51
|
Cytolytic assessment of hyperacute rejection and production of nuclear transfer embryos using hCD46-transgenic porcine embryonic germ cells. ZYGOTE 2009; 17:101-8. [DOI: 10.1017/s096719940800511x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SummaryHuman complement regulatory protein hCD46 may reduce the hyperacute rejection (HAR) in pig-to-human xenotransplantation. In this study, anhCD46gene was introduced into porcine embryonic germ (EG) cells. Treatment of human serum did not affect the survival of hCD46-transgenic EG cells, whereas the treatment significantly reduced the survival of non-transgenic EG cells (p< 0.01). The transgenic EG cells presumably capable of alleviating HAR were transferred into enucleated oocytes. Among 235 reconstituted oocytes, 35 (14.9%) developed to the blastocyst stage. Analysis of individual embryos indicated that 80.0% (28/35) of embryos contained the transgene hCD46. The result of the present study demonstrates resistance of hCD46-transgenic EG cells against HAR, and the usefulness of the transgenic approach may be predicted by this cytolytic assessment prior to actual production of transgenic pigs. Subsequently performed EG cell nuclear transfer gave rise to hCD46-transgenic embryos. Further study on the transfer of these embryos to recipients may produce hCD46-transgenic pigs.
Collapse
|
52
|
Niu X, Gupta K, Yang JT, Shamblott MJ, Levchenko A. Physical transfer of membrane and cytoplasmic components as a general mechanism of cell-cell communication. J Cell Sci 2009; 122:600-10. [DOI: 10.1242/jcs.031427] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Recent evidence from different research areas has revealed a novel mechanism of cell-cell communication by spontaneous intercellular transfer of cellular components (ICT). Here we studied this phenomenon by co-culturing different cells that contain distinct levels of proteins or markers for the plasma membrane or cytoplasm. We found that a variety of transmembrane proteins are transferable between multiple cell types. Membrane lipids also show a high efficiency of intercellular transfer. Size-dependent cytoplasmic transfer allows exchange of cytoplasmic macromolecules up to 40 kDa between somatic cells, and up to 2000 kDa between uncommitted human precursor cells and human umbilical vein endothelial cells. Protein transfer, lipid transfer and cytoplasmic component transfer can occur simultaneously and all require direct cell-cell contact. Analyses of the properties of ICT, together with a close examination of cell-cell interactions, suggest that the spontaneous ICT of different cellular components might have a common underlying process: transient local membrane fusions formed when neighboring cells undergo close cell-cell contact.
Collapse
Affiliation(s)
- Xinle Niu
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Kshitiz Gupta
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Joy T. Yang
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Michael J. Shamblott
- Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Andre Levchenko
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
53
|
Layton DS, Bean AG, Dodge NM, Strom ADG, Sandrin MS, Ierino FL. Differential cytokine expression and regulation of human anti-pig xenogeneic responses by modified porcine dendritic cells. Xenotransplantation 2009; 15:257-67. [PMID: 18957048 DOI: 10.1111/j.1399-3089.2008.00477.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Porcine dendritic cells (DC) are likely to be pivotal cells in the initiation of stimulatory and potential tolerogenic responses to xenoantigens, however, there are limited studies characterizing these antigen presenting cells. METHODS Porcine PBMC (CD172a(+)) were cultured with GM-CSF and IL-4 and phenotype and functional capabilities assessed. Lipopolysaccharide (LPS), IL-10, and IL-3 were added to the GM-CSF/IL-4 DC cultures to determine phenotypic and functional changes. Quantitative real-time polymerase chain reaction (PCR) for key cytokines was performed and the modified porcine DC were further assessed by primary mixed lymphocyte reaction to determine the effect of LPS, IL-10, and IL-3 on stimulatory capability. RESULTS Porcine PBMC (CD172(+)) cultured with GM-CSF and IL-4 produced cells with DC morphology, which were major histocompatability complex (MHC) class II(+), CD14(-/lo), and CD1a(lo). Addition of IL-10 or IL-3 to GM-CSF/IL-4 DC cultures produced cells with lower levels of MHC class II and higher levels of antigen uptake consistent with less mature DC. Quantitative real-time PCR of DC showed the addition of IL-10 induced an increase in IL-10 mRNA, no detectable IL-12, and reduced IL-6 mRNA. The addition of IL-3 to DC cultures decreased IL-12, IL-6 and tumor necrosis factor (TNF), with no change in IL-10 mRNA. GM-CSF/IL-4 DC induced strong human lymphocyte proliferation, compared with significantly reduced stimulatory capacity induced by IL-10 and IL-3 treated DC cultures. CONCLUSIONS The profound effect on differential DC cytokine profile and reduced human anti-pig responses has important therapeutic implications in xenotransplantation. The mechanism of altered regulation warrants further investigation.
Collapse
|
54
|
Byrne GW, Stalboerger PG, Davila E, Heppelmann CJ, Gazi MH, McGregor HCJ, LaBreche PT, Davies WR, Rao VP, Oi K, Tazelaar HD, Logan JS, McGregor CGA. Proteomic identification of non-Gal antibody targets after pig-to-primate cardiac xenotransplantation. Xenotransplantation 2009; 15:268-76. [PMID: 18957049 DOI: 10.1111/j.1399-3089.2008.00480.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Experience with non-antigenic galactose alpha1,3 galactose (alphaGal) polymers and development of alphaGal deficient pigs has reduced or eliminated the significance of this antigen in xenograft rejection. Despite these advances, delayed xenograft rejection (DXR) continues to occur most likely due to antibody responses to non-Gal endothelial cell (EC) antigens. METHODS To gauge the diversity of the non-Gal antibody response we used antibody derived from CD46 transgenic heterotopic cardiac xenografts performed without T-cell immunosuppression, Group A (n = 4) and Gal knockout (GT-KO) heart transplants under tacrolimus and sirolimus immunosuppression, Group B (n = 8). Non-Gal antibody was measured by flow cytometry and by western blots using GT-KO EC membrane antigens. A nanoLC/MS/MS analysis of proteins recovered from 2D gels was used to identify target antigens. RESULTS Group A recipients exhibited a mixed cellular and humoral rejection. Group B recipients mainly exhibited classical DXR. Western blot analysis showed a non-Gal antibody response induced by GT+ and GT-KO hearts to an overlapping set of pig aortic EC membrane antigens. Proteomic analysis identified 14 potential target antigens but failed to define several immunodominant targets. CONCLUSIONS These experiments indicate that the non-Gal antibody response is directed to a number of stress response and inflammation related pig EC antigens and a few undefined targets. Further analysis of these antibody specificities using alternative methods is required to more fully define the repertoire of non-Gal antibody responses.
Collapse
Affiliation(s)
- Guerard W Byrne
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Cozzi E, Bosio E, Seveso M, Rubello D, Ancona E. Xenotransplantation as a model of integrated, multidisciplinary research. Organogenesis 2009; 5:288-96. [PMID: 19568350 PMCID: PMC2659370 DOI: 10.4161/org.7578] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 11/19/2008] [Indexed: 11/19/2022] Open
Abstract
Xenotransplantation was proposed a long time ago as a possible solution to the world-wide shortage of human organs. For years, researchers in this field have almost exclusively directed their efforts towards combating the immunological barrier that precluded long-term xenograft survival. Studies have been conducted in both small and large animal models and the most relevant results have been obtained in pre-clincal studies, specifically those utilising the pig-to-nonhuman primate combination. In this context, a better understanding of the immunological mechanisms underlying the rejection of a xenograft have allowed the identification of specific targets of intervention that have resulted in considerable improvements in survival of porcine organs or cells in nonhuman primates. However it has also become apparent that if xenotransplantation has to enter the clinical arena, a multidisciplinary approach will be needed to comprehensively tackle the different issues related to the use of a xenograft to cure human disease.In this regard, the safety, ethics and regulatory aspects of xenotransplantation are currently being aggressively addressed to enable the initiation of xenotransplantation with a favourable risk/benefit ratio.
Collapse
Affiliation(s)
- Emanuele Cozzi
- Direzione Sanitaria; Padua General Hospital; Padua, Italy; Department of Surgical and Gastroenterological Sciences; University of Padua; Padua, Italy; CORIT (Consorzio per la Ricerca sul Trapianto d'Organi); Padua, Italy; Department of Nuclear Medicine; PET Centre; S. Maria della Misericordia Hospital; Rovigo, Italy; Clinica Chirurgica III; Padua General Hospital; Padua, Italy
| | - Erika Bosio
- Direzione Sanitaria; Padua General Hospital; Padua, Italy; Department of Surgical and Gastroenterological Sciences; University of Padua; Padua, Italy; CORIT (Consorzio per la Ricerca sul Trapianto d'Organi); Padua, Italy; Department of Nuclear Medicine; PET Centre; S. Maria della Misericordia Hospital; Rovigo, Italy; Clinica Chirurgica III; Padua General Hospital; Padua, Italy
| | - Michela Seveso
- Direzione Sanitaria; Padua General Hospital; Padua, Italy; Department of Surgical and Gastroenterological Sciences; University of Padua; Padua, Italy; CORIT (Consorzio per la Ricerca sul Trapianto d'Organi); Padua, Italy; Department of Nuclear Medicine; PET Centre; S. Maria della Misericordia Hospital; Rovigo, Italy; Clinica Chirurgica III; Padua General Hospital; Padua, Italy
| | - Domenico Rubello
- Direzione Sanitaria; Padua General Hospital; Padua, Italy; Department of Surgical and Gastroenterological Sciences; University of Padua; Padua, Italy; CORIT (Consorzio per la Ricerca sul Trapianto d'Organi); Padua, Italy; Department of Nuclear Medicine; PET Centre; S. Maria della Misericordia Hospital; Rovigo, Italy; Clinica Chirurgica III; Padua General Hospital; Padua, Italy
| | - Ermanno Ancona
- Direzione Sanitaria; Padua General Hospital; Padua, Italy; Department of Surgical and Gastroenterological Sciences; University of Padua; Padua, Italy; CORIT (Consorzio per la Ricerca sul Trapianto d'Organi); Padua, Italy; Department of Nuclear Medicine; PET Centre; S. Maria della Misericordia Hospital; Rovigo, Italy; Clinica Chirurgica III; Padua General Hospital; Padua, Italy
| |
Collapse
|
56
|
Abstract
PURPOSE OF REVIEW We review recent insights into the mechanisms and prevalence of accommodation. Accommodation refers to an acquired resistance of an organ graft to humoral injury and rejection. RECENT FINDINGS Accommodation has been postulated to reflect changes in antibodies, control of complement and/or acquired resistance to injury by antibodies, complement or other factors. We discuss the importance of these mechanisms, highlighting new conclusions. SUMMARY Accommodation may be a common, perhaps the most common, outcome of organ transplantation and, in some systems, a predictable outcome of organ xenotransplantation. Further understanding of how accommodation is induced and by what mechanisms it is manifest and maintained could have a profound impact on transplantation in general and perhaps on other fields.
Collapse
Affiliation(s)
- Raymond J Lynch
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
57
|
Han K, Lee JE, Kwon SJ, Park SY, Shim SH, Kim H, Moon JH, Suh CS, Lim HJ. Human amnion-derived mesenchymal stem cells are a potential source for uterine stem cell therapy. Cell Prolif 2008; 41:709-25. [PMID: 18823496 DOI: 10.1111/j.1365-2184.2008.00553.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Human amnion is an easy-to-obtain novel source of human mesenchymal stem cells, which poses little or no ethical dilemmas. We have previously shown that human amnion-derived mesenchymal (HAM) cells exhibit certain mesenchymal stem cell-like characteristics with respect to expression of stem cell markers and differentiation potentials. MATERIALS AND METHODS In this study, we further characterized HAM cells' potential for in vivo therapeutic application. RESULTS Flow cytometric analyses of HAM cells show that they express several stem cell-related cell surface markers, including CD90, CD105, CD59, CD49d, CD44 and HLA-ABC, but not CD45, CD34, CD31, CD106 or HLA-DR. HAM cells at the 10th passage showed normal karyotype. More interestingly, the AbdB-like HOXA genes HOXA9, HOXA10 and HOXA11 that are expressed in the mesenchyme of the developing female reproductive tract and pregnant uteri are also expressed in HAM cells, suggesting similarities between these two mesenchymal cell types. Progesterone receptor is also highly expressed in HAM cells and expression of genes or proteins in HAM cells could be manipulated with the aid of lentivirus technology or cell-permeable peptides. To test potentials of HAM cells for in vivo application, we introduced enhanced green fluorescence protein (EGFP)-expressing HAM cells to mice by intrauterine infusion (into uteri) or by intravenous injection (into the circulation). Presence of EGFP-expressing cells within the uterine mesenchyme after intrauterine infusion or in lungs after intravenous injection was noted within 1-4 weeks. CONCLUSIONS Collectively, these results suggest that HAM cells are a potential source of mesenchymal stem cells with therapeutic potential.
Collapse
Affiliation(s)
- K Han
- Department of Biomedical Science and Technology, Research Center for Drugs, Institute of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Low molecular weight dextran sulfate as complement inhibitor and cytoprotectant in solid organ and islet transplantation. Mol Immunol 2008; 45:4084-94. [DOI: 10.1016/j.molimm.2008.07.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Accepted: 07/15/2008] [Indexed: 11/19/2022]
|
59
|
Saethre M, Schneider MKJ, Lambris JD, Magotti P, Haraldsen G, Seebach JD, Mollnes TE. Cytokine secretion depends on Galalpha(1,3)Gal expression in a pig-to-human whole blood model. THE JOURNAL OF IMMUNOLOGY 2008; 180:6346-53. [PMID: 18424758 DOI: 10.4049/jimmunol.180.9.6346] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Transplants from alpha1,3-galactosyltransferase (Gal) gene-knockout pigs to nonhuman primates are largely protected from hyperacute but not acute humoral xenograft rejection. The present study investigates the role of Gal in cytokine responses using a novel pig-to-human whole blood in vitro model, developed for species-specific analysis of porcine and human cytokines. Porcine (n = 7) and human (n = 27) cytokines were measured using ELISA or multiplex technology, respectively. Porcine aortic endothelial cells from control (Gal(+/+)) and Gal-deficient (Gal(-/-)) pigs were incubated with human lepirudin anticoagulated whole blood from healthy donors. E-selectin expression was measured by flow cytometry. The C3 inhibitor compstatin and a C5aR antagonist were used to study the role of complement. Cytokine species specificity was documented, enabling detection of 2 of 7 porcine cytokines and 13 of 27 human cytokines in one single sample. Gal(+/+) porcine aortic endothelial cells incubated with human whole blood showed a marked complement C5b-9 dependent up-regulation of E-selectin and secretion of porcine IL-6 and IL-8. In contrast, Gal(-/-) cells responded with E-selectin and cytokine expression which was so weak that the role of complement could not be determined. Human IL-6, IL-8, IFN-gamma, MIP-1alpha, MIP-1beta, eotaxin, and RANTES were detected in the Gal(+/+) system, but virtually no responses were seen in the Gal(-/-) system (p = 0.03). The increase in human cytokine release was largely complement dependent and, in contrast to the porcine response, mediated through C5a. Species-specific analysis of cytokine release revealed a marked, complement-dependent response when Gal(+/+) pig cells were incubated with human whole blood, compared with Gal(-/-) cells which induced virtually no cytokine release.
Collapse
Affiliation(s)
- Marit Saethre
- Institute of Immunology, Rikshospitalet University Hospital and Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
60
|
González-Stawinski GV, Tan CD, Smedira NG, Starling RC, Rodríguez ER. Decay-accelerating Factor Expression May Provide Immunoprotection Against Antibody-mediated Cardiac Allograft Rejection. J Heart Lung Transplant 2008; 27:357-61. [DOI: 10.1016/j.healun.2008.01.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 11/06/2007] [Accepted: 01/02/2008] [Indexed: 11/25/2022] Open
|
61
|
Petersen B, Carnwath JW, Niemann H. The perspectives for porcine-to-human xenografts. Comp Immunol Microbiol Infect Dis 2008; 32:91-105. [PMID: 18280567 DOI: 10.1016/j.cimid.2007.11.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2007] [Indexed: 10/22/2022]
Abstract
The shortage of donated human organs for transplantation continues to be a life threatening problem for patients suffering from complete organ failure. Although this gap is increasing due to the demographic changes in aging Western populations, it is generally accepted that international trading in human organ is not an ethical solution. Alternatives to the use of human organs for transplantation must be developed and these alternatives include stem cell therapy, artificial organs and organs from other species, i.e. xenografts. For practical reasons but most importantly because of its physiological similarity with humans, the pig is generally accepted as the species of choice for xenotransplantation. Nevertheless, before porcine organs can be used in human xenotransplantation, it is necessary to make a series of precise genetic modifications to the porcine genome, including the addition of genes for factors which suppress the rejection of transplanted porcine tissues and the inactivation or removal of undesirable genes which can only be accomplished at this time by targeted recombination and somatic nuclear transfer. This review will give an insight into the advances in transgenic manipulation and cloning in pigs--in the context of porcine-to-human xenotransplantation.
Collapse
Affiliation(s)
- Bjoern Petersen
- Institute of Farm Animal Genetics (FLI), Department of Biotechnology, Hoeltystrasse 10, 31535 Neustadt, Germany
| | | | | |
Collapse
|
62
|
Kirk AD, Elster EA. Immunology of Transplantation. Surgery 2008. [DOI: 10.1007/978-0-387-68113-9_81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
63
|
|
64
|
Reduced Fibrin Deposition and Intravascular Thrombosis in hDAF Transgenic Pig Hearts Perfused With Tirofiban. Transplantation 2007; 84:1667-76. [DOI: 10.1097/01.tp.0000295742.45413.dc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
65
|
Gonzalez-Stawinski GV, Cook DJ, Chui J, Gupta S, Navia JL, Hoercher K, Taylor DO, Yamani MH, Starling RC, Smedira NG. A Comparative Analysis Between Survivors and Nonsurvivors with Antibody Mediated Cardiac Allograft Rejection. J Surg Res 2007; 142:233-8. [PMID: 17727888 DOI: 10.1016/j.jss.2007.04.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 04/19/2007] [Accepted: 04/20/2007] [Indexed: 11/15/2022]
Abstract
BACKGROUND Antibody mediated rejection (AMR) is an important cause of graft loss in the post heart transplant period. The following study was conducted to determine differences between survivors and nonsurvivors who developed post heart transplant AMR. METHODS We retrospectively reviewed the charts of patients who received a heart transplant between January 1993 and December 2002. Patients with biopsy proven AMR were identified. This group was divided into survivors and nonsurvivors. Groups were compared with regards to demographics, T-cell flow panel of reactive antibodies (PRA), flow cross-matches (anti-donor HLA Class I and II), and short- and long-term outcomes. Results of endomyocardial biopsies were collected to allow calculation of sensitivity, specificity, negative- and positive predictive values as well as accuracy of immunoglobulins and complement split products in association to death. RESULTS A total of 65 patients (8.9%) were diagnosed with AMR. Mean age was 48 y (range: 8-68 y) and 53.8% were males. Episodes of hemodynamic instability associated with AMR were observed in 37% of patients. Only two deaths were directly attributed to acute AMR. Nearly 20% of AMR patients developed transplant coronary artery disease. Univariate analysis identified T-PRA (P < 0.001), mean T-cell molecules of equivalent soluble fluorochrome (MESF) (P < 0.001) and mean B-cell MESF (P < 0.001) as possible factors associated with death. Neither demographics of complement split products were associated to late death. CONCLUSION When studying patients with AMR, pretransplant T-PRA, T-cell, and B-cell MESF may identify individuals at risk of late death.
Collapse
|
66
|
Saethre M, Baumann BC, Fung M, Seebach JD, Mollnes TE. Characterization of natural human anti-non-gal antibodies and their effect on activation of porcine gal-deficient endothelial cells. Transplantation 2007; 84:244-50. [PMID: 17667817 DOI: 10.1097/01.tp.0000268815.90675.d5] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The generation of Galalpha1-3Gal (Gal) transferase deficient pigs has increased the interest in non-Gal antigens potentially representing important targets for xenoreactive antibody binding leading to xenograft rejection. The present study addressed the levels and immunoglobulin isotypes of preformed human anti-non-Gal antibodies and their potential to activate porcine endothelial cells. METHODS Porcine endothelial cells lacking the Gal epitope (Gal-/-) were used to measure immunoglobulin (Ig) M and IgG subclass anti-non-Gal antibodies, using sera from 80 blood donors and pooled human AB serum. Antibodies specific for the non-Gal Hanganutziu-Deicher (HD) xenoantigen were measured by enzyme-linked immunosorbent assay. Activation of Gal-/- and Gal+/+ endothelial cells by human serum was measured, in the presence or absence of complement inhibitors, by E-selectin cell-surface expression using flow cytometry. RESULTS Anti-non-Gal antibody levels varied considerably among individual sera and comprised approximately 10% of total anti-porcine antibodies without sex or age differences. Among the IgG subclasses only IgG1 and IgG2 were detected. Human serum-induced E-selectin expression on Gal-/- cells was less than 20% compared with Gal+/+ cells, correlated with anti-HD IgM and IgG antibody levels (P=0.027 and 0.032, respectively), and was largely complement-independent in accordance with the lack of IgG3 anti-non-Gal antibodies. In contrast, E-selectin upregulation on Gal+/+ cells was reduced in complement blocking experiments. CONCLUSION Preformed anti-non-Gal antibodies, in particular anti-HD antibodies, were present in all human sera samples, activated porcine endothelial cells, and may therefore play a role in xenograft rejection using organs from GalT-/- pigs.
Collapse
Affiliation(s)
- Marit Saethre
- Institute of Immunology, Rikshospitalet-Radiumhospitalet Medical Center and Faculty of Medicine, University of Oslo, Oslo, Norway.
| | | | | | | | | |
Collapse
|
67
|
Cozzi E, Seveso M, Hutabba S, Fabris S, Cavicchioli L, Ancona E. An Update on Xenotransplantation. Vet Res Commun 2007; 31 Suppl 1:15-25. [PMID: 17682842 DOI: 10.1007/s11259-007-0002-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Xenotransplantation is one of the possible avenues currently being explored to address the shortage problem of human organs. With this in mind, this article will briefly review the current situation with respect to the immunological, physiological and biosafety aspects related to the transplantation of pig organs into primates. Acute humoral xenograft rejection (AHXR) currently remains the central immunological obstacle and the development of strategies for both a better control of the elicited anti-pig humoral immune response or the prevention of the onset of coagulation disorders that accompany AHXR are the two primary focuses of research. To date, porcine xenografts have been shown to sustain the life of nonhuman primates for several months. Such preclinical studies have also demonstrated the absence of insurmountable physiological incompatibilities between pig and primate. In addition, reassuring findings regarding biosafety aspects have been generated and pro-active research aimed at the identification of an organ source with a higher safety profile is also underway. These advancements, in conjunction with ongoing research in pig genetic engineering, immunosuppression and tolerance are expected to further extend the survival of porcine xenografts transplanted into primates. However, until further physiological, efficacy and safety data are generated in relevant primate models, clinical xenotransplantation should not be considered.
Collapse
Affiliation(s)
- E Cozzi
- CORIT (Consorzio per la Ricerca sul Trapianto d'Organi), Padua, Italy.
| | | | | | | | | | | |
Collapse
|
68
|
Abstract
The innate immune system not only participates in host defence but also contributes to the control of adaptive immune responses. Complement and Toll-like receptors (TLR) are key components of innate immunity. Emerging evidence suggests their activation is involved in all major aspects of transplantation. This paper reviews the current understanding of how the complement and TLR on impact transplant injury.
Collapse
Affiliation(s)
- Tao Lin
- Department of Nephrology and Transplantation, King's College London School of Medicine at Guy's, King's College and St Thomas' Hospitals, London, UK
| | | | | |
Collapse
|
69
|
Abstract
Cell surface proteins containing covalently linked lipids associate with specialized membrane domains. Morphogens like Hedgehog and Wnt use their lipid anchors to bind to lipoprotein particles and employ lipoproteins to travel through tissues. Removal of their lipid anchors or decreasing lipoprotein levels give rise to adverse Hedgehog and Wnt signaling. Some parasites can also transfer their glycosylphosphatidylinositol-anchored surface proteins to host lipoprotein particles. These antigen-loaded lipoproteins spread throughout the circulation, and probably hamper an adequate immune response by killing neutrophils. Together, these findings imply a widespread role for lipoproteins in intercellular transfer of lipid-anchored surface proteins, and may have various physiological consequences. Here, we discuss how lipid-modified proteins may be transferred to and from lipoproteins at the cellular level.
Collapse
Affiliation(s)
- Sylvia Neumann
- Department of Membrane Enzymology, Bijvoet Center and Institute of Biomembranes, Utrecht University, Utrecht, The Netherlands
| | | | | |
Collapse
|
70
|
Horton PJ, Chaudhury P, Rochon C, Metrakos P, Tchervenkov J. Should trials of liver xenotransplantation proceed in toxic fulminant hepatic failure? Xenotransplantation 2006; 13:483. [PMID: 17059571 DOI: 10.1111/j.1399-3089.2006.00337.x] [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: 11/28/2022]
|
71
|
|
72
|
Black SM, Grehan JF, Rivard AL, Benson BA, Wahner AE, Koch AE, Levay-Young BK, Dalmasso AP. Porcine Endothelial Cells and Iliac Arteries Transduced with AdenoIL-4 Are Intrinsically Protected, through Akt Activation, against Immediate Injury Caused by Human Complement. THE JOURNAL OF IMMUNOLOGY 2006; 177:7355-63. [PMID: 17082655 DOI: 10.4049/jimmunol.177.10.7355] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Vascular endothelial cells (ECs) can be injured in a variety of pathologic processes that involve activated complement. We reported previously that porcine ECs incubated with exogenous IL-4 or IL-13 are protected from cytotoxicity by human complement and also from apoptosis by TNF-alpha. The resistance to complement consists of an intrinsic mechanism that is lost a few days after cytokine removal. In our current study, we investigated whether transfer of the IL-4 gene into porcine ECs in vitro and into porcine vascular tissues in vivo would induce efficient and durable protection from human complement. We found that ECs transduced with adenoIL-4 or adenoIL-13 exhibited continuous production of the cytokine and prolonged protection from complement-mediated killing. IL-4 also protected ECs from activation: ECs incubated with IL-4 did not develop cell retraction and intercellular gaps upon stimulation with sublytic complement. The endothelium and subendothelium of pig iliac arteries that were transduced with the IL-4 gene were effectively protected from complement-dependent immediate injury after perfusion with human blood. However, after similar perfusion, the endothelium was immediately lost from arteries that were transduced with a control adenovirus. The protection was not due to up-regulation of the complement regulators decay accelerating factor, membrane cofactor protein, and CD59, or to reduced complement activation, but required the participation of Akt. Although our studies model protection in pig-to-primate xenotransplantation, our findings of IL-4 induction of Akt-mediated protection may be more broadly applicable to EC injury as manifested in ischemia-reperfusion, allotransplantation, and various vascular diseases.
Collapse
Affiliation(s)
- Sylvester M Black
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Manzi L, Montaño R, Abad MJ, Arsenak M, Romano E, Taylor P. Expression of human soluble complement receptor 1 by a pig endothelial cell line inhibits lysis by human serum. Xenotransplantation 2006; 13:75-9. [PMID: 16497215 DOI: 10.1111/j.1399-3089.2005.00265.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The importance of complement activation and naturally occurring anti-pig antibodies in the hyperacute rejection (HAR) observed in models of pig-to-human xenotransplantation is well established. To overcome this, much effort has been dedicated to preparing transgenic pigs by knocking out Galalpha(1-3)Gal expression in these animals, or knocking in the expression of human complement regulatory proteins (CRPs), such as CD59 or decay accelerating factor. A soluble form of another membrane CRP, complement receptor type 1 (CR1), has also been shown to inhibit complement activation. Here, we show that transfection of a pig endothelial cell line with a truncated form of human soluble complement receptor 1 (sCR1) almost completely protected these cells from complement-mediated lysis by human AB serum. Pigs genetically manipulated to express human sCR1 may represent an additional strategy to inhibit HAR of pig-to-human transplanted organs.
Collapse
Affiliation(s)
- Lorna Manzi
- Laboratory of Cellular and Molecular Pathology, Centre for Experimental Medicine, Venezuelan Institute for Scientific Research (IVIC), Caracus, Venezuela
| | | | | | | | | | | |
Collapse
|
74
|
Davila E, Byrne GW, LaBreche PT, McGregor HCJ, Schwab AK, Davies WR, Rao VP, Oi K, Tazelaar HD, Logan JS, McGregor CGA. T-cell responses during pig-to-primate xenotransplantation. Xenotransplantation 2006; 13:31-40. [PMID: 16497210 DOI: 10.1111/j.1399-3089.2005.00258.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
UNLABELLED Xenotransplantation using porcine organs may resolve a chronic shortage of donor organs for clinical transplantation if significant immunological barriers can be overcome. To determine the potential role of T lymphocytes in Xenograft (Xg) rejection, we transplanted transgenic hCD46 porcine hearts heterotopically into baboon recipients. METHODS Recipients were treated to deplete anti-Gal antibody with a non-antigenic alpha-Gal polyethylene glycol polymer (TPC) (n = 2), TPC plus rituximab (anti-CD20) (n = 1) or were untreated (n = 1). None of the recipients received T-cell immunosuppression. RESULTS All Xgs failed within 7 days and showed evidence of a mixed humoral and cellular rejection process. Cellular infiltration consisting primarily of CD4+ T cells and few CD8+ T cells. Proliferation and cytotoxicity assays showed sensitization of CD4+ and CD8+ T cells that reacted with porcine IFN-gamma (pIFN-gamma)-stimulated porcine aortic endothelial cells (PAEC). The CD4+ lymphocytes displayed greater cytotoxicity than CD8+ cells. An increased frequency of PAEC-specific interleukin (IL) 2 and IFN-gamma-secreting T cells was observed, suggesting a Th1 cytokine bias. An increase in the percentage of circulating CD4+CD28- cells was observed at the time of rejection and over 50% of the CD4+ cells recovered from residual pig tissue at necropsy lacked CD28 expression. CONCLUSIONS These findings show that lymphocytes are efficiently stimulated by PAEC antigens and can mediate direct tissue destruction. These studies (1) provide an insight into the potential of cellular-mediated cardiac Xg rejection, (2) show for the first time the induction of cytotoxic pig-specific CD4+CD28- lymphocytes and (3) provide a rational basis for determining different modes of immunosuppression to treat Xg rejection.
Collapse
Affiliation(s)
- Eduardo Davila
- Department of Pediatrics, Louisiana State University Health Science Center, Stanley S. Scott Cancer Center, New Orleans, LA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Miyagawa S, Fukuta D, Kitano E, Kobayashi C, Fumimoto Y, Shirasu A, Hattori H, Shirakura R, Fukuzawa M. Effect of tandem forms of DAF(CD55) on complement-mediated xenogeneic cell lysis. Xenotransplantation 2006; 13:433-9. [PMID: 16925667 DOI: 10.1111/j.1399-3089.2006.00331.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND It is difficult to produce a transgenic animal with high expression of decay-accelerating factor (CD55: DAF) or other molecules. The purpose of this study was to assess the effect of tandem forms of DAF on a xenogeneic cell membrane against human complement. METHODS cDNAs of the delta-Short Consensus Repeat (SCR) 1-DAF, the double-DAF, the triple-DAF, and the tetra-DAF with a FLAG-tag were established. Chinese hamster ovary (CHO) cell lines and a pig endothelial cell (PEC) line expressing these molecules were established. The amelioration of complement-mediated lysis by the transfectant molecules on these cells was examined. The CHO cell transfectants were also incubated with normal human serum, and the amount of C3 deposited was determined by FACS analysis. RESULTS Stable CHO cells and PEC transfectants, in which each molecule was clearly expressed, and Western blots showed that each band corresponded to the expected molecular weight. The extent of amelioration of complement-mediated lysis by these four molecules was then examined. A clear tendency was found, as follows: The higher the tandem number of DAF, the greater was the effect on cytotoxicity. Additional experiments focusing on triple-DAF and tetra-DAF did not indicate any significant difference in complement-mediated lysis. Consistent with the complement-regulatory ability, the inhibitory effect of the deposition of C3 fragments by these molecules was closely related to the degree of amelioration. CONCLUSION These data indicate that tandem DAF, especially a triple-DAF, is a very effective form for protecting against complement activation.
Collapse
Affiliation(s)
- Shuji Miyagawa
- Division of Organ Transplantation, Department of Molecular Therapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Wang J, Zhang B, Fang J, Sujino K, Li H, Otter A, Hindsgaul O, Palcic MM, Wang PG. Frontal Affinity Chromatography Coupled to Mass Spectrometry: An Effective Method for KdDetermination and Screening of α‐Gal Derivatives Binding to Anti‐Gal Antibodies (IgG). J Carbohydr Chem 2006. [DOI: 10.1081/car-120025323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Jianqiang Wang
- a Department of Chemistry , University of Alberta , Edmonton, Alberta, T6G 2G2, Canada
- b Department of Chemistry , Wayne State University , Detroit, Michigan, 48202, USA
- c Triad Therapeutics, Inc. , 9381 Judicial Drive, San Diego, California, 92121, USA
| | - Boyan Zhang
- a Department of Chemistry , University of Alberta , Edmonton, Alberta, T6G 2G2, Canada
| | - Jianwen Fang
- b Department of Chemistry , Wayne State University , Detroit, Michigan, 48202, USA
| | - Keiko Sujino
- a Department of Chemistry , University of Alberta , Edmonton, Alberta, T6G 2G2, Canada
| | - Hong Li
- a Department of Chemistry , University of Alberta , Edmonton, Alberta, T6G 2G2, Canada
| | - Albin Otter
- a Department of Chemistry , University of Alberta , Edmonton, Alberta, T6G 2G2, Canada
| | - Ole Hindsgaul
- a Department of Chemistry , University of Alberta , Edmonton, Alberta, T6G 2G2, Canada
| | - Monica M. Palcic
- a Department of Chemistry , University of Alberta , Edmonton, Alberta, T6G 2G2, Canada
| | - Peng George Wang
- b Department of Chemistry , Wayne State University , Detroit, Michigan, 48202, USA
| |
Collapse
|
77
|
Lee JH, Lee HJ, Nahm KM, Jeon HY, Hwang WS, Paik NW, Rho HM. Effects of combined expression of human complement regulatory proteins and H-transferase on the inhibition of complement-mediated cytolysis in porcine embryonic fibroblasts. Transplant Proc 2006; 38:1618-21. [PMID: 16797369 DOI: 10.1016/j.transproceed.2006.02.129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Indexed: 11/19/2022]
Abstract
The expression of human complement regulatory proteins (CRP) and H-transferase (HT) in porcine cells is one of the strategies for suppression of hyperacute rejection (HAR) of xenotransplants in human recipients. In this study, we investigated the inhibitory effect of combined expression of human complement regulators and HT on human serum-mediated cytolysis in porcine embryonic fibroblasts. For the combinated expression of human CRPs in transformed pig cells, cDNAs of human DAF, MCP, and CD59 were cloned into the same insertional plasmid under the control of pCMV IE and LTR. The double combination of CRPs, hDAF-hMCP, and hMCP-hCD59 survived over 50% in the presence of 50% human serum, compared to the control. Moreover, the cell viability was increased more than 65% and 80% in the combination of human DAF-CD59 and DAF-MCP-CD59, respectively. In addition, the combination of HT gene to hDAF-hCD59 vector increased the viability close to 80%, similar to the triple combination of CRPs. These observations suggest that the combined expression of human CRPs and HT in the same insertional vector may be more effective in protecting porcine cells from human complement-mediated cytolysis.
Collapse
Affiliation(s)
- J H Lee
- Indang Institute of Molecular Biology, Inje University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
78
|
Saethre M, Lea T, Borgen MS, Fiane AE, Michaelsen TE, Thorsby E, Haraldsen G, Mollnes TE. Human complement-activating immunoglobulin (Ig)G3 antibodies are essential for porcine endothelial cell activation. Xenotransplantation 2006; 13:215-23. [PMID: 16756564 DOI: 10.1111/j.1399-3089.2006.00289.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Complement-activating naturally occurring anti-porcine endothelial cell antibodies (Abs) are responsible for hyperacute rejection in porcine-to-primate transplantation, whereas the role of complement in acute vascular rejection, characterized by type II endothelial cell activation, is less well understood. We previously demonstrated a correlation between porcine type II endothelial cell activation, as detected by E-selectin expression, and human immunoglobulin (Ig)G3 anti-Gal alpha1-3Gal (Gal) Abs, which was not seen for IgG1, IgG2 or IgG4. The present study was undertaken to investigate whether there is a causal relationship between human anti-porcine IgG3 Abs and porcine endothelial cell activation. METHODS IgG3 was isolated employing a Protein A column to 98.3% purity. Porcine endothelial cells were incubated with isolated human IgG3 or the combination of IgG1, IgG2 and IgG4. E-selectin expression and complement activation were investigated by flow cytometry and Western blotting, respectively. RESULTS Purified IgG3, in contrast to the other IgG subclasses, induced a substantial increase in E-selectin expression. This activation was accompanied by complement activation as detected by C3 cleavage, and was abolished by heat inactivation or by adding the complement inhibitor FUT-175. Depletion of anti-Gal Abs reduced E-selectin expression by 60%, consistent with the presence of complement-activating anti-porcine non-Gal Abs of the IgG3 subclass. CONCLUSIONS Collectively, these data strengthen the hypothesis that human anti-porcine endothelial cell Abs of the IgG3 subclass are essential for endothelial cell activation in porcine-to-human species grafts and demonstrate such activation to be partly independent of Gal epitopes.
Collapse
Affiliation(s)
- Marit Saethre
- Institute of Immunology, Rikshospitalet University Hospital, University of Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Sprong H, Suchanek M, van Dijk SM, van Remoortere A, Klumperman J, Avram D, van der Linden J, Leusen JHW, van Hellemond JJ, Thiele C. Aberrant receptor-mediated endocytosis of Schistosoma mansoni glycoproteins on host lipoproteins. PLoS Med 2006; 3:e253. [PMID: 16942390 PMCID: PMC1502155 DOI: 10.1371/journal.pmed.0030253] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Accepted: 04/04/2006] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Bilharzia is one of the major parasitic infections affecting the public health and socioeconomic circumstances in (sub) tropical areas. Its causative agents are schistosomes. Since these worms remain in their host for decades, they have developed mechanisms to evade or resist the immune system. Like several other parasites, their surface membranes are coated with a protective layer of glycoproteins that are anchored by a lipid modification. METHODS AND FINDINGS We studied the release of glycosyl-phosphatidylinositol (GPI)-anchored proteins of S. mansoni and found them in the circulation associated with host lipoprotein particles. Host cells endocytosed schistosomal GPI-anchored proteins via their lipoprotein receptor pathway, resulting in disturbed lysosome morphology. In patients suffering from chronic schistosomiasis, antibodies attacked the parasite GPI-anchored glycoproteins that were associated with the patients' own lipoprotein particles. These immunocomplexes were endocytosed by cells carrying an immunoglobulin-Fc receptor, leading to clearance of lipoproteins by the immune system. As a consequence, neutral lipids accumulated in neutrophils of infected hamsters and in human neutrophils incubated with patient serum, and this accumulation was associated with apoptosis and reduced neutrophil viability. Also, Trypanosoma brucei, the parasite that causes sleeping sickness, released its major GPI-anchored glycoprotein VSG221 on lipoprotein particles, demonstrating that this process is generalizable to other pathogens/parasites. CONCLUSIONS Transfer of parasite antigens to host cells via host lipoproteins disrupts lipid homeostasis in immune cells, promotes neutrophil apoptosis, may result in aberrant antigen presentation in host cells, and thus cause an inefficient immune response against the pathogen.
Collapse
Affiliation(s)
- Hein Sprong
- Department of Membrane Enzymology, Bijvoet Center, Utrecht University, Utrecht, Netherlands
- * To whom correspondence should be addressed. E-mail: (HS); (CT)
| | - Monika Suchanek
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Suzanne M van Dijk
- Department of Membrane Enzymology, Bijvoet Center, Utrecht University, Utrecht, Netherlands
- Department of Cell Biology, Utrecht University, Utrecht, Netherlands
| | | | - Judith Klumperman
- Department of Cell Biology, Utrecht University, Utrecht, Netherlands
| | - Diana Avram
- Department of Biochemistry of Lipids, Utrecht University, Utrecht, Netherlands
| | - Joke van der Linden
- Immunotherapy Laboratory, Department of Immunology, University Medical Center Utrecht, Netherlands
| | - Jeanette H. W Leusen
- Immunotherapy Laboratory, Department of Immunology, University Medical Center Utrecht, Netherlands
| | - Jaap J van Hellemond
- Department of Biochemistry and Cell Biology, Institute of Biomembranes, Utrecht University, Utrecht, Netherlands
| | - Christoph Thiele
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * To whom correspondence should be addressed. E-mail: (HS); (CT)
| |
Collapse
|
80
|
Moore DJ, Markmann JF, Deng S. Avenues for immunomodulation and graft protection by gene therapy in transplantation. Transpl Int 2006; 19:435-45. [PMID: 16771864 DOI: 10.1111/j.1432-2277.2006.00314.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Organ transplantation represents the only definitive therapy for many causes of end-organ failure. However, the universal success of this therapy is limited by chronic allograft rejection, the side effects of chronic immunosuppressive therapy, and a severe shortage of donor organs. Presently, the success of solid-organ transplantation depends on the continuous administration of toxic and nonspecific immunosuppressive agents, therapies that present risks for opportunistic infection, malignancy, and a variety of agent-specific side effects. To promote the use of transplantation with limited risk of long-term sequelae, three dominant research challenges emerge: (i) elimination of the need for exogenous immunosuppression by immunological tolerance induction; (ii) prevention of chronic rejection/graft dysfunction; and (iii) expansion of available organs for transplantation. Gene therapy may provide significant advances and solutions in each of these areas. Rejection of the graft in the immediate post-transplant period has been attacked through the transfer of immunomodulatory molecules in addition to tolerance inducing approaches. Chronic graft rejection may be similarly addressed through permanent tolerance induction or alternatively through the introduction of molecules to resist chronic graft damage. Genetic manipulation of stem cells may ultimately produce transgenic animals to serve as tissue donors to overcome the limited donor organ supply. This review will highlight ongoing developments in the translation of gene therapy approaches to the challenges inherent in transplantation.
Collapse
Affiliation(s)
- Daniel J Moore
- Department of Pediatrics, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
81
|
Gaca JG, Appel JZ, Lukes JG, Gonzalez-Stawinski GV, Lesher A, Palestrant D, Logan JS, Love SD, Holzknecht ZE, Platt JL, Parker W, Davis RD. Effect of an anti-C5a monoclonal antibody indicates a prominent role for anaphylatoxin in pulmonary xenograft dysfunction. Transplantation 2006; 81:1686-94. [PMID: 16794535 DOI: 10.1097/01.tp.0000226063.36325.02] [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] [Indexed: 10/24/2022]
Abstract
BACKGROUND In contrast to renal or cardiac xenografts, the inhibition of complement using cobra venom factor (CVF) accelerates pulmonary xenograft failure. By activating C3/C5 convertase, CVF depletes complement while additionally generating C5a and other anaphylatoxins, to which pulmonary xenografts may be uniquely susceptible. The current study investigates the role of C5a in pulmonary xenograft failure in baboons. METHODS Left orthotopic pulmonary xenografts using swine lungs expressing human CD46 were performed in baboons receiving: I) no other treatment (n=4), II) immunodepletion (n=5), and III) immunodepletion plus a single dose of mouse anti-human C5a monoclonal antibody (anti-C5a, 0.6 mg/kg administered intravenously) (n=3). The extent to which anti-C5a inhibits baboon C5a was assessed in vitro using a hemolytic reaction involving baboon serum and porcine red blood cells and by ELISA. RESULTS Baboons in Group III exhibited significantly prolonged xenograft survival (mean=722+/-121 min, P=0.02) compared to baboons in Group I (mean=202+/-24 min) and Group II (mean=276+/-79 min). Furthermore, baboons in Groups I and II experienced pronounced hemodynamic compromise requiring inotropic support whereas those in Group III remained hemodynamically stable throughout experimentation without the need for additional pharmacologic intervention. CONCLUSIONS These findings indicate that C5a exacerbates pulmonary xenograft injury and compromises recipient hemodynamic status. Moreover, blockade of anaphylatoxins, such as C5a, offers a promising approach for future investigations aimed at preventing pulmonary xenograft injury in baboons.
Collapse
Affiliation(s)
- Jeffrey G Gaca
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Weaver JG, McGregor CG, Tazelaar HD, Badley AD. Rejection severity directly correlates with myocyte apoptosis in pig-to-baboon cardiac xenotransplantation. J Heart Lung Transplant 2006; 24:841-7. [PMID: 15982611 PMCID: PMC1282520 DOI: 10.1016/j.healun.2004.05.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Revised: 05/16/2004] [Accepted: 05/17/2004] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The process by which cardiac myocytes die during xenograft rejection is incompletely understood. The presence of cardiac myocyte apoptosis in discordant xenotransplant models has been noted, yet no investigators have examined whether a relationship between myocyte apoptosis and rejection severity exists. Thus, we chose to further investigate this observation. METHODS Eight explanted pig-to-baboon cardiac grafts with varying severities of rejection, as determined by hematoxylin and eosin histology, were examined for apoptosis by transmission electron microscopy (TEM) and TUNEL (terminal deoxynucleotide transferase-mediated digoxigenin-dUTP nick-end labeling) immunohistochemistry. In addition, Western blot analysis for the cleavage of the apoptosis regulatory proteins pro-caspase 8 and 3 was performed. RESULTS Transmission electron microscopy revealed that a severely rejected graft displayed widespread condensation of nuclear chromatin, which is a characteristic morphologic feature of apoptosis. TUNEL staining verified this observation and allowed for the quantification of myocyte apoptosis in each graft. Subsequent linear regression analysis of the extent of myocyte apoptosis and rejection severity revealed a direct correlation (R(2)=0.757, p=0.005). In addition, Western blot analysis demonstrated that myocyte apoptosis involves the cleavage of pro-caspase 8 and 3. CONCLUSIONS Myocyte death in rejecting pig-to-baboon cardiac xenografts occurs through an apoptotic pathway and directly correlates with the severity of graft rejection. Further studies aimed at elucidating the apoptotic stimulus are therefore warranted. Moreover, our data suggest that antiapoptotic strategies may be of benefit in the treatment of xenograft rejection.
Collapse
Affiliation(s)
- Joel G.R. Weaver
- From the Division of Infectious Diseases
- Division of General Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | - Andrew D. Badley
- From the Division of Infectious Diseases
- Program in Translational Immunovirology and Biodefense, Mayo Clinic and Foundation, Rochester, Minnesota; and the
- Reprint requests: Andrew D. Badley, MD, FRCPC, Division of Infectious Diseases, Translational Program in Immunovirology and Biodefense, Mayo Clinic, 200 First Street NW, Rochester, MN 55905. Telephone: 507-284-3747. Fax: 507-284-3757. E-mail:
| |
Collapse
|
83
|
Shimizu A, Yamada K. Pathology of renal xenograft rejection in pig to non-human primate transplantation. Clin Transplant 2006; 20 Suppl 15:46-52. [PMID: 16848876 DOI: 10.1111/j.1399-0012.2006.00550.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Xenotransplantation has the potential to alleviate the critical shortage of organs for transplantation in humans. Miniature swine are a promising donor species for xenotransplantation. However, when swine organs are transplanted into primates, hyperacute rejection (HAR), acute humoral xenograft rejection (AHXR), acute cellular xenograft rejection (ACXR), and chronic xenograft rejection prevent successful engraftment. Developing a suitable regimen for preventing xenograft rejection requires the ability to accurately diagnosis the severity and type of rejection in the graft. For this purpose, histopathology remains the most definitive and reliable tool. We discuss here the characteristic features of xenograft rejection in a preclinical pig-to-non-human primate transplantation model. In miniature swine to baboon xenotransplantation, marked interstitial hemorrhage develops in HAR, and renal microvascular injury develops with multiple platelet-fibrin microthrombi in both HAR and AHXR. T-cell-mediated cellular immunity plays an important role in ACXR. Chronic humoral and cellular rejection may induce chronic xenograft rejection, and will be a major cause of graft loss in discordant xenotransplantation.
Collapse
Affiliation(s)
- Akira Shimizu
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
84
|
Abstract
The transplantation of animal organs into humans as a way of treating organ failure has been pursued for 100 years. Clinical xenotransplantation, as such, has always failed because the transplanted organ is rejected by the recipient. Recent advances in transplant immunology have revealed some mechanisms underlying the rejection of xenografts, and these discoveries have sparked efforts to use genetic engineering of animals and therapeutics directed at the recipient to overcome this problem. This paper reviews the current understanding of the mechanisms of xenograft rejection and efforts to overcome rejection and other hurdles.
Collapse
Affiliation(s)
- Robert Zhong
- Department of Surgery, University of Western Ontario, London, Ontario and
| | - Jeffrey L. Platt
- Transplantation Biology and the Departments of Surgery, Immunology and Pediatrics Mayo Clinic College of Medicine, Rochester, Minnesota
- Address correspondence to: Jeffrey L. Platt, M.D., Mayo Clinic, Transplantation Biology, 200 First Street SW, Medical Science 2-66, Rochester, MN 55905, Phone: (507) 538-0313, Fax: (507) 284-4957, E-mail:
| |
Collapse
|
85
|
Affiliation(s)
- Anthony J F d'Apice
- Immunology Research Centre, St Vincent's Health, Fitzroy, Vic., Australia. @svhm.org.au
| |
Collapse
|
86
|
Qin X, Ferris S, Hu W, Guo F, Ziegeler G, Halperin JA. Analysis of the promoters and 5'-UTR of mouse Cd59 genes, and of their functional activity in erythrocytes. Genes Immun 2006; 7:287-97. [PMID: 16541098 DOI: 10.1038/sj.gene.6364296] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The complement regulatory protein CD59 inhibits formation of the membrane attack complex (MAC), the terminal effector of the complement system. There are two mouse Cd59 genes in mice but only one in humans. In the work reported here we (a) mapped the promoter regions of both mCd59a and mCd59b genes, (b) identified two different promoters for each mCd59 gene, (c) defined a previously unrecognized additional exon 1 in each mCd59 gene, (d) identified that each mCd59 gene expresses two different tissue-specific transcripts that differ in their 5'-UTR, and (e) confirmed the presence of mCd59b mRNA in multiple tissues. At the functional level, comparison of the sensitivity of mCd59ab(-/-) and mCd59a(-/-) red blood cells to MAC-mediated lysis revealed that mCd59b protects RBC from MAC-mediated lysis, at least in the setting of mCd59a deficiency. Together these findings indicate that the mCd59 genes may have complex and perhaps different regulatory mechanisms in different tissues.
Collapse
Affiliation(s)
- X Qin
- Department of Medicine, Division of Hematology and Oncology, Brigham and Women's Hospital, Boston, MA, USA.
| | | | | | | | | | | |
Collapse
|
87
|
Mitsuhashi N, Fischer-Lougheed J, Shulkin I, Kleihauer A, Kohn DB, Weinberg KI, Starnes VA, Kearns-Jonker M. Tolerance induction by lentiviral gene therapy with a nonmyeloablative regimen. Blood 2006; 107:2286-93. [PMID: 16291598 DOI: 10.1182/blood-2005-03-1172] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractAntibodies (Abs) directed at the Galα1,3Galβ1,4GlcNAc-R (αGal) carbohydrate epitope initiate xenograft rejection. Previously, we have shown that bone marrow transplantation (BMT) with lentivirus-mediated gene transfer of porcine α1,3 galactosyltransferase (GalT) is able to induce tolerance to αGal-expressing heart grafts following a lethal dose of irradiation. Here we show the first demonstration of permanent survival of αGal+ hearts following transplantation with autologous, lentivirus-transduced BM using a nonmyeloablative regimen. Autologous BM from GalT knockout (GalT–/–) mice was transduced with a lentiviral vector expressing porcine GalT and transplanted into sublethally irradiated (3 Gy) GalT–/– mice. Chimerism in the peripheral blood cells (PBCs) remained low but was higher in the BM, especially within the stromal cell population. Mice reconstituted with GalT did not produce anti-αGal Abs over time. We immunized these mice with αGal-expressing cells and assessed humoral immune responses. Anti-αGal xenoantibodies were not produced in mice reconstituted with GalT, but normal Ab responses to other xenoantigens were detected. Mice reconstituted with GalT accepted αGal+ heart grafts over 100 days. Transduction with lentiviral vectors results in chimerism at levels sufficient to induce long-term tolerance under nonmyeloablative conditions.
Collapse
Affiliation(s)
- Noboru Mitsuhashi
- Childrens Hospital of Los Angeles, Department of Cardiothoracic Surgery, Los Angeles, CA 90027, USA
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Abstract
The involvement of complement in the pathogenesis of a great number of partly life threatening diseases defines the importance to develop inhibitors which specifically interfere with its deleterious action. Endogenous soluble complement-inhibitors, antibodies or low molecular weight antagonists, either blocking key proteins of the cascade reaction or neutralizing the action of the complement-derived anaphylatoxins have successfully been tested in various animal models over the past years. Promising results consequently led to first clinical trials. This review is focused on different approaches for the development of inhibitors, on their site of action in the cascade, on possible indications for complement inhibition based on experimental animal data, and on potential side effects of such treatment.
Collapse
Affiliation(s)
- Tom E Mollnes
- Institute of Immunology, Rikshospitalet University Hospital and University of Oslo, N-0027 Oslo, Norway.
| | | |
Collapse
|
89
|
Robertson C, Booth SA, Beniac DR, Coulthart MB, Booth TF, McNicol A. Cellular prion protein is released on exosomes from activated platelets. Blood 2006; 107:3907-11. [PMID: 16434486 DOI: 10.1182/blood-2005-02-0802] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cellular prion protein (PrP(C)) is a glycophosphatidylinositol (GPI)-anchored protein, of unknown function, found in a number of tissues throughout the body, including several blood components of which platelets constitute the largest reservoir in humans. It is widely believed that a misfolded, protease-resistant form of PrP(C), PrP(Sc), is responsible for the transmissible spongiform encephalopathy (TSE) group of fatal neurodegenerative diseases. Although the pathogenesis of TSEs is poorly understood, it is known that PrP(C) must be present in order for the disease to progress; thus, it is important to determine the physiologic function of PrP(C). Resolving the location of PrP(C) in blood will provide valuable clues as to its function. PrP(C) was previously shown to be on the alpha granule membrane of resting platelets. In the current study platelet activation led to the transient expression of PrP(C) on the platelet surface and its subsequent release on both microvesicles and exosomes. The presence of PrP(C) on platelet-derived exosomes suggests a possible mechanism for PrP(C) transport in blood and for cell-to-cell transmission.
Collapse
Affiliation(s)
- Catherine Robertson
- Department of Oral Biology, University of Manitoba, 780 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0W2
| | | | | | | | | | | |
Collapse
|
90
|
Banz Y, Rieben R. Endothelial cell protection in xenotransplantation: looking after a key player in rejection. Xenotransplantation 2006; 13:19-30. [PMID: 16497209 DOI: 10.1111/j.1399-3089.2005.00266.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The endothelium, as an organ at the interface between the intra- and extravascular space, actively participates in maintaining an anti-inflammatory and anti-coagulant environment under physiological conditions. Severe humoral as well as cellular rejection responses, which accompany cross-species transplantation of vascularized organs as well as ischemia/reperfusion injury, primarily target the endothelium and disrupt this delicate balance. Activation of pro-inflammatory and pro-coagulant pathways often lead to irreversible injury not only of the endothelial layer but also of the entire graft, with ensuing rejection. This review focuses on strategies targeted at protecting the endothelium from such damaging effects, ranging from genetic manipulation of the donor organ to soluble, as well as membrane-targeted, protective strategies.
Collapse
Affiliation(s)
- Yara Banz
- Department of Clinical Research, University of Bern, Switzerland
| | | |
Collapse
|
91
|
Feng G, Yangpei Z, Yanjun J, Yingli W, Yingxia T, Shuguang T. Down-regulation of αGal epitopes by co-transfection of α1,3-galactosidase gene and α1,2-fucosyltransferase gene. CHINESE SCIENCE BULLETIN-CHINESE 2005. [DOI: 10.1007/bf02899642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
92
|
McGregor CGA, Davies WR, Oi K, Teotia SS, Schirmer JM, Risdahl JM, Tazelaar HD, Kremers WK, Walker RC, Byrne GW, Logan JS. Cardiac xenotransplantation: recent preclinical progress with 3-month median survival. J Thorac Cardiovasc Surg 2005; 130:844-51. [PMID: 16153938 DOI: 10.1016/j.jtcvs.2005.04.017] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2005] [Revised: 04/07/2005] [Accepted: 04/07/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Transplantation is limited by a lack of human organ donors. Organs derived from animals, most likely the pig, represent a potential solution to this problem. For the heart, 90-day median graft survival of life-supporting pig hearts transplanted to nonhuman primates has been considered a reasonable standard for entry into the clinical arena. Overcoming the immune barrier to successful cardiac xenotransplantation is most appropriately first explored with the non-life-supporting heterotopic model. METHODS We performed a series of 7 heterotopic heart transplantations from CD46 transgenic pigs to baboons using a combination of therapeutic agents largely targeted at controlling the synthesis of anti-pig antibodies. Rituximab (anti-CD20) and Thymoglobulin (rabbit antithymocyte globulin [ATG]; SangStat Medical Corp, Fremont, Calif) were used as induction therapy. Baseline immunosuppression consisted of splenectomy, tacrolimus, sirolimus, steroids, and TPC (an anti-Gal antibody therapeutic). Rejection events were not treated. RESULTS By using Kaplan-Meier analysis, median graft survival was 96 days (range, 15-137 days; 95% confidence interval, 38-99 days). Only 2 grafts were lost as a result of rejection, as defined by cessation of graft palpation. There was no evidence of a consumptive coagulopathy, infectious complications were treatable, and no posttransplantation lymphoproliferative disorders occurred. No cellular infiltration was observed. CONCLUSIONS This study reports the longest median survival to date (96 days) of pig hearts transplanted heterotopically into baboons. Duplication of these results in the orthotopic life-supporting position could bring cardiac xenotransplantation to the threshold of clinical application.
Collapse
Affiliation(s)
- Christopher G A McGregor
- Division of Cardiovascular Surgery, Department of Surgery, Mayo Clinic, Rochester, Minn 55905, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Banz Y, Cung T, Korchagina EY, Bovin NV, Haeberli A, Rieben R. Endothelial cell protection and complement inhibition in xenotransplantation: a novel in vitro model using whole blood. Xenotransplantation 2005; 12:434-43. [PMID: 16202066 DOI: 10.1111/j.1399-3089.2005.00239.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Studying the interactions between xenoreactive antibodies, complement and coagulation factors with the endothelium in hyperacute and acute vascular rejection usually necessitates the use of in vivo models. Conventional in vitro or ex vivo systems require either serum, plasma or anti-coagulated whole blood, making analysis of coagulation-mediated effects difficult. Here a novel in vitro microcarrier-based system for the study of endothelial cell (EC) activation and damage, using non-anticoagulated whole blood is described. Once established, the model was used to study the effect of the characterized complement- and coagulation inhibitor dextran sulfate (DXS, MW 5000) for its EC protective properties in a xenotransplantation setting. METHODS Porcine aortic endothelial cells (PAEC), grown to confluence on microcarrier beads, were incubated with non-anticoagulated whole human blood until coagulation occurred or for a maximum of 90 min. PAEC-beads were either pre- or co-incubated with DXS. Phosphate buffered saline (PBS) experiments served as controls. Fluid phase and surface activation markers for complement and coagulation were analyzed as well as binding of DXS to PAEC-beads. RESULTS Co- as well as pre-incubation of DXS, followed by washing of the beads, significantly prolonged time to coagulation from 39 +/- 12 min (PBS control) to 74 +/- 23 and 77 +/- 20 min, respectively (P < 0.005 vs. PBS). DXS treatment attenuated surface deposition of C1q, C4b/c, C3b/c and C5b-9 without affecting IgG or IgM deposition. Endothelial integrity, expressed by positivity for von Willebrand Factor, was maintained longer with DXS treatment. Compared with PBS controls, both pre- and co-incubation with DXS significantly prolonged activated partial thromboplastin time (>300 s, P < 0.05) and reduced production of thrombin-antithrombin complexes and fibrinopeptide A. Whilst DXS co-incubation completely blocked classical pathway complement activity (CH50 test) DXS pre-incubation or PBS control experiments showed no inhibition. DXS bound to PAEC-beads as visualized using fluorescein-labeled DXS. CONCLUSIONS This novel in vitro microcarrier model can be used to study EC damage and the complex interactions with whole blood as well as screen ''endothelial protective'' substances in a xenotransplantation setting. DXS provides EC protection in this in vitro setting, attenuating damage of ECs as seen in hyperacute xenograft rejection.
Collapse
Affiliation(s)
- Yara Banz
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
94
|
Sun H, Chen G, Liu W, Kubelik D, Yang H, White DJ, Zhong R, Garcia B. The Influence of Baseline Expression of Human Decay Accelerating Factor Transgene on Graft Survival and Acute Humoral Xenograft Rejection. Transplantation 2005; 80:1331-9. [PMID: 16314803 DOI: 10.1097/01.tp.0000177649.30721.31] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Transgenic pigs expressing human decay accelerating factor (hDAF) have been widely used as donors in various non-human primate transplant models. Despite the use of similar immunosuppressive protocols, there is marked variation in graft survival among centres. The present study was undertaken to determine whether the level of hDAF expression in the pig kidney correlates with the degree of rejection and duration of graft survival. METHODS hDAF transgenic pigs were provided from two suppliers: Guelph Imutran Centre (G) and Harlan Sprague Dawley (H). Following a bilateral nephrectomy, a single hDAF pig kidney was implanted in the baboon, which was subsequently treated with conventional immunosuppressive protocols. The pig's contralateral kidney was collected to provide baseline data. The severity of acute humoral xenograft rejection (AHXR) was graded as stage I-III. hDAF expression was measured using morphologic analysis comparing the contralateral and grafted kidneys at the endpoint. RESULTS Baseline hDAF expression in kidneys from pigs provided by supplier G was significantly higher than that from supplier H (P<0.01). Furthermore, the survival of baboons receiving grafts from G pigs was significantly longer than those receiving grafts from H pigs (P<0.05). In addition, reduction of hDAF expression at the endpoint was associated with a higher degree of AHXR. Severe apoptosis or necrosis was found in grafts with AHXR II-III. CONCLUSIONS Pig kidneys from different suppliers have variable baseline hDAF expression, which may have an influence on graft survival. Reduced expression of hDAF in the terminal graft was associated with the severity of rejection.
Collapse
Affiliation(s)
- Hongtao Sun
- Department of Pathology, University of Western Ontario, and Multi-Organ Transplant Program, London Health Sciences Centre, London, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Wenzel K, Zabojszcza J, Carl M, Taubert S, Lass A, Harris CL, Ho M, Schulz H, Hummel O, Hubner N, Osterziel KJ, Spuler S. Increased Susceptibility to Complement Attack due to Down-Regulation of Decay-Accelerating Factor/CD55 in Dysferlin-Deficient Muscular Dystrophy. THE JOURNAL OF IMMUNOLOGY 2005; 175:6219-25. [PMID: 16237120 DOI: 10.4049/jimmunol.175.9.6219] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dysferlin is expressed in skeletal and cardiac muscles. However, dysferlin deficiency results in skeletal muscle weakness, but spares the heart. We compared intraindividual mRNA expression profiles of cardiac and skeletal muscle in dysferlin-deficient SJL/J mice and found down-regulation of the complement inhibitor, decay-accelerating factor/CD55, in skeletal muscle only. This finding was confirmed on mRNA and protein levels in two additional dysferlin-deficient mouse strains, A/J mice and Dysf-/- mice, as well as in patients with dysferlin-deficient muscular dystrophy. In vitro, the absence of CD55 led to an increased susceptibility of human myotubes to complement attack. Evidence is provided that decay-accelerating factor/CD55 is regulated via the myostatin-SMAD pathway. In conclusion, a novel mechanism of muscle fiber injury in dysferlin-deficient muscular dystrophy is demonstrated, possibly opening therapeutic avenues in this to date untreatable disorder.
Collapse
Affiliation(s)
- Katrin Wenzel
- Myology Research Group, Department of Neurology, Charité University Hospital, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Chen W, Zhou D, Torrealba JR, Waddell TK, Grant D, Zhang L. Donor Lymphocyte Infusion Induces Long-Term Donor-Specific Cardiac Xenograft Survival through Activation of Recipient Double-Negative Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:3409-16. [PMID: 16116235 DOI: 10.4049/jimmunol.175.5.3409] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Previous studies have shown that pretransplant donor lymphocyte infusion (DLI) can enhance xenograft survival. However, the mechanism by which DLI induces xenograft survival remains obscure. Using T cell subset-deficient mice as recipients we show that CD4+, but not CD8+, T cells are necessary to mediate the rejection of concordant cardiac xenografts. Adoptive transfer of naive CD4+ T cells induces rejection of accepted cardiac xenografts in CD4-/- mice. This rejection can be prevented by pretransplant DLI in the absence of any other treatment. Furthermore, we demonstrate that DLI activates alphabeta-TCR+CD3+CD4-CD8- double-negative (DN) regulatory T (Treg) cells in xenograft recipients, and that DLI-activated DN Treg cells can inhibit the proliferation of donor-specific xenoreactive CD4+ T cells in vitro. More importantly, adoptive transfer of DLI-activated DN Treg cells from xenograft recipients can suppress the proliferation of xenoreactive CD4+ T cells and their ability to produce IL-2 and IFN-gamma in vivo. Adoptive transfer of DLI-activated DN Treg cells also prevents CD4+ T cell-mediated cardiac xenograft rejection in an Ag-specific fashion. These data provide direct evidence that DLI can activate recipient DN Treg cells, which can induce donor-specific long-term cardiac xenograft survival by suppressing the proliferation and function of donor-specific CD4+ T cells in vivo.
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Laboratory Medicine and Pathobiology, Multi Organ Transplantation Program, Toronto General Research Institute, University Health Network, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
97
|
O'Connell PJ. Thrombotic microangiopathy: the next big hurdle for xenotransplantation. J Am Soc Nephrol 2005; 16:2529-30. [PMID: 16093447 DOI: 10.1681/asn.2005070735] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
98
|
Eto N, Kojima I, Uesugi N, Inagi R, Miyata T, Fujita T, Johnson RJ, Shankland SJ, Nangaku M. Protection of endothelial cells by dextran sulfate in rats with thrombotic microangiopathy. J Am Soc Nephrol 2005; 16:2997-3005. [PMID: 16093450 DOI: 10.1681/asn.2005020137] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The characteristic features of thrombotic microangiopathy (TMA) include glomerular and peritubular capillary endothelial cell injury in association with loss of heparan sulfate proteoglycans on the cell surface and thrombus formation, followed by subsequent ischemic tubulointerstitial damage. It therefore was hypothesized that dextran sulfate (DXS) may protect the kidney against endothelial damage in a model of TMA. TMA was induced in rats by renal artery perfusion of an antiglomerular endothelial antibody, followed by the administration of DXS or vehicle. Renal damage was assessed by histologic analysis and measurements of blood urea nitrogen and creatinine. Whereas control rats developed severe renal failure with extensive glomerular and tubular injury, administration of DXS significantly protected renal function and preserved the glomerular endothelium and peritubular capillaries. The beneficial effect of DXS could be attributed to the ability of DXS to protect endothelial cells from coagulation and complement activation, as demonstrated by the histologic analysis. In addition, binding of the administered DXS to the surface of the glomerular endothelium was confirmed in TMA rats, suggesting that DXS acts as a "repair coat" of injured glomerular endothelium. In conclusion, DXS protects the kidney from experimental TMA. This protection may be mediated by DXS's binding directly to the surface of glomerular endothelium and amelioration of coagulation, complement activation, and cellular matrix loss.
Collapse
Affiliation(s)
- Nobuaki Eto
- Division of Nephrology and Endocrinology, University of Tokyo, School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Shimizu A, Yamada K, Yamamoto S, Lavelle JM, Barth RN, Robson SC, Sachs DH, Colvin RB. Thrombotic Microangiopathic Glomerulopathy in Human Decay Accelerating Factor–Transgenic Swine-to-Baboon Kidney Xenografts. J Am Soc Nephrol 2005; 16:2732-45. [PMID: 16049072 DOI: 10.1681/asn.2004121148] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Models of pig-to-baboon xenografting were examined to identify the mechanisms and pathologic characteristics of acute humoral xenograft rejection (AHXR). Thymus and kidney (composite thymokidney) from human decay accelerating factor-transgenic swine were transplanted into baboons (n = 16) that were treated with an immunosuppressive regimen that included extracorporeal immunoadsorption of anti-alphaGal antibody and inhibition of complement activation. Morphologic and immunohistochemical studies were performed on protocol biopsies and graftectomy samples. All renal xenografts avoided hyperacute rejection. However, graft rejection coincided with the increase of anti-alphaGal antibody in the recipient's circulation. The 16 xenografts studied were divided into two groups dependent on the rapid return (group 1) or gradual return (group 2) of anti-alphaGal antibody after immunoadsorption. In group 1 (n = 6), grafts were rejected to day 27 with development of typical AHXR, characterized by marked interstitial hemorrhage and thrombotic microangiopathy in the renal vasculature. In group 2 (n = 10), grafts also developed thrombotic microangiopathy affecting mainly the glomeruli by day 30 but also showed minimal evidence of interstitial injury and hemorrhage. In the injured glomeruli, IgM and C4d deposition, subsequent endothelial cell death and activation with upregulation of von Willebrand factor and tissue factor, and a decrease of CD39 expression developed with the formation of fibrin-platelet multiple microthrombi. In this model, the kidney xenografts, from human decay accelerating factor-transgenic swine, in baboons undergo AHXR. In slowly evolving AHXR, graft loss is associated with the development of thrombotic microangiopathic glomerulopathy. Also, anti-alphaGal IgM deposition and subsequent complement activation play an important role in the mechanism of glomerular endothelial injury and activation and the formation of multiple microthrombi.
Collapse
Affiliation(s)
- Akira Shimizu
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Capey S, van den Berg CW. Porcine complement regulators protect aortic smooth muscle cells poorly against human complement-induced lysis and proliferation: consequences for xenotransplantation. Xenotransplantation 2005; 12:217-26. [PMID: 15807772 DOI: 10.1111/j.1399-3089.2005.00217.x] [Citation(s) in RCA: 10] [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 Accelerated atherosclerosis after transplantation has been observed and is characterized by smooth muscle cell proliferation in the graft. Porcine cells are frequently used in models of atherosclerosis and porcine organs are considered for use in transplantation. Complement (C) activation is known to play a major role in rejection of xenografts and is also considered to play a role in the development of atherosclerosis. The aim of this study was to investigate the expression and function of membrane bound regulators of complement (CReg) on porcine aortic smooth muscle cells (PASMC). METHODS The PASMC were assessed for expression of CReg and susceptibility to lysis by human C by flow-cytometry. The effect of various cytokines on CReg expression and C-susceptibility was investigated. The ability of human C to induce cell proliferation was assessed using the Alamar blue assay. RESULTS The PASMC only express the CReg membrane cofactor protein (MCP) and CD59 on their cell surface. MCP expression was increased by interleukin (IL)-4. In contrast to porcine aortic endothelial cells (PAEC), PASMC were found to be surprisingly sensitive to C-mediated lysis, mainly due to a low level of expression of CD59. Human C-induced proliferation of PASMC, which was dependent on complete membrane attack complex (MAC) formation. CONCLUSIONS Endogenously expressed CReg on PASMC poorly protect these cells to human C. Human C can induce proliferation of PASMC. In order to prevent accelerated atherosclerosis in porcine xenografts, increased levels of CReg not only have to be obtained on the endothelial cells but also on the smooth muscle cells.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/physiology
- Aorta/cytology
- Aorta/metabolism
- CD55 Antigens/metabolism
- CD59 Antigens/physiology
- Cell Division/physiology
- Cell Membrane/metabolism
- Cells, Cultured
- Complement Membrane Attack Complex/biosynthesis
- Complement System Proteins/physiology
- Cytoprotection
- Humans
- Interleukin-4/pharmacology
- Membrane Cofactor Protein
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- RNA, Messenger/metabolism
- Swine/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Steven Capey
- Department of Pharmacology, Therapeutics and Toxicology, Wales Heart Research Institute, Cardiff University, Wales College of Medicine, Cardiff, CF144XN, UK
| | | |
Collapse
|