Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Transplant Proc ; 48(6): 2200-7, 2016.
Article in English | MEDLINE | ID: mdl-27569971

ABSTRACT

Islet transplantation can potentially cure type 1 diabetes mellitus, but it is limited by a shortage of human donors as well as by islet graft destruction by inflammatory and thrombotic mechanisms. A possible solution to these problems is to use genetically modified pig islets. Endothelial protein C receptor (EPCR) enhances protein C activation and regulates coagulation, inflammation, and apoptosis. We hypothesized that human EPCR (hEPCR) expression on donor islets would improve graft survival and function. Islets from an hEPCR transgenic mouse line strongly expressed the transgene, and hEPCR expression was maintained after islet isolation. Islets were transplanted from hEPCR mice and wild-type (WT) littermates into diabetic mice in a marginal-dose syngeneic intraportal islet transplantation model. The blood glucose level normalized within 5 days in 5 of 7 recipients of hEPCR islets, compared with only 2 of 7 recipients of WT islets (P < .05). Transplanted hEPCR islets had better preserved morphology and more intense insulin staining than WT grafts, and they retained transgene expression. The improved engraftment compared with WT islets suggests that inflammation and coagulation associated with the transplant process can be reduced by hEPCR expression on donor tissue.


Subject(s)
Antigens, CD/metabolism , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation , Receptors, Cell Surface/metabolism , Transplants/metabolism , Animals , Apoptosis , Blood Glucose/analysis , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 1/blood , Endothelial Protein C Receptor , Graft Survival , Humans , Insulin/analysis , Male , Mice , Mice, Transgenic , Protective Agents/metabolism , Protein C/metabolism , Swine
2.
Am J Transplant ; 14(4): 797-805, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24502456

ABSTRACT

Differential protein glycosylation in the donor and recipient can have profound consequences for transplanted organs, as evident in ABO-incompatible transplantation and xenotransplantation. In this study, we investigated the impact of altered fucosylation on graft acceptance by using donor mice overexpressing human α1,2-fucosyltransferase (HTF). Skin and heart grafts from HTF transgenic mice were rapidly rejected by otherwise completely matched recipients (median survival times 16 and 14 days, respectively). HTF skin transplanted onto mice lacking T and B cells induced an natural killer cell-mediated innate rejection crisis that affected 50-95% of the graft at 10-20 days. However, in the absence of adaptive immunity, the residual graft recovered and survived long-term (>100 days). Experiments using "parked" grafts or MHC class II-deficient recipients suggested that indirect rather than direct antigen presentation plays a role in HTF skin graft rejection, although the putative antigen(s) was not identified. We conclude that altered glycosylation patterns on donor tissue can trigger a powerful rejection response comprising both innate and adaptive components. This has potential implications for allotransplantation, in light of increasing recognition of the variability of the human glycome, and for xenotransplantation, where carbohydrate remodeling has been a lynchpin of donor genetic modification.


Subject(s)
Fucosyltransferases/metabolism , Graft Rejection/etiology , Heart Transplantation/adverse effects , Major Histocompatibility Complex/physiology , Skin Transplantation/adverse effects , Transplantation, Heterologous/adverse effects , Animals , Antigen Presentation/immunology , Female , Fucosyltransferases/genetics , Glycosylation , Graft Rejection/mortality , Graft Rejection/pathology , Graft Survival , Humans , Immunoenzyme Techniques , Killer Cells, Natural/immunology , Lymphocyte Depletion , Male , Mice , Mice, Knockout , Mice, Transgenic , Prognosis , Risk Factors , Survival Rate , T-Lymphocytes/immunology , Tissue Donors , Transplantation, Homologous , Galactoside 2-alpha-L-fucosyltransferase
3.
Am J Transplant ; 12(9): 2363-72, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22681753

ABSTRACT

Thrombosis and inflammation are major obstacles to successful pig-to-human solid organ xenotransplantation. A potential solution is genetic modification of the donor pig to overexpress molecules such as the endothelial protein C receptor (EPCR), which has anticoagulant, anti-inflammatory and cytoprotective signaling properties. Transgenic mice expressing human EPCR (hEPCR) were generated and characterized to test this approach. hEPCR was expressed widely and its compatibility with the mouse protein C pathway was evident from the anticoagulant phenotype of the transgenic mice, which exhibited a prolonged tail bleeding time and resistance to collagen-induced thrombosis. hEPCR mice were protected in a model of warm renal ischemia reperfusion injury compared to wild type (WT) littermates (mean serum creatinine 39.0 ± 2.3 µmol/L vs. 78.5 ± 10.0 µmol/L, p < 0.05; mean injury score 31 ± 7% vs. 56 ± 5%, p < 0.05). Heterotopic cardiac xenografts from hEPCR mice showed a small but significant prolongation of survival in C6-deficient PVG rat recipients compared to WT grafts (median graft survival 6 vs. 5 days, p < 0.05), with less hemorrhage and edema in rejected transgenic grafts. These data indicate that it is possible to overexpress EPCR at a sufficient level to provide protection against transplant-related thrombotic and inflammatory injury, without detrimental effects in the donor animal.


Subject(s)
Antigens, CD/metabolism , Endothelium, Vascular/metabolism , Glycoproteins/metabolism , Models, Animal , Receptors, Cell Surface/metabolism , Animals , Endothelial Protein C Receptor , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Reperfusion Injury/prevention & control
4.
Am J Transplant ; 10(12): 2586-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20840479

ABSTRACT

The vascular ectonucleotidases CD39[ENTPD1 (ectonucleoside triphosphate diphosphohydrolase-1), EC 3.6.1.5] and CD73[EC 3.1.3.5] generate adenosine from extracellular nucleotides. CD39 activity is critical in determining the response to ischemia-reperfusion injury (IRI), and CD39 null mice exhibit heightened sensitivity to renal IRI. Adenosine has multiple mechanisms of action in the vasculature including direct endothelial protection, antiinflammatory and antithrombotic effects and is protective in several models of IRI. Mice transgenic for human CD39 (hCD39) have increased capacity to generate adenosine. We therefore hypothesized that hCD39 transgenic mice would be protected from renal IRI. The overexpression of hCD39 conferred protection in a model of warm renal IRI, with reduced histological injury, less apoptosis and preserved serum creatinine and urea levels. Benefit was abrogated by pretreatment with an adenosine A2A receptor antagonist. Adoptive transfer experiments showed that expression of hCD39 on either the vasculature or circulating cells mitigated IRI. Furthermore, hCD39 transgenic kidneys transplanted into syngeneic recipients after prolonged cold storage performed significantly better and exhibited less histological injury than wild-type control grafts. Thus, systemic or local strategies to promote adenosine generation and signaling may have beneficial effects on warm and cold renal IRI, with implications for therapeutic application in clinical renal transplantation.


Subject(s)
Antigens, CD/biosynthesis , Apyrase/biosynthesis , Reperfusion Injury/prevention & control , Adenosine/metabolism , Animals , Cold Ischemia , Humans , Kidney Cortex Necrosis/prevention & control , Mice , Mice, Transgenic , Models, Animal
5.
Am J Transplant ; 10(2): 242-50, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20055798

ABSTRACT

Thrombomodulin (TBM) is an important vascular anticoagulant that has species specific effects. When expressed as a transgene in pigs, human (h)TBM might abrogate thrombotic manifestations of acute vascular rejection (AVR) that occur when GalT-KO and/or complement regulator transgenic pig organs are transplanted to primates. hTBM transgenic mice were generated and characterized to determine whether this approach might show benefit without the development of deleterious hemorrhagic phenotypes. hTBM mice are viable and are not subject to spontaneous hemorrhage, although they have a prolonged bleeding time. They are resistant to intravenous collagen-induced pulmonary thromboembolism, stasis-induced venous thrombosis and pulmonary embolism. Cardiac grafts from hTBM mice to rats treated with cyclosporine in a model of AVR have prolonged survival compared to controls. hTBM reduced the inflammatory reaction in the vein wall in the stasis-induced thrombosis and mouse-to-rat xenograft models and reduced HMGB1 levels in LPS-treated mice. These results indicate that transgenic expression of hTBM has anticoagulant and antiinflammatory effects that are graft-protective in murine models.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cyclosporine/pharmacology , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Rats , Swine , Thrombomodulin , Transgenes/drug effects
7.
Xenotransplantation ; 7(4): 237-46, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11081758

ABSTRACT

Pig-to-primate vascularized xenografts undergo hyperacute rejection (HAR). This results from pre-formed xenoreactive antibodies directed against galactose-alpha1,3-galactose (alphaGal) in the donor organ and activation of the complement cascade. We describe an in vivo murine model of HAR using a BALB/c mice system devoid of histocompatibility or complement differences between donor and recipient to investigate in isolation, the effects of alphaGal epitope and anti-alphaGal antibody interactions in causing rejection of vascularized heart transplants. Gal KO mice were immunized with rabbit red blood cell membranes to induce high anti-alphaGal antibody titers that were predominantly IgM by ELISA (enzyme-linked immunosorbent assay). When alphaGal-expressing mice hearts were transplanted heterotopically into these recipients (n= 12), 67% of grafts rejected within 24 h, the majority within 16 h with histological features of HAR. In contrast, none of the grafts in the non-immunized Gal KO recipient control group (n=11) underwent HAR. Interestingly, approximately 50% of the remaining grafts in both the immunized and non-immunized Gal KO recipient group were rejected between 7 and 27 days by a rejection process characterized by a dense infiltrate of macrophage/monocytes, perivascular cuffing and tissue destruction similar to recent descriptions of delayed xenograft rejection (DXR). In addition, some grafts (21.5%) continued to survive in the immunized Gal KO recipients despite the presence of anti-alphaGal antibody and normal complement activity and these showed well-preserved myocardium when harvested whilst still functioning well at days 30 or 90. No rejection was seen when Gal KO donors were used in this system (n=4), nor when alphaGal-expressing BALB/c hearts were transplanted into alphaGal-expressing BALB/c recipients (n=5). This in vivo small animal model offers the opportunity to test a variety of strategies to overcome HAR prior to more resource intensive pig-to-primate studies, and may provide insights into the processes similar to DXR and accommodation.


Subject(s)
Disaccharides/immunology , Galactosyltransferases/metabolism , Graft Rejection/immunology , Heart Transplantation/immunology , Acute Disease , Animals , Antibodies, Heterophile/blood , Arterioles/pathology , Coronary Vessels/pathology , Epitopes/immunology , Erythrocyte Transfusion , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Graft Rejection/pathology , Heart Transplantation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Rabbits
8.
Transplantation ; 70(1): 168-74, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919596

ABSTRACT

BACKGROUND: Investigations of the role of CD4 T lymphocytes in allograft rejection and tolerance have relied on the use of mouse models with a deficiency in CD4 cells. However, in mice treated with depleting monoclonal antibody (mAb) and in MHC class II knockout (KO) mice, there are residual populations of CD4 cells. CD4 KO mice had increased CD4- CD8-TCRalphabeta+ helper T cells, and both strains of KO mice could reject skin allografts at the normal rate. In this study, transgenic mice with no peripheral CD4 cells were the recipients of skin and heart allografts. Results were compared with allograft survival in CD4 and MHC class II KO mice. METHODS: GK5 (C57BL/6 bml mice transgenic for a chimeric anti-CD4 antibody) had no peripheral CD4 cells. These mice, and CD4 and class II KO mice, received BALB/c or CBA skin or cardiac allografts. Some GK5 mice were treated with anti-CD8 mAb to investigate the role of CD8 cells in rejection. CD4 and CD8 cells were assessed by FACS and immunohistochemistry. RESULTS: BALB/c skin on GK5 mice had a mean survival time +/- SD of 24+/-6 days, compared with 9+/-2 days in wild-type mice. Anti-CD8 mAb prolonged this to 66+/-7 days. BALB/c skin survived 10+/-2 days on class II KO and 14+/-2 days on CD4 KO, both significantly less than the survival seen on GK5 recipients (P<0.001). BALB/c hearts survived >100 days in GK5 recipients and in wild-type recipients treated with anti-CD4 mAb at the time of grafting, in contrast to a mean survival time of 10+/-2 days in untreated wild-type mice. Immunohistochemistry revealed that long-term surviving heart allografts from the GK5 recipients had CD8 but no CD4 cellular infiltrate. These hearts showed evidence of transplant vasculopathy. CONCLUSIONS: The GK5 mice, with a complete absence of peripheral CD4 cells, provide the cleanest available model for investigating the role of CD4 lymphocytes in allograft rejection. Prolonged skin allograft survival in these mice compared with CD4 and MHC class II KO recipients was clearly the result of improved CD4 depletion. Nevertheless, skin allograft rejection, heart allograft infiltration, and vascular disease, mediated by CD8 cells, developed in the absence of peripheral CD4 T cells.


Subject(s)
CD4 Antigens/physiology , CD4-Positive T-Lymphocytes/physiology , Graft Survival , Animals , CD8-Positive T-Lymphocytes/physiology , Heart Transplantation/immunology , Histocompatibility Antigens Class II/physiology , Immunoglobulin G/blood , Mice , Mice, Inbred Strains , Mice, Knockout , Skin Transplantation/immunology , Transplantation, Homologous
9.
Xenotransplantation ; 7(1): 42-7, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10809056

ABSTRACT

Antibodies directed against galactose-alpha1,3-galactose (alphaGal) are believed to play an important role in the pathogenesis of delayed xenograft rejection (DXR). This study was designed to determine whether alpha1,3-galactosyltransferase-deficient (Gal KO) mice can naturally acquire a sufficient anti-alphaGal titre to cause the delayed type rejection of alphaGal-expressing hearts. Gal KO mice of various ages were assessed for anti-alphaGal antibody levels. alphaGal-expressing hearts were transplanted heterotopically into these mice and monitored daily. Rejecting and surviving hearts were evaluated histologically. In Gal KO mice greater than 6-month-old, 64% had an anti-alphaGal antibody titre above the background level. When wild-type alphaGal-expressing hearts were transplanted into this group, 45% of grafts rejected within 5 to 13 days. Histological examination of the rejected hearts displayed marked tissue damage and an inflammatory infiltrate of predominantly macrophage/monocytes. Surviving grafts showed preserved morphology. Like humans, Gal KO mice naturally develop anti-alphaGal antibodies with age. The titre in these mice was sufficient to cause a "delayed-type" rejection of a significant proportion of alphaGal-expressing cardiac grafts. This model thus provides an opportunity to investigate the role of naturally acquired anti-alphaGal antibodies in the pathogenesis of DXR.


Subject(s)
Disaccharides/immunology , Graft Rejection/immunology , Isoantibodies/physiology , Models, Immunological , Transplantation, Heterologous/immunology , Transplantation, Homologous/immunology , Animals , Disaccharides/genetics , Female , Graft Rejection/pathology , Heart Transplantation/immunology , Heart Transplantation/pathology , Humans , Immunity, Innate/genetics , Isoantibodies/biosynthesis , Isoantibodies/blood , Isoantibodies/genetics , Male , Mice , Mice, Knockout , Transplantation, Heterologous/pathology , Transplantation, Homologous/pathology
10.
Transpl Immunol ; 8(3): 203-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11147701

ABSTRACT

To compare the effectiveness of transgenic and systemic monoclonal antibody therapy for pancreas transplantation, vascularised segmental pancreas allografts from wild-type or transgenic pancreatic tissue that secreted monoclonal anti-CD4 were placed in CBA recipients in which diabetes had been induced chemically by streptozotocin (STZ, non-autoimmune diabetes). In untreated CBA recipients, wild-type BALB/c or C57BL/6 bml pancreas transplants were rejected in a mean survival time (MST) of 27 and 30 days, respectively. BALB/c and C57BL/6 graft survival improved when recipients were given a short course of T cell depleting monoclonal anti-CD4 antibody, (GK 1.5, 2 mg total on days -1, 0, 1, 2 with grafting on day 0) with MST +/- S.D. of 71 +/- 29 and 44 +/- 36 days, respectively. Thus, transient depletion of CD4 was effective in delaying pancreas allograft rejection in these strain combinations. The use of C57BL/6 bml mice transgenic for a rat anti-CD4 antibody (GK5 mice) as pancreas donors provided allografts that secreted sufficient anti-CD4 antibody to cause CD4 T cell depletion in the recipients (CD4 cells decreased from 30 to < 5% of small lymphocytes). This degree of depletion was not sustained and the CD4 recovery inversely correlated with graft survival. Mice with > 20% CD4 cells in the splenic lymphocyte population 4 weeks post-transplant rejected their grafts (3 of 10 mice). However, in 7 of 10 mice CD4 cells remained low (< 15%) and allografts survived for > 80 days. The GK5 allografts survived significantly longer than those from non-transgenic bml controls (MST 83 +/- 32 days, compared with 30 days, P < 0.0005). This survival time was similar to that of BALB/c allografts in CBA recipients treated with a high dose of anti-CD4 antibody. Thus, transgenic secretion of anti-CD4 antibody by the pancreas allograft was very effective in prolonging its survival.


Subject(s)
Antibodies, Monoclonal/biosynthesis , CD4 Antigens/immunology , Graft Enhancement, Immunologic/methods , Graft Survival/immunology , Pancreas Transplantation/immunology , Animals , Antibodies, Monoclonal/genetics , Antilymphocyte Serum/biosynthesis , Antilymphocyte Serum/genetics , Diabetes Mellitus, Experimental/surgery , Graft Survival/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Pancreas Transplantation/pathology , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , T-Lymphocytes/immunology , Time Factors , Transplantation Immunology , Transplantation, Homologous
11.
Microsurgery ; 19(6): 272-4, 1999.
Article in English | MEDLINE | ID: mdl-10469441

ABSTRACT

This study describes a new method for joining the donor ureter to the recipient bladder during mouse kidney transplantation. The donor left kidney was harvested using methods previously published, except that bladder tissue was not harvested with the end of the ureter. The recipient left kidney was removed and the donor kidney was attached using end-to-side anastomosis. The recipient bladder was pierced with a 21-gauge needle allowing curved forceps to be inserted through the bladder, to pull through the ureter, and the periuretal tissue was stitched to the exterior wall of the bladder. The donor ureter was allowed to retract inside the bladder. Following a right nephrectomy, grafts were monitored by blood serum creatinine and urea. With a technical success rate of 83%, this technique reduced donor harvest time by 20 minutes and ureter attachment time by 15 minutes making it the best method available for mouse kidney transplantation.


Subject(s)
Kidney Transplantation/methods , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Suture Techniques , Time Factors , Ureter/surgery , Urinary Bladder/surgery
12.
Transpl Immunol ; 7(1): 37-44, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10375076

ABSTRACT

The study was designed to compare second heart and skin grafts and in vitro assays as a means of assessing peripheral tolerance in C57BL/6 mice. Vascularized heterotopic BALB/c hearts were placed in C57BL/6 recipients treated with anti-CD4, GK1.5 (1 mg total per 20 g mouse i.p. on days 0, 1, 2, 3). Those mice in which hearts survived for >60 days were challenged with donor and third-party (CBA) skin grafts or with second heart grafts, of donor or third-party origin, attached to the carotid artery and jugular vein. In vitro alloreactivity was assessed by mixed lymphocyte reactions (MLR) and cell mediated lympholysis (CML) using recipient spleen cells. Parenchymal damage, cellular infiltration and vascular disease were assessed from the histology of long-term allografts and isografts. Allografts in untreated recipients were rapidly rejected while isografts survived > 100 days. Primary allografts in anti-CD4 treated recipients also survived > 100 days, as did donor strain secondary heart transplants given at >60 days after the first graft. Third-party hearts were rapidly rejected, as were donor and third-party skin grafts placed on recipients with long-term allografts. These recipients showed low MLR response to both donor and third-party stimulators and donor-specific suppression of CML at 60 days post graft. Long-surviving heart allografts all showed evidence of parenchymal damage and vascular intimal thickening. Thus in the BALB/c to C57BL/6 donor-recipient strain combination, hearts, but not skin grafts, could be used to demonstrate peripheral tolerance, which seemed to be both organ and major histocompatibility complex (MHC) specific. Despite long survival, BALB/c hearts all showed evidence of parenchymal damage and vascular intimal thickening, a sign of chronic rejection.


Subject(s)
CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Heart Transplantation/immunology , Immune Tolerance/immunology , Skin Transplantation/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Graft Survival/immunology , Lymphocyte Depletion/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Tissue Donors , Transplantation, Homologous
14.
Transpl Immunol ; 7(4): 207-13, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10638833

ABSTRACT

In order to reduce the toxic effects of the T cell activating anti-CD3 monoclonal antibody, 145-2C11, F(ab')2 fragments were prepared by pepsin digestion. These fragments were then used as non-immunosuppressive carriers for the cytotoxic drug idarubicin (IDA), to reduce toxicity of both the monodonal antibodies (mAb) and the drug and to increase the specificity of drug delivery. The IDA-145-2C11 F(ab')2 immunoconjugate was tested for specificity by fluorometry. 145-2C11 intact antibody, 145-2C11 F(ab')2 and IDA conjugates of the antibody and F(ab')2 were used to treat CBA recipients of BALB/c vascularized cardiac allografts. Mice with hearts surviving >100 days were challenged with donor and third party (C57BL/6) skin grafts. Although both antibody and F(ab')2 blocked the binding of 145-2C11-FITC to CBA spleen cells, only the intact antibody caused sustained depletion of CD3 cells in vivo. 145-2C11 F(ab')2 blocked cell surface CD3 within 30 min, but was cleared in 24 h without depletion of CD3 cells from the spleen. In BALB/c to CBA cardiac allografts (rejected in 12-17 days), IDA-145-2C11 F(ab')2 (0.2 mg/20 g mouse i.p. at the time of transplantation) induced >100 days' allograft survival and specific tolerance, in contrast to the equivalent dose of 145-2C11 F(ab')2 (mean survival 25 days). Hearts from IDA-145-2C11 F(ab')2-treated mice at >100 days showed decreased cellular infiltration and less chronic vascular disease than long-surviving hearts from mice treated with an alternative antibody, KT3. Thus, F(ab')2 prepared from 145-2C11 provided a suitable CD3-specific, nonimmunosuppressive carrier for IDA. This immunoconjugate was more effective against both acute and chronic rejection than other conjugates or whole antibody. IDA-145-2C11 F(ab')2 is an effective, nontoxic tolerogen in the mouse cardiac allograft model.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Coronary Disease/immunology , Heart Transplantation/immunology , Idarubicin/therapeutic use , Immune Tolerance/immunology , Immunoconjugates/therapeutic use , Immunoglobulin Fab Fragments/therapeutic use , Animals , Antibiotics, Antineoplastic/toxicity , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/toxicity , CD3 Complex/immunology , CD3 Complex/metabolism , Chronic Disease , Coronary Disease/pathology , Coronary Disease/prevention & control , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/immunology , Heart Transplantation/pathology , Idarubicin/toxicity , Immunoconjugates/toxicity , Mice , Mice, Inbred Strains
15.
Transplantation ; 66(6): 748-54, 1998 Sep 27.
Article in English | MEDLINE | ID: mdl-9771838

ABSTRACT

BACKGROUND: The key role of anti-galactose alpha1,3-galactose (anti-alphaGal) xenoantibodies in initiating hyperacute xenograft rejection has been clearly demonstrated using a variety of in vitro and in vivo approaches. However, the role of anti-alphaGal antibodies in mediating post-hyperacute rejection mechanisms, such as antibody-dependent cellular cytoxicity, remains to be determined, primarily because of the lack of a small animal model with which to study this phenomena. METHODS: Hearts from wild-type mice were transplanted heterotopically into alpha1,3-galactosyltransferase knockout (Gal KO) mice, which like humans develop antibodies to the disaccharide galactose alpha1,3-galactose (Gal). At the time of rejection, hearts were examined histologically to determine the mechanism of rejection. RESULTS: Hearts from wild-type mice transplanted into high-titer anti-alphaGal recipients were rejected in 8-13 days. Histological examination demonstrated a cellular infiltrate consisting of macrophages (80-90%), natural killer cells (5-10%), and T cells (1-5%). In contrast, wild-type hearts transplanted into low anti-Gal titer recipients demonstrated prolonged (>90 day) survival. However, a significant proportion (30-40%) of these underwent a minor rejection episode between 10 and 13 days, but then recovered ("accommodated"). CONCLUSIONS: The results of this study suggest that the Gal KO mouse is a useful small animal vascularized allograft model, in which the role of anti-alphaGal antibody in graft rejection can be studied in isolation from other rejection mechanisms. The titer of anti-alphaGal antibody was found to be the critical determinant of rejection. The histopathological features of rejection in this model are very similar to other models of delayed xenograft rejection, in both the timing and composition of the cellular infiltrate. The Gal KO mouse therefore provides a new rodent model, which will aid in the identification of the distinct components involved in the pathogenesis of delayed xenograft rejection.


Subject(s)
Antibodies/immunology , Disaccharides/immunology , Galactosyltransferases/genetics , Graft Rejection/immunology , Transplantation, Heterologous/immunology , Animals , Antibodies, Protozoan/immunology , Female , Galactosyltransferases/metabolism , Graft Rejection/genetics , Graft Rejection/pathology , Heart Transplantation/immunology , Leishmania major/immunology , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Myocardium/pathology
16.
Diabetes ; 47(9): 1399-405, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9726227

ABSTRACT

Spontaneously diabetic nonobese diabetic (NOD/Lt) mice were treated with anti-T-cell monoclonal antibodies (mAbs) at the time of grafting with vascularized segmental pancreas isografts. Recipients were either untreated or given anti-CD4 and/or anti-CD8 mAbs (0.5 mg/20-g mouse on each of 4 consecutive days), which reduced target cell levels to <5% of normal. Graft function was monitored by measuring blood glucose (BG) levels. Transplants were removed for histological examination when BG returned to >20 mmol/l for two consecutive readings. Isografts from 3- to 4-week-old prediabetic mice placed in untreated diabetic NOD mice ceased functioning in 9-13 days with a mean survival time (MST) +/- SD of 10 +/- 2. Treatment with anti-CD4 prolonged survival significantly (MST = 61 +/- 35 days, P < 0.05 compared with untreated control mice). Anti-CD8 treatment was less effective, but it still significantly improved graft survival (MST = 24 +/- 9 days, P < 0.05 compared with untreated control mice). Anti-CD8 plus anti-CD4 treatment was highly effective in inhibiting autoimmune destruction of the grafts (MST = 97 +/- 8 days). This clearly demonstrates that transient inactivation of most T-cells with anti-CD4 plus anti-CD8 mAbs effectively controls autoimmune disease in the isograft, despite recovery of CD4 and CD8 T-cells to normal levels. Although insulitis developed in the long-term grafts, insulitis scores did not increase between 33 and 100 days, and none of the mice progressed to IDDM in 100 days. Histology showed a predominantly peri-islet T-cell and macrophage infiltrate with ductal expression of the cytokines interleukin (IL)-4, IL-2, and interferon-gamma. There was little infiltrate or expression of cytokines within the islets. Thus, mAb treatment at the time of grafting allowed isograft survival and prevented progression from insulitis to beta-cell destruction.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Survival/immunology , Immunosuppression Therapy/methods , Lymphocyte Depletion/methods , Pancreas Transplantation/immunology , Animals , Female , Insulin/biosynthesis , Islets of Langerhans/physiology , Mice , Mice, Inbred NOD , Pancreas Transplantation/pathology , Pancreas Transplantation/physiology , Time Factors , Transplantation, Isogeneic
17.
Immunol Cell Biol ; 76(6): 563-5, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9893036

ABSTRACT

To investigate the effect of IL-4 deletion on cardiac allograft survival, vascularized BALB/c cardiac allografts were placed in C57BL/6, 129Sv x C57BL/6 (IL-4 +/+) or 129Sv x C57BL/6 IL-4 knockout mice (IL-4-/-). Untreated recipients rejected allografts in < 15 days while isografts survived indefinitely (> 100 days). Treatment with anti-CD4 (GK1.5) for 4 days at the time of allografting increased mean survival to > 100 days in C57BL/6, 90+/-16 days in 129Sv x C57BL/6 (IL-4 +/+) and 68 +/- 36 days in 129Sv x C57BL/6 (IL-4-/-) recipients. Although there was a trend towards shorter survival times in the IL-4-/- mAb-treated mice, survival in the three recipient groups was not significantly different (P = 0.07). A 30-day course of anti-CD4 did not further prolong BALB/c heart survival. All long-surviving hearts had histological evidence of parenchymal damage and transplant vascular disease. None of these recipients developed antigen-specific tolerance, since both donor and third party skin graft challenges were rejected when challenged at > 60 days post-graft and all primary grafts failed by 120 days. Thus the effects of IL-4 deletion were subtle and were seen only with low doses of immunosuppression in this high responder strain combination.


Subject(s)
Graft Survival/immunology , Heart Transplantation/immunology , Interleukin-4/immunology , Animals , Antibodies, Monoclonal , Antibody Specificity , Immune Tolerance/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Rabbits , Skin Transplantation/immunology , Time Factors
18.
Transplantation ; 64(5): 684-90, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9311703

ABSTRACT

BACKGROUND: In testing new anti-CD3 agents for transplantation tolerance induction, an anti-CD3 monoclonal antibody was used as a carrier for the cytotoxic drug idarubicin (IDA). METHODS: Anti-CD3 (KT3) was covalently coupled with IDA, producing the IDA-KT3 immunoconjugate, which was tested for specificity by fluorometry and for inhibition of proliferation of CD3+ E3 cells ([3H]thymidine uptake). KT3 and IDA-KT3 were used to treat CBA recipients of BALB/c vascularized cardiac allografts. Mice with hearts surviving >100 days were challenged with donor and third-party (C57BL/6) skin. RESULTS: Conjugation to IDA did not reduce binding of KT3 to E3 cells, although the toxicity of IDA was reduced by conjugation. In BALB/c to CBA cardiac allografts (rejected in 12-17 days), both KT3 and IDA-KT3 (0.25-0.5 mg/20 g mouse i.p. at the time of transplantation) induced tolerance. Hearts survived >100 days and skin graft challenge showed indefinite survival of donor grafts but not third-party grafts. KT3 was less toxic, as measured by tumor necrosis factor-a release and blood glucose levels, than equivalent dosages of 145-2C11. At lower dosages (0.1 mg/20 g mouse), KT3-treated animals rejected BALB/c allografts in 15 to 19 days, but IDA-KT3 induced long survival (>100 days) and donor-specific tolerance in 5 of 6 mice. CONCLUSIONS: Coupling IDA to anti-CD3 reduced the in vivo toxicity of IDA and improved the immunosuppressive performance of KT3, reducing the side effects seen with other anti-CD3 agents. IDA-KT3 is a new, effective, nontoxic tolerogen in this donor-recipient combination.


Subject(s)
CD3 Complex/immunology , Idarubicin/immunology , Immune Tolerance/immunology , Immunoconjugates/pharmacology , Isoantigens/immunology , Animals , Antibodies, Monoclonal/toxicity , Cell Division/drug effects , Epitopes , Graft Survival/immunology , Heart Transplantation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Skin Transplantation/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL