We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Immunogenicity analysis of decellularized cardiac scaffolds after transplantation into rats

    Cailing Tong

    School of Life Science, Xiamen University, Fujian, 361102, China

    Organ Transplantation Institute, Medical College, Xiamen University, Fujian, 361102, China

    Key Laboratory of Organ & Tissue Regeneration, Fujian Province, Fujian, 61102, China

    ,
    Junjie Xia

    Organ Transplantation Institute, Medical College, Xiamen University, Fujian, 361102, China

    Key Laboratory of Organ & Tissue Regeneration, Fujian Province, Fujian, 61102, China

    ,
    Baiyi Xie

    Organ Transplantation Institute, Medical College, Xiamen University, Fujian, 361102, China

    Key Laboratory of Organ & Tissue Regeneration, Fujian Province, Fujian, 61102, China

    ,
    Minghui Li

    Organ Transplantation Institute, Medical College, Xiamen University, Fujian, 361102, China

    Key Laboratory of Organ & Tissue Regeneration, Fujian Province, Fujian, 61102, China

    ,
    Feifei Du

    Organ Transplantation Institute, Medical College, Xiamen University, Fujian, 361102, China

    Key Laboratory of Organ & Tissue Regeneration, Fujian Province, Fujian, 61102, China

    ,
    Cheng Li

    Organ Transplantation Institute, Medical College, Xiamen University, Fujian, 361102, China

    Key Laboratory of Organ & Tissue Regeneration, Fujian Province, Fujian, 61102, China

    ,
    Yaguang Li

    Organ Transplantation Institute, Medical College, Xiamen University, Fujian, 361102, China

    Key Laboratory of Organ & Tissue Regeneration, Fujian Province, Fujian, 61102, China

    ,
    Zhonggui Shan

    *Author for correspondence: Tel.: +086 137 7993 1737;

    E-mail Address: szgdoctor@126.com

    Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Fujian, 361003, China

    &
    Zhongquan Qi

    **Author for correspondence: Tel: +086 138 0600 1271;

    E-mail Address: oti@xmu.edu.cn

    Organ Transplantation Institute, Medical College, Xiamen University, Fujian, 361102, China

    Key Laboratory of Organ & Tissue Regeneration, Fujian Province, Fujian, 61102, China

    Published Online:https://doi.org/10.2217/rme-2018-0139

    Aim: Cardiac extracellular matrix (cECM) scaffolds are promising biomaterials for clinical applications. Our aim is to determine the immunogenicity of decellularized scaffolds from different sources for use as artificial organs during organ transplantation. Materials & methods: We transplanted Lewis rats with syngeneic (Lewis rat cECM), allogeneic (BN rat cECM) or xenogeneic (hamster cECM) decellularized cardiac scaffolds. Acute vascular and cellular rejection was quantified by immunohistochemistry and immune cell infiltration. Results: BN rat and hamster hearts were rejected following transplantation. BN and hamster cECMs had similarly low immunogenicity compared with Lewis rat cECMs and did not lead to increased rejection. Conclusion: We found that scaffolds from all sources did not induce vascular or cellular rejection and exhibited low immunogenicity.

    References

    • 1 Alghazali KM, Nima ZA, Hamzah RN et al. Bone-tissue engineering: complex tunable structural and biological responses to injury, drug delivery, and cell-based therapies. Drug Metab. Rev. 47(4), 1–24 (2015).
    • 2 Karunaratne A, Xi L, Bentley L et al. Multiscale alterations in bone matrix quality increased fragility in steroid induced osteoporosis. Bone 84, 15–24 (2016).
    • 3 Santiago-Medina P, Sundaram PA, Diffoot-Carlo N. Titanium oxide: a bioactive factor in osteoblast differentiation. Int. J. Dent. 2015, 357653 (2015).
    • 4 Chapple CR, Osman NI, Mangera A et al. Application of tissue engineering to pelvic organ prolapse and stress urinary incontinence. Low. Urin. Tract Symptoms 7(2), 63–70 (2015).
    • 5 Li Z, Zhang M. Chitosan-alginate as scaffolding material for cartilage tissue engineering. J. Biomed. Mater. Res. Part A 75(2), 485–493 (2005).
    • 6 Glowacki J, Mizuno S. Collagen scaffolds for tissue engineering. Biopolymers 89(5), 338–344 (2008).
    • 7 Hoque ME, Nuge T, Yeow TK, Nordin N, Prasad RGSV. Gelatin based scaffolds for tissue engineering – a review. Polym. Res. J. 9(1), 15–32 (2014).
    • 8 Zhu J, Marchant RE. Design properties of hydrogel tissue-engineering scaffolds. Expert Rev. Med. Devices 8(5), 607–626 (2011).
    • 9 Nayak KK, Gupta P. In vitro biocompatibility study of keratin/agar scaffold for tissue engineering. Int. J. Biol. Macromol. 81, 1–10 (2015).
    • 10 Stupp SI, Ciegler GW. Organoapatites: materials for artificial bone. I. Synthesis and microstructure. J. Biomed. Mater. Res. 26(2), 169–183 (1992).
    • 11 Cahn F KT. Generation of an artificialskin construct containing a non-degradable fiber mesh: a potential transcutaneous interface. Biomed Mater. 3(3), 1–17 (2008).
    • 12 Quint C, Kondo Y, Manson RJ et al. Decellularized tissue-engineered blood vessel as an arterial conduit. Proc. Natl Acad. Sci. USA 108(22), 9214–9219 (2011).
    • 13 Schaner PJ, Martin ND, Tulenko TN et al. Decellularized vein as a potential scaffold for vascular tissue engineering. J. Vasc. Surg. 40(1), 146–153 (2004).
    • 14 Ling C, Li Q, Brown ME et al. Bioengineered vocal fold mucosa for voice restoration. Sci. Transl. Med. 7(314), 314ra187–314ra187 (2015).
    • 15 Ott HC, Matthiesen TS, Goh S-K et al. Perfusion-decellularized matrix: using nature's platform to engineer a bioartificial heart. Nat. Med. 14(2), 213–221 (2008).
    • 16 Uzarski JS, Xia Y, Belmonte JCI, Wertheim JA. New strategies in kidney regeneration and tissue engineering. Curr. Opin. Nephrol. Hypertens. 23(4), 399–405 (2014).
    • 17 Palakkan AA, Hay DC, Anil Kumar PR, Kumary TV, Ross JA. Liver tissue engineering and cell sources: issues and challenges. Liver Int. 33(5), 666–676 (2013).
    • 18 Faulk DM, Wildemann JD, Badylak SF. Decellularization and cell seeding of whole liver biologic scaffolds composed of extracellular matrix. J. Clin. Exp. Hepatol. 5(1), 69–80 (2015).
    • 19 Atala A. Tissue engineering of human bladder. Br. Med. Bull. 97, 81–104 (2011).
    • 20 Kador KE, Montero RB, Venugopalan P et al. Tissue engineering the retinal ganglion cell nerve fiber layer. Biomaterials 34(17), 4242–4250 (2013).
    • 21 Yu Y, Alkhawaji A, Ding Y, Mei J. Decellularized scaffolds in regenerative medicine. Oncotarget 7(36), 58671–58683 (2016).
    • 22 Rana D, Zreiqat H, Benkirane-Jessel N, Ramakrishna S, Ramalingam M. Development of decellularized scaffolds for stem cell-driven tissue engineering. J. Tissue Eng. Regen. Med. 11(4), 942–965 (2017).
    • 23 Tapias LF, Ott HC. Decellularized scaffolds as a platform for bioengineered organs. Curr. Opin. Organ Transplant. 19(2), 145–152 (2014).
    • 24 Hussein KH, Park K-M, Kang K-S, Woo H-M. Biocompatibility evaluation of tissue-engineered decellularized scaffolds for biomedical application. Mater. Sci. Eng. C 67, 766–778 (2016).
    • 25 Crapo PM, Medberry CJ, Reing JE et al. Biologic scaffolds composed of central nervous system extracellular matrix. Biomaterials 33(13), 3539–3547 (2012).
    • 26 Billingham ME, Cary NR, Hammond ME et al. A working formulation for the standardization of nomenclature in the diagnosis of heart and lung rejection: Heart Rejection Study Group. The International Society for Heart Transplantation. J. Heart Transplant. 9(6), 587–593 (2017).
    • 27 Go AS, Mozaffarian D, Roger VL et al. Heart disease and stroke statistics – 2014 update: a report from the American Heart Association. Circulation 129(3), e28 (2014).
    • 28 Moroni F, Mirabella T. Decellularized matrices for cardiovascular tissue engineering. Am. J. Stem Cells 3(1), 1–20 (2014).
    • 29 Weymann A, Patil NP, Sabashnikov A et al. Bioartificial heart: a human-sized porcine model – the way ahead. PLoS ONE 9(11), e111591 (2014).
    • 30 Boeer U, Buettner FFR, Klingenberg M et al. Immunogenicity of intensively decellularized equine carotid arteries is conferred by the extracellular matrix protein collagen type VI. PLoS ONE 9(8), e105964 (2014).
    • 31 Ayala García MA, González Yebra B, López Flores AL, Guaní Guerra E. The major histocompatibility complex in transplantation. J. Transplant. 2012, 1–7 (2012).
    • 32 Hricik DE. Transplant immunology and immunosuppression: core curriculum 2015. Core Curric. Nephrol. 65(6), 956–966 (2015).
    • 33 Moreau A, Varey E, Anegon I, Cuturi M-C. Effector mechanisms of rejection. Cold Spring Harb. Perspect. Med. 3(11), 1–33 (2013).
    • 34 Hancock WW, Gao W, Shemmeri N et al. Immunopathogenesis of accelerated allograft rejection in sensitized recipients: humoral and nonhumoral mechanisms. Transplantation 73(9), 1392–1397 (2002).
    • 35 Sánchez-Fueyo A, Strom TB. Immunologic basis of graft rejection and tolerance following transplantation of liver or other solid organs. Gastroenterology 140(1), 51–64 (2011).
    • 36 Michaels PJ, Fishbein MC, Colvin RB. Humoral rejection of human organ transplants. Springer Semin. Immunopathol. 25(2), 119–140 (2003).
    • 37 Ingulli E. Mechanism of cellular rejection in transplantation. Pediatr. Nephrol. 25(1), 61–74 (2010).
    • 38 Zhao Y, Zhang S, Zhou J et al. Biomaterials The development of a tissue-engineered artery using decellularized scaffold and autologous ovine mesenchymal stem cells. Biomaterials 31(2), 296–307 (2010).
    • 39 Jiao Z-X, Leng Y, Xia J-J et al. As2 O3 combined with leflunomide prolongs heart xenograft survival via suppressing the response of Th1, Th2, and B cells in a rat model. Xenotransplantation 23(3), 237–248 (2016).
    • 40 Oberhuber R, Cardini B, Kofler M et al. Murine cervical heart transplantation model using a modified cuff technique. J. Vis. Exp. (92), e50753 (2014).
    • 41 Xiao F, Chong AS, Foster P et al. Leflunomide controls rejection in hamster to rat cardiac xenografts. Transplantation 58(7), 828–834 (1994).
    • 42 Billingham ME, Cary NR, Hammond ME et al. A working formulation for the standardization of nomenclature in the diagnosis of heart and lung rejection: Heart Rejection Study Group. The International Society for Heart Transplantation. J. Heart Transplant. 9(6), 587–593 (1990).
    • 43 Platt JL. Antibodies in transplantation. Discov. Med. 10(51), 125–133 (2010).
    • 44 Lynch RJ, Platt JL. Accommodation in organ transplantation. Curr. Opin. Organ Transplant. 13(2), 165–170 (2008).
    • 45 Chang AT, Platt JL. The role of antibodies in transplantation. Transplant. Rev. (Orlando). 23(4), 191–198 (2009).
    • 46 LaRosa DF, Rahman AH, Turka LA. The innate immune system in allograft rejection and tolerance. J. Immunol. 178(12), 7503–7509 (2007).
    • 47 Lakkis FG, Lechler RI. Origin and biology of the allogeneic response. Cold Spring Harb. Perspect. Med. 3(8), a014993 (2013).
    • 48 Dijke EI, Platt JL, Blair P et al. B cells in transplantation. J. Heart Lung Transplant. 35(6), 704–710 (2016).
    • 49 Kloc M, Ghobrial RM. Chronic allograft rejection: a significant hurdle to transplant success. Burn. Trauma. 2(1), 3–10 (2014).
    • 50 Badylak SF, Gilbert TW. Immune response to biologic scaffold materials. Semin. Immunol. 20(2), 109–116 (2008).
    • 51 McKeown DW, Bonser RS, Kellum JA. Management of the heartbeating brain-dead organ donor. Br. J. Anaesth. 108, i96–i107 (2012).
    • 52 Linden PK. History of solid organ transplantation and organ donation. Crit. Care Clin. 25(1), 165–184 (2009).
    • 53 Saidi RF, Hejazii Kenari SK. Challenges of organ shortage for transplantation: solutions and opportunities. Int. J. Organ Transplant. Med. 5(3), 87–96 (2014).
    • 54 Beyar R. Challenges in organ transplantation. Rambam Maimonides Med. J. 2(2), e0049 (2011).
    • 55 Calne R. Clinical transplantation: current problems, possible solutions. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 360(1461), 1797–1801 (2005).
    • 56 Murphy PG, Smith M. Towards a framework for organ donation in the UK. Br. J. Anaesth. 108(Suppl. 1), i56–i67 (2012).
    • 57 Rudge C, Matesanz R, Delmonico FL, Chapman J. International practices of organ donation. Br. J. Anaesth. 108, i48–i55 (2012).
    • 58 Tam PPL, Rossant J, Skarnes WC, Beddington RS. Mouse embryonic chimeras: tools for studying mammalian development. Development 130(25), 6155–6163 (2003).
    • 59 Isotani A, Hatayama H, Kaseda K, Ikawa M, Okabe M. Formation of a thymus from rat ES cells in xenogeneic nude mouse – rat ES chimeras. Genes Cells 16(4), 397–405 (2011).
    • 60 Eckardt S, McLaughlin KJ, Willenbring H. Mouse chimeras as a system to investigate development, cell and tissue function, disease mechanisms and organ regeneration. Cell Cycle 10(13), 2091–2099 (2011).
    • 61 Conner DA. Chimeric mouse production by microinjection. In: Current Protocols in Molecular Biology. John Wiley & Sons, Inc., NJ, USA, 23.7.1–23.7.20 (2001).
    • 62 Gardner RL, Johnson MH. Investigation of early mammalian development using interspecific chimaeras between rat and mouse. Nat. New Biol. 246(151), 86–89 (1973).
    • 63 Wu J, Platero-Luengo A, Sakurai M et al. Interspecies chimerism with mammalian pluripotent stem cells. Cell 168(3), 473–486.e15 (2017).
    • 64 Mueller S, Prelle K, Rieger N et al. Chimeric pigs following blastocyst injection of transgenic porcine primordial germ cells. Mol. Reprod. Dev. 54(3), 244–254 (1999).
    • 65 Akimkin V, Beer M, Blome S et al. New chimeric porcine coronavirus in swine feces, Germany, 2012. Emerg. Infect. Dis. 22(7), 1314–1315 (2016).
    • 66 Behringer RR. Human-animal chimeras in biomedical research. Cell Stem Cell. 1(3), 259–262 (2007).
    • 67 Aasen T, Raya A, Barrero MJ et al. Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nat. Biotechnol. 26(11), 1276–1284 (2008).
    • 68 Cong L, Ran FA, Cox D et al. Multiplex genome engineering using CRISPR/Cas systems. Science (80-.). 339(6121), 819–823 (2013).
    • 69 Cyranoski D, Ledford H. International outcry over genome-edited baby claim. Nature 563, 607–608 (2018).
    • 70 Hyun I, Taylor P, Testa G et al. Ethical standards for human-to-animal chimera experiments in stem cell research. Cell Stem Cell. 1(2), 159–163 (2007).
    • 71 van Sandwijk MS, Bemelman FJ, Ten Berge IJM. Immunosuppressive drugs after solid organ transplantation. Neth. J. Med. 71(6), 281–289 (2013).
    • 72 van Gelder T, van Schaik RH, Hesselink DA. Pharmacogenetics and immunosuppressive drugs in solid organ transplantation. Nat. Rev. Nephrol. 10(12), 725–731 (2014).
    • 73 Starzl TE, Makowka L. Organ transplantation – then and now. Hosp. Physician 23(8), 28–33, 36 (1987).
    • 74 Griesemer A, Yamada K, Sykes M. Xenotransplantation: immunological hurdles and progress toward tolerance. Immunol. Rev. 258(1), 241–258 (2014).
    • 75 Nie X, Wang D-A. Decellularized orthopaedic tissue-engineered grafts: biomaterial scaffolds synthesised by therapeutic cells. Biomater. Sci. 6(11), 2798–2811 (2018).
    • 76 Stoppel WL, Hu D, Domian IJ, Kaplan DL, Black LD. Anisotropic silk biomaterials containing cardiac extracellular matrix for cardiac tissue engineering. Biomed. Mater. 10(3), 034105 (2015).
    • 77 Chen W, Konkel JE. TGF-beta and ‘adaptive’ Foxp3(+) regulatory T cells. J. Mol. Cell Biol. 2(1), 30–36 (2010).