Journal Home Online First Current Issue Archive For Authors Journal Information 中文版

Engineering >> 2022, Volume 19, Issue 12 doi: 10.1016/j.eng.2022.05.004

Leading Approaches to Vascularize Kidney Constructs in Tissue Engineering

Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, NC 27157, USA

Received: 2021-09-06 Revised: 2022-04-14 Accepted: 2022-05-04 Available online: 2022-05-24

Next Previous

Abstract

There is an unprecedented need for new treatments for renal failure, as the incidence of this disease is increasing disproportionately to advancements in therapies. Current treatments are limited by the availability of viable organs, for which there is a worldwide lack. These treatment modalities also require a substantial amount of infrastructure, significantly limiting the access to care in most countries. Kidney tissue engineering approaches promise to develop alternative solutions that address many of the inadequacies in current care. Although many advancements have been made—primarily in the past decade—in biofabrication and whole-organ tissue engineering, many challenges remain. One major hindrance to the progress of current tissue engineering approaches is establishing successful vascularization of developed engineered tissue constructs. This review focuses on the recent advancements that address the vascular challenge, including the biofabrication of vasculature, whole-organ engineering through decellularization and recellularization approaches, microscale organogenesis, and vascularization using organoids in the context of kidney tissue engineering. We also highlight the specific challenges that remain in developing successful strategies capable of clinical translation.

Figures

Fig. 1

Fig. 2

Fig. 3

References

[ 1 ] US Renal Data System. 2017 USRDS annual data report: epidemiology of kidney disease in the United States. Bethesda, MD: National Institute of Health, National Institute of Diabetes and Digestive and Kidney Diseases; 2017.

[ 2 ] US Department of Human and Health Services. OPTN/SRTR 2018 annual data report: kidney. Minneapolis, MN: Chronic Disease Research Group of the Hennepin Healthcare Research Institute; 2018. link1

[ 3 ] Maggiore U, Budde K, Heemann U, Hilbrands L, Oberbauer R, Oniscu GC, et al.; the ERA-EDTA DESCARTES Working Group. Long-term risks of kidney living donation: review and position paper by the ERA-EDTA DESCARTES Working Group. Nephrol Dial Transpl 2017;32(2):216–23. link1

[ 4 ] Matas AJ, Hays RE, Ibrahim HN. Long-term non-end-stage renal disease risks after living kidney donation. Am J Transplant 2017;17(4):893–900. link1

[ 5 ] Matas AJ, Halbert RJ, Barr ML, Helderman JH, Hricik DE, Pirsch JD, et al. Life satisfaction and adverse effects in renal transplant recipients: a longitudinal analysis. Clin Transplant 2002;16(2):113–21. link1

[ 6 ] Yanik EL, Chinnakotla S, Gustafson SK, Snyder JJ, Israni AK, Segev DL, et al. Effects of maintenance immunosuppression with sirolimus after liver transplant for hepatocellular carcinoma. Liver Transplant 2016;22(5):627–34. link1

[ 7 ] Bodro M, Sanclemente G, Lipperheide I, Allali M, Marco F, Bosch J, et al. Impact of antibiotic resistance on the development of recurrent and relapsing symptomatic urinary tract infection in kidney recipients. Am J Transplant 2015;15(4):1021–7. link1

[ 8 ] Sprangers B, Nair V, Launay-Vacher V, Riella LV, Jhaveri KD. Risk factors associated with post-kidney transplant malignancies: an article from the Cancer-Kidney International Network. Clin Kidney J 2018;11(3):315–29. link1

[ 9 ] Luo HL, Chiang PH, Cheng YT, Chen YT. Propensity-matched survival analysis of upper urinary tract urothelial carcinomas between end-stage renal disease with and without kidney transplantation. BioMed Res Int 2019;2019:2979142. link1

[10] Ortiz A. The unaccomplished mission of reducing mortality in patients on kidney replacement therapy. Clin Kidney J 2020;13(6):948–51. link1

[11] Rouwkema J, Khademhosseini A. Vascularization and angiogenesis in tissue engineering: beyond creating static networks. Trends Biotechnol 2016;34 (9):733–45. link1

[12] Moffat DB, Fourman J. A vascular pattern of the rat kidney. 1963. J Am Soc Nephrol 2001;12(3):624–32.

[13] Eremina V, Sood M, Haigh J, Nagy A, Lajoie G, Ferrara N, et al. Glomerularspecific alterations of VEGF-A expression lead to distinct congenital and acquired renal diseases. J Clin Invest 2003;111(5):707–16. link1

[14] Seifu DG, Purnama A, Mequanint K, Mantovani D. Small-diameter vascular tissue engineering. Nat Rev Cardiol 2013;10(7):410–21. link1

[15] Baker SC, Rohman G, Hinley J, Stahlschmidt J, Cameron NR, Southgate J. Cellular integration and vascularisation promoted by a resorbable, particulate-leached, cross-linked poly(e-caprolactone) scaffold. Macromol Biosci 2011;11(5):618–27. link1

[16] Thein-Han W, Xu HH. Prevascularization of a gas-foaming macroporous calcium phosphate cement scaffold via coculture of human umbilical vein endothelial cells and osteoblasts. Tissue Eng Pt A 2013;19(15–16):1675–85. link1

[17] Singh S, Wu BM, Dunn JC. The enhancement of VEGF-mediated angiogenesis by polycaprolactone scaffolds with surface cross-linked heparin. Biomaterials 2011;32(8):2059–69. link1

[18] Simón-Yarza T, Formiga FR, Tamayo E, Pelacho B, Prosper F, Blanco-Prieto MJ. Vascular endothelial growth factor-delivery systems for cardiac repair: an overview. Theranostics 2012;2(6):541–52. link1

[19] Park IS, Kim SH, Kim YH, Kim IH, Kim SH. A collagen/smooth muscle cellincorporated elastic scaffold for tissue-engineered vascular grafts. J Biomater Sci-Polym E 2009;20(11):1645–60. link1

[20] Hu Q, Shen Z, Zhang H, Liu S, Feng R, Feng J, et al. Designed and fabrication of triple-layered vascular scaffold with microchannels. J Biomater Sci-Polym E 2021;32(6):714–34. link1

[21] Zhao L, Li X, Yang L, Sun L, Mu S, Zong H, et al. Evaluation of remodeling and regeneration of electrospun PCL/fibrin vascular grafts in vivo. Mat Sci Eng CMaster 2021;118:111441. link1

[22] Carvalho DJ, Feijão T, Neves MI, da Silva RMP, Barrias C. Directed selfassembly of spheroids into modular vascular beds for engineering large tissue constructs. Biofabrication 2020;13(3):035008. link1

[23] Gauvin R, Ahsan T, Larouche D, Lévesque P, Dubé J, Auger FA, et al. A novel single-step self-assembly approach for the fabrication of tissue-engineered vascular constructs. Tissue Eng Pt A 2010;16(5):1737–47. link1

[24] Datta P, Ayan B, Ozbolat IT. Bioprinting for vascular and vascularized tissue biofabrication. Acta Biomater 2017;51:1–20. link1

[25] Ng HY, Lee KA, Kuo CN, Shen YF. Bioprinting of artificial blood vessels. Int J Bioprinting 2018;4(2):140. link1

[26] Sarker MD, Naghieh S, Sharma NK, Ning L, Chen X. Bioprinting of vascularized tissue scaffolds: influence of biopolymer, cells, growth factors, and gene delivery. J Healthc Eng 2019;2019:9156921. link1

[27] Skardal A, Zhang J, Prestwich GD. Bioprinting vessel-like constructs using hyaluronan hydrogels crosslinked with tetrahedral polyethylene glycol tetracrylates. Biomaterials 2010;31(24):6173–81. link1

[28] Skardal A, Zhang J, McCoard L, Xu X, Oottamasathien S, Prestwich GD. Photocrosslinkable hyaluronan-gelatin hydrogels for two-step bioprinting. Tissue Eng Pt A 2010;16(8):2675–85. link1

[29] Li L, Qin S, Peng J, Chen A, Nie Y, Liu T, et al. Engineering gelatin-based alginate/carbon nanotubes blend bioink for direct 3D printing of vessel constructs. Int J Biol Macromol 2020;145:262–71. link1

[30] Hinton TJ, Jallerat Q, Palchesko RN, Park JH, Grodzicki MS, Shue HJ, et al. Three-dimensional printing of complex biological structures by freeform reversible embedding of suspended hydrogels. Sci Adv 2015;1(9):e1500758. link1

[31] Yu Y, Zhang Y, Martin JA, Ozbolat IT. Evaluation of cell viability and functionality in vessel-like bioprintable cell-laden tubular channels. J Biomech Eng 2013;135(9):091011. link1

[32] Dolati F, Yu Y, Zhang Y, De Jesus AM, Sander EA, Ozbolat IT. In vitro evaluation of carbon-nanotube-reinforced bioprintable vascular conduits. Nanotechnology 2014;25(14):145101. link1

[33] Gao Q, He Y, Fu JZ, Liu A, Ma L. Coaxial nozzle-assisted 3D bioprinting with built-in microchannels for nutrients delivery. Biomaterials 2015;61:203–15. link1

[34] Attalla R, Puersten E, Jain N, Selvaganapathy PR. 3D bioprinting of heterogeneous bi- and tri-layered hollow channels within gel scaffolds using scalable multi-axial microfluidic extrusion nozzle. Biofabrication 2018;11(1):015012. link1

[35] Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol 2014;32(8):773–85. link1

[36] Lee VK, Lanzi AM, Haygan N, Yoo SS, Vincent PA, Dai G. Generation of multiscale vascular network system within 3D hydrogel using 3D bio-printing technology. Cell Mol Bioeng 2014;7(3):460–72. link1

[37] Lee VK, Kim DY, Ngo H, Lee Y, Seo L, Yoo SS, et al. Creating perfused functional vascular channels using 3D bio-printing technology. Biomaterials 2014;35 (28):8092–102. link1

[38] Kesari P, Xu T, Boland T. Layer-by-layer printing of cells and its application to tissue engineering. MRS Online Proc Libr 2004;845:5–11. link1

[39] Cui X, Boland T, D’Lima DD, Lotz MK. Thermal inkjet printing in tissue engineering and regenerative medicine. Recent Pat Drug Deliv Formul 2012;6 (2):149–55. link1

[40] Arai K, Iwanaga S, Toda H, Genci C, Nishiyama Y, Nakamura M. Threedimensional inkjet biofabrication based on designed images. Biofabrication 2011;3(3):034113. link1

[41] Nakamura M, Kobayashi A, Takagi F, Watanabe A, Hiruma Y, Ohuchi K, et al. Biocompatible inkjet printing technique for designed seeding of individual living cells. Tissue Eng 2005;11(11–12):1658–66. link1

[42] Christensen K, Xu C, Chai W, Zhang Z, Fu J, Huang Y. Freeform inkjet printing of cellular structures with bifurcations. Biotechnol Bioeng 2015;112 (5):1047–55. link1

[43] Xu C, Chai W, Huang Y, Markwald RR. Scaffold-free inkjet printing of three-dimensional zigzag cellular tubes. Biotechnol Bioeng 2012;109 (12):3152–60. link1

[44] Jensen PK, Steven K. Influence of intratubular pressure on proximal tubular compliance and capillary diameter in the rat kidney. Pflugers Arch 1979;382 (2):179–87. link1

[45] Tisher CC. Functional anatomy of the kidney. Hosp Prac 1978;13(5):53–65. link1

[46] Kerjaschki D. Dysfunctions of cell biological mechanisms of visceral epithelial cell (podocytes) in glomerular diseases. Kidney Int 1994;45(2):300–13. link1

[47] Salant DJ. The structural biology of glomerular epithelial cells in proteinuric diseases. Curr Opin Nephrol Hypertens 1994;3(6):569–74. link1

[48] Wartiovaara J, Ofverstedt LG, Khoshnoodi J, Zhang J, Mäkelä E, Sandin S, et al. Nephrin strands contribute to a porous slit diaphragm scaffold as revealed by electron tomography. J Clin Invest 2004;114(10):1475–83. link1

[49] Kriz W, Koepsell H. The structural organization of the mouse kidney. Z Anat Entwicklungsgesch 1974;144(2):137–63. link1

[50] Kriz W, Bachmann S. Pre- and post-glomerular arterioles of the kidney. J Cardiovasc Pharmacol 1985;7(Suppl 3):S24–30. link1

[51] Haraldsson B, Nyström J, Deen WM. Properties of the glomerular barrier and mechanisms of proteinuria. Physiol Rev 2008;88(2):451–87. link1

[52] Tröndle K, Rizzo L, Pichler R, Koch F, Itani A, Zengerle R, et al. Scalable fabrication of renal spheroids and nephron-like tubules by bioprinting and controlled self-assembly of epithelial cells. Biofabrication 2021;13 (3):035019. link1

[53] Singh NK, Han W, Nam SA, Kim JW, Kim JY, Kim YK, et al. Three-dimensional cell-printing of advanced renal tubular tissue analogue. Biomaterials 2020;232:119734. link1

[54] Nakayama KH, Batchelder CA, Lee CI, Tarantal AF. Decellularized rhesus monkey kidney as a three-dimensional scaffold for renal tissue engineering. Tissue Eng Pt A 2010;16(7):2207–16. link1

[55] Nakayama KH, Lee CC, Batchelder CA, Tarantal AF. Tissue specificity of decellularized rhesus monkey kidney and lung scaffolds. PLoS ONE 2013;8 (5):e64134. link1

[56] He M, Callanan A. Comparison of methods for whole-organ decellularization in tissue engineering of bioartificial organs. Tissue Eng Part B 2013;19 (3):194–208. link1

[57] Orlando G, Farney AC, Iskandar SS, Mirmalek-Sani SH, Sullivan DC, Moran E, 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):363–70. link1

[58] Crapo PM, Gilbert TW, Badylak SF. An overview of tissue and whole organ decellularization processes. Biomaterials 2011;32(12):3233–43. link1

[59] Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell 2006;126(4):677–89. link1

[60] Engler AJ, Sweeney HL, Discher DE, Schwarzbauer JE. Extracellular matrix elasticity directs stem cell differentiation. J Musculoskelet Neuronal Interact 2007;7(4):335. link1

[61] Damaraju SM, Shen Y, Elele E, Khusid B, Eshghinejad A, Li J, et al. Threedimensional piezoelectric fibrous scaffolds selectively promote mesenchymal stem cell differentiation. Biomaterials 2017;149:51–62. link1

[62] Reilly GC, Engler AJ. Intrinsic extracellular matrix properties regulate stem cell differentiation. J Biomech 2010;43(1):55–62. link1

[63] Pennesi G, Scaglione S, Giannoni P, Quarto R. Regulatory influence of scaffolds on cell behavior: how cells decode biomaterials. Curr Pharm Biotechnol 2011;12(2):151–9. link1

[64] 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. link1

[65] 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. link1

[66] Caralt M, Uzarski JS, Iacob S, Obergfell KP, Berg N, Bijonowski BM, et al. Optimization and critical evaluation of decellularization strategies to develop renal extracellular matrix scaffolds as biological templates for organ engineering and transplantation. Am J Transplant 2015;15 (1):64–75. link1

[67] Lichtenberg A, Cebotari S, Tudorache I, Sturz G, Winterhalter M, Hilfiker A, et al. Flow-dependent re-endothelialization of tissue-engineered heart valves. J Heart Valve Dis 2006;15(2):287–93. Discussion 293–4. link1

[68] Ross EA, Williams MJ, Hamazaki T, Terada N, Clapp WL, Adin C, et al. Embryonic stem cells proliferate and differentiate when seeded into kidney scaffolds. J Am Soc Nephrol 2009;20(11):2338–47. link1

[69] Ross EA, Abrahamson DR, St John P, Clapp WL, Williams MJ, Terada N, et al. Mouse stem cells seeded into decellularized rat kidney scaffolds endothelialize and remodel basement membranes. Organogenesis 2012;8 (2):49–55. link1

[70] Bonandrini B, Figliuzzi M, Papadimou E, Morigi M, Perico N, Casiraghi F, et al. Recellularization of well-preserved acellular kidney scaffold using embryonic stem cells. Tissue Eng Pt A 2014;20(9–10):1486–98. link1

[71] Ko IK, Abolbashari M, Huling J, Kim C, Mirmalek-Sani SH, Moradi M, et al. Enhanced re-endothelialization of acellular kidney scaffolds for whole organ engineering via antibody conjugation of vasculatures. Technology 2014;2 (3):243–53. link1

[72] Baptista PM, Siddiqui MM, Lozier G, Rodriguez SR, Atala A, Soker S. The use of whole organ decellularization for the generation of a vascularized liver organoid. Hepatology 2011;53(2):604–17. link1

[73] Robertson MJ, Dries-Devlin JL, Kren SM, Burchfield JS, Taylor DA. Optimizing recellularization of whole decellularized heart extracellular matrix. PLoS ONE 2014;9(2):e90406. link1

[74] Wang M, Bao L, Qiu X, Yang X, Liu S, Su Y, et al. Immobilization of heparin on decellularized kidney scaffold to construct microenvironment for antithrombosis and inducing reendothelialization. Sci China Life Sci 2018;61(10):1168–77. link1

[75] Huling J, Min SI, Kim DS, Ko IK, Atala A, Yoo JJ. Kidney regeneration with biomimetic vascular scaffolds based on vascular corrosion casts. Acta Biomater 2019;95:328–36. link1

[76] Robert B, St John PL, Abrahamson DR. Direct visualization of renal vascular morphogenesis in Flk1 heterozygous mutant mice. Am J Physiol 1998;275(1): F164–72. link1

[77] Loughna S, Hardman P, Landels E, Jussila L, Alitalo K, Woolf AS. A molecular and genetic analysis of renalglomerular capillary development. Angiogenesis 1997;1(1):84–101. link1

[78] Hyink DP, Abrahamson DR. Origin of the glomerular vasculature in the developing kidney. Semin Nephrol 1995;15(4):300–14. link1

[79] Hyink DP, Tucker DC, St John PL, Leardkamolkarn V, Accavitti MA, Abrass CK, et al. Endogenous origin of glomerular endothelial and mesangial cells in grafts of embryonic kidneys. Am J Physiol 1996;270(5 Pt 2):F886–99. link1

[80] Hyink D. Three-dimensional imaging of fetal mouse kidneys. Methods Mol Biol 2012;886:87–93. link1

[81] Munro DAD, Hohenstein P, Davies JA. Cycles of vascular plexus formation within the nephrogenic zone of the developing mouse kidney. Sci Rep 2017;7 (1):3273. link1

[82] Breier G, Clauss M, Risau W. Coordinate expression of vascular endothelial growth factor receptor-1 (flt-1) and its ligand suggests a paracrine regulation of murine vascular development. Dev Dyn 1995;204(3):228–39. link1

[83] Bartlett CS, Jeansson M, Quaggin SE. Vascular growth factors and glomerular disease. Annu Rev Physiol 2016;78(1):437–61. link1

[84] Sison K, Eremina V, Baelde H, Min W, Hirashima M, Fantus IG, et al. Glomerular structure and function require paracrine, not autocrine, VEGF–VEGFR-2 signaling. J Am Soc Nephrol 2010;21(10):1691–701. link1

[85] Vargas-Valderrama A, Messina A, Mitjavila-Garcia MT, Guenou H. The endothelium, a key actor in organ development and hPSC-derived organoid vascularization. J Biomed Sci 2020;27(1):67. link1

[86] Freedman BS, Brooks CR, Lam AQ, Fu H, Morizane R, Agrawal V, et al. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat Commun 2015;6(1):8715. link1

[87] Low JH, Li P, Chew EGY, Zhou B, Suzuki K, Zhang T, et al. Generation of human PSC-derived kidney organoids with patterned nephron segments and a de novo vascular network. Cell Stem Cell 2019;25(3):373–87.e9. link1

[88] Homan KA, Gupta N, Kroll KT, Kolesky DB, Skylar-Scott M, Miyoshi T, et al. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat Methods 2019;16(3):255–62. link1

[89] Rogers SA, Lowell JA, Hammerman NA, Hammerman MR. Transplantation of developing metanephroi into adult rats. Kidney Int 1998;54(1):27–37. link1

[90] Rogers SA, Hammerman MR. Transplantation of rat metanephroi into mice. Am J Physiol Regul Integr Comp Physiol 2001;280(6):R1865–9. link1

[91] Rogers SA, Liapis H, Hammerman MR. Transplantation of metanephroi across the major histocompatibility complex in rats. Am J Physiol Regul Integr Comp Physiol 2001;280(1):R132–6. link1

[92] Rogers SA, Talcott M, Hammerman MR. Transplantation of pig metanephroi. ASAIO J 2003;49(1):48–52. link1

[93] Dekel B, Amariglio N, Kaminski N, Schwartz A, Goshen E, Arditti FD, et al. Engraftment and differentiation of human metanephroi into functional mature nephrons after transplantation into mice is accompanied by a profile of gene expression similar to normal human kidney development. J Am Soc Nephrol 2002;13(4):977–90. link1

[94] Dekel B, Burakova T, Arditti FD, Reich-Zeliger S, Milstein O, Aviel-Ronen S, et al. Human and porcine early kidney precursors as a new source for transplantation. Nat Med 2003;9(1):53–60. link1

[95] Imberti B, Corna D, Rizzo P, Xinaris C, Abbate M, Longaretti L, et al. Renal primordia activate kidney regenerative events in a rat model of progressive renal disease. PLoS ONE 2015;10(3):e0120235. link1

[96] Xinaris C, Benedetti V, Rizzo P, Abbate M, Corna D, Azzollini N, et al. In vivo maturation of functional renal organoids formed from embryonic cell suspensions. J Am Soc Nephrol 2012;23(11):1857–68. link1

[97] Takebe T, Enomura M, Yoshizawa E, Kimura M, Koike H, Ueno Y, et al. Vascularized and complex organ buds from diverse tissues via mesenchymal cell-driven condensation. Cell Stem Cell 2015;16(5):556–65. link1

[98] Takasato M, Er PX, Chiu HS, Little MH. Generation of kidney organoids from human pluripotent stem cells. Nat Protoc 2016;11(9):1681–92. link1

[99] Bantounas I, Ranjzad P, Tengku F, Silajdzˇic´ E, Forster D, Asselin MC, et al. Generation of functioning nephrons by implanting human pluripotent stem cell-derived kidney progenitors. Stem Cell Rep 2018;10(3):766–79. link1

[100] Taguchi A, Nishinakamura R. Higher-order kidney organogenesis from pluripotent stem cells. Cell Stem Cell 2017;21(6):730–46.e6. link1

[101] Sharmin S, Taguchi A, Kaku Y, Yoshimura Y, Ohmori T, Sakuma T, et al. Human induced pluripotent stem cell-derived podocytes mature into vascularized glomeruli upon experimental transplantation. J Am Soc Nephrol 2016;27(6):1778–91. link1

[102] Van den Berg CW, Ritsma L, Avramut MC, Wiersma LE, van den Berg BM, Leuning DG, et al. Renal subcapsular transplantation of PSC-derived kidney organoids induces neo-vasculogenesis and significant glomerular and tubular maturation in vivo. Stem Cell Rep 2018;10(3):751–65. link1

[103] Francipane MG, Han B, Oxburgh L, Sims-Lucas S, Li Z, Lagasse E. Kidney-in-alymph node: a novel organogenesis assay to model human renal development and test nephron progenitor cell fates. J Tissue Eng Regen Med 2019;13(9):1724–31. link1

[104] Komori J, Boone L, deWard A, Hoppo T, Lagasse E. The mouse lymph node as an ectopic transplantation site for multiple tissues. Nat Biotechnol 2012;30 (10):976–83. link1

[105] Francipane MG, Lagasse E. Maturation of embryonic tissues in a lymph node: a new approach for bioengineering complex organs. Organogenesis 2014;10 (3):323–31. link1

[106] Francipane MG, Lagasse E. The lymph node as a new site for kidney organogenesis. Stem Cells Transl Med 2015;4(3):295–307. link1

[107] Francipane MG, Lagasse E. Regenerating a kidney in a lymph node. Pediatr Nephrol 2016;31(10):1553–60. link1

[108] Rogers SA, Hammerman MR. Transplantation of metanephroi after preservation in vitro. Am J Physiol Regul Integr Comp Physiol 2001;281(2): R661–5. link1

Related Research