Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
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 ; 15(2): 358-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25612490

ABSTRACT

Galactosyl-transferase KO (GalT-KO) pigs represent a potential solution to xenograft rejection, particularly in the context of additional genetic modifications. We have performed life supporting kidney xenotransplantation into baboons utilizing GalT-KO pigs transgenic for human CD55/CD59/CD39/HT. Baboons received tacrolimus, mycophenolate mofetil, corticosteroids and recombinant human C1 inhibitor combined with cyclophosphamide or bortezomib with or without 2-3 plasma exchanges. One baboon received a control GalT-KO xenograft with the latter immunosuppression. All immunosuppressed baboons rejected the xenografts between days 9 and 15 with signs of acute humoral rejection, in contrast to untreated controls (n = 2) that lost their grafts on days 3 and 4. Immunofluorescence analyses showed deposition of IgM, C3, C5b-9 in rejected grafts, without C4d staining, indicating classical complement pathway blockade but alternate pathway activation. Moreover, rejected organs exhibited predominantly monocyte/macrophage infiltration with minimal lymphocyte representation. None of the recipients showed any signs of porcine endogenous retrovirus transmission but some showed evidence of porcine cytomegalovirus (PCMV) replication within the xenografts. Our work indicates that the addition of bortezomib and plasma exchange to the immunosuppressive regimen did not significantly prolong the survival of multi-transgenic GalT-KO renal xenografts. Non-Gal antibodies, the alternative complement pathway, innate mechanisms with monocyte activation and PCMV replication may have contributed to rejection.


Subject(s)
Boronic Acids/therapeutic use , Complement C1 Inhibitor Protein/therapeutic use , Galactosyltransferases/genetics , Graft Survival/physiology , Heterografts , Kidney Transplantation , Plasma Exchange , Pyrazines/therapeutic use , Animals , Animals, Genetically Modified , Autoimmune Diseases , Bortezomib , Cytomegalovirus/physiology , Galactosyltransferases/deficiency , Gene Knockout Techniques , Immunity, Innate/physiology , Immunosuppressive Agents/therapeutic use , Kidney/surgery , Kidney/virology , Models, Animal , Papio anubis , Sus scrofa , Virus Replication/physiology
3.
Am J Transplant ; 14(6): 1300-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24842781

ABSTRACT

The instant blood-mediated inflammatory reaction (IBMIR) is a major obstacle to the engraftment of intraportal pig islet xenografts in primates. Higher expression of the galactose-α1,3-galactose (αGal) xenoantigen on neonatal islet cell clusters (NICC) than on adult pig islets may provoke a stronger reaction, but this has not been tested in the baboon model. Here, we report that WT pig NICC xenografts triggered profound IBMIR in baboons, with intravascular clotting and graft destruction occurring within hours, which was not prevented by anti-thrombin treatment. In contrast, IBMIR was minimal when recipients were immunosuppressed with a clinically relevant protocol and transplanted with NICC from αGal-deficient pigs transgenic for the human complement regulators CD55 and CD59. These genetically modified (GM) NICC were less susceptible to humoral injury in vitro than WT NICC, inducing significantly less complement activation and thrombin generation when incubated with baboon platelet-poor plasma. Recipients of GM NICC developed a variable anti-pig antibody response, and examination of the grafts 1 month after transplant revealed significant cell-mediated rejection, although scattered insulin-positive cells were still present. Our results indicate that IBMIR can be attenuated in this model, but long-term graft survival may require more effective immunosuppression or further donor genetic modification.


Subject(s)
Blood , Graft Rejection , Islets of Langerhans Transplantation , Transplantation, Heterologous , Animals , Antibodies/blood , Cattle , Papio
4.
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
5.
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
6.
Transplant Proc ; 43(9): 3426-30, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22099813

ABSTRACT

Galactosyl-transferase knockout (GT-KO) pigs represent the latest major progress to reduce immune reactions in xenotransplantation. However, their organs are still subject to rapid humoral rejection involving complement activation requiring the ongoing development of further genetic modifications in the pig. In a pig-to-baboon renal transplantation setting, we have used donor pigs that are not only GT-KO, but also transgenic for human CD55 (hCD55), hCD59, hCD39, and fucosyl-transferase (hHT). We studied kidney xenograft survival, physiological and immunologic parameters, xenogeneic rejection characteristics, as well as viral transmission aspects among two groups of baboons: control animals (n = 2), versus those (n = 4) treated with a cocktail of cyclophosphamide, tacrolimus, mycophenolate mofetil, steroids, and a recombinant human C1 inhibitor. Whereas control animals showed clear acute humoral rejection at around day 4, the treated animals showed moderately improved graft survival with rejection at around 2 weeks posttransplantation. Biopsies showed signs of acute vascular rejection (interstitial hemorrhage, glomerular thrombi, and acute tubular necrosis) as well as immunoglobulin (Ig)M and complement deposition in the glomerular and peritubular capillaries. The low level of preformed non-Gal-α1.3Gal IgM detected prior to transplantation increased at 6 days posttransplantation, whereas induced IgG appeared after day 6. No porcine endogenous retrovirus (PERV) transmission was detected in any transplanted baboon. Thus, surprisingly, organs from the GT-KO, hCD55, hCD59, hCD39, and hHT transgenic donors did not appear to convey significant protection against baboon anti-pig antibodies and complement activation, which obviously continue to be significant factors under a suboptimal immunosuppression regimen. The association, timing, and doses of immunosuppressive drugs remain critical. They will have to be optimized to achieve longer graft survivals.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , CD55 Antigens/metabolism , CD59 Antigens/metabolism , Fucosyltransferases/metabolism , Kidney Transplantation/immunology , Transplantation, Heterologous/immunology , Animals , Animals, Genetically Modified , Endogenous Retroviruses/metabolism , Graft Rejection , Graft Survival , Humans , Immunoglobulin G/chemistry , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/methods , Papio , Swine , Time Factors , Transplantation, Heterologous/methods
7.
Am J Physiol Heart Circ Physiol ; 300(5): H1960-70, 2011 May.
Article in English | MEDLINE | ID: mdl-21335461

ABSTRACT

Antioxidant enzymatic pathways form a critical network that detoxifies ROS in response to myocardial stress or injury. Genetic alteration of the expression levels of individual enzymes has yielded mixed results with regard to attenuating in vivo myocardial ischemia-reperfusion injury, an extreme oxidative stress. We hypothesized that overexpression of an antioxidant network (AON) composed of SOD1, SOD3, and glutathione peroxidase (GSHPx)-1 would reduce myocardial ischemia-reperfusion injury by limiting ROS-mediated lipid peroxidation and oxidative posttranslational modification (OPTM) of proteins. Both ex vivo and in vivo myocardial ischemia models were used to evaluate the effect of AON expression. After ischemia-reperfusion injury, infarct size was significantly reduced both ex vivo and in vivo, ROS formation, measured by dihydroethidium staining, was markedly decreased, ROS-mediated lipid peroxidation, measured by malondialdehyde production, was significantly limited, and OPTM of total myocardial proteins, including fatty acid-binding protein and sarco(endo)plasmic reticulum Ca(²+)-ATPase (SERCA)2a, was markedly reduced in AON mice, which overexpress SOD1, SOD3, and GSHPx-1, compared with wild-type mice. These data demonstrate that concomitant SOD1, SOD3, and GSHPX-1 expression confers marked protection against myocardial ischemia-reperfusion injury, reducing ROS, ROS-mediated lipid peroxidation, and OPTM of critical cardiac proteins, including cardiac fatty acid-binding protein and SERCA2a.


Subject(s)
Antioxidants/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Oxidative Stress/physiology , Protein Processing, Post-Translational/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Glutathione Peroxidase/metabolism , Lipid Peroxidation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
8.
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
9.
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
10.
Am J Transplant ; 8(6): 1101-12, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18444940

ABSTRACT

Incompatibility between pig thrombomodulin (TM) and primate thrombin is thought to be an important factor in the development of microvascular thrombosis in rejecting pig-to-primate xenografts. To examine this interaction at the molecular level, we cloned pig TM and measured its ability to bind human thrombin and act as a cofactor for the activation of human protein C and TAFI. The 579-residue pig TM protein showed approximately 69% sequence identity to human TM. Within the EGF domains necessary for binding of thrombin (EGF56), protein C (EGF4) and TAFI (EGF3), all of the amino acids previously identified as critical for the function of human TM, with the exception of Glu-408 in EGF5, were conserved in pig TM. Comparison of transfected cells expressing pig or human TM demonstrated that both proteins bound human thrombin and inhibited its procoagulant activity. However, pig TM was a poor cofactor for the activation of human protein C and TAFI, with domain swapping showing that EGF5 was the most important determinant of compatibility. Thus, while pig TM may be capable of binding thrombin generated in the vicinity of xenograft endothelium, its failure to promote the activation of human protein C remains a significant problem.


Subject(s)
Protein C/metabolism , Thrombin/metabolism , Thrombomodulin/metabolism , Transplantation, Heterologous/adverse effects , Animals , Carboxypeptidase B2/metabolism , Coenzymes/metabolism , Enzyme Activation , Graft Rejection/metabolism , Humans , Microcirculation , Protein Binding , Swine , Thrombosis/metabolism
11.
Transpl Immunol ; 14(1): 9-16, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15814276

ABSTRACT

The human genome possesses pseudogenes for the enzyme alpha1,3 galactosyltransferase and hence, human cells and tissues do not express the Galalpha terminated trisaccharide structure Galalpha1-3Galbeta1-4GlcNAc, the so-called alpha-Gal epitope. Circulating antibodies specific for this carbohydrate epitope are, however, present in high amounts in humans. It has previously been hypothesized that the antibody production is induced by the presence of the alpha-Gal epitope in the cell walls of the enteric flora, especially Enterobacteriaceae spp. However, in mice, in which the epitope has been deleted by targeted mutation of the gal-transferase gene, alpha-Gal antibodies do not appear without prior immunization, although the mice through their growth probably have been exposed to a normal bacterial flora of e.g. Enterobacteriaceae spp. It is unknown whether there are different types of immune reactions to antigenic carbohydrate expressing bacteria and whether there are discrepancies in the enteric flora between these knockout mice and their wild type litter mates. In this study the enteric flora of alpha-Gal knockout and wild type mice was compared both in relation to the prevalence of different types of bacteria in the two groups of mice, as well as in relation to the expression of the epitope on the surface of Enterobacteriaceae spp. Our results showed that the enteric flora did not differ significantly between knockout and wild type mice and that it was comparable to the flora known to be present in the intestines of other mice. All Enterobacteriaceae spp. examined expressed the alpha-Gal epitope no matter whether they were isolated from knockout or wild type mice. It is, therefore, discussed whether it is more reasonable to assume that alpha-Gal antibodies in mammals that do not produce alpha1,3 galactosyltransferase such as in the knock mice and in humans are the result of another antigen stimulant than these common representatives of the enteric flora, that we isolated from the two types of mice. Possible candidates for a carrier in humans could be bacteria or viruses not isolated from barrier-bred mice.


Subject(s)
Antigens, Bacterial/metabolism , Trisaccharides/metabolism , Animals , Bacteria/genetics , Bacteria/immunology , Bacteria/isolation & purification , Cecum/immunology , Cecum/microbiology , Female , Genitalia/immunology , Genitalia/microbiology , Male , Mice , Mice, Knockout , Respiratory System/immunology , Respiratory System/microbiology , Trisaccharides/genetics , Trisaccharides/immunology
12.
Transplant Proc ; 37(9): 4103-6, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16386637

ABSTRACT

INTRODUCTION: The search for alternative sources for transplant organs leads us to the search for animals as an inexhaustible source of organs. The objective of this study was to analyze whether livers from polytransgenic pigs expressing the human complement regulatory proteins CD55 (hDAF), CD59, and alfa alpha1,2-fucosyltransferase (H-transferase), protected against hyperacute rejection after orthotopic liver xenotransplantation to a baboon and also to study pig liver function in a nonhuman primate. MATERIALS AND METHODS: Nine liver transplants from pig to baboon were divided into two groups: a control group (n = 4) of genetically unmodified pigs and an experimental group (n = 5) of pigs transgenic for CD55, CD59, and H-transferase as donors. All the donating piglets obtained through hysterectomy were maintained in specific pathogen-free conditions. The selection of transgenic pig donors followed demonstration of transgene expression using monoclonal antibodies (antiCD55, antiCD59) and immunohistological studies on liver biopsies. RESULTS: All animals in the control group developed hyperacute rejection with survival rates less than 16 hours without function of transplanted livers. In the experimental group none of the animals suffered hyperacute rejection. Survival in this group was between 13 and 24 hours. The livers were functional, producing bile and maintaining above 35% prothrombin activity. Only in one case was there primary dysfunction of the xenograft. CONCLUSION: Polytransgenic livers for complement regulatory proteins prevent hyperacute rejection when xenotransplanted into a baboon.


Subject(s)
Graft Rejection/prevention & control , Graft Survival/immunology , Liver Transplantation/immunology , Transplantation, Heterologous/immunology , Acute Disease , Animals , Animals, Genetically Modified , CD55 Antigens/analysis , CD55 Antigens/genetics , CD59 Antigens/analysis , CD59 Antigens/genetics , Fucosyltransferases/genetics , Humans , Papio , Swine
13.
Am J Transplant ; 2(6): 520-5, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12118895

ABSTRACT

Delayed rejection of pig kidney xenografts by primates is associated with vascular injury that may be accompanied by a form of consumptive coagulopathy in recipients. Using a life-supporting pig-to-baboon renal xenotransplantation model, we have tested the hypothesis that treatment with recombinant human antithrombin III would prevent or at least delay the onset of rejection and coagulopathy. Non-immunosuppressed baboons were transplanted with transgenic pig kidneys expressing the human complement regulators CD55 and CD59. Recipients were treated with an intravenous infusion of antithrombin III eight hourly (250 units per kg body weight), with or without low molecular weight heparin. Antithrombin-treated recipients had preservation of normal renal function for 4-5 days, which was twice as long as untreated animals, and developed neither thrombocytopenia nor significant coagulopathy during this period. Thus, recombinant antithrombin III may be a useful therapeutic agent to ameliorate both early graft damage and the development of systemic coagulation disorders in pig-to-human xenotransplantation.


Subject(s)
Antithrombin III/pharmacology , Disseminated Intravascular Coagulation/prevention & control , Graft Rejection/prevention & control , Kidney Transplantation/adverse effects , Transplantation, Heterologous/adverse effects , Animals , Humans , Papio , Swine
15.
Transpl Immunol ; 9(1): 51-6, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11680572

ABSTRACT

Previously, we demonstrated that combination CTLA4-Fc and anti-CD40L mAb treatment results in tolerance to concordant, cellular islet xenografts. The aim of this study was to determine its effectiveness in a model of fetal pig pancreas (FPP) xenotransplantation. Survival of FPP fragment grafts were compared to the survival of rat islet or cardiac xenografts following short term CTLA4-Fc and anti-CD40L mAb treatment. Rat islet and FPP fragment grafts survived long-term. However, rat cardiac grafts were rejected by 52-91 days. Both rat islet and FPP grafts showed similar histology with intact islet structures and adjacent 'nests' of lymphocytes. Concordant vascularised rat hearts showed extensive polymorphonuclear infiltrate, concentric vasculitis and a perivascular infiltrate predominantly of CD8+ T cells. This suggests that this therapy is effective for prolonging islet xenografts and demonstrates that the cellular mechanism of rejection for vascularised and non-vascularised xenografts are different.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Differentiation/therapeutic use , CD28 Antigens/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , Heart Transplantation/immunology , Immunoconjugates , Islets of Langerhans Transplantation/immunology , Mice/immunology , Rats/immunology , Swine/immunology , Transplantation, Heterologous/immunology , Abatacept , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD , CTLA-4 Antigen , Coronary Vessels/pathology , Diabetes Mellitus, Experimental/surgery , Drug Evaluation, Preclinical , Graft Rejection/immunology , Graft Rejection/pathology , Male , Mice, Inbred CBA , Myocardium/pathology , Neutrophils/immunology , Organ Specificity , Pancreas/blood supply , Pancreas/embryology , Rats, Inbred Strains , Species Specificity , T-Lymphocytes, Cytotoxic/immunology , Vasculitis/etiology , Vasculitis/immunology , Vasculitis/pathology
16.
Euro Surveill ; 6(2): 26-30, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11682709

ABSTRACT

A study on the coverage of influenza vaccination among elderly people was carried out. Fity-six per cent of those interviewed had been vaccinated (weighted coverage 50%), compared with the target of 75%, but higher rates were reported in those older than 74 years and those with chronic diseases.


Subject(s)
Influenza Vaccines/administration & dosage , Vaccination/statistics & numerical data , Aged , Chronic Disease , Female , Health Care Surveys , Humans , Italy , Male , Sex Distribution
17.
Gene Ther ; 8(12): 897-904, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11426329

ABSTRACT

The involvement of the vascular endothelium in a large number of diseases supports the importance of vascular-specific gene delivery for their treatment. The hereditary hemorrhagic telangiectasia type 1 is an example of a vascular inherited disease (OMIM 187300). This is an autosomal dominant vascular disorder originated by mutations in the endoglin gene and associated with frequent epistaxis, telangiectases, gastrointestinal bleedings, and arteriovenous malformations in brain, lung and liver. Here, we address for the first time the possibility of using in vivo gene transfer to target endoglin expression to the vasculature. The promoter of the endothelial gene, ICAM-2, was used to generate transgenic animals which demonstrated endothelial expression of endoglin. Next, the promoters of the human endothelial genes, endoglin and ICAM-2, were inserted upstream of the human endoglin cDNA, and the resulting constructs were systemically or locally delivered, demonstrating endoglin expression in the vessel walls of liver, lung and skin. These gene transfer experiments represent an initial step in the treatment of the hereditary hemorrhagic telangiectasia type 1 by gene therapy, and suggest that endoglin and ICAM-2 promoters can be used to deliver other genes to the endothelium specifically.


Subject(s)
Antigens, CD/genetics , Cell Adhesion Molecules/genetics , Endothelium, Vascular/metabolism , Gene Targeting/methods , Promoter Regions, Genetic , Telangiectasia, Hereditary Hemorrhagic/therapy , Vascular Cell Adhesion Molecule-1/genetics , Animals , Blotting, Western/methods , Endoglin , Gene Expression , Genetic Therapy/methods , Humans , Immunohistochemistry/methods , Liver/metabolism , Lung/metabolism , Mice , Mice, Transgenic , Receptors, Cell Surface , Skin/metabolism , Vascular Cell Adhesion Molecule-1/analysis
18.
Transgenic Res ; 10(6): 523-31, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11817540

ABSTRACT

A retrospective analysis of transgenesis rates obtained in seven pronuclear microinjection programs was undertaken to determine if a relationship existed between the amount of DNA injected and transgenesis rates in the pig. Logistic regression analysis showed that as the concentration of DNA injected increased from 1 to 10 ng/microl, the number of transgenics when expressed as a proportion of the number liveborn (integration rate) increased from 4% to an average of 26%. A similar relationship was found when the number of molecules of DNA injected per picolitre was analysed. No evidence was obtained to suggest either parameter influenced integration rate in mice when the same constructs were injected. The number of transgenics liveborn when expressed as a proportion of ova injected (efficiency rate), increased as DNA concentration increased up to 7.5 ng/microl and then decreased at 10 ng/microl for both species suggesting that at this concentration DNA (or possible contaminants) may have influenced embryo survival. The relationship between efficiency and the number of molecules injected per picolitre was complex suggesting that the concentration at which DNA was injected was a better determinant of integration and efficiency rates. In conclusion, the present study suggests that transgenes need to be injected at concentrations of between 5 and 10 ng/microl to maximise integration and efficiency rates in pigs.


Subject(s)
Animals, Genetically Modified , Gene Transfer Techniques , Mice, Transgenic , Animals , Blotting, Southern , DNA/analysis , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Retrospective Studies , Swine , Transgenes
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...