US20110311494A1 - Methods of reprogramming renal cells - Google Patents

Methods of reprogramming renal cells Download PDF

Info

Publication number
US20110311494A1
US20110311494A1 US13/203,277 US201013203277A US2011311494A1 US 20110311494 A1 US20110311494 A1 US 20110311494A1 US 201013203277 A US201013203277 A US 201013203277A US 2011311494 A1 US2011311494 A1 US 2011311494A1
Authority
US
United States
Prior art keywords
renal
cells
cell
differentiated
renal cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/203,277
Inventor
Dekel Benjamin
Orit Harari-Shteinberg
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tel HaShomer Medical Research Infrastructure and Services Ltd
Original Assignee
Tel HaShomer Medical Research Infrastructure and Services Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tel HaShomer Medical Research Infrastructure and Services Ltd filed Critical Tel HaShomer Medical Research Infrastructure and Services Ltd
Priority to US13/203,277 priority Critical patent/US20110311494A1/en
Assigned to TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD. reassignment TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DEKEL, BENJAMIN, HARARI-SHTEINBERG, ORIT
Publication of US20110311494A1 publication Critical patent/US20110311494A1/en
Assigned to TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD. reassignment TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD. CORRECTIVE ASSIGNMENT TO CORRECT THE INCORRECT INVENTOR NAME OF ORIT HARARI-SHTEINBERG PREVIOUSLY RECORDED ON REEL 026884 FRAME 0452. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT INDICATING THE CORRECT INVENTOR NAME OF ORIT HARARI-STEINBERG. Assignors: DEKEL, BENJAMIN, HARARI-STEINBERG, ORIT
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/22Urine; Urinary tract, e.g. kidney or bladder; Intraglomerular mesangial cells; Renal mesenchymal cells; Adrenal gland
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0687Renal stem cells; Renal progenitors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/06Anti-neoplasic drugs, anti-retroviral drugs, e.g. azacytidine, cyclophosphamide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/065Modulators of histone acetylation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/415Wnt; Frizzeled
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/25Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from renal cells, from cells of the urinary tract

Definitions

  • the renal cell population of the present invention for the preparation of a medicament for the treatment of a renal disorder.
  • Methylation state of the Six2 promoter was analyzed by MeDIP; GAPDH was used as negative control and D4Z4 as positive control. To evaluate the level of DNA methylation, the ratio of PCR products from immunoprecipitated DNA vs. input DNA was calculated.
  • FIGS. 8A-C are graphs and photographs illustrating Over-expression (OE) of Oct4 pluripotency gene in HAK cells.
  • Human Oct4 gene was introduced into HAK cells by lentiviral infection enabling OE of Oct4 simultaneously with a puromycin (Puro) resistance gene.
  • Puro puromycin
  • Oct4 transcript levels were up regulated following infection, subsequent effect was observed following Puro exposure as shown by qRT-PCR (A-B).
  • A-B qRT-PCR
  • Growing infected cells on Puro selection resulted in homogenous culture over-expressing Oct4 showing morphologic ‘switch’ into foci of small round cells (C).
  • the present invention in some embodiments thereof, relates to isolated populations of renal progenitor cells and methods of generating and using same.
  • FIGS. 1A-T , 2 A-T and 5 A-D Whilst reducing the present invention to practice, the present inventors showed that epigenetic ‘transcriptional’ reprogramming of HAK cells via VPA or TSA/5-Aza treatment can increase the expression of pluripotency-associated genes as demonstrated by qRT-PCR analysis ( FIGS. 1A-T , 2 A-T and 5 A-D), Western blot analysis ( FIG. 3 ) and flow cytometry analysis ( FIGS. 4A-N ).
  • pluripotency associated gene refers to a gene which is upregulated at least two fold, more preferably at least 5 fold in a pluripotent cell as compared to a non-pluripotent cell.
  • genes belonging to the klf (Knippel-like factor) family include, for example Klf1(NM — 010635, mouse and NM — 006563, human), Klf2 (NM — 008452, mouse and NM — 016270, human), Klf4 (NM — 010637, mouse and NM — 004235, human) and Klf5 (NM — 009769, mouse and NM — 001730, human).
  • renal stem cell associated gene refers to a gene that is upregulated at least 2 fold and more preferably at least 5 fold in a renal stem cell compared with a differentiated renal cell.
  • six2 and sall1 are up-regulated.
  • the present inventors have shown that if a Wnt antagonist is used in combination with a chromatin modifying agent, a synergistic effect is seen whereby the upreguation of renal stem cell specific genes in the cells is even more apparent ( FIGS. 7A-R ).
  • Wnt antagonists e.g. DKK1 and sFRP1
  • the present invention also contemplates other Wnt antagonists such as siRNAs, RNAzymes, DNAzymes, miRNAs and the like. Such wnt antagonists are typically transfected into the cells as described herein below.
  • a “pharmaceutical composition” refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • kidney development progresses through mesenchymal to epithelial conversion of the MM (Vimentin + ) to epithelial progenitors (E-cadherin + )
  • the present inventors analyzed whether the reinduction of Six2 and Osr1 and pluripotency genes recapitulates early renal development and is associated with Vimentin upregulation.
  • significant downregulation of Vimentin was found with no effect on E-cadherin levels ( FIGS. 1I-J ).
  • human fetal kidney cells derived from mid-gestation kidneys were evaluated.

Abstract

A method of reprogramming a differentiated renal cell towards a progenitor phenotype is disclosed. The method comprises up-regulating in the differentiated renal cell an expression of at least one pluripotency associated gene and/or at least one renal stem cell associated gene, thereby reprogramming the differentiated renal cell towards a progenitor phenotype. Cell populations generated thereby and uses thereof are also disclosed.

Description

    FIELD AND BACKGROUND OF THE INVENTION
  • The present invention, in some embodiments thereof, relates to isolated populations of renal progenitor cells and methods of generating and using same.
  • The fundamental functional unit of the kidney required for urine formation is the nephron. The number of nephrons in the human adult kidney (HAK) ranges from 300,000 to a million. The human metanephros (direct embryonic precursor tissue of the adult kidney) appears at the 5th week of gestation and renal stem/progenitor cells in a discrete region of the metanephros termed the metanephric/nephrogenic mesenchyme (MM) are induced to undergo mesenchymal-to-epithelial transition (MET) and form all epithelial cell types of the nephron until 34 weeks of gestation. These include glomerular (parietal, visceral) and tubular (proximal, distal) epithelia (which can be detected by segment-specific markers) but not collecting ducts. Thus, endowment of new nephrons is restricted to prenatal development in humans, while in rodents it persists only until the immediate postnatal period (up to 2 weeks postnatal).
  • Specifying the renal progenitor population throughout development are a unique combination of transcription factors, including the Hox11 paralogs, Osr1, Pax2, Eya1, Wt1, Sall1, Six2, and Cited1. These genes considered early markers of kidney progenitor cells are silenced, at least in part, by epigenetic modifications once nephrogenesis commences [1]. Epigenetic modifications responsible for transcriptional and lineage control are reversible and can be therefore manipulated. For example: methylation of lysine 27 on histone subunit H3 (H3K27me) by the Polycomb complex is associated with transcriptional repression, whereas methylation of lysine 4 on H3 (H3K4me) and any lysine acetylation of histones is associated with gene activation. Of all, recent experiments have established that Six2 and Osr1 are required to sustain a true committed stem cell in the MM that is capable of self-renewing and of differentiating towards different types of nephron epithelia [2-4].
  • Genetic diseases affecting the various cell types of the nephron are the leading cause of end-stage renal disease (ESRD) in the pediatric population which requires renal replacement therapy (dialysis, transplantation) and carries extremely high morbidity and mortality rates. Mutations in genes that specify the renal progenitor cell pool (see above) can lead to either congenital renal hypoplasia (too few nephrons) or to the appearance of malformed nephrons (congenital renal dysplasia). These can be viewed as ‘kidney stem cell diseases’. In contrast, mutations in genes that participate in the function of a highly specialized cell type in the kidney result in loss of that specific task. One such example are genetic defects in structural proteins of the glomerular podocyte (podocin, nephrin), an epithelial cell type responsible for the filtration barrier, which lead to massive spillage of protein to the urine and to the appearance of nephrotic syndrome, focal segmental glomerulosclerosis and ESRD (ref). In contrast to acquired childhood nephrotic syndrome which is sensitive to steroid treatment, the inherited disease is steroid-resistant.
  • The goal of renal regenerative medicine is to create an unlimited supply of human cells resembling the renal progenitors residing in the MM so as be able to replenish diseased ones in renal hypoplasia/dysplasia, affording enhanced nephron development or replace glomerular podocytes by inoculating a cell population containing the podocyte progenitor cell.
  • Derivation of renal stem/progenitors can be achieved by various strategies. One strategy is to sort them out from developing human kidneys via specific surface markers (similar to hematopoietic stem cells of the hematopoietic system). Another strategy is to induce differentiation of pluripotent cells (allogeneic human ES) along the renal lineage. A third strategy is to dedifferentiate autologous cell sources such as human adult kidney (HAK) cells into renal stem/progenitors.
  • Yamanaka and colleagues have shown the ability to reprogram cells into a pluripotent phenotype via the over-expression of specific factors, Oct4, c-Myc, Sox2, Klf4 [5]. Recently, Melton and colleagues reported the in vivo reprogramming of adult pancreatic exocrine cells to beta-cells using a strategy of re-expressing key developmental regulators of the endocrine pancreas [6], indicating a general paradigm for directing cell reprogramming and trans-differentiation across two mature cell types without reversion to a pluripotent stem cell state.
  • Metsuyanim et al teaches that the cessation of nephrogenesis is coincident with dramatic down-regulation of the renal progenitor genes, which is associated in part with epigenetic silencing [1].
  • U.S. Patent Application No. 20060177925 teaches multipotent renal progenitor cells (MRPC) that are antigen positive for vimentin and Oct-4, and are antigen negative for zona occludens, cytokeratin, and major histocompatibility Class I and II molecules.
  • SUMMARY OF THE INVENTION
  • According to an aspect of some embodiments of the present invention there is provided a method of reprogramming a differentiated renal cell towards a progenitor phenotype, the method comprising up-regulating in the differentiated renal cell an expression of at least one pluripotency associated gene and/or at least one renal stem cell associated gene, thereby reprogramming the differentiated renal cell towards a progenitor phenotype.
  • According to an aspect of some embodiments of the present invention there is provided a renal cell population generated according to the method of the present invention.
  • According to an aspect of some embodiments of the present invention there is provided a method of repairing or regenerating a renal tissue in a subject in need thereof the method comprising administering to the subject a therapeutically effective amount of the renal cell population of the present invention, thereby repairing or regenerating renal tissue in a subject.
  • According to an aspect of some embodiments of the present invention there is provided a method of monitoring differentiation state of a renal cell, the method comprising determining in the renal cell expression of a pluripotency associated gene selected from the group consisting of Oct4, Nanog and klf4, and/or at least one renal stem cell associated gene selected from the group consisting of Six2, Osr1, Pax2, Sal1 and cited 1, wherein expression of the gene above a predetermined threshold is indicative of a renal stem cell.
  • According to an aspect of some embodiments of the present invention there is provided a use of the renal cell population of the present invention, for the preparation of a medicament for the treatment of a renal disorder.
  • According to an aspect of some embodiments of the present invention there is provided a method of monitoring differentiation state of a renal cell, the method comprising determining in the renal cell expression of a pluripotency associated gene selected from the group consisting of Oct4, Nanog and klf4, and/or at least one renal stem cell associated gene selected from the group consisting of Six2, Osr1, Pax2, Sal1 and cited 1, wherein expression of the gene above a predetermined threshold is indicative of a renal stem cell.
  • According to some embodiments of the invention, the method further comprises isolating the cell with the progenitor phenotype.
  • According to some embodiments of the invention, the at least one pluripotency associated gene is selected from the group consisting of Oct4, Nanog and klf4.
  • According to some embodiments of the invention, the at least one renal stem cell associated gene is selected from the group consisting of Six2, Osr1, Pax2, Sal1 and cited 1.
  • According to some embodiments of the invention, the upregulating is effected by contacting the differentiated renal cell with at least one chromatin modifying agent.
  • According to some embodiments of the invention, the upregulating is effected by transfecting the differentiated renal cell with a nucleic acid construct comprising a nucleic acid sequence of the at least one pluripotency associated gene and/or the at least one renal stem cell associated gene.
  • According to some embodiments of the invention, the at least one chromatin modifying agent is selected from the group consisting of valproic acid, 5-aza-2′-deoxycytidine and TSA.
  • According to some embodiments of the invention, the differentiated renal cell is a human differentiated renal cell.
  • According to some embodiments of the invention, the differentiated renal cell is an adult differentiated renal cell.
  • According to some embodiments of the invention, the method further comprises contacting the differentiated renal cell with a Wnt antagonist.
  • According to some embodiments of the invention, the method further comprises isolating the renal stem cell.
  • According to some embodiments of the invention, the renal cell is a human renal cell.
  • According to some embodiments of the invention, the renal cell is an adult renal cell.
  • Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • Some embodiments of the invention are herein described, by way of example only, with reference to the accompanying drawings. With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of embodiments of the invention. In this regard, the description taken with the drawings makes apparent to those skilled in the art how embodiments of the invention may be practiced.
  • FIGS. 1A-T are bar graphs illustrating quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis of human adult (A-J) and fetal (K-T) kidney cells subjected to TSA/AzaC (24 h). Shown are nephric-progenitor—(A-E and K-O), pluripotency/self renewal—(F-H and P-R) and mesenchymal-epithelial transition (MET, I-J and S-T) genes. At least three independent samples of human kidneys were used. Normalization was performed against GAPDH expression (endogenous control) and RQ (relative quantification) calculated relative to the untreated controls. Data were calculated as average±SD. *, p<0.05; **, p<0.05.
  • FIGS. 2A-T are bar graphs illustrating qRT-PCR analysis of human adult (A-J) and fetal (K-T) kidney cells subjected to VPA (24 h, three different concentrations). Shown are nephric-progenitor—(A-E and K-O), pluripotency/self renewal—(F-H and P-R) and mesenchymal-epithelial transition (MET, I-J and S-T) genes. At least three independent samples of human kidneys were used. Data were calculated as average±SD. *, p<0.05; **, p<0.05 after logarithmic transformation versus untreated controls.
  • FIG. 3 is a photograph of a Western blot analysis for SIX2 and WT1 in VPA treated human adult and fetal kidney cells, showing elevated SIX2 but not WT1 protein levels in both cell types following treatment. Melanoma cells were used as negative control and alpha-tubulin and GAPDH were used for loading control.
  • FIGS. 4A-N are graphs illustrating flow cytometric analyses of embryonic renal progenitor surface markers (NCAM, PSA-NCAM) and other markers such as CD133 in human kidney cells following treatment with 24 h −4 mM VPA. At least three independent samples of human kidneys were used. Shown are (FIGS. 4A-L) Representative FACS analysis and (FIGS. 4M-N) fold induction in expression levels demonstrating significant elevation of PSA-NCAM, NCAM and to a lesser extent of CD133.
  • FIGS. 5A-D are graphs illustrating temporal expression patterns of Six2 and Osr1 mRNA in HAK cells undergoing culture passages and subjected to constant VPA exposure (const VPA), 24-hour exposure to 4 mM VPA (VPA) and control samples (CNT). Total RNA was isolated from the HAK cells in P1-P3 and transcript levels of were analyzed by qRT-PCR. Shown are two examples of independent human kidney cells.
  • FIGS. 5E-J are photographs illustrating epithelial-like cell morphology in VPA constant treated cells compared to fibroblastic appearance in transient VPA exposure and control groups.
  • FIG. 6A is a scheme of CpG island region in the Six2 promoter. Marked sections refer to the tested region upstream to the stared transcription starting site (TSS) (the un-stared TSS is an alternative one).
  • FIGS. 6B-F are bar graphs illustrating the results of CHIP assays. FIG. 6B-E: Comparison of histone modifications between VPA treated and untreated human kidney cells. DNA immunoprecipitated with antibodies directed against meH3K4, meH3K27 and AcH4 was analyzed by qRT-PCR utilizing primers from different regions of the Six2 promoter and one region of the Wt1 promoter. To evaluate the level of histone acetylation and methylation, the ratio of PCR products from immunoprecipitated DNA vs. input DNA was calculated. CHIP assays for meH3K4 are shown using two different antibodies. FIG. 6F: Methylation state of the Six2 promoter was analyzed by MeDIP; GAPDH was used as negative control and D4Z4 as positive control. To evaluate the level of DNA methylation, the ratio of PCR products from immunoprecipitated DNA vs. input DNA was calculated.
  • FIGS. 7A-B are bar graphs illustrating qRT-PCR analysis of the Wnt pathway genes, Wnt4, frizzled7 (FZD7) and β-catenin human adult and fetal kidney cells subjected to VPA. Data were calculated as average±SD. *, p<0.05; **, p<0.05 after logarithmic transformation versus untreated controls.
  • FIGS. 7C-M are photographs illustrating β-catenin immunofluorescence in adult and fetal kidney after treatment with valproic acid for 24 hours; Human epithelial cell line with (C) or without (D) primary antibody was used as positive or antibody control, respectively. Human fibroblast cell line (E) was used as negative control. β-catenin expression in adult or fetal kidney before (F, G and H, I) and after (J, K and L, M) treatment, respectively is shown in low (×63) and high magnification (×100), showing strong and extensive expression in VPA treated cells, as well as nuclear localization in the adult kidney (J). Bar represents 20 μM and cell nucleus was stained with Hoechst 33342 (Blue) in all samples tested.
  • FIGS. 7N-Q are bar graphs illustrating quantitative reverse transcription-polymerase chain reaction analysis of Six2 and Osr1 following the addition of Wnt pathway antagonists, DKK1 and sFRP1 to non-treated and VPA treated human and fetal kidney cells. Representative experiment of three yielding similar results.
  • FIG. 7R is a bar graph illustrating qRT-PCR analysis of Six2 expression, in human adult and fetal kidney cells following the addition of Wnt pathway antagonists, DKK1 and sFRP1 to non-treated and VPA treated human and fetal kidney cells. A comparison in Six2 levels between the various treatment groups of adult and fetal kidney cells showed, as expected, higher Six2 in HFK cells, combined VPA/Wnt inhibitor treatment of HAK cells resulted in Six2 re-induction to similar and even higher levels to those of control developing human kidney cells
  • FIGS. 8A-C are graphs and photographs illustrating Over-expression (OE) of Oct4 pluripotency gene in HAK cells. Human Oct4 gene was introduced into HAK cells by lentiviral infection enabling OE of Oct4 simultaneously with a puromycin (Puro) resistance gene. As expected, Oct4 transcript levels were up regulated following infection, subsequent effect was observed following Puro exposure as shown by qRT-PCR (A-B). Growing infected cells on Puro selection resulted in homogenous culture over-expressing Oct4 showing morphologic ‘switch’ into foci of small round cells (C).
  • FIGS. 9A-F are bar graphs illustrating qRT-PCR analysis of Oct4 OE HAK cells with or without exposure to VPA. OE of Oct4 induced Six2 and Osr1 but not Pax2 and Sall1. However, the combination of Oct4 OE with VPA exposure shows higher Six2 expression pattern compared to each of the single manipulations. Concomitant changes in EMT genes, down-regulation of E-cad and up-regulation of Vim, indicated that the observed morphologic ‘switch’ is likely to be associated with EMT (reverse embryogenesis) from which re-programming and de-differentiation may progress.
  • FIGS. 10A-F are photographs illustrating the morphology of HAK following Oct4 OE at passages 5, 7 and 9.
  • FIGS. 11A-E are bar graphs illustrating qRT-PCR analysis of particular genes following Oct4 OE at passages 5, 7 and 9.
  • DESCRIPTION OF EMBODIMENTS OF THE INVENTION
  • The present invention, in some embodiments thereof, relates to isolated populations of renal progenitor cells and methods of generating and using same.
  • Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not necessarily limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways.
  • Renal failure, whether arising from an acute or chronic decline in renal function, is a severe condition that can result in substantial or complete failure of the filtration, reabsorption, endocrine and homeostatic functions of the kidney. It is therefore desirable to obtain cells such as stem cells capable of developing into cells that could supply some or all of the functions of the kidney.
  • The goal of renal regenerative medicine is to create an unlimited supply of human cells resembling the renal progenitors residing in the metanephric/nephrogenic mesenchyme (MM) so as be able to replenish diseased ones in renal hypoplasia/dysplasia, affording enhanced nephron development or replace glomerular podocytes by inoculating a cell population containing the podocyte progenitor cell.
  • In an attempt to generate such cell populations, the present inventors experimented with numerous protocols in order to dedifferentiate human adult kidney (HAK) cells.
  • Whilst reducing the present invention to practice, the present inventors showed that epigenetic ‘transcriptional’ reprogramming of HAK cells via VPA or TSA/5-Aza treatment can increase the expression of pluripotency-associated genes as demonstrated by qRT-PCR analysis (FIGS. 1A-T, 2A-T and 5A-D), Western blot analysis (FIG. 3) and flow cytometry analysis (FIGS. 4A-N).
  • Further the present inventors surprisingly found that over-expression of a pluripotency associated gene via genetic manipulation in HAK cells did not cause the cells to fully dedifferentiate into induced pluripotent cells (iPS) (that would in turn have to be re-differentiated into the renal lineage) but rather directly induce renal progenitors, as illustrated by qRT-PCR analysis and immunohistochemistry (FIGS. 8A-C). A combination of epigenetic reprogramming using chromatin modifying agents and genetic manipulations induced a robust phenotypic switch manifested as epithelial to mesenchymal transition (EMT) simulating reverse embryogenesis in the kidney, as illustrated by qRT-PCR (FIGS. 9A-F).
  • Thus, according to one aspect of the present invention there is provided a method of reprogramming a differentiated renal cell towards a progenitor phenotype, the method comprising up-regulating in the differentiated renal cell an expression of at least one pluripotency associated gene and/or at least one renal stem cell associated gene, thereby reprogramming the differentiated renal cell towards a progenitor phenotype.
  • It will be appreciated that the differentiation status of cells is a continuous spectrum, with a terminally differentiated state at one end of this spectrum and a de-differentiated state (omnipotent state) at the other end.
  • Accordingly, the phrase “differentiated renal cell” as used herein, refers to a cell derived from the kidney that has progressed down the development pathway further than a progenitor cell. According to one embodiment, the differentiated renal cells comprise a specialized function and form (e.g., epithelial cells, endothelial cells, mesangial cells, vascular smooth muscle cells, and pericytes). The cells may be terminally differentiated or partially differentiated. According to an embodiment of this aspect of the present invention, the differentiated renal cells express genes associated with pluripotency (e.g. Oct4, Nanog and klf4) or genes associated with renal stem cells (e.g. any of the markers Hox11 paralogs, Osr1, Pax2, Eya1, Wt1, Sall1, Six2 and Cited1) to a lesser extent than stem cells (e.g. less than half the amount, less than one quarter of the amount and more preferably less than one tenth the amount that is expressed in stem cells).
  • Typically, differentiated cells are not capable of giving rise to a clone of cells.
  • The cells may be primary cells such as those freshly isolated from an animal (e.g. during a biopsy), or may be derived from a cell line (immortalized cells). In an exemplary aspect, the differentiated cells are mammalian cells, such as, for example, human cells or mouse cells. Differentiated cells used in the present invention may be in the form of a group or tissue.
  • The term “dedifferentiating” refers to any movement of the differentiation status of a cell along the spectrum toward a less differentiated state. For example, dedifferentiating includes reversing a multipotent cell back to a pluripotent cell, and reversing a terminally differentiated cell back to either a multipotent cell or a pluripotent cell. In another embodiment, dedifferentiation of a differentiated cell turns the cell back to a multipotent state.
  • The term “progenitor phenotype” refers to a state of a cell whereby it is committed to differentiating towards at least one type of nephron epithelia and also is capable of self-renewing. Typically, renal progenitor cells express some of the phenotypic markers that are characteristic of renal lineages. Typically, they do not produce progeny of other embryonic germ layers when cultured by themselves in vitro, unless dedifferentiated or reprogrammed. It will be appreciated that it is not implied that each of the cells within the population have the capacity of forming more than one type of progeny, although individual cells that are multipotent renal progenitor cells may be present.
  • According to one embodiment of this aspect of the present invention, the renal progenitor cells of the present invention express Six2 and/or Osr1. According to yet another embodiment of this aspect of the present invention, the renal progenitor cells of the present invention express Six2, Osr1, Pax2 and vimentin. According to yet another embodiment of this aspect of the present invention, the renal progenitor cells of the present invention do not express e-cadherin. According to yet another embodiment of this aspect of the present invention, the renal progenitor cells of the present invention are capable of giving rise to a clone of cells.
  • As mentioned, the method of the present invention is effected by up-regulating in the differentiated renal cell an expression of at least one pluripotency associated gene and/or at least one renal stem cell associated gene.
  • As used herein, the phrase “pluripotency associated gene” refers to a gene which is upregulated at least two fold, more preferably at least 5 fold in a pluripotent cell as compared to a non-pluripotent cell.
  • Examples of pluripotent associated genes include those that belong to the Oct family, the Sox family, klf family, Myc family, Lin family and Nanog (mouse NM028016 and human NM024865).
  • Examples of genes belonging to the Oct family include, for example, Oct3/4 (NM013633, mouse and NM002701, human), Oct1A (NM198934, mouse and NM002697, human), Oct6 (NM011141, mouse and NM002699, human), and the like. Oct3/4 is a transcription factor belonging to the POU family, and is reported as a marker of undifferentiated cells (Okamoto et al., Cell 60:461-72, 1990). Oct3/4 is also reported to participate in the maintenance of pluripotency (Nichols et al., Cell 95:379-91, 1998).
  • Examples of genes belonging to the Sox (SRY-box containing) family include, for example Sox1 (NM009233, mouse and NM005986, human), Sox3 (NM009237, mouse and NM005634, human), Sox7 (NM011446, mouse and NM031439, human), Sox15 (NM009235, mouse and NM006942, human), Sox17 (NM011441, mouse and NM022454, human) and Sox18 (NM009236, mouse and NM018419, human), and a preferred example includes Sox2 (NM011443, mouse and NM003106, human).
  • Examples of genes belonging to the klf (Knippel-like factor) family include, for example Klf1(NM010635, mouse and NM006563, human), Klf2 (NM008452, mouse and NM016270, human), Klf4 (NM010637, mouse and NM004235, human) and Klf5 (NM009769, mouse and NM001730, human).
  • Examples of genes belonging to the Myc (myeolcytomatosis oncogene) family include, for example C-Myc (NM010849, mouse and NM002467, human), N-Myc (NM008709, mouse and NM005378, human) and L-Myc (NM008506, mouse and NM005376, human).
  • Examples of genes belonging to the Lin (myeolcytomatosis oncogene) family include, for example Lin28b (NM001031772, mouse and NM001004317, human).
  • The phrase “renal stem cell associated gene” refers to a gene that is upregulated at least 2 fold and more preferably at least 5 fold in a renal stem cell compared with a differentiated renal cell.
  • Examples of renal stem cell associated genes include, but are not limited to six2 (NM016932-accession number:AF136939), osr1 (NM145260.2), pax2 (NM003987.3, NM000278.3, NM003988.3, NM003989.3, NM003990.3) sall1 (NM002968) and cited 1 (NM001144885.1, NM001144886.1, NM001144887.1 NM004143.3).
  • It will be appreciated that the present invention contemplates up-regulation of any combination of the above mentioned genes and any number of the above mentioned genes.
  • According to one embodiment osr1 and six2 are upregulated.
  • According to yet another embodiment six2 and pax2 are up-regulated.
  • According to still another embodiment six2 and sall1 are up-regulated.
  • According to yet another embodiment, pax2 and sall1 are up-regulated.
  • According to still another embodiment osr1, six2 and pax2 are up-regulated.
  • Various methods are contemplated in order to up-regulate expression of the above described genes.
  • According to one embodiment up-regulation is effected by contacting the cells with at least one chromatin modifying agent.
  • According to one embodiment, the chromatin modifying agent of the present invention is a histone deacetylase inhibitor.
  • Examples of chromatin modifying agents include, but are not limited to TSA, sodium butyrate, 5-aza-2′-deoxycytidine, valproic acid, vorinostat, LBH-589, apicidin, TPX-HA analogue, CI-994, MS-275, MGCD0103, and derivatives or analogues of the above-mentioned. Further examples of chromatin modifying agents are provided in U.S. Patent Application No. 20090275032, incorporated herein by reference.
  • According to one embodiment, the differentiated cells of the present invention are dedifferentiated in a culture medium containing the chromatin modifying agent for at least 24 hours. Preferably, the chromatin modifying agent is present in the culture medium during the entire propagation phase.
  • The present inventors have shown that if a Wnt antagonist is used in combination with a chromatin modifying agent, a synergistic effect is seen whereby the upreguation of renal stem cell specific genes in the cells is even more apparent (FIGS. 7A-R). Thus, the present invention conceives of addition of Wnt antagonists (e.g. DKK1 and sFRP1) to the culture medium in combination with the chromatin modifying agents. The present invention also contemplates other Wnt antagonists such as siRNAs, RNAzymes, DNAzymes, miRNAs and the like. Such wnt antagonists are typically transfected into the cells as described herein below.
  • Another method contemplated by the present inventors in order to up-regulate expression of the genes mentioned above is by genetic manipulation. This may be effected in combination with (concomitantly with, prior to or following) chromatin modifying agents or instead of chromatin modifying agents.
  • The genes are typically introduced into the cell in the form of a vector, i.e., a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. The vector can be a viral vector (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced. Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Expression vectors are capable of directing the expression of genes to which they are operably linked.
  • The recombinant expression vectors that can be introduced into renal stem cells include one or more regulatory sequences that are operably linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, “operably linked” is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the cell in which the gene is to be expressed, the level of expression of protein desired, etc.
  • Vector DNA can be introduced into cells via conventional transformation or transfection techniques. As used herein, the terms “transformation” and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (supra), and other laboratory manuals.
  • For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., for resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Preferred selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding the transgene or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
  • It will be appreciated that the cells of the present invention may be transfected with additional genes in order to confer additional properties upon them. This may be effected concomitantly with transfection of the pluripotency associated genes and/or renal stem cell associated genes or following same. For example, genes encoding erythropoeitin or insulin can be introduced into the cells of the present invention. For treatment of anemia associated with renal failure or diabetes it can be useful to introduce into a patient a stem cells modified to express erythropoeitin or insulin. The renal stem cells can be stably or transiently transfected with DNA encoding any therapeutically useful polypeptide.
  • The renal cells of the invention can also be provided with a transgene encoding VEGF or some other factor that can promote growth and or differentiation of cells.
  • Other candidate genes for gene therapy include, for example, genes encoding the alpha 5 chain of type IV collagen (COL4A5), polycystin, alpha-galactosidase A, thiazide-sensitive sodium chloride cotransporter (NCCT), nephrin, actinin, or aquaporin 2.
  • Following the dedifferentiation procedure, renal progenitor cells may be isolated. According to one embodiment, the renal progenitor cells are isolated by flow cytometry using antibodies against proteins that are preferentially expressed in renal progenitor cells and a fluorescence-activated cell sorter (FACS). Co-filed PCT Application Agent References, 48270, filed on 25 Feb. 2010, which claims the benefit under 119(e) of U.S. Provisional Patent Application No. 61/202,425 filed 26 Feb. 2009 to the present inventors, incorporated herein by reference teaches a variety of markers that can be used for this analysis including for example NCAM and ALDH/ALDHbright.
  • As used herein, the term “flow cytometry” refers to an assay in which the proportion of a material (e.g. renal cells comprising a particular maker) in a sample is determined by labeling the material (e.g., by binding a labeled antibody to the material), causing a fluid stream containing the material to pass through a beam of light, separating the light emitted from the sample into constituent wavelengths by a series of filters and mirrors, and detecting the light.
  • A multitude of flow cytometers are commercially available including for e.g. Becton Dickinson FACScan and FACScalibur (BD Biosciences, Mountain View, Calif.). Antibodies that may be used for FACS analysis are taught in Schlossman S, Boumell L, et al, [Leucocyte Typing V. New York: Oxford University Press; 1995] and are widely commercially available.
  • Once isolated, cells of the present invention may be cultured and allowed to proliferate. According to one embodiment the cells are maintained in serum free medium. According to another embodiment the cells are allowed to proliferate in serum containing medium. Optionally the cells may then be directed to differentiate into a desired lineage.
  • The developmental potential of progenitor cells thus obtained can be investigated using methods which are well known in the art. For example by injection into other organs (liver, muscle, heart and bone marrow) to test their multipotency Clarke et al. describes protocols for investigating the development potential of stem cells (Clarke et al. 2000 Science 288:1660).
  • Methods of analyzing the differentiation status of the dedifferentiated cells of the present invention is by analyzing the expression levels of particular genes—e.g. those associated with a differentiated state, those associated with a pluripotent state and/or those associated with a progenitor state.
  • Methods which can be used to analyze expression levels of genes include for example qRT-PCR, Northern blot analysis, Western blot analysis, immunohistochemistry.
  • The renal progenitor cells of the invention can be used to supplement or substitute for kidney cells that have been destroyed or have reduced function. Thus, they can be used to treat patients having poor or no kidney function. The renal stem cells of the invention or cells derived from the renal stem cells of the invention may be capable of performing the filtration and reabsorptive/secretive functions of the kidney.
  • Thus according to an aspect of the present invention there is provided a method of treating a renal damage in a subject in need thereof comprising administering to the damaged kidney of the subject a therapeutically effective amount of any of the isolated population of cells, thereby treating the renal disease in the subject.
  • Cells of the present invention can be used to treat any form of acute or chronic kidney disease, diabetic nephropathy, renal disease associated with hypertension, hypertensive acute tubular injury (ischemic, toxic), interstitial nephritis, congenital anomalies (Aplasia/dysplasia/obstructive uropathy/reflux nephropathy); hereditary conditions (Juvenile nephronophtisis, ARPCKD, Alport, Cystinosis, Primary Hyperoxaluria); Glomerulonephritides (Focal Segmental Glomerulosclerosis); Multisystem Diseases (SLE, HSP, HUS).
  • The cells may be administered per se or as part of a pharmaceutical composition where they are mixed with a suitable carrier or excipient.
  • As used herein a “pharmaceutical composition” refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • Herein the term “active ingredient” refers to the renal progenitor cells (or cells differentiated therefrom) accountable for the biological effect.
  • Hereinafter, the phrases “physiologically acceptable carrier” and “pharmaceutically acceptable carrier” which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound. An adjuvant is included under these phrases.
  • Herein the term “excipient” refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • The renal progenitor cells or cells derived from the renal progenitors cells can be administered into a subject such as surgically or by infusion. For example, renal progenitor cells are injected in vivo into the blood stream or directly into a kidney that is in the postischemic recovery phase. This can be tested easily in an animal model predictive of ischemic kidney damage, the renal pedicle of an anesthetized mouse is clamped for 30 minutes to induce kidney ischemia. Renal progenitor cells are then injected into the juxtamedullary region (approximately 2000 cells at a depth of 2-4 mm). After 2 weeks of recovery, immunohistochemical analysis is used as described above to look for differentiated cells surface markers GP330, Tamm-Horfall, Dolichos Biflorous, and the like. Post-incorporation differentiation status can then be compared to pre-injection marker status.
  • Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • For any preparation used in the methods of the invention, the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fing1, et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p. 1).
  • The number of transplanted cells and intervals between transplantations may be adjusted individually to provide a sufficient biological effect (minimal effective concentration, MEC). The MEC will vary for each preparation, but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
  • Depending on the severity and responsiveness of the condition to be treated, transplantations can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
  • The amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • The progenitor cells of the invention or cells derived from the stem cells of the invention (e.g., epithelial cells endothelial cells, mesangial cells, vascular smooth muscle cells, and pericytes) can be used to construct artificial kidney systems. Such a system can be based on a hollow fiber filtration system.
  • In one example of a filtration device, the progenitor cells of the invention or differentiated progeny thereof are grown on the interior of hollow fibers having relatively high hydraulic conductivity (i.e., ultrafiltration coefficient). The hollow fiber passes through a chamber that is provided with a filtrate outlet port. Arterial blood containing metabolic waste and other unwanted material is introduced into one end of the hollow fiber through an inlet port. Blood passed through the fiber and exits the other end of the fiber through an outlet port where it passed into the patient's vascular venous flow. As blood passes through the fiber, filtrate pass through the cells lining the interior of the fiber and through the hollow fiber itself. This filtrate then passes out of the chamber containing the fiber through the filtrate outlet port. The device preferably includes many such hollow fibers each of which can be in its own chamber. Alternatively many, many hollow fibers (100-100,000 or even more) can be bundled together in a single chamber.
  • The cells of the invention can be used to create a tubule-processing device. In such a device the stem cells of the invention or differentiated cells derived from the stem cells of the invention can be grown in a layer on the exterior of the semipermeable hollow fiber. The fiber is placed in a chamber that is provided with an inlet port and an outlet port. As ultrafiltrate from filtered blood flows through the chamber, reabsorbant passes through the cell layer and through the wall of the fiber into the lumen of the fiber from which it can be directed back into the patient's systemic circulation. Ultrafiltrate that is not reabsorbed passes through the outlet port of the chamber.
  • In the devices described above, it can be desirable to coat the fiber surface that will bear the cell layer with extracellular matrix components. For example, the fiber can be coated with materials such as collagen (e.g., Type I collagen or Type IV collagen), proteoglycan, fibronectin, and laminin or combinations thereof. It can be desirable to combine various cell types on the inner or outer surface of the fibers. For example, it can be desirable to include endothelial cells and pericyte, vascular smooth muscle cells or mesangial cells or fibroblasts or combinations thereof. It can also be useful to provide a feeder layer of cells, e.g., irradiated fibroblasts or other cells that can provide soluble factors and structural support to cells they are indirectly or directly in contact with.
  • The above-described filtration system and the above-described tubule processing system can be combined to create an artificial kidney. Such systems are described in U.S. Pat. No. 6,150,164, hereby incorporated by reference. A number of suitable materials for forming the hollow fiber are described in U.S. Pat. No. 6,150,164, hereby incorporated by reference.
  • The present invention provides a method of using the progenitor cells of the present invention to characterize cellular responses to biologic or pharmacologic agents involving isolating the cells as described, expanding the cells to establish a plurality of progenitors, contacting the progenitor cultures with one or more biologic or pharmacologic agents, identifying one or more cellular responses to the one or more biologic or pharmacologic agents, and comparing the one or more cellular responses of the cultures. Tissue culture techniques known to those of skill in the art allow mass culture of hundreds of thousands of cell samples from different individuals, providing an opportunity to perform rapid screening of compounds suspected to be, for example, teratogenic or mutagenic.
  • As used herein the term “about” refers to ±10%.
  • The terms “comprises”, “comprising”, “includes”, “including”, “having” and their conjugates mean “including but not limited to”. This term encompasses the terms “consisting of” and “consisting essentially of”.
  • The phrase “consisting essentially of” means that the composition or method may include additional ingredients and/or steps, but only if the additional ingredients and/or steps do not materially alter the basic and novel characteristics of the claimed composition or method.
  • As used herein, the singular form “a”, “an” and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a compound” or “at least one compound” may include a plurality of compounds, including mixtures thereof.
  • Throughout this application, various embodiments of this invention may be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
  • Whenever a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases “ranging/ranges between” a first indicate number and a second indicate number and “ranging/ranges from” a first indicate number “to” a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals therebetween.
  • As used herein the term “method” refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
  • As used herein, the term “treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition.
  • It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements.
  • Various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below find experimental support in the following examples.
  • EXAMPLES
  • Reference is now made to the following examples, which together with the above descriptions, illustrate some embodiments of the invention in a non limiting fashion.
  • Generally, the nomenclature used herein and the laboratory procedures utilized in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are thoroughly explained in the literature. See, for example, “Molecular Cloning: A laboratory Manual” Sambrook et al., (1989); “Current Protocols in Molecular Biology” Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al., “Current Protocols in Molecular Biology”, John Wiley and Sons, Baltimore, Md. (1989); Perbal, “A Practical Guide to Molecular Cloning”, John Wiley & Sons, New York (1988); Watson et al., “Recombinant DNA”, Scientific American Books, New York; Birren et al. (eds) “Genome Analysis: A Laboratory Manual Series”, Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057; “Cell Biology: A Laboratory Handbook”, Volumes I-III Cellis, J. E., ed. (1994); “Culture of Animal Cells—A Manual of Basic Technique” by Freshney, Wiley-Liss, N.Y. (1994), Third Edition; “Current Protocols in Immunology” Volumes I-III Coligan J. E., ed. (1994); Stites et al. (eds), “Basic and Clinical Immunology” (8th Edition), Appleton & Lange, Norwalk, Conn. (1994); Mishell and Shiigi (eds), “Selected Methods in Cellular Immunology”, W. H. Freeman and Co., New York (1980); available immunoassays are extensively described in the patent and scientific literature, see, for example, U.S. Pat. Nos. 3,791,932; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; 4,098,876; 4,879,219; 5,011,771 and 5,281,521; “Oligonucleotide Synthesis” Gait, M. J., ed. (1984); “Nucleic Acid Hybridization” Hames, B. D., and Higgins S. J., eds. (1985); “Transcription and Translation” Hames, B. D., and Higgins S. J., eds. (1984); “Animal Cell Culture” Freshney, R. I., ed. (1986); “Immobilized Cells and Enzymes” IRL Press, (1986); “A Practical Guide to Molecular Cloning” Perbal, B., (1984) and “Methods in Enzymology” Vol. 1-317, Academic Press; “PCR Protocols: A Guide To Methods And Applications”, Academic Press, San Diego, Calif. (1990); Marshak et al., “Strategies for Protein Purification and Characterization—A Laboratory Course Manual” CSHL Press (1996); all of which are incorporated by reference as if fully set forth herein. Other general references are provided throughout this document. The procedures therein are believed to be well known in the art and are provided for the convenience of the reader. All the information contained therein is incorporated herein by reference.
  • GENERAL MATERIALS AND METHODS
  • Tissue samples: human tissues samples were collected according to the Helsinki requirements. Human fetal kidney cells (HFK) were collected from elective abortions at fetal gestational age ranged from 15 to 19 weeks. Normal human adult kidney cell (HAK) samples were retrieved from borders of RCC tumors from partial nephrectomy patients.
  • Establishment of primary cultures from human kidney tissues: Collected tissues were minced in HBSS, soaked in IMDM Medium (Invitrogen) supplemented with 0.1% collagenase II (Invitrogen). The digested tissue was then gradually forced through a 100 μm, 70 μm and 50 μm cell strainer to achieve a single cell suspension, and cultured in growth medium supplemented with FBS 10%, L-Glutamin 1%, Pen-Strep 1% and growth factors: 50 ng/ml of bFGF, 50 ng/ml of EGF and 5 ng/ml of SCF (R&D systems).
  • Cell treatment: Cells were treated for 24 hours with growth medium supplemented with 1, 2, or 4 mM VPA (Sigma) or with H2O for control sample. Otherwise, cells were treated for 24 hours with growth medium supplemented with the combination of 75 μM TSA (Sigma) and 250 μM 5-AZA (Sigma) or with 100% Ethanol and Acetic Acid (Acetic Acid: H2O 1:1) for control sample. In some experiments Wnt pathway inhibitors were used in conjunction with VPA as following: cells were treated for 72 hours with growth medium supplemented with 3 μg/ml DKK1(R&D systems) or with 7 μg/ml sFRP1 (R&D systems). 24 hours prior to harvesting VPA at a concentration of 4 mM was added to the cell culture.
  • Flow cytometry: Cells were detached from culture plated with non-enzymatic cell dissociation solution (Sigma-Aldrich). Cells (1×105 in each reaction) were suspended in 50 μl of FACS buffer [0.5% BSA and 0.02% sodium azid in PBS (Sigma-Aldrich and Invitrogen, respectively)] and blocked with FcR Blocking Reagent (MiltenyiBiotec) and human serum (1:1) for 15 minutes. Cells were then incubated for 45 minutes with the following primary antibodies: CD24-PE, NCAM1-PE, (both from eBioscience), CD133-APC, PSA-NCAM (both from MiltenyiBiotec), FZD7-biotin, NTRK2 (both from R&D Systems) or a matching isotype control. Cells were washed with FACS buffer, and incubated for 30 minutes with a secondary antibody if needed: goat anti mouse Streptavidin-APC (BD) or goat anti mouse Alexa Fluor 647 (Invitrogen). Cell's labeling was detected using FACSCalibur (BD). Flow cytometry results were analyzed using FlowJo analysis software. Viable cells were gated by both their FSC/SSC profile and 7AAD (eBioscience) exclusion.
  • Quantitative reverse transcription-PCR: Total RNA from kidney tissue cultured cells was isolated using the TRIZOL reagent (Invitrogen) according to the manufacturer's protocols. cDNA synthesis was carried out using Verso kit (Thermo scientific) with Random primers. Quantitative RT PCR was performed using an ABI7900HT sequence detection system (Perkin-Elmer/Applied Biosystems) in the presence of SYBR green (SYBR green PCR kit; Qiagen). The sequences of the specific primers used for PCR are summarized in Table 1. Each analysis reaction was performed in duplicates or triplicates. GapDH was used as an endogenous control throughout all experimental analyses. Analysis was performed using the −ΔΔCt method, which determines fold changes in gene expression relative to a comparator sample.
  • TABLE 1 
    Gene Sequence
    SIX2 5′-CCA AGG AAA GGG AGA ACA ACG-3′-SEQ ID NO: 1
    5′-GCT GGA TGA TGA GTG GTC TGG-3′-SEQ ID NO: 2
    WT1 5′-GCT GTC CCA CTT ACA GAT GCA-3′-SEQ ID NO: 3
    5′-TCA AAG CGC CAG CTG GAG TTT-3′-SEQ ID NO: 4
    PAX2 5′-CCC AGC GTC TCT TCC ATC A-3′-SEQ ID NO: 5
    5′-GGC GTT GGG TGG AAA GG-3′-SEQ ID NO: 6
    OSR1 5′-TGT ATG GTT TCA GCG CGT TG-3′-SEQ ID NO: 7
    5′-GGG TTG AAT GAC ATG AGG GAA-3′-SEQ ID NO: 8
    SALL1 5′-CAA TCT TAA GGT ACA CAT GGG CAC-3′-SEQ ID NO: 9
    5′-TGC CTC CTA GAA ATG TCA TGG G-3′-SEQ ID NO: 10
    E-CADHERIN 5′-AGT GCC AAC TGG ACC ATT CA-3′-SEQ ID NO: 11
    5′-TCT TTG ACC ACC GCT CTC CT-3′-SEQ ID NO: 12
    VIMENTIN 5′-ACA CCC TGC AAT CTT TCA GAC A-3′-SEQ ID NO: 13
    5′-GAT TCC ACT TTG CGT TCA AGG T-3′-SEQ ID NO: 14
    OCT4 5′-GAG AAC CGA GTG AGA GGC AAC C-3′-SEQ ID NO: 15
    5′-CAT AGT CGC TGC TTG ATC GCT TG-3′-SEQ ID NO: 16
    NANOG 5′-AAT ACC TCA GCC TCC AGC AGA TG-3′-SEQ ID NO: 17
    5′-TGC GTC ACA CCA TTG CTA TTC TTC-3′-SEQ ID NO: 18
    5′-ACC AGG CAC TAC CGT AAA CAC A-3′-SEQ ID NO: 19
    KLF4 5′-GGT CCG ACC TGG AAA ATG CT-3′-SEQ ID NO: 20
    5′-ACC AGG CAC TAC CGT AAA CAC A-3′-SEQ ID NO: 39
    GAPDH 5′-TCCACCACCCTGTTGCTGTA-3′-SEQ ID NO: 40
    5′-TCCACCACCCTGTTGCTGTA-3′-SEQ ID NO: 41
  • Chromatin Immunoprecipitation assay (ChIP): 10×106 cells adult kidney cells were grown. Untreated cells (control) or cells treated with VPA (4 mM) were cross-linked with 1% formaldehayde for 10 min at RT in culture medium. The cells were washed and harvested in cold phosphate-buffered saline (PBS) containing protease inhibitors (Complete mini, Roche Applied Science) and Pepstatin (Sigma). The cells were then washed with Buffer B (20 mM Hepes pH 7.6, 0.25% Triton-x, 10 mM EDTA, 0.5 mM EGTA) and Buffer C (50 mM Hepes pH 7.6, 150 mM NaCl, 1 mM EDTA, 0.5 mM EGTA) were resuspended in 300 μl of SDS lysis buffer [1% SDS, 10 mM EDTA and 50 mM Tris-HCl (pH 8.1)], and incubated on ice for 10 minutes. Lysates were sonicated with 8×10 second bursts and debris were removed by centrifugation for 10 minutes at 1000 g, at 4° C. Supernatants were diluted 10-fold in ChIP dilution buffer [0.01% SDS, 1.1% Triton X-100, 1.2 mM EDTA, 16.7 mM Tris-HCl (pH 8.1) and 167 mM NaCl] and pre-cleared by incubating with 40 μl of protein A+salmon sperm beads (Upstate Biotechnology) for 30 minutes at 4° C. Beads were pelleted for 1 minute at 1000 g at 4° C. A total of 10 μl of the supernatant was saved as input, and the rest was divided into equal aliquots and incubated by rocking with either control Ab (IgG) or with of specific antibodies; Trimethyl-Histon 3K27 antibody (Upstate Biotechnology), anti acetylatedhistone H4 (Upstate Biotechnology) over night at 4° C. A total of 60 μl of protein A+salmon sperm beads was added, and the samples were rocked for 2 hours at 4° C. The complexes on the beads were washed for 5 minutes at 4° C. with the following buffers: low salt [0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl (pH 8.1) and 150 mM NaCl], high salt [0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl (pH 8.1), 500 mM NaCl], LiCl wash [0.25 M LiCl, 1% Nonidet P-40, 1% deoxycholate, 1 mM EDTA and 10 mM Tris-HCl (pH 8.1)], and twice with TE [10 mM Tris-HCl and 1 mM EDTA (pH 8.0)]. Immune complexes were eluted twice with 200 μl of elution buffer [1% SDS and 0.1 M NaHCO3) for 15 minutes at room temperature. The samples were de-crosslinked by adding 16 μl of 5 M NaCl, 8 μl of 0.5 M EDTA (pH 6.5) and 16 μl of 1M Tris-HCl (pH 8.1) and incubated O.N at 65° C. Proteinase K (10 mg/ml) and the samples were incubated for additional 2 hours at 55° C. Immunoprecipitated DNA was recovered by phenol/chloroform extraction and ethanol precipitation and was analyzed by quantitative RT PCR. Primer sets of Six2, WT1, and GapDH promoters are summarized in Table 2, herein below.
  • Methylation Depletion Immunoprecipitation (MeDIP): 10 μg of sonicated genomic DNA (100-400 bp in length) was denatured, incubated O/N at 4° C. with 10 μg/μl of anti-methyl cytosine antibody (Diagenode, Belgium), and subsequently with 60 μl of protein A fast flow beads (Upstate Biotechnologies) for 2 hours at 4° C. The beads were washed and incubated with digestion buffer (50 mM Tris-HCl pH 8.0, 10 mM EDTA, 0.5% SDS) and proteinase K for 3 hours and the DNA extracted by phenol-chloroform and EtOH precipitation. The samples were tested by quantitative RT PCR with the indicated primers at table 2. The sonicated DNA served as input.
  • TABLE 2
    Primer Sequence
    six2 CpG-1157 5′-AAGCAAAAAACAGGACCCCC-3′-SEQ ID NO: 21
    5′-AACGGAGGCAAGATTCCCA-3′-SEQ ID NO: 22
    six2 CpG-955 5′-GAAGCCCCACCACCGTCCTAGA-3′-SEQ ID NO: 23
    5′-ACTTAACCCCACGGGTCCCACA-3′-SEQ ID NO: 24
    six2 CpG-731 5′-GAAGTCGATTCTCCGGCGT-3′-SEQ ID NO: 25
    5′-CCCACCCCCATCCTAGAA AC-3′-SEQ ID NO: 26
    six2 CpG-541 5′-GGGTAGAATGTGCCCGGTGAACAGA-3′-SEQ ID NO: 27
    5′-AGGGAAGGCGGAGACCGTTTAAGG-3′-SEQ ID NO: 28
    six2 CpG-412 5′-GCTGCCCAAACTTTCTTCCCCTG-3′-SEQ ID NO: 29
    5′-CGAAAAGGGGTGGATCGAGGTG-3′-SEQ ID NO: 30
    six2 CpG-92 5′-CGAGGCTCGGGTTACCAGT-3′ SEQ ID NO: 31
    5′-CCCTGATTGGTCCGGTTATCT-3′ SEQ ID NO: 32
    WT1 CpG 5′-GCGGCGCTCGAGAAGTTAT-3′ SEQ ID NO: 33
    5′-GGTCTCAGAACCGAGTGGGAG-3′ SEQ ID NO: 34
    GapDH 5′-TCGACAGTCAGCCGCATCT-3′ SEQ ID NO: 35
    5′-CTAGCCTCCCGGGTTTCTCT-3′ SEQ ID NO: 36
    D4Z4 1168 5′-TCGCTCTGGTCTTCTACGTGG-3′ SEQ ID NO: 37
    5′-AGTCTTGAGTGTGCCAGGCC-3′ SEQ ID NO: 38
  • Western Blot: AK, FK (treated and untreated) and Melanoma cells were harvested with Tripsin/EDTA. Cell extracts were prepared with lysis buffer (50 mM Tris-HCl, 150 mM NaCl, 0.1 mM EDTA, 0.5 mM DTT, 1.5 mM MgCl, 0.5% Triton-X) and their concentrations were detected by BCA protein assay kit (Thermo scientific). 100 μg of total proteins were heated at 95° C. for 5 min with laemmli sample buffer (Bio-rad) and then were loaded on SDS-10% polyacrylamide gel. After electrophoresis, proteins were transferred to nitrocellulose membrane that was blocked with 5% nonfat dry milk overnight at 4° C. The membrane was reacted with the required antibodies, namely Six2 (Affinity Bioreagents), WT1 (Santa Cruz), goat anti-rabbit IgG (Jackson). The membrane was reacted with ECL substrate (Thermo scientific) and was exposed to medical x-ray film (Fuji).
  • Immunofluorescence staining: AK, FK (treated and untreated) human epithelial cell line and fibroblast cell line were fixed with acetone at −20° C. for seven minutes and washed with PBS. Cells were blocked with 7% human serum in PBS for 15 minutes followed by incubation with β-catenin antibody (1:150) (Chemicon international) for 60 minutes. Cells were washed and then incubated with secondary antibody with Cy-3 conjugated anti-mouse IgG (1:200) (Jackson) for 30 minutes. For nuclear staining, cells were washed with PBS containing Hochest (Dako). The presence of β-catenin was examined under a Zeiss confocal fluorescence microscope.
  • Creation of Oct4 OE culture by lentivital infection: 293T cells were co-transfected by calcium phosphate with pSin-EF2-Oct4-Pur vector (Addgene, Yu J et al. Science. 2007, a retroviral vector expressing Oct4, or control retroviral empty vector, both plasmids contain puromycin resistance sequences) and pUMVC (encodes Gag-Pol) and pVSVG (encodes envelope protein of VSV). After 8 hours, the supernatants were replaced with fresh medium. Two days post transfection, supernatants of transfected cells were filtered, and Polybrene (hexadimethrine bromide; Sigma) was added to a final concentration of 8 μg/ml. These viral preparations were used to infect human adult kidney cells for 6 hours, after which fresh medium was added. This infection procedure was repeated on the next day. Three days post infection, the cells were selected for puromycin (puro) resistance at a concentration of 1.5 ug/ml. Cells were analyzed in the presence or absence of VPA treatment.
  • Creation of renal progenitor gene OE culture by viral infection. In order to create and optimize iRPCs, the present inventors propose to over-express developmental transcription factors specifying the embryonic renal progenitor pool (‘renal progenitor genes’) in human kidney cells via a viral system (e.g. lentivirus). The factors will be fluorescently tagged and/or associated with eukaryotic antibiotic resistance so as to enable isolation the desired cells. Three plasmids have been designed: Six2-IRES-GFP, Osr1-mCherry-Neomycin and mCherry-Pax2-Puro. These genes will be to over-expressed individually or in various combinations including in the presence or absence of the pluripotency/self-renewal gene Oct4. Infection will be carried out on primary human cortical renal cell cultures, mesenchymal stem cells and on the more defined RPTECs. Putative iRPCs generated from OE of single or multiple factors, will be then test in vitro in order to find the optimal genetic combination affording induction of progenitors from adult tissue.
  • Example 1 TSA/AzaC Treatment
  • The following experiments were performed in order to determine whether epigenetically repressed genes could be restored by treatment with a combination of the demethylating agent 5-aza-2′-deoxycytidine and the HDAC inhibitor TSA. Thus, human adult kidney cells were initially cultured in the presence or absence of TSA/AzaC for 24 hours. Changes in gene expression in at least three independent samples of human adult kidneys were analyzed after 24-h TSA/AzaC treatment by quantitative RT-PCR. In all cases a robust re-activation pattern was found including Six2 and Osr1 but not Sall1, Wt1 and Pax2 (FIGS. 1A-E). A wide-range of Six2 reinduction levels was noted (five-fold to 25-fold), probably due to primary culture heterogeneity. Several pluripotency/self-renewal genes, among which are Oct4, Nanog and Klf4, have been characterized to be crucial for somatic cell reprogramming. Therefore, the present inventors determined whether TSA/AzaC treatment brings about changes in their expression. Interestingly, expression of the ES cell transcription factors Nanog and Klf4 was significantly upregulated in human adult kidney cells, although Oct4 levels were unchanged (FIGS. 1F-H). Because kidney development progresses through mesenchymal to epithelial conversion of the MM (Vimentin+) to epithelial progenitors (E-cadherin+), the present inventors analyzed whether the reinduction of Six2 and Osr1 and pluripotency genes recapitulates early renal development and is associated with Vimentin upregulation. However, significant downregulation of Vimentin was found with no effect on E-cadherin levels (FIGS. 1I-J). Next, human fetal kidney cells derived from mid-gestation kidneys were evaluated. At this stage of nephrogenesis, there are opposing factors that govern the effects of treatment; on one hand the relative proportion of MM-progenitor cells is rather low compared to more differentiated epithelial cell types. In contrast, renal progenitor genes are still expected to be more abundantly expressed and less methylated at their promoters and thus less influenced. Altogether, a similar pattern of expression was found in treated human fetal kidney cells, upregulation of Six2 and Osr1 and reduction in Wt1, Pax2 and Sall1 levels (FIGS. 1K-O). In addition, induction of the pluripotency genes, Nanog and Klf4 was observed, similar to the adult kidney as well as of Oct4 (FIGS. 1P-R). Analysis of Vimentin/E-cadherin showed significant elevation of E-cadherin and reduction in Vimentin levels (FIGS. 1S-T).
  • Example 2 VPA Treatment
  • The following experiments were performed in order to determine the effects of VPA, an HDAC inhibitor on kidney cells. Changes in renal progenitor gene levels were assessed initially in adult human kidneys following a 24-hour VPA exposure. In contrast to TSA/AzaC, activation of Six2 was much similar among independent samples and a dose-dependent reinduction of Six2 could be observed in the presence of VPA (five-fold increase) (FIG. 2A). Furthermore, in addition to Six2, both Osr1 and Sall1 were found to be upregulated in a dose-dependent manner, while Pax2 levels were mildly elevated and Wt1 reduced (FIGS. 2A-E). Analysis of the pluripotency genes showed Nanog and Klf4 to be significantly up-regulated in all VPA concentrations, and Oct4 particularly in VPA-2 mM, while Vimentin/E-cadherin analysis demonstrated Vimentin levels to be mostly unchanged while E-cadherin mildly elevated (VPA-2 mM, 4 mM) (FIGS. 2F-J).
  • Almost exact findings were noted in human fetal kidney cells exposed to VPA; highest induction in Six2 levels (four-fold) and to a lesser extent of Osr1 and Sall1, with dose-dependent activation, concomitant with reduction of Wt1 and mild elevation in Pax2 levels (FIGS. 2K-O). Furthermore, levels of all pluripotency genes, Nanog, Klf4 and Oct4 (two-five fold increase at 4 mM VPA), and also E-cadherin were found to be significantly elevated along with the renal progenitor genes (FIGS. 2P-T).
  • Example 3 Changes in Protein Levels Following VPA Treatment
  • Having established that similar to TSA/AzaC, VPA treatment results in activation of specific renal progenitor genes as well as pluripotency genes, but also induces Sall1 and that these changes fluctuate less across human kidney samples, the present inventors analyzed changes at the protein level following exposure to 2 and 4 mM VPA. For western analysis the present inventors concentrated on Six2 protein as it was most substantially induced and it has a major role in specifying the renal stem cell population in the MM. Wt1, which is not induced at the gene level, was concomitantly analyzed. Western blots performed on both human fetal and adult kidney cells 24-h after VPA exposure clearly demonstrated an increment in Six2 protein levels in both types of cells (4 mM) and no changes in Wt1, both following their gene expression pattern (FIG. 3). Because certain surface markers characterize renal progenitor cells, FACS analysis of human adult and fetal kidney cells was performed so as to determine whether VPA exposure can also alter their expression levels. NCAM and its embryonic form, PSA-NCAM, both expressed on stem/progenitor cells of the condensed mesenchyme and on MM-derived progenitors showed significant upregulation in expression levels, with dramatic elevation (5-10-fold) in PSA-NCAM expression, especially in the adult kidney (FIGS. 4A-H). Surface markers such as CD133, that has been previously suggested to be expressed in adult renal stem cells and is widely distributed in either fetal and adult kidneys, showed milder changes (CD133) (FIGS. 4I-L).
  • Example 4 Six2 and Osr1 are Maintained Upon Tissue Culture Passage and Constant VPA Treatment
  • Maintaining re-induction of Six2/Osr1 in cultured human kidney cells is potentially important if such cells were to be propagated in vitro for the purpose of cell therapy. The present inventors therefore determined whether Six2 and Osr1 are maintained upon tissue culture passage of adult cells. Two treatment protocols were employed; the first, 24-hour exposure to 4 mM VPA and the second constant exposure to VPA. It was found that while as expected gene levels were re-induced in the P0 cells following the transient VPA protocol with Six2 more profoundly so, in later passages they remained similar to controls. In contrast, constant VPA treatment resulted in significant Six2 and Osr1 re-induction upon culture passage and Six2 levels were even further up-regulated in P3 cells (FIG. 5AD). This was associated with an appearance of epithelial-like cell morphology compared with control cells and those transiently induced which acquired a fibroblastic morphology, in accordance with reports that human kidney cells may undergo transition into myofibroblasts (FIG. 5E-J).
  • Example 5 Epigenetic Modification of the Six2 Promoter
  • To test whether HDAC inhibition following VPA treatment functions in part through chromatin remodeling, ChIP was used to examine the post-transcriptionally modified state of the histones at the Six2 promoter. Recent evidence has indicated that unique patterns of histone modifications are responsible for transcriptional and lineage control. In particular, methylation of lysine 27 on histone subunit H3 (H3K27me) by the Polycomb complex is associated with transcriptional repression, whereas methylation of lysine 4 on H3 (H3K4me) is associated with gene activation. More generally, lysine acetylation of histones is also associated with transcriptional activation. Accordingly, multiple loci at the Six2 promoter region were examined for such changes by CHIP analysis. The promoter sites examined were designed at consecutive distance from the 5′ transcription start site (TSS) (from ˜90 bp upstream to ˜1300 bp) (FIG. 6A). The Wt1 gene, which in contrast with Six2, was found to be modestly affected by VPA exposure was concomitantly analyzed. In addition to the chromatin state, DNA methylation profiles at the Six2 and Wt1 promoter regions were determined so as to detect epigenetic modifications at the DNA level. Treatment with VPA 4 mM significantly increased acetylated histones H4 in multiple Six2 promoter regions (upstream to the TSS: −92, −412, −541, −731, −955 and −1157) accompanied with significant increases in H3K4me but no change in H3K27me levels (FIGS. 6B-E). In contrast with Six2, a modest elevation in repressive H3K27me in the Wt1 promoter region was observed, but not H3K4me, likely to be responsible for little changes in gene expression (FIGS. 6B-E). In addition, MeDIP revealed DNA hypomethylation at the Six2 promoter loci regardless of VPA treatment, while methylation on Wt1 promoter was minimally effected by the treatment (FIG. 6F). Although the observed histone acetylation is directly modulated by HDAC inhibitor treatment, changes in methylation in both histones and DNA likely result from secondary effects. These results indicated that reactivation of Six2 is linked to epigenetic modification of histones at its promoter region leading to gene induction.
  • Example 6 Wnt Pathway Modulation Synergizes with VPA to Induce Six2/Osr1 Re-Induction
  • It has been suggested that in addition its major role as an HDAC inhibitor, VPA can inhibit GSK3β enabling the activation of the canonical Wnt pathway. Indeed, quantitative RT PCR analysis showed an elevation in β-catenin mRNA levels in VPA-treated human kidney cells (FIGS. 7A-B), and immunoflorescence disclosed cyoplasmic and nuclear accumulation of β-catenin compared to non-treated cells (FIGS. 7C-M). In addition, a strong up-regulation of Wnt4 mRNA was found (FIGS. 7A-B), which has been shown to drive differentiation in the MM progenitor population by opposing Six2 [7, 8](ref,). To evaluate the contribution of VPA-induced Wnt activation on renal progenitor gene expression, Wnt antagonists' sFRP1 and DKK1 were added to VPA and their effect was analyzed. When analyzing the sole effects of DKK1 and sFRP1 on Six2 expression in AK and FK cells, elevated Six2 levels in FK cells were observed, in which the Wnt pathway is likely to be more operative (FIGS. 7N-Q). sFRP1 had a synergistic effect on Six2 expression in both human adult and fetal kidney cells, while the synergistic effect of DKK1 was noted mostly in adult kidney cells. Both sFRP1 and DKK1 had a synergistic affect on Osr1 expression in both cells type; with little effect of the Wnt antagonists alone (FIGS. 7N-Q). While a comparison in Six2 levels between the various treatment groups of adult and fetal kidney cells showed, as expected, higher Six2 in FK cells, combined VPA/Wnt inhibitor treatment of AK cells resulted in Six2 re-induction to similar and even higher levels to those of control developing human kidney cells (FIG. 7R).
  • Example 7 Genetic Manipulation of HAK Cells
  • Having observed that epigenetic modulation of HAK cells results in re-activation of six2/osr1 but is not sufficient to induce epithelial to mesenchymal transition (EMT), but on the contrary preserves the epithelial phenotype, the present inventors began to analyze the additive effects of genetic manipulation on HAK cells. Since analyses demonstrated (see for example FIGS. 1G-H; 1Q-R) Nanog to be consistently up-regulated following epigenetic modification while Oct4 was less induced and even remained unchanged in HAK cells treated with VPA, the present inventors reasoned to test the contribution of overexpressing (OE) Oct4 via introduction of the human Oct4 gene into HAK cells in the presence or absence of epigenetic manipulation.
  • Human Oct4 gene was introduced into HAK cells by viral infection (FIGS. 8A-C) enabling over-expression of Oct4 simultaneously with VPA treatment. The cells were analyzed for renal ‘stemness genes’ and EMT. As illustrated in FIGS. 9A-F, over-expression of Oct4 re-induced the MM stem cell genes along with EMT.
  • Example 8 Phenotypic and Functional (Clonogenicity, Self-Renewal, Expansion and Multipotentiality) In Vitro Characterization of Putative iRPCs
  • While reactivation of dormant renal-progenitor-genes and loss of segment-specific markers along with EMT phenotypic switch are important steps in induction of human renal progenitors, functional assays demonstrating stem/progenitor properties in vitro are crucial. Thus, following verification of the initial criteria of all putative human induced renal progenitor cells (iRPCs), they will be tested for clonogenicity, self renewal, expansion and multipotentiality. Single-cell clones raised will be phenotypically characterized by qRT-PCR, FACS and immunostaining (outlined above) for renal embryonic gene and surface marker expression, EMT and segment-specific markers. Additional methodologies are outlined herein below:
  • Clonogenicity/self renewal/expansion. A single cell clonogenicity assay will be performed for human kidney derived cells as described in Pode-Shakked N. et al 2008. Briefly, cells at limiting dilution concentration are plated in matrigel (BD) coated 96-well micro well plates in of culture media and are further expanded. The number of colonized wells is recorded after 3-4 weeks.
  • Multipotentiality The potency of the induced renal progenitor cells (iRPCs) will be analyzed at the single cell level for the mesoderm lineage in general (fat, bone, smooth muscle, endothelium) as described by Dekel at al (JASN, 2006) and more specifically for renal segment-specification. If iRPCs are induced into ‘true’ MM-progenitors they may carry potential to differentiate into more than one nephron epithelial cell type. Wnt4 will be supplied as a well-known inductive signal for the differentiation of MM-progenitors, and single progenitors will be cultured with NIH3T3 cells stably expressing Wnt4. The protocol involves a 7 day induction period and which after the presence of types of epithelial cells that exist in glomeruli and renal tubules will be examined by immunostaining with EMT and segment-specific markers.
  • Example 9 Six2 and Osr1 are Maintained Upon Tissue Culture Passage of OE Oct4 Human Adult Kidney Cells
  • Maintaining re-induction of Six2/Osr1 and the progenitor-state in cultured human kidney cells is highly important if such cells were to be propagated and expanded in vitro for the purpose of cell therapy. The present inventors therefore determined whether the morphologic switch accompanied by Six2 and Osr1 upregulation is maintained upon tissue culture passage of OE Oct4 HAK cells. It was found that OE Oct4 HAK cells can be continually propagated (P5, P7, P9) while showing a smaller round cell compared to spindle-shaped fibroblastic morphology in control AK and up-regulation of OSR1 and SIX2, indicative of maintaining a human renal progenitor state (FIGS. 10A-F and 11A-E).
  • REFERENCES
    • 1. Metsuyanim, S., et al., Accumulation of malignant renal stem cells is associated with epigenetic changes in normal renal progenitor genes. Stem Cells, 2008. 26(7): p. 1808-17.
    • 2. Boyle, S., et al., Fate mapping using Cited1-CreERT2 mice demonstrates that the cap mesenchyme contains self-renewing progenitor cells and gives rise exclusively to nephronic epithelia. Dev Biol, 2008. 313(1): p. 234-45.
    • 3. Kobayashi, A., et al., Six2 defines and regulates a multipotent self-renewing nephron progenitor population throughout mammalian kidney development. Cell Stem Cell, 2008. 3(2): p. 169-81.
    • 4. Self, M., et al., Six2 is required for suppression of nephrogenesis and progenitor renewal in the developing kidney. Embo J, 2006. 25(21): p. 5214-28.
    • 5. Yamanaka, S., Induction of pluripotent stem cells from mouse fibroblasts by four transcription factors. Cell Prolif, 2008. 41 Suppl 1: p. 51-6.
    • 6. Zhou, W. and C. R. Freed, Adenoviral gene delivery can reprogram human fibroblasts to induced pluripotent stem cells. Stem Cells, 2009. 27(11): p. 2667-74.
    • 7. Kispert, A., S. Vainio, and A. P. McMahon, Wnt-4 is a mesenchymal signal for epithelial transformation of metanephric mesenchyme in the developing kidney. Development, 1998. 125(21): p. 4225-34.
    • 8. Nishinakamura, R., Stem cells in the embryonic kidney. Kidney Int, 2008. 73(8): p. 913-7.

Claims (17)

1. A method of reprogramming a differentiated renal cell towards a progenitor phenotype, the method comprising up-regulating in the differentiated renal cell an expression of at least one pluripotency associated gene and/or at least one renal stem cell associated gene, thereby reprogramming the differentiated renal cell towards a progenitor phenotype.
2. The method of claim 1, further comprising isolating the cell with the progenitor phenotype.
3. The method of claim 1, wherein said at least one pluripotency associated gene is selected from the group consisting of Oct4, Nanog and klf4.
4. The method of claim 1, wherein said at least one renal stem cell associated gene is selected from the group consisting of Six2, Osr1, Pax2, Sal1 and cited 1.
5. The method of claim 1, wherein said upregulating is effected by contacting the differentiated renal cell with at least one chromatin modifying agent.
6. The method of claim 1, wherein said upregulating is effected by transfecting the differentiated renal cell with a nucleic acid construct comprising a nucleic acid sequence of said at least one pluripotency associated gene and/or said at least one renal stem cell associated gene.
7. The method of claim 5, wherein said at least one chromatin modifying agent is selected from the group consisting of valproic acid, 5-aza-2′-deoxycytidine and TSA.
8. The method of claim 1, wherein said differentiated renal cell is a human differentiated renal cell.
9. The method of claim 1, wherein said differentiated renal cell is an adult differentiated renal cell.
10. The method of claim 5, further comprising contacting the differentiated renal cell with a Wnt antagonist.
11. A renal cell population generated according to the method of claim 1.
12. A method of repairing or regenerating a renal tissue in a subject in need thereof the method comprising administering to the subject a therapeutically effective amount of the renal cell population of claim 11, thereby repairing or regenerating renal tissue in a subject.
13. (canceled)
14. A method of monitoring differentiation state of a renal cell, the method comprising determining in the renal cell expression of a pluripotency associated gene selected from the group consisting of Oct4, Nanog and klf4, and/or at least one renal stem cell associated gene selected from the group consisting of Six2, Osr1, Pax2, Sal1 and cited 1, wherein expression of said gene above a predetermined threshold is indicative of a renal stem cell.
15. The method of claim 12, further comprising isolating the renal stem cell.
16. The method of claim 12, wherein said renal cell is a human renal cell.
17. The method of claim 12, wherein said renal cell is an adult renal cell.
US13/203,277 2009-02-26 2010-02-25 Methods of reprogramming renal cells Abandoned US20110311494A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/203,277 US20110311494A1 (en) 2009-02-26 2010-02-25 Methods of reprogramming renal cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US20242609P 2009-02-26 2009-02-26
US20242509P 2009-02-26 2009-02-26
US13/203,277 US20110311494A1 (en) 2009-02-26 2010-02-25 Methods of reprogramming renal cells
PCT/IL2010/000159 WO2010097794A1 (en) 2009-02-26 2010-02-25 Methods of reprogramming renal cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2010/000159 A-371-Of-International WO2010097794A1 (en) 2009-02-26 2010-02-25 Methods of reprogramming renal cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/027,256 Division US9770469B2 (en) 2009-02-26 2013-09-16 Methods of reprogramming renal cells

Publications (1)

Publication Number Publication Date
US20110311494A1 true US20110311494A1 (en) 2011-12-22

Family

ID=42124475

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/203,282 Active 2030-04-13 US8828722B2 (en) 2009-02-26 2010-02-25 Isolated populations of renal stem cells and methods of isolating and using same
US13/203,277 Abandoned US20110311494A1 (en) 2009-02-26 2010-02-25 Methods of reprogramming renal cells
US14/027,256 Expired - Fee Related US9770469B2 (en) 2009-02-26 2013-09-16 Methods of reprogramming renal cells
US14/479,385 Expired - Fee Related US9808488B2 (en) 2009-02-26 2014-09-08 Isolated populations of renal stem cells and methods of isolating and using same
US15/728,508 Abandoned US20180042965A1 (en) 2009-02-26 2017-10-10 Isolated populations of renal stem cells and methods of isolating and using same

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/203,282 Active 2030-04-13 US8828722B2 (en) 2009-02-26 2010-02-25 Isolated populations of renal stem cells and methods of isolating and using same

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/027,256 Expired - Fee Related US9770469B2 (en) 2009-02-26 2013-09-16 Methods of reprogramming renal cells
US14/479,385 Expired - Fee Related US9808488B2 (en) 2009-02-26 2014-09-08 Isolated populations of renal stem cells and methods of isolating and using same
US15/728,508 Abandoned US20180042965A1 (en) 2009-02-26 2017-10-10 Isolated populations of renal stem cells and methods of isolating and using same

Country Status (4)

Country Link
US (5) US8828722B2 (en)
EP (3) EP2401363B1 (en)
IL (3) IL214783A (en)
WO (2) WO2010097794A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8828722B2 (en) 2009-02-26 2014-09-09 Tel Hashomer Medical Research Infrastructure And Services Ltd. Isolated populations of renal stem cells and methods of isolating and using same
WO2020223660A1 (en) * 2019-05-02 2020-11-05 Timothy Bertram Treatment of kidney disease in subjects with kidney and/or urinary tract anomalies
US10870834B2 (en) 2010-05-13 2020-12-22 Tel Hashomer Medical Research Infrastructure And Services Ltd. Isolated populations of adult renal cells and methods of isolating and using same
US11225642B2 (en) * 2013-06-11 2022-01-18 Kyoto University Method for producing renal progenitor cells

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103695469B (en) * 2013-11-12 2017-01-11 中山大学 Method for preparing CD44 gene defect mouse induced pluripotent stem cells
WO2015118300A1 (en) * 2014-02-07 2015-08-13 British Telecommunications Public Limited Company Communications interface
JP6587288B2 (en) 2014-06-25 2019-10-09 テル ハショマー メディカル リサーチ インフラストラクチャー アンド サーヴィシーズ リミテッド Its use for the identification as well as the diagnosis and treatment of cancer stem cells
AU2016321015B2 (en) * 2015-09-11 2021-09-30 Astellas Pharma Inc. Method for producing renal progenitor cells
US11447769B2 (en) * 2018-03-27 2022-09-20 The Trustees Of The University Of Pennsylvania Modified immune cells having enhanced function and methods for screening for same
AU2021377272A1 (en) * 2020-11-13 2023-06-22 Timothy A. Bertram Enriched bioactive renal cell populations, characteristics and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040014209A1 (en) * 2002-01-23 2004-01-22 Lassar Andrew B. Compositions and methods for modulating cell differentiation
US20070031966A1 (en) * 2005-07-18 2007-02-08 Regents Of The University Of Michigan Renal progenitor cells from embryonic stem cells

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL154600B (en) 1971-02-10 1977-09-15 Organon Nv METHOD FOR THE DETERMINATION AND DETERMINATION OF SPECIFIC BINDING PROTEINS AND THEIR CORRESPONDING BINDABLE SUBSTANCES.
NL154598B (en) 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
NL154599B (en) 1970-12-28 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING SPECIFIC BINDING PROTEINS AND THEIR CORRESPONDING BINDABLE SUBSTANCES, AND TEST PACKAGING.
US3901654A (en) 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3853987A (en) 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
NL171930C (en) 1972-05-11 1983-06-01 Akzo Nv METHOD FOR DETERMINING AND DETERMINING BITES AND TEST PACKAGING.
US3850578A (en) 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3935074A (en) 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US3984533A (en) 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
US5011771A (en) 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US6150164A (en) 1996-09-30 2000-11-21 The Regents Of The University Of Michigan Methods and compositions of a bioartificial kidney suitable for use in vivo or ex vivo
US6984522B2 (en) * 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
US6458588B1 (en) 2001-01-31 2002-10-01 The General Hospital Corporation Renal stem cells and uses thereof
JP2005050666A (en) 2003-07-28 2005-02-24 Tokai Rubber Ind Ltd Hose material for fuel cell and hose for fuel cell using the same
CA2536909A1 (en) * 2003-08-29 2005-03-10 Regents Of The University Of Minnesota Kidney derived stem cells and methods for their isolation, differentiation and use
US20050260623A1 (en) 2004-02-27 2005-11-24 Michigan State University Oct-4 and GJIC expression as markers for adult human stem cells and cancer cell precursors
US20090275032A1 (en) 2005-08-01 2009-11-05 Nupotential, Inc. Reprogramming a cell by inducing a pluripotent gene through use of an HDAC modulator
WO2007027905A2 (en) 2005-08-31 2007-03-08 Science And Technology Corporation @ Unm Human renal stem cells
TW200925513A (en) 2007-12-11 2009-06-16 Prodisc Technology Inc LED lamp structure for reducing multiple shadows
WO2010097794A1 (en) 2009-02-26 2010-09-02 Tel Hashomer Medical Research Infrastructure And Services Ltd. Methods of reprogramming renal cells
PL2569417T3 (en) 2010-05-13 2018-12-31 Tel Hashomer Medical Research Infrastructure And Services Ltd. Isolated populations of adult renal cells and methods of isolating and using same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040014209A1 (en) * 2002-01-23 2004-01-22 Lassar Andrew B. Compositions and methods for modulating cell differentiation
US20070031966A1 (en) * 2005-07-18 2007-02-08 Regents Of The University Of Michigan Renal progenitor cells from embryonic stem cells

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Jones J et al. 2007. Genomics of renal cell cancer: the biology behind and the therapy ahead. Clin Cancer Res 13:685s-692s. *
Kretzler M et al. 2001. Integrin-linked kinase as a candidate downstreameffector in proteinuria. FASEB J 15: 1843-1845. *
Milutinovic S et al. 2007. Valproate induces widespread epigenetic reprogramming which involves demethylation of specific genes. Carcinogenesis 28: 560-571. *
Senanayake U et al. 2013. The pluripotent renal stem cell regulator SIX2 is activated in renal neoplasms and influences cellular proliferation and migration. Human Pathol 44: 336-345. *
Yoshikawa M et al. 2007. Inhibition of Histone Deacetylase Activity Suppresses Epithelial-to-Mesenchymal Transition Induced by TGF-_1 in Human Renal Epithelial Cells. J Am Soc Nephrol 18: 58-65. *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8828722B2 (en) 2009-02-26 2014-09-09 Tel Hashomer Medical Research Infrastructure And Services Ltd. Isolated populations of renal stem cells and methods of isolating and using same
US9770469B2 (en) 2009-02-26 2017-09-26 Tel Hashomer Medical Research Infrastructure And Services Ltd. Methods of reprogramming renal cells
US9808488B2 (en) 2009-02-26 2017-11-07 Tel Hashomer Medical Research Infrastructure And Services Ltd. Isolated populations of renal stem cells and methods of isolating and using same
US10870834B2 (en) 2010-05-13 2020-12-22 Tel Hashomer Medical Research Infrastructure And Services Ltd. Isolated populations of adult renal cells and methods of isolating and using same
US11225642B2 (en) * 2013-06-11 2022-01-18 Kyoto University Method for producing renal progenitor cells
WO2020223660A1 (en) * 2019-05-02 2020-11-05 Timothy Bertram Treatment of kidney disease in subjects with kidney and/or urinary tract anomalies

Also Published As

Publication number Publication date
EP2465920A3 (en) 2013-02-27
EP2401362B1 (en) 2017-04-12
US9808488B2 (en) 2017-11-07
US8828722B2 (en) 2014-09-09
WO2010097793A2 (en) 2010-09-02
US20150139963A1 (en) 2015-05-21
EP2465920B1 (en) 2017-04-12
US20110311495A1 (en) 2011-12-22
US20140011280A1 (en) 2014-01-09
EP2401362A2 (en) 2012-01-04
IL216043A0 (en) 2011-12-29
WO2010097794A1 (en) 2010-09-02
EP2401363A1 (en) 2012-01-04
EP2401363B1 (en) 2018-08-22
IL214837B (en) 2018-11-29
IL214783A0 (en) 2011-11-30
WO2010097793A3 (en) 2010-11-25
EP2465920A2 (en) 2012-06-20
US20180042965A1 (en) 2018-02-15
IL214783A (en) 2015-05-31
US9770469B2 (en) 2017-09-26
IL214837A0 (en) 2011-11-30

Similar Documents

Publication Publication Date Title
US9770469B2 (en) Methods of reprogramming renal cells
Tang et al. Direct reprogramming rather than iPSC-based reprogramming maintains aging hallmarks in human motor neurons
JP6708617B2 (en) Generation of reprogrammed pluripotent cells
KR101657318B1 (en) Multipotent/pluripotent cells and methods
US8962331B2 (en) Method of making induced pluripotent stem cell from adipose stem cells using minicircle DNA vectors
US10870834B2 (en) Isolated populations of adult renal cells and methods of isolating and using same
US20190390167A1 (en) Method for producing cartilage cells induced to be differentiated from stem cells
US20220213444A1 (en) Compositions and methods for cellular reprogramming
US20230390339A1 (en) Urine-derived epithelial cell cultures, nephrospheroids derived therefrom and methods of producing and using same
Zhang et al. Epigenetic regulation of p63 blocks squamous-to-neuroendocrine transdifferentiation in esophageal development and malignancy
Borkent Roadblocks and Facilitators of Reprogramming to Pluripotency
Cheung Deciphering X-chromosome Inactivation and the Role of MECP2e1 in Rett Syndrome Patient Induced Pluripotent Stem Cells

Legal Events

Date Code Title Description
AS Assignment

Owner name: TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND S

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DEKEL, BENJAMIN;HARARI-SHTEINBERG, ORIT;REEL/FRAME:026884/0452

Effective date: 20110830

AS Assignment

Owner name: TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND S

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE INCORRECT INVENTOR NAME OF ORIT HARARI-SHTEINBERG PREVIOUSLY RECORDED ON REEL 026884 FRAME 0452. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT INDICATING THE CORRECT INVENTOR NAME OF ORIT HARARI-STEINBERG;ASSIGNORS:DEKEL, BENJAMIN;HARARI-STEINBERG, ORIT;REEL/FRAME:030889/0046

Effective date: 20110830

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION