate MYC and/or downregulate p53.


In the transformation phase, the treated cells are cultured for a sufficient length of time in conventional cell culture medium, for example, Dulbecco's Modified Eagle's medium (DMEM). Preferably, the cells are cultured for at least 7 days in this first step, to about 80% confluence. The cells then replated and expanded in HCM for a period of about 15 to 30 days, preferably for about 18-30 days, and more preferably, for about 18 days (expansion phase), and then transferred to modified William's E medium for a period of about 5 days (maturation phase), following which induced hepatocytes are harvested. Preferably, p53 siRNA is downregulated at the end of the expansion phase, for example at about day 20-30 post infection, preferably, at about day 25 post infection, before the cells are transferred into the modified William's E medium (FIG. 1E). We observe silence of p53 siRNA around 25 days post infection. The silence is mainly caused by the introduction of hepatic transcription factors. For example, HNF4A and CEBPA can substantially decrease proliferative rate of iHeps. Furthermore, the self-establishment of endogenous hepatic maturation signaling network also attenuate the reliability of exogenous expression of other transcription factors (FIG. 2).


The method includes a step confirming that the non-hepatocytes have acquired hepatocyte-like properties, using morphological and functional characteristics as well as gene expression.


Morphological confirmation methods include the confirmation of morphological characteristics specific for hepatocytes such as cells having a plurality of nuclei observed by a phase microscope and granules rich in cytoplasm observed by an electron microscope, in particular, the presence of glycogen granules.


Treated cells can also be identified as induced hepatocytes using one or more of the following characteristics: their ability to express ALB at a level comparable to that of primary human hepatocytes; expression of one or more of the five major cytochrome P450 enzymes, CYP3A4, CYP1A2, CYP2C9, and CYP2C19; expression of phase II enzyme or phase II transporter selected from the group consisting of UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, GSTA1, UGT2B7, UGT2515, MGST1, NNMT, NTCP, OATP1B3, MRP6, MRP2, FMO5, MAOA, MAOB, and EPHX1. Successful induction can be confirmed by the presence of an epithelial marker and the absence of a marker for the cell which is being induced. For example, where the cell being induced is a fibroblast, additional indication that the cells has been induced into a hepatocyte-like cell can be expression of at least one epithelial cell marker, for example, E-cadherin, and absence of expression of the fibroblast marker genes such as COLIA1, PDGFRB, THY1 and α-fetoprotein as measured for example by RT-PCR.


A. Upregulating Hepatocyte Inducing Factors and MYC


Hepatocyte inducing factors and MYC are upregulated by contacting the non-hepatocyte with factors which upregulate gene expression and or protein levels/activity of the Hepatocyte inducing Factors and MYC. These factors include, but are not limited to nucleic acids, proteins and small molecules.


For example, upregulation may be accomplished by exogenously introducing the nucleic acids encoding the hepatocyte inducing Factor(s) and optionally, MYC, into the non-hepatocyte (host cell). The nucleic acid may be homologous or heterologous. The nucleic acid molecule can be DNA or RNA, preferably, mRNA. Preferably, the nucleic acid molecule is introduced into the non-hepatocyte cell by lentiviral expression.


The host cell is transformed to overexpress at least one hepatocyte inducing factor selected from the group consisting of HNF1A, HNF4A, HNF6, ATF5, PROX1, and CEBPA. Preferably, the cell is additionally transformed overexpress the proliferation factor MYC. In some embodiments the cell is transformed to express at least 2, at least 3, at least 4 or at least 5 of the hepatocyte inducing factors. In a preferred embodiment, the cell is transformed to overexpress all 6 Hepatocyte inducing factors.


Vectors containing nucleic acids to be expressed can be transferred into host cells. Nucleic acids can be transfected into mammalian cells by techniques including, for example, calcium phosphate co-precipitation, DEAE-dextran-mediated transfection, lipofection, electroporation, or microinjection. The Ex vivo methods disclosed herein can include, for example, the steps of harvesting cells from a subject/donor, culturing the cells, transducing them with an expression vector, and maintaining the cells under conditions suitable for expression of the encoded polypeptides. These methods are known in the art of molecular biology.


Upregulation may also be accomplished by treating the cells with factors known to increase expression of genes encoding the Hepatocyte inducing factors/MYC and/or factors known to increase the corresponding protein levels. For example, Zhao, et al., Cell Res., 23(1):157-161 (2013), disclose a method for promoting the emergence of PROX1 and HNF6-expressing cells from hESCs using the induction factors FGF7, BMP2 and BMP4. Known factors, including small molecules and/or proteins which upregulate Hepatocyte inducing factors gene expression or protein levels can also be use.


B. Downregulating p53


p53 can be downregulated by treating cells to downregulate p53 gene expression, mRNA levels or protein levels. This step includes contacting the cells with any molecule that is known to downregulate p53 gene expression, mRNA or protein levels, including but not limited to nucleic acid molecules, small molecules and protein.


p53 gene expression can be inhibited using a functional nucleic acid, or vector encoding the same, selected from the group consisting of antisense oligonucleotides, siRNA, shRNA, miRNA, EGSs, ribozymes, and aptamers. Preferably, p53 gene expression is inhibited using siRNA, shRNA, or miRNA.


1. RNA Interference


In some embodiments, P53 gene expression is inhibited through RNA interference. Gene expression can also be effectively silenced in a highly specific manner through RNA interference (RNAi). This silencing was originally observed with the addition of double stranded RNA (dsRNA) (Fire, et al. (1998) Nature, 391:806-11; Napoli, et al. (1990) Plant Cell 2:279-89; Hannon, (2002) Nature, 418:244-51). Once dsRNA enters a cell, it is cleaved by an RNase III-like enzyme, Dicer, into double stranded small interfering RNAs (siRNA) 21-23 nucleotides in length that contains 2 nucleotide overhangs on the 3′ ends (Elbashir, et al. (2001) Genes Dev., 15:188-200; Bernstein, et al. (2001) Nature, 409:363-6; Hammond, et al. (2000) Nature, 404:293-6). In an ATP dependent step, the siRNAs become integrated into a multi-subunit protein complex, commonly known as the RNAi induced silencing complex (RISC), which guides the siRNAs to the target RNA sequence (Nykanen, et al. (2001) Cell, 107:309-21). At some point the siRNA duplex unwinds, and it appears that the antisense strand remains bound to RISC and directs degradation of the complementary mRNA sequence by a combination of endo and exonucleases (Martinez, et al. (2002) Cell, 110:563-74). However, the effect of iRNA or siRNA or their use is not limited to any type of mechanism.


Short Interfering RNA (siRNA) is a double-stranded RNA that can induce sequence-specific post-transcriptional gene silencing, thereby decreasing or even inhibiting gene expression. In one example, a siRNA triggers the specific degradation of homologous RNA molecules, such as mRNAs, within the region of sequence identity between both the siRNA and the target RNA. For example, WO 02/44321 discloses siRNAs capable of sequence-specific degradation of target mRNAs when base-paired with 3′ overhanging ends, herein incorporated by reference for the method of making these siRNAs.


Sequence specific gene silencing can be achieved in mammalian cells using synthetic, short double-stranded RNAs that mimic the siRNAs produced by the enzyme dicer (Elbashir, et al. (2001) Nature, 411:494 498) (Ui-Tei, et al. (2000) FEBS Lett 479:79-82). siRNA can be chemically or in vitro-synthesized or can be the result of short double-stranded hairpin-like RNAs (shRNAs) that are processed into siRNAs inside the cell. Synthetic siRNAs are generally designed using algorithms and a conventional DNA/RNA synthesizer. Suppliers include Ambion (Austin, Tex.), ChemGenes (Ashland, Mass.), Dharmacon (Lafayette, Colo.), Glen Research (Sterling, Va.), MWB Biotech (Esbersberg, Germany), Proligo (Boulder, Colo.), and Qiagen (Vento, The Netherlands). siRNA can also be synthesized in vitro using kits such as Ambion's SILENCER® siRNA Construction Kit.


The production of siRNA from a vector is more commonly done through the transcription of a short hairpin RNAse (shRNAs). Kits for the production of vectors comprising shRNA are available, such as, for example, Imgenex's GENESUPPRESSOR™ Construction Kits and Invitrogen's BLOCK-IT™ inducible RNAi plasmid and lentivirus vectors.


2. Antisense


p53 gene expression can be inhibited by antisense molecules. Antisense molecules are designed to interact with a target nucleic acid molecule through either canonical or non-canonical base pairing. The interaction of the antisense molecule and the target molecule is designed to promote the destruction of the target molecule through, for example, RNAse H mediated RNA-DNA hybrid degradation. Alternatively the antisense molecule is designed to interrupt a processing function that normally would take place on the target molecule, such as transcription or replication. Antisense molecules can be designed based on the sequence of the target molecule. There are numerous methods for optimization of antisense efficiency by finding the most accessible regions of the target molecule. Exemplary methods include in vitro selection experiments and DNA modification studies using DMS and DEPC. It is preferred that antisense molecules bind the target molecule with a dissociation constant (Kd) less than or equal to 10−6, 10−8, 10−10, or 10−12.


An “antisense” nucleic acid sequence (antisense oligonucleotide) can include a nucleotide sequence that is complementary to a “sense” nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to the p53 encoding mRNA. Antisense nucleic acid sequences and delivery methods are well known in the art (Goodchild, Curr. Opin. Mol. Ther., 6(2):120-128 (2004); Clawson, et al., Gene Ther., 11(17):1331-1341 (2004)). The antisense nucleic acid can be complementary to an entire coding strand of a target sequence, or to only a portion thereof. An antisense oligonucleotide can be, for example, about 7, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, or more nucleotides in length.


An antisense nucleic acid sequence can be designed such that it is complementary to the entire p53 mRNA sequence, but can also be an oligonucleotide that is antisense to only a portion of the p53 mRNA. An antisense nucleic acid can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used. The antisense nucleic acid also can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).


Other examples of useful antisense oligonucleotides include an alpha-anomeric nucleic acid. An alpha-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual beta-units, the strands run parallel to each other (Gaultier et al., Nucleic Acids. Res. 15:6625-6641 (1987)). The antisense nucleic acid molecule can also comprise a 2′-o-methylribonucleotide (Inoue et al. Nucleic Acids Res. 15:6131-6148 (1987)) or a chimeric RNA-DNA analogue (Inoue et al. FEBS Lett., 215:327-330 (1987)).


3. Aptamers


In some embodiments, the inhibitory molecule is an Aptamer. Aptamers are molecules that interact with a target molecule, preferably in a specific way. Aptamers can bind the target molecule with a very high degree of specificity. For example, aptamers have been isolated that have greater than a 10,000 fold difference in binding affinities between the target molecule and another molecule that differ at only a single position on the molecule. Because of their tight binding properties, and because the surface features of aptamer targets frequently correspond to functionally relevant parts of the protein target, aptamers can be potent biological antagonists. Typically aptamers are small nucleic acids ranging from 15-50 bases in length that fold into defined secondary and tertiary structures, such as stem-loops or G-quartets. Aptamers can bind small molecules, such as ATP and theophiline, as well as large molecules, such as reverse transcriptase and thrombin. Aptamers can bind very tightly with Kd's from the target molecule of less than 10−12 M. It is preferred that the aptamers bind the target molecule with a Kd less than 10−6, 10−8, 10−10, or 10−12. It is preferred that the aptamer have a Kd with the target molecule at least 10, 100, 1000, 10,000, or 100,000 fold lower than the Kd with a background binding molecule. It is preferred when doing the comparison for a molecule such as a polypeptide, that the background molecule be a different polypeptide.


4. Ribozymes


p53 gene expression can be inhibited using ribozymes. Ribozymes are nucleic acid molecules that are capable of catalyzing a chemical reaction, either intramolecularly or intermolecularly. It is preferred that the ribozymes catalyze intermolecular reactions. There are a number of different types of ribozymes that catalyze nuclease or nucleic acid polymerase type reactions which are based on ribozymes found in natural systems, such as hammerhead ribozymes. There are also a number of ribozymes that are not found in natural systems, but which have been engineered to catalyze specific reactions de novo. Preferred ribozymes cleave RNA or DNA substrates, and more preferably cleave RNA substrates. Ribozymes typically cleave nucleic acid substrates through recognition and binding of the target substrate with subsequent cleavage. This recognition is often based mostly on canonical or non-canonical base pair interactions. This property makes ribozymes particularly good candidates for target specific cleavage of nucleic acids because recognition of the target substrate is based on the target substrates sequence.


5. Triplex Forming Oligonucleotides


p53 gene expression can be inhibited using triplex forming molecules. Triplex forming functional nucleic acid molecules are molecules that can interact with either double-stranded or single-stranded nucleic acid. When triplex molecules interact with a target region, a structure called a triplex is formed in which there are three strands of DNA forming a complex dependent on both Watson-Crick and Hoogsteen base-pairing. Triplex molecules are preferred because they can bind target regions with high affinity and specificity. It is preferred that the triplex forming molecules bind the target molecule with a Kd less than 10−6, 10−8, 10−10, or 10−12.


6. External Guide Sequences


p53 expression can be inhibited using external guide sequences. External guide sequences (EGSs) are molecules that bind a target nucleic acid molecule forming a complex, which is recognized by RNase P, which then cleaves the target molecule. EGSs can be designed to specifically target a RNA molecule of choice. RNAse P aids in processing transfer RNA (tRNA) within a cell. Bacterial RNAse P can be recruited to cleave virtually any RNA sequence by using an EGS that causes the target RNA:EGS complex to mimic the natural tRNA substrate. Similarly, eukaryotic EGS/RNAse P-directed cleavage of RNA can be utilized to cleave desired targets within eukaryotic cells. Representative examples of how to make and use EGS molecules to facilitate cleavage of a variety of different target molecules are known in the art.


7. ShRNA


p53 expression can be inhibited using small hairpin RNAs (shRNAs), and expression constructs engineered to express shRNAs. Transcription of shRNAs is initiated at a polymerase III (pol III) promoter, and is thought to be terminated at position 2 of a 4-5-thymine transcription termination site. Upon expression, shRNAs are thought to fold into a stem-loop structure with 3′ UU-overhangs; subsequently, the ends of these shRNAs are processed, converting the shRNAs into siRNA-like molecules of about 21 nucleotides (Brummelkamp et al., Science 296:550-553 (2002); Lee et al., Nature Biotechnol. 20:500-505 (2002); Miyagishi and Taira, Nature Biotechnol. 20:497-500 (2002); Paddison et al., Genes Dev. 16:948-958 (2002); Paul et al., Nature Biotechnol. 20:505-508 (2002); Sui (2002) supra; Yu et al., Proc. Natl. Acad. Sci. USA 99(9):6047-6052 (2002).


C. Delivery Vehicles


Methods of making and using vectors for in vivo expression of functional nucleic acids such as antisense oligonucleotides, siRNA, shRNA, miRNA, EGSs, ribozymes, and aptamers are known in the art.


For example, the delivery vehicle can be a viral vector, for example a commercially available preparation, such as an adenovirus vector (Quantum Biotechnologies, Inc. (Laval, Quebec, Canada). The viral vector delivery can be via a viral system, such as a retroviral vector system which can package a recombinant retroviral genome. The recombinant retrovirus can then be used to infect and thereby deliver to the infected cells nucleic acid encoding the hepatocyte inducing factor(s). The exact method of introducing the altered nucleic acid into the host cell is, of course, not limited to the use of retroviral vectors. Other techniques are widely available for this procedure including the use of adenoviral vectors, adeno-associated viral (AAV) vectors, lentiviral vectors, pseudotyped retroviral vectors, and others described in (Soofiyani, et al., Advanced Pharmaceutical Bulletin, 3(2):249-255 (2013). Viruses can be modified to enhance safety, increase specific uptake, and improve efficiency (see, for example, Zhang, et al., Chinese J Cancer Res., 30(3):182-8 (2011), Miller, et al., FASEB J, 9(2):190-9 (1995), Verma, et al., Annu Rev Biochem., 74:711-38 (2005)).


Physical transduction techniques can also be used, such as liposome delivery and receptor-mediated and other endocytosis mechanisms (see, for example, Schwartzenberger et al., Blood, 87:472-478 (1996)). Commercially available liposome preparations such as LIPOFECTIN, LIPOFECTAMINE (GIBCO-BRL, Inc., Gaithersburg, Md.), SUPERFECT (Qiagen, Inc. Hilden, Germany) and TRANSFECTAM (Promega Biotec, Inc., Madison, Wis.), as well as other liposomes developed according to procedures standard in the art are well known. In addition, nucleic acid or vectors encoding the hepatocyte inducing factors can be delivered in vivo by electroporation as well as by means of a sonoporation. During electroporation electric pulses are applied across the cell membrane to create a transmembrane potential difference, allowing transient membrane permeation and transfection of nucleic acids through the destabilized membrane (Soofiyani, et al., Advanced Pharmaceutical Bulletin, 3(2):249-255 (2013)). Sonoporation combines the local application of ultrasound waves and the intravascular or intratissue administration of gas microbubbles to transiently increase the permeability of vessels and tissues (Escoffre, et al., Curr Gene Ther., 13(1):2-14 (2013)). Electroporation and ultrasound based techniques are targeted transfection methods because the electric pulse or ultrasound waves can be focused on a target tissue or organ and hence gene delivery and expression should be limited to thereto. Expression or overexpression of the disclosed hepatocyte inducing factors accomplished with any of these or other commonly used gene transfer methods, including, but not limited to hydrodynamic injection, use of a gene gun.


IV. Method of Using

The studies disclosed herein show that human hepatocytes with drug metabolic function can be generated by lineage reprogramming, thus providing a cell resource for pharmaceutical applications.


A. In Vitro and Research Applications


(1) Drug Testing


Liver parenchymal cells play a key role in drug development because the liver plays a central role in the metabolic activity of the drug. At present, the main cause of failure of a drug candidate is its ADME (absorption, distribution, metabolism, excretion) is not ideal. An essential part of drug discovery research is to the metabolic and toxicological effects of the candidate drug on liver cells, human liver parenchymal cells with full participation of drug metabolism. Currently the main hepatocytes used for in vitro drug development are human adult primary hepatocytes. Due to their limited sources, and the difficulty of maintaining primary hepatocyte function in vitro is difficult to maintain, their application in drug development is quite limited, hiHeps disclosed herein which express phase I, II and III drug-metabolizing enzymes can be used in vitro drug metabolism studies.


(ii) Research


The problem encountered in studies involving infectious diseases is the lack of adequate animal models. hiHeps can be used to construct humanized mouse models for study of infectious diseases, for example, hepatitis B and C infections. These animal models can provide a reliable in vivo platform for use in the development of vaccines and drugs for treating infectious diseases, particularly diseases that infect the liver.


B. In Vivo Applications


Liver failure and loss of function is one of the most severe consequences of liver disease. Because of its rapid onset, rapid progression, liver transplantation is the primary means of treatment of these diseases. However, donor scarcity presents a serious problem and many patients die while waiting for liver transplantation.


The studies disclosed herein show that transplanted hiHeps repopulate up to 30% of the livers of Tet-uPA/Rag2−/−γc−/− mice and secrete more than 300 mg/ml human albumin in vivo. Thus, hiHeps can be used in the treatment of liver failure and loss of function diseases, for example.


Transplanting isolated iHeps by percutaneous or transjugular infusion into the portal vein, or injecting into the splenic pulp or the peritoneal cavity, is a less invasive procedure compared with liver transplantation. The iHeps are preferably obtained from the same animal being treated. As the host liver is not removed or resected, the loss of graft function should not worsen liver function. Furthermore, isolated iHeps could be, potentially, cryopreserved for ready access. The iHeps can be used as a vehicle for ex vivo gene therapy for example, for rescuing patients from radiation-induced liver damage resulting from radiotherapy for liver tumors. iHeps can be transplanted into a recipient organism using a carrier such as a matrix known for transplantation of hepatocytes. For example, Zhou, et al., Liver Transpl., 17(4):418-27 (2011) discloses the use of decellularized liver matrix (DLM) as a carrier for hepatocyte transplantation. Schwartz, et al., Int. J. Gastroentrol., 10(1): discloses isolating liver and pancreas cells from tissue samples, seeding onto a poly-L-lactic acid matrix and re-implanting into the mesentery of the same patient.


hiHeps can also be used in the bio-artificial liver support systems. Bioartificial liver support system based on the disclosed cells are constructed to temporarily replace the main function of liver failure (remove hazardous substances, provide the liver synthetic biologically active substances), to stabilize and improve the patient's internal environment, until a suitable donor source for transplantation is available. Methods for making bioartifical liver are disclosed for example in U.S. Publication No. 2008/0206733.


V. Kits

Kits for inducing in vitro reprogramming of non-hepatocytes into induced heptocytes with functional hepatocyte metabolic properties are disclosed. The kit includes factors which up-regulate hepatocyte inducing factors HNF1A, HNF6, HNF4A, ATF5, PROX1, CEPBA, and/or MYC and factors which downregulate p53 gene expression and/or protein activity. In one embodiment, the kit includes any DNA sequence of HNF1A, HNF6, HNF4A, ATF5, PROX1, CEPBA, and/or MYC and DNA sequence to downregulate p53 gene expression. In a preferred embodiment, the kit includes lentiviruses which overexpress HNF1A, HNF6, HNF4A, ATF5, PROX1, CEPBA, and/or MYC gene and nucleic acid which inhibits p53 gene expression.


Examples

Materials and Methods


Human Primary Cell Isolation and Culture


The present study was approved by the Clinical Research Ethics Committee of China-Japan Friendship Hospital (Ethical approval No: 2009-50), Stem Cell Research Oversight of Peking University (SCRO201103-03) and conducted according to the principles of the Declaration of Helsinki.


Human embryonic skins and fetal liver tissues at 14 gestational weeks were obtained from abortion with informed patient consent. Fetal liver cells were obtained as previously described (Lilja et al., 64:1240-1248 (1997)). The fetal liver tissue was cut into 1-3 mm3 fragments for digestion in 10 ml medium (RPMI 1640) supplemented with 1 mg/ml collagenase IV (Gibco). Digestion was performed at 37° C. for 15-20 min and erythrocytes were eliminated by slow-speed centrifugation. Cells were washed with RPMI 1640 medium for 3 times. Trypan blue exclusion estimated that cell viability was 90%.


Fresh human embryonic skin tissue (HEF) and ex vivo human adult foreskin tissue (HFF) were sterilized with 75% aqueous ethanol and washed with phosphate buffered saline (PBS). The tissue was carefully separated from subcutaneous tissue with ophthalmic scissors. The tissue was washed several times with PBS, small tissue blocks were seeded in a petri dish, and placed in an incubator at 37° C., 5% CO2. Two hours later, the following were added: DMEM high glucose medium (purchased from Hyclone company, product catalog No. SH30022.01B), 15% fetal bovine serum (FBS), 0.1 mM β-mercaptoethanol, 1% non-essential amino acids, and 1 mM Glutamate, 8 units/ml gentamicin). Cells were digested with 0.25% trypsin and 0.02% EDTA at room temperature for 5 minutes. Cells were seeded at 1:3 in the above-described DMEM high glucose medium in a new Petri dish. Medium was changed every two days, and cells were passaged 1:3 every 4 days to obtain human fibroblasts (derived from fetal skin) and human fibroblasts (derived from adult foreskin). Human skin fibroblasts get to about 80% confluence following cell culture for about 5-7 days.


Human primary hepatocytes were isolated from human donor livers not used for liver transplantation, following informed consent (Seglen, 13:29-83 (1976)) and cultured with HCM (LONZA).


Generation of hiHeps


This study was approved by the Clinical Research Ethics Committee of the China-Japan Friendship Hospital (ethical approval 2009-50) and Stem Cell Research Oversight of Peking University (SCRO201103-03), and conducted according to the principles of the Declaration of Helsinki.


Human fibroblasts were infected overnight and cultured in DMEM plus 10% fetal bovine serum for 1 week before transfer into hepatocyte culture medium (HCM) (Lonza) for expansion.


One day before viral infection, human fibroblasts were seeded at 20,000 cells/well into 12-well cell culture plates containing mammalian somatic cell culture medium, and cultured at 37° C. and 5% carbon dioxide culture for 12 hours; then thereto was added the following lentivirus expression vectors: lentivirus expression vectors expressing HNF1A, HNF6, HNF4A, ATF5, PROX1, CEBPA and MYC, respectively and a lentivirus expressing a DNA(s) for inhibiting the expression of p53, 10 μl for HNF1A, 10 μl for HNF6, 6 μl for HNF4A, 10 μl for ATF5, 3 μl for PROX1, 3 μl for CEBPA, 10 μl for MYC and 10 μl for p53 (lentivirus for inhibiting the expression of p53). The medium was changed after 20 hours, after which the medium was changed every day. Cells were cultured for 7 days in DMEM and then transferred into HCM.


After 3 weeks of culture, HCM was replaced by modified William's E medium (Beijing Vitalstar Biotechnology). Cells were passaged every 4 days, and human hepatocyte-like cells were harvested after 30 days. A schematic for hiHep reprogramming is shown in FIG. 1E.


Growth Curve and Doubling Times


For MTT assays, the induced cells of expansion stage and maturation stage were plated into 96-well plate (1000 cells per well) and cultured in HCM (before p53 siRNA-GFP silence) or modified WEM (after p53 siRNA-GFP silence) separately for 7 days. MTT assay was done at each day according to the manufacturer's instructions (Vybrant® MTT Cell Proliferation Assay Kit, Invitrogen). To calculate the doubling time of the induced cells in the expansion stage, the induced cells in the expansion stage (before p53 siRNA-GFP silence) were plated at the density of 30000 cells per well, and cultured in 12-well plate coated with matrigel. The growth rate was determined by counting the number of cells using a hemacytometer as a function of time. Data from the exponential phase of growth (data points at 12, 24, 36 and 48 h) were used to obtain an exponential growth curve. Doubling time (Td) was then obtained using the formula: Td=t*ln 2/ln(Nt/N0) where Nt is the cell number at time t; NO is the cell number at the initial time.


Hepatic Differentiation


Human embryonic stem cells (hESCs, ES cell line H1, WiCell research institute) were maintained on irradiated mouse embryonic fibroblasts in hESCs medium (Thomson et al., Science 282:1145-1147 (1998)). hESCs were differentiated into hepatocytes as previously reported (Zhao et al., Cell Res 23:157-161 (2013)).


Molecular Cloning, Lentivirus Production and Transduction


Complementary DNAs of transcriptional factors are amplified from the human full-length TrueClones™ (Origene) and inserted into pCDH-EF1-MCS-T2A-Puro (System Biosciences) according to user's manual (for each of lentivirus expression vectors of HNF1A, HNF6, HNF4A, ATF5, PROX1, and CEBPA, SEQ ID NOs: 1-6 are inserted into restriction enzyme sites of pCDH-EF1-MCS-T2A-Puro, respectively). Lentivirus expression vector of MYC is constructed by inserting SEQ ID NO:7 into restriction enzyme sites (Xho I and EcoR I) of expression vector pLL-IRES-Puro (Zhao Y et al., Cell Stem Cell. 2008 Nov. 6; 3(5): 475-9; available from Beijing Vitalstar Biotechnology, Ltd. or Peking University. For full sequence information, see http://www.sciencegateway.org/protocols/lentivirus/pllmap.html). Lentivirus for inhibiting the expression of p53 is constructed as follows: DNA molecule for interfering with the expression of p53 is inserted into restriction enzyme sites (Hpa I and Xho I) of expression vector pll3.7 (Rubinson and Dillon et al., Nature Genetics, 2003; available from Beijing Vitalstar Biotechnology, Ltd. or Peking University). The DNA molecule for interfering with the expression of p53 is obtained by annealing with a sense chain (5′-TGACTCCAGTGGTAATCTACTTCAAGAGAGTAGATTACCACTGGA GTCTTTTTTC-3′) and a antisense chain (5′-TC GAGAAAAAAGACTCCAGTGGTAATCTACTCTCTTG AAGTAGATTACCACTGGAGT CA-3′). Virus package is conducted as described previously (Zhao et al., Cell Stem Cell, 3:475-479 (2008)). Human fibroblasts are infected in DMEM (Hyclone) with 10% fetal bovine serum, containing 10 μg/ml polybrene for 12 hours. The fibroblasts were replated seven days post infection and cultured in HCM (LONZA). At about 25 days post infection when p53 siRNA was silenced as indicated by a GFP reporter, hiHeps were cultured in modified William's E Medium (Vitalstar Biotechnology).


Albumin ELISA, Periodic Acid-Schiff (PAS) Staining, Indocyanine Green (ICG) Uptake and Release, Low-Density Lipoprotein (LDL) Uptake and Oil Red Staining


Human Albumin was measured using the Human Albumin ELISA Quantitation kit (Bethyl Laboratory). The PAS staining system was purchased from Sigma-Aldrich. Cultures were fixed with 4% paraformaldehyde (DingGuo) and stained according to the manufacturer's instructions. ICG uptake and release was performed as previously described (Cai et al., Hepatology 45:1229-1239 (2007)). For LDL uptake assay, 10 μg/ml DiI-Ac-LDL (Invitrogen) was incubated with hiHeps for 4 h at 37° C. and observed by fluorescence microscopy. For lipid detection, cultures were fixed with 4% paraformaldehyde and treated with 60% isopropanol for 5 min. Then the isopropanol was removed and Oil Red O working solution was added and incubated for 15 min at room temperature. Then the Oil Red O was removed and cultures rinsed with until clear.


CYP Metabolism Assay


Drug metabolic activity was evaluated using the traditional suspension method as previously described (Gebhardt et al., Drug Metab. Rev. 35:145-213 (2003)). hiHeps were cultured in the medium with 50 mM rifampicin, 50 mMb-naphthoflavone, and 1 m Mphenobarbital for 72 hr and refreshed every 24 hr. Cell viability of dissociated hiHeps, HepG2 cells, ES-Heps, fibroblasts, and freshly isolated primary human hepatocytes was measured by trypan blue. One milliliter of prewarmed incubation medium (William's E medium, 10 mM HEPES [pH 7.4], 2 mM GlutaMAX) was added per 1 3 106 total cells (cell suspension). The substrate solutions were prepared with the same incubation medium [400 mM testosterone, 10 mM midazolam, 200 mM phenacetin, 1 mM bupropion, 500 mM (S)-mephenytoin, 50 mM diclofenac]. The reactions were started by mixing 250 ml of the substrate solution with 250 ml of cell suspension in a 5 ml polystyrene round-bottom tube (BD Falcon). The tubes were put in an orbital shaker in the incubator and the shaker speed was adjusted to 210 rpm. After a 15-240 min incubation at 37° C., the tubes were centrifuged at room temperature to collect the supernatant. The reactions were stopped by addition of sample aliquots to tubes containing triple the volume of quenching solvent (methanol) and frozen at −80° C. Isotope-labeled reference metabolites were used as internal standards. Internal reference metabolites for testosterone, midazolam, (S)-mephenytoin, diclofenac, bupropion, and phenacetin are 6b-hydroxytestosterone-[D7], hydroxymidazolam-[13C3], 40-hydroxymephenytoin-[D3], 40-hydroxydiclofenac-[13C6], hydroxybupropion-[D6], and acetomidophenol-[13C2, 15N], respectively. The metabolites were used to make standard curves for the metabolite analyses. Standard metabolites were 6b-hydroxytestosterone, 10-hydroxymidazolam, hydroxybupropion, 40-hydroxydiclofenac, (±)-40-hydroxymephenytoin, and acetaminophen. The metabolites were quantified by Pharmaron using validated traditional LC-MS methods. The results are expressed as picomoles of metabolite formed per minute and per million cells. Chemicals were purchased from Sigma including b-naphthoflavone, rifampicin, testosterone, midazolam, diclofenac, and phenacetin. Standard metabolites and internal reference metabolites were purchased from BD Biosciences. Phenobarbital was a kind gift from Jinning Lou.


qRT-PCR and RT-PCR


Total RNA was isolated by RNeasy Micro Kit (Qiagen) and then reverse-transcribed with SuperScript® III First-Strand Synthesis (Invitrogen). RT-PCR was performed with 2×EasyTaq PCR SuperMix (TransGen) following the manufacturer's instructions. Primers used for specific detection of endogenous gene expression are shown in Tables 1 and 2.









TABLE 1







Primers used for specific detection of endogenous genes


in FIG. 2A









Gene
Forward Primer (5′→3′)
Reverse Primer (5′→3′)





CEBPA
AGCATTGCCTAGGAACACGA
CCCCAGGATCAAAAGTAATCCCA



A (SEQ ID NO: 8)
(SEQ ID NO: 9)





FOXA1
TACTCCTTCAACCACCCGTTC
GCTATGCCAGACAAACCCC (SEQ



(SEQ ID NO: 10)
ID NO: 11)





FOXA2
CCTACGAACAGGTGATGCAC
GATTTCTTCTCCCTTGCGTCT



T (SEQ ID NO: 12)
(SEQ ID NO: 13)





FOXA3
CGCCCTACAACTTCAACCAC
GATCAGGCCCCAAGAGCTTC



(SEQ ID NO: 14)
(SEQ ID NO: 15)





HNF1A
GCCTCTTCCTCCCAGTAACCA
TATCCCACGAAGCAGCGACA



(SEQ ID NO: 16)
(SEQ ID NO: 17)





HNF4A
AGAAAGAGGCAGACCATCCA
TCCCTGCATACTCCTTGAAGC



C (SEQ ID NO: 18)
(SEQ ID NO: 19)





HNF6
GCAGCTCCAATTCAGGCAAC
CATCATTTGTCTTGCCAAGTCG



(SEQ ID NO: 20)
(SEQ ID NO: 21)





LRH1
CAGATGCCGGAAAACATGCA
CTTAAGTCCATTGGCTCGGAT



A (SEQ ID NO: 22)
(SEQ ID NO: 23)





COL1A1
GGACACCACCCTCAAGAGCC
GTCATGCTCTCGCCGAACCAG



(SEQ ID NO: 24)
(SEQ ID NO: 25)





PDGFRB
ATTCCATGCCGAGTAACAGA
AGTTGACCACCTCATTCCCGAT



CCC (SEQ ID NO: 26)
(SEQ ID NO: 27)





THY1
GCGATTATCTACCCACGTCCA
ACAGACCATGTCCGTGCTA (SEQ



C (SEQ ID NO: 28)
ID NO: 29)





PROX1
CCGAACTGCCTACAAGAGC
AAGGCAGAAAGAAAACAACCA



(SEQ ID NO: 30)
(SEQ ID NO: 31)





GAPDH
TCTTCCAGGAGCGAGATCCC
TGGTCATGAGTCCTTCCACGAT



T (SEQ ID NO: 32)
(SEQ ID NO: 33)
















TABLE 2







Primers used for specific detection of exogenous genes in


FIG. 2B









Gene
Forward Primer (5′→3′)
Reverse Primer (5′→3′)





CEBPA
TGCCTCCTGAACTGCGTCC
GCTCCGCCTCGTAGAAGTCG



(SEQ ID NO: 34)
(SEQ ID NO: 35)





HNF1A
CCGTCTAGGTAAGTTTAAAG
CTCCGGGTAGTAGCTCCAC (SEQ



CTC (SEQ ID NO: 36)
ID NO: 37)





HNF4A
CCGTCTAGGTAAGTTTAAAG
GTGTCATTGCCCATCGTCA (SEQ



CTC (SEQ ID NO: 38)
ID NO: 39)





HNF6
CCGTCTAGGTAAGTTTAAAG
CCGATCGCTTCCATGGTCAG (SEQ



CTC (SEQ ID NO: 40)
ID NO: 41)





PROX1
CCGTCTAGGTAAGTTTAAAG
CGTCCTTTTCACTCCAATGTCA



CTC (SEQ ID NO: 42)
(SEQ ID NO: 43)





ATF5
CCGTCTAGGTAAGTTTAAAG
GTGAAATCAACTCGCTCAGTC



CTC (SEQ ID NO: 44)
(SEQ ID NO: 45)









qRT-PCR was performed using Power SYBR® Green PCR Master Mix (Applied Biosystems) on MX3000P Sequence Detection System (Stratagene). Primers used are shown in Table 3.









TABLE 3







Primers used for qRT-PCR, Related to FIG. 3









Gene
Forward Primer (5′→3′)
Reverse Primer (5′→3′)





ALB
GCACAGAATCCTTGGTGA
ATGGAAGGTGAATGTTTCA



ACAG (SEQ ID NO: 46)
GCA (SEQ ID NO: 47)





CEBPA
ACAAGAACAGCAACGAG
CATTGTCACTGGTCAGCTC



TACCG (SEQ ID NO: 48)
CA (SEQ ID NO: 49)





FOXA1
GTGGCTCCAGGATGTTAG
AGGCCTGAGTTCATGTTGC



GA (SEQ ID NO: 50)
T (SEQ ID NO: 51)





FOXA2
CGACTGGAGCAGCTACTA
TACGTGTTCATGCCGTTCAT



TGC (SEQ ID NO: 52)
(SEQ ID NO: 53)





FOXA3
CTGGCCGAGTGGAGCTAC
AGGGGGATAGGGAGAGCT



TA (SEQ ID NO: 54)
TA (SEQ ID NO: 55)





HNF1A
CCATCCTCAAAGAGCTGG
GTGCTGCTGCAGGTAGGAC



AG (SEQ ID NO: 56)
T (SEQ ID NO: 57)





HNF4A
CCAAAACCCTCGTCGACA
TTCTCAAATTCCAGGGTGG



TG (SEQ ID NO: 58)
TGTA (SEQ ID NO: 59) 





HNF6
TGTGGAAGTGGCTGCAG
TGTGAAGACCAACCTGGGC



GA (SEQ ID NO: 60)
T (SEQ ID NO: 61)





ONECUT2
CGAACACTCTTCGCCATC
GTTGCTGACGGTTGTGAGC



TTC (SEQ ID NO: 62)
TC (SEQ ID NO: 63)





PROX1
ACAGGGCTCTGAACATGC
GGCATTGAAAAACTCCCGT



AC (SEQ ID NO: 64)
A (SEQ ID NO: 65)





LRH1
CGAGTGGGCCAGGAGTA
CGGTAAATGTGGTCGAGGA



GTA (SEQ ID NO: 66)
T (SEQ ID NO: 67)





GATA4
CCCGACACCCCAATCTC
CAGGCGTTGCACAGATAGT



(SEQ ID NO: 68)
G (SEQ ID NO: 69)





GATA6
CCAACTTCCACCTCTTCT
TCTTGACCCGAATACTTGA



AACTCAG (SEQ ID NO: 70)
GCTC (SEQ ID NO: 71)





ATF5
CTATGAGGTCCTTGGGGG
CTCGCTCAGTCATCCAGTC



AG (SEQ ID NO: 72)
A (SEQ ID NO: 73)





USF1
ACAGTTGGAGAAAATCG
ATCCGAGGAACTGGTCCTT



GCA (SEQ ID NO: 74)
T (SEQ ID NO: 75)





USF2
TTGATGGAACCAGAACA
AGCTGGACGATCCAGTTGT



CCC (SEQ ID NO: 76)
T (SEQ ID NO: 77)





XBP1
GTGAGCTGGAACAGCAA
CCAAGCGCTGTCTTAACT



GTG (SEQ ID NO: 78)
C (SEQ ID NO: 79)





ZHX2
GGTCTGGATGTACCGACT
AAAATTGGAATGGCACCAA



GC (SEQ ID NO: 80)
C (SEQ ID NO: 81)





NFIA
ACCCCATCACATAGGGGT
TAATGTCAGCGTCACTTGG



TT (SEQ ID NO: 82)
C (SEQ ID NO: 83)





PXR
TTGCCCATCGAGGACCAG
GTCTCCGCGTTGAACACTG



AT (SEQ ID NO: 84)
T (SEQ ID NO: 85)





CAR
GTCCCACCTGCCCCTTTG
AGTGGCGCCTCTGAGTCTT



(SEQ ID NO: 86)
G (SEQ ID NO: 87)





FXR
CAGGATTTCAGACTTTGG
CTTCAACCGCAGACCCTTT



ACCAT (SEQ ID NO: 88)
C (SEQ ID NO: 89)





PPARA
AGAGATTTCGCAATCCAT
ACTGGTATTCCGTAAAGCC



CGG (SEQ ID NO: 90)
AAAG (SEQ ID NO: 91)





AHR
ACATCACCTACGCCAGTC
CGCTTGGAAGGATTTGACT



G(SEQ ID NO: 92)
TGA (SEQ ID NO: 93)





PPARG
TACTGTCGGTTTCAGAAATG
GTCAGCGGACTCTGGATTCAG



CC (SEQ ID NO: 94)
(SEQ ID NO: 95)





PPARD
GTGATCCACGACATCGAGAC
TGCACGCTGATCTCCTTGTAG



A (SEQ ID NO: 96)
(SEQ ID NO: 97)





LXRA
CCTTCAGAACCCACAGAGAT
ACGCTGCATAGCTCGTTCC



CC (SEQ ID NO: 98)
(SEQ ID NO: 99)





VDR
TCTCCAATCTGGATCTGAGT
ACAGCTCTAGGGTCACAGAAG



GAA (SEQ ID NO: 100)
(SEQ ID NO: 101)





GR
CCAACGGTGGCAATGTGAA
CCAAGGACTCTCATTCGTCTCT



AT (SEQ ID NO: 102)
T (SEQ ID NO: 103)





CYP2E1
CTGACCACCCTCCGGAACTA
GGCCTTGGGTCTTCCTGAGT



T (SEQ ID NO: 104)
(SEQ ID NO: 105)





CYP2D6
GTGTCCAACAGGAGATC
CACCTCATGAATCACGGCA



GACG (SEQ ID NO: 106)
GT (SEQ ID NO: 107)





CYP2C19
GAAGAGGAGCATTGAGG
GCCCAGGATGAAAGTGGG



ACCG (SEQ ID NO: 108)
AT (SEQ ID NO: 109)





CYP2C9
GCCACATGCCCTACACAG
TAATGTCACAGGTCACTGC



ATG (SEQ ID NO: 110)
ATGG (SEQ ID NO: 111)





CYP1A2
CTTCGTAAACCAGTGGCA
AGGGCTTGTTAATGGCAGT



GG (SEQ ID NO: 112)
G (SEQ ID NO: 113)





CYP3A4
AGCCTGGTGCTCCTCTAT
CCCTTATGGTAGGACAAAA



CT (SEQ ID NO: 114)
T (SEQ ID NO: 115)





CYP2B6
CCGGGGATATGGTGTGAT
CCGAAGTCCCTCATAGTGG



CTT (SEQ ID NO: 116)
TC (SEQ ID NO: 117)





CYP2A6
GAGTTCCTGTCACTGTTG
GTCCTGGCAGGTGTTTCAT



CG (SEQ ID NO: 118)
C (SEQ ID NO: 119)





UGT1A1
CCATCATGCCCAATATGG
CCACAATTCCATGTTCTCC



TT (SEQ ID NO: 120)
A (SEQ ID NO: 121)





UGT1A3
GCCAACAGGAAGCCACT
CAGCAATTGCCATAGCTTT



ATC (SEQ ID NO: 122)
C (SEQ ID NO: 123)





UGT1A4
AACGGGAAGCCACTATCT
TCAGCAATTGCCATAGCTT



CA (SEQ ID NO: 124)
TC (SEQ ID NO: 125)





UGT1A6
AATTTCCTAAAGGCCGGT
TTGATCCCAAAGAGAAAAC



CA (SEQ ID NO: 126)
CA (SEQ ID NO: 127)





UGT1A9
ACTATCCCAAACCCGTGA
ACCACAATTCCATGTTCTC



TG (SEQ ID NO: 128)
CA (SEQ ID NO: 129)





UGT2B7
AACGTAATTGCATCAGCC
GGTCATTCTGGGGTATCCA



CT (SEQ ID NO: 130)
C (SEQ ID NO: 131)





UGT2B15
GTTTTCTCTGGGGTCGAT
ATTTGGCTTCTTGCCATCAA



GA (SEQ ID NO: 132)
(SEQ ID NO: 133)





NAT2
CAGCCTAGTTCCTGGTTG
GGATCTGGTGCTCAAGAAT



CT (SEQ ID NO: 134)
G (SEQ ID NO: 135)





BCRP
CTGAGATCCTGAGCCTTT
AAGCCATTGGTGTTTCCTT



GG (SEQ ID NO: 136)
G (SEQ ID NO: 137)





OATP1B1
TTCAATCATGGACCAAAA
TGAGTGACAGAGCTGCCAA



TCAA (SEQ ID NO: 138)
G (SEQ ID NO: 139)





OATP1B3
GAAAACAAGACGCTGCA
TCCTTTCTATTTGAGTGATG



ATG (SEQ ID NO: 140)
GAAA (SEQ ID NO: 141)





NTCP
AGGGGGACATGAACCTC
AGGTCCCCATCATAGATCC



AG (SEQ ID NO: 142)
C (SEQ ID NO: 143)





GAPDH
TGCACCACCAACTGCTTA
GGCATGGACTGTGGTCATG



GC (SEQ ID NO: 144)
AG (SEQ ID NO: 145)









Primer for 18s rRNA was purchased from QIAGEN. Quantified values were normalized against the input determined by two housekeeping genes (GAPDH or RRN18S). For the positive control in qRT-PCR, five different batches of fresh isolated primary human hepatocytes were collected in RNAprotect (Qiagen) and stored at −20° C. Total RNA was isolated and then reverse-transcribed to cDNA as described above. Equal volumes of cDNA obtained from five different batches of freshly isolated primary human hepatocytes were mixed to be taken as the positive control.


Immunofluorescence and Flow Cytometric Analysis


Cells or tissue sections were fixed in 4% paraformaldehyde (Dingguo) at room temperature for 15 minutes and blocked with PBS containing 0.25% Triton X-100 and 5% normal donkey serum (Jackson ImmunoResearch Laboratories, Inc) at room temperature for 1 hour or at 4° C. overnight. Samples were incubated with primary antibodies at 4° C. overnight, washed three times with PBS and then incubated with appropriate secondary antibodies for 1 hour at room temperature in the dark. Nuclei were stained with DAPI (Roche). Experiments were repeated for three times and typical results were shown. The primary antibodies used for immuno-fluorescence are as follows: rabbit anti CYP3A4, rabbit anti CYP2C9, rabbit anti YP1A2, rabbit anti CYP2E1, rabbit anti CYP2D6 (all from AbD Serotec), Goat anti ALB (Bethyl Laboratories, INC), Rabbit anti NR5A2/LRH1 (Abeam), Rabbit anti COL1A1 (Abeam), Mouse anti E-CAD (Abeam), Mouse anti human nuclei (Millipore). The secondary antibodies used for immunofluorescence are as follows: DyLight® 550 Donkey anti rabbit and DyLight® 550 Donkey anti goat (from Abeam), DyLight 488 donkey anti goat Dylight 549 donkey anti goat, DyLight 488 donkey anti mouse, Dylight 549 donkey anti mouse, DyLight 488 donkey anti rabbit, Dylight 549 donkey anti rabbit (all from Jackson ImmunoResearch Laboratories). Flow cytometric assays were conducted as reported previously (Zhao et al., Cell Res., 23:157-161 (2013)).


RNA-Sequence Analysis


Total RNA was isolated from HEFs, HepG2 cells, ES-Heps, hiHeps and freshly isolated primary human hepatocytes. RNA sequencing libraries were prepared with the Illumina TruSeq RNA Sample Preparation Kit. The fragmented and randomly primed 200-bp paired-end libraries were sequenced on Illumina HiSeq 2000 sequencing system.


Toxicity Assays.


hiHeps were incubated with various concentrations of compounds dissolved in culture medium for 24 h. Cell viability was measured by MTT assay (Invitrogen) following the manufacturer's instructions and as described previously (Khetani and Bhatia, Nat Biotechnol 26, 120-126 (2008)).


Animals and Transplantation


Tet-uPA/Rag2−/−/γc−/− mice on a BALB/c background were purchased from Beijing Vitalstar Biotechnology. hiHeps, ES-Heps, and primary human hepatocytes (2×106 cells/animal) were injected into the spleens of the mice. Blood samples were collected and human ALBUMIN was quantified using the Human Albumin ELISA Quantitation kit (Bethyl Laboratories). Livers of recipient mice were embedded in OCT compound (Sakura) and then frozen in liquid nitrogen. Cryostat sections (10 mm) were stained.


Statistical Analysis


For statistical analysis, a two-tailed unpaired t test was used. Results are expressed as mean±SD. p values are as follows: *p<0.05; **p<0.01; ***p<0.001.


Accession Numbers


RNA-sequencing data have been deposited in the NCBI Gene Expression Omnibus database under accession number GSE54066.


Results


Identification of Factors that Induce Hepatic Fate


To identify the combination of transcription factors that induce human embryonic fibroblasts (HEFs) into hepatocytes, a pool of transcription factors (Table 4) that were previously shown to be expressed in human hepatocytes and are crucial to the determination of hepatic cell fate was selected (Nagaoka and Duncan, Prog. Mol. Biol Transl Sci., 97:79-101 (2010); Zaret, Nat. Rev. Genet., 9:329-340 (2008)).









TABLE 4







Transcription Factors Analyzed in Freshly Isolated Primary


Human Hepatocyte










Gene
GeneBank Accession







FOXA1
NM_004496



FOXA2
NM_021784



PROX1
NM_001270616



CEBPA
NM_004364



HNF1A
NM_000545



HNF4A
NM_178849



HNF6
NM_004498



GATA6
NM_005257



PPARA
NM_005036



ZHX2
NM_014943



LRH1
NM_205860



ONECUT2
NM_004852



ATF5
NM_001193646



USF2
NM_003367



USF1
NM_007122



ZGPAT
NM_032527



NF1A
NM_001134673










Previous studies also showed that proliferation arrest and cell death are general barriers to cell reprogramming (Huang et al., Nature, 475:386-389 (2011); Zhao et al., Cell Stem Cell, 3:475-479 (2008)). Thus, MYC was employed in the reprogramming process, as well as p53 small interfering RNA (siRNA) was employed in the reprogramming process. Briefly, HNF1A and HNF4A are preferentially considered because of their critical role in both embryonic and adult liver among the 17 transcription factors. Then additional factors were screened using a “2+1” strategy by the addition of one candidate factor at a time to the combination of HNF1A and HNF4A.


The data showed that HNF6, cooperating with HNF4A and HNF1A, can result in a high percentage of Albumin (ALB)-positive cells within 20 days (data not shown). These three factor induced hepatocyte-like cells (3H cells) exhibited some hepatic properties, including glycogen synthesis and low-density lipoprotein (LDL) uptake (data not shown). However, the expression level of ALB in these cells was extremely low (FIG. 1A). Moreover, the expression of the major cytochrome P450 enzymes in hepatocytes was not detected in these cells (data not shown). Therefore, the 3H cells appear to be functionally immature, implying that additional factors are required for their full maturation.


Identification of Factors that Generate Mature Hepatocytes


To identify the factors capable of inducing the functional maturation of hepatocyte-like cells, a global gene expression analysis was performed on 3H cells, freshly isolated primary human hepatocytes (F-HEPs), and fetal liver cells. Differential expression of several hepatic transcription factors, including CEBPA, ATF5, and PROX1, was observed among the three samples (data not shown). These three genes were expressed at relatively low levels in the 3H cells and in fetal hepatocytes compared to the levels in adult hepatocytes. This difference was further confirmed by quantitative PCR (FIGS. 1B and 1C). Among these genes, PROX1 was shown in a recent study to be a key transcription factor that is critical in the metabolic maturation of hepatocytes (Zhao et al., Cell Res., 23:157-161 (2013)). CEBPA and ATF5 are highly abundant liver-enriched transcription factors, indicating the importance of transcriptional regulation in hepatic function. Furthermore, a gene expression study showed that these three genes were highly expressed in F-HEPs (FIG. 1D). Collectively, these data showed that overexpressing these factors can lead to the functional maturation of 3H cells.


To generate mature human hepatocytes from fibroblasts, the three factors with CEBPA, PROX1, and ATF5, were combined, and overexpressed in HEFs following the scheme shown in FIG. 1E. A dramatic morphological change of fibroblasts into epithelial cells was observed in 1 week. These cells proliferated rapidly in hepatocyte culture medium (HCM), with the doubling time ranging from 9 to 11 hr (FIG. 1F). At 2 weeks post infection, the replated cells showed the typical morphology of primary human hepatocytes (data not shown). At about 25 days postinfection, p53 siRNA was silenced, as indicated by a GFP reporter (data not shown), and the induced cells were transferred to a modified William's E medium (FIGS. 1E and 1F). Quantitative PCR results showed that the induced hepatocyte-like cells expressed ALB at a level that was comparable to that of primary human hepatocytes (FIG. 1G), which was significantly higher than that of 3H cells (FIG. 1A). The reprogramming efficiency was further analyzed and found that 90% of the induced cells were ALB positive and nearly 100% were α-1 antitrypsin (AAT) positive (FIGS. 1H and 1I). The secretion of ALB was dramatically enhanced and was comparable to that of primary human hepatocytes (FIG. 1J). Furthermore, the four major cytochrome P450 enzymes, CYP3A4, CYP1A2, CYP2C9, and CYP2C19, were also expressed in the induced cells as detected by immunostaining (data not shown). Removal of any of these six factors would lead to a substantial decrease in the expression of drug metabolic enzymes and transporters (FIG. 1K). These results indicate that functional hepatic properties were obtained in these induced hepatocyte-like cells, which were termed hiHeps.


hiHeps Possess the Typical Characteristics of Human Hepatocytes


To evaluate hepatic fate conversion, typical hepatic features were first analyzed. Immunofluorescence microscopy showed that the epithelial marker E-cadherin (ECAD) was coexpressed with ALB in hiHeps (data not shown). In addition, the fibroblast marker COL1A1 was not detected (data not shown). These results indicate a successful mesenchymal-epithelial transition in hiHeps. Next, endogenous hepatic transcription network activation in hiHeps was further examined using RT-PCT.


The RT-PCR results showed that the endogenous expression of FOXA1, FOXA2, and FOXA3 (Zaret et al., Nat. Rev. Genet., 9:329-340 (2008)) was activated in iHeps (FIG. 2A). LRH1, another core transcription factor involved in the hepatic cross-regulatory network (Nagaoka and Duncan, Prog. Mol. Biol Transl Sci., 97:79-101 (2010)), was also endogenously expressed in hiHeps (FIG. 2A).


The expression of FOXA2 and LRH1 was confirmed using immunofluorescence (data not shown). Additionally, fibroblast marker genes, including COL1A1, PDGFRB, and THY1, were not detected in hiHeps (FIG. 2A). In accordance with p53 siRNA silencing exogenous expression of HNF1A, HNF6, HNF4A, ATF5, PROX1, and CEBPA was silenced in hiHeps (FIG. 2B). The primers used in FIG. 2A can specifically identify endogenous transcripts of HNF1A, HNF4A, PROX1 and CEBPA. These primers are designed to bind to the unique 5′UTR or 3′UTR of endogenous transcripts rather than coding sequences. In addition, MYC was decreased in iHeps to a level lower than that of freshly isolated primary human hepatocytes, as revealed by quantitative RT-PCR (qRT-PCR) (FIG. 2C). Collectively, these data indicate that hiHeps gain a hepatic transcription network.


Next, hiHeps was evaluated for functional characteristics of human hepatocytes. hiHeps were competent for LDL uptake (data not shown). In addition, hiHeps could incorporate indocyanine green (ICG) from the medium and exclude the absorbed ICG after withdrawal (data not shown). Oil red O staining in hiHeps showed an accumulation of fatty droplets, and Periodic Acid-Schiff (PAS) staining indicated glycogen synthesis (data not shown). Similar to human adult hepatocytes, hiHeps were AFP negative (data not shown). G banding analysis revealed that hiHeps had a normal karyotype after 7 weeks of culture (data not shown). Besides HEFs, similar results were obtained when adult foreskin fibroblasts were converted as described herein using the same factors (data not shown). Collectively, these results indicate that hiHeps exhibit typical hepatic functional features.


The global gene expression patterns in hiHeps and F-HEPs were compared by RNA sequencing. Principle component analysis and hierarchical clustering analysis revealed that hiHeps established from different donors were clustered with human hepatocytes and separated from human fibroblasts, HepG2 cells, and human embryonic stem cell (ESC)-derived hepatocytes (ES-Heps) (data not shown). Indeed, hepatic transcription factors were upregulated (As it is depicted in FIG. 2A, these factors are FOXA1, FOXA2, FOXA3, CEBPA, HNF1A, HNF4A, PROX1 and LRH1) and the expression of fibroblast signature genes (As it is depicted in FIG. 2A, these factors are PDGFB1, THY1 and COL1A1) was downregulated in hiHeps (data not shown). Additionally, hiHeps displayed the gene expression patterns of hepatocytes in a set of genes involved in lipoprotein, cholesterol, fat, glucose, and drug metabolism (data not shown). Altogether, these results indicate that hiHeps show a similar expression profile to primary human hepatocytes.


Establishment of the Central Network of Drug Metabolism in hiHeps


To evaluate whether hiHeps expressed key enzymes in drug metabolism, the expression in hiHeps of five key CYP enzymes, CYP1A2, CYP2B6, CYP2C9, CYP2C19, and CYP3A4 in hiHeps was quantitatively confirmed. The five key CYPs are major phase I enzymes that account for 60% of human drug oxidation (Zhou et al., Drug Metab. Rev., 41:89-295 (2009)). As the positive control, pooled F-HEPs from five individual donors were used. Notably, comparable mRNA levels of these major CYPs could be detected in hiHeps and F-HEPs, in contrast to their expression in hepatocytes derived from human ESCs and HepG2 cells (FIG. 3A). Next, hiHeps were analyzed for the presence of phase II enzymes and phase III transporters, which are important for the excretion of xenobiotic drugs. The expression levels of these genes were similar to those in F-HEPs (FIGS. 3B-3D). Additionally, hiHeps expressed a broad spectrum of phase I and phase II metabolic enzymes and phase III transporters (FIG. 3E). Collectively, these findings show that the central network of drug metabolism was successfully established in hiHeps and resembled that of pooled freshly isolated primary human hepatocytes.


Level of Key Drug Metabolic Activities in hiHeps is Comparable to that in Freshly Isolated Primary Human Hepatocytes


To evaluate the drug metabolic activities of hiHeps, the studies first focused on CYP3A4. Using ultraperformance liquid chromatography-tandem mass spectrometry technology, the drug metabolic activity of CYP3A4 in hiHeps was detected by using two structurally different substrates, testosterone and midazolam. Because of the remarkable interindividual variability in drug clearance, two batches of freshly isolated primary human hepatocytes were used as the positive control. In contrast to the HepG2 cell line, ES-Heps, and HEFs, hiHeps were able to metabolize the two CYP3A4-selective substrates efficiently and the metabolism efficiency is comparable to the metabolism seen with freshly isolated hepatocytes (F-HEPs) (FIG. 4A). Zhao, et al. disclose that ES-Heps express CYP3A4 with activities at levels that are lower than those seen in 25-week-old fetal hepatocytes and human adult primary hepatocytes (Zhao, et al., Cell Res., 23:157-161 (2013)). Furthermore, the metabolic activities of CYP1A2 and CYP2B6 in hiHeps were found to be comparable to that of F-HEPs (FIG. 4A). The activities of CYP2C9 and CYP2C19 in hiHeps were approximately 30% of F-HEPs (FIG. 4A). The metabolic activities of all these CYP enzymes in hiHeps were at least 100-fold higher than that of ES-Heps. These data indicate that hiHeps exhibit comparable metabolic activities of the key CYP enzymes to those of freshly isolated primary human hepatocytes.


To further evaluate the functional central network of drug metabolism in hiHeps, the expression of nuclear receptors between hiHeps and F-HEPs, which are critical in regulating the expression of metabolizing enzymes, was compared. Nuclear receptors that are responsible for the xenobiotic metabolizing system were expressed in hiHeps (FIG. 3F). Moreover, hiHeps responded to the standard inducers of CYP3A4, CYP1A2, and CYP2B6 at the mRNA level (FIG. 4B). Taken together, these data show a functional establishment of the nuclear receptor network in hiHeps.


To assess the potential application of hiHeps in studying hepatotoxicity, acute toxicity of model hepatotoxins were quantified. As hepatotoxicity is the most common adverse event resulting in drug failure (Sahi et al., Curr. Drug Discov. Technol., 7:188-198 2010), the sensitivity of drug toxicity is a key index for the potential application of human hepatocytes in drug discovery. hiHeps showed a level of sensitivity comparable to that of primary human hepatocytes when incubated with a series of model hepatotoxins (FIG. 4C), showing the potential of using hiHeps for testing drug toxicity.


Repopulation of Tet-uPA/Rag2−/−γc−/− Mouse Liver with hiHeps


To investigate the capacity of hiHeps to repopulate mouse liver, Tet-uPA (urokinase-type plasminogen activator)/Rag2−/−/γc−/− mice were injected intrasplenically with hiHeps (Song et al., Am. J. Pathol., 175:1975-1983 (2009)). The secretion of human Albumin in mouse serum increased gradually and the highest level reached was 313 mg/ml at 7 weeks after hiHep transplantation (FIGS. 5A-5C), which was 1,000-fold higher than ES-Heps and comparable to primary human hepatocytes (FIG. 5B). To analyze the engraftment efficiency, hepatocytes were isolated from whole liver of two mice and measured by flow cytometry analysis. The repopulation efficiency was about 30% in the mouse that secreted 313 mg/ml human Albumin (FIG. 4C). No tumorigenesis was observed 2 months after hiHep transplantation. Grafts of hiHeps were also analyzed. Six weeks after transplantation, clusters of cells expressing human ALB were observed in the recipient mice (data not shown). To confirm the metabolic function of hiHeps in vivo, CYP expression was analyzed. The expression of major CYPs including CYP3A4, CYP2C9, CYP1A2, CYP2E1, CYP2C19, and CYP2D6 (data not shown) indicated that hiHeps can be functional in vivo. Collectively, these results show that hiHeps can robustly repopulate the liver of Tet-uPA/Rag2−/−/γc−/− mice and were functional in vivo.


Discussion


These studies show that human hiHeps are readily and reproducibly generated from HEFs using a combination of hepatic fate conversion factors HNF1A, HNF4A, and HNF6 together with the maturation factors ATF5, PROX1, and CEBPA. Similar to primary human hepatocytes, hiHeps exhibit many typical hepatic features, including their epithelial morphology, expression of hepatocyte specific markers, basic functional properties of hepatocytes, and global gene expression patterns. Importantly, an integral spectrum of phase I and phase II drug-metabolizing enzymes and phase III drug transporters is well established in hiHeps. Furthermore, transplanted hiHeps can repopulate up to 30% of the livers of Tet-uPA/Rag2−/−/γc−/− mice and secrete more than 300 mg/ml human albumin in vivo. This data shows that human hepatocytes with drug-metabolizing functions can be generated from fibroblasts using lineage reprogramming. One key question in lineage reprogramming is how to obtain fully functional cells. In hepatic transdifferentiation, mouse induced hepatocyte-like cells were generated with several important hepatic characteristics, through the expression of hepatic fate determination factors in fibroblasts (Huang et al., 2011; Sekiya and Suzuki, Nature, 475:390-393 (2011)). However, incomplete hepatocyte differentiation and expression of certain hepatoblast markers by hiHeps are compatible with an immature or progenitor-like state (Willenbring, Cell Stem Cell, 9:89-91 (2011)). These studies also show that that certain hepatic fate determination factors could reprogram HEFs into hepatocyte-like cells. However, these cells are not functional until the addition of three additional factors (FIGS. 1G-1J). The additional three factors promote further metabolic maturation of hiHeps (data not shown). Thus, hepatic fate determination and hepatic functional maturation may be governed by different master genes and are somewhat independent of each other. To obtain fully functional cells, the ectopic expression of cell fate determination factors may not be sufficient, and additional functional maturation factors are required to promote this process.


The drug metabolic capacity of human hepatocytes is one of the most important functions that distinguish hepatocytes from other lineages and has broad applications in drug development. Efforts to differentiate human pluripotent stem cells into hepatocytes have resulted in cells that were functionally immature. A recent study showed that human ES-Heps express CYP1A2 and CYP3A4 (Zhao et al., Cell Res., 23:157-161 (2013)). However, the activities of these two CYP enzymes were significantly lower than that of primary hepatocytes. In another study, differentiated hepatocytes exhibited CYP3A4 and CYP1A2 activities only comparable to that of cultured primary hepatocytes (Ogawa et al., Development, 140:3285-3296 2013). However, a number of liver-essential functions were progressively lost with time in cultured primary hepatocytes (Elaut et al., Curr. Drug Metab. 7:629-660 (2006)). In the studies disclosed herein, the gold standard, freshly isolated primary human hepatocytes, was used as the positive control. The hiHeps disclosed herein express the key phase I (CYP3A4, CYP2C9, CYP2C19, CYP2B6, and CYP1A2) and phase II drug-metabolizing enzymes and phase III drug transporters at a level comparable to that of freshly isolated primary human hepatocytes. Importantly, the metabolic activities of the five CYP enzymes in hiHeps were comparable to those in freshly isolated primary human hepatocytes, indicating the potential application of hiHeps in evaluating drugs metabolized by these CYP enzymes (FIG. 4A). The expression of endogenous nuclear receptors related to xenobiotic metabolizing systems was also detected in these cells (Nakata et al., Drug Metab. Pharmacokinet., 21:437-457 (2006)) (FIG. 3F). Moreover, the expression of CYP3A4, CYP1A2, and CYP2B6 was increased by the standard inducers (FIG. 4B). In addition, because integrated metabolism pathways (phase I and phase II enzymes and phase III drug transporters) in hepatocytes are of vital importance for drug discovery (Castell et al., Expert Opin. Drug Metab. Toxicol. 2:183-212 (2006)), the drug metabolic network of hiHeps was closely analyzed. The expression pattern of genes encoding the drug metabolizing markers was similar to that in primary human hepatocytes, implying an upregulation of the drug metabolic network in hiHeps (FIGS. 3A-3F). Collectively, these results indicate the integral establishment of the central network of functional drug metabolism in hiHeps, making these cells a potential alternative for preclinical screening assays.


Another key characteristic of human hepatocytes in drug development is their sensitivity to drug toxicity. Human hepatocytes derived from human pluripotent stem cells have a relatively low sensitivity to drug toxicity (Zhao et al., Cell Res., 23:157-161 (2013)). By contrast, the sensitivity of hiHeps disclosed herein to multiple model hepatotoxins is comparable to that of primary human hepatocytes (FIG. 4C). Thus, hiHeps can be a valuable alternative cell resource in hepatotoxicity assays for new drug discovery. Importantly, our results demonstrate that the induced cells could be expanded at a large scale at an early stage (FIG. 1F), and the function of hiHeps could be maintained for 16 days (FIG. 4D). Considering the reprogramming efficiency (FIGS. 1H and 1I), more than 1011 functional hi-Heps can be obtained starting from 104 of fibroblasts (data not shown). These results show that hiHeps could be used in a practical manner for pharmaceutical development.


Hepatocyte transplantation is a promising alternative to orthotopic liver transplantation (Dhawan et al., Nat Rev Gastroenterol Hepatol, 7:288-298 (2010)). However, the limited supply of donor organs that can provide good-quality cells remains a major challenge. In the studies described herein, hiHeps were able to repopulate mouse liver robustly and secreted up to 313 mg/ml human ALBUMIN, which is two orders of magnitude higher than recent studies using human hepatocytes derived from human embryonic stem cells (FIGS. 5A and 5B) (Takebe et al., Nature, 499:481-484 (2013); Woo et al., Gastroenterology, 142:602-611 (2012)). Furthermore, transplanted hiHeps expressed major CYP enzymes (data not shown), indicating that hiHeps retained drug metabolic capabilities in vivo. Collectively, hiHeps can serve as a potential cell source for the establishment of a humanized mouse model and hepatocyte transplantation.


In conclusion, human hepatocytes were generated with drug metabolizing functions using the combined expression of cell fate determination factors and cell maturation factors. The generation of functional human hepatocytes with lineage reprogramming provides a way to obtain well-characterized, reproducible, and functional human hepatocytes for pharmaceutical applications.

Claims
  • 1. A method for inducing human fibroblast cells into induced hepatocytes-like cells (iHeps), comprising the steps of: (a) introducing one or more nucleic acid sequences encoding six factors, wherein the six factors are Hepatocyte nuclear factor 1-alpha (HNF1A), Hepatocyte nuclear factor 4-alpha (HNF4A), Hepatocyte nuclear factor 6-alpha (HNF6), Activating transcription factor 5 (ATF5), Prospero homeobox protein (PROX1), and CCAAT/enhancer-binding protein alpha (CEBPA), into isolated fibroblasts;(b) culturing the cells from the step (a) in a somatic cell medium;(c) expanding the cells from the step (b) in a hepatocyte cell culture medium; and(d) culturing the cells from the step (c) in a hepatocyte maturation medium wherein expression of the six factors is effective to induce formation of iHeps, wherein the iHeps do not express α-fetoprotein, and wherein the iHeps secret albumin, and are capable of taking up LDL, incorporating indocyanine green (ICG), synthesizing and storing glycogen and accumulating fat.
  • 2. The method of claim 1, wherein the step (a) further comprises treating the cells to upregulate MYC and downregulate p53.
  • 3. The method of claim 2, wherein the step (a) comprises transfecting the cells with a vector expressing p53 siRNA and inhibiting the expression of p53 siRNA at the end of the step (c).
  • 4. The method of claim 1, wherein in the step (a) the cells are transformed with nucleic acids as set forth by SEQ ID NOs: 1-7, respectively.
  • 5. The method of claim 1, wherein in the step (b) the cells are cultured in the somatic cell culture medium for a period of at least 7 days.
  • 6. The method of claim 1 wherein in the step (c) the cells are cultured in the hepatocyte cell culture medium for a period of about 15 to 30 days, preferably, 18-30 days, more preferably about 18 days.
  • 7. The method of claim 1 wherein in the step (d) the cells are cultured in the hepatocyte maturation medium for a period of at least 5 days.
  • 8. The method of claim 1, wherein the fibroblast cells are derived from a mammal, the method further comprising identifying iHeps by detecting the expression of at least one hepatic marker selected from the group consisting of albumin, Cytochrome P450 (CYP)3A4 and CYPB6, glycogen synthesis and storage, and/or fatty droplet accumulation.
  • 9. The method of claim 8, wherein the mammal is selected from the group consisting of human, rat, mouse, monkey, dog, cat, cattle, rabbit, horse and pig.
  • 10. The method of claim 1, wherein the iHeps express at least one drug metabolizing enzyme selected from the group consisting of CYP3A4, CYPB6, CYP1A2, CYP2C9, CYP2C19, or combinations thereof and optionally, wherein MYC expression level in the iHeps is lower than the MYC expression level found in hepatocytes obtained from the corresponding organism.
  • 11. The method of claim 1, wherein the non-hepatocyte cells are fibroblast cells, and optionally wherein the iHeps express E-cadherin, the iHeps do not express one or more of the fibroblast marker genes selected from COL1A1, PDGFRB, THY1 and α-fetoprotein, or a combination thereof.
  • 12. The method of claim 1, wherein the iHeps at least one drug metabolic phase II enzyme or phase II transporter selected from the group consisting of CYP1A2, CYP2C9, CYP2C19, UDP glucuronosyltransferase (UGT)1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, GSTA1, UGT2B7, UGT2515, Microsomal glutathione-S-transferase 1 (MGST1), nicotinamide N-methyltransferase (NNMT), NTCP, organic anion-transporting polypeptide 1B3 (OATP1B3), Multidrug resistance protein(MRP)6, MRP2, Flavin-containing monooxygenase 5 (FMO5), Monoamine oxidase (MAO)A, MAOB, and epoxide hydrolase 1 (EPHX1).
  • 13. The method of claim 1, wherein the metabolic activity of at least one of CYP3A4, CYPB6, CYP1A2, CYP2C9, and CYP2C19 is at least 50% higher than the activity of the same enzyme in ES-Heps (embryonic stem cell (ESC)-derived hepatocytes) obtained from the same organism.
  • 14. The method of claim 13, wherein the metabolic activity of at least one of CYP3A4, CYPB6, CYP1A2, CYP2C9, and CYP2C19 is 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100% or more, higher than the activity in ES-Heps.
  • 15. The method of claim 13, wherein the metabolic activity of at least one of CYP3A4, CYPB6, CYP1A2, CYP2C9, and CYP2C19 is at least 100-fold higher than that of ES-Heps.
  • 16. A method for inducing human fibroblast cells into induced hepatocytes-like cells (iHeps), comprising the steps of: (a) introducing one or more nucleic acid sequences encoding HNF1A, HNF4A, HNF6, ATF5, PROX1, and CEBPA, into isolated human fibroblasts;(b) culturing the cells from the step (a) in a somatic cell medium;(c) expanding the cells from the step (b) in a hepatocyte cell culture medium; and(d) culturing the cells from the step (c) in a hepatocyte maturation medium such that iHeps form, wherein the iHeps do not express α-fetoprotein, and wherein the iHeps secret albumin, and are capable of taking up LDL, incorporating indocyanine green (ICG), synthesizing and storing glycogen and accumulating fat;the method further comprising identifying iHeps by detecting the expression of at least one hepatic marker selected from the group consisting of albumin, Cytochrome P450 (CYP)3A4 and CYPB6, glycogen synthesis and storage, and/or fat accumulation,wherein the mammal is a human.
  • 17. A method for inducing human fibroblast cells into induced hepatocytes-like cells (iHeps), comprising the steps of: (a) introducing one or more nucleic acid sequences encoding HNF1A, HNF4A, HNF6, ATF5, PROX1, and CEBPA, into isolated human fibroblasts;(b) culturing the cells from the step (a) in a somatic cell medium;(c) expanding the cells from the step (b) in a hepatocyte cell culture medium; and(d) culturing the cells from the step (c) in a hepatocyte maturation medium such that iHeps form, wherein the iHeps do not express α-fetoprotein, and wherein the iHeps secret albumin, and are capable of taking up LDL, incorporating indocyanine green (ICG), synthesizing and storing glycogen and accumulating fat.
Priority Claims (1)
Number Date Country Kind
201410048337.X Feb 2014 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2015/072232 2/4/2015 WO
Publishing Document Publishing Date Country Kind
WO2015/120776 8/20/2015 WO A
US Referenced Citations (6)
Number Name Date Kind
20110195056 Pryor Aug 2011 A1
20110280844 Yu Nov 2011 A1
20120196360 Okita Aug 2012 A1
20120231490 Mizuguchi Sep 2012 A1
20130071365 Suzuki Mar 2013 A1
20140242595 Yu Aug 2014 A1
Foreign Referenced Citations (6)
Number Date Country
2013252081 Jun 2013 JP
0244321 Jan 2002 WO
2011016588 Feb 2011 WO
2011130402 Oct 2011 WO
2012058868 May 2012 WO
2014039768 Mar 2014 WO
Non-Patent Literature Citations (33)
Entry
Sekiya (Nature, 475: 1-6, 2011) (Year: 2011).
Sekiya (Nature, 475: 1-6, 2011) supplemental materials.
Abe, et al., “N-terminal hydrophobic amino acids of activating transcription factor 5 (ATF5) protein confer interleukin 1β (IL-1β)-induced stabilization” J. Biol. Chem., 289(7):3888-900 (2014.).
Cai, et al., “Directed differentiation of human embryonic stem cells into functional hepatic cells,” Hepatology, 45(5):1229-39 (2007).
Courtois, et al., “Interaction of a liver-specific nuclear factor with the fibrinogen and alpha 1-antitrypsin promoters” Science, 238(4827):688-92 (1987).
Dhawan, et al., “Human hepatocyte transplantation: current experience and future challenges” Nat Rev Gastroenterol Hepatol, 7:288-98 (2010).
Gebhardt, et al., “New hepatocyte in vitro systems for drug metabolism: metabolic capacity and recommendations for application in basic research and drug development, standard operation procedures” Drug Metab. Rev. 35:145-213 ( 2003).
GENBANK Accession No. NM_001290746.1 , “Homo sapiens activating transcription factor 5 (ATF5), transcript variant 3, mRNA” 5 pages, first appeared Mar. 19, 2014, updated Oct. 8, 2016, retrieved Dec. 12, 2016.
Gonzalez, “Regulation of hepatocyte nuclear factor 4 alpha-mediated transcription” Drug Metab. Pharmacokinet., 23(1):2-7 (2008).
Huang , et al., “Induction of functional hepatocyte-like cells from mouse fibroblast by defined factors” Nature, 475:386-9 (2011).
Martinez, et al.,“Single-stranded antisense siRNAs guide target RNA cleavage in RNAi” Cell, 110:563-74 (2002).
Ogawa, et al., “Three-dimensional culture and cAMP signaling promote the maturation of human pluripotent stem cell-derived hepatocytes” Development, 140:3285-96 (2013).
Sahi, et al., “Hepatocytes as a tool in drug metabolism, transport and safety evaluations in drug discovery” Curr. Drug Discov.Technol., 7:188-98 (2010).
Schwartzenberger, et al., “Targeted gene transfer to human hematopoietic progenitor cell lines through the c-kit receptor” Blood, 87:472-8 (1996).
Seglen, “Preparation of isolated rat liver cells” Methods Cell Biol., 13:29-83 (1976).
Sekiya and Suzuki, “Direct conversion of mouse fibroblasts to hepatocyte-like cells by defined factors” Nature, 475:390-3 (2011).
Sladeck, et al., “Liver-enriched transcription factor HNF-4 is a novel member of the steroid hormone receptor superfamily” Genes Dev., 4(12B): 2353-65 (1990).
Song, et al., “A mouse model of inducible liver injury caused by tet-on regulated urokinase for studies of hepatocyte transplantation” Am. J. Pathol., 175:1975-83 (2009).
Takebe, et al., “Vascularized and functional human liver from an iPSC-derived organ bud transplant” Nature, 499:481-4 (2013).
Vierbuchen and Wernig, “Molecular roadblocks for cellular reprogramming” Mol. Cell, 47: 827-38 (2012).
Willenbring, “A simple code for installing hepatocyte function” Cell Stem Cell, 9:89-91 (2011).
Woo, et al., “Direct and indirect contribution of human embryonic stem cell-derived hepatocyte-like cells to liver repair in mice” Gastroenterology, 142:602-11 (2012).
Zhao, et al., “Two supporting factors greatly improve the efficiency of human iPSC generation” Cell Stem Cell, 3:475-9 (2008).
Zhao, et al., “Promotion of the efficient metabolic maturation of human pluripotent stem cell-derived hepatocytes by correcting specification defects” Cell Res., 23(1):157-61 (2013).
International Search Report for corresponding PCT application PCT/CN2015/072232 dated May 11, 2015.
Ji, et al., “Cell fate conversation: Direct induction of hepatocyte-like cells from fibroblasts”, J Cellular Biochem., 114(2):256-65 (2013).
Kigiso, et al., “Transdifferentiation of human fibroblasts into hepatocyte-like cells by defined transcriptional factors”, Hepatology Intl, 7(3):937-44 (2013).
Extended European Search Report for corresponding European application EP15148953 dated Jun. 2, 2017.
Canadian Office Action dated Jul. 4, 2018 Canadian Patent Application No. 2,936,526.
Wilkening, Stahl, and Bader—“Comparison of Primary Human Hepatocytes and Hepatoma Cell Line HEPG2 with regard to their Biotransformation Properties”—The American Society for Pharmacology and Experimental Therapeutics, vol. 31, No. 8, DMD 31: 1035-1042—(2003).
Office Action CA (Canada) 2,939,525 dated Jun. 18, 2019.
Office Action AU (Australia) 2015218082 dated Apr. 15, 2020.
Office Action CA (Canada) 2,939,525 dated Jun. 4, 2020.
Related Publications (1)
Number Date Country
20170218333 A1 Aug 2017 US