1
|
Sykes M, Sachs DH. Transplanting organs from pigs to humans. Sci Immunol 2020; 4:4/41/eaau6298. [PMID: 31676497 DOI: 10.1126/sciimmunol.aau6298] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022]
Abstract
The success of organ transplantation is limited by the complications of immunosuppression, by chronic rejection, and by the insufficient organ supply, and thousands of patients die every year while waiting for a transplant. With recent progress in xenotransplantation permitting porcine organ graft survival of months or even years in nonhuman primates, there is renewed interest in its potential to alleviate the organ shortage. Many of these advances are the result of our heightened capacity to modify pigs genetically, particularly with the development of CRISPR-Cas9-based gene editing methodologies. Although this approach allows the engineering of pig organs that are less prone to rejection, the clinical application of xenotransplantation will require the ability to avoid the ravages of a multifaceted attack on the immune system while preserving the capacity to protect both the recipient and the graft from infectious microorganisms. In this review, we will discuss the potential and limitations of these modifications and how the engineering of the graft can be leveraged to alter the host immune response so that all types of immune attack are avoided.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, NY, USA. .,Department of Microbiology and Immunology, Columbia University Medical Center, NY, USA.,Department of Surgery, Columbia University Medical Center, NY, USA
| | - David H Sachs
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, NY, USA.,Department of Surgery, Columbia University Medical Center, NY, USA
| |
Collapse
|
2
|
Sykes M. IXA Honorary Member Lecture, 2017: The long and winding road to tolerance. Xenotransplantation 2018; 25:e12419. [PMID: 29913040 DOI: 10.1111/xen.12419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Abstract
The last 15 years or so have seen exciting progress in xenotransplantation, with porcine organ grafts surviving months or even years in non-human primates. These advances reflect the application of new scientific knowledge, improved immunosuppressive agents, and genetic engineering. The field has recently enjoyed a renaissance of interest and hope, largely due to the exponential increase in our capacity to genetically engineer porcine source animals. However, immune responses to xenografts are very powerful and widespread clinical application of xenotransplantation will depend on the ability to suppress these immune responses while preserving the capacity to protect both the recipient and the graft from infectious microorganisms. Our work over the last three decades has aimed to engineer the immune system of the recipient in a manner that achieves specific tolerance to the xenogeneic donor while preserving otherwise normal immune function. Important proofs of principle have been obtained, first in rodents, and later in human immune systems in "humanized mice" and finally in non-human primates, demonstrating the capacity and potential synergy of mixed xenogeneic chimerism and xenogeneic thymic transplantation in tolerizing multiple arms of the immune system. Considering the fact that clinical tolerance has recently been achieved for allografts and the even greater importance of avoiding excessive immunosuppression for xenografts, it is my belief that it is both possible and imperative that we likewise achieve xenograft tolerance. I expect this to be accomplished through the availability of targeted approaches to recipient immune conditioning, understanding of immunological mechanisms of tolerance, advanced knowledge of physiological incompatibilities, and the availability of inbred miniature swine with optimized use of genetic engineering.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA.,Department of Microbiology & Immunology, Columbia University, New York, NY, USA.,Department of Surgery, Columbia University, New York, NY, USA
| |
Collapse
|
3
|
Griesemer A, Yamada K, Sykes M. Xenotransplantation: immunological hurdles and progress toward tolerance. Immunol Rev 2015; 258:241-58. [PMID: 24517437 DOI: 10.1111/imr.12152] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The discrepancy between organ need and organ availability represents one of the major limitations in the field of transplantation. One possible solution to this problem is xenotransplantation. Research in this field has identified several obstacles that have so far prevented the successful development of clinical xenotransplantation protocols. The main immunologic barriers include strong T-cell and B-cell responses to solid organ and cellular xenografts. In addition, components of the innate immune system can mediate xenograft rejection. Here, we review these immunologic and physiologic barriers and describe some of the strategies that we and others have developed to overcome them. We also describe the development of two strategies to induce tolerance across the xenogeneic barrier, namely thymus transplantation and mixed chimerism, from their inception in rodent models through their current progress in preclinical large animal models. We believe that the addition of further beneficial transgenes to Gal knockout swine, combined with new therapies such as Treg administration, will allow for successful clinical application of xenotransplantation.
Collapse
Affiliation(s)
- Adam Griesemer
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | | | |
Collapse
|
4
|
Cellular Immune Responses to Xenografts. Xenotransplantation 2014. [DOI: 10.1128/9781555818043.ch5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
5
|
Fasler-Kan E, Barteneva NS, Ketterer S, Wunderlich K, Reschner A, Nurzhanova A, Flammer J, Huwyler J, Meyer P. Human cytokines activate JAK-STAT signaling pathway in porcine ocular tissue. Xenotransplantation 2013; 20:469-80. [PMID: 24289470 PMCID: PMC4235432 DOI: 10.1111/xen.12070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 09/20/2013] [Indexed: 12/15/2022]
Abstract
Background The JAK/STAT (Janus Tyrosine Kinase, Signal Transducers and Activators of Transcription) pathway is associated with cytokine or growth factor receptors and it is critical for growth control, developmental regulation and homeostasis. The use of porcine ocular cells as putative xenotransplants appears theoretically possible. The aim of this study was to investigate the response of various porcine ocular cells in vitro to human cytokines in regard to the activation of JAK-STAT signaling pathways. Methods Porcine lens epithelial cells, pigmented iris epithelial cells and pigmented ciliary body cells were used in this study. These cells were isolated from freshly enucleated porcine eyes by enzymatic digestion. Cultured cells between passages 3–8 were used in all experiments. Electromobility shift assay (EMSA), proliferation assay, immunofluorescence staining and flow cytometry were used to evaluate the JAK-STAT signaling pathway in these cells. Results JAK/STAT signaling pathways could be activated in porcine pigmented epithelial ciliary body cells, in pigmented iris epithelial cells and in lens epithelial cells in response to porcine and human interferons and cytokines. All cells showed very strong STAT1 activation upon stimulation with porcine interferon-gamma. Porcine ocular cells also respond to human cytokines; IFN-alpha induced strong activation of STAT1 in EMSA, flow cytometry and immunofluorescence experiments whereas activation of STAT3 was less strong in EMSA, but strong in flow cytometry and immunofluorescence. Human recombinant IL-6 activated STAT3 and human IL-4 activated STAT6. With the help of immunofluorescence assay and flow cytometry we observed nuclear localization of STAT proteins after activation of porcine ocular cells with cytokines and interferons. Human IFN-α had an inhibitory effect on porcine ocular cells in proliferation assays. Conclusion Our study demonstrated that some types of human cytokines and interferon activate intracellular JAK-STAT signaling pathways in porcine ocular cells. We hypothesize that direct stimulation of the JAK-STAT pathway in porcine cells in response to human cytokines will lead to complications or failure, if pig-to-human ocular tissue xenotransplantation were to be carried out. For successful xenotransplantation among other obstacles there must be new approaches developed to regulate signaling pathways.
Collapse
Affiliation(s)
- Elizaveta Fasler-Kan
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Institute of Chemistry and Bioanalytics, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Mandell RB, Flick R, Staplin WR, Kaniewski LD, Carzoli AK, Manuszak RP, Wang J, Rossi GR, Vahanian NN, Link CJ. The αGal HyperAcute(®) Technology: enhancing immunogenicity of antiviral vaccines by exploiting the natural αGal-mediated zoonotic blockade. Zoonoses Public Health 2011; 56:391-406. [PMID: 19486321 DOI: 10.1111/j.1863-2378.2008.01191.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The αGal HyperAcute(®) Technology exploits a robust zoonotic blockade to enhance potency of antiviral vaccines. Naturally acquired immunity against the common αGal epitope [galactose-alpha(1,3)-galactose-beta(1,4)N-acetylglucosamine-R (Gal-α(1,3)-Gal-β(1,4)-GlcNAc-R)] is facilitated by the loss of a key enzyme in the epitope's biosynthetic pathway. As human cells are devoid of this epitope, chronic stimulus from gut flora leads to high levels of circulating anti-αGal antibodies and the development of a robust immune pathway. As the αGal epitope is immediately recognized as foreign, the naturally acquired αGal immune pathway in humans serves as a strong barrier to zoonotic infection. The αGal HyperAcute(®) Technology takes advantage of this natural process to facilitate the rapid presentation of modified antigens to antigen-presenting cells, leading to a strong immune response. The evolutionary immunity to αGal ensures that the presence of αGal epitopes on antigens will lead to a robust immune response involving cross-activation of T(H)1 immunity, characterized by cytokine secretion and increased phagocytic activity, and T(H)2 immunity characterized by high antibody titres. αGal epitopes can be applied to antiviral vaccines by biological, enzymatic or chemical means. Several detection methods that directly and indirectly verify αGal addition are discussed. Enhanced immunogenicity (humoral and cellular) of αGal-modified vaccines is shown for several antiviral vaccine candidates. αGal modification of antiviral vaccine components leads to enhanced immunogenicity. The existing body of literature describing the utility of αGal epitopes as a safe and robust immunostimulatory and -modulatory agent in humans supports the basis for applying the αGal HyperAcute(®) Technology to the improvement of antiviral vaccines, both new and currently approved.
Collapse
Affiliation(s)
- R B Mandell
- BioProtection Systems Corporation, Ames, IA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
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: 6.7] [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
|
8
|
Human leukocyte transmigration across Galalpha(1,3)Gal-negative porcine endothelium is regulated by human CD18 and CD99. Transplantation 2009; 87:491-9. [PMID: 19307784 DOI: 10.1097/tp.0b013e318195fb8d] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND In pig-to-human xenotransplantation cross-species receptor interactions mediate cellular infiltration and rejection of porcine grafts. However, the mechanisms responsible for recruitment of human leukocyte subsets across porcine endothelial cells (EC) remain largely unknown. Here, we investigated the role of CD99, CD18, and Galalpha(1,3)Gal (Gal) in this process. METHODS Adhesion and transmigration of human peripheral blood mononuclear cell (PBMC) subsets on Gal and Gal porcine EC (pEC) and on human EC was analyzed using a two-compartment system separated by a permeable membrane. The mechanisms of human PBMC recruitment to pEC were investigated by blocking cell surface receptors and by differentially measuring adhesion and transendothelial migration (TEM). RESULTS Blocking of CD18, but not CD99, decreased human PBMC adhesion on pEC, whereas blocking of CD18 or CD99 strongly reduced the subsequent human PBMC TEM across pEC. The inhibitory effect of CD99 blockade was slightly stronger across pEC as compared with human EC. A critical role for Gal in TEM of human monocytes, B, natural killer (NK), NK/T, and T cells was excluded by evaluating TEM across pEC derived from Gal and Gal pigs. CONCLUSIONS CD99 and CD18, but not Gal, play a critical role in human monocyte and lymphocyte TEM across pEC, and their respective porcine ligands may serve as targets to specifically inhibit human leukocyte recruitment in pig-to-human xenotransplantation.
Collapse
|
9
|
Sachs DH, Sykes M, Yamada K. Achieving tolerance in pig-to-primate xenotransplantation: reality or fantasy. Transpl Immunol 2008; 21:101-5. [PMID: 19059481 DOI: 10.1016/j.trim.2008.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Accepted: 11/18/2008] [Indexed: 02/06/2023]
Abstract
Because the immunologic differences between species are far greater than those within species, it is likely that the amount of immunosuppression that would be required for successful xenografting would be so much greater than that now used for allografting, that the side-effects and complications would be unacceptable. Tolerance approaches to xenotransplantation would overcome this concern. Studies in humanized mouse models have demonstrated that human T cells can be tolerized to porcine xenografts, providing important proofs of principle of the potential feasibility of pig-to-primate xenograft tolerance. The results available from studies of pig-to-primate xenotransplantation to date have demonstrated that while chronic immunosuppressive drugs have not completely avoided either T cell responses or humoral rejection, approaches directed toward tolerance induction have been encouraging with regard to avoiding immunization at both of these levels.
Collapse
Affiliation(s)
- David H Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School Boston, Massachusetts, USA.
| | | | | |
Collapse
|
10
|
Gibbons C, Sykes M. Manipulating the immune system for anti-tumor responses and transplant tolerance via mixed hematopoietic chimerism. Immunol Rev 2008; 223:334-60. [PMID: 18613846 PMCID: PMC2680695 DOI: 10.1111/j.1600-065x.2008.00636.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
SUMMARY Stem cells (SCs) with varying potentiality have the capacity to repair injured tissues. While promising animal data have been obtained, allogeneic SCs and their progeny are subject to immune-mediated rejection. Here, we review the potential of hematopoietic stem cells (HSCs) to promote immune tolerance to allogeneic and xenogeneic organs and tissues, to reverse autoimmunity, and to be used optimally to cure hematologic malignancies. We also review the mechanisms by which hematopoietic cell transplantation (HCT) can promote anti-tumor responses and establish donor-specific transplantation tolerance. We discuss the barriers to clinical translation of animal studies and describe some recent studies indicating how they can be overcome. The recent achievements of durable mixed chimerism across human leukocyte antigen barriers without graft-versus-host disease and of organ allograft tolerance through combined kidney and bone marrow transplantation suggest that the potential of this approach for use in the treatment of many human diseases may ultimately be realized.
Collapse
Affiliation(s)
- Carrie Gibbons
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | |
Collapse
|
11
|
Manzano M, Bueno P, Rueda R, Ramirez-Tortosa CL, Prieto PA, Lopez-Pedrosa JM. Intestinal toxicity induced by 5-fluorouracil in pigs: a new preclinical model. Chemotherapy 2007; 53:344-55. [PMID: 17785971 DOI: 10.1159/000107724] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Accepted: 08/06/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND The goal of this study was to develop an animal model of intestinal injury induced by 5-fluorouracil (5-FU) in pigs. METHODS Six domestic pigs were used as control (healthy group) and another 6 malnourished pigs orally received 5-FU (treated group). After 4 weeks of treatment, pigs were sacrificed and jejunum, ileum and colon were isolated for histological, immunological and biochemical analyses. RESULTS 5-FU caused a decrease in the intestinal mass. Disaccharidase, and phosphate alkaline activities, and glutathione redox cycle were disrupted by 5-FU. Histopathological alterations in the crypts and villous were greater in the small intestine than in the colon. 5-FU decreased the number of peripheral and intestinal leukocytes, promoting an increase in T-cytotoxic cells and a decrease in T-helper and B cells. CONCLUSION This pig model of intestinal dysfunction closely mimics the common side effects of cancer chemotherapy in humans, and provides a useful tool for evaluating novel antimucotoxic agents.
Collapse
Affiliation(s)
- M Manzano
- Strategic R&D Department, Abbott Nutrition International, Granada, Spain
| | | | | | | | | | | |
Collapse
|
12
|
Colvin GA, Lambert JF, Dooner MS, Cerny J, Quesenberry PJ. Murine allogeneic in vivo stem cell homing(,). J Cell Physiol 2007; 211:386-91. [PMID: 17167771 PMCID: PMC1986762 DOI: 10.1002/jcp.20945] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Stem cell homing has been studied in syngeneic models and appears to be rapid (<1 h) and dependent on cellular adhesion and migration factors. We utilized a full H2-mismatched transplantation model to determine the basics of allogeneic homing. C57BL/6J Lin-Sca-1+ cells were labeled with CFSE and injected in non-myeloablated BALB/c mice. Fluorescent cell detection was via high-speed FACS analysis. Alternatively, B6.SJL whole bone marrow cells were injected in lethally irradiated BALB/c mice (10 Gy). One, 3, 6, and 24 h after transplant, marrow was harvested and cells were either plated for high proliferative potential colony-forming cell (HPP-CFC) assay or secondarily injected into myeloablated (8 Gy) C57BL/6J mice using 10% competing C57BL/6J marrow. Chimerism was evaluated at 8 weeks. CFSE+ cells were detected in the bone marrow 1, 3, and 6 h after injection. The numbers were moderately lower when compared to syngeneic homing possibly due to strain effect. Conversely, utilizing a surrogate or secondary assay, we observed a decline of secondary engraftment of harvested cells over time, but not of HPP-CFC. Combining experiments and normalizing the 1-h time point to 100% (to allow comparison), we observed a mean relative engraftment of 87 +/- 29%, 72 +/- 21%, 84 +/- 35% of the 1 h level at 3, 6, and 24 h respectively. HPP-CFC assay showed no significant variation as a homing surrogate over 1-6 h. These data indicate a rapid homing into allogeneic recipients with a plateau at 1 h. The decline of secondary engraftability over time may indicate a phenotype alteration of homed cells.
Collapse
Affiliation(s)
- Gerald A Colvin
- Department of Research, Roger Williams Medical Center, Providence, Rhode Island, USA.
| | | | | | | | | |
Collapse
|
13
|
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
|
14
|
Jin R, Greenwald A, Peterson MD, Waddell TK. Human monocytes recognize porcine endothelium via the interaction of galectin 3 and alpha-GAL. THE JOURNAL OF IMMUNOLOGY 2006; 177:1289-95. [PMID: 16818789 DOI: 10.4049/jimmunol.177.2.1289] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Monocytes are one of the key inflammatory cells recruited to xenografts and play an important role in delayed xenograft rejection. Previous studies have demonstrated the ability of monocytes to bind to the major xenoantigen Gal-alpha(1,3)Gal-beta(1,4)GlcNAc-R; however, the receptor that mediates this interaction has yet to be identified. We provide evidence that it is Galectin-3, a approximately 30-kDa lectin that recognizes beta-galactosides (Gal-beta(1-3/4)GlcNAc) and plays diverse roles in many physiological and pathological events. Human monocyte binding is strikingly increased on porcine aortic endothelial cells (PAEC), which express high levels of Gal-alpha(1,3)Gal-beta(1,4)GlcNAc-R, compared with human aortic endothelial cells. Human monocytes obtained from healthy donors bind to Gal-alpha(1,3)Gal-beta(1,4)GlcNAc-R at variable intensities. This variation of binding intensity was consistent and reproducible in individual donors. Galectin-3 is mainly expressed in human monocytes, not lymphocytes. Purified Galectin-3 is able to bind directly to Gal-alpha(1,3)Gal-beta(1,4)GlcNAc-R. Galectin-3 can also be affinity isolated from monocytes (and not lymphocytes) using an Gal-alpha(1,3)Gal-beta(1,4)GlcNAc-R-biotin/streptavidin-bead pull-down system. Soluble Galectin-3 binds preferentially to PAEC vs human aortic endothelial cells, and this binding can be inhibited by lactose, indicating dependence on the carbohydrate recognition domain of Galectin-3. Gal-alpha(1,3)Gal-beta(1,4)GlcNAc-R is at least partly responsible for this phenomenon, as binding decreased after digestion of PAEC with alpha-galactosidase. Furthermore, monocytes pretreated with a blocking anti-Galectin-3 Ab show decreased adhesion to PAEC when compared with isotype control in a parallel plate flow chamber perfusion assay. Thus, we conclude that Galectin-3 expressed in human monocytes is a receptor for the major xenoantigen (Gal-alpha(1,3)Gal-beta(1,4)GlcNAc-R), expressed on porcine endothelial cells.
Collapse
MESH Headings
- Animals
- Antigens, Heterophile/biosynthesis
- Antigens, Heterophile/immunology
- Antigens, Heterophile/metabolism
- Binding Sites, Antibody
- Cells, Cultured
- Down-Regulation/immunology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Epitopes/metabolism
- Galectin 3/antagonists & inhibitors
- Galectin 3/biosynthesis
- Galectin 3/immunology
- Galectin 3/metabolism
- Humans
- Lectins/metabolism
- Ligands
- Monocytes/cytology
- Monocytes/immunology
- Monocytes/metabolism
- Protein Binding/immunology
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Swine
- Trisaccharides/metabolism
Collapse
Affiliation(s)
- Rongyu Jin
- Division of Thoracic Surgery, Department of Surgery, Toronto General Hospital Research Institute, University Health Network, University of Toronto, 200 Elizabeth Street, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
15
|
Simon AR, Dalla-Riva C, Kühn C, Tessmann R, Meder I, Martin U, Haverich A. Adhesive functions of both chains of VLA-integrins are not fully conserved across the human-porcine species barrier: implications for xenotransplantation. Xenotransplantation 2005; 12:473-80. [PMID: 16202071 DOI: 10.1111/j.1399-3089.2005.00251.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND A possible solution to the shortage of organs for transplantation would be the use of swine as source animals. As current immunosuppressive protocols cannot prevent rejection of these organs, super-selective immunosuppression or the induction of donor-specific central tolerance represent two promising approaches. Central tolerance induction involves bone marrow transplantation, and depends on intrathymic deletion of donor reactive host cells by donor antigen-presenting cells. In super-selective immunosuppression, the aim would be to block specific adhesive interactions on one species side only, leaving the other species side unaffected. As both processes depend on the interaction of adhesion molecules with their ligands, we investigated whether the beta1-integrins, which play roles in hematopoiesis as well as in rejection, can successfully interact across the swine-to-human species barrier. METHODS We employed static cell-to-extracellular protein and cell-to-cell adhesion assays, using different cell types and monoclonal antibody as well as peptide-fragments to analyze conservation of cross-species adhesive interactions. RESULTS We found that porcine and human cells interact differently with their cross-species ligands than their own and that the adhesive function of the beta1-chain does not seem to be fully conserved across the species barrier. CONCLUSIONS Integrin functions are not fully conserved across the pig-to-human species barrier. While the development of multi-transgenic pigs, whose integrins interact with human ligands in a more ''human-like'' manner may be necessary to facilitate tolerance induction, these facts give rise to new possibilities concerning super-selective immunosuppression.
Collapse
Affiliation(s)
- André R Simon
- Department of Thoracic- and Cardiovascular Surgery, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
16
|
Giovino MA, Wang H, Sykes M, Yang YG. Role of VLA-4 and VLA-5 in ex vivo maintenance of human and pig hematopoiesis in human stroma-supported long-term cultures. Exp Hematol 2005; 33:363-70. [PMID: 15730860 DOI: 10.1016/j.exphem.2004.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Revised: 10/08/2004] [Accepted: 11/18/2004] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The advantage of recipient hematopoiesis over that of xenogeneic donors poses a fundamental obstacle to the induction of xenograft tolerance through mixed hematopoietic chimerism. Here we explore the role of beta1 integrins in maintenance of human vs porcine hematopoiesis within a human hematopoietic environment. METHODS Porcine and human c-kit+ bone marrow cells were purified and cultured on human bone marrow stroma for 6 weeks. The role of VLA-4 and VLA-5 in the maintenance of porcine vs human hematopoiesis in this human stroma-supported long-term bone marrow culture (LTBMC) system was evaluated by using blocking mAbs that bind to both species. RESULTS Blocking VLA-4 with HP2/1 inhibited both human and porcine hematopoiesis, whereas anti-VLA-5 (SAM-1) suppressed the function of human, but not porcine, hematopoietic cells. In mixed LTBMC of porcine and human cells on a human stroma, porcine hematopoietic cells were at a competitive disadvantage, as seen by a rapid decline in cellularity, including clonogenic progenitors. This disadvantage was substantially overcome by the addition of SAM-1. Furthermore, human, but not porcine, cell adhesion to human fibronectin was inhibited by arginine-glycine-aspartic acid (RGD) peptides. CONCLUSION Taken together, these results indicate that VLA-4 plays critical role for porcine hematopoiesis in a human hematopoietic environment, and raise the possibility that porcine VLA-5 might be unable to bind the respective human ligand and/or to initiate adequate post-ligand-binding signaling. Thus, VLA-5 may provide a potential target for developing approaches to improve porcine hematopoiesis in human recipients.
Collapse
|
17
|
Yang YG. Application of xenogeneic stem cells for induction of transplantation tolerance: present state and future directions. ACTA ACUST UNITED AC 2004; 26:187-200. [PMID: 15378269 DOI: 10.1007/s00281-004-0159-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2004] [Accepted: 04/18/2004] [Indexed: 01/22/2023]
Abstract
Xenotransplantation using pig organs provides a possible solution to the severe shortage of allogeneic organ donors, one of the major limiting factors in clinical transplantation. However, because of the greater antigenic differences that exist between different species than within a species, the immune response to xenografts is much more vigorous than to allografts. Thus, tolerance induction is essential to the success of clinical xenotransplantation. Tolerance induced by mixed hematopoietic chimerism across the MHC barrier is remarkably robust, but its ability to induce tolerance across highly disparate xenogeneic barriers remains poorly studied. None of the current available regimens of host conditioning, which permit hematopoietic stem cell engraftment and chimerism induction in allogeneic or closely related (concordant) xenogeneic combinations, has been demonstrated to be effective in establishing porcine hematopoietic chimerism in a discordant xenogeneic species. Unlike bone marrow transplantation within the same species, the innate immune system and the species specificity of cytokines and adhesion molecules essential to hematopoiesis pose formidable obstacles to the establishment of donor hematopoiesis across discordant xenogeneic barriers. The genetic incompatibility between species may also impede xenograft tolerance induction by mixed chimerism. While we remain far from achieving tolerance in clinical xenotransplantation, recent studies using a transgenic mouse model have proven the principle that mixed hematopoietic chimerism may induce mouse and human T cell tolerance to porcine xenografts. This review article focuses on the barriers to porcine hematopoietic engraftment in highly disparate xenogeneic species and the possible application of mixed hematopoietic chimerism to xenograft tolerance induction.
Collapse
Affiliation(s)
- Yong-Guang Yang
- Bone Marrow Transplantation Section, Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, MGH-East, Building 149, 13th Street, Boston, MA 02129, USA.
| |
Collapse
|
18
|
Crosby K, Yatko C, Dersimonian H, Pan L, Edge ASB. A novel monoclonal antibody inhibits the immune response of human cells against porcine cells: identification of a porcine antigen homologous to CD58. Transplantation 2004; 77:1288-94. [PMID: 15114100 DOI: 10.1097/01.tp.0000120377.57543.d8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Human CD58 is an adhesion molecule that interacts with CD2 on lymphocytes. We describe here an antibody that blocks responses of human peripheral blood mononuclear cells (PBMCs) to porcine cells and reacts with a porcine protein with homology to CD58. METHODS Antibodies were isolated with a screen for inhibition of the human antiporcine response. One of these antibodies was used for immunoaffinity purification of a protein that was identified by molecular weight determination, endoglycosidase sensitivity, and microsequencing analysis as a porcine homologue of CD58. RESULTS The antigen recognized by this antibody was a cell surface protein of relative molecular mass (Mr)=45,000 containing N-linked carbohydrate chains. Immunoaffinity purification of this protein and microsequencing revealed homology to sheep CD58 as well as sequences that were common to this protein and both sheep and human CD58. The protein was widely distributed on porcine cells, including lymphocytes, endothelial cells, muscle cells, and neuronal cells. This antibody efficiently inhibited lysis of porcine targets by human PBMCs in addition to preventing proliferation of the human PBMCs in response to the porcine cells. CONCLUSIONS The CD2 interaction with porcine cells is important for the efficient recognition of porcine tissue, and inhibition of the human antiporcine immune response with the antibody is likely to be caused by the disruption of the human CD2 interaction with this porcine homologue of CD58. The antibody may prove to be useful for the blocking of this interaction without interfering with other functions of T cells.
Collapse
|
19
|
Baumann BC, Forte P, Hawley RJ, Rieben R, Schneider MKJ, Seebach JD. Lack of Galactose-α-1,3-Galactose Expression on Porcine Endothelial Cells Prevents Complement-Induced Lysis but Not Direct Xenogeneic NK Cytotoxicity. THE JOURNAL OF IMMUNOLOGY 2004; 172:6460-7. [PMID: 15128838 DOI: 10.4049/jimmunol.172.10.6460] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The galactose-alpha-1,3-galactose (alphaGal) carbohydrate epitope is expressed on porcine, but not human cells, and therefore represents a major target for preformed human anti-pig natural Abs (NAb). Based on results from pig-to-primate animal models, NAb binding to porcine endothelial cells will likely induce complement activation, lysis, and hyperacute rejection in pig-to-human xenotransplantation. Human NK cells may also contribute to innate immune responses against xenografts, either by direct recognition of activating molecules on target cells or by FcgammaRIII-mediated xenogeneic Ab-dependent cellular cytotoxicity (ADCC). The present study addressed the question as to whether the lack of alphaGal protects porcine endothelial cells from NAb/complement-induced lysis, direct xenogeneic NK lysis, NAb-dependent ADCC, and adhesion of human NK cells under shear stress. Homologous recombination, panning, and limiting dilution cloning were used to generate an alphaGal-negative porcine endothelial cell line, PED2*3.51. NAb/complement-induced xenogeneic lysis of PED2*3.51 was reduced by an average of 86% compared with the alphaGal-positive phenotype. PED2*3.51 resisted NK cell-mediated ADCC with a reduction of lysis ranging from 30 to 70%. However, direct xenogeneic lysis of PED2*3.51, mediated either by freshly isolated or IL-2-activated human NK cells or the NK cell line NK92, was not reduced. Furthermore, adhesion of IL-2-activated human NK cells did not rely on alphaGal expression. In conclusion, removal of alphaGal leads to a clear reduction in complement-induced lysis and ADCC, but does not resolve adhesion of NK cells and direct anti-porcine NK cytotoxicity, indicating that alphaGal is not a dominant target for direct human NK cytotoxicity against porcine cells.
Collapse
MESH Headings
- Animals
- Antibodies, Heterophile/metabolism
- Antibody-Dependent Cell Cytotoxicity/genetics
- Antigens, Heterophile/immunology
- Antigens, Heterophile/physiology
- Binding Sites, Antibody/genetics
- Cell Adhesion/genetics
- Cell Adhesion/immunology
- Cell Line
- Cell Line, Transformed
- Clone Cells
- Complement System Proteins/physiology
- Cytotoxicity, Immunologic/genetics
- Disaccharides/deficiency
- Disaccharides/genetics
- Disaccharides/immunology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Humans
- Immune Tolerance/genetics
- Killer Cells, Natural/immunology
- Stress, Mechanical
- Swine
Collapse
Affiliation(s)
- Bettina C Baumann
- Department of Internal Medicine, Laboratory for Transplantation Immunology, University Hospital, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
20
|
Holgersson J, Ehrnfelt C, Hauzenberger E, Serrander L. Leukocyte endothelial cell interactions in pig to human organ xenograft rejection. Vet Immunol Immunopathol 2002; 87:407-15. [PMID: 12072266 DOI: 10.1016/s0165-2427(02)00094-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In cases where antibody- and complement-mediated hyperacute rejection (HAR) of vascularized organ xenografts has been prevented, acute vascular rejection (AVR) and acute T cell-mediated rejection (ACR) cause graft destruction. Infiltration of leukocytes (innate and graft-primed T cells) into the graft execute the latter two rejection modalities. The leukocyte extravasation process, which is a prerequisite for graft infiltration, is governed by adhesion molecules, including the selectin, integrin and immunoglobulin protein families, and the chemokine protein family. The compatibility between porcine endothelial cell and human leukocyte adhesion molecules was investigated in dynamic adhesion and static transendothelial migration assays. The effect of human anti-pig antibodies on human leukocyte adhesion to, and transendothelial migration across, porcine endothelium was assessed under dynamic and static conditions, respectively. In contrast to previously published results, no difference in the ability of neutrophils to adhere to pig and human endothelium was observed. Furthermore, no evident quantitative or qualitative differences in the capacity of human and porcine endothelium to support transendothelial migration of human leukocytes (T, B and natural killer (NK) cells, monocytes, and neutrophils) could be detected. The presence of human anti-pig antibodies (Abs) modulated the migration of leukocytes across porcine endothelium, as well as neutrophil adhesion to porcine endothelium under conditions of flow. Antibodies specific for pig endothelial adhesion molecules can potentially be used as species (graft)-specific immunosuppressive reagents in order to prevent cellular organ xenograft rejection.
Collapse
Affiliation(s)
- Jan Holgersson
- Division of Clinical Immunology, Karolinska Institutet, F79 Huddinge University Hospital AB, S-141 86 Stockholm, Sweden.
| | | | | | | |
Collapse
|
21
|
Theodore PR, Simon AR, Warrens AN, Sackstein R, Sykes M. Porcine mononuclear cells adhere to human fibronectin independently of very late antigen-5: implications for donor-specific tolerance induction in xenotransplantation. Xenotransplantation 2002; 9:277-89. [PMID: 12060464 DOI: 10.1034/j.1399-3089.2002.01086.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
To combat the shortage of donor organs, transplantation across species barriers has been proposed. Induction of tolerance would overcome the substantial immunologic barriers to xenotransplantation and would avoid the chronic use of immunosuppressive agents. Successful transplantation of hematopoietic cells induces robust specific tolerance to donor antigens in allogeneic and xenogeneic models. The beta1 integrin class of adhesion molecules and their interactions with extracellular matrix components are thought to be integral to the engraftment and maturation of hematopoietic stem cells. We therefore examined the efficacy of porcine very late antigen-5 (VLA-5) and VLA-4 interactions with the human extracellular matrix (ECM) protein, fibronectin. Peripheral blood mononuclear cells (PBMCs) from humans and miniature swine were flourochrome labeled and adhesion to plates coated with whole human fibronectin (whFN) or its 120 KDa fragment containing the VLA-5 binding region was determined. Flow cytometry and immuno- precipitation were used to identify a monoclonal antibody that cross-reacted on porcine VLA-5. Human and pig PBMC adhesion to human fibronectin (hFN) or 120 kDa fragment-coated plates was assessed following incubation with control ab, anti-VLA-4, anti-VLA-5, or soluble fibronectin. Using rabbit complement, cells expressing VLA-5 were purged from PBMC preparations before performing the adhesion assay. Porcine and human PBMC both adhered to hFN in a divalent cation-dependent and activation-dependent manner. Adhesion to hFN of human but not pig PBMC was blocked by anti-VLA-5 monoclonal antibody SAM-1, although this mAb immunoprecipitated a heterodimeric cell surface molecule (155/135 kDa) resembling VLA-5 from pig PBMC. Complement-mediated depletion of VLA-5-expressing cells ablated specific binding of human but not porcine cells to hFN and its 120 kDa fragment. Addition of soluble fibronectin was capable of blocking adhesion of PBMC of both species to hFN. Anti-VLA-4 reduced the binding of PBMC from both species to hFN to a similar extent. Human and pig cells can specifically adhere to hFN and its 120 kDa fragment, suggesting that this critical cell-ECM interaction is preserved across species. While human cells exclusively use VLA-5 for binding to the 120 kDa fragment, porcine cells could not be shown to adhere to whFN or its 120 kDa fragment via VLA-5. However, porcine VLA-4 is capable of mediating adhesion to human FN. We conclude that disparities in the adhesive interactions of beta1 integrins may be a barrier to the use of porcine hematopoietic stem cell transplantation as a means of inducing donor-specific tolerance in the pig to human species combination.
Collapse
Affiliation(s)
- Pierre R Theodore
- Transplantation Biology Research Center, Surgical Service, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
22
|
Abe M, Qi J, Sykes M, Yang YG. Mixed chimerism induces donor-specific T-cell tolerance across a highly disparate xenogeneic barrier. Blood 2002; 99:3823-9. [PMID: 11986242 DOI: 10.1182/blood.v99.10.3823] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Induction of tolerance is likely to be essential for successful xenotransplantation because immune responses across xenogeneic barriers are vigorous. Although mixed hematopoietic chimerism leads to stable donor-specific tolerance in allogeneic and closely related xenogeneic (eg, rat-to-mouse) combinations, the ability of this approach to induce tolerance across a highly disparate xenogeneic barrier has not yet been demonstrated. In this study, we investigated the immune responses of murine T cells that developed in mice with pre-established porcine hematopoietic chimerism. Our results show for the first time that induction of porcine hematopoietic chimerism can eliminate the development of antiporcine donor responses in a highly disparate xenogeneic species. Porcine hematopoietic chimeras showed donor-specific nonresponsiveness in the mixed lymphocyte reaction, lack of antidonor IgG antibody production, and acceptance of donor skin grafts. Thus, mixed chimerism is capable of inducing tolerance in a highly disparate xenogeneic combination and may have clinical potential to prevent xenograft rejection. (Blood. 2002;99:3823-3829)
Collapse
Affiliation(s)
- Masahiro Abe
- Transplantation Biology Research Center, Surgical Service, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
| | | | | | | |
Collapse
|
23
|
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
|
24
|
Forte P, Pazmany L, Matter-Reissmann UB, Stussi G, Schneider MK, Seebach JD. HLA-G inhibits rolling adhesion of activated human NK cells on porcine endothelial cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6002-8. [PMID: 11698480 DOI: 10.4049/jimmunol.167.10.6002] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human NK cells adhere to and lyse porcine endothelial cells (pEC) and therefore may contribute to the cell-mediated rejection of vascularized pig-to-human xenografts. Since MHC class I molecules inhibit the cytotoxic activity of NK cells, the expression of HLA genes in pEC has been proposed as a potential solution to overcome NK cell-mediated xenogeneic cytotoxicity. HLA-G, a minimally polymorphic HLA class I molecule that can inhibit a wide range of NK cells, is an especially attractive candidate for this purpose. In this study we tested whether the expression of HLA-G on pEC inhibits the molecular mechanisms that lead to adhesion of human NK cells to pEC and subsequent xenogeneic NK cytotoxicity. To this end two immortalized pEC lines (2A2 and PED) were stably transfected with HLA-G1. Rolling adhesion of activated human NK cells to pEC monolayers and xenogeneic cytotoxicity against pEC mediated by polyclonal human NK lines as well as NK clones were inhibited by the expression of HLA-G. The adhesion was partially reversed by masking HLA-G on pEC with anti-HLA mAbs or by masking the HLA-G-specific inhibitory receptor ILT-2 on NK cells with the mAb HP-F1. The inhibition of NK cytotoxicity by HLA-G was only partially mediated by ILT-2, indicating a role for other unknown NK receptors. In conclusion, transgenic expression of HLA-G may be useful to prevent human NK cell responses to porcine xenografts, but is probably not sufficient on its own. Moreover, the blocking of rolling adhesion by HLA-G provides evidence for a novel biological function of HLA molecules.
Collapse
Affiliation(s)
- P Forte
- Department of Internal Medicine, Laboratory for Transplantation Immunology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
25
|
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: 1.3] [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
|
26
|
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
|
27
|
Seebach JD, Schneider MK, Comrack CA, LeGuern A, Kolb SA, Knolle PA, Germana S, DerSimonian H, LeGuern C, Sachs DH. Immortalized bone-marrow derived pig endothelial cells. Xenotransplantation 2001; 8:48-61. [PMID: 11208191 DOI: 10.1034/j.1399-3089.2001.00075.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Primary cultures of porcine endothelial cells (EC) can only be maintained for a limited number of passages. To facilitate studies of xenogeneic human anti-pig immune responses in vitro, pig microvascular bone-marrow (BM) and macrovascular aortic EC were obtained from our herd of partially inbred miniature swine, homozygous for the major histocompatibility locus, and immortalized with a modified SV40 large T vector. The resulting BM-derived (2A2) and aortic (PEDSV.15) immortalized EC lines showed unlimited growth and EC phenotype as indicated by expression of von Willebrand Factor (vWF) and low density lipoprotein (LDL) receptors as well as by formation of typical cobblestone monolayers. Ultrastructural studies revealed morphological similarities in primary and immortalized EC. Flow cytometry analysis demonstrated constitutive SLA class I expression by all lines whereas SLA class II was only expressed after stimulation with porcine IFNgamma. Furthermore, pig CD34 mRNA was detected by Northern blot analysis in primary and immortalized aortic EC but not in 2A2. Both EC lines expressed a number of myeloid markers, adhesion molecules and xenoantigens, the latter being determined by binding of human natural antibodies. Gene transfer into the porcine EC lines was successfully performed by electroporation or calcium-phosphate transfection, as well as by adenoviral infection. Finally, the functional similarity between primary and immortalized EC was demonstrated in adhesion and cytotoxicity assays. Together, these results suggest that 2A2 and PEDSV. 15 represent valuable tools to study both human cellular and humoral immune responses in vitro against pig EC derived from microvascular and large vessels.
Collapse
Affiliation(s)
- J D Seebach
- Department of Internal Medicine, University Hospital Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Borbolla JR, Nájera A, López-Hernández MA, González M, Siller A, Rubio B, Garcés O, Delgado JL, Collados MT. Correlation coefficients between several parameters and CD34+ cell yield in peripheral blood stem cell harvesting by apheresis. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2001; 10:17-8. [PMID: 11276354 DOI: 10.1089/152581601750098138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
29
|
Jiménez-Marín A, Garrido JJ, de Andrés-Cara DF, Morera L, Barbancho MJ, Llanes D. Molecular cloning and characterization of the pig homologue to human CD29, the integrin beta1 subunit. Transplantation 2000; 70:649-55. [PMID: 10972224 DOI: 10.1097/00007890-200008270-00019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND CD29 is the beta1 subunit, a member of the integrin gene superfamily that function as receptor for cell adhesion molecules of the extracellular matrix. Porcine integrin beta1 subunit is involved in rejection of pig-to-human tissue xenografts as target of the natural antibodies present in the human serum. Moreover since CD29, as part of the beta1 integrins very late antigen 4 (VLA-4) and VLA-6, is involved in homing and differentiation of haematopoietic progenitor cells, its characterization in pig is critical to study the interaction of porcine adhesion molecules with human ligands in the induction of donor-specific tolerance toward porcine antigens, a process extremely desirable to prevent rejection of xenogeneic organs. METHODS The porcine CD29 cDNA has been isolated from a cDNA library and its structure determined. In addition, reverse transcription-polymerase chain reaction (RT-PCR) was performed to determine the expression of CD29 in different tissues. RESULTS The nucleotide sequence of the porcine cDNA includes an open reading frame encoding a polypeptide of 798 amino acids. Expression analysis showed that porcine CD29 is expressed in all lymphoid tissues tested and, in lower amounts, in nonlymphoid tissues. Pig CD29 deduced amino acid sequence displays extensive conservation compared with CD29 sequences from other species and a common structural feature with all the other CD29 molecules analyzed in mammals, including the 12 potential N-glycosilation sites. Punctual changes between human and swine CD29 molecule into the ligand binding domain, and/or into the regulatory domain, suggest potential differences between human and porcine CD29 relative to the human CD29 ligands. CONCLUSIONS Cloning of the swine CD29 gene offers a new tool for an alternative protocol of removing xenoreactive antibodies in the recipient. In addition, the determination of the differences between human and swine CD29 will help to understand the adhesion molecule-ligand interactions and their function across the swine-human barrier in xenotransplantation.
Collapse
Affiliation(s)
- A Jiménez-Marín
- Departamento de Genética, facultad de Veterinaria, Córdaba, Spain
| | | | | | | | | | | |
Collapse
|
30
|
Dalla Riva C, Tessmann R, Kuehn C, Martin U, Diehl A, Steinhoff G, Haverich A, Simon AR. VLA-Integrin beta-1 chain function is not fully conserved between the human and porcine species: implications for xenotransplantation. Transplant Proc 2000; 32:1054-5. [PMID: 10936350 DOI: 10.1016/s0041-1345(00)01116-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- C Dalla Riva
- Leibniz Research Laboratories for Artificial Organs and Biotechnology, Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Homing of Human Cells in the Fetal Sheep Model: Modulation by Antibodies Activating or Inhibiting Very Late Activation Antigen-4–Dependent Function. Blood 1999. [DOI: 10.1182/blood.v94.7.2515.419k15_2515_2522] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mechanisms by which intravenously (IV)-administered hematopoietic cells home to the bone marrow (BM) are poorly defined. Although insightful information has been obtained in mice, our knowledge about homing of human cells is very limited. In the present study, we investigated the importance of very late activation antigen (VLA)-4 in the early phases of lodgment of human CD34+progenitors into the sheep hematopoietic compartment after in utero transplantation. We have found that preincubation of donor cells with anti–VLA-4 blocking antibodies resulted in a profound reduction of human cell lodgment in the fetal BM at 24 and 48 hours after transplantation, with a corresponding increase of human cells in the peripheral circulation. Furthermore, IV infusion of the anti–VLA-4 antibody at later times (posttransplantation days 21 to 24) resulted in redistribution or mobilization of human progenitors from the BM to the peripheral blood. In an attempt to positively modulate homing, we also pretreated human donor cells with an activating antibody to β1 integrins. This treatment resulted in increased lodgment of donor cells in the fetal liver, presumably for hemodynamic reasons, at the expense of the BM. Given previous involvement of the VLA-4/vascular cell adhesion molecule (VCAM)-1 adhesion pathway in homing and mobilization in the murine system, our present data suggest that cross-reacting ligands (likely VCAM-1) for human VLA-4 exist in sheep BM, thereby implicating conservation of molecular mechanisms of homing and mobilization across disparate species barriers. Thus, information from xenogeneic models of human hematopoiesis and specifically, the human/sheep model of in utero transplantation, may provide valuable insights into human hematopoietic transplantation biology.
Collapse
|
32
|
Nasu K, Whyte A, Green SJ, Evans PC, Kilshaw PJ. Alpha-galactosyl-mediated activation of porcine endothelial cells: studies on CD31 and VE-cadherin in adhesion and signaling. Transplantation 1999; 68:861-7. [PMID: 10515388 DOI: 10.1097/00007890-199909270-00020] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Ligation of alpha-galactosyl epitopes on endothelial cells by naturally occurring human antibodies causes hyperacute rejection in porcine-to-human xenotransplantation. The alpha-galactosyl-specific lectin Bandeiraea simplicifolia isolectin B4 (IB4) has been reported to trigger endothelial "gap" formation and tyrosine phosphorylation of an unidentified 130-kDa protein. We have studied two 130-kDa junctional adhesion molecules, CD31 and VE-cadherin, in porcine aortic endothelial cells (PAECs) during IB4-mediated activation. The cellular distribution of these molecules, their susceptibility to tyrosine phosphorylation, and their capacity to bind IB4 or natural human antibodies have been determined. METHODS Porcine CD31 and VE-cadherin were cloned. Recombinant proteins and monoclonal antibodies were prepared. The distribution and phosphorylation of CD31 and VE-cadherin in confluent PAECs activated with IB4 or human serum were studied by confocal microscopy and Western blotting, respectively. RESULTS IB4 caused rapid redistribution of CD31 and VE-cadherin away from cell junctions and tyrosine-phosphorylation of CD31 but not VE-cadherin. A monoclonal antibody to CD31 also triggered tyrosine phosphorylation of this molecule, but brief exposure of PAECs to normal human serum did not. Tyrosine-phosphorylated CD31 complexed with SHP2 and other unidentified phosphoproteins. Both IB4 and natural human antibodies bound to porcine CD31 but not to VE-cadherin. Cell adhesion tests showed that porcine and human CD31 are functionally incompatible. CONCLUSIONS Endothelial cell retraction during IB4-mediated activation of PAECs is associated with rapid loss of CD31 and VE-cadherin from cell junctions. CD31 becomes strongly tyrosine-phosphorylated and forms a cell signaling complex, which may have a significant role in the response of the xenograft vascular endothelium.
Collapse
Affiliation(s)
- K Nasu
- The Babraham Institute, Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|
33
|
Affiliation(s)
- H Rabb
- Division of Nephrology, Hennepin County Medical Center, University of Minnesota Medical School, Minneapolis, USA
| | | |
Collapse
|
34
|
Simon AR, Warrens AN, Sykes M. Efficacy of adhesive interactions in pig-to-human xenotransplantation. IMMUNOLOGY TODAY 1999; 20:323-30. [PMID: 10379051 DOI: 10.1016/s0167-5699(99)01485-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Successful xenotransplantation depends on many factors, one being the interactions of cross-species adhesion molecule-ligand pairs. Depending on the approach used to facilitate xenotransplantation, these interactions can play differing roles. Here, André Simon, Anthony Warrens and Megan Sykes review the existing information on pig-to-human adhesive interactions and its implication for different approaches to pig-to-human xenotransplantation.
Collapse
Affiliation(s)
- A R Simon
- Bone Marrow Transplantation Section, Transplantation Biology Research Center, Surgical Service, Massachusetts General Hospital/Harvard Medical School, MGH East, Building 149-5102, 13th Street, Boston, MA 02129, USA.
| | | | | |
Collapse
|
35
|
Warrens AN, Simon AR, Theodore PR, Sykes M. Human-porcine receptor-ligand compatibility within the immune system: relevance for xenotransplantation. Xenotransplantation 1999; 6:75-8. [PMID: 10431783 DOI: 10.1034/j.1399-3089.1999.00020.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Immune responses to xenografts are likely to be highly dependent on the efficiency of molecular interactions between the donor and the recipient species. This brief review summarizes what is currently known about the compatibilities across the human-porcine species barrier of the molecular interactions that are important in the immune response.
Collapse
Affiliation(s)
- A N Warrens
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston 02129, USA
| | | | | | | |
Collapse
|
36
|
Carter AS, Cerundolo L, Koo DD, Rust NA, Morris PJ, Fuggle SV. Cross-species reactivity of a panel of antibodies with monkey and porcine tissue. Xenotransplantation 1999; 6:123-30. [PMID: 10431789 DOI: 10.1034/j.1399-3089.1999.00014.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The continuing shortage of organs available for transplantation limits the number of patients able to benefit from this highly successful form of therapy. Interest in alternative sources of organs has now turned towards the pig because of its physiological similarity to human. There is a requirement therefore for reagents not only for research purposes but possibly for studying xenotransplants in the clinical situation in the future. In this study, we have concentrated on determining the cross-species reactivity of a large panel of antibodies directed against human leukocyte markers, testing peripheral blood leukocytes and also including renal tissue to determine non-leukocyte cross-reactivity. A total of 63 out of 127 antibodies cross-reacted with cynomolgus monkey cells. Twenty of these antibodies stained similar populations of leukocytes to human, whereas the remaining 43 reacted with clearly different populations. The majority of antibodies (108/127) were unreactive with porcine leukocytes, reflecting the evolutionary differences between pig and man. Of the 19 antibodies cross-reactive with porcine cells, seven reacted with similar proportions of leukocytes to human, whereas the remaining 12 antibodies stained entirely different populations. The most interesting, and potentially most useful, antibodies were four that reacted with human, cynomolgus monkey and porcine tissue in a similar manner, suggesting that the epitopes recognized are present on similar molecules. These antibodies were directed against CD29 (MEM1O1A, K20) and CD18 (BU87, 7E4), the common beta1- and beta2-integrin subunits respectively. This study demonstrates that there are antigens common to cynomolgus monkey, pig and man that react with currently available antibodies. Nevertheless, when determining cross-species reactivity of human antibodies, it is important to consider the possibility that there may be additional non-leukocyte reactivity in other tissues.
Collapse
Affiliation(s)
- A S Carter
- Nuffield Department of Surgery, University of Oxford, UK
| | | | | | | | | | | |
Collapse
|
37
|
Auchincloss H. Literature update 1998, part 3. Xenotransplantation 1999; 6:66-71. [PMID: 10355734 DOI: 10.1034/j.1399-3089.1999.00013.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- H Auchincloss
- Transplantation Unit, Surgical Services, Massachusetts General Hospital, Boston 02114, USA
| |
Collapse
|