1
|
Helder MRK, Stoyles NJ, Tefft BJ, Hennessy RS, Hennessy RRC, Dyer R, Witt T, Simari RD, Lerman A. Xenoantigenicity of porcine decellularized valves. J Cardiothorac Surg 2017; 12:56. [PMID: 28716099 PMCID: PMC5514525 DOI: 10.1186/s13019-017-0621-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/12/2017] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The xenoantigenicity of porcine bioprosthetic valves is implicated as an etiology leading to calcification and subsequent valve failure. Decellularization of porcine valves theoretically could erase the antigenicity of the tissue leading to more durable prosthetic valves, but the effectiveness of decellularization protocols in regard to completely removing antigens has yet to be verified. Our hypothesis was that decellularization would remove the more abundant α-gal antigens but not remove all the non α-gal antigens, which could mount a response. METHODS Porcine aortic valves were decellularized with 1% sodium dodecyl sulfate for 4 days. Decellularized cusps were evaluated for α-gal epitopes by ELISA. To test for non α-gal antigens, valves were implanted into sheep. Serum was obtained from the sheep preoperatively and 1 week, 1 month, and 2 months postoperatively. This serum was utilized for anti-porcine antibody staining and for quantification of anti-pig IgM and IgG antibodies and complement. RESULTS Decellularized porcine cusps had 2.8 ± 2.0% relative α-gal epitope as compared to fresh porcine aortic valve cusps and was not statistically significantly different (p = 0.4) from the human aortic valve cusp which had a 2.0 ± 0.4% relative concentration. Anti-pig IgM and IgG increased postoperatively from baseline levels. Preoperatively anti-pig IgM was 27.7 ± 1.7 μg/mL and it increased to 71.9 ± 12.1 μg/mL average of all time points postoperatively (p = 0.04). Preoperatively anti-pig IgG in sheep serum was 44.9 ± 1.5 μg/mL and it increased to 72.6 ± 6.0 μg/mL average of all time points postoperatively (p = 0.01). There was a statistically significant difference (p = 0.00007) in the serum C1q concentration before valve implantation (2.5 ± 0.2 IU/mL) and at averaged time points after valve implantation (5.3 ± 0.3 IU/mL). CONCLUSIONS Decellularization with 1% sodium dodecyl sulfate does not fully eliminate non α-gal antigens; however, significant reduction in α-gal presence on decellularized cusps was observed. Clinical implications of the non α-gal antigenic response are yet to be determined. As such, evaluation of any novel decellularized xenografts must include rigorous antigen testing prior to human trials.
Collapse
Affiliation(s)
| | - Nicholas J. Stoyles
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905 USA
| | - Brandon J. Tefft
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905 USA
| | - Ryan S. Hennessy
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905 USA
| | - Rebecca R. C. Hennessy
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905 USA
| | - Roy Dyer
- Divisions of Immunochemical Core Lab, Mayo Clinic, Rochester, MN USA
| | - Tyra Witt
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905 USA
| | - Robert D. Simari
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905 USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905 USA
| |
Collapse
|
2
|
Vadori M, Cozzi E. Immunological challenges and therapies in xenotransplantation. Cold Spring Harb Perspect Med 2014; 4:a015578. [PMID: 24616201 DOI: 10.1101/cshperspect.a015578] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Xenotransplantation, or the transplantation of cells, tissues, or organs between different species, was proposed a long time ago as a possible solution to the worldwide shortage of human organs and tissues for transplantation. In this setting, the pig is currently seen as the most likely candidate species. In the last decade, progress in this field has been remarkable and includes a better insight into the immunological mechanisms underlying the rejection process. Several immunological hurdles nonetheless remain, such as the strong antibody-mediated and innate or adaptive cellular immune responses linked to coagulation derangements, precluding indefinite xenograft survival. This article reviews our current understanding of the immunological mechanisms involved in xenograft rejection and the potential strategies that may enable xenotransplantation to become a clinical reality in the not-too-distant future.
Collapse
Affiliation(s)
- Marta Vadori
- CORIT (Consortium for Research in Organ Transplantation), Legnaro, 35020 Padua, Italy
| | | |
Collapse
|
3
|
Abstract
The shortage of human organs for transplantation has focused research on the possibility of transplanting pig organs into humans. Many factors contribute to the failure of a pig organ graft in a primate. A rapid innate immune response (natural anti-pig antibody, complement activation, and an innate cellular response; e.g., neutrophils, monocytes, macrophages, and natural killer cells) is followed by an adaptive immune response, although T-cell infiltration of the graft has rarely been reported. Other factors (e.g., coagulation dysregulation and inflammation) appear to play a significantly greater role than in allotransplantation. The immune responses to a pig xenograft cannot therefore be controlled simply by suppression of T-cell activity. Before xenotransplantation can be introduced successfully into the clinic, the problems of the innate, coagulopathic, and inflammatory responses will have to be overcome, most likely by the transplantation of organs from genetically engineered pigs. Many of the genetic manipulations aimed at protecting against these responses also reduce the adaptive response. The T-cell and elicited antibody responses can be prevented by the biological and/or pharmacologic agents currently available, in particular, by costimulation blockade-based regimens. The exogenous immunosuppressive regimen may be significantly reduced by the presence of a graft from a pig transgenic for a mutant (human) class II transactivator gene, resulting in down-regulation of swine leukocyte antigen class II expression, or from a pig with "local" vascular endothelial cell expression of an immunosuppressive gene (e.g., CTLA4-Ig). The immunomodulatory efficacy of regulatory T cells or mesenchymal stromal cells has been demonstrated in vitro but not yet in vivo.
Collapse
|
4
|
Hara H, Witt W, Crossley T, Long C, Isse K, Fan L, Phelps CJ, Ayares D, Cooper DKC, Dai Y, Starzl TE. Human dominant-negative class II transactivator transgenic pigs - effect on the human anti-pig T-cell immune response and immune status. Immunology 2013; 140:39-46. [PMID: 23566228 DOI: 10.1111/imm.12107] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 03/26/2013] [Accepted: 04/03/2013] [Indexed: 12/13/2022] Open
Abstract
Swine leucocyte antigen (SLA) class II molecules on porcine (p) cells play a crucial role in xenotransplantation as activators of recipient human CD4(+) T cells. A human dominant-negative mutant class II transactivator (CIITA-DN) transgene under a CAG promoter with an endothelium-specific Tie2 enhancer was constructed. CIITA-DN transgenic pigs were produced by nuclear transfer/embryo transfer. CIITA-DN pig cells were evaluated for expression of SLA class II with/without activation, and the human CD4(+) T-cell response to cells from CIITA-DN and wild-type (WT) pigs was compared. Lymphocyte subset numbers and T-cell function in CIITA-DN pigs were compared with those in WT pigs. The expression of SLA class II on antigen-presenting cells from CIITA-DN pigs was significantly reduced (40-50% reduction compared with WT; P < 0·01), and was completely suppressed on aortic endothelial cells (AECs) even after activation (100% suppression; P < 0·01). The human CD4(+) T-cell response to CIITA-DN pAECs was significantly weaker than to WT pAECs (60-80% suppression; P < 0·01). Although there was a significantly lower frequency of CD4(+) cells in the PBMCs from CIITA-DN (20%) than from WT (30%) pigs (P < 0·01), T-cell proliferation was similar, suggesting no significant immunological compromise. Organs and cells from CIITA-DN pigs should be partially protected from the human cellular immune response.
Collapse
Affiliation(s)
- Hidetaka Hara
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW Immunological barriers still preclude clinical xenotransplantation. The protective role of CD4(+)CD25(+)Foxp3(+) T-regulatory cells (Treg) in allotransplantation is well described and, therefore, could represent a promising therapeutical tool for xenotransplantation. This review addresses the latest findings on Treg in xenotransplantation research. RECENT FINDINGS In vivo, costimulation blockade-based strategies including anti-CD154 monoclonal antibodies (mAbs) in combination with rapamycin or anti-LFA-1 mAb prolonged both concordant and discordant islets xenografts survival in a Treg-dependent manner. In vitro, IL-10 secretion was shown to be critical for the suppression of xenogeneic responses mediated by Treg. Moreover, transgenic expression of inducible costimulator-immunoglobulin or PD-L1 on porcine endothelial cells inhibited human T-cell proliferation in vitro and was associated with the induction of Treg and IL-10 secretion. CXCR3 mediated the recruitment of Treg to pig endothelium. Finally, the recruitment of human Treg was enhanced by the immobilization of human CCL17 on pig endothelium. SUMMARY There is increasing evidence for the potential of CD4(+)CD25(+)Foxp3(+) Treg to protect xenografts. Induction of Treg in recipients and/or recruitment of human Treg to pig endothelium may represent novel strategies to prevent cell-mediated rejection in pig-to-human xenotransplantation.
Collapse
|
6
|
Anderson AJ, Haus DL, Hooshmand MJ, Perez H, Sontag CJ, Cummings BJ. Achieving stable human stem cell engraftment and survival in the CNS: is the future of regenerative medicine immunodeficient? Regen Med 2011; 6:367-406. [PMID: 21548741 DOI: 10.2217/rme.11.22] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There is potential for a variety of stem cell populations to mediate repair in the diseased or injured CNS; in some cases, this theoretical possibility has already transitioned to clinical safety testing. However, careful consideration of preclinical animal models is essential to provide an appropriate assessment of stem cell safety and efficacy, as well as the basic biological mechanisms of stem cell action. This article examines the lessons learned from early tissue, organ and hematopoietic grafting, the early assumptions of the stem cell and CNS fields with regard to immunoprivilege, and the history of success in stem cell transplantation into the CNS. Finally, we discuss strategies in the selection of animal models to maximize the predictive validity of preclinical safety and efficacy studies.
Collapse
Affiliation(s)
- Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, 845 Health Science Road, UC Irvine, Irvine, CA 92697-1705, USA.
| | | | | | | | | | | |
Collapse
|
7
|
Tahara H, Ide K, Basnet N, Tanaka Y, Ohdan H. Determination of the precursor frequency and the reaction intensity of xenoreactive human T lymphocytes. Xenotransplantation 2010; 17:188-96. [PMID: 20636539 DOI: 10.1111/j.1399-3089.2010.00575.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND It is acknowledged that the response of human T cells to xenogeneic targets is more potent than that to allogeneic targets. However, it is not clear whether the more vigorous T cell response to xenoantigens than to alloantigens is attributable to a higher frequency or stronger reaction of xenoreactive T cells. METHODS We determined the precursor frequencies (PFs) and stimulation indexes (SIs) of xenoreactive human T cells by performing a mixed lymphocyte reaction (MLR) assay using a carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeling technique. Irradiated porcine or human peripheral blood mononuclear cells (PBMCs)used as stimulator cells--were cultured with CFSE-labeled human PBMCs--used as responder cells. RESULTS The SIs of the xenoreactive CD4(+) T cells were significantly higher than those of the alloreactive CD4(+) T cells, whereas the PFs of the alloreactive and xenoreactive CD4(+) T cell precursors were almost identical, suggesting a stronger reaction by a single xenoreactive CD4(+) T cell. In contrast, the SIs of the xenoreactive CD8(+) T cells did not differ from those of the alloreactive CD4(+) T cells, and the PFs of the allo- and xenoreactive CD8(+) T cell precursors were also identical. Addition of a soluble human CD47-Fc fusion protein in the porcine-to-human MLR assay caused a statistically significant reduction of the SIs of the xenoreactive CD4(+) T cells. Such an alteration was abrogated by further addition of blocking antibodies (Abs) against either human CD47 or signal regulatory protein-alpha in the porcine-to-human MLR assay. Addition of human CD47-Fc after the depletion of non-T cells from the population of human responder PBMCs in this MLR assay did not influence the SIs of the xenoreactive CD4(+) T cells. CONCLUSIONS The more vigorous T cell response to xenoantigens than to alloantigens is possibly attributable to a stronger reaction of xenoreactive T cells; the interspecies incompatibility of CD47 may contribute to such xenoreactive CD4(+) T cell responses via an indirect pathway.
Collapse
Affiliation(s)
- Hiroyuki Tahara
- Department of Surgery, Division of Frontier Medical Science, Programs for Biomedical Research, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | |
Collapse
|
8
|
Ekser B, Cooper DKC. Overcoming the barriers to xenotransplantation: prospects for the future. Expert Rev Clin Immunol 2010; 6:219-30. [PMID: 20402385 PMCID: PMC2857338 DOI: 10.1586/eci.09.81] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cross-species transplantation (xenotransplantation) has immense potential to solve the critical need for organs, tissues and cells for clinical transplantation. The increasing availability of genetically engineered pigs is enabling progress to be made in pig-to-nonhuman primate experimental models. Potent pharmacologic immunosuppressive regimens have largely prevented T-cell rejection and a T-cell-dependent elicited antibody response. However, coagulation dysfunction between the pig and primate is proving to be a major problem, and this can result in life-threatening consumptive coagulopathy. This complication is unlikely to be overcome until pigs expressing a human 'antithrombotic' or 'anticoagulant' gene, such as thrombomodulin, tissue factor pathway inhibitor or CD39, become available. Progress in islet xenotransplantation has been more encouraging, and diabetes has been controlled in nonhuman primates for periods in excess of 6 months, although this has usually been achieved using immunosuppressive protocols that might not be clinically applicable. Further advances are required to overcome the remaining barriers.
Collapse
Affiliation(s)
- Burcin Ekser
- Thomas E Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA, and Department of Surgery and Organ Transplantation, University of Padua, Padua, Italy
| | - David KC Cooper
- Thomas E Starzl Transplantation Institute, University of Pittsburgh Medical Center, Starzl Biomedical Science Tower, W1543, 200 Lothrop Street, Pittsburgh, PA 15261, USA, Tel.: +1 412 383 6961, Fax: +1 412 624 1172,
| |
Collapse
|
9
|
Pierson RN, Dorling A, Ayares D, Rees MA, Seebach JD, Fishman JA, Hering BJ, Cooper DKC. Current status of xenotransplantation and prospects for clinical application. Xenotransplantation 2009; 16:263-80. [PMID: 19796067 PMCID: PMC2866107 DOI: 10.1111/j.1399-3089.2009.00534.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Xenotransplantation is one promising approach to bridge the gap between available human cells, tissues, and organs and the needs of patients with diabetes or end-stage organ failure. Based on recent progress using genetically modified source pigs, improving results with conventional and experimental immunosuppression, and expanded understanding of residual physiologic hurdles, xenotransplantation appears likely to be evaluated in clinical trials in the near future for some select applications. This review offers a comprehensive overview of known mechanisms of xenograft injury, a contemporary assessment of preclinical progress and residual barriers, and our opinions regarding where breakthroughs are likely to occur.
Collapse
Affiliation(s)
- Richard N Pierson
- Division of Cardiac Surgery, Department of Surgery, University of Maryland, Baltimore VAMC, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Suppressive efficacy and proliferative capacity of human regulatory T cells in allogeneic and xenogeneic responses. Transplantation 2008; 86:1452-62. [PMID: 19034017 DOI: 10.1097/tp.0b013e318188acb0] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND An understanding of the mechanisms that suppress the human anti-pig cellular response is key for xenotransplantation. We have compared the ability of human regulatory T cells (Tregs) to suppress xenogeneic and allogeneic responses in vitro. METHODS Human peripheral blood mononuclear cells (PBMC), CD4+ T cells, or CD4+ CD25- T cells were stimulated with irradiated human or wild type (WT) or alpha1,3-galactosyltransferase gene-knockout (GT-KO) pig PBMC in the presence or absence of human CD4+ CD25 high Tregs. In separate experiments, 5- (and 6)-carboxyfluorescein diacetate succinimidyl ester-labeled human CD4+ T cells were stimulated with human or pig PBMC. The expansion and precursor frequencies of allo- and xenoreactive Tregs were assessed by labeling with FoxP3 mAb and flow cytometric analysis. RESULTS The responses of human PBMC, CD4+ T cells, and CD4+ CD25- T cells to pig PBMC were stronger than to human PBMC (P<0.05). Human anti-GT-KO responses were weaker than anti-WT responses (P<0.05). Human CD4+ CD25 high Tregs suppressed proliferation of CD4+ CD25- T cells to both human and pig PBMC stimulator cells with the same efficiency. Alloreactive CD4+ CD25+ FoxP3 high responder T cells proliferated more than their xenoreactive counterparts (P<0.05), although xenoreactive CD4+ CD25+ T cells proliferated more than alloreactive cells (P<0.05). There was no difference in precursor frequency between allo- and xeno-reactive CD4+ CD25+ FoxP3 high cells. CONCLUSIONS Human T-cell responses to pig cells are stronger than to allogeneic cells. The human response to GT-KO PBMC is weaker than to WT PBMC. Although human Tregs can suppress both responses, expansion of CD4+ CD25+ FoxP3 high cells against pig PBMC is weaker than against human PBMC. More human Tregs may be required to suppress the stronger xenogeneic response.
Collapse
|
11
|
Cooper DKC, Dorling A, Pierson RN, Rees M, Seebach J, Yazer M, Ohdan H, Awwad M, Ayares D. Alpha1,3-galactosyltransferase gene-knockout pigs for xenotransplantation: where do we go from here? Transplantation 2007; 84:1-7. [PMID: 17627227 DOI: 10.1097/01.tp.0000260427.75804.f2] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The ability to genetically engineer pigs that no longer express the Galalpha1,3Gal (Gal) oligosaccharide has been a significant step toward the clinical applicability of xenotransplantation. Using a chronic immunosuppressive regimen based on costimulatory blockade, hearts from these pigs have survived from 2 to 6 months in baboons. Graft failure was predominantly from the development of a thrombotic microangiopathy. Potential contributing factors include the presence of preformed anti-nonGal antibodies or the development of low levels of elicited antibodies to nonGal antigens, natural killer (NK) cell or macrophage activity, and inherent coagulation dysregulation between pigs and primates. The breeding of pigs transgenic for an "anticoagulant" gene, such as human tissue factor pathway inhibitor, hirudin, or CD39, or lacking the gene for the prothrombinase, fibrinogen-like protein-2, is anticipated to inhibit the change in the endothelium to a procoagulant state that takes place in the pig organ after transplantation. The identification of the targets for anti-nonGal antibodies and/or human macrophages might allow further genetic modification of the pig, and xenogeneic NK cell recognition and activation may be inhibited by the transgenic expression of human leukocyte antigen molecules and/or by blocking the function of activating NK receptors. The ultimate goal of induction of T-cell tolerance may be possible only if these hurdles in the coagulation system and innate immunity can be overcome.
Collapse
Affiliation(s)
- David K C Cooper
- The Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zhang B, Ge B, Xia X, Liu J, Sun L, Rao E, Zhao Y. Compatibility of porcine and human interleukin 2: implications for xenotransplantation. Xenotransplantation 2007; 13:423-32. [PMID: 16925666 DOI: 10.1111/j.1399-3089.2006.00329.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Xenotransplantation provides a possible solution to the severe shortage of allogeneic organ donors. The pig, which shares many physiological similarities with humans, makes it an optimal species for preclinical experimentation and clinical applications. Interleukin 2 (IL2) is a potent growth factor secreted primarily by T helper lymphocytes and it is vital to the cellular expansion required for a productive immune response and the development and peripheral expansion of CD4+CD25+ regulatory T cells. Therefore, it is essential to understand of the compatibility of IL2 between pigs and humans. METHODS We first compared the cDNA and protein sequences and the crystal structures of human and porcine IL2 and IL2 receptors, respectively. The effect of IL2 to induce T cell proliferation was determined by 3H-thymidine incorporation and cell cycle detection. RESULTS Porcine IL2 induced very limited proliferation of human lymphocytes while it functioned well on porcine lymphocytes. Human IL2 had remarkably reduced effects on porcine lymphocytes whereas it worked well on human lymphocytes. CONCLUSION Our present study showed that the interaction of IL2 and IL2R across species might have defects. Together with the wide physiological functions of IL2, our data indicated that physiological disorders could be caused by the poor function of xenogeneic donor IL2 on host cells in full hematopoietic chimera. Our data suggested an additional potential advantage for the mixed xenogeneic chimeras.
Collapse
Affiliation(s)
- Baojun Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Transplantation Biology Research Division, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Recently, Galalpha1-3Galbeta1-4GlcNAc (Gal) knockout (k/o) pigs have been developed using genetic cloning technologies. This remarkable achievement has generated great enthusiasm in xenotransplantation studies. This review summarizes the current status of nonhuman primate experiments using Gal k/o pig organs. Briefly, when Gal k/o pig organs are transplanted into primates, hyperacute rejection does not occur. Although graft survival has been prolonged up to a few months in some cases, the overall results were not better than those using Gal-positive pig organs with human complement regulatory protein transgenes. Gal k/o pig kidneys rapidly developed rejection which was associated with increased anti-non-Gal antibodies. Although the precise mechanisms of Gal k/o pig organ rejection are not clear, it could result from incomplete deletion of Gal, up-regulation of new antigen (non-Gal antigen) and/or production of non-Gal antibodies. Future work in xenotransplantation should place emphasis on further modification of donors, such as combining human complement regulatory genes with Gal k/o, deleting non-Gal antigens and adding protective/surviving genes or a gene that inhibits coagulation. Induction of donor-specific T- and B-cell tolerance and promotion of accommodation are also warranted.
Collapse
Affiliation(s)
- R Zhong
- Department of Surgery, University of Western Ontario, Multi-Organ Transplant Program, London Health Sciences Centre, Transplantation Group, Robarts Research Institute, London, Ontario, Canada
| |
Collapse
|
14
|
Abstract
Solid organ transplantation is limited by an insufficient number of organs to meet the needs of a growing population of patients with end-organ failure. A second problem is that, after successful transplantation, many organs fail owing in large part to chronic immunologic injury or so called "chronic rejection". In other circumstances, the organ "outlives" the recipient, often because the recipient succumbs to medical conditions related to chronic immunosuppression. This review focuses on two future therapies that could solve these problems, specifically, tolerance induction to permit long-term patient and graft survival and xenotransplantation to provide an unlimited supply of donor organs. The primary focuses are the most promising ongoing transplantation research that could be encountered by clinical surgeons in the near future.
Collapse
Affiliation(s)
- Matthew J Weiss
- Transplantation Biology Research Center and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | | | | |
Collapse
|
15
|
Pierson RN. Primate T-cell responses to porcine antigens: implications for clinical xenotransplantation. Xenotransplantation 2006; 13:14-8. [PMID: 16497208 DOI: 10.1111/j.1399-3089.2005.00268.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Richard N Pierson
- Department of Surgery, University of Maryland Medical System Staff Surgeon, Baltimore VAMC, Baltimore, MD, USA.
| |
Collapse
|
16
|
Abstract
Solid organ transplantation has been, by most measures, a phenomenal success. Nonetheless, the field is plagued by extreme shortages of available organs from a very limited number of donors. One potential solution to this organ availability crisis is the use of animals as organ donors for humans (xenotransplantation). Though the concept remains theoretical, significant advances are being made in the field of genetics and in our understanding of the immunological barriers to xenotransplantation. With these advances also comes increased knowledge about the potential risks of xenotransplants, especially disease transmission. The eventual clinical application of animal-to-human transplants will require a careful, balanced appraisal of these issues.
Collapse
Affiliation(s)
- M F Levy
- Transplantation Services, Baylor University Medical Center, Dallas, Texas 75246, USA
| |
Collapse
|
17
|
|
18
|
Wu G, Pfeiffer S, Schröder C, Zhang T, Nguyen BN, Lea W, Kelishadi S, Atkinson JB, Schuurman HJ, White DJG, Azimzadeh AM, Pierson RN. Local or short-term systemic costimulatory molecule blockade prolongs rat corneal allograft survival. Xenotransplantation 2005; 12:197-208. [PMID: 15807770 DOI: 10.1111/j.1399-3089.2005.00221.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Costimulatory molecule blockade with antibody-based immunosuppressive agents has been shown to prolong the survival of many types of allograft. The effects were evaluated of local costimulatory molecule blockade with different CTLA4-Ig constructs and of systemic, short-term treatment with an anti-CD28 monoclonal antibody on orthotopic corneal allograft survival in the rat. METHODS Adult Fischer-344 rats underwent Wistar-Furth orthotopic corneal grafts. The rats were treated with two different CTLA4-fusion proteins administered intraocularly in the perioperative period, or systemically with anti-CD28 monoclonal antibody JJ319. Corneal graft survival was determined by daily slit-lamp examination. The day of rejection was defined as the first postoperative day on which the iris margin was no longer clearly visible through the corneal graft. RESULTS Local administration of CTLA4-fusion protein with mutated immunoglobulin constant region domains via a single perioperative intraocular injection prolonged corneal graft survival modestly but significantly (P < 0.05), in contrast to a CTLA4-fusion protein with wild-type immunoglobulin domains, which had no effect on graft survival (P > 0.5). Systemic short-term administration of 400 microg total of an anti-CD28 monoclonal antibody also prolonged corneal graft survival significantly (P < 0.05) and was more effective than systemic administration of 2 mg total of CTLA4-fusion protein (P < 0.05). CONCLUSIONS Local administration of CTLA4-fusion protein with mutated (non-functional) immunoglobulin domains or systemic administration of anti-CD28 monoclonal antibody can prolong corneal allograft survival in the rat.
Collapse
Affiliation(s)
- Guosheng Wu
- University of Maryland and Baltimore VAMC, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Report on the State of the Science in Xenotransplantation [Executive Summary]. Biotechnol Law Rep 2005. [DOI: 10.1089/blr.2005.24.382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
20
|
Mirenda V, Golshayan D, Read J, Berton I, Warrens AN, Dorling A, Lechler RI. Achieving permanent survival of islet xenografts by independent manipulation of direct and indirect T-cell responses. Diabetes 2005; 54:1048-55. [PMID: 15793243 DOI: 10.2337/diabetes.54.4.1048] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recent success in pancreatic islet allotransplantation has raised expectations but has equally highlighted the acute shortage of donor tissue. The use of xenogeneic tissue would help to address this shortage; however, strong cellular immunity limits the application of this approach. T-cell responses to xenogeneic tissues involve recognition of intact species-mismatched major histocompatibility complex (MHC) molecules, the direct pathway, and xenogeneic proteins presented as peptides by responder-type MHC molecules, the indirect pathway. In this study, we exploited the species difference to selectively and sequentially inhibit direct and indirect xenoresponses after transplantation of porcine islets into mice. Selective inhibition of the direct response was achieved using porcine CTLA4-Ig, which binds preferentially to pig versus mouse B7 molecules. Selective inhibition of the indirect response was achieved using murine CTLA4-Ig, which binds preferentially to mouse B7 molecules. Administration of porcine CTLA4-Ig alone caused modest prolongation of islet survival. Injection of murine CTLA4-Ig alone had a minimal effect. However, the injection of the porcine fusion protein early and the murine homolog late after grafting led to permanent survival of the porcine islets, in the absence of any other immunosuppression. These results suggest that a similar approach could have clinical utility in porcine islet xenotransplantation.
Collapse
Affiliation(s)
- Vincenzo Mirenda
- Department of Immunology, Division of Medicine, Imperial College London, UK
| | | | | | | | | | | | | |
Collapse
|
21
|
Andres A, Toso C, Morel P, Bosco D, Bucher P, Oberholzer J, Mathe Z, Mai G, Wekerle T, Berney T, Bühler LH. Macrophage depletion prolongs discordant but not concordant islet xenograft survival. Transplantation 2005; 79:543-9. [PMID: 15753843 DOI: 10.1097/01.tp.0000151764.39095.ca] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND T cells and macrophages play a major role in the rejection of xenografted islets. Depending on the phylogenetic disparity, direct or indirect antigen presentation is predominant. The aim of this study was to analyze in vitro the predominance of direct or indirect presentation by depleting antigen-presenting cells in concordant and discordant xenogeneic combinations. In vivo, we analyzed the effect of macrophage depletion on concordant and discordant islet xenograft survival to assess in which combination this strategy can be used as therapeutic tool. MATERIALS AND METHODS In vitro, we performed mouse anti-rat and anti-human mixed lymphocyte reactions (MLR) after depletion of responder or stimulator antigen-presenting cells by magnetic sorting. In vivo, streptozotocin-induced diabetic C57BL/6 mice were treated by gadolinium chloride (GdCl) to deplete macrophages, and rat or human islets were transplanted under the kidney capsule. Islet function was followed by glycemia, and xenografts were analyzed at regular intervals for histology and immunohistochemistry. RESULTS Mouse anti-rat MLR showed a predominant direct antigen presentation pathway, whereas in mouse anti-human MLR, direct and indirect pathways were similarly involved. Survival of rat islets was not modified by GdCl therapy. In contrast, survival of human islets was significantly prolonged in GdCl-treated mice. Macrophage infiltration was decreased in concordant and discordant GdCl-treated xenografts at day 4 compared with controls. At day 15, macrophage, CD4+, and CD8+ cell infiltration was similar in all groups. CONCLUSIONS Our results indicate that direct antigen presentation is dominant in the rejection mechanism of concordant islet xenografts and cannot be influenced by host macrophage depletion. Both direct and indirect antigen presentation are involved in rejection of discordant xenogeneic islets. Macrophage depletion should only be considered as therapeutic tool for discordant islet xenotransplantation.
Collapse
Affiliation(s)
- Axel Andres
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Andres A, Toso C, Morel P, Bosco D, Bucher P, Oberholzer J, Mathe Z, Mai G, Wekerle T, Berney T, Bühler LH. Phylogenetic disparity influences the predominance of direct over indirect pathway of antigen presentation in islet xenotransplantation. Transplant Proc 2005; 37:463-5. [PMID: 15808677 DOI: 10.1016/j.transproceed.2004.11.074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND Cellular immunity plays a major role in rejection of xenografted islets. Depending on the phylogenetical disparity, direct or indirect antigen presentation is predominant. The aim of this study was to analyze in vitro the predominance of direct or indirect presentation, and in vivo the effect of macrophage depletion on concordant and discordant islet xenograft survival. MATERIALS AND METHODS In vitro, we performed mouse antirat and mouse antihuman mixed lymphocyte reactions (MLR) after depletion of responder or stimulator antigen-presenting cells. In vivo, streptozotocin-induced diabetic C57BL/6 mice were treated by gadolinium chloride to deplete macrophages and rat or human islets were transplanted under the kidney capsule. Islet function was followed by glycemia and xenografts were analyzed at regular intervals for histology. RESULTS Mouse antirat MLR showed a predominant direct antigen presentation pathway, whereas in mouse antihuman MLR, direct and indirect pathways were similarly involved. Survival of rat islets was not modified by GdCl therapy. In contrast, survival of human islets was significantly prolonged in GdCl-treated mice. Macrophage infiltration was decreased in concordant and discordant GdCl-treated xenografts at day 4, compared to controls. At day 15, macrophage infiltration was similar in all groups. DISCUSSION Our results indicate that direct antigen presentation is dominant in rejection of concordant islet xenografts and cannot be influenced by host macrophage depletion. Both direct and indirect antigen presentation are involved in rejection of discordant xenogeneic islets. Macrophage depletion or inhibition should be considered as therapeutic tool for discordant islet xenotransplantation.
Collapse
Affiliation(s)
- A Andres
- Surgical Research Unit, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Houser SL, Kuwaki K, Knosalla C, Dor FJMF, Gollackner B, Cheng J, Shimizu A, Schuurman HJ, Cooper DKC. Thrombotic microangiopathy and graft arteriopathy in pig hearts following transplantation into baboons. Xenotransplantation 2004; 11:416-25. [PMID: 15303978 DOI: 10.1111/j.1399-3089.2004.00155.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Acute humoral xenograft rejection (AHXR) is an immunologic barrier in pig-to-baboon organ transplantation (Tx). We report microvascular thrombosis and myocardial necrosis in a series of cardiac xenografts. METHODS Ten baboons underwent heterotopic heart Tx from pigs transgenic for human decay-accelerating factor. Recipients were treated with soluble Gal glycoconjugates and multiple immunosuppressive agents. Grafts were removed when palpable contractions stopped. Stained tissue sections from harvested grafts were analyzed by light and fluorescence microscopy. RESULTS Xenograft survival ranged from 4 to 139 (mean 37, median 27) days. Some histology was typical for AHXR (n = 4; median survival 22 days). Hemorrhage and edema were only focal in the longer-surviving grafts (n = 4, median survival 54 days). All grafts had multiple platelet-rich fibrin thrombi occluding myocardial vessels. Ischemic damage was manifested by contraction band necrosis in four grafts, myocytolysis in eight, coagulative necrosis in nine, and patchy myocyte dropout in all grafts. A notable paucity of interstitial mononuclear cells was observed in all grafts. Marked intimal thickening resembling that of allograft vasculopathy was observed in one graft. Immunofluorescence showed immunoglobulin (Ig)G and/or IgM deposition in five grafts. Multivessel C4d deposition appeared in seven grafts. Significant C3 deposition was absent. CONCLUSIONS Cardiac xenograft survival in the pig-to-baboon model can be significantly prolonged by vigorous immunosuppressive treatment of recipient animals. Additional efforts to block humoral activation of graft endothelial cells and/or to overcome species-specific molecular coagulation pathway incompatibilities may prevent the development of microvascular thrombosis and myocardial infarction. Cardiac xenograft vasculopathy (chronic rejection) can occur with prolonged graft survival.
Collapse
Affiliation(s)
- Stuart L Houser
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
One novel solution to the shortage of human organs available for transplantation envisions 'growing' new organs in situ via xenotransplantation of developing anlagen from animal embryos. We and others have shown that renal progenitor cells (metanephroi) transplanted into animal hosts undergo organogenesis (differentiation and growth), become vascularized by blood vessels of host origin and exhibit excretory function. Metanephroi can be stored for up to 3 days in vitro prior to transplantation with no impairment in growth or function post-implantation. Metanephroi can be transplanted across both concordant (rat to mouse) and highly disparate (pig to rodent) xenogeneic barriers. Here we review studies exploring the potential therapeutic use of embryonic kidney transplantation as a means to achieve renal organogenesis.
Collapse
Affiliation(s)
- Marc R Hammerman
- Departments of Medicine, and Cell Biology and Physiology, Renal Division, Box 8126, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA.
| |
Collapse
|
26
|
Kuwaki K, Knosalla C, Dor FJMF, Gollackner B, Tseng YL, Houser S, Mueller N, Prabharasuth D, Alt A, Moran K, Cheng J, Behdad A, Sachs DH, Fishman JA, Schuurman HJ, Awwad M, Cooper DKC. Suppression of natural and elicited antibodies in pig-to-baboon heart transplantation using a human anti-human CD154 mAb-based regimen. Am J Transplant 2004; 4:363-72. [PMID: 14961988 DOI: 10.1111/j.1600-6143.2004.00353.x] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Natural and elicited antipig antibodies (Abs) lead to acute humoral xenograft rejection (AHXR). Ten baboons underwent heterotopic heart transplantation (Tx) from human decay-accelerating factor (hDAF) pigs. Depletion of anti-Galalpha1, 3Gal (Gal) Abs was achieved by the infusion of a Gal glycoconjugate from day-1. Immunosuppression included induction of antithymocyte globulin, thymic irradiation, and cobra venom factor, and maintenance with a human antihuman CD154 mAb, mycophenolate mofetil, and methylprednisolone; heparin and prophylactic ganciclovir were also administered. Pig heart survival ranged from 4 to 139 (mean 37, median 27) days, with three functioning for >50 days. Graft failure (n = 8) was from classical AHXR [4], thrombotic microangiopathy [3], or intragraft thrombosis [1], with death (n = 2) from pneumonia [1], or possible drug toxicity (with features of thrombotic microangiopathy) [1]. Anti-Gal Abs (in microg/mL) were depleted by Gal glycoconjugate before graft implantation from means of 41.3 to 6.3 (IgM) and 12.4-4.6 (IgG), respectively, and at graft excision were 6.3 and 1.7 microg/mL, respectively. No elicited Abs developed, and no cellular infiltration was seen. The treatment regimen was effective in maintaining low anti-Gal Ab levels and in delaying or preventing AHXR. The combination of costimulatory blockade and heparin with Tx of a Gal-negative pig organ may prolong graft survival further.
Collapse
Affiliation(s)
- Kenji Kuwaki
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Within a relatively short time span, a significant number of barriers to xeno-transplantation have been identified and potential solutions generated; however, the survival rates for pig-to-primate heart transplantation remain modest at best, with the longest functioning heterotopic heart transplant surviving only 99 days and the longest functioning orthotopic heart transplant surviving only 39 days. A great deal of improvement in immunological strategies will be needed to make xeno-transplantation a clinical reality. The most exciting prospect in the near term is the use of organs from homozygous alphaGal knockout pigs. The diversity of the biological pathways involved in the total spectrum of xenograft rejection, however, makes it highly likely that the clinical feasibility of xeno-transplantation will depend on a multipronged approach that incorporates the advantages of genetically eliminating the alphaGal epitope on hyperacute and acute xenograft rejection and the advantages of tolerance induction on cellular and chronic xenograft rejection.
Collapse
Affiliation(s)
- Ruediger Hoerbelt
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | |
Collapse
|
28
|
Rijkelijkhuizen JKRA, Haanstra KG, Wubben J, Töns A, Roos A, van Gijlswijk-Janssen DJ, Ringers J, Bouwman E, Jonker M. T-cell-specific immunosuppression results in more than 53 days survival of porcine islets of langerhans in the monkey. Transplantation 2004; 76:1359-68. [PMID: 14627916 DOI: 10.1097/01.tp.0000085290.60182.6b] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Transplantation of islets of Langerhans can restore insulin production in diabetic patients. Because of the shortage of human donor organs, transplantation of porcine islets may be an alternative solution. The present study was aimed at the characterization of rejection mechanisms of porcine islets transplanted into eight nondiabetic monkeys under the kidney capsule. METHODS Cultured adult pig islets were used, which showed no expression of the galactose(alpha1,3)galactose epitope, major histocompatibility complex class II, or CD45, and no binding of antibodies or complement after exposure to monkey serum. Immunosuppression consisted of cyclophosphamide, cyclosporine A (CsA), and steroids (group 1); or antithymocyte globulin, anti-interleukin-2 receptor antibody, CsA, and steroids (group 2). In three animals of group 2, islets were also transplanted in the portal vein. RESULTS Although all monkeys had preformed anti-pig antibodies, no correlation was found between antibody titers and rejection and no deposition of antibodies or complement was observed in the grafts. Group 1 showed islets up to day 11, followed by T-cell infiltration and rejection at approximately day 14. In group 2, two monkeys showed infiltrates consisting predominantly of T cells starting at approximately day 29, whereas two monkeys showed well-preserved islets without infiltration up to day 53. In the livers of the three monkeys that also received islets intraportally and were resectioned on days 21, 33, and 49, no islets could be detected. CONCLUSIONS This study demonstrates that cultured adult pig islets can survive in the monkey for more than 53 days without signs of rejection under standard immunosuppression.
Collapse
|
29
|
Paris MCJ, Snow M, Cox SL, Shaw JM. Xenotransplantation: a tool for reproductive biology and animal conservation? Theriogenology 2004; 61:277-91. [PMID: 14662128 DOI: 10.1016/s0093-691x(03)00234-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The transplantation of reproductive organs, including ovaries and ovarian tissue, was pioneered over 100 years ago. In the 1960s, ovarian grafting was used as a tool to investigate ovarian function, but with the recent development of more effective cryopreservation protocols for ovarian tissue, germline preservation and propagation have now become realistic goals. This review describes progress in ovarian banking and ovarian tissue transplantation, with emphasis on how fresh and frozen ovarian tissue can be used in assisted reproduction for both humans and animals. This paper focuses most closely on the potential value of xenotransplantation, the transplantation of gonads from one species to another, to conserve rare and endangered species. Specific attention is drawn to the use of xenotransplantation as a strategy for generating viable gametes that can be used to produce live fertile offspring. Other upcoming xenogeneic technologies that may be of potential significance in animal conservation, such as transplantation of whole ovaries or isolated growing follicles, and even male germ cells, are discussed.
Collapse
Affiliation(s)
- Monique C J Paris
- Murdoch Children's Research Institute, Flemington Road, Parkville 3052, Vic, Australia.
| | | | | | | |
Collapse
|
30
|
Sun Y, Langnas AN, Zhao Y. The interaction defect of accessory molecules is responsible for the poor ex vivo response to human antigens of mouse T helper cells. Scand J Immunol 2003; 58:59-66. [PMID: 12828559 DOI: 10.1046/j.1365-3083.2003.01281.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mouse T cells fail to respond to xenogeneic pig and human antigens using the direct antigen-presenting pathway. The poor response by mouse CD8 cells is because of multiple defects in the molecular interactions between mouse CD8 cells and xenogeneic antigen-presenting cells (APCs). Using human CD4/DR3+, mouse CD4-/major histocompatibility complex (MHC) class II - mice, we investigated the defects in molecular interaction responsible for the poor response to xenogeneic antigens by naïve mouse CD4+ cells. Mouse CD4 cells failed to respond to human leucocyte antigen (HLA)-DR3 expressed on mouse APCs but developed a strong response to alloantigens, indicating a defect in the interaction between mouse CD4 and HLA-DR3 molecules. Human CD4+/mouse CD4- mouse T cells respond poorly to human and pig APCs but not allogeneic APCs, indicating that accessory molecular interactions are deficient across highly separated species. Adding mouse interleukin-2 (IL-2) to the mixed lymphocyte reaction system did not improve the poor response to human or pig antigens by mouse or human CD4+/mouse CD4- mouse T cells. Therefore, multiple molecular interactions deficient between mouse CD4 cells and human or pig APCs may lead to the poor response to xenogeneic antigens by naïve mouse CD4 cells.
Collapse
Affiliation(s)
- Y Sun
- Department of Surgery, University of Nebraska Medical Center, The Lied Transplant Center, Omaha, NE 68198, USA
| | | | | |
Collapse
|
31
|
Hara H, Ohdan H, Tokita D, Onoe T, Zhou W, Asahara T. Construction of ectopic xenogeneic bone marrow structure associated with persistent multi-lineage mixed chimerism by engraftment of rat bone marrow plugs into mouse kidney capsules. Xenotransplantation 2003; 10:259-66. [PMID: 12694546 DOI: 10.1034/j.1399-3089.2003.02026.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Poor bone marrow (BM) engraftment in a xenogeneic combination results at least in part from the limited engraftment capacity of BM-derived stromal cells, which support hematopoietic repopulation in a species-specific fashion. We attempted to construct a BM stromal microenvironment by engraftment of BM plug fragments into kidney capsules in a rat-to-mouse combination. BM plugs from F344/N Jcl-rnu/rnu (F344 nu) rats were transplanted into the kidney capsules of C.B-17 scid/scid (C.B-17 scid) mice treated with rabbit anti-asialo-GM1 serum to deplete natural killer (NK) cells and then with 3 Gy of whole body irradiation. As a conventional control, an equivalent amount of F344 nu bone marrow cells (BMCs) was intravenously injected into C.B-17 scid mice treated with a similar conditioning regimen. In both mouse recipients of rat BM plug engraftment in the kidney capsules and recipients of intravenous injection of rat BMC suspension, comparable extents of donor rat class I+ cells were persistently detected in the peripheral blood. However, the differentiation of rat-derived B cells in the mouse recipients of rat BM plugs was more rapid than that in the recipients of rat BMC suspension. In the late phase (10 weeks after BM transplantation), the percentage of rat-derived T cells (CD4+ cells) in the mouse recipients of rat BM plugs was significantly higher than that in the recipients of rat BMC suspension. At this time point, ectopic BM structure consisting of bone, mesenchymal cells, and hematopoietic progenitors was constructed in the kidney capsules of mice that received rat BM plugs. Most of the cells in the ectopic BM were derived from the donor rat. Thus, engraftment of BM plugs into the kidney capsules results in the construction of a donor-derived BM microenvironment, facilitating multilineage mixed xenogeneic chimerism.
Collapse
Affiliation(s)
- Hidetaka Hara
- Department of Surgery, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Bonenfant C, Vallée I, Sun J, Brossay A, Thibault G, Guillaumin JM, Lebranchu Y, Bardos P, Butler JE, Watier H. Analysis of human CD4 T lymphocyte proliferation induced by porcine lymphoblastoid B cell lines. Xenotransplantation 2003; 10:107-19. [PMID: 12588644 DOI: 10.1034/j.1399-3089.2003.01062.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND This study was undertaken to characterize the two porcine lymphoblastoid cell lines L23 and L35, derived from a pig inoculated by the retrovirus Tsukuba-1, and to determine how they induce a strong human lymphocyte proliferation. METHODS Phenotypic characterization was performed by flow cytometry and reverse transcriptase-polymerase chain reaction analyses. Xenogeneic mixed lymphocyte reactions (XMLR) were performed using unfractionated human peripheral blood mononuclear cells (huPBMC) and purified CD4+ T lymphocytes as responding cells, in the presence of blocking antibodies and fusion proteins. RESULTS The immunoglobulin genes were demonstrated to be rearranged in L23 and L35 cell lines, in agreement with the expression of a B cell phenotype. Both induced a similar proliferation of huPBMCs and purified human CD4+ lymphocytes from adult or cord blood (naïve cells). Proliferation of CD4+ T lymphocytes was completely blocked by anti-SLA-DR plus anti-SLA-DQ mAbs, excluding human lymphocyte transformation by porcine viruses. The frequency of proliferative precursors was inconsistent with that induced by a retroviral superantigen but similar to classical direct xenoantigen presentation as observed with other porcine antigen-presenting cells. Extensive analysis of costimulatory signals led to the identification of the CD28 pathway, in agreement with membrane expression of B7 molecules on L23 and L35 cells, and of the CD2 pathway in L35 cells. CONCLUSION These two porcine lymphoblastoid cell lines have been further characterized and clearly identified as belonging to the B cell lineage. By expressing major histocompatibility complex class II antigens and costimulatory molecules, they induce a vigorous proliferative response of human CD4+ lymphocytes through a direct presentation pathway.
Collapse
Affiliation(s)
- Christine Bonenfant
- EA 3249 Cellules Hématoponaïétiques, Hémostase et Greffe and IFR 120 Imagerie et Exploration Fonctionnelles, Université François Rabelais, Tours cedex, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen W, Ford MS, Young KJ, Cybulsky MI, Zhang L. Role of double-negative regulatory T cells in long-term cardiac xenograft survival. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:1846-53. [PMID: 12574350 DOI: 10.4049/jimmunol.170.4.1846] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A novel subset of CD3(+)CD4(-)CD8(-) (double negative; DN) regulatory T cells has recently been shown to induce donor-specific skin allograft acceptance following donor lymphocyte infusion (DLI). In this study, we investigated the effect of DLI on rat to mouse cardiac xenotransplant survival and the ability of DN T cells to regulate xenoreactive T cells. B6 mice were given either DLI from Lewis rats, a short course of depleting anti-CD4 mAb, both DLI and anti-CD4 treatment together, or left untreated. DLI alone did not prolong graft survival when compared with untreated controls. Although anti-CD4-depleting mAb alone significantly prolonged graft survival, grafts were eventually rejected by all recipients. However, the combination of DLI and anti-CD4 treatment induced permanent cardiac xenograft survival. We demonstrate that recipients given both DLI and anti-CD4 treatment had a significant increase in the total number of DN T cells in their spleens when compared with all other treatment groups. Furthermore, DN T cells harvested from the spleens of DLI plus anti-CD4-treated mice could dose-dependently inhibit the proliferation of syngeneic antidonor T cells. Suppression mediated by these DN T cells was specific for antidonor T cells as T cells stimulated by third-party Ags were not suppressed. These results demonstrate for the first time that a combination of pretransplant DLI and anti-CD4-depleting mAb can induce permanent survival of rat to mouse cardiac xenografts and that DN T regulatory cells play an important role in preventing long-term concordant xenograft rejection through the specific suppression of antidonor T cells.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens/metabolism
- Antigens, Surface
- CD3 Complex/biosynthesis
- CD4 Antigens/immunology
- CD4 Antigens/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD8 Antigens/metabolism
- CD8-Positive T-Lymphocytes/immunology
- Cell Division/genetics
- Cell Division/immunology
- Epitopes, T-Lymphocyte/immunology
- Graft Enhancement, Immunologic/methods
- Graft Survival/genetics
- Graft Survival/immunology
- Heart Transplantation/immunology
- Immune Tolerance/genetics
- Immunosuppression Therapy
- Injections, Intraperitoneal
- Interphase/immunology
- Lectins, C-Type
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Lymphocyte Transfusion
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- NK Cell Lectin-Like Receptor Subfamily B
- Proteins/metabolism
- Rats
- Rats, Inbred Lew
- Rats, Inbred WF
- Spleen/cytology
- Spleen/metabolism
- Spleen/transplantation
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Transplantation Conditioning
- Transplantation, Heterologous/immunology
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Laboratory Medicine and Pathobiology, Multi Organ Transplantation Program, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, M5G 2C4 Canada
| | | | | | | | | |
Collapse
|
34
|
Zhu T, Zhang L, Shen SO, Guo H, Xia SS, Chen ZK. Reconstitution with DC-depleted splenocytes failed to reject the well-established hamster heart xenografts in nude mice. Transplant Proc 2003; 35:526. [PMID: 12591515 DOI: 10.1016/s0041-1345(02)03815-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- T Zhu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
To determine whether pig metanephroi grow and differentiate after allotransplantation or xenotransplantation across a highly disparate barrier, we implanted metanephroi from embryonic day 28 (E28) pig embryos into the omentum of unilaterally nephrectomized adult pigs or C57Bl/6J mice (hosts). Some mouse hosts received anti-CD45RB, anti-CD154, and anti-CD11a (costimulatory blockade). E28 pig metanephroi were < 0.2 mm in diameter and contained only metanephric blastema and segments of ureteric bud. Pig metanephroi transplanted into pigs underwent growth and differentiation of nephrons over a 2 week period without the need for costimulatory blockade of hosts. In contrast, pig metanephroi did not grow or differentiate in mice that received no costimulatory blockade. However, by 2 weeks posttransplantation in mice in which costimulation was blocked, metanephroi from E28 pigs had enlarged, become vascularized, and had formed mature tubules and glomeruli. By 3 weeks posttransplantation in mice, metanephroi had grown to the point that they were approximately half the volume of the native mouse kidney. Here we show that growth and development of pig metanephroi occurs posttransplantation across an allogeneic or highly disparate xenogeneic barrier.
Collapse
Affiliation(s)
- Sharon A Rogers
- Department of Medicine, George M. O'Brien Kidney and Urological Disease Center, Renal Division, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | | | | |
Collapse
|
36
|
Hara H, Ohdan H, Mizunuma K, Zhou W, Tanaka Y, Fudaba Y, Tashiro H, Asahara T. The role of mouse stroma in rat haematopoiesis in xenogeneic rat/mouse bone marrow transplantation chimeras. Transplant Proc 2002; 34:2759. [PMID: 12431598 DOI: 10.1016/s0041-1345(02)03399-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- H Hara
- Second Department of Surgery, Hiroshima University Faculty of Medicine, Hiroshima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Xenotransplantation has the potential to deliver an unlimited supply of organs for transplantation. However, this promise has yet to translate into clinical application, despite substantial research efforts in the last decade. Although increasing numbers of studies are being performed in relevant pre-clinical (pig-to-primate) transplantation models, so far these have highlighted the apparent elusiveness of long-term xenograft survival. Humoral rejection remains the main obstacle to success, but control of T cell-mediated rejection will be a problem in the future and there are major concerns about the possible transmission of porcine endogenous retroviruses (PERV) and other infectious agents. This article reviews recent advances in the understanding of acute vascular rejection (AVR), acute T cell-mediated rejection and PERV transmission and highlights some of the strategies that may prove successful in overcoming these problems. Although progress has been slow, the promise of an inexhaustible supply of organs is sufficient reason to continue research in these areas. Assuming the specific problem of AVR can be ameliorated by one of a number of strategies currently under investigation, there are grounds to believe that xenotransplantation will become a clinical reality. Pig xenografts, currently grounded, might eventually fly!
Collapse
Affiliation(s)
- Anthony Dorling
- Department of Immunology, Faculty of Medicine, Imperial College School of Science, Technology and Medicine, Hammersmith Hospital London, UK.
| |
Collapse
|
38
|
Gorczynski RM, Hu J, Chen Z, Kai Y, Lei J. A CD200FC immunoadhesin prolongs rat islet xenograft survival in mice. Transplantation 2002; 73:1948-53. [PMID: 12131694 DOI: 10.1097/00007890-200206270-00018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND A solubilized form of the CD200 molecule, CD200Fc, has been shown to suppress allograft rejection and development of collagen-induced arthritis in mice. We investigated whether the same molecule could prolong survival of rat islet xenografts. METHODS Streptozocin-treated mice, receiving injections with anti-asialo-GM1 antibody, received rat islets ( approximately 400/mouse) under the kidney capsule or injected into the portal vein, along with rapamycin treatment. Thereafter mice received injections of CD200Fc (10 microg/mouse/injection) or control mouse IgG2. Blood glucose was monitored daily. Some mice received additional injections of anti-CD200/-CD200R monoclonal antibodies. RESULTS Portal vein delivery of islets led to more extended resolution of diabetes than did transplantation under the kidney capsule. CD200Fc further prolonged survival in either case, an effect abolished by anti-CD200 or F(ab')2 anti-CD200R mAbs, but not by whole anti-CD200R (anti-CD200R Ig). Spleen cells taken from CD200Fc-treated mice showed polarization to type-2 cytokine production (interleukin-4, interleukin-10) on restimulation with rat splenocytes in culture, in comparison to cells from control mice (type-1 cytokines, interlulin-2, interferon-gamma). CONCLUSION CD200:CD200R interactions are important in regulating rat islet xenograft survival.
Collapse
Affiliation(s)
- R M Gorczynski
- The Toronto Hospital, University Health Network and Department of Surgery, University of Toronto, Toronto, Canada M5G2C4
| | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- D H Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Department of Medicine, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts 02129, USA
| | | | | | | |
Collapse
|
40
|
Schneider MKJ, Strasser M, Gilli UO, Kocher M, Moser R, Seebach JD. Rolling adhesion of human NK cells to porcine endothelial cells mainly relies on CD49d-CD106 interactions. Transplantation 2002; 73:789-96. [PMID: 11907429 DOI: 10.1097/00007890-200203150-00023] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Acute vascular rejection in pig-to-primate xenotransplantation involves recognition and damage of porcine (po) endothelial cells (EC) by human (hu) leukocytes, probably including natural killer (NK) cells. To study such interactions we analyzed rolling and static adhesion of hu NK cells to po EC. METHODS The effects of blocking hu and po adhesion molecules on the adhesion hu NK cells to po EC monolayers was analyzed under shear stress (10 min, 37 degrees C, 0.7 dynes/cm2) or under static conditions (10 min, 37 degrees C). All used cell populations were phenotypically characterized by flow cytometry. RESULTS Blocking of CD106 on po EC or its ligand CD49d on hu NK cells decreased rolling adhesion of both fresh and activated hu NK cells by more than 75%. Masking of CD62L on fresh but not activated hu NK resulted in a 44% decrease in rolling adhesion, in line with the diminished cell surface expression of CD62L upon activation. Antibodies to CD31, CD54, CD62E, and CD62P on EC or CD11a, CD18, and CD162 on NK cells had only minor effects on rolling adhesion. The adhesion of the FcgammaRIII- hu NK cell line NK92 to po EC was inhibited by 95% after masking po CD106 whereas antibodies to po CD31, CD54, CD62E, or CD62P had no effect, thereby excluding effects of Fc-receptor-dependent binding of hu NK cells to po EC. Static adhesion of activated NK cells was reduced by approximately 60% by blocking either CD49d or CD106, by 47% by blocking CD11a, and by 82% upon simultaneous blocking of CD11a and CD49d. CONCLUSIONS Interactions between hu CD49d and po CD106 are crucial for both rolling and firm adhesion of hu NK cells to po EC and thus represent attractive targets for specific therapeutic interventions to prevent NK cell-mediated responses against po xenografts.
Collapse
Affiliation(s)
- Mårten K J Schneider
- Laboratory for Transplantation Immunology, University Hospital Zürich, CH-8091 Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Abstract
The limited availability of donor organs restricts the number of kidney transplantations performed per year. One novel solution to this shortage envisions 'growing' new kidneys in situ via xenotransplantation of renal primordia. To be successful, such an approach would require the following criteria to be met: that transplants are 'grown' in a location that renders their subsequent excretory function possible; that developed transplants have normal morphology and function; that transplanted metanephroi become well integrated into the host, in terms of inducing a state of immune tolerance and being vascularized by host vessels; and that source material for metanephros transplants is easy to access. This review summarizes recent work addressing these issues.
Collapse
|
43
|
Bühler L, Alwayn IP, Basker M, Oravec G, Thall A, White-Scharf ME, Sachs DH, Awwad M, Cooper DK. CD40-CD154 pathway blockade requires host macrophages to induce humoral unresponsiveness to pig hematopoietic cells in baboons. Transplantation 2001; 72:1759-68. [PMID: 11740385 DOI: 10.1097/00007890-200112150-00009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The effect of CD154 blockade and macrophage depletion or inhibition on baboon humoral and cellular immune responses to pig antigens was studied in a pig-to-baboon peripheral blood mobilized progenitor cell (PBPC) transplantation model aimed at inducing tolerance. We infused pig PBPCs in baboons pretreated with a nonmyeloablative regimen along with murine anti-human CD154 monoclonal antibody (mAb) and macrophage-depleting or -inhibiting agents. Group 1 baboons (n=2) underwent a nonmyeloablative regimen and immunoadsorption of anti-Gal(alpha)1,3Gal (Gal) antibody (Ab) before intravenous infusion of high doses (1.3-4.6 x 10(10)cells/kg) of PBPCs. In group 2 (n=5), cyclosporine was replaced by 8 doses of anti-CD154 mAb over 14 days. Group 3 (n=3) received the group 2 regimen plus medronate liposomes (n=2) or commercially available human intravenous immunoglobulin G depleted of anti-Gal Ab (n=1) to deplete/inhibit recipient macrophages. Group 1 developed sensitization to Gal and also developed new Ab to non-Gal porcine antigens within 10 to 20 days. In group 2, no sensitization to Gal or non-Gal determinants was seen, but Gal-reactive antibodies did return to their preleukocyte transplantation levels. CD154 blockade, therefore, induced humoral unresponsiveness to pig cells. In group 3, sensitization to Gal was seen in all three baboons at 20 days, and Abs against new porcine determinants developed in one baboon. The depletion or inhibition of host macrophages, therefore, prevented the induction of humoral unresponsiveness by CD154 blockade. These results suggest that CD154 blockade induces humoral unresponsiveness by a mechanism that involves the indirect pathway of antigen presentation. In vitro investigation of baboon anti-pig mixed lymphocyte reaction confirmed that only the indirect pathway is efficiently blocked by anti-CD154 mAb. The mechanism in which blockade of the CD40-CD154 pathway induces its effect remains to be determined, but it could involve the generation of regulatory cells capable of suppressing the direct pathway.
Collapse
Affiliation(s)
- L Bühler
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Schneider MK, Forte P, Seebach JD. Adhesive interactions between human NK cells and porcine endothelial cells. Scand J Immunol 2001; 54:70-5. [PMID: 11439150 DOI: 10.1046/j.1365-3083.2001.00966.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Human natural killer (NK) cells are able to adhere to xenogeneic porcine endothelial cells (EC) and evidence from in vitro studies as well as animal models suggests a potential role for NK cells in the cellular recognition and damage of porcine xenogeneic tissues. One possible explanation for the observed NK cell-mediated xenogeneic cytotoxicity against porcine EC is the molecular incompatibility between porcine major histocompatibility complex (MHC) class I molecules and MHC-specific inhibitory receptors on human NK cells. In this review we attempt to summarize the current knowledge concerning adhesive interactions between human NK cells and porcine EC under special considerations of the cross-species receptor-ligand interactions. Methodological differences in assessing adhesion between various studies are reviewed and comparisons to the syngeneic/allogeneic adhesion mechanisms are made. Finally, the therapeutic potential of blocking antibodies and transgenic HLA expression in preventing NK-cell adhesion and xenogeneic cytotoxicity is discussed.
Collapse
Affiliation(s)
- M K Schneider
- Laboratory for Transplantation Immunology, University Hospital Zürich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | | | | |
Collapse
|
45
|
Tereb DA, Kirkiles-Smith NC, Kim RW, Wang Y, Rudic RD, Schechner JS, Lorber MI, Bothwell AL, Pober JS, Tellides G. Human T cells infiltrate and injure pig coronary artery grafts with activated but not quiescent endothelium in immunodeficient mouse hosts. Transplantation 2001; 71:1622-30. [PMID: 11435975 DOI: 10.1097/00007890-200106150-00023] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND We have previously demonstrated that human artery grafts transplanted to immunodeficient mice are infiltrated and injured by unsensitized allogeneic human T cells. We extended our investigations to human anti-porcine xenoresponses in this model. METHODS Pig coronary artery segments were interposed into the infrarenal aorta of severe combined immunodeficiency/beige mice. After 7 days, certain recipients were reconstituted with human leukocytes and/or treated with proinflammatory cytokines. The grafts were harvested after 1-70 days and examined by histology, immunohistochemistry, and morphometry. RESULTS Pig artery grafts from untreated mice had no evidence of injury, leukocytic infiltrate, or endothelial cell activation up to 70 days postoperatively, despite deposition of murine complement. Host reconstitution with human peripheral blood mononuclear cells resulted in a discrete population of circulating T cells that did not infiltrate or injure the grafts up to 28 days after adoptive transfer. Administration of porcine interferon-gamma for up to 28 days sustained the expression of graft vascular cell adhesion molecule-1 and major histocompatibility complex antigens, but did not initiate recruitment of human leukocytes. In contrast, treatment with human tumor necrosis factor for 7 days induced the de novo expression of porcine E-selectin by graft endothelial cells and elicited human T cell infiltration and human peripheral blood mononuclear cell-dependent vascular injury. CONCLUSIONS The human peripheral blood mononuclear cell-severe combined immunodeficiency/beige mouse model identifies a significant difference between human T cell allogeneic and xenogeneic responses in vivo. Xenografts with quiescent endothelium are not infiltrated or injured by T cells under the same conditions in which allografts are rejected. Activation of pig coronary artery endothelial cells by human tumor necrosis factor, but not porcine interferon-gamma, elicits cellular xenoresponses.
Collapse
Affiliation(s)
- D A Tereb
- Section of Cardiothoracic Surgery, Yale University School of Medicine, 121 FMB, 333 Cedar Street, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ohdan H, Yang YG, Swenson KG, Kitamura H, Sykes M. T cell and B cell tolerance to GALalpha1,3GAL-expressing heart xenografts is achieved in alpha1,3-galactosyltransferase-deficient mice by nonmyeloablative induction of mixed chimerism. Transplantation 2001; 71:1532-42. [PMID: 11435961 DOI: 10.1097/00007890-200106150-00009] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND We have previously demonstrated that mixed xenogeneic chimerism and donor-specific T-cell tolerance can be induced in the rat-to-mouse species combination by using a relatively nontoxic, nonmyeloablative conditioning regimen. However, natural antibodies (NAbs) against Galalpha1,3Gal (Gal) pose an additional major barrier to pig-to-human vascularized xenograft acceptance. METHODS To determine whether the mixed chimerism approach could also overcome this humoral barrier, T cell-depleted rat (GalT+/+) bone marrow cells (BMC) were transplanted to alpha1,3-galactosyltransferase deficient (GalT-/-) mice conditioned with a nonmyeloablative regimen, consisting of transient T cell and natural killer (NK) cell depletion, 3 Gy whole body irradiation, and 7 Gy thymic irradiation. RESULTS By giving a high dose (180x106) of rat BMC, persistent mixed chimerism could be induced in GalT-/- mice, although the level of donor-type hematopoietic repopulation declined over time. Induction of mixed chimerism was associated with a rapid disappearance of anti-Gal and anti-rat NAb in the sera. Both anti-Gal Ab-producing cells and B cells with receptors recognizing Gal were undetectable in mixed chimeras, even when the chimerism levels declined, suggesting that a very low level of chimerism could effectively maintain B-cell tolerance to Gal, probably by clonal deletion and/or receptor editing. Mixed chimeras accepted subsequently transplanted donor-type rat hearts (>100 days) without immunosuppressive therapy, whereas delayed vascular and even hyperacute rejection of rat hearts occurred in conditioned control GalT-/- mice. Cellular rejection occurred by 5-6 days in conditioned control wild-type mice. CONCLUSIONS These findings demonstrate that induction of mixed chimerism with a nonmyeloablative regimen can prevent vascularized xenograft rejection by cellular and anti-Gal Ab-dependent pathways in GalT+/+-to-GalT-/- species combinations.
Collapse
Affiliation(s)
- H Ohdan
- Bone Marrow Transplantation Section, Transplantation Biology Research Center, Massachusetts General Hospital, MGH East, Building 149-5102, 13th Street, Boston, MA 02129, USA
| | | | | | | | | |
Collapse
|
47
|
Lalain S, Clémenceau B, Gouin E, Saï P. In vitro co-incubation of pig islet cells with xenogeneic human blood mononuclear cells causes loss of insulin release during perifusion: involvement of non-T-cell- and T-cell-mediated mechanisms. Hum Immunol 2001; 62:607-14. [PMID: 11390035 DOI: 10.1016/s0198-8859(01)00251-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Because the different steps of the human cellular immune rejection of pig islets are still poorly understood, our previous work concerned the intensity and mechanisms of the proliferation of human peripheral blood mononuclear cells (PBMC) to adult pig islet cells (PIC). As lymphocyte proliferation is not indicative of alteration of PIC, the present in vitro study evaluated cell-mediated immune effectors possibly involved in impairment of adult PIC. A test was thus developed, based on perifusion analysis of the alteration of insulin release from PIC incubated with different human cells. Compared to PIC incubation alone or with autologous pig splenocytes, seven-day co-incubation with whole human peripheral blood mononuclear cells (PBMC) (n = 18) led to almost complete abolition of basal and stimulated insulin releases (p < 0.0001). This effect could not be reversed by extensive sequential washes before perifusion of PIC, and the number of PIC was decreased by 78% after seven-day co-incubation with PBMC. PBMC are a complex mixture of cells involved in different xenogeneic mechanisms, and two components of this PIC impairment were then detected separately. First, the effect of PBMC against PIC was decreased (p < 0.0001) after removal of either MHC class II+ or CD14+ cells from PBMC. On the contrary, decreasing effect (p < 0.001) on insulin secretion was observed when only plastic-adherent or CD14+ cells were co-incubated with PIC. Additionally, alteration of insulin release from PIC cultured with PBMC or plastic-adherent cells was abolished dose-dependently (p < 0.0001 and p < 0.04, respectively) by gadolinium chloride (which inhibits macrophages), but not modified by cyclosporin A or mycophenolate mofetil which did not alter insulin release from PIC but blocked the proliferation of PBMC against PIC. A second mechanism was also detected, since co-incubation of PIC with purified human T cells remixed with antigen-presenting cells led to a decrease (p < 0.0001) of insulin release. This model based on the alteration of dynamic basal and stimulated insulin secretion provides detailed account of in vitro human cell-mediated impairment of PIC. It shows that the xenogeneic effect of whole mononuclear cells was strong and rapid. A crucial role was played by MHC class II+, CD14+, and plastic-adherent cells. Two mechanisms appear to be responsible for the role of these cells: 1) early direct effect, potentially involved in vivo in primary nonfunction of islets aggressed by monocytes/macrophages; and 2) the presentation of PIC xenoantigens leading to impairment by T lymphocytes, which may be involved in in vivo specific cellular rejection.
Collapse
Affiliation(s)
- S Lalain
- Cellular and Molecular Immuno-Endocrinology, University/INRA/ENVN, Nantes, France
| | | | | | | |
Collapse
|
48
|
Abstract
To determine whether transplanted metanephroi grow and differentiate after implantation into the omentum in hosts of a different species, we implanted metanephroi from embryonic day 15 (E15) rat embryos into uninephrectomized mice (hosts). Some host mice received human CTLA4Ig (hCTLA4Ig), anti-CD45RB, and anti-CD154 (tolerance-inducing agents). E15 metanephroi contained only metanephric blastema, segments of ureteric bud, and primitive nephrons with no glomeruli. Rat metanephroi did not grow or differentiate in mice that received no tolerance-inducing agents. However, by 2 wk posttransplantation in mice that received hCTLA4Ig, anti-CD45RB, and anti-CD154, metanephroi from E15 rats had enlarged, become vascularized, and formed mature tubules and glomeruli. Rat metanephroi contained cells that stained specifically for mouse CD31, a marker for sprouting endothelial cells. Some rat glomerular capillary loops stained positively for mouse CD31. Here, we show that chimeric kidneys develop from metanephroi transplanted rat→mouse and that glomeruli are vascularized, at least in part, by host vessels.
Collapse
Affiliation(s)
- S A Rogers
- George M. O'Brien Kidney and Urological Disease Center, Renal Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
49
|
Abstract
Achieving transplantation tolerance is an important goal in the effort to reduce long-term morbidity and mortality in organ transplant recipients. Robust, lifelong, donor-specific tolerance can be reliably achieved by induction of mixed chimerism in various animal models. To date, the clinical application of these proto-cols has been impeded partly by the potential toxicity of the required host conditioning regimens and the lack of successful studies in large animals. This article reviews the progress achieved in recent years in developing considerably milder conditioning protocols in rodents, and in extending some of these models to achieve permanent mixed chimerism and tolerance in large animals. Advances in the induction of xenogeneic tolerance through mixed chimerism are also discussed.
Collapse
Affiliation(s)
- T Wekerle
- Department of Surgery, Vienna General Hospital, University of Vienna, Währingergürtel 18, Vienna, A-1090 Austria.
| | | |
Collapse
|
50
|
Abstract
The number of kidney transplantations performed per year is restricted by the limited availability of donor organs. One possible solution to this shortage is the use of renal xenografts. However, the transplantation of xenografts is complicated by hyperacute and acute rejection. A second possible solution is to 'grow a kidney' from a transplanted renal anlage. It has been postulated that the host immune response might be attenuated after the transplantation of such an anlage (metanephros) instead of a developed kidney. Transplanted metanephroi become chimeric organs in that their blood supply originates, at least partly, from the host. It is possible to transplant a developing metanephros, without the use of immunosuppression, from one rat to another. Transplanted metanephroi grow, differentiate, become vascularized, and function in host rats. 'Growing kidneys' via the transplantation of metanephroi may hold promise as a novel therapeutic approach to the treatment of chronic renal failure.
Collapse
Affiliation(s)
- M R Hammerman
- George M O'Brien Kidney and Urological Disease Center, Department of Medicine, Washington University School of Medicine, St Louis, Missouri 63110, USA.
| |
Collapse
|