US20130059325A1 - Isolated populations of adult renal cells and methods of isolating and using same - Google Patents

Isolated populations of adult renal cells and methods of isolating and using same Download PDF

Info

Publication number
US20130059325A1
US20130059325A1 US13/697,531 US201113697531A US2013059325A1 US 20130059325 A1 US20130059325 A1 US 20130059325A1 US 201113697531 A US201113697531 A US 201113697531A US 2013059325 A1 US2013059325 A1 US 2013059325A1
Authority
US
United States
Prior art keywords
cells
cell
renal
adult
kidney
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
US13/697,531
Other languages
English (en)
Inventor
Benjamin Dekel
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.)
Ramot at Tel Aviv University Ltd
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 US13/697,531 priority Critical patent/US20130059325A1/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
Publication of US20130059325A1 publication Critical patent/US20130059325A1/en
Assigned to TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD., RAMOT AT TEL-AVIV UNIVERSITY LTD. reassignment TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD.
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: BUZHOR, ELLA, HARARI-STEINBERG, ORIT
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • 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
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • 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
    • C12N2501/30Hormones
    • C12N2501/33Insulin
    • 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
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • C12N2501/392Sexual steroids
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/30Synthetic polymers
    • C12N2533/40Polyhydroxyacids, e.g. polymers of glycolic or lactic acid (PGA, PLA, PLGA); Bioresorbable polymers

Definitions

  • the present invention in some embodiments thereof, relates to isolated populations of adult renal cells and methods of isolating and using same.
  • the kidney is a vital organ in mammals, responsible for fluid homeostasis, waste excretion, and hormone production.
  • ESRD end-stage renal disease
  • Dialysis is the major treatment modality for ESRD, but it has significant limitations in terms of morbidity, mortality, and cost. Allogenic kidney transplantation provides significant benefits in terms of mortality and is ultimately less costly, but is hampered by a severe shortage of available donor organs.
  • Acute renal failure is also quite common, having a mortality rate that ranges from 20 to 70%. For a number of reasons, including aggressive care of an older patient population, the mortality rate due to ARF has not changed over the past 20 years despite advances in technology and therapies.
  • kidney disease has a variety of individual types, they appear to converge into a few pathways of disease progression.
  • the functional unit of the kidney is the nephron.
  • tissues such as bone or glandular epithelia which retain significant capacity for regeneration
  • novel therapies for kidney disease including artificial organs, genetic engineering, and cell therapy.
  • tissue-specific multipotential stem cells including the skin, the hematopoietic system and the intestine.
  • the kidney In contrast to these rapidly-cycling organs, the kidney has a low rate of cell turnover under steady state conditions and it's regenerative capacity is limited. Extra-renal tissue-specific stem cells, including those of the bone marrow do not harbor nephrogenic potential, motivating the search for an adult kidney stem cell.
  • nephron progenitor population residing in the metanephric mesenchyme (MM) and more specifically in the condensed mesenchyme (CM) is entirely exhausted with the completion of nephrogenesis (human-34 th gestational week, mice-2 weeks postnatal) and therefore no progenitor population with similar nephrogenic potential to the MM/CM exists in the adult kidney (6, 7).
  • a population may exist with a more restricted potential than the CM (for instance a progenitor cell type for proximal tubular cells).
  • This cell type is likely to arise from within the epithelial tubular compartment as Humphreys et al (8) demonstrated by lineage tracing that the cells responsible for tubular repopulation after kidney ischemia are of tubular origin, thereby excluding an extra-tubular source.
  • Murine studies have elucidated early markers specifying the epithelial renal progenitor population including a unique combination of transcription factors such as Hox11 paralogs, Osr1, Pax2, Eya1, Wt1, Sall1, Six2, and Cited1 (9). These early renal progenitor markers have been mostly shown to down-regulate with cessation of nephrogenesis in both murine (6) and human kidneys (7).
  • Bussolati et al [American Journal of Pathology. 2005; 166:545-555] teaches isolation and characterization of CD133+ cells derived from normal adult human kidney and suggest that this cell population represent a multipotent adult resident stem cell population that may contribute to the repair of renal injury.
  • an isolated cell population of human adult kidney cells comprising at least 80% adult renal stem cells having a NCAM+ signature.
  • a method of isolating human adult renal cells comprising enriching for a subpopulation of renal cells from an adult renal tissue, the subpopulation of renal cells having a NCAM+ signature, wherein the enriching is effected such that at least 80% of the adult renal cells are of the subpopulation of renal cells.
  • a method of determining clonogenic potential of an adult renal cell population comprising:
  • a cell culture comprising a culture medium and the isolated cell population comprising at least 80% adult renal stem cells having a NCAM+ signature.
  • 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 the isolated cell population comprising at least 80% adult renal stem cells having a NCAM+ signature, thereby treating the renal disease in the subject.
  • a method of identifying an agent capable of regulating differentiation of a renal stem cell comprising contacting the isolated population of cells comprising at least 80% adult renal stem cells having a NCAM+ signature with an agent, wherein a change in developmental phenotype is indicative of the agent capable of regulating differentiation of the renal stem cells.
  • a method of generating a nephrospheroid comprising culturing human adult kidney cells in a culture medium under non-adherent conditions, thereby generating the nephrospheroid.
  • an isolated nephrospheroid comprising human adult kidney cells.
  • a cell culture comprising a culture medium and an isolated population of nephrospheroids, the nephrospheroids comprising human adult kidney cells.
  • a method of identifying an agent capable of regulating differentiation of a renal stem cell comprising contacting an isolated population of nephrospheroids with an agent, the nephrospheroids comprising human adult kidney cells, wherein a change in developmental phenotype is indicative of the agent capable of regulating differentiation of the renal stem cells.
  • 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 an isolated population of nephrospheroids, the nephrospheroids comprising human adult renal cells, thereby treating the renal disease in the subject.
  • the enriching is effected by detecting surface marker expression of NCAM.
  • the cells are seeded on a scaffold.
  • the method further comprises dispersing the human adult kidney cells prior to culturing.
  • the medium further comprises epidermal growth factor (EGF) and fibroblast growth factor (FGF).
  • EGF epidermal growth factor
  • FGF fibroblast growth factor
  • the medium further comprises insulin and progesterone.
  • the medium is devoid of serum.
  • the medium comprises serum.
  • the method further comprises expanding human adult kidney cells in a culture medium under adherent conditions prior to the culturing.
  • the culture medium comprises serum.
  • the isolated nephrospheroid is characterized by enhanced expression of at least one polypeptide selected from the group consisting of sal1, pax2, six2 and WT1 as compared to the adult kidney cells grown under adherent conditions.
  • the isolated nephrospheroid is characterized by enhanced expression of each of sal1, pax2, six2 and WT1 as compared to the adult kidney cells grown under adherent conditions.
  • the isolated nephrospheroid is generated in a serum-free medium.
  • the isolated nephrospheroid is generated in serum-containing medium.
  • FIG. 1 is a diagram of the experimental design for ascertaining culture conditions for isolation of adult renal progenitor cells.
  • Adult kidney tissues were collected from patients nephrectomized due to localized renal tumors. The tissues were digested to a single cell suspension. Cells were grown using either in Serum containing media (SCM) or Serum-free media (SFM). Upon receiving confluent adherent culture (after approximately 7 days), cells were harvested and subjected to limiting dilutions, RNA extraction, sphere formation and continuous adherent culture assays. FKCM-Fetal kidney conditioned media, LD-Limiting Dilution.
  • SCM Serum containing media
  • SFM Serum-free media
  • FIGS. 2A-F are photographs illustrating the growth pattern of hAK cells in two different growth media: SCM and SFM. After one day in culture only a few cells adhered, a few days later these cells started to expand, demonstrating a different pattern of expansion; in SFM expansion was concentric and defined, whereas in SCM, expansion was in less organized manner.
  • FIGS. 3A-J are photographs illustrating adult kidney cell culture characteristics.
  • passage 1 low-passage cultures
  • LTA staining a predominance of proximal tubular cells
  • DBA staining a minority of collecting duct' cells
  • hKEpCs positively stain for markers
  • negative staining is seen in human foreskin fibroblasts (HFF).
  • HFF human foreskin fibroblasts
  • RPTEC renal proximal tubular epithelial cells exclusively stain for LTA. Positively stained cells are green. Nuclei are stained with DAPI (blue).
  • FIGS. 4A-F are photographs illustrating adult kidney cell culture characteristics at passages P3-P5. The cells positively stain for both markers along the represented passages. Nuclei are stained with DAPI (blue).
  • FIGS. 5A-E are bar graphs illustrating the results of the qRT-PCR analysis of renal stem/progenitor genes (SAL1, SIX2, WT1 and PAX2) and pluripotency gene Nanog in SCM and SFM expanded hKEpC cultures of passages P0-P2.
  • SAL1, SIX2, WT1 and PAX2 renal stem/progenitor genes
  • FIGS. 6A-D are graphs and photographs illustrating the formation of hKEpC spheroids.
  • FIGS. 6A and B Representative micrographs of p2 and p6 spheroid morphology (Sph P2 and Sph P6 respectively) obtained from the same hKEpC origin. While P2 spheroids are less organized, P6 spheroids are more condensed, well organixed and demonstrate true sphere morphology.
  • FIGS. 6C and D Quantitative representation of P2 vs. P6 spheroid formation from 2 ⁇ 10 4 cells/2 ml.
  • FIG. 6C represents spheroid number formed, showing significantly higher spheroid formation at P6.
  • FIG. 6A and B Representative micrographs of p2 and p6 spheroid morphology (Sph P2 and Sph P6 respectively) obtained from the same hKEpC origin. While P2 spheroids are less organized, P6
  • 6D represents the number of spheroids formed according to spheroid size, showing that the small size spheroids (less than 15 ⁇ m), rather than the bigger ones (more than 15 ⁇ m) predominantly contribute to the number difference between the P2 vs. P6 spheroids (represented in FIG. 6C ).
  • Graphs represent mean values from the tripicates from 3 different tissue donors.
  • FIG. 7 are photographs illustrating the characterization of hKEpC spheroid origin.
  • hKEpC cells grown as a monolayer were infected by lenitvirus-based vecgtors, carrying the gene for either green fluorescent protein (GFP, green) or m-cherry (red). Fluorescent cells were mixed at an exact ratio of 1:1 and subjected to low-attachment conditions to allow formation of spheroids.
  • Upper panel spheroid formation after 7 days in culture ( ⁇ 20).
  • Lower panel spheroid formation after 6 weeks in culture ( ⁇ 10).
  • FIGS. 8A-J are photographs and graphs illustrating that low attachment conditions induce higher expression of renal progenitor genes.
  • FIGS. 8A-B Spheroid-like structures formed in the low attachment conditions from hAK cells.
  • FIG. 8C Adherent hAK culture;
  • FIG. 8D Relative quantification RT-PCR analysis shows higher nanog and fetal kidney progenitor genes expression in the low-attachment conditions (originated from adherent grown in SCM) in comparison to the adherent culture of the p1 hAK cells in SCM. *, p ⁇ 0.05; **, p ⁇ 0.05 after logarithmic transformation; FIGS. 8E-H .
  • FIG. 8I Elevated transcript levels of Gpc3, in P1 spheroid cells (generated after expansion as a monolayer in SCM) compared to P1 monolayer culture expanded in SCM. P is less than 0.05 after logarithmic transformation.
  • FIG. 8J Elevated progenitor and pluripotency genes transcript levels of P6 spheroid cells (generated after expansion as a monolayer in SCM) compared to P6 monolayer culture expanded in SCM. SCM-serum containing medium. SFM-serum free medium.
  • FIG. 9A is a bar graph illustrating surface marker expression and ALDH activity in spheroids vs adherent culture. Spheroids have enchanced ALDH acivity in comparison to adherent culture of hAK cells. Epithelial (EpCAM, CD24), mesenchymal cell (CD44) markers and CD133 has no difference in both culture conditions.
  • FIG. 9B are time lapse microscopy photographs of the spheroid formation.
  • hKEpC suspension was seeded on the PolyHEMA precoated plates.
  • Micrographs were to taken by the CSN 410 Zeiss microscope ( ⁇ 10) with 3 minute intervals.
  • Upper and lower planes show two representative events of cell collisions and aggregation in the process of spheroid formation.
  • FIG. 9C are representative dot plots showing enhanced ALDH1 activity in spheoids compared to monolayer culture.
  • ALDH1 enzymatic activity was detected using ALDEFLUOR assay.
  • DEAB was used to inhibit the reaction of ALDH with the ALDEFLUOR reagent, providing a negative control.
  • FIGS. 10A-C illustrate the results of microarray analysis of hKEpC spheroids vs. monolayer cells originating from 3 adult kidney (AK) donors.
  • A Unsupervised hierarchical clustering separated samples into two different groups: spheroids and monolayer counterparts;
  • B Hierarchical clustering of differentially expressed genes. Genes that were either up- (477 genes) or down-regulated (348 genes) in spheroids (Sph AK1-3) at least twofold compared with their monolayer culture counterparts (Mono AK1-3);
  • C Forest plot of the cellular processes gene groups representing percent of up- (red) and down (green)-regulated genes.
  • FIGS. 11A-B illustrate hKEpC spheroid characterization and proliferation.
  • FIG. 11A is a photograph illustrating hematoxylin&eosin staining of paraffin embedded spheroids.
  • FIG. 11B is a photograph illustrating immunofluorescence analysis of NCAM and Ki67 of the spheroids. Paraffin embedded spheroids were stained with NCAM (green), Ki67 (red) and Hecht (blue). Low proliferation was observed as evidenced from the low Ki67 staining. Also, low NCAM staining was observed in agreement with FACS analysis.
  • FIGS. 13A-D are photomicrographs illustrating clone morphology.
  • SCM and FKCM clones originated from 1 cell/well, SFM-clones originated from 5 cells/well. FKCM clones were more viable and confluent in comparison to SCM.
  • FIGS. 14A-C are graphs illustrating FACs sorting of NCAM1 expressing hAK cells.
  • FIGS. 15A-E are bar graphs comparing gene expression between NCAM1+ and NCAM1-cell fractions by quantitative RT-PCR analysis.
  • Cultured hAK cells sorted according to NCAM1 overexpress the renal ‘stemness’ genes: (a) renal epithelial progenitor genes, wt1, pax2, six2 (Osr1 was also found to be significantly up-regulated, data not shown); (b) Wnt pathway and renal progenitor surface markers, CTNNB 1, FZD7, NCAM1 and ACTR2b; (c) polycomb group, EZH2.
  • FIGS. 16A-C are bar graphs illustrating the clonogenic potential of hAK NCAM+ cells. Both positive and negative fraction's cells were plated at 1 and 5 cells per well dilution. NCAM+ cells show high clonogenic potential in all concentrations. Graphs represent three experiments originating from three different hAK tissues.
  • FIG. 16D is a graph illustrating the results of a MTS proliferation assay performed on hAK cells sorted according to NCAM1. Both positive and negative cell fractions were analyzed 4, 5 and 7 days following sorting. NCAM+ cells showed lower proliferation capacity. Representative graph of three experiments, data represents mean of triplicates. *, p ⁇ 0.05.
  • FIGS. 16E-J are photomicrographs illustrating the results of a spheroid formation assay performed on NCAM + and NCAM ⁇ fractions sorted from low-passage cultures and expanded in vitro.
  • NCAM + cells show ability to form well-defined spheroids, as opposed to NCAM ⁇ cells, which lack that ability, after 7 days in low-attachment conditions.
  • FIGS. 17A-K are photographs illustrating the results of an in vivo analysis of spheroid and monolayer hKEpC in the chick embryo. 0.43 ⁇ 10 6 cells, derived from dissociated spheroid and monolayer were grafted on the CAM.
  • FIGS. 9A-B CAM grafts (arrowheads) generated from (A) spheroids and (B) monolayer cells, 7 days following grafting.
  • grafts originated from P2 cultures of spheroids (C), monolayer (D) and from P6 cultures of spheroids (E) and monolayer (F) cells, (G) whole spheroids (P2) ( ⁇ 20) demonstrating extensive tubule formation exclusively by hKEpC spheroids.
  • FIGS. 18A-D are photomicrographs illustrating that P2 hKEpC spheroids generate segment-specific tubules.
  • Immunoperoxidase (brown) staining of ( ⁇ 20 arrowheads) for segment-specific markers (18A) LTA, (18B) anti-Tamm horsfall glycoprotein (THG) and (18C) DBA.
  • FIG. 18D Immunofluorescent DBA staining (red) nuclei counterstained with Hoechst (blue). Original magnification ⁇ 20.
  • FIGS. 19A-C are photomicrographs of immunoperoxidase (brown) staining of ( ⁇ 20 arrowheads) for segment specific markers (19A) LTA, (19B) THG and (C) DBA. Original magnification ⁇ 20.
  • FIGS. 20A-F are photographs illustrating the results of an in vivo tubulogenesis assay.
  • A Picture of an explant of hAK cells 7 days after engraftment. Grafting was performed in matrigel B. Fluorescence of the explant (cells were labeled with CFSE prior to grating enabling the detection of a fluorescent signal). Histological analysis of 7-day grafts (H&E) revealed tubular regeneration by low cell numbers (0.43 ⁇ 10 6 cells/egg) only when grafting dissociated hAK spheroids (C) or hAK NCAM+ cells (D). Tubular structures are highlighted in boxes. Similar cell number of adherent cultured hAK cells did not generate tubular structures (E). Control HEK 293 (F) or mesenchymal stem cells (not shown) did not generate tubular structures and remained as undifferentiated masses.
  • the present invention in some embodiments thereof, relates to isolated populations of adult renal cells and methods of isolating and using same.
  • Renal failure 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 progenitor or stem cells capable of developing into renal cells that could substitute for some or all of the functions of the kidney.
  • hAK Human adult kidney
  • Sphere structures are multicellular globes that develop from cells that survive anchorage-independent conditions in vitro, such as growth in ultra-low attachment plates. Unlike monolayer-based cultures, these structures carry the advantage of mirroring the 3D cellular context. Furthermore, sphere-forming assays have been shown to be a useful means for maintenance and expansion of putative stem/progenitor cell populations.
  • the present inventor therefore sought to investigate for the first time primary human kidney cells grown in suspension culture over non-adherent plastic surfaces as opposed to monolayer expanded cells.
  • the present inventor isolated primary human renal cells from kidney surgical samples, established heterogeneous cultures of human kidney epithelial cells (hKEpC) and demonstrated to their ability to efficiently generate 3D aggregates or spheroids.
  • NCAM cell surface progenitor marker
  • NCAM + enriched adult renal cells overexpressed early renal epithelial progenitor markers (Six2, Osr1, Sall1, Pax2 and Wt1) and early surface antigens (FZD7, AVR2b) (11), polycomb group (Bmi-1, Ezh2), Wnt pathway (Beta-catenin, FZD7) as well the pluripotency marker, Oct4 ( FIGS. 9A-E ), indicating the presence of stem/progenitor cells.
  • the NCAM + subpopulation was highly clonogenic ( FIGS. 16A-C ) and further comprised sphere generating capabilities ( FIGS. 16E-J ) further indicating the presence of stem/progenitor cells.
  • an isolated cell population of human adult kidney cells comprising at least 50%, 60%, 70% 80% or 90% adult renal stem having a NCAM+ signature.
  • the term “isolated” means that a cell population is removed from its natural environment.
  • the term “purified,” means that a cell population is essentially free from any other cell type (e.g., feeder fibroblasts).
  • renal stem cell refers to a cell which is not terminally differentiated as a renal cell but which has the ability to differentiate into specialized cell having one or more structural and/or functional aspects of a physiologic kidney. According to specific embodiments the renal stem cells are not embryonic stem cells.
  • At least 50%, 60%, 70% 80% or 90% of the renal stem cells have a NCAM+CD133+ signature.
  • At least 50%, 60%, 70% 80% or 90% of the renal stem cells have a NCAM+CD133 ⁇ signature.
  • At least 50%, 60%, 70% 80% or 90% of the renal stem cells have a NCAM+CD24+ signature.
  • At least 50%, 60%, 70% 80% or 90% of the renal stem cells have a NCAM+CD24 ⁇ signature.
  • At least 50%, 60%, 70% 80% or 90% of the renal stem cells have a NCAM+ nestin+ signature.
  • At least 50%, 60%, 70% 80% or 90% of the renal stem cells have a NCAM+ nestin ⁇ signature.
  • NCAM+ populations of the present invention further comprise a gene expression profile as provided in FIGS. 15A-E . Assaying expression of any of the genes of the provided expression profile may be used to qualify cells of the NCAM+, signature as further described herein below.
  • the present invention further provides for a method of isolating the aforementioned cells. This is effected by enriching for a subpopulation of renal cells from a human adult renal tissue, the subpopulation of renal cells having an NCAM+ signature.
  • the kidney may comprise a whole kidney or fragments thereof (e.g., renal capsule).
  • the cells of the adult kidney are of a heterogeneous population.
  • the cells of the adult kidney may be dispersed prior to selection.
  • Exemplary agents that may be used to disperse the kidney cells include collagenase, dispase and trypsin.
  • the cells of the adult kidney are expanded prior to sorting.
  • the cells are cultured for less than three passages, more preferably for less than two passages.
  • NCAM1 (3 variants): NM — 181351, NM — 000615, NM — 001076682; FZD7: NM — 003507; CD24: NM — 013230; CD133 (PROM1): NM — 006017; NTRK2: AF410902; PSA-NCAM, Polysialylated NCAM1 same ID as NCAM1; ACVRIIB: NM — 001106; ROR2 (2 variants): M97639 NM, — 004560; oct4 (POU5F1): NM — 203289 NM — 002701; six2: NM — 016932 (accession number:AF136939); sal11: NM — 002968; ctnnb1 NM — 001098210 (NM — 001098209 XM — 001133660 XM — 001133664 XM —
  • Antibodies for the above mentioned cell markers are commercially available. Examples include but are not limited to, NCAM1 (eBioscience), EPCAM (MiltenyiBiotec), FZD7 (R&D Systems), CD24 (eBioscience), CD133 (MiltenyiBiotec), NTRK2 (R&D Systems), PSA-NCAM (MiltenyiBiotec) ACVRIIB (R&D Systems), ROR2 (R&D Systems), nestin (Abcam).
  • the term “enriching” refers to a procedure which allows the specific subpopulation of renal cells to comprise at least about 50%, preferably at least about 70%, more preferably at least about 80%, about 95%, about 97%, about 99% or more renal stem cells having the desired signature (e.g. NCAM+).
  • the enriching may be effected using known cell sorting procedures such as by using a fluorescence-activated cell sorter (FACS).
  • FACS fluorescence-activated cell sorter
  • 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 material e.g. renal cells comprising a particular maker
  • Another method of cell sorting is magnetic cell sorting as further described in the Examples section below.
  • the enriching may also be effected by depleting of non-relevant subpopulations such as renal stromal cells or interstitium (interstitial) cells.
  • cells of the present invention may be cultured and their “stemness” properties may be further analyzed as described below.
  • clonogenicity is a function of stem cells
  • the cells may be analyzed for their clonogenic potential.
  • the present inventors have shown that isolated adult renal cells having an NCAM+ signature are highly clonogenic.
  • the present inventor has discovered that culturing cells at low dilution in serum-comprising medium, preferably in the presence of conditioned medium from human fetal kidney cells is an optimal way to ascertain clonogenic potential. By counting the number of clones formed after a predetermined time (e.g. one month), one can determine the clonogenic potential of a renal cell population.
  • a predetermined time e.g. one month
  • An exemplary method for obtaining conditioned medium from human fetal kidney cells is by combining (e.g. in q 1:1 ratio) SCM and SCM from FK cultures of passages 1 to 3.
  • the ability to form spheres is also a function of stem cells. Accordingly, the cells may be analyzed for their sphere-forming potential.
  • the present inventors have shown that isolated adult renal cells having an NCAM+ signature have a high sphere forming potential.
  • Another way to confirm the presence of renal stem cells is by testing for expression of stem cell-specific genes.
  • An upregulation of such genes infers the presence of renal stem cells.
  • Such genes include, but are not limited to Six2 (NM — 016932-accession number: AF136939), osr1 (NM — 145260.2), Pax2 (NM — 003987.3 NM — 000278.3, NM — 003988.3, NM — 003989.3, NM — 003990.3), Sall1 (NM — 002968) and Cited 1 (NM — 001144885.1, NM — 001144886.1, NM — 001144887.1 NM — 004143.3).
  • Methods for analyzing for the expression of stem cell-specific genes include RT-PCR, Northern blot, Western blot, flow cytometry and the like.
  • the present inventor has found optimal conditions for culturing adult kidney cells such that they form spheroids.
  • the present inventor found that these spheroids expressed stem cell-specific genes to a greater extent than adult kidney cells that were cultured under adherent conditions.
  • a method of generating a nephrospheroid comprising culturing adult kidney cells under non-adherent conditions, thereby generating the nephrospheroid.
  • nephrospheroid refers to a 3 dimensional (spherical or partially) aggregate of kidney cells. It may also be referred to as a tubular organoid. The nephrospheroid comprises at least two cell types and is not derived from a single cell-type (i.e. is not of a clonal origin).
  • the nephrospheroid is capable of generating proximal distal tubules and collecting ducts when allowed to differentiate in vivo following grafting to the chorioallantoic membrane (CAM) of the chick embryo.
  • CAM chorioallantoic membrane
  • the nephrospheroid is not capable of generating proximal distal tubules and collecting ducts when allowed to differentiate in vivo following grafting to the chorioallantoic membrane (CAM) of the chick embryo.
  • CAM chorioallantoic membrane
  • non-adherent conditions refers to conditions in which the cells do not attach to the surface of a container in which they are cultured such that a substantial portion of the cells can be removed from the surface of the container by mechanical manipulations that do not cause significant damage to the cells. It is understood that the cells can still be retained in or on a non-adherent matrix (e.g., on Hydrogel spheres) and be removed from the surface of the container. Such manipulations include, for example, to gentle agitation, massage, or manual manipulation of the container, or rinsing the container with growth media As used herein, a substantial portion of the cells to be removed is at least 70%, preferably at least 75%, 80% or 85%, more preferably at least 90% or 95%.
  • Manipulations that cause damage to the cells can be identified by determining the viability of the cells before and after manipulation, for example by trypan blue staining. Mechanical manipulations should cause damage to less than 20%, preferably less than 15%, or 10%, more preferably less than 5%, 2%, or 1% of the cells. Numerous methods are known for culturing cells under non-adherent conditions. These include growth of cells encapsulated in matrices such as Hydrogel and Matrigel, on in between layers of agarose, or in TeflonTM bags. An exemplary hydrogel which may be used is PolyHEMA. It will be appreciated that the cells can grow in contact with the non-adherent matrices, but do not adhere to plastic culture containers.
  • matrices such as Hydrogel and Matrigel
  • An exemplary hydrogel which may be used is PolyHEMA. It will be appreciated that the cells can grow in contact with the non-adherent matrices, but do not adhere to plastic culture containers.
  • Contemplated culture mediums include, but are not limited to IMDM (Invitrogen) or DMEM (Invitrogen).
  • the culture medium is devoid of serum.
  • the medium may comprise additional components which further encourage the cells to form spheroids.
  • the medium may further comprise growth factors such as epidermal growth factor (EGF) and fibroblast growth factor (FGF).
  • growth factors such as epidermal growth factor (EGF) and fibroblast growth factor (FGF).
  • EGF epidermal growth factor
  • FGF fibroblast growth factor
  • Other contemplated components include insulin and progesterone.
  • the cells are dispersed as described herein above.
  • the adult kidney cells are cultured prior to forming the spheroids in order to expand the number of cells.
  • the adult kidney cells are expanded in serum containing medium for about 4, 5, 6, 7 or more passages under adherent conditions prior to generation of the spheroids.
  • adherent conditions refers to conditions in which the cells attach to the surface of a container in which they are cultured such that a substantial portion of the cells cannot be removed from the surface of the container by mechanical manipulations that do not cause significant damage to the cells.
  • the present inventor generated nephrospheroids and proceeded to characterize these structures.
  • an isolated nephrospheroid may be characterized by enhanced expression of at least one polypeptide selected from the group consisting of sal1, pax2, six2 and WT1 or combinations thereof, as compared to identical adult kidney cells grown under adherent conditions.
  • an isolated nephrospheroid may be characterized by enhanced expression of each of sal1, pax2, six2 and WT1 as compared to identical adult kidney cells grown under adherent conditions.
  • enhanced expression refers to an increase in expression by at least 1.5 fold, more preferably at least 2 fold and even more preferably at least 3 fold.
  • the cell populations of the present invention are typically allowed to proliferate under conditions that preserve their stem/progenitor cell phenotype.
  • Cell populations of the present invention can be genetically modified to express a transgene. This may be used to increase survival of the cells, render them immortalized or differentiated to a desired lineage. Examples of such transgenes and methods of introducing the same are provided below.
  • 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.
  • genes encoding erythropoeitin or insulin can be introduced into a kidney stem cell.
  • a stem cells modified to express erythropoeitin or insulin can be introduced into a patient.
  • the renal stem cells can be stably or transiently transfected with DNA encoding any therapeutically useful polypeptide.
  • the cell populations 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.
  • genes can be driven by an inducible promoter so that levels of the transgen can be regulated.
  • These inducible promoter systems may include a mutated ligand binding domain of the human estrogen receptor (ER) attached to the protein to be produced. This would require that the individual ingest tamoxifen to allow expression of the protein.
  • Alternatives are tetracyclin on or off systems, RU486, and a rapamycin inducible system.
  • An additional method to obtain relatively selective expression is to use tissue specific promoters. For instance, one could introduce a transgene driven by the KSP-cadherin, nephrin or uromodulin-specific promoter.
  • Cells isolated or generated by the method described herein can be genetically modified by introducing DNA or RNA into the cell by a variety of methods known to those of skill in the art. These methods are generally grouped into four major categories: (1) viral transfer, including the use of DNA or RNA viral vectors, such as retroviruses (including lentiviruses), Simian virus 40 (SV40), adenovirus, Sindbis virus, and bovine papillomavirus for example; (2) chemical transfer, including calcium phosphate transfection and DEAE dextran transfection methods; (3) membrane fusion transfer, using DNA-loaded membrane vesicles such as liposomes, red blood cell ghosts, and protoplasts, for example; and (4) physical transfer techniques, such as microinjection, electroporation, or direct “naked” DNA transfer.
  • viral transfer including the use of DNA or RNA viral vectors, such as retroviruses (including lentiviruses), Simian virus 40 (SV40), adenovirus, Sindbis virus, and bovine
  • Cells can be genetically altered by insertion of pre-selected isolated DNA, by substitution of a segment of the cellular genome with pre-selected isolated DNA, or by deletion of or inactivation of at least a portion of the cellular genome of the cell. Deletion or inactivation of at least a portion of the cellular genome can be accomplished by a variety of means, including but not limited to genetic recombination, by antisense technology (which can include the use of peptide nucleic acids, or PNAs), or by ribozyme technology, for example. Insertion of one or more pre-selected DNA sequences can be accomplished by homologous recombination or by viral integration into the host cell genome.
  • the desired gene sequence can also be incorporated into the cell, particularly into its nucleus, using a plasmid expression vector and a nuclear localization sequence. Methods for directing polynucleotides to the nucleus have been described in the art.
  • the genetic material can be introduced using promoters that will allow for the gene of interest to be positively or negatively induced using certain chemicals/drugs, to be eliminated following administration of a given drug/chemical, or can be tagged to allow induction by chemicals (including but not limited to the tamoxifen responsive mutated estrogen receptor) for expression in specific cell compartments (including but not limited to the cell membrane).
  • Calcium phosphate transfection which relies on precipitates of plasmid DNA/calcium ions, can be used to introduce plasmid DNA containing a target gene or polynucleotide into isolated or cultured cells. Briefly, plasmid DNA is mixed into a solution of calcium chloride, then added to a solution which has been phosphate-buffered. Once a precipitate has formed, the solution is added directly to cultured cells. Treatment with DMSO or glycerol can be used to improve transfection efficiency, and levels of stable transfectants can be improved using bis-hydroxyethylamino ethanesulfonate (BES). Calcium phosphate transfection systems are commercially available (e.g., ProFection from Promega Corp., Madison, Wis.).
  • DEAE-dextran transfection which is also known to those of skill in the art, may be preferred over calcium phosphate transfection where transient transfection is desired, as it is often more efficient.
  • microinjection can be particularly effective for transferring genetic material into the cells.
  • the developmental potential of the cell populations 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).
  • the cell populations may also be grated into chick embryos so as to ascertain their developmental potential as described in the Examples section herein below.
  • the cell populations 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 cell populations of the invention or cells derived there from may be capable of performing the filtration and reabsorptive/secretive functions of the kidney.
  • 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 the isolated population of cells described herein, 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 present inventor contemplates administration of single cell suspensions of dissociated spheroid-cells, partly dissociated spheroid-cells or non-dissociated spheroid cells.
  • the cells may be administered per se or as part of a pharmaceutical composition where they are mixed with a suitable carrier or excipient.
  • 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.
  • active ingredient refers to the renal progenitor cells (or cells differentiated therefrom) accountable for the biological effect.
  • 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.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • the cell populations or cells derived can be administered into a subject such as surgically or by infusion.
  • a subject such as surgically or by infusion.
  • renal cells are injected in vivo 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 stem 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.
  • the cells of the invention can be used to construct artificial kidney systems.
  • Such a system can be based on a hollow fiber filtration system.
  • the stem 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.
  • 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.
  • 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 (i.e. a scaffold).
  • the fiber is placed in a chamber that is provided with an inlet port and an outlet port.
  • 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.
  • 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 present invention provides a method of using the cell populations of the present invention to characterize cellular responses to biologic or pharmacologic agents involving isolating the cells as described s, culture expanding the cells to establish a plurality of MRPC cultures, contacting the MRPC 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.
  • 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.
  • a compound or “at least one compound” may include a plurality of compounds, including mixtures thereof.
  • 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.
  • 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.
  • 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.
  • 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.
  • SCM serum containing medium
  • SFM serum free medium
  • SCM comprised IMDM 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).
  • SFM comprised 500 ml DMEM:F12 (ratio 1:1, Invitrogen), 1% Pen-strep, 2 ml B27 supplement (Gibco), 4 ⁇ g/ml heparin, 1% Non essential Amino acids (invitrogen), 1% of sodium pyruvate (invitrogen), 1% L-glutamine, 1 ml Lipid mix (Sigma), 5 ml N2 supplement 100X (Gibco), 5 ml growth factor mix (for 200 ml of growth factor mix: 100 ml DMEM:F12, 4 ml 30% glucose, 200 mg transferin, 50 mg insulin in 20 ml of water, 19.33 mg putrescine in 20 ml ddw, 200 ⁇ l sodium selanite (0.3 mM stock), 20 ⁇ l progesterone (2 mM stock)), FGF 10 ng/ml, EGF 20 ng/ml.
  • FKCM Fetal kidney conditioned medium
  • Monolayer cells were detached from culture plates with 0.25% trypsin (Gibco), spheroids were collected and dissociated by 5-10 min digestion with Accutase (Sigma-Aldrich). Viable cell number was determined using Trypan blue staining (Invitrogen). Cells (1 ⁇ 10 5 in each reaction) were suspended in 50 ⁇ l 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, Auburn, USA) and human serum (1:1) for 15 min at 4° C. Cells were then incubated for 45 min with a respective antibody or a matching isotype control.
  • FACS buffer 0.5% BSA and 0.02% sodium azid in PBS (Sigma-Aldrich and Invitrogen, respectively)
  • CD56 (NCAM) microbeads (Miltenyi Biotec) were used for single marker cell separation. Positive and negative fractions were separated using Mini or MidiMACS cell columns (Miltenyi Biotech) according to the manufacturer's protocols. Briefly, cell suspension was obtained and, for the removal of clumps, was passed through a 30 ⁇ m nylon mesh. Cells were labeled by adding 20 ⁇ l CD56 microbeads per 10 7 total cells for 15 minutes in refrigerator. Then the cells were washed, resuspended and magnetically separated. For increased purity, the fractions were passed a second time through fresh columns Separated cells were plated for limiting dilution, differentiation assays and FACS analysis. A part of cells was used for RNA extraction.
  • Cells were harvested as described above, filtered through a 30 ⁇ m nylon mesh before final centrifugation, then re-suspended in flow cytometry buffer consisting of 2 mM bovine serum albumin (BSA; Sigma-Aldrich) and 10% sodium azide in PBS. Cells were labeled with anti NCAM:PE (eBioscience) or other needed antibody. Fluorescence-activated cell sorter FACSAria and the FACSDiva software (BD Biosciences) were used in order to enrich for cells expressing these markers.
  • BSA bovine serum albumin
  • PE eBioscience
  • Quantitative real time reverse transcription PCR (qPCR) reactions were carried out to determine fold changes in expression of the selected renal ‘stemness’ genes (57) as well of differentiation markers in the sorted hAK cells.
  • nephron segment-specific genes were analyzed: Aminopeptidase-A (ENPEP), Aquaporin-1(AQP1), Aquaporin-3 (AQP3), Na/CL co-transporter (NCCT), Podocin; renal stem/progenitor genes: PAX2, SALL1, SIX2, WT1 and pluripotency gene: NANOG.
  • Limiting dilution assay was performed on separated cell fractions NCAM1 positive vs. NCAM1 negative. Briefly, sorted cells were plated in 96-well micro well plates (Greiner Bio-One) in 150 ⁇ l of culture media, at 0.3 or 1 cells per well dilution. The low cell concentration was achieved by serial dilutions reaching 1000 cells per ml. The number of colonized wells was recorded after one month.
  • Sections, 4- ⁇ m thick, from whole blocks of normal hAK were cut for immunohistochemistry. The sections were processed within 1 week to avoid oxidation of antigens. Before immunostaining, sections were treated with 10 mM citrate buffer, PH 6.0 for 10 min at 97° C. in a microwave oven for antigen retrieval, followed by treatment of 3% H 2 O 2 for 10 minutes. The slides were subsequently stained by the labeled—(strept) avidin-biotin (LAB-SA) method using a histostain plus kit (Zymed). Anti-human CD56 antibody (LifeSpan Biosciences, Inc.) and anti-FZD7 antibody, at a dilution of 1:50, were used. Controls were prepared by omitting the primary antibodies or by substituting the primary antibodies with goat IgG isotype. The immunoreaction was visualized by an HRP-based chromogen/substrate system, (Zymed).
  • Spheroids were collected, fixed in PFA 4%, embedded in agarose gel and then in paraffin. Immunocytochemistry for Ki67 (mammalian-specific monoclonal rabbit antibody, Lab Vision clone SP6) was performed on the sections containing spheroids using microwave antigen retrieval. Detection was performed with Alexa-594 anti-rabbit antibodies (Molecular Probes), and slides were counterstained with Hoechst.
  • Ki67 mammalian-specific monoclonal rabbit antibody, Lab Vision clone SP6
  • Detection was performed with Alexa-594 anti-rabbit antibodies (Molecular Probes), and slides were counterstained with Hoechst.
  • Tubular segments were identified by use of the following markers: Proximal tubule (PT) with Fluorescein labeled Lotus tetragonolobus lectin (LTL), collecting duct (CD) with Fluorescein labeled Dolichos biflorus agglutinin (DBA) 1:200 for 30 minutes (Vector Laboratories); distal tubules and thick ascending limb of Henle with anti-Tamm-Horsfall Glycoprotein antibody (anti-THG) (Millipore, Chemicon), secondary antibody used for this staining NorthernLights anti-sheep IgG-NL637 (R&D systems).
  • PT Proximal tubule
  • LDL Lotus tetragonolobus lectin
  • CD collecting duct
  • DBA Dolichos biflorus agglutinin 1:200 for 30 minutes (Vector Laboratories)
  • Fertile chicken eggs were obtained from a commercial supplier, and incubated at 37° C. at 60-70% humidity in a forced-draft incubator. At 3 days of incubation, an artificial air sac was established dropping the CAM. A window was opened in the shell, and the CAM exposed on 9 or 10th day of incubation.
  • AK cells derived from AK adherent vs spheroid cultures or NCAM+/ ⁇ sup-populations. AK cells were suspended in 50 ⁇ l medium and Matrigel (1:1 by volume) and pipetted into a plastic ring placed on the membrane. The egg was then sealed with adhesive tape and returned to the incubator.
  • Double-immunocytochemistry for Ki67 (mammalian-specific monoclonal rabbit antibody, Lab Vision clone SP6) and NCAM (mouse monoclonal, Santa Cruz) was performed on the sections containing AK cells using microwave antigen retrieval. Detection was performed with Alexa-488 anti-mouse and Alexa-594 anti-rabbit antibodies (Molecular Probes), and slides were counterstained with Hoechst, and all serial sections were examined. Photomicrographs were made with digital cameras (CFW-1312M and CFW-1612C, Scion Corporation) on Olympus SZX12 and BX51 microscopes. All changes in the images (contrast, brightness, gamma, sharpening) were made evenly across the entire field, and no features were removed or added digitally.
  • HEK293 cells were initially transformed.
  • HEK293 cells were maintained in Dulbecco's modified Eagle's medium supplemented with 10% fetal calf serum, L-glutamine, penicillin and streptomycin (Biological Industries, Israel), at 37° C. and 5% CO2.
  • Cells were transfected using calcium phosphate with three lentiviral vectors, pHR-CMV-GFP/m-Cherry (7.5 ⁇ g), ⁇ R8.2 (5 ⁇ g) and pMD2.G (2.5 ⁇ g). After 6 h, the supernatants were replaced with 5 ml of fresh medium.
  • RNA from each sample was used to prepare biotinylated target DNA, according to manufacturer's recommendations.
  • the target cDNA generated from each sample was processed as per manufacturer's recommendation using an Affymetrix GeneChip Instrument System (ur12).
  • the quality and amount of starting RNA was confirmed using an agarose gel or by Bioanalyser (Agilent). After scanning, array images were assessed by eye to confirm scanner alignment and the absence of significant bubbles or scratches on the chip surface. The signals derived from the array will be assessed using various quality assessment metrics. Details of quality control measures can be found at (url1).
  • Gene level RMA sketch algorithm (Affymetrix Expression Console and Partek Genomics Suite 6.2) was used for crude data generation. Significantly changed genes were filtered as changed by at least 2 fold (P value 0.05). Genes were filtered and analysed using unsupervised hierarchical cluster analysis, and supervised hierarchical cluster analysis (Partek Genomics Suite and Spotfire DecisionSite for Functional Genomics; Somerville, Mass.) to get a first assessment of the data. Further processing included functional analysis and over-representation calculations based on Gene Ontology and publication data: DAVID (www.apps1.niaid.nih gov/David/upload.asp), Ingenuity, Database for Annotation (GO), Visualization, and Integrated Discovery. Over-representation calculations are done using Ease (DAVID).
  • hKEpC were seeded on the poly-HEMA pre-coated plate. Photomicrographs were taken every 3 minutes by CLSN 410 Zeiss microscope ( ⁇ 10) in DIC mode. Images were stacked to the movie file by ImageJ 1.42q software.
  • Results are expressed as the mean values ⁇ STDEV.
  • tissue was dissociated into a single cell suspension and cultured in low densities in T75 flasks so as to allow clonal growth (see scheme in FIG. 1 ).
  • SCM serum containing media
  • SFM serum free media
  • proximal tubule aminopeptidase-A (ENPEP) and Aquaporin-(AQP1)-collecting duct—Aquaporin-3 (AQP3), distal convoluted tubule—NA/Cl co-transporter (NCCT), podocyte—Podocin.
  • ENPEP proximal tubule
  • Aquaporin-(AQP1)-collecting duct Aquaporin-3
  • NCCT distal convoluted tubule
  • podocyte podocin.
  • FIGS. 5A-E Analysis of renal progenitor gene expression in the heterogeneous P0 adherent SCM and SFM cultures showed similar gene levels ( FIGS. 5A-E ). Cells were then harvested and propagated and expanded as adherent cultures in SFM/SCM or subjected to sphere formation and limiting dilutions to assess for clonogenicity in various culture conditions (see FIG. 1 ).
  • culture conditions that support proliferation of human kidney cells that form spheroids may represent a strategy for isolation of cells with progenitor potential. Accordingly, heterogeneous P0 adherent SCM and SFM cultures originating from five hAK samples were subjected to low attachment conditions—specifically they were seeded on polyHEMA plates at a density of 20-40,000 viable cells/ml.
  • nephrospheroids nephrospheroids
  • hKEpC spheroids 100-130 micrometer in diameter
  • kidney-derived cells from two donors were grown as a monolayer and stably labeled with either red or green fluorescent proteins using lentivirus-based vectors, directing constitutive expression of mCherry and GFP, respectively.
  • lentivirus-based vectors directing constitutive expression of mCherry and GFP, respectively.
  • cells were propagated P2-P3. Fluorescent hKEpC were detached, mixed at a ratio of 1:1 and subjected to low-attachment conditions at low densities 10 4 cells/well to generate spheroids. Continuous microscopic examination from 7-10 days to six weeks after seeding revealed that spheroids contained both red and green cells.
  • FIG. 7 More than 75% of cells in each kidney-spheroid were comprised of one color ( FIG. 7 ), suggesting that aggregation into hKEpC spheroids had occurred and that cells were not entirely clonally derived.
  • time-lapse microscopy was utilized to follow initial events after hKEpC seeding (2 ⁇ 10 4 cells/well) in non-adherent conditions, as cells were filmed every 3 minutes for 48 hours. Cell collision and aggregation were noticed to occur within five hours after seeding, indicating this as the initiating process for spheroid formation.
  • the present inventors initially determined whether the generation of hKEpC spheroids promoted the expression of ‘sternness’ genes.
  • FIG. 9A Flow cytometry was used ( FIG. 9A ) to analyze the percentage of cells expressing the epithelial, renal and mesenchymal stem cell antigens EpCAM, CD24, CD133, CD44 (15) in spheroid and monolayer hKEpC. High expression levels of CD24 and CD44 (80-100% of cells) in both spheroids and monolayer cells was found ( FIG. 9A and FIG. 9B ), while EpCAM and CD133 levels found to be further elevated in spheroids indicating mostly an EpCAM + CD24 + CD133 + CD44 + phenotype of spheroid cells.
  • aldehyde dehydrogenase1 an enzyme which increased activity has been detected in stem/progenitor cell populations, showed significantly higher levels in hKEpC spheroid cells compared to monolayer counterparts ( FIG. 9C ). It was found that 29.93 ⁇ 11.78% of spheroid cells displayed high levels of ALDH1 activity, compared to 8.06 ⁇ 4.53% of monolayer cells.
  • hKEpC spheroids have a distinct antigenic profile with enhanced ALDH1 activity.
  • Global transcriptional changes associated with kidney spheroid formation Having illuminated specific characteristics of hKEpC spheroids the present inventors wanted to asses on a global level the transcriptional alternations taking place in relation to with spheroid formation. For this spheroid and monolayer hKEpC were generated from three different human adult kidney sources and their global gene expression profile using oligonucleotide microarrays were compared.
  • Unsupervised clustering (Partek 6.5) of the entire human microarray data set clearly distinguished among samples separating them into two major groups: hKEpC spheroids and hKEpC grown as monolayer and indicating a different biological entity and fundamental difference in gene expression patterns ( FIG. 10A ). Kidney spheroids were closer to each other rather than to their monolayer counterpart of the same adult kidney origin. 825 genes differentially expressed by spheroid and monolayer hKEpC (>2 fold change, ANOVA, P ⁇ 0.05) were identified. These included 477 genes upregulated and 348 downregulated in spheroids compared to monolayer cells ( FIG. 10B ). The 20 genes most highly expressed in hKEpC spheroids and monolayer cells are respectively shown in Table 2.
  • the Gene Ontology (GO) enrichment analysis tool and DAVID were used. Up and down regulated genes in hKEpC spheroids were categorized into cellular processes, according to Partek ( FIG. 10C ) and DAVID (Table 3), showing the most significantly elevated genes to group into cell-cell adhesion/ECM/cell recognition, ion transport, regulation of cellular component biogenesis, while down-regulated genes were related to cell growth/mitosis/cell cycle and cell locomotion.
  • Table 4 further elaborates 23 genes categorized in biological adhesion, which were up-regulated in spheroid cells (DAVID, p ⁇ 0.00001).
  • PKHD1 polycystic kidney Localized predominantly at 3.38 and hepatic the disease 1 apical domain of polarized epithelial cells, suggesting it may be involved in the tubulogenesis and/or maintenance of duct-lumen architecture.
  • PCDHB2 protocadherin The extracellular domains 2.94 beta 2 interact in a homophilic manner to specify differential cell-cell connections.
  • CDH16 cadherin 16 cell adhesion molecule 2.99 KSP-cadherin AGT angiotensinogen Essential component of the 5.67 (serpin peptidase renin-angiotensin system inhibitor, (RAS) clade A, member 8)
  • ITGB6 integrin beta 6 Integrin alpha-V/beta-6 is a 3.41 receptor for fibronectin and cytotactin.
  • VNN1 vanin 1 May play a role in 4.79 oxidative-stress response
  • RHOB ras homolog gene Mediates apoptosis in 2.58 family member B neoplastically transformed cells after DNA damage. Affects cell adhesion and growth factor signaling in transformed cells.
  • GPI glycosylphosphatidylinositol
  • Extracellular matrix protein 15.33 CHL1 cell adhesion cell adhesion molecule 4.58 molecule with homology to L1CAM CEACAM1 carcinoembryonic cell adhesion molecule 2.07 antigen-related cell adhesion molecule
  • hKEpC spheroids generated a quiescent niche enriched in cell-cell and cell matrix interactions.
  • the quiescent nature of spheroids was confirmed by analysis of proliferating cells in whole spheroids fixed and embedded in paraffin and stained for hematoxylin and eosin ( FIG. 11A ) and for the cell proliferation marker, Ki-67 ( FIG. 11B ).
  • All hKEpC spheroids exhibited a low proliferation index of ⁇ 10% of Ki-67-positive cells per spheroid/section, indicative of the quiescent nature of the spheroids.
  • NCAM1 Expressing Cells Isolated from Heterogeneous hAK Cultures are Highly Clonogenic and Preferentially Form Spheres
  • the present inventors determined surface markers that could identify cells within the heterogeneous hAK cultures preferentially exhibiting these characteristics. Accordingly, cell subpopulations positive for surface markers which have been shown to mark the renal progenitor population of the developing human kidney such as NCAM1 and FZD7 (10, 11) were sorted. NCAM1 which during nephrogenesis is localized to cells of the MM and its early derivatives, including condensed mesenchyme and early nephron, is not expressed in the adult kidney in vivo (11, 17, 18).
  • NCAM+ cells Efficient fractionation of NCAM+ cells was achieved with FACS sorting ( FIGS. 14A-C ) and to a lesser extent via microbeads.
  • Analysis of renal ‘stemness’ genes in NCAM+ cells compared to NCAM ⁇ fraction obtained from heterogeneous cultures of five different hAK revealed overexpression of the early renal epithelial progenitor markers (Six2, Osr1, Sall1, Pax2 and Wt1) and early surface antigens (FZD7, AVR2b) (11), polycomb group (Bmi-1, Ezh2), Wnt pathway (Beta-catenin, FZD7) as well the pluripotency marker, Oct4 ( FIGS. 15A-E ).
  • NCAM+ and NCAM ⁇ spheres showed similar elevated gene levels when compared to adherent NCAM+ cells (data not shown).
  • NCAM strongly enriched for sphere-forming capability in low-passage heterogeneous cultures.
  • sphere-formation irrespective of NCAM expression enriches for the renal progenitor genes.
  • hKEpC spheroids have enhanced renal “sternness” profile and recapitulate a microenvironment rich in ECM and cell contact molecules the present inventors tested whether this leads to improved functional potency to generate renal structures. Accordingly, human cell grafting was performed onto the chorioallantoic membrane (CAM) of the chick embryo and their fate 7 days post-implantation was analyzed ( FIG. 17A-k ). Chick embryos were grafted with either whole hKEpC spheroids, or single cell suspensions of dissociated spheroid-cells (immediately after disassociation) and monolayer hKEpC. The suspended cells are especially important as they represent an injectable form of cells.
  • CAM chorioallantoic membrane
  • FIG. 17G Implantation of whole human kidney-spheroids onto the CAM resulted in tubule formation.
  • Comparison of single cell implantation of spheroid and monolayer hKEpC demonstrated that grafts generated from spheroid cells were much bigger then their counterparts ( FIGS. 17A-B ).
  • H&E staining revealed robust tubule formation capacity by spheroid-cells, e.g. 0.43 ⁇ 10 6 spheroid cells induced formation of multiple tubular structures, while few tubuli were observed in grafts generated by similar numbers of monolayer hKEpC ( FIGS. 17C-D ).
  • tubular structures that were formed by dissociated spheroid hKEpC, graft sections were stained for segment-specific tubular markers (LTA, proximal; THG, distal; DBA, distal/collecting). It was found that reconstituted renal structures showed LTA, THG and DBA positive tubules and were reminiscent of a wide adult human tubular spectrum ( FIGS. 18A-D ). To clarify specificity of DBA expression immunofluorescent staining was performed and DBA(+) tubules were found to comprise a portion of the reconstituted tubules ( FIG. 18D ).
  • Spheroid-cells obtained from high-passage cultures also showed more than one type of differentiated tubules with positive staining of the THG and DBA markers and to a much lesser extent LTA staining ( FIGS. 19A-C ).
  • hKEpC spheroids enhance functional potency for tubule formation.
  • NCAM+ sorted, adherent cells The regenerative ability of NCAM+ sorted, adherent cells was also analyzed. In this experiment, strong tubular reconstitution by 0.43 ⁇ 10 6 NCAM + cells was observed with the NCAM ⁇ fraction failing to form similar structures ( FIGS. 20A-F ). Thus, low numbers of both spheroid- and sorted NCAM+ cells can recapitulate kidney structures in vivo indicative of high renal potential.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Urology & Nephrology (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US13/697,531 2010-05-13 2011-05-12 Isolated populations of adult renal cells and methods of isolating and using same Abandoned US20130059325A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/697,531 US20130059325A1 (en) 2010-05-13 2011-05-12 Isolated populations of adult renal cells and methods of isolating and using same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US33420610P 2010-05-13 2010-05-13
US201161457613P 2011-05-02 2011-05-02
US13/697,531 US20130059325A1 (en) 2010-05-13 2011-05-12 Isolated populations of adult renal cells and methods of isolating and using same
PCT/IL2011/000376 WO2011141914A1 (fr) 2010-05-13 2011-05-12 Populations isolées de cellules rénales adultes et procédés d'isolement et d'utilisation desdites populations

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2011/000376 A-371-Of-International WO2011141914A1 (fr) 2010-05-13 2011-05-12 Populations isolées de cellules rénales adultes et procédés d'isolement et d'utilisation desdites populations

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/045,784 Division US10870834B2 (en) 2010-05-13 2018-07-26 Isolated populations of adult renal cells and methods of isolating and using same

Publications (1)

Publication Number Publication Date
US20130059325A1 true US20130059325A1 (en) 2013-03-07

Family

ID=44515147

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/697,531 Abandoned US20130059325A1 (en) 2010-05-13 2011-05-12 Isolated populations of adult renal cells and methods of isolating and using same
US16/045,784 Active 2031-06-25 US10870834B2 (en) 2010-05-13 2018-07-26 Isolated populations of adult renal cells and methods of isolating and using same

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/045,784 Active 2031-06-25 US10870834B2 (en) 2010-05-13 2018-07-26 Isolated populations of adult renal cells and methods of isolating and using same

Country Status (6)

Country Link
US (2) US20130059325A1 (fr)
EP (1) EP2569417B1 (fr)
CA (1) CA2799307C (fr)
ES (1) ES2672918T3 (fr)
PL (1) PL2569417T3 (fr)
WO (1) WO2011141914A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015134610A1 (fr) * 2014-03-06 2015-09-11 Yale University Procédés pour le traitement de la néphropathie diabétique
US9770469B2 (en) 2009-02-26 2017-09-26 Tel Hashomer Medical Research Infrastructure And Services Ltd. Methods of reprogramming renal cells
US20200377860A1 (en) * 2015-09-03 2020-12-03 The Brigham And Women's Hospital, Inc. Three-dimensional differentiation of epiblast spheroids to kidney organoids models stage-specific epithelial physiology, morphogenesis, and disease
US10900022B2 (en) 2013-06-14 2021-01-26 The University Of Queensland Renal progenitor cells
US11268950B2 (en) 2016-09-28 2022-03-08 Organovo, Inc. Use of engineered renal tissues in assays

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6995112B2 (ja) 2017-04-06 2022-02-04 日機装株式会社 細胞の培養方法及び細胞支持複合体の製造方法

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL154600B (nl) 1971-02-10 1977-09-15 Organon Nv Werkwijze voor het aantonen en bepalen van specifiek bindende eiwitten en hun corresponderende bindbare stoffen.
NL154598B (nl) 1970-11-10 1977-09-15 Organon Nv Werkwijze voor het aantonen en bepalen van laagmoleculire verbindingen en van eiwitten die deze verbindingen specifiek kunnen binden, alsmede testverpakking.
NL154599B (nl) 1970-12-28 1977-09-15 Organon Nv Werkwijze voor het aantonen en bepalen van specifiek bindende eiwitten en hun corresponderende bindbare stoffen, alsmede testverpakking.
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 (nl) 1972-05-11 1983-06-01 Akzo Nv Werkwijze voor het aantonen en bepalen van haptenen, alsmede testverpakkingen.
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
US6235551B1 (en) 1997-12-31 2001-05-22 Micron Technology, Inc. Semiconductor device including edge bond pads and methods
US6458588B1 (en) 2001-01-31 2002-10-01 The General Hospital Corporation Renal stem cells and uses thereof
US20040014209A1 (en) 2002-01-23 2004-01-22 Lassar Andrew B. Compositions and methods for modulating cell differentiation
JP2005050666A (ja) 2003-07-28 2005-02-24 Tokai Rubber Ind Ltd 燃料電池用ホース材料およびそれを用いてなる燃料電池用ホース
US20070031966A1 (en) 2005-07-18 2007-02-08 Regents Of The University Of Michigan Renal progenitor cells from embryonic stem cells
WO2007027905A2 (fr) 2005-08-31 2007-03-08 Science And Technology Corporation @ Unm Cellules souches renales humaines
TW200925513A (en) 2007-12-11 2009-06-16 Prodisc Technology Inc LED lamp structure for reducing multiple shadows
EP2401362B1 (fr) 2009-02-26 2017-04-12 Tel HaShomer Medical Research Infrastructure and Services Ltd. Populations isolées de cellules souches rénales et leurs procédés d'isolement et d'utilisation

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Eccles et al., Comparative in Situ Hybridization Analysis of PAX2, PAX8, and WT1 Gene Transcription in Human Fetal Kidney and Wilms' Tumours, American Journal of Pathology, Vol. 146, No. 1, January 1995. *
Garvin et al., The In Vitro Growth, Heterotransplantation, and Immunohistochemical Characterization of the Blastemal Component of Wilms' Tumor, American Journal of Pathology, Vol. 129, No. 2, November 1987. *
Pode-Shakked et al. 2009 cover page. downloaded from http://onlinelibrary.wiley.com/doi/10.1111/j.1582-4934.2008.00607.x/full. p.1 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US10900022B2 (en) 2013-06-14 2021-01-26 The University Of Queensland Renal progenitor cells
US12060580B2 (en) 2013-06-14 2024-08-13 The University Of Queensland Renal progenitor cells
WO2015134610A1 (fr) * 2014-03-06 2015-09-11 Yale University Procédés pour le traitement de la néphropathie diabétique
US20200377860A1 (en) * 2015-09-03 2020-12-03 The Brigham And Women's Hospital, Inc. Three-dimensional differentiation of epiblast spheroids to kidney organoids models stage-specific epithelial physiology, morphogenesis, and disease
US11268950B2 (en) 2016-09-28 2022-03-08 Organovo, Inc. Use of engineered renal tissues in assays
US11982668B2 (en) 2016-09-28 2024-05-14 Organovo, Inc. Use of engineered renal tissues in assays

Also Published As

Publication number Publication date
ES2672918T3 (es) 2018-06-18
PL2569417T3 (pl) 2018-12-31
CA2799307C (fr) 2019-07-02
WO2011141914A1 (fr) 2011-11-17
US10870834B2 (en) 2020-12-22
CA2799307A1 (fr) 2011-11-17
EP2569417B1 (fr) 2018-04-18
EP2569417A1 (fr) 2013-03-20
US20180346882A1 (en) 2018-12-06

Similar Documents

Publication Publication Date Title
US10870834B2 (en) Isolated populations of adult renal cells and methods of isolating and using same
Buzhor et al. Kidney spheroids recapitulate tubular organoids leading to enhanced tubulogenic potency of human kidney-derived cells
Bahmad et al. Sphere-formation assay: three-dimensional in vitro culturing of prostate cancer stem/progenitor sphere-forming cells
Buzhor et al. Reactivation of NCAM1 defines a subpopulation of human adult kidney epithelial cells with clonogenic and stem/progenitor properties
Zheng et al. Most C6 cells are cancer stem cells: evidence from clonal and population analyses
US9808488B2 (en) Isolated populations of renal stem cells and methods of isolating and using same
Wei et al. Human skeletal muscle-derived stem cells retain stem cell properties after expansion in myosphere culture
US20200291361A1 (en) Composition and method
Zhu et al. Optimization of the conditions of isolation and culture of dairy goat male germline stem cells (mGSC)
Chou et al. Fetal hepatic progenitors support long-term expansion of hematopoietic stem cells
KR20190112090A (ko) 다능성줄기세포의 분화 제어 방법
KR20220004126A (ko) 유도 다능성 줄기 세포로부터 생체내에서 기능적이고 환자-특이적인 흉선 조직의 생성
Duss et al. Mesenchymal precursor cells maintain the differentiation and proliferation potentials of breast epithelial cells
CA2837462A1 (fr) Systemes de tubule proximal bioartificiel et leurs procedes d'utilisation
JP2023550036A (ja) 富化された生体活性腎細胞集団、その特徴及び使用
Omer et al. OCT4 induces long-lived dedifferentiated kidney progenitors poised to redifferentiate in 3D kidney spheroids
Dai et al. Isolation, Characterization, and Safety Evaluation of Human Skin‐Derived Precursors from an Adherent Monolayer Culture System
Dunville et al. Laminin 511 and WNT signalling sustain prolonged expansion of hiPSC-derived hippocampal progenitors
US20230390339A1 (en) Urine-derived epithelial cell cultures, nephrospheroids derived therefrom and methods of producing and using same
Meregalli STUDY OF MULTIPOTENT RENAL PKHHIGH STEM-LIKE CELLS, ISOLATED FROM HUMAN NEPHROSPHERES: REGENERATIVE ABILITIES AND TRANSCRIPTOMIC PROFILE
Silver Investigations into the biology of the extracellular matrix in neurogenesis, brain injury, and cancer
Wagner et al. Thomas Höfner, Christian Eisen, Corinna Klein, Teresa Rigo-Watermeier, 2 Stephan M. Goeppinger, 4 Anna Jauch, 5 Brigitte Schoell, 5 Vanessa Vogel, Elisa Noll, Wilko Weichert, 4 Irene Baccelli, Anja Schillert, 2
Thorgeirsson et al. Wnt and the Cancer Niche: Paracrine Interactions with Gastrointestinal Cancer Cells Undergoing Asymmetric Cell Division
Helgudóttir Ummyndunaráhrif HER2 yfirtjáningar í undirhópum þekjufrumna úr brjóstkirtli

Legal Events

Date Code Title Description
AS Assignment

Owner name: TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND S

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DEKEL, BENJAMIN;REEL/FRAME:029324/0777

Effective date: 20110630

AS Assignment

Owner name: RAMOT AT TEL-AVIV UNIVERSITY LTD., ISRAEL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD.;REEL/FRAME:040705/0399

Effective date: 20161123

Owner name: TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND S

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND SERVICES LTD.;REEL/FRAME:040705/0399

Effective date: 20161123

AS Assignment

Owner name: TEL HASHOMER MEDICAL RESEARCH INFRASTRUCTURE AND S

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HARARI-STEINBERG, ORIT;BUZHOR, ELLA;REEL/FRAME:042369/0725

Effective date: 20141022

STCB Information on status: application discontinuation

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