Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Stem cells in urology

Abstract

The shortage of donors for organ transplantation has stimulated research on stem cells as a potential resource for cell-based therapy in all human tissues. Stem cells have been used for regenerative medicine applications in many organ systems, including the genitourinary system. The potential applications for stem cell therapy have, however, been restricted by the ethical issues associated with embryonic stem cell research. Instead, scientists have explored other cell sources, including progenitor and stem cells derived from adult tissues and stem cells derived from the amniotic fluid and placenta. In addition, novel techniques for generating stem cells in the laboratory are being developed. These techniques include somatic cell nuclear transfer, in which the nucleus of an adult somatic cell is placed into an oocyte, and reprogramming of adult cells to induce stem-cell-like behavior. Such techniques are now being used in tissue engineering applications, and some of the most successful experiments have been in the field of urology. Techniques to regenerate bladder tissue have reached the clinic, and exciting progress is being made in other areas, such as regeneration of the kidney and urethra. Cell therapy as a treatment for incontinence and infertility might soon become a reality. Physicians should be optimistic that regenerative medicine and tissue engineering will one day provide mainstream treatment options for urologic disorders.

Key Points

  • Regenerative medicine and tissue engineering strategies are becoming a viable option for replacing diseased or damaged organs, particularly in the urinary tract

  • Adult stem cells would be an ideal source of autologous cells for production of new organs, but because they can be difficult to isolate and expand in vitro, their use has been limited

  • Embryonic stem cells provide an almost limitless source of cells for regenerative applications, but ethical concerns limit their use; thus, techniques such as somatic cell nuclear transfer and reprogramming are being developed so that embryonic-like stem cells can be obtained without the destruction of an embryo

  • Amniotic fluid and placenta have been shown to contain pluripotent cells that can be driven to differentiate into a wide variety of specific cell types, and these cells might be useful for tissue engineering applications

  • Tissue engineering strategies for regenerating components of the urinary tract, such as the kidney, bladder, and urethra, are extremely promising; tissue engineering strategies for repairing urethral tissue are in clinical use, and laboratory-grown bladders have been implanted into patients

  • While more study is needed, regenerative medicine and tissue engineering might one day produce mainstream treatments for organ failure

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Somatic cell nuclear transfer is a procedure that is used to create embryonic stem cells that are genetically identical to a specific individual.
Figure 2: Different methods of generating male and female gametes from embryonic stem cells.

Similar content being viewed by others

References

  1. Weiner LP (2008) Definitions and criteria for stem cells. Methods Mol Biol 438: 3–8

    PubMed  Google Scholar 

  2. Jain KK (2005) Ethical and regulatory aspects of embryonic stem cell research. Expert Opin Biol Ther 5: 153–162

    CAS  PubMed  Google Scholar 

  3. Ballas CB et al. (2002) Adult bone marrow stem cells for cell and gene therapies: implications for greater use. J Cell Biochem Suppl 38: 20–28

    PubMed  Google Scholar 

  4. Jiao J and Chen DF (2008) Induction of neurogenesis in nonconventional neurogenic regions of the adult central nervous system by niche astrocyte-produced signals. Stem Cells 26: 1221–1230

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Taupin P (2006) Therapeutic potential of adult neural stem cells. Recent Patents CNS Drug Discov 1: 299–303

    CAS  Google Scholar 

  6. Jensen UB et al. (2008) A distinct population of clonogenic and multipotent murine follicular keratinocytes residing in the upper isthmus. J Cell Sci 121: 609–617

    CAS  PubMed  Google Scholar 

  7. Crisan M et al. (2008) Purification and long-term culture of multipotent progenitor cells affiliated with the walls of human blood vessels: myoendothelial cells and pericytes. Methods Cell Biol 86: 295–309

    CAS  PubMed  Google Scholar 

  8. Devine SM (2002) Mesenchymal stem cells: will they have a role in the clinic? J Cell Biochem Suppl 38: 73–79

    PubMed  Google Scholar 

  9. Jiang Y et al. (2002) Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418: 41–49

    CAS  PubMed  Google Scholar 

  10. Spradling A et al. (2001) Stem cells find their niche. Nature 414: 98–104

    CAS  PubMed  Google Scholar 

  11. Caplan AI (2007) Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol 213: 341–347

    CAS  PubMed  Google Scholar 

  12. da Silva Meirelles L et al. (2008) In search of the in vivo identity of mesenchymal stem cells. Stem Cells [10.1634/stemcells.2007-1122]

  13. Duan X et al. (2007) Disrupted-In-Schizophrenia 1 regulates integration of newly generated neurons in the adult brain. Cell 130: 1146–1158

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Crisan M et al. (2008) A reservoir of brown adipocyte progenitors in human skeletal muscle. Stem Cells [10.1634/stemcells.2008-0325]

  15. Luttun A et al. (2006) Differentiation of multipotent adult progenitor cells into functional endothelial and smooth muscle cells. Curr Protoc Immunol 22: Unit 22F.9

    PubMed  Google Scholar 

  16. Mimeault M and Batra SK (2008) Recent progress on tissue-resident adult stem cell biology and their therapeutic implications. Stem Cell Rev 4: 27–49

    PubMed  Google Scholar 

  17. Ikeda E et al. (2008) Multipotent cells from the human third molar: feasibility of cell-based therapy for liver disease. Differentiation 76: 495–505

    CAS  PubMed  Google Scholar 

  18. Nolen-Walston RD et al. (2008) Cellular kinetics and modeling of bronchioalveolar stem cell response during lung regeneration. Am J Physiol Lung Cell Mol Physiol 294: L1158–L1165

    CAS  PubMed  Google Scholar 

  19. In't Anker PS et al. (2003) Mesenchymal stem cells in human second-trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potential. Haematologica 88: 845–852

    Google Scholar 

  20. Hristov M et al. (2008) Adult progenitor cells in vascular remodeling during atherosclerosis. Biol Chem 389: 837–844

    CAS  PubMed  Google Scholar 

  21. Martin GR (1981) Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad Sci USA 78: 7634–7638

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Thomson JA et al. (1998) Embryonic stem cell lines derived from human blastocysts. Science 282: 1145–1147

    CAS  PubMed  Google Scholar 

  23. Polgár K et al. (1989) Characterization of rapidly adhering amniotic fluid cells by combined immunofluorescence and phagocytosis assays. Am J Hum Genet 45: 786–792

    PubMed  PubMed Central  Google Scholar 

  24. Priest RE et al. (1978) Origin of cells in human amniotic fluid cultures: ultrastructural features. Lab Invest 39: 106–109

    CAS  PubMed  Google Scholar 

  25. In't Anker PS et al. (2003) Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 102: 1548–1549

    CAS  Google Scholar 

  26. Tsai MS et al. (2004) Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum Reprod 19: 1450–1456

    PubMed  Google Scholar 

  27. Prusa AR et al. (2004) Neurogenic cells in human amniotic fluid. Am J Obst Gynecol 191: 309–314

    Google Scholar 

  28. Schmidt D et al. (2007) Prenatally fabricated autologous human living heart valves based on amniotic fluid derived progenitor cells as single cell source. Circulation 116 (Suppl 1): I64–I70

    PubMed  Google Scholar 

  29. De Coppi P et al. (2007) Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 25: 100–106

    CAS  PubMed  Google Scholar 

  30. French AJ et al. (2008) Development of human cloned blastocysts following somatic cell nuclear transfer with adult fibroblasts. Stem Cells 26: 485–493

    CAS  PubMed  Google Scholar 

  31. Rideout WM III et al. (2001) Nuclear cloning and epigenetic reprogramming of the genome. Science 293: 1093–1098

    PubMed  Google Scholar 

  32. Rideout WM III et al. (2002) Correction of a genetic defect by nuclear transplantation and combined cell and gene therapy. Cell 109: 17–27

    PubMed  Google Scholar 

  33. Hochedlinger K and Jaenisch R (2002) Monoclonal mice generated by nuclear transfer from mature B and T donor cells. Nature 415: 1035–1038

    CAS  PubMed  Google Scholar 

  34. Eggan K et al. (2004) Mice cloned from olfactory sensory neurons. Nature 428: 44–49

    CAS  PubMed  Google Scholar 

  35. Hochedlinger K and Jaenisch R (2002) Nuclear transplantation: lessons from frogs and mice. Curr Opin Cell Biol 14: 741–748

    CAS  PubMed  Google Scholar 

  36. Hochedlinger K and Jaenisch R (2003) Nuclear transplantation, embryonic stem cells, and the potential for cell therapy. N Engl J Med 349: 275–286

    CAS  PubMed  Google Scholar 

  37. Simerly C et al. (2003) Molecular correlates of primate nuclear transfer failures. Science 300: 297

    PubMed  Google Scholar 

  38. Hwang WS et al. (2004) Evidence of a pluripotent human embryonic stem cell line derived from a cloned blastocyst. Science 303: 1669–1674

    CAS  PubMed  Google Scholar 

  39. Hwang WS et al. (2005) Patient-specific embryonic stem cells derived from human SCNT blastocysts. Science 308: 1777–1783

    CAS  PubMed  Google Scholar 

  40. Byrne JA et al. (2007) Producing primate embryonic stem cells by somatic cell nuclear transfer. Nature 450: 497–502

    CAS  PubMed  Google Scholar 

  41. Vajta G (2007) Somatic cell nuclear transfer in its first and second decades: successes, setbacks, paradoxes and perspectives. Reprod Biomed Online 15: 582–590

    PubMed  Google Scholar 

  42. Chung Y et al. (2006) Embryonic and extraembryonic stem cell lines derived from single mouse blastomeres. Nature 439: 216–219

    CAS  PubMed  Google Scholar 

  43. Zhang X et al. (2006) Derivation of human embryonic stem cells from developing and arrested embryos. Stem Cells 24: 2669–2676

    CAS  PubMed  Google Scholar 

  44. Geber S and Sampaio M (1999) Blastomere development after embryo biopsy: a new model to predict embryo development and to select for transfer. Hum Reprod 14: 782–786

    CAS  PubMed  Google Scholar 

  45. Hardy K (1993) Development of human blastocysts in vitro . In Preimplantation Embryo Development 184–199 (Ed. Bavister BD) New York: Springer–Verlag

    Google Scholar 

  46. Landry DW and Zucker HA et al. (2004) Embryonic death and the creation of human embryonic stem cells. J Clin Invest 114: 1184–1186

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Martinez F et al. (2002) Caspase activity in preimplantation human embryos is not associated with apoptosis. Hum Reprod 17: 1584–1590

    CAS  PubMed  Google Scholar 

  48. Hurlbut WB (2005) Altered nuclear transfer as a morally acceptable means for the procurement of human embryonic stem cells. Perspect Biol Med 48: 211–228

    PubMed  Google Scholar 

  49. Meissner A and Jaenisch R (2006) Generation of nuclear transfer-derived pluripotent ES cells from cloned Cdx2-deficient blastocysts. Nature 439: 212–215

    CAS  PubMed  Google Scholar 

  50. Vauhkonen M et al. (2008) Helicobacter pylori infection induces a reversible expression of the CDX2 transcription factor protein in human gastric epithelium. Scand J Gastroenterol 43: 915–921

    CAS  PubMed  Google Scholar 

  51. Wernig M et al. (2007) In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448: 318–324

    CAS  PubMed  Google Scholar 

  52. Takahashi K and Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126: 663–676

    CAS  PubMed  Google Scholar 

  53. Takahashi K et al. (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131: 861–872

    CAS  PubMed  Google Scholar 

  54. Blelloch R et al. (2006) Reprogramming efficiency following somatic cell nuclear transfer is influenced by the differentiation and methylation state of the donor nucleus. Stem Cells 24: 2007–2013

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Lysaght MJ (2002) Maintenance dialysis population dynamics: current trends and long-term implications. J Am Soc Nephrol 13 (Suppl 1): S37–S40

    PubMed  Google Scholar 

  56. Humes HD et al. (1999) Replacement of renal function in uremic animals with a tissue-engineered kidney. Nat Biotechnol 17: 451–455

    CAS  PubMed  Google Scholar 

  57. MacKay SM et al. (1998) Tissue engineering of a bioartificial renal tubule. ASAIO J 44: 179–183

    CAS  PubMed  Google Scholar 

  58. Aebischer P et al. (1987) The bioartificial kidney: progress towards an ultrafiltration device with renal epithelial cells processing. Life Support Syst 5: 159–168

    CAS  PubMed  Google Scholar 

  59. Ip TK et al. (1988) Cellular control of membrane permeability: implications for a bioartificial renal tubule. ASAIO Trans 34: 351–355

    CAS  PubMed  Google Scholar 

  60. Humes HD (2000) Bioartificial kidney for full renal replacement therapy. Semin Nephrol 20: 71–82

    CAS  PubMed  Google Scholar 

  61. Amiel GE et al. (2000) Renal therapy using tissue-engineered constructs and gene delivery. World J Urol 18: 71–79

    CAS  PubMed  Google Scholar 

  62. Lanza RP et al. (1996) Encapsulated cell technology. Nat Biotechnol 14: 1107–1111

    CAS  PubMed  Google Scholar 

  63. Lanza RP et al. (2002) Generation of histocompatible tissues using nuclear transplantation. Nat Biotechnol 20: 689–696

    CAS  PubMed  Google Scholar 

  64. Humes HD et al. (1996) Tubulogenesis from isolated single cells of adult mammalian kidney: clonal analysis with a recombinant retrovirus. Am J Physiol. Renal Physiol 271: 42–49

    Google Scholar 

  65. Qiao J et al. (1999) Branching morphogenesis independent of mesenchymal-epithelial contact in the developing kidney. Proc Natl Acad Sci USA 96: 7330–7335

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Aboushwareb T et al. (2008) Erythropoietin producing cells for potential cell therapy. World J Urol 26: 295–300

    CAS  PubMed  Google Scholar 

  67. Cilento BG et al. (1994) Phenotypic and cytogenetic characterization of human bladder urothelia expanded in vitro . J Urol 152: 665–670

    CAS  PubMed  Google Scholar 

  68. Atala A et al. (2006) Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet 367: 1241–1246

    PubMed  Google Scholar 

  69. El-Kassaby A et al. (2008) Randomized comparative study between buccal mucosal and acellular bladder matrix grafts in complex anterior urethral strictures. J Urol 179: 1432–1436

    PubMed  Google Scholar 

  70. El-Kassaby AW et al. (2003) Urethral stricture repair with an off-the-shelf collagen matrix. J Urol 169: 170–173

    CAS  PubMed  Google Scholar 

  71. De Filippo RE et al. (2002) Urethral replacement using cell seeded tubularized collagen matrices. J Urol 168: 1789–1792

    CAS  PubMed  Google Scholar 

  72. Orabi HO et al. (2008) Tissue engineered tubularized urethra for surgical reconstruction: a pre-clinical study [abstract]. FASEB J 22: 581.6

    Google Scholar 

  73. Cannon TW et al. (2003) Improved sphincter contractility after allogenic muscle-derived progenitor cell injection into the denervated rat urethra. Urology 62: 958–963

    PubMed  Google Scholar 

  74. Peyromaure M et al. (2004) Fate of implanted syngenic muscle precursor cells in striated urethral sphincter of female rats: perspectives for treatment of urinary incontinence. Urology 64: 1037–1041

    PubMed  Google Scholar 

  75. Lee JY et al. (2004) Long term effects of muscle-derived stem cells on leak point pressure and closing pressure in rats with transected pudendal nerves. Mol Cells 18: 309–313

    CAS  PubMed  Google Scholar 

  76. Mitterberger M et al. (2008) Adult stem cell therapy of female stress urinary incontinence. Eur Urol 53: 169–175

    PubMed  Google Scholar 

  77. Strasser H et al. (2007) Transurethral ultrasonography-guided injection of adult autologous stem cells versus transurethral endoscopic injection of collagen in treatment of urinary incontinence. World J Urol 25: 385–392

    CAS  PubMed  Google Scholar 

  78. Strasser H et al. (2007) Autologous myoblasts and fibroblasts versus collagen for treatment of stress urinary incontinence in women: a randomised controlled trial. Lancet 369: 2179–2186

    CAS  PubMed  Google Scholar 

  79. Hoshi A et al. (2008) Reconstruction of radical prostatectomy-induced urethral damage using skeletal muscle-derived multipotent stem cells. Transplantation 85: 1617–1624

    PubMed  Google Scholar 

  80. Mitterberger M et al. (2008) Myoblast and fibroblast therapy for post-prostatectomy urinary incontinence: 1-year followup of 63 patients. J Urol 179: 226–231

    PubMed  Google Scholar 

  81. Strasser H et al. (2007) Transurethral ultrasound guided injection of autologous myoblasts and fibroblasts in treatment of incontinence after prostate surgery [abstract #1182]. J Urol 177 (Suppl): 390

    Google Scholar 

  82. Kleinert S and Horton R (2008) Retraction—autologous myoblasts and fibroblasts for treatment of stress urinary incontinence: a randomised controlled trial. Lancet 372: 789–790

    PubMed  Google Scholar 

  83. Bhatt RI et al. (2003) Novel method for the isolation and characterisation of the putative prostatic stem cell. Cytometry A 54: 89–99

    PubMed  Google Scholar 

  84. Lawson DA et al. (2007) Isolation and functional characterization of murine prostate stem cells. Proc Natl Acad Sci USA 104: 181–186

    CAS  PubMed  Google Scholar 

  85. Lawson DA and Witte ON (2007) Stem cells in prostate cancer initiation and progression. J Clin Invest 117: 2044–2050

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Hübner K et al. (2003) Derivation of oocytes from mouse embryonic stem cells. Science 300: 1251–1256

    PubMed  Google Scholar 

  87. Toyooka Y et al. (2004) Embryonic stem cells can form germ cells in vitro . Proc Natl Acad Sci USA 100: 11457–11462

    Google Scholar 

  88. Kanatsu-Shinohara M and Shinohara T (2007) Culture and genetic modification of mouse germline stem cells. Ann NY Acad Sci 1120: 59–71

    PubMed  Google Scholar 

  89. Machluf M et al. (2000) Controlled release of therapeutic agents: slow delivery and cell encapsulation. World J Urol 18: 80–83

    CAS  PubMed  Google Scholar 

  90. Machluf M et al. (2003) Microencapsulation of Leydig cells: a system for testosterone supplementation. Endocrinology 144: 4975–4979

    CAS  PubMed  Google Scholar 

  91. Zuk PA et al. (2001) Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 7: 211–228

    CAS  PubMed  Google Scholar 

  92. Shah NM et al. (1996) Alternative neural crest cell fates are instructively promoted by TGFbeta superfamily members. Cell 85: 331–343

    CAS  PubMed  Google Scholar 

  93. Poulsom R et al. (2001) Bone marrow contributes to renal parenchymal turnover and regeneration. J Pathol 195: 229–235

    CAS  PubMed  Google Scholar 

  94. Brinster RL and Avarbock MR (1994) Germline transmission of donor haplotype following spermatogonial transplantation. Proc Natl Acad Sci USA 91: 11303–11307

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Lo KC et al. (2004) De novo testosterone production in luteinizing hormone receptor knockout mice after transplantation of leydig stem cells. Endocrinology 145: 4011–4015

    CAS  PubMed  Google Scholar 

  96. Geijsen N et al. (2004) Derivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 427: 148–154

    CAS  PubMed  Google Scholar 

  97. Strasser H et al. (2004) Stem cell therapy for urinary incontinence [German]. Urologe A 43: 1237–1241

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors wish to thank Dr Jennifer L Olson for editorial assistance with this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anthony Atala.

Ethics declarations

Competing interests

T Aboushwareb has declared that he has conducted research supported by Tengion. A Atala has declared that he has acted as a consultant for and holds stock in Tengion and Plureon, companies involved in developing tissue engineering and stem cell technologies.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Aboushwareb, T., Atala, A. Stem cells in urology. Nat Rev Urol 5, 621–631 (2008). https://doi.org/10.1038/ncpuro1228

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncpuro1228

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing