240
Views
12
CrossRef citations to date
0
Altmetric
Reviews

Kidney transplantation, bioengineering and regeneration: an originally immunology-based discipline destined to transition towards ad hoc organ manufacturing and repair

, , , , , , , , , , , , , , , & show all
Pages 169-182 | Received 23 Jan 2015, Accepted 21 Oct 2015, Published online: 04 Dec 2015

References

  • Reference annotations
  • • of interest
  • •• of considerable interest
  • UNOS/OPTN Data. Available from: http://optn.transplant.hrsa.gov/latestData/rptData.asp.
  • Matas AJ, Smith JM, Skeans MA, et al. OPTN/SRTR 2011 annual data report: kidney. Am J Transplant. 2013;13(Suppl 1):11–46.
  • Proposal to Substantially Revise the National Kidney Allocation System (Kidney Transplantation Committee) [Internet; cited 2015 Nov 27]. Available from: http://optn.transplant.hrsa.gov/PublicComment/pubcommentPropSub_311.pdf.
  • 2012 United States Renal Data System Annual Report [Internet]. 2012 Atlas of CKD & ESRD [cited 2015 Nov 27]. Available from: http://www.usrds.org/atlas.aspx, p. 283–294
  • Womer KL, Kaplan B. Recent developments in kidney transplantation–a critical assessment. Am J Transplant. 2009;9:1265–1271.
  • Bohl DL, Brennan DC. BK virus nephropathy and kidney transplantation. Clin J Am Soc Nephrol. 2007;2(Suppl 1):S36–46.
  • Randhawa P, Brennan DC. BK virus infection in transplant recipients: an overview and update. Am J Transplant. 2006;6:2000–2005.
  • Brennan DC, Agha I, Bohl DL, et al. Incidence of BK with tacrolimus versus cyclosporine and impact of preemptive immunosuppression reduction. Am J Transplant. 2005;5:582–594.
  • Solez K, Colvin RB, Racusen L, et al. Banff 07 classification of renal allograft pathology: updates and future directions. Am J Transplant. 2008;8:753–760.
  • Cornell LD, Smith RN, Colvin RB. Kidney transplantation: mechanisms of rejection and acceptance. Annu Rev Pathol. 2008;3:189–220.
  • Colvin RB. Antibody-mediated renal allograft rejection: diagnosis and pathogenesis. J Am Soc Nephrol. 2007;18:1046–1056.
  • Solez K, Colvin RB, Racusen LC, et al. Banff ‘05 meeting report: differential diagnosis of chronic allograft injury and elimination of chronic allograft nephropathy (‘CAN’). Am J Transplant. 2007;7:518–526.
  • Zou Y, Stastny P, Susal C, et al. Antibodies against MICA antigens and kidney-transplant rejection. N Engl J Med. 2007;357:1293–1300.
  • Stratta RJ, Rohr MS, Sundberg AK, et al. Intermediate-term outcomes with expanded criteria deceased donors in kidney transplantation: a spectrum or specter of quality? Ann Surg. 2006;243:594–601. Discussion 601–603.
  • Metzger RA, Delmonico FL, Feng S, et al. Expanded criteria donors for kidney transplantation. Am J Transplant. 2003;3(Suppl 4):114–125.
  • Ojo AO, Hanson JA, Meier-Kriesche H, et al. Survival in recipients of marginal cadaveric donor kidneys compared with other recipients and wait-listed transplant candidates. J Am Soc Nephrol. 2001;12:589–597.
  • Port FK, Bragg-Gresham JL, Metzger RA, et al. Donor characteristics associated with reduced graft survival: an approach to expanding the pool of kidney donors. Transplantation. 2002;74:1281–1286.
  • Pascual J, Zamora J, Pirsch JD. A systematic review of kidney transplantation from expanded criteria donors. Am J Kidney Dis. 2008;52:553–586.
  • Rao PS, Schaubel DE, Guidinger MK, et al. A comprehensive risk quantification score for deceased donor kidneys: the kidney donor risk index. Transplantation. 2009;88:231–236.
  • Abt PL, Fisher CA, Singhal AK. Donation after cardiac death in the US: history and use. J Am Coll Surg. 2006;203:208–225.
  • Howard RJ, Schold JD, Cornell DL. A 10-year analysis of organ donation after cardiac death in the United States. Transplantation. 2005;80:564–568.
  • Perico N, Cattaneo D, Sayegh MH, et al. Delayed graft function in kidney transplantation. Lancet. 2004;364:1814–1827.
  • Farney AC, Hines MH, Al-Geizawi S, et al. Lessons learned from a single center’s experience with 134 donation after cardiac death donor kidney transplants. J Am Coll Surg. 2011;212:440–451. Discussion 451–453.
  • Ojo AO, Wolfe RA, Held PJ, et al. Delayed graft function: risk factors and implications for renal allograft survival. Transplantation. 1997;63:968–974.
  • Singh RP, Farney AC, Rogers J, et al. Kidney transplantation from donation after cardiac death donors: lack of impact of delayed graft function on post-transplant outcomes. Clin Transplant. 2011;25:255–264.
  • Yarlagadda SG, Coca SG, Formica RN Jr, et al. Association between delayed graft function and allograft and patient survival: a systematic review and meta-analysis. Nephrol Dial Transplant. 2009;24:1039–1047.
  • Kayler LK, Mohanka R, Basu A, et al. Single versus dual renal transplantation from donors with significant arteriosclerosis on pre-implant biopsy. Clin Transplant. 2009;23:525–531.
  • Kucirka LM, Ros RL, Subramanian AK, et al. Provider response to a rare but highly publicized transmission of HIV through solid organ transplantation. Arch Surg. 2011;146:41–45.
  • Kucirka LM, Singer AL, Segev DL. High infectious risk donors: what are the risks and when are they too high? Curr Opin Organ Transplant. 2011;16:256–261.
  • Freeman RB, Cohen JT. Transplantation risks and the real world: what does ‘high risk’ really mean? Am J Transplant. 2009;9:23–30.
  • Lonze BE, Dagher NN, Liu M, et al. Outcomes of renal transplants from Centers for Disease Control and Prevention high-risk donors with prospective recipient viral testing: a single-center experience. Arch Surg. 2011;146:1261–1266.
  • Sharma A, Fisher RA, Cotterell AH, et al. En bloc kidney transplantation from pediatric donors: comparable outcomes with living donor kidney transplantation. Transplantation. 2011;92:564–569.
  • Thomusch O, Tittelbach-Helmrich D, Meyer S, et al. Twenty-year graft survival and graft function analysis by a matched pair study between pediatric en bloc kidney and deceased adult donors grafts. Transplantation. 2009;88:920–925.
  • Bhayana S, Kuo YF, Madan P, et al. Pediatric en bloc kidney transplantation to adult recipients: more than suboptimal? Transplantation. 2010;90:248–254.
  • Laurence JM, Sandroussi C, Lam VW, et al. Utilization of small pediatric donor kidneys: a decision analysis. Transplantation. 2011;91:1110–1113.
  • Sureshkumar KK, Patel AA, Arora S, et al. When is it reasonable to split pediatric en bloc kidneys for transplantation into two adults? Transplant Proc. 2010;42:3521–3523.
  • Anil Kumar MS, Khan SM, Jaglan S, et al. Successful transplantation of kidneys from deceased donors with acute renal failure: three-year results. Transplantation. 2006;82:1640–1645.
  • Greenstein SM, Moore N, McDonough P, et al. Excellent outcome using “impaired” standard criteria donors with elevated serum creatinine. Clin Transplant. 2008;22:630–633.
  • Lin NC, Yang AH, King KL, et al. Results of kidney transplantation from high-terminal creatinine donors and the role of time-zero biopsy. Transplant Proc. 2010;42:3382–3386.
  • Montgomery RA, Warren DS, Segev DL, et al. HLA incompatible renal transplantation. Curr Opin Organ Transplant. 2012;17:386–392.
  • Gloor J, Stegall MD. Sensitized renal transplant recipients: current protocols and future directions. Nat Rev Nephrol. 2010;6:297–306.
  • Reinsmoen NL, Lai CH, Vo A, et al. Evolving paradigms for desensitization in managing broadly HLA sensitized transplant candidates. Discov Med. 2012;13:267–273.
  • Montgomery RA, Locke JE, King KE, et al. ABO incompatible renal transplantation: a paradigm ready for broad implementation. Transplantation. 2009;87:1246–1255.
  • Masterson R, Hughes P, Walker RG, et al. ABO incompatible renal transplantation without antibody removal using conventional immunosuppression alone. Am J Transplant. 2014;14:2807–2813.
  • Rees MA, Kopke JE, Pelletier RP, et al. A nonsimultaneous, extended, altruistic-donor chain. N Engl J Med. 2009;360:1096–1101.
  • Segev DL, Veale JL, Berger JC, et al. Transporting live donor kidneys for kidney paired donation: initial national results. Am J Transplant. 2011;11:356–360.
  • Cravedi P, Perna A, Ruggenenti P, et al. Mycophenolate mofetil versus azathioprine in organ transplantation. Am J Transplant. 2009;9:2856–2857.
  • Remuzzi G, Lesti M, Gotti E, et al. Mycophenolate mofetil versus azathioprine for prevention of acute rejection in renal transplantation (MYSS): a randomised trial. Lancet. 2004;364:503–512.
  • Ponticelli C. The pros and cons of mTOR inhibitors in kidney transplantation. Expert Rev Clin Immunol. 2014;10(2):295–305.
  • Peddi VR, Wiseman A, Chavin K, et al. Review of combination therapy with mTOR inhibitors and tacrolimus minimization after transplantation. Transplant Rev. 2013;27:97.
  • Brennan DC, Daller JA, Lake KD, et al. Rabbit antithymocyte globulin versus basiliximab in renal transplantation. N Engl J Med. 2006;355:1967–1977.
  • Shapiro R, Ellis D, Tan HP, et al. Alemtuzumab pre-conditioning with tacrolimus monotherapy in pediatric renal transplantation. Am J Transplant. 2007;7:2736–2738.
  • Farney AC, Doares W, Rogers J, et al. A randomized trial of alemtuzumab versus antithymocyte globulin induction in renal and pancreas transplantation. Transplantation. 2009;88:810–819.
  • Hanaway MJ, Woodle ES, Mulgaonkar S, et al. Alemtuzumab induction in renal transplantation. N Engl J Med. 2011;364(20):1909–1919.

•• The authors showed that biopsy-confirmed acute rejection was less frequent with alemtuzumab than with conventional therapy.

  • Saull HE, Enderby CY, Gonwa TA, et al. Comparison of alemtuzumab vs. antithymocyte globulin induction therapy in primary non-sensitized renal transplant patients treated with rapid steroid withdrawal. Clin Transplant. 2015 Jul;29(7):573–580.
  • Ekberg H, Tedesco-Silva H, Demirbas A, et al. Reduced exposure to calcineurin inhibitors in renal transplantation. N Engl J Med. 2007;357:2562–2575.
  • Schena FP, Pascoe MD, Alberu J, et al. Conversion from calcineurin inhibitors to sirolimus maintenance therapy in renal allograft recipients: 24-month efficacy and safety results from the CONVERT trial. Transplantation. 2009;87:233–242.
  • Srinivas TR, Meier-Kriesche HU. Minimizing immunosuppression, an alternative approach to reducing side effects: objectives and interim result. Clin J Am Soc Nephrol. 2008;3(Suppl 2):S101–16.
  • Rostaing L, Vincenti F, Grinyó J, et al. Long-term belatacept exposure maintains efficacy and safety at 5 years: results from the long-term extension of the BENEFIT study. Am J Transplant. 2013;13(11):2875–2883.

•• This study demonstrated a sustained renal function benefit associated with Belatacept with improvements over analogous cyclosporine therapy.

  • Charpentier B, Medina Pestana JO, Del C Rial M, et al Long-term exposure to belatacept in recipients of extended criteria donor kidneys. Am J Transplant. 2013;12:2884–2891.

•• This paper further underscored the benefits of belatacept therapy over cyclosporine in a cohort of extended criteria transplants.

  • Satyananda V, Shapiro R. Belatacept in kidney transplantation. Current Opin Organ Transplant. 2014;19:573–577.
  • Su VC, Harrison J, Rogers C, et al. Belatacept: a new biologic and its role in kidney transplantation. Ann Pharmacother. 2012;46:57–67.
  • Little MH. Regrow or repair: potential regenerative therapies for the kidney. J Am Soc Nephrol. 2006;17(9):2390–2401.
  • Orlando G, Wood KJ, Soker S, et al. How regenerative medicine may contribute to the achievement of an immunosuppression-free state. Transplantation. 2011;92:36–38.
  • Witzgall R, Brown D, Schwarz C, et al. Localization of proliferating cell nuclear antigen, vimentin, c-Fos and clusterin in the postischemic kidney. Evidence for a heterogenous genetic response among nephron segments and a large pool of mitotically active and dediffferentiated cells. J Clin Invest. 1994;93:2175–2188.
  • Kusaba T, Lalli M, Kramann R, et al. Differentiated kidney epithelial cells repair injured proximal tubule. Proc Natl Acad Sci USA. 2014;111(4):1527–1532.
  • Humphreys BD, Duffield JS, Bonventre JV. Renal stem cells in recovery from acute kidney injury. Minerva Urol Nefrol. 2006;58(4):329–337.
  • Bussolati B, Camussi G. Stem cells in acute kidney injury. Contrib Nephrol. 2007;156:250–258.
  • Eirin A, Lerman LO. Mesenchymal stem cell treatment for chronic renal failure. Stem Cell Res Ther. 2014;5(4):83.
  • Katari R, Peloso A, Orlando G. Tissue engineering and regenerative medicine: Semantic considerations for an evolving paradigm. Front Bioeng Biotechnol. 2014;2:57.
  • Kim D, Dressler GR. Nephrogenic factors promote differentiation of mouse embryonic stem cells into renal epithelial. J Am Soc Nephrol. 2005;16:3527–3534.
  • Narayanan K, Schumacher KM, Tasnim F, et al. Human embryonic stem cells differentiate into functional renal proximal tubular-like cells. Kidney Int. 2013;83(4):593–603.
  • Takasato M, Er PX, Becroft M, et al. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat Cell Biol. 2014;16(1):118–126.

•• In this report, the authors successfully demonstrated the potential of human embryonic stem cells to recapitulate renal function spontaneously in the controlled setting.

  • Rumballe BA, Georgas KM, Combes AN, et al. Nephron formation adopts a novel spatial topology at cessation of nephrogenesis. Dev Biol. 2011;360(1):110–122.
  • Pollock CA. Toward a bioartificial kidney: will embryonic stem cells be the answer? Kidney Int. 2013;83(4):543–545.
  • Denker HW. Potentiality of embryonic stem cells: an ethical problem even with stem cell source. J Med Ethics. 2006;32:665–671.
  • Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–872.
  • Mae S, Shono A, Shiota F, et al. Monitoring and robust induction of nephrogenic intermediate mesoderm from human pluripotent stem cells. Nat Commun. 2013;4:1367.
  • Araoka T, Mae S, Kurose Y, 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.
  • Taguchi A, Kaku Y, Ohmori T, 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.
  • Lee PY, Chien Y, Chiou GY, et al. Induced pluripotent stem cells without c-Myc attenuate acute kidney injury via downregulating the signaling of oxidative stress and inflammation in ischemia-reperfusion rats. Cell Transplantation. 2012;21:2569–2585.
  • Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8:726–736.
  • Burdon TJ, Paul A, Noiseux N, et al. Bone marrow stem cell derived paracrine factors for regenerative medicine: current perspective and therapeutic potential. Bone Marrow Res. 2011;2011:207326.
  • Humphreys BD, Bonventre JV. Mesenchymal stem cells in acute kidney injury. Annu Rev Med. 2008;59:311–325.
  • Lange C, Togel F, Ittrich H, et al. Administered mesenchymal stem cells enhance recovery from ischemia/reperfusion induced acute renal failure in rats. Kidney Int. 2005;4:1613–1617.
  • Morigi M, Introna M, Imberti B, et al. Human bone marrow mesenchymal stem cells accelerate recovery of acute renal injury and prolong survival in mice. Stem Cells. 2008;26:2075–2082.
  • Hayakawa M, et al. Role of bone marrow stem cells in the healing process of mouse experimental glomerulonephritis. Pediatr Res. 2005;58:323–328.
  • Li J, et al. The contribution of bone marrow-derived cells to the development of renal interstitial fibrosis. Stem Cells. 2007;25:697–706.
  • Qian H, et al. Bone marrow-derived mesenchymal stem cells ameliorate rat acute failure by differentiation into renal tubular epithelial-like cells. Int J Mol Med. 2008;22:325–332.
  • Perry J, et al. Type IV collagen induces podocytic features in bone marrow stromal stem cells in vitro. J Am Soc Nephrol. 2006;17:66–76.
  • Herrera MB, Bussolati B, Bruno S, et al. Mesenchymal stem cells contribute to the renal repair of acute tubular epithelial injury. Int J Mol Med. 2004;14:1035–1041.
  • Bruno S, Grange C, Collino F, et al. Microvescicoles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS One. 2012;7:e33115.
  • Zhao JJ, Liu JL, Liu L, et al. Protection of mesenchymal stem cells on acute kidney injury. Mol Med Rep. 2014;9:91–96.
  • Hipp J, Atala A. Sources of stem cells for regenerative medicine. Stem Cell Rev. 2008;4:3–11.
  • De Coppi P, Bartsch G, Siddiqui MM, et al. Isolation of amniotic stem cell line with potential for therapy. Nat Biotechnol. 2007;25:100–106.
  • Perin L, Giuliani S, Jin D, et al. Renal differentiation of amniotic fluid stem cells. Cell Prolif. 2007;40:936–948.
  • Hauser PV, de Fazio R, Bruno S, et al. Stem cells derived from human amniotic fluid contribute to acute kidney injury recovery. Am J Pathol. 2010;177:2011–2021.
  • Rota C, Imberti B, Pozzobon M, et al. Human amniotic fluid stem cell preconditioning improves their regenerative potential. Stem Cells Dev. 2012;21:1911–1923.
  • Sedrakyan S, da Sacco S, Milanesi A, et al. Injection of amniotic fluid stem cells delays progression of renal fibrosis. J Am Soc Nephrol. 2012;23:661–673.
  • Sun D, Bu L, Liu C, et al. Therapeutic effects of human amniotic fluid-derived stem cells on renal interstitial fibrosis in a murine model of unilateral ureteral obstruction. PLoS One. 2013;8:e65042.
  • Orlando G, Wood KJ, De Coppi P, et al. Regenerative medicine as applied to general surgery. Ann Surg. 2012;255(5):867–880.
  • Badylak SF, Weiss DJ, Caplan A, et al. Engineered whole organs and complex tissues. Lancet. 2012;379(9819):943–952.
  • Gilbert TW, Sellaro TL, Badylak SF. Decellularization of tissues and organs. Biomaterials. 2006;27(19):3675–3683.
  • Bissell MJ, Hall HG, Parry G. How does the extracellular matrix direct gene expression? J Theor Biol. 1982;99:31–68.
  • Chen SS, Fitzgerald W, Zimmerberg J, et al. Cell-cell and cell-extracellular matrix interactions regulate embryonic stem cell differentiation. Stem Cells. 2007;25(3):553–561.
  • Ross EA, Williams MJ, Hamazaki T, et al. Embryonic stem cells proliferate and differentiate when seeded into kidney scaffolds. J Am Soc Nephrol. 2009;20(11):2338–2347.
  • Nakayama KH, Lee CC, Batchelder CA, et al. Tissue specificity of decellularized rhesus monkey kidney and lung scaffolds. PLoS One. 2013;8(5):e64134.
  • Park KM, Woo HM. Systemic decellularization for multi-organ scaffolds in rats. Transplant Proc. 2012;44(4):1151–1154.
  • Orlando G, Farney A, Sullivan DC, et al. Production and implantation of renal extracellular matrix scaffolds from porcine kidneys as a platform for renal bioengineering investigations. Ann Surg. 2012;256(2):363–370.
  • Song JJ, Guyette JP, Gilpin SE, et al. Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat Med. 2013;19(5):646–651.

•• The authors were able to bioengineer a functional kidney using the decellularization method and orthotopically implant the graft which eventually produced rudimentary urine.

  • Orlando G, Booth C, Wang Z, et al. Discarded human kidneys as a source of ECM scaffold for kidney regeneration technologies. Biomaterials. 2013;24:5915–5925.

•• Confronting the real shortage of transplantable human kidneys, the authors identified discarded kidneys as a potentially realistic platform for realizable bioengineering applications.

  • Petrosyan A, Orlando G, Peloso A, et al. Understanding the bioactivity of stem cells seeded on ECM produced from discarded human kidneys. CellR4. 2015;3(1):e1401.
  • Peloso A, Petrosyan A, Da Sacco S, et al. Renal ECM scaffolds from discarded kidneys maintain glomerular morphometry and vascular resilience, and retains critical growth factors. Transplantation. 2015;99(9):1807–1816.
  • Salvatori M, Peloso A, Katari R, et al. Semi-xenotransplantation: the regenerative medicine-based approach to immunosuppression-free transplantation and to meet the organ demand. Xenotransplantation. 2015 Jan–Feb;22(1):1–6
  • Orlando G, Soker S, Stratta RJ. Organ bioengineering and regeneration as the new Holy Grail for organ transplantation. Ann Surg. 2013;258(2):221–232.
  • Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci. 2010;123(Pt 24):4195–4200.
  • Yokoo T, Ohashi T, Shen JS, et al. Human mesenchymal stem cells in rodent whole-embryo culture are reprogrammed to contribute to kidney tissues. Proc Natl Acad Sci USA. 2005;102(9):3296–3300.
  • Anglani F, Mezzabotta F, Ceol M, et al. The regenerative potential of the kidney: what can we learn from developmental biology? Stem Cell Rev. 2010;6(4):650–657.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.