J Korean Med Sci.  2008 Jun;23(3):514-520. 10.3346/jkms.2008.23.3.514.

Mechanism of Humoral and Cellular Immune Modulation Provided by Porcine Sertoli Cells

Affiliations
  • 1Xenotransplantation Research Center, Seoul National University, Seoul, Korea.
  • 2Department of Thoracic and Cardiovascular Surgery, Seoul National University, Seoul, Korea.
  • 3Laboratory of Immunology, Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea. jrl@plaza.snu.ac.kr
  • 4Transplantation Research Institute, Medical Research Center, Seoul National University, Seoul, Korea.
  • 5Department of Thoracic and Cardiovascular Surgery, Sejong General Hospital, Sejong Heart Institute, Bucheon, Korea.
  • 6Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.

Abstract

The understanding of main mechanisms that determine the ability of immune privilege related to Sertoli cells (SCs) will provide clues for promoting a local tolerogenic environment. In this study, we evaluated the property of humoral and cellular immune response modulation provided by porcine SCs. Porcine SCs were resistant to human antibody and complement-mediated formation of the membrane attack complex (38.41+/-2.77% vs. 55.02+/-5.44%, p=0.027) and cell lysis (42.95+/-1.75% vs. 87.99 +/-2.25%, p<0.001) compared to immortalized aortic endothelial cells, suggesting that porcine SCs are able to escape cellular lysis associated with complement activation by producing one or more immunoprotective factors that may be capable of inhibiting membrane attack complex formation. On the other hand, porcine SCs and their culture supernatant suppressed the up-regulation of CD40 expression (p<0.05) on DCs in the presence of LPS stimulation. These novel findings, as we know, suggest that immune modulatory effects of porcine SCs in the presence of other antigen can be obtained from the first step of antigen presentation. These might open optimistic perspectives for the use of porcine SCs in tolerance induction eliminating the need for chronic immunosuppressive drugs.

Keyword

Porcine Sertoli Cells; Immune Privilege; Humoral Immune Response; Cellular Immune Response; Tolerance

MeSH Terms

Animals
Antibodies, Heterophile/immunology
Antibody Formation/*immunology
Antigens, CD40/immunology
Aorta/cytology
Cell Line, Transformed
Cell Survival/immunology
Complement Membrane Attack Complex/immunology
Complement System Proteins/immunology
Dendritic Cells/cytology/immunology
Endothelial Cells/cytology/immunology
Epitopes/immunology
Humans
Immune Tolerance/*immunology
Immunity, Cellular/*immunology
Male
Mice
Mice, Inbred C57BL
Sertoli Cells/cytology/*immunology
Swine
*Tissue Engineering
Transplantation, Heterologous

Figure

  • Fig. 1 Expression of Gal-α1,3-Gal (Gal) on immortalized porcine Sertoli (NPSCi) or aortic endothelial (PECi) cells. Cells cultured as a monolayer on chamber slides and fixed for 2 hr with absolute cold ethanol were immunostained for IB4 lectin (brown). Most of NPSCi (A) and PECi (B) cells are positive to the Gal epitope. FACS analysis of non-fixed NPSCi (C) and PECi (D) cells stained with FITC-conjugated IB4 lectin (1:50) for 1 hr to detect Gal (solid line). Also, there are no significant differences in the binding of human IgM to binding of human xenoreactive antibodies to NPSCi (E, 76.95±6.03%) or PECi (F, 80.76±2.44%) cells (p>0.05).

  • Fig. 2 Formation of membrane attack complex (MAC) on the immortalized porcine Sertoli (NPSCi, A, 38.41±2.77%) and aortic endothelial (PECi, B, 55.02±5.44%) cells in the presence of normal human serum. Both cell lines show the significant difference in the formation of MAC (p=0.027). Gray histograms correspond to the control cells that were not pre-incubated with human serum, and solid histograms represent specific staining of human C5b-9. The data are presented as mean±SD of three individual experiments in triplicate.

  • Fig. 3 Susceptibility of immortalized porcine Sertoli (NPSCi) or aortic endothelial (PECi) cells to human natural antibody- and complement-mediated lysis. Negative control values (media alone) for NPSCi and PECi cells were arbitrarily set equal to 100%, and the relative percentage of cell viability was calculated for each condition. The data are presented as the mean±SD of three individual experiments in triplicate. Means bearing different superscript letters are significantly different at p<0.05. M, media alone; M+C, media+rabbit complement; iNHS, heat-inactivated normal human serum; NSH, normal human serum.

  • Fig. 4 The effects of immortalized porcine Sertoli cells on the activation of dendritic cells. Immortalized porcine SCs (NPSCi cells) do not activate DCs (empty bar) but inhibit cell surface expression of CD40 in the presence of LPS. The data are presented as the mean±SD of three individual experiments in triplicate. Means bearing different superscript letters are significantly different at p<0.05. MFI, mean fluorescence index; ciDC, immature DC differentiated in the presence of Dexa and D3; iDC, immature DC; LPS, lipopolysaccharide.

  • Fig. 5 The effects of culture supernatant of immortalized porcine Sertoli cells (iSPNT) on the activation of dendritic cells (DCs) in the presence of LPS. CD40 expression on DCs was significantly inhibited in the presence of iSPNT. But iSPNT did not affect the expression of CD80 and I-Ab molecules on the surface of LPS stimulated DCs. The data are presented as the mean±SD of three individual experiments in triplicate. Means bearing different superscript letters are significantly different at p<0.05. MFI, mean fluorescence index; iDC, immature DC; LPS, lipopolysaccharide.


Reference

1. Dufour JM, Rajotte RV, Seeberger K, Kin T, Korbutt GS. Long-term survival of neonatal porcine Sertoli cells in non-immunosuppressed rats. Xenotransplantation. 2003. 10:577–586.
Article
2. Lee HM, Oh BC, Lim DP, Lee DS, Cho J, Lee G, Lee JR. Establishment and characterization of porcine Sertoli cell line for the study of xenotransplantation. Xenotransplantation. 2007. 14:112–118.
Article
3. Lee HM, Oh BC, Lim DP, Lee DS, Cho J, Lee G, Lee JR. Role of complement regulatory proteins in the survival of murine allo-transplanted Sertoli cells. J Korean Med Sci. 2007. 22:277–282.
Article
4. Bailey R, Griswold MD. Clusterin in the male reproductive system: localization and possible function. Mol Cell Endocrinol. 1999. 151:17–23.
Article
5. van den Berg CW, Morgan BP. Complement-inhibiting activities of human CD59 and analogues from rat, sheep, and pig are not homologously restricted. J Immunol. 1994. 152:4095–4101.
6. van den Berg CW, Perez de la Lastra JM, Llanes D, Morgan BP. Purification and characterization of the pig analogue of human membrane cofactor protein (CD46/MCP). J Immunol. 1997. 158:1703–1709.
7. Suarez-Pinzon W, Korbutt GS, Power R, Hooton J, Rajotte RV, Rabinovitch A. Testicular sertoli cells protect islet beta-cells from autoimmune destruction in NOD mice by a transforming growth factor-beta1-dependent mechanism. Diabetes. 2000. 49:1810–1818.
Article
8. Bellgrau D, Gold D, Selawry H, Moore J, Franzusoff A, Duke RC. A role for CD95 ligand in preventing graft rejection. Nature. 1995. 377:630–632.
Article
9. Yang H, Wright JR Jr. Co-encapsulation of Sertoli enriched testicular cell fractions further prolongs fish-to-mouse islet xenograft survival. Transplantation. 1999. 67:815–820.
10. Halberstadt C, Emerich DF, Gores P. Use of Sertoli cell transplants to provide local immunoprotection for tissue grafts. Expert Opin Biol Ther. 2004. 4:813–825.
Article
11. Reis e Sousa C. Activation of dendritic cells: translating innate into adaptive immunity. Curr Opin Immunol. 2004. 16:21–25.
Article
12. Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines. Cell. 2001. 106:255–258.
13. Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol. 2003. 21:685–711.
14. Morelli AE, Thomson AW. Dendritic cells: regulators of alloimmunity and opportunities for tolerance induction. Immunol Rev. 2003. 196:125–146.
Article
15. Dufour JM, Hamilton M, Rajotte RV, Korbutt GS. Neonatal porcine Sertoli cells inhibit human natural antibody-mediated lysis. Biol Reprod. 2005. 72:1224–1231.
16. Sadeghi K, Wessner B, Laggner U, Ploder M, Tamandl D, Friedl J, Zugel U, Steinmeyer A, Pollak A, Roth E, Boltz-Nitulescu G, Spittler A. Vitamin D3 down-regulates monocyte TLR expression and triggers hyporesponsiveness to pathogen-associated molecular patterns. Eur J Immunol. 2006. 36:361–370.
17. Zhu M, Wang SS, Xia ZX, Cao RH, Chen D, Huang YB, Liu B, Chen ZK, Chen S. Inhibition of xenogeneic response in porcine endothelium using RNA interference. Transplantation. 2005. 79:289–296.
Article
18. Jenne DE, Tschopp J. Molecular structure and functional characterization of a human complement cytolysis inhibitor found in blood and seminal plasma: identity to sulfated glycoprotein 2, a constituent of rat testis fluid. Proc Natl Acad Sci USA. 1989. 86:7123–7127.
Article
19. Kirszbaum L, Sharpe JA, Murphy B, d'Apice AJ, Classon B, Hudson P, Walker ID. Molecular cloning and characterization of the novel, human complement-associated protein, SP-4040: a link between the complement and reproductive systems. EMBO J. 1989. 8:711–718.
Article
20. Bozas SE, Kirszbaum L, Sparrow RL, Walker ID. Several vascular complement inhibitors are present on human sperm. Biol Reprod. 1993. 48:503–511.
21. Hinchliffe SJ, Rushmere NK, Hanna SM, Morgan BP. Molecular cloning and functional characterization of the pig analogue of CD59: relevance to xenotransplantation. J Immunol. 1998. 160:3924–3932.
22. Maher SE, Pflugh DL, Larsen NJ, Rothschild MF, Bothwell AL. Structure/function characterization of porcine CD59: expression, chromosomal mapping, complement-inhibition, and costimulatory activity. Transplantation. 1998. 66:1094–1100.
23. Perez de la Lastra JM, Harris CL, Hinchliffe SJ, Holt DS, Rushmere NK, Morgan BP. Pigs express multiple forms of decay-accelerating factor (CD55), all of which contain only three short consensus repeats. J Immunol. 2000. 165:2563–2573.
Article
24. Thompson CB. Distinct roles for the costimulatory ligands B7-1 and B7-2 in T helper cell differentiation? Cell. 1995. 81:979–982.
Article
25. Hanninen A, Martinez NR, Davey GM, Heath WR, Harrison LC. Transient blockade of CD40 ligand dissociates pathogenic from protective mucosal immunity. J Clin Invest. 2002. 109:261–267.
26. Martin E, O'Sullivan B, Low P, Thomas R. Antigen-specific suppression of a primed immune response by dendritic cells mediated by regulatory T cells secreting interleukin-10. Immunity. 2003. 18:155–167.
Article
27. Frasca L, Scotta C, Lombardi G, Piccolella E. Both maturation and survival of human dendritic cells are impaired in the presence of anergic/suppressor T cells. Clin Dev Immunol. 2003. 10:61–65.
Article
28. Brown RD, Pope B, Murray A, Esdale W, Sze DM, Gibson J, Ho PJ, Hart D, Joshua D. Dendritic cells from patients with myeloma are numerically normal but functionally defective as they fail to up-regulate CD80 (B7-1) expression after huCD40LT stimulation because of inhibition by transforming growth factor-beta1 and interleukin-10. Blood. 2001. 98:2992–2998.
29. Hegde S, Pahne J, Smola-Hess S. Novel immunosuppressive properties of interleukin-6 in dendritic cells: inhibition of NF-kappaB binding activity and CCR7 expression. FASEB J. 2004. 18:1439–1441.
Full Text Links
  • JKMS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr