Skip to main content

Stem Cell Therapy and Tissue Engineering in Urogenital Diseases

  • Chapter
  • First Online:
Advances in Stem Cell Therapy

Part of the book series: Stem Cell Biology and Regenerative Medicine ((STEMCELL))

  • 1399 Accesses

Abstract

The urogenital tract is the target of numerous congenital and acquired disorders that affect quality of life and may threaten patients’ lives. Although the genitourinary tract has its own endogenous progenitor/stem cell depot for repair, in several situations, the defect or functional impairment is beyond the cells’ restorative capacity. Hence, there will be a need to replenish the deficient cells and associated function and/or to replace the diseased tissue or organ. Regenerative medicine, including tissue engineering, has emerged as a potential therapeutic option with extensive applications. It involves the use of cells, scaffolds, or both to repair the kidney, urinary tract, and genital organs. Stem cells come in many types and from variable sources, with the embryonic, adult, amniotic fluid, and induced pluripotent stem cells being the most commonly explored and used. In this chapter, we will review the advances and future potentials of stem cells for regeneration and repair of the genitourinary tract. Mechanisms of actions, preclinical and clinical trials, and types of therapies will be described. We will point to the challenges that face their use and to the possible avenues to overcome these obstacles.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Abbreviations

AMDC:

Autologous muscle-derived stem cell

ASC:

Adipose tissue-derived stem cell

BMP:

Bone morphogenetic protein

BMSC:

Bone marrow-derived stem cell

ECM:

Extracellular matrix

ED:

Erectile dysfunction

EFG:

Epidermal growth factor

EPCs:

Endothelial progenitor cells

ESCs:

Embryonic stem cells

HGF:

Hepatocyte growth factor

HO-1:

Heme-oxygenase-1

HSC:

Hematopoietic stem cell

IGF:

Insulin-like growth factor

iPSCs:

Induced pluripotent stem cells

MSCs:

Mesenchymal stem cells

PD:

Peyronie’s disease

PDE-5:

Phosphodiesterase-5

PLA:

Polylactic acid

SMC:

Smooth muscle cell

STING:

Subureteric transurethral injection

SUI:

Stress urinary incontinence

TA:

Tunica albuginea

UCB:

Umbilical cord blood

UC:

Urothelial cell

UVJ:

Ureterovesical junction

VD:

Voiding dysfunction

VEGF:

Vascular endothelial growth factor

VUR:

Vesicoureteral reflux

References

  1. Till JE, Mc CE. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat Res. 1961;14:213–22.

    Article  CAS  PubMed  Google Scholar 

  2. Vaziri H, Dragowska W, Allsopp RC, Thomas TE, Harley CB, Lansdorp PM. Evidence for a mitotic clock in human hematopoietic stem cells: loss of telomeric DNA with age. Proc Natl Acad Sci U S A. 1994;91(21):9857–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Allsopp RC, Chang E, Kashefi-Aazam M, Rogaev EI, Piatyszek MA, Shay JW, et al. Telomere shortening is associated with cell division in vitro and in vivo. Exp Cell Res. 1995;220(1):194–200.

    Article  CAS  PubMed  Google Scholar 

  4. Krause DS, Theise ND, Collector MI, Henegariu O, Hwang S, Gardner R, et al. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell. 2001;105(3):369–77.

    Article  CAS  PubMed  Google Scholar 

  5. Yin T, Li L. The stem cell niches in bone. J Clin Invest. 2006;116(5):1195–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–7.

    Article  CAS  PubMed  Google Scholar 

  7. Tuan RS, Boland G, Tuli R. Adult mesenchymal stem cells and cell-based tissue engineering. Arthritis Res Ther. 2003;5(1):32–45.

    Article  CAS  PubMed  Google Scholar 

  8. da Silva ML, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119(Pt 11):2204–13.

    Google Scholar 

  9. Lenoir N. Europe confronts the embryonic stem cell research challenge. Science. 2000;287(5457):1425–7.

    Article  CAS  PubMed  Google Scholar 

  10. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76.

    Article  CAS  PubMed  Google Scholar 

  11. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72.

    Article  CAS  PubMed  Google Scholar 

  12. Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448(7151):313–7.

    Article  CAS  PubMed  Google Scholar 

  13. Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, Hochedlinger K, et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature. 2007;448(7151):318–24.

    Article  CAS  PubMed  Google Scholar 

  14. Jaenisch R, Young R. Stem cells, the molecular circuitry of pluripotency and nuclear reprogramming. Cell. 2008;132(4):567–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Miura K, Okada Y, Aoi T, Okada A, Takahashi K, Okita K, et al. Variation in the safety of induced pluripotent stem cell lines. Nat Biotechnol. 2009;27(8):743–5.

    Article  CAS  PubMed  Google Scholar 

  16. Nakagawa M, Takizawa N, Narita M, Ichisaka T, Yamanaka S. Promotion of direct reprogramming by transformation-deficient Myc. Proc Natl Acad Sci U S A. 2010;107(32):14152–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Friedenstein AJ, Piatetzky II S, Petrakova KV. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol. 1966;16(3):381–90.

    CAS  PubMed  Google Scholar 

  18. Dennis JE, Merriam A, Awadallah A, Yoo JU, Johnstone B, Caplan AI. A quadripotential mesenchymal progenitor cell isolated from the marrow of an adult mouse. J Bone Miner Res. 1999;14(5):700–9.

    Article  CAS  PubMed  Google Scholar 

  19. Kadiyala S, Young RG, Thiede MA, Bruder SP. Culture expanded canine mesenchymal stem cells possess osteochondrogenic potential in vivo and in vitro. Cell Transplant. 1997;6(2):125–34.

    Article  CAS  PubMed  Google Scholar 

  20. Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J Cell Biochem. 1997;64(2):278–94.

    Article  CAS  PubMed  Google Scholar 

  21. Sanchez-Ramos J, Song S, Cardozo-Pelaez F, Hazzi C, Stedeford T, Willing A, et al. Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp Neurol. 2000;164(2):247–56.

    Article  CAS  PubMed  Google Scholar 

  22. Resende RR, da Costa JL, Kihara AH, Adhikari A, Lorencon E. Intracellular Ca2+ regulation during neuronal differentiation of murine embryonal carcinoma and mesenchymal stem cells. Stem Cells Dev. 2010;19(3):379–94.

    Article  CAS  PubMed  Google Scholar 

  23. Resende RR, Adhikari A, da Costa JL, Lorencon E, Ladeira MS, Guatimosim S, et al. Influence of spontaneous calcium events on cell-cycle progression in embryonal carcinoma and adult stem cells. Biochim Biophys Acta. 2010;1803(2):246–60.

    Article  CAS  PubMed  Google Scholar 

  24. Young RG, Butler DL, Weber W, Caplan AI, Gordon SL, Fink DJ. Use of mesenchymal stem cells in a collagen matrix for Achilles tendon repair. J Orthop Res. 1998;16(4):406–13.

    Article  CAS  PubMed  Google Scholar 

  25. Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, Stornaiuolo A, Cossu G, et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science. 1998;279(5356):1528–30.

    Article  CAS  PubMed  Google Scholar 

  26. Ghoraishizadeh S, Ghorishizadeh A, Ghoraishizadeh P, Daneshvar N, Boroojerdi MH. Application of nanoscaffolds in mesenchymal stem cell-based therapy. Adv Regen Med. 2014;2014:14.

    Google Scholar 

  27. Hoogduijn MJ, Dor FJ. Mesenchymal stem cells in transplantation and tissue regeneration. Front Immunol. 2011;2:84.

    PubMed  PubMed Central  Google Scholar 

  28. Vaegler M, Lenis AT, Daum L, Amend B, Stenzl A, Toomey P, et al. Stem cell therapy for voiding and erectile dysfunction. Nat Rev Urol. 2012;9(8):435–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103(11):1204–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chermansky CJ, Cannon TW, Torimoto K, Fraser MO, Yoshimura N, de Groat WC, et al. A model of intrinsic sphincteric deficiency in the rat: electrocauterization. Neurourol Urodyn. 2004;23(2):166–71.

    Article  PubMed  Google Scholar 

  31. Carr LK, Robert M, Kultgen PL, Herschorn S, Birch C, Murphy M, et al. Autologous muscle derived cell therapy for stress urinary incontinence: a prospective, dose ranging study. J Urol. 2013;189(2):595–601.

    Article  PubMed  Google Scholar 

  32. Cruz M, Dissaranan C, Cotleur A, Kiedrowski M, Penn M, Damaser M. Pelvic organ distribution of mesenchymal stem cells injected intravenously after simulated childbirth injury in female rats. Obstet Gynecol Int. 2012;2012:612946.

    PubMed  Google Scholar 

  33. Kim SO, Na HS, Kwon D, Joo SY, Kim HS, Ahn Y. Bone-marrow-derived mesenchymal stem cell transplantation enhances closing pressure and leak point pressure in a female urinary incontinence rat model. Urol Int. 2011;86(1):110–6.

    Article  PubMed  Google Scholar 

  34. Humphreys BD, Bonventre JV. Mesenchymal stem cells in acute kidney injury. Annu Rev Med. 2008;59:311–25.

    Article  CAS  PubMed  Google Scholar 

  35. Lange C, Togel F, Ittrich H, Clayton F, Nolte-Ernsting C, Zander AR, et al. Administered mesenchymal stem cells enhance recovery from ischemia/reperfusion-induced acute renal failure in rats. Kidney Int. 2005;68(4):1613–7.

    Article  PubMed  Google Scholar 

  36. Zoja C, Garcia PB, Rota C, Conti S, Gagliardini E, Corna D, et al. Mesenchymal stem cell therapy promotes renal repair by limiting glomerular podocyte and progenitor cell dysfunction in adriamycin-induced nephropathy. Am J Physiol Renal Physiol. 2012;303(9):F1370–81.

    Article  CAS  PubMed  Google Scholar 

  37. Kunter U, Rong S, Djuric Z, Boor P, Muller-Newen G, Yu D, et al. Transplanted mesenchymal stem cells accelerate glomerular healing in experimental glomerulonephritis. J Am Soc Nephrol. 2006;17(8):2202–12.

    Article  CAS  PubMed  Google Scholar 

  38. Franquesa M, Herrero E, Torras J, Ripoll E, Flaquer M, Goma M, et al. Mesenchymal stem cell therapy prevents interstitial fibrosis and tubular atrophy in a rat kidney allograft model. Stem Cells Dev. 2012;21(17):3125–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Togel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C. Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am J Physiol Renal Physiol. 2005;289(1):F31–42.

    Article  PubMed  CAS  Google Scholar 

  40. Togel F, Weiss K, Yang Y, Hu Z, Zhang P, Westenfelder C. Vasculotropic, paracrine actions of infused mesenchymal stem cells are important to the recovery from acute kidney injury. Am J Physiol Renal Physiol. 2007;292(5):F1626–35.

    Article  CAS  PubMed  Google Scholar 

  41. Imberti B, Morigi M, Tomasoni S, Rota C, Corna D, Longaretti L, et al. Insulin-like growth factor-1 sustains stem cell mediated renal repair. J Am Soc Nephrol. 2007;18(11):2921–8.

    Article  CAS  PubMed  Google Scholar 

  42. Xinaris C, Morigi M, Benedetti V, Imberti B, Fabricio AS, Squarcina E, et al. A novel strategy to enhance mesenchymal stem cell migration capacity and promote tissue repair in an injury specific fashion. Cell Transplant. 2013;22(3):423–36.

    Article  CAS  PubMed  Google Scholar 

  43. Bi B, Guo J, Marlier A, Lin SR, Cantley LG. Erythropoietin expands a stromal cell population that can mediate renoprotection. Am J Physiol Renal Physiol. 2008;295(4):F1017–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zarjou A, Kim J, Traylor AM, Sanders PW, Balla J, Agarwal A, et al. Paracrine effects of mesenchymal stem cells in cisplatin-induced renal injury require heme oxygenase-1. Am J Physiol Renal Physiol. 2011;300(1):F254–62.

    Article  CAS  PubMed  Google Scholar 

  45. Liu H, Liu S, Li Y, Wang X, Xue W, Ge G, et al. The role of SDF-1-CXCR4/CXCR7 axis in the therapeutic effects of hypoxia-preconditioned mesenchymal stem cells for renal ischemia/reperfusion injury. PLoS One. 2012;7(4), e34608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Herrera MB, Bussolati B, Bruno S, Morando L, Mauriello-Romanazzi G, Sanavio F, et al. Exogenous mesenchymal stem cells localize to the kidney by means of CD44 following acute tubular injury. Kidney Int. 2007;72(4):430–41.

    Article  CAS  PubMed  Google Scholar 

  47. Kunter U, Rong S, Boor P, Eitner F, Muller-Newen G, Djuric Z, et al. Mesenchymal stem cells prevent progressive experimental renal failure but maldifferentiate into glomerular adipocytes. J Am Soc Nephrol. 2007;18(6):1754–64.

    Article  CAS  PubMed  Google Scholar 

  48. Miki T, Lehmann T, Cai H, Stolz DB, Strom SC. Stem cell characteristics of amniotic epithelial cells. Stem Cells. 2005;23(10):1549–59.

    Article  CAS  PubMed  Google Scholar 

  49. Parolini O, Alviano F, Bagnara GP, Bilic G, Buhring HJ, Evangelista M, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international Workshop on Placenta Derived Stem Cells. Stem Cells. 2008;26(2):300–11.

    Article  PubMed  Google Scholar 

  50. Tsai MS, Lee JL, Chang YJ, Hwang SM. Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum Reprod. 2004;19(6):1450–6.

    Article  PubMed  Google Scholar 

  51. In’t Anker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FH, Willemze R, et al. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood. 2003;102(4):1548–9.

    Article  Google Scholar 

  52. Underwood MA, Gilbert WM, Sherman MP. Amniotic fluid: not just fetal urine anymore. J Perinatol. 2005;25(5):341–8.

    Article  PubMed  Google Scholar 

  53. Prusa AR, Marton E, Rosner M, Bernaschek G, Hengstschlager M. Oct-4-expressing cells in human amniotic fluid: a new source for stem cell research? Hum Reprod. 2003;18(7):1489–93.

    Article  PubMed  Google Scholar 

  54. De Coppi P, Bartsch Jr G, Siddiqui MM, Xu T, Santos CC, Perin L, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol. 2007;25(1):100–6.

    Article  PubMed  CAS  Google Scholar 

  55. Peister A, Mellad JA, Larson BL, Hall BM, Gibson LF, Prockop DJ. Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood. 2004;103(5):1662–8.

    Article  CAS  PubMed  Google Scholar 

  56. Wexler SA, Donaldson C, Denning-Kendall P, Rice C, Bradley B, Hows JM. Adult bone marrow is a rich source of human mesenchymal ‘stem’ cells but umbilical cord and mobilized adult blood are not. Br J Haematol. 2003;121(2):368–74.

    Article  PubMed  Google Scholar 

  57. Lee OK, Kuo TK, Chen WM, Lee KD, Hsieh SL, Chen TH. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood. 2004;103(5):1669–75.

    Article  CAS  PubMed  Google Scholar 

  58. Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, et al. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells. 2004;22(7):1330–7.

    Article  PubMed  Google Scholar 

  59. Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109(1):235–42.

    Article  CAS  PubMed  Google Scholar 

  60. Udagawa N, Takahashi N, Akatsu T, Tanaka H, Sasaki T, Nishihara T, et al. Origin of osteoclasts: mature monocytes and macrophages are capable of differentiating into osteoclasts under a suitable microenvironment prepared by bone marrow-derived stromal cells. Proc Natl Acad Sci U S A. 1990;87(18):7260–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Suda T, Takahashi N, Martin TJ. Modulation of osteoclast differentiation. Endocr Rev. 1992;13(1):66–80.

    CAS  PubMed  Google Scholar 

  62. Bendall LJ, Kortlepel K, Gottlieb DJ. Human acute myeloid leukemia cells bind to bone marrow stroma via a combination of beta-1 and beta-2 integrin mechanisms. Blood. 1993;82(10):3125–32.

    CAS  PubMed  Google Scholar 

  63. Conget PA, Minguell JJ. Phenotypical and functional properties of human bone marrow mesenchymal progenitor cells. J Cell Physiol. 1999;181(1):67–73.

    Article  CAS  PubMed  Google Scholar 

  64. Ribeiro A, Laranjeira P, Mendes S, Velada I, Leite C, Andrade P, et al. Mesenchymal stem cells from umbilical cord matrix, adipose tissue and bone marrow exhibit different capability to suppress peripheral blood B, natural killer and T cells. Stem Cell Res Ther. 2013;4(5):125.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7(2):211–28.

    Article  CAS  PubMed  Google Scholar 

  66. Lindroos B, Suuronen R, Miettinen S. The potential of adipose stem cells in regenerative medicine. Stem Cell Rev. 2011;7(2):269–91.

    Article  PubMed  Google Scholar 

  67. Bailey AM, Kapur S, Katz AJ. Characterization of adipose-derived stem cells: an update. Curr Stem Cell Res Ther. 2010;5(2):95–102.

    Article  CAS  PubMed  Google Scholar 

  68. Suga H, Matsumoto D, Eto H, Inoue K, Aoi N, Kato H, et al. Functional implications of CD34 expression in human adipose-derived stem/progenitor cells. Stem Cells Dev. 2009;18(8):1201–10.

    Article  CAS  PubMed  Google Scholar 

  69. Zimmerlin L, Donnenberg VS, Pfeifer ME, Meyer EM, Peault B, Rubin JP, et al. Stromal vascular progenitors in adult human adipose tissue. Cytometry A. 2010;77(1):22–30.

    PubMed  PubMed Central  Google Scholar 

  70. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275(5302):964–7.

    Article  CAS  PubMed  Google Scholar 

  71. Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, et al. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 1999;85(3):221–8.

    Article  CAS  PubMed  Google Scholar 

  72. Nolan DJ, Ciarrocchi A, Mellick AS, Jaggi JS, Bambino K, Gupta S, et al. Bone marrow-derived endothelial progenitor cells are a major determinant of nascent tumor neovascularization. Genes Dev. 2007;21(12):1546–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, et al. Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med. 2005;353(10):999–1007.

    Article  CAS  PubMed  Google Scholar 

  74. Kilarski WW, Samolov B, Petersson L, Kvanta A, Gerwins P. Biomechanical regulation of blood vessel growth during tissue vascularization. Nat Med. 2009;15(6):657–64.

    Article  CAS  PubMed  Google Scholar 

  75. Stewart K, Bouchard M. Coordinated cell behaviours in early urogenital system morphogenesis. Semin Cell Dev Biol. 2014;36:13–20.

    Article  CAS  PubMed  Google Scholar 

  76. Bertram JF, Douglas-Denton RN, Diouf B, Hughson MD, Hoy WE. Human nephron number: implications for health and disease. Pediatr Nephrol. 2011;26(9):1529–33.

    Article  PubMed  Google Scholar 

  77. Quaggin SE, Kreidberg JA. Development of the renal glomerulus: good neighbors and good fences. Development. 2008;135(4):609–20.

    Article  CAS  PubMed  Google Scholar 

  78. Krause D, Cantley LG. Bone marrow plasticity revisited: protection or differentiation in the kidney tubule? J Clin Invest. 2005;115(7):1705–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Smeets B, Moeller MJ. Parietal epithelial cells and podocytes in glomerular diseases. Semin Nephrol. 2012;32(4):357–67.

    Article  CAS  PubMed  Google Scholar 

  80. Romagnani P, Lasagni L, Remuzzi G. Renal progenitors: an evolutionary conserved strategy for kidney regeneration. Nat Rev Nephrol. 2013;9(3):137–46.

    Article  CAS  PubMed  Google Scholar 

  81. Matsumoto K, Mizuno S, Nakamura T. Hepatocyte growth factor in renal regeneration, renal disease and potential therapeutics. Curr Opin Nephrol Hypertens. 2000;9(4):395–402.

    Article  CAS  PubMed  Google Scholar 

  82. Humes HD, Cieslinski DA, Coimbra TM, Messana JM, Galvao C. Epidermal growth factor enhances renal tubule cell regeneration and repair and accelerates the recovery of renal function in postischemic acute renal failure. J Clin Invest. 1989;84(6):1757–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Miller SB, Martin DR, Kissane J, Hammerman MR. Insulin-like growth factor I accelerates recovery from ischemic acute tubular necrosis in the rat. Proc Natl Acad Sci U S A. 1992;89(24):11876–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Homma T, Sakai M, Cheng HF, Yasuda T, Coffey Jr RJ, Harris RC. Induction of heparin-binding epidermal growth factor-like growth factor mRNA in rat kidney after acute injury. J Clin Invest. 1995;96(2):1018–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Nakagawa T, Sasahara M, Haneda M, Kataoka H, Nakagawa H, Yagi M, et al. Role of PDGF B-chain and PDGF receptors in rat tubular regeneration after acute injury. Am J Pathol. 1999;155(5):1689–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Vukicevic S, Basic V, Rogic D, Basic N, Shih MS, Shepard A, et al. Osteogenic protein-1 (bone morphogenetic protein-7) reduces severity of injury after ischemic acute renal failure in rat. J Clin Invest. 1998;102(1):202–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Yanagita M, Okuda T, Endo S, Tanaka M, Takahashi K, Sugiyama F, et al. Uterine sensitization-associated gene-1 (USAG-1), a novel BMP antagonist expressed in the kidney, accelerates tubular injury. J Clin Invest. 2006;116(1):70–9.

    Article  CAS  PubMed  Google Scholar 

  88. Maeshima A, Nojima Y, Kojima I. The role of the activin-follistatin system in the developmental and regeneration processes of the kidney. Cytokine Growth Factor Rev. 2001;12(4):289–98.

    Article  CAS  PubMed  Google Scholar 

  89. Li B, Cohen A, Hudson TE, Motlagh D, Amrani DL, Duffield JS. Mobilized human hematopoietic stem/progenitor cells promote kidney repair after ischemia/reperfusion injury. Circulation. 2010;121(20):2211–20.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Stokman G, Leemans JC, Claessen N, Weening JJ, Florquin S. Hematopoietic stem cell mobilization therapy accelerates recovery of renal function independent of stem cell contribution. J Am Soc Nephrol. 2005;16(6):1684–92.

    Article  CAS  PubMed  Google Scholar 

  91. Iwasaki M, Adachi Y, Minamino K, Suzuki Y, Zhang Y, Okigaki M, et al. Mobilization of bone marrow cells by G-CSF rescues mice from cisplatin-induced renal failure, and M-CSF enhances the effects of G-CSF. J Am Soc Nephrol. 2005;16(3):658–66.

    Article  CAS  PubMed  Google Scholar 

  92. Togel F, Isaac J, Westenfelder C. Hematopoietic stem cell mobilization-associated granulocytosis severely worsens acute renal failure. J Am Soc Nephrol. 2004;15(5):1261–7.

    Article  PubMed  Google Scholar 

  93. Dekel B, Shezen E, Even-Tov-Friedman S, Katchman H, Margalit R, Nagler A, et al. Transplantation of human hematopoietic stem cells into ischemic and growing kidneys suggests a role in vasculogenesis but not tubulogenesis. Stem Cells. 2006;24(5):1185–93.

    Article  CAS  PubMed  Google Scholar 

  94. Duffield JS, Park KM, Hsiao LL, Kelley VR, Scadden DT, Ichimura T, et al. Restoration of tubular epithelial cells during repair of the postischemic kidney occurs independently of bone marrow-derived stem cells. J Clin Invest. 2005;115(7):1743–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Lin F, Moran A, Igarashi P. Intrarenal cells, not bone marrow-derived cells, are the major source for regeneration in postischemic kidney. J Clin Invest. 2005;115(7):1756–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Humphreys BD, Valerius MT, Kobayashi A, Mugford JW, Soeung S, Duffield JS, et al. Intrinsic epithelial cells repair the kidney after injury. Cell Stem Cell. 2008;2(3):284–91.

    Article  CAS  PubMed  Google Scholar 

  97. Chen YT, Sun CK, Lin YC, Chang LT, Chen YL, Tsai TH, et al. Adipose-derived mesenchymal stem cell protects kidneys against ischemia-reperfusion injury through suppressing oxidative stress and inflammatory reaction. J Transl Med. 2011;9:51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Eirin A, Zhu XY, Krier JD, Tang H, Jordan KL, Grande JP, et al. Adipose tissue-derived mesenchymal stem cells improve revascularization outcomes to restore renal function in swine atherosclerotic renal artery stenosis. Stem Cells. 2012;30(5):1030–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Ebrahimi B, Eirin A, Li Z, Zhu XY, Zhang X, Lerman A, et al. Mesenchymal stem cells improve medullary inflammation and fibrosis after revascularization of swine atherosclerotic renal artery stenosis. PLoS One. 2013;8(7), e67474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Shi H, Patschan D, Dietz GP, Bahr M, Plotkin M, Goligorsky MS. Glial cell line-derived neurotrophic growth factor increases motility and survival of cultured mesenchymal stem cells and ameliorates acute kidney injury. Am J Physiol Renal Physiol. 2008;294(1):F229–35.

    Article  CAS  PubMed  Google Scholar 

  101. Yuan L, Wu MJ, Sun HY, Xiong J, Zhang Y, Liu CY, et al. VEGF-modified human embryonic mesenchymal stem cell implantation enhances protection against cisplatin-induced acute kidney injury. Am J Physiol Renal Physiol. 2011;300(1):F207–18.

    Article  CAS  PubMed  Google Scholar 

  102. Kim DS, Cho HJ, Choi HR, Kwon SB, Park KC. Isolation of human epidermal stem cells by adherence and the reconstruction of skin equivalents. Cell Mol Life Sci. 2004;61(21):2774–81.

    Article  CAS  PubMed  Google Scholar 

  103. Morizane R, Monkawa T, Fujii S, Yamaguchi S, Homma K, Matsuzaki Y, et al. Kidney specific protein-positive cells derived from embryonic stem cells reproduce tubular structures in vitro and differentiate into renal tubular cells. PLoS One. 2014;8(6), e64843.

    Article  PubMed  CAS  Google Scholar 

  104. Vigneau C, Polgar K, Striker G, Elliott J, Hyink D, Weber O, et al. Mouse embryonic stem cell-derived embryoid bodies generate progenitors that integrate long term into renal proximal tubules in vivo. J Am Soc Nephrol. 2007;18(6):1709–20.

    Article  CAS  PubMed  Google Scholar 

  105. Chade AR, Zhu X, Lavi R, Krier JD, Pislaru S, Simari RD, et al. Endothelial progenitor cells restore renal function in chronic experimental renovascular disease. Circulation. 2009;119(4):547–57.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Chade AR, Zhu XY, Krier JD, Jordan KL, Textor SC, Grande JP, et al. Endothelial progenitor cells homing and renal repair in experimental renovascular disease. Stem Cells. 2010;28(6):1039–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Hauser PV, De Fazio R, Bruno S, Sdei S, Grange C, Bussolati B, et al. Stem cells derived from human amniotic fluid contribute to acute kidney injury recovery. Am J Pathol. 2010;177(4):2011–21.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Sedrakyan S, Da Sacco S, Milanesi A, Shiri L, Petrosyan A, Varimezova R, et al. Injection of amniotic fluid stem cells delays progression of renal fibrosis. J Am Soc Nephrol. 2012;23(4):661–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Perin L, Sedrakyan S, Giuliani S, Da Sacco S, Carraro G, Shiri L, et al. Protective effect of human amniotic fluid stem cells in an immunodeficient mouse model of acute tubular necrosis. PLoS One. 2010;5(2), e9357.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Mae S, Shono A, Shiota F, Yasuno T, Kajiwara M, Gotoda-Nishimura N, et al. Monitoring and robust induction of nephrogenic intermediate mesoderm from human pluripotent stem cells. Nat Commun. 2013;4:1367.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Araoka T, Mae S, Kurose Y, Uesugi M, Ohta A, Yamanaka S, et al. Efficient and rapid induction of human iPSCs/ESCs into nephrogenic intermediate mesoderm using small molecule-based differentiation methods. PLoS One. 2014;9(1), e84881.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Taguchi A, Kaku Y, Ohmori T, Sharmin S, Ogawa M, Sasaki H, et al. Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells. Cell Stem Cell. 2014;14(1):53–67.

    Article  CAS  PubMed  Google Scholar 

  113. Song JJ, Guyette JP, Gilpin SE, Gonzalez G, Vacanti JP, Ott HC. Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat Med. 2013;19(5):646–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Burgkart R, Tron A, Prodinger P, Culmes M, Tuebel J, van Griensven M, et al. Decellularized kidney matrix for perfused bone engineering. Tissue Eng Part C Methods. 2014;20(7):553–61.

    Article  PubMed  Google Scholar 

  115. Sullivan DC, Mirmalek-Sani SH, Deegan DB, Baptista PM, Aboushwareb T, Atala A, et al. Decellularization methods of porcine kidneys for whole organ engineering using a high-throughput system. Biomaterials. 2012;33(31):7756–64.

    Article  CAS  PubMed  Google Scholar 

  116. Orlando G, Booth C, Wang Z, Totonelli G, Ross CL, Moran E, et al. Discarded human kidneys as a source of ECM scaffold for kidney regeneration technologies. Biomaterials. 2013;34(24):5915–25.

    Article  CAS  PubMed  Google Scholar 

  117. Aggarwal S, Moggio A, Bussolati B. Concise review: stem/progenitor cells for renal tissue repair: current knowledge and perspectives. Stem Cells Transl Med. 2013;2(12):1011–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Takasato M, Er PX, Becroft M, Vanslambrouck JM, Stanley EG, Elefanty AG, et al. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat Cell Biol. 2014;16(1):118–26.

    Article  CAS  PubMed  Google Scholar 

  119. Simaioforidis V, de Jonge P, Sloff M, Oosterwijk E, Geutjes P, Feitz WF. Ureteral tissue engineering: where are we and how to proceed? Tissue Eng Part B Rev. 2013;19(5):413–9.

    Article  CAS  PubMed  Google Scholar 

  120. de Jonge PK, Simaioforidis V, Geutjes PJ, Oosterwijk E, Feitz WF. Recent advances in ureteral tissue engineering. Curr Urol Rep. 2015;16(1):465.

    Article  PubMed  Google Scholar 

  121. Fu Q, Deng CL, Zhao RY, Wang Y, Cao Y. The effect of mechanical extension stimulation combined with epithelial cell sorting on outcomes of implanted tissue-engineered muscular urethras. Biomaterials. 2014;35(1):105–12.

    Article  CAS  PubMed  Google Scholar 

  122. Shi JG, Fu WJ, Wang XX, Xu YD, Li G, Hong BF, et al. Tissue engineering of ureteral grafts by seeding urothelial differentiated hADSCs onto biodegradable ureteral scaffolds. J Biomed Mater Res A. 2012;100(10):2612–22.

    Article  PubMed  CAS  Google Scholar 

  123. Liao W, Yang S, Song C, Li X, Li Y, Xiong Y. Construction of ureteral grafts by seeding bone marrow mesenchymal stem cells and smooth muscle cells into bladder acellular matrix. Transplant Proc. 2013;45(2):730–4.

    Article  CAS  PubMed  Google Scholar 

  124. Zhao Z, Yu H, Xiao F, Wang X, Yang S, Li S. Differentiation of adipose-derived stem cells promotes regeneration of smooth muscle for ureteral tissue engineering. J Surg Res. 2012;178(1):55–62.

    Article  CAS  PubMed  Google Scholar 

  125. Hicks RM. The function of the golgi complex in transitional epithelium. Synthesis of the thick cell membrane. J Cell Biol. 1966;30(3):623–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Andersson KE, Arner A. Urinary bladder contraction and relaxation: physiology and pathophysiology. Physiol Rev. 2004;84(3):935–86.

    Article  CAS  PubMed  Google Scholar 

  127. Fowler CJ, Griffiths D, de Groat WC. The neural control of micturition. Nat Rev Neurosci. 2008;9(6):453–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Lang R, Liu G, Shi Y, Bharadwaj S, Leng X, Zhou X, et al. Self-renewal and differentiation capacity of urine-derived stem cells after urine preservation for 24 hours. PLoS One. 2013;8(1), e53980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Cotsarelis G, Cheng SZ, Dong G, Sun TT, Lavker RM. Existence of slow-cycling limbal epithelial basal cells that can be preferentially stimulated to proliferate: implications on epithelial stem cells. Cell. 1989;57(2):201–9.

    Article  CAS  PubMed  Google Scholar 

  130. Kurzrock EA, Lieu DK, Degraffenried LA, Chan CW, Isseroff RR. Label-retaining cells of the bladder: candidate urothelial stem cells. Am J Physiol Renal Physiol. 2008;294(6):F1415–21.

    Article  CAS  PubMed  Google Scholar 

  131. Zhang H, Lin G, Qiu X, Ning H, Banie L, Lue TF, et al. Label retaining and stem cell marker expression in the developing rat urinary bladder. Urology. 2012;79(3):746.e1–6.

    PubMed  Google Scholar 

  132. Turedi S, Incealtin O, Hos G. Complications associated with ureterosigmoidostomy—colon carcinoma and ascendens infection resulting in nephrectomy: a case report. Acta Chir Belg. 2009;109(4):531–3.

    Article  CAS  PubMed  Google Scholar 

  133. Salom EM, Mendez LE, Schey D, Lambrou N, Kassira N, Gomez-Marn O, et al. Continent ileocolonic urinary reservoir (Miami pouch): the University of Miami experience over 15 years. Am J Obstet Gynecol. 2004;190(4):994–1003.

    Article  PubMed  Google Scholar 

  134. Orabi H, Bouhout S, Morissette A, Rousseau A, Chabaud S, Bolduc S. Tissue engineering of urinary bladder and urethra: advances from bench to patients. ScientificWorldJournal. 2013;2013:154564.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Donnenberg VS, Zimmerlin L, Rubin JP, Donnenberg AD. Regenerative therapy after cancer: what are the risks? Tissue Eng Part B Rev. 2010;16(6):567–75.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Subramaniam R, Hinley J, Stahlschmidt J, Southgate J. Tissue engineering potential of urothelial cells from diseased bladders. J Urol. 2011;186(5):2014–20.

    Article  CAS  PubMed  Google Scholar 

  137. Zhang R, Jack GS, Rao N, Zuk P, Ignarro LJ, Wu B, et al. Nuclear fusion-independent smooth muscle differentiation of human adipose-derived stem cells induced by a smooth muscle environment. Stem Cells. 2012;30(3):481–90.

    Article  PubMed  CAS  Google Scholar 

  138. Tian H, Bharadwaj S, Liu Y, Ma PX, Atala A, Zhang Y. Differentiation of human bone marrow mesenchymal stem cells into bladder cells: potential for urological tissue engineering. Tissue Eng Part A. 2010;16(5):1769–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Shi JG, Fu WJ, Wang XX, Xu YD, Li G, Hong BF, et al. Transdifferentiation of human adipose-derived stem cells into urothelial cells: potential for urinary tract tissue engineering. Cell Tissue Res. 2012.

    Google Scholar 

  140. Gandhi D, Molotkov A, Batourina E, Schneider K, Dan H, Reiley M, et al. Retinoid signaling in progenitors controls specification and regeneration of the urothelium. Dev Cell. 2013;26(5):469–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Liu J, Huang J, Lin T, Zhang C, Yin X. Cell-to-cell contact induces human adipose tissue-derived stromal cells to differentiate into urothelium-like cells in vitro. Biochem Biophys Res Commun. 2009;390(3):931–6.

    Article  CAS  PubMed  Google Scholar 

  142. Zhang M, Peng Y, Zhou Z, Zhou J, Wang Z, Lu M. Differentiation of human adipose-derived stem cells co-cultured with urothelium cell line toward a urothelium-like phenotype in a nude murine model. Urology. 2013;81(2):465.e15–22.

    PubMed  Google Scholar 

  143. Jack GS, Zhang R, Lee M, Xu Y, Wu BM, Rodriguez LV. Urinary bladder smooth muscle engineered from adipose stem cells and a three dimensional synthetic composite. Biomaterials. 2009;30(19):3259–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Moad M, Pal D, Hepburn AC, Williamson SC, Wilson L, Lako M, et al. A novel model of urinary tract differentiation, tissue regeneration, and disease: reprogramming human prostate and bladder cells into induced pluripotent stem cells. Eur Urol. 2013;64(5):753–61.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Xue Y, Cai X, Wang L, Liao B, Zhang H, Shan Y, et al. Generating a non-integrating human induced pluripotent stem cell bank from urine-derived cells. PLoS One. 2013;8(8), e70573.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Osborn SL, Kurzrock EA. Production of urothelium from pluripotent stem cells for regenerative applications. Curr Urol Rep. 2015;16(1):466.

    Article  PubMed  Google Scholar 

  147. Kim JH, Lee SR, Song YS, Lee HJ. Stem cell therapy in bladder dysfunction: where are we? And where do we have to go? Biomed Res Int. 2013;2013:930713.

    PubMed  PubMed Central  Google Scholar 

  148. Song YS, Lee HJ, Doo SH, Lee SJ, Lim I, Chang KT, et al. Mesenchymal stem cells overexpressing hepatocyte growth factor (HGF) inhibit collagen deposit and improve bladder function in rat model of bladder outlet obstruction. Cell Transplant. 2012;21(8):1641–50.

    Article  PubMed  Google Scholar 

  149. Nitta M, Tamaki T, Tono K, Okada Y, Masuda M, Akatsuka A, et al. Reconstitution of experimental neurogenic bladder dysfunction using skeletal muscle-derived multipotent stem cells. Transplantation. 2010;89(9):1043–9.

    Article  PubMed  Google Scholar 

  150. Orabi H, Goulet C, Rousseau A, Fradette J, Bolduc S. Adipose derived stem cells for treatment of lower genitourinary dysfunction. J Stem Cell Res Ther. 2014;4(4):190.

    Google Scholar 

  151. Levanovich PE, Diokno A, Hasenau DL, Lajiness M, Pruchnic R, Chancellor MB. Intradetrusor injection of adult muscle-derived cells for the treatment of underactive bladder: pilot study. Int Urol Nephrol. 2015;47(3):465–7.

    Article  PubMed  Google Scholar 

  152. Diamond DA, Caldamone AA. Endoscopic correction of vesicoureteral reflux in children using autologous chondrocytes: preliminary results. J Urol. 1999;162(3 Pt 2):1185–8.

    CAS  PubMed  Google Scholar 

  153. Pichler R, Klima G, Richter E, Marksteiner R, Mayr V, Skradski V, et al. Autologous fibroblast transplantation at the vesico-ureteral junction as potential reconstructive cell replacement in an animal model. World J Urol. 2013;31(1):169–74.

    Article  PubMed  Google Scholar 

  154. Wu S, Wang Z, Bharadwaj S, Hodges SJ, Atala A, Zhang Y. Implantation of autologous urine derived stem cells expressing vascular endothelial growth factor for potential use in genitourinary reconstruction. J Urol. 2011;186(2):640–7.

    Article  CAS  PubMed  Google Scholar 

  155. Rogers RG. Clinical practice. Urinary stress incontinence in women. N Engl J Med. 2008;358(10):1029–36.

    Article  CAS  PubMed  Google Scholar 

  156. Waetjen LE, Liao S, Johnson WO, Sampselle CM, Sternfield B, Harlow SD, et al. Factors associated with prevalent and incident urinary incontinence in a cohort of midlife women: a longitudinal analysis of data: study of women’s health across the nation. Am J Epidemiol. 2007;165(3):309–18.

    Article  PubMed  Google Scholar 

  157. Hou JC, Alhalabi F, Lemack GE, Zimmern PE. Outcome of transvaginal mesh and tape removed for pain only. J Urol. 2014;192(3):856–60.

    Article  PubMed  Google Scholar 

  158. Khan ZA, Nambiar A, Morley R, Chapple CR, Emery SJ, Lucas MG. Long-term follow-up of a multicentre randomised controlled trial comparing tension-free vaginal tape, xenograft and autologous fascial slings for the treatment of stress urinary incontinence in women. BJU Int. 2015;115(6):968–77.

    Article  PubMed  Google Scholar 

  159. Kerr LA. Bulking agents in the treatment of stress urinary incontinence: history, outcomes, patient populations, and reimbursement profile. Rev Urol. 2005;7 Suppl 1:S3–11.

    PubMed  PubMed Central  Google Scholar 

  160. Kirchin V, Page T, Keegan PE, Atiemo K, Cody JD, McClinton S. Urethral injection therapy for urinary incontinence in women. Cochrane Database Syst Rev. 2012;2, CD003881.

    Google Scholar 

  161. Wu G, Song Y, Zheng X, Jiang Z. Adipose-derived stromal cell transplantation for treatment of stress urinary incontinence. Tissue Cell. 2011;43(4):246–53.

    Article  PubMed  Google Scholar 

  162. Lin G, Wang G, Banie L, Ning H, Shindel AW, Fandel TM, et al. Treatment of stress urinary incontinence with adipose tissue-derived stem cells. Cytotherapy. 2010;12(1):88–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Zhao W, Zhang C, Jin C, Zhang Z, Kong D, Xu W, et al. Periurethral injection of autologous adipose-derived stem cells with controlled-release nerve growth factor for the treatment of stress urinary incontinence in a rat model. Eur Urol. 2011;59(1):155–63.

    Article  CAS  PubMed  Google Scholar 

  164. Kim BS, Chun SY, Lee JK, Lim HJ, Bae JS, Chung HY, et al. Human amniotic fluid stem cell injection therapy for urethral sphincter regeneration in an animal model. BMC Med. 2012;10:94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Chun SY, Kwon JB, Chae SY, Lee JK, Bae JS, Kim BS, et al. Combined injection of three different lineages of early-differentiating human amniotic fluid-derived cells restores urethral sphincter function in urinary incontinence. BJU Int. 2014;114(5):770–83.

    Article  PubMed  Google Scholar 

  166. Liu G, Wang X, Sun X, Deng C, Atala A, Zhang Y. The effect of urine-derived stem cells expressing VEGF loaded in collagen hydrogels on myogenesis and innervation following after subcutaneous implantation in nude mice. Biomaterials. 2013;34(34):8617–29.

    Article  CAS  PubMed  Google Scholar 

  167. Zou XH, Zhi YL, Chen X, Jin HM, Wang LL, Jiang YZ, et al. Mesenchymal stem cell seeded knitted silk sling for the treatment of stress urinary incontinence. Biomaterials. 2010;31(18):4872–9.

    Article  CAS  PubMed  Google Scholar 

  168. Shi LB, Cai HX, Chen LK, Wu Y, Zhu SA, Gong XN, et al. Tissue engineered bulking agent with adipose-derived stem cells and silk fibroin microspheres for the treatment of intrinsic urethral sphincter deficiency. Biomaterials. 2014;35(5):1519–30.

    Article  CAS  PubMed  Google Scholar 

  169. De Filippo RE, Yoo JJ, Atala A. Urethral replacement using cell seeded tubularized collagen matrices. J Urol. 2002;168(4 Pt 2):1789–92; discussion 92–3.

    Article  PubMed  Google Scholar 

  170. Orabi H, AbouShwareb T, Zhang Y, Yoo JJ, Atala A. Cell-seeded tubularized scaffolds for reconstruction of long urethral defects: a preclinical study. Eur Urol. 2013;63(3):531–8.

    Article  PubMed  Google Scholar 

  171. Raya-Rivera A, Esquiliano DR, Yoo JJ, Lopez-Bayghen E, Soker S, Atala A. Tissue-engineered autologous urethras for patients who need reconstruction: an observational study. Lancet. 2011;377(9772):1175–82.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Bhargava S, Patterson JM, Inman RD, MacNeil S, Chapple CR. Tissue-engineered buccal mucosa urethroplasty-clinical outcomes. Eur Urol. 2008;53(6):1263–9.

    Article  PubMed  Google Scholar 

  173. Rousseau A, Fradette J, Bernard G, Gauvin R, Laterreur V, Bolduc S. Adipose-derived stromal cells for the reconstruction of a human vesical equivalent. J Tissue Eng Regen Med. 2013;9(11):E135–43.

    Article  PubMed  CAS  Google Scholar 

  174. Frimberger D, Morales N, Shamblott M, Gearhart JD, Gearhart JP, Lakshmanan Y. Human embryoid body-derived stem cells in bladder regeneration using rodent model. Urology. 2005;65(4):827–32.

    Article  PubMed  Google Scholar 

  175. Huang HJ, Zhang JM, Chu HH. Feasibility of constructing tissue-engineered corpus cavernous urethra with rabbit bone marrow mesenchymal stem cells. Zhongguo Zuzhi Gongcheng Yanjiu Yu Linchuang Kangfu. 2007;11:2665–8.

    CAS  Google Scholar 

  176. Wu S, Liu Y, Bharadwaj S, Atala A, Zhang Y. Human urine-derived stem cells seeded in a modified 3D porous small intestinal submucosa scaffold for urethral tissue engineering. Biomaterials. 2011;32(5):1317–26.

    Article  PubMed  CAS  Google Scholar 

  177. Perovic S. Phalloplasty in children and adolescents using the extended pedicle island groin flap. J Urol. 1995;154(2 Pt 2):848–53.

    Article  CAS  PubMed  Google Scholar 

  178. Mulhall JP, Schiff J, Guhring P. An analysis of the natural history of Peyronie’s disease. J Urol. 2006;175(6):2115–8; discussion 8.

    Article  PubMed  Google Scholar 

  179. Young VL, Khouri RK, Lee GW, Nemecek JA. Advances in total phalloplasty and urethroplasty with microvascular free flaps. Clin Plast Surg. 1992;19(4):927–38.

    CAS  PubMed  Google Scholar 

  180. Nukui F, Okamoto S, Nagata M, Kurokawa J, Fukui J. Complications and reimplantation of penile implants. Int J Urol. 1997;4(1):52–4.

    Article  CAS  PubMed  Google Scholar 

  181. Ma L, Yang Y, Sikka SC, Kadowitz PJ, Ignarro LJ, Abdel-Mageed AB, et al. Adipose tissue-derived stem cell-seeded small intestinal submucosa for tunica albuginea grafting and reconstruction. Proc Natl Acad Sci U S A. 2012;109(6):2090–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Feldman HA, Goldstein I, Hatzichristou DG, Krane RJ, McKinlay JB. Impotence and its medical and psychosocial correlates: results of the Massachusetts Male Aging Study. J Urol. 1994;151(1):54–61.

    CAS  PubMed  Google Scholar 

  183. Hatzimouratidis K, Hatzichristou DG. A comparative review of the options for treatment of erectile dysfunction: which treatment for which patient? Drugs. 2005;65(12):1621–50.

    Article  CAS  PubMed  Google Scholar 

  184. Lue TF. Erectile dysfunction. N Engl J Med. 2000;342(24):1802–13.

    Article  CAS  PubMed  Google Scholar 

  185. Qiu X, Fandel TM, Ferretti L, Albersen M, Orabi H, Zhang H, et al. Both immediate and delayed intracavernous injection of autologous adipose-derived stromal vascular fraction enhances recovery of erectile function in a rat model of cavernous nerve injury. Eur Urol. 2012;62(4):720–7.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Orabi H, Lue TF. 1014 Adipose derived stem cells ameliorate diabetic erectile dysfunction in three dimensional culture model of cavernous tissue. J Urol. 2013;189(4), e416.

    Article  Google Scholar 

  187. Garcia MM, Fandel TM, Lin G, Shindel AW, Banie L, Lin CS, et al. Treatment of erectile dysfunction in the obese type 2 diabetic ZDF rat with adipose tissue-derived stem cells. J Sex Med. 2010;7(1 Pt 1):89–98.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Ryu JK, Tumurbaatar M, Jin HR, Kim WJ, Kwon MH, Piao S, et al. Intracavernous delivery of freshly isolated stromal vascular fraction rescues erectile function by enhancing endothelial regeneration in the streptozotocin-induced diabetic mouse. J Sex Med. 2012;9(12):3051–65.

    Article  PubMed  Google Scholar 

  189. Kim IG, Piao S, Lee JY, Hong SH, Hwang TK, Kim SW, et al. Effect of an adipose-derived stem cell and nerve growth factor-incorporated hydrogel on recovery of erectile function in a rat model of cavernous nerve injury. Tissue Eng Part A. 2013;19(1–2):14–23.

    Article  CAS  PubMed  Google Scholar 

  190. Jeong HH, Piao S, Ha JN, Kim IG, Oh SH, Lee JH, et al. Combined therapeutic effect of udenafil and adipose-derived stem cell (ADSC)/brain-derived neurotrophic factor (BDNF)-membrane system in a rat model of cavernous nerve injury. Urology. 2013;81(5):1108.e7–14.

    Article  PubMed  Google Scholar 

  191. Lin G, Albersen M, Harraz AM, Fandel TM, Garcia M, McGrath MH, et al. Cavernous nerve repair with allogenic adipose matrix and autologous adipose-derived stem cells. Urology. 2011;77(6):1509.e1–8.

    Article  PubMed  Google Scholar 

  192. Ying C, Hu W, Cheng B, Yang M, Zheng X, Wang X. Erectile function restoration after repair of resected cavernous nerves by adipose-derived stem cells combined with autologous vein graft in rats. Cell Mol Neurobiol. 2014;34(3):393–402.

    Article  PubMed  Google Scholar 

  193. Lin G, Qiu X, Fandel T, Banie L, Wang G, Lue TF, et al. Tracking intracavernously injected adipose-derived stem cells to bone marrow. Int J Impot Res. 2011;23(6):268–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. You D, Jang MJ, Lee J, Suh N, Jeong IG, Sohn DW, et al. Comparative analysis of periprostatic implantation and intracavernosal injection of human adipose tissue-derived stem cells for erectile function recovery in a rat model of cavernous nerve injury. Prostate. 2013;73(3):278–86.

    Article  CAS  PubMed  Google Scholar 

  195. Orabi H, Lin G, Ferretti L, Lin CS, Lue TF. Scaffoldless tissue engineering of stem cell derived cavernous tissue for treatment of erectile function. J Sex Med. 2012;9(6):1522–34.

    Article  CAS  PubMed  Google Scholar 

  196. Qiu X, Villalta J, Ferretti L, Fandel TM, Albersen M, Lin G, et al. Effects of intravenous injection of adipose-derived stem cells in a rat model of radiation therapy-induced erectile dysfunction. J Sex Med. 2012;9(7):1834–41.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Yokonishi T, Sato T, Komeya M, Katagiri K, Kubota Y, Nakabayashi K, et al. Offspring production with sperm grown in vitro from cryopreserved testis tissues. Nat Commun. 2014;5:4320.

    Article  CAS  PubMed  Google Scholar 

  198. Kim ED, Crosnoe L, Bar-Chama N, Khera M, Lipshultz LI. The treatment of hypogonadism in men of reproductive age. Fertil Steril. 2013;99(3):718–24.

    Article  CAS  PubMed  Google Scholar 

  199. Machluf M, Orsola A, Boorjian S, Kershen R, Atala A. Microencapsulation of Leydig cells: a system for testosterone supplementation. Endocrinology. 2003;144(11):4975–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ingrid Saba .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2017 Springer International Publishing Switzerland

About this chapter

Cite this chapter

Saba, I., Ramsay, S., Bolduc, S., Orabi, H. (2017). Stem Cell Therapy and Tissue Engineering in Urogenital Diseases. In: El-Badri, N. (eds) Advances in Stem Cell Therapy. Stem Cell Biology and Regenerative Medicine. Humana Press, Cham. https://doi.org/10.1007/978-3-319-29149-9_10

Download citation

Publish with us

Policies and ethics