Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Transplant Proc ; 48(6): 2200-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27569971

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 1/cirurgia , Transplante das Ilhotas Pancreáticas , Receptores de Superfície Celular/metabolismo , Transplantes/metabolismo , Animais , Apoptose , Glicemia/análise , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Tipo 1/sangue , Receptor de Proteína C Endotelial , Sobrevivência de Enxerto , Humanos , Insulina/análise , Masculino , Camundongos , Camundongos Transgênicos , Substâncias Protetoras/metabolismo , Proteína C/metabolismo , Suínos
2.
Am J Transplant ; 15(2): 358-70, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25612490

RESUMO

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.


Assuntos
Ácidos Borônicos/uso terapêutico , Proteína Inibidora do Complemento C1/uso terapêutico , Galactosiltransferases/genética , Sobrevivência de Enxerto/fisiologia , Xenoenxertos , Transplante de Rim , Troca Plasmática , Pirazinas/uso terapêutico , Animais , Animais Geneticamente Modificados , Doenças Autoimunes , Bortezomib , Citomegalovirus/fisiologia , Galactosiltransferases/deficiência , Técnicas de Inativação de Genes , Imunidade Inata/fisiologia , Imunossupressores/uso terapêutico , Rim/cirurgia , Rim/virologia , Modelos Animais , Papio anubis , Sus scrofa , Replicação Viral/fisiologia
3.
Am J Transplant ; 14(6): 1300-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24842781

RESUMO

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.


Assuntos
Sangue , Rejeição de Enxerto , Transplante das Ilhotas Pancreáticas , Transplante Heterólogo , Animais , Anticorpos/sangue , Bovinos , Papio
4.
Am J Transplant ; 12(9): 2363-72, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22681753

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Endotélio Vascular/metabolismo , Glicoproteínas/metabolismo , Modelos Animais , Receptores de Superfície Celular/metabolismo , Animais , Receptor de Proteína C Endotelial , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Traumatismo por Reperfusão/prevenção & controle
5.
Am J Transplant ; 10(2): 242-50, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20055798

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Ciclosporina/farmacologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Ratos , Suínos , Trombomodulina , Transgenes/efeitos dos fármacos
6.
Am J Transplant ; 8(6): 1101-12, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18444940

RESUMO

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.


Assuntos
Proteína C/metabolismo , Trombina/metabolismo , Trombomodulina/metabolismo , Transplante Heterólogo/efeitos adversos , Animais , Carboxipeptidase B2/metabolismo , Coenzimas/metabolismo , Ativação Enzimática , Rejeição de Enxerto/metabolismo , Humanos , Microcirculação , Ligação Proteica , Suínos , Trombose/metabolismo
7.
Am J Transplant ; 2(6): 520-5, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12118895

RESUMO

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.


Assuntos
Antitrombina III/farmacologia , Coagulação Intravascular Disseminada/prevenção & controle , Rejeição de Enxerto/prevenção & controle , Transplante de Rim/efeitos adversos , Transplante Heterólogo/efeitos adversos , Animais , Humanos , Papio , Suínos
9.
Xenotransplantation ; 7(4): 237-46, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11081758

RESUMO

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.


Assuntos
Dissacarídeos/imunologia , Galactosiltransferases/metabolismo , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Doença Aguda , Animais , Anticorpos Heterófilos/sangue , Arteríolas/patologia , Vasos Coronários/patologia , Epitopos/imunologia , Transfusão de Eritrócitos , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Rejeição de Enxerto/patologia , Transplante de Coração/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Coelhos
10.
Transplantation ; 69(12): 2504-15, 2000 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-10910270

RESUMO

BACKGROUND: The genetic modification of pigs is a powerful strategy that may ultimately enable successful xenotransplantation of porcine organs into humans. METHODS: Transgenic pigs were produced by microinjection of gene constructs for human complement regulatory proteins CD55 and CD59 and the enzyme alpha1,2-fucosyltransferase (H-transferase, HT), which reduces expression of the major xenoepitope galactose-alpha1,3-galactose (alphaGal). Kidneys from CD55/HT and CD55/CD59/HT transgenic pigs were transplanted into nephrectomised, nonimmunosuppressed adult baboons. RESULTS: In several lines of transgenic pigs, CD55 and CD59 were expressed strongly in all tissues examined, whereas HT expression was relatively weak and did not significantly reduce alphaGal. Control nontransgenic kidneys (n=4) grafted into baboons were hyperacutely rejected within 1 hr. In contrast, kidneys from CD55/HT pigs (n=2) were rejected after 30 hr, although kidneys from CD55/CD59/HT pigs (n=6) maintained function for up to 5 days. In the latter grafts, infiltration by macrophages, T cells, and B cells was observed at days 3 and 5 posttransplantation. The recipients developed thrombocytopenia and abnormalities in coagulation, manifested in increased clotting times and an elevation in the plasma level of the fibrin degradation product D-dimer, within 2 days of transplantation. Treatment with low molecular weight heparin prevented profound thrombocytopenia but not the other aspects of coagulopathy. CONCLUSIONS: Strong expression of CD55 and CD59 completely protected porcine kidneys from hyperacute rejection and allowed a detailed analysis of xenograft rejection in the absence of immunosuppression. Coagulopathy appears to be a common feature of pig-to-baboon renal transplantation and represents yet another major barrier to its clinical application.


Assuntos
Transtornos da Coagulação Sanguínea/etiologia , Antígenos CD59/fisiologia , Fucosiltransferases/fisiologia , Rejeição de Enxerto , Transplante de Rim/imunologia , Transplante Heterólogo/imunologia , Animais , Antígenos CD59/análise , Antígenos CD59/genética , Fucosiltransferases/genética , Imuno-Histoquímica , Terapia de Imunossupressão , Rim/patologia , Transplante de Rim/efeitos adversos , Camundongos , Papio , Suínos , Galactosídeo 2-alfa-L-Fucosiltransferase
11.
Xenotransplantation ; 7(1): 42-7, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10809056

RESUMO

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.


Assuntos
Dissacarídeos/imunologia , Rejeição de Enxerto/imunologia , Isoanticorpos/fisiologia , Modelos Imunológicos , Transplante Heterólogo/imunologia , Transplante Homólogo/imunologia , Animais , Dissacarídeos/genética , Feminino , Rejeição de Enxerto/patologia , Transplante de Coração/imunologia , Transplante de Coração/patologia , Humanos , Imunidade Inata/genética , Isoanticorpos/biossíntese , Isoanticorpos/sangue , Isoanticorpos/genética , Masculino , Camundongos , Camundongos Knockout , Transplante Heterólogo/patologia , Transplante Homólogo/patologia
12.
Transplantation ; 66(6): 748-54, 1998 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-9771838

RESUMO

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.


Assuntos
Anticorpos/imunologia , Dissacarídeos/imunologia , Galactosiltransferases/genética , Rejeição de Enxerto/imunologia , Transplante Heterólogo/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Feminino , Galactosiltransferases/metabolismo , Rejeição de Enxerto/genética , Rejeição de Enxerto/patologia , Transplante de Coração/imunologia , Leishmania major/imunologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Miocárdio/patologia
13.
Transplantation ; 65(12): 1599-604, 1998 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-9665076

RESUMO

BACKGROUND: Organs from transgenic animals with high-level endothelial expression of the human complement regulatory factors CD55 and CD59 are significantly protected from human complement-mediated injury. Elimination or reduction of the major xenoepitope alphaGal, achieved by knocking out the alpha1,3-galactosyltransferase gene (Gal KO) or expressing human alpha1,2-fucosyltransferase (H transferase or HTF), also affords protection, although to a lesser degree. In this study, we examined whether the protection provided by strong CD55 and CD59 expression can be augmented by the Gal KO or HTF modifications. METHODS: Hearts from four groups of mice (wild type, CD55/CD59, CD55/CD59/HTF, and CD55/CD59/Gal KO) were perfused ex vivo with 40% human plasma. Mean heart work for each group was compared over a 60-min period. RESULTS: Wild-type hearts ceased to function effectively within 15 min of plasma addition. CD55/CD59 hearts displayed prolonged survival and maintained approximately 10% maximum work at the end of perfusion. Introduction of Gal KO or HTF onto the CD55/CD59 background resulted in a further prolongation, with work maintained at 20-30% of the maximum level. CONCLUSIONS: We used an ex vivo model to demonstrate that eliminating alphaGal expression further prolongs the function of mouse hearts expressing high levels of CD55 and CD59. In addition, we showed that reducing alphaGal by expressing HTF is equally as effective in prolonging CD55/CD59 heart function as knocking out Gal transferase, thus providing a feasible strategy for translating these advances to the pig.


Assuntos
Antígenos CD55/análise , Antígenos CD59/análise , Fucosiltransferases/fisiologia , Galactosiltransferases/fisiologia , Rejeição de Enxerto/prevenção & controle , Transplante de Coração , Animais , Galactosiltransferases/genética , Humanos , Camundongos , Camundongos Knockout , Miocárdio/imunologia , Miocárdio/patologia , Transplante Heterólogo , Galactosídeo 2-alfa-L-Fucosiltransferase
14.
Transplantation ; 65(6): 832-7, 1998 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-9539096

RESUMO

BACKGROUND: The expression of human alpha1,2-fucosyltransferase (H-transferase, HT) has been proposed as an alternative strategy to alpha1,3-galactosyltransferase (GT) gene knockout, which is not currently feasible in pigs, to reduce the galactose-alpha1,3-galactose (Gal) epitope expression. HT expression has recently been shown in transgenic mice and pigs to significantly reduce Gal expression on a variety of cells; however, its ability to do so on endothelial cells and its effectiveness at prolonging xenograft survival are yet to be determined. METHODS: HT-transgenic, Gal knockout (Gal KO) mice, and mice containing both genetic modifications (HT-transgenic/Gal KO) were tested for H-substance and Gal expression on splenocytes and endothelial cells by flow cytometric analysis. In addition, the hearts of these mice were perfused ex vivo with 6% human plasma, and the effect on cardiac function was determined. RESULTS AND CONCLUSION: H-substance expression was detected on both splenocytes and endothelial cells of HT-transgenic mice. The level of H-substance expression was not affected by the presence or absence of GT in the cells, consistent with HT being dominant over GT. The ability of HT expression to reduce Gal expression was highly variable depending on the cell type. Gal expression on splenocytes was almost completely eliminated, whereas on endothelial cells, substantial Gal remained despite a 70% reduction. When perfused ex vivo with human plasma, hearts from HT-transgenic, Gal KO, and HT-transgenic/Gal KO mice demonstrated a similar prolongation in survival, compared with wild-type controls. Therefore, as far as hyperacute rejection is concerned, HT expression may be as effective as Gal KO in protecting against xenoantibody and complement mediated injury. However, the effect of residual Gal on non-hyperacute rejection responses remains to be determined.


Assuntos
Fucosiltransferases/metabolismo , Rejeição de Enxerto , Transplante de Coração/imunologia , Lectinas de Plantas , Animais , Antígenos de Superfície/imunologia , Dissacarídeos/imunologia , Dissacarídeos/metabolismo , Endotélio Vascular/enzimologia , Endotélio Vascular/imunologia , Humanos , Lectinas , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/imunologia , Baço/enzimologia , Baço/imunologia , Transplante Heterólogo , Galactosídeo 2-alfa-L-Fucosiltransferase
15.
Transplantation ; 64(6): 882-8, 1997 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-9326415

RESUMO

BACKGROUND: Hyperacute rejection (HAR) currently prevents the use of pigs as organ donors for humans. It is now generally accepted that the key instigators of HAR are naturally occurring xenoantibodies against the terminal disaccharide galactose alpha1,3-galactose (Gal), and the species incompatibility between human complement and porcine complement regulatory molecules. Using two in vitro models and an ex vivo mouse heart perfusion model, we have shown previously that cells and tissues from Gal knockout (Gal KO) and transgenic mice expressing the human cell surface complement regulator decay-accelerating factor (DAF/CD55) are partially, but not completely, protected from human complement-mediated injury. METHODS: In the present study, Gal KO mice were crossed with DAF transgenic mice and bred to homozygosity (DAF/Gal KO). Isolated splenocytes were incubated with human serum, and the protective effect of DAF and Gal KO was assessed by measuring complement deposition and cell lysis. Hearts perfused ex vivo with human plasma were examined for human antibody and complement deposition, and assessed functionally by measuring work performed by the heart. RESULTS: Splenocytes from DAF/Gal KO mice were found to be more resistant to complement-mediated injury than cells from either DAF transgenic or Gal KO mice. In addition, hearts from DAF/Gal KO mice, when perfused with human plasma, displayed prolonged survival compared with hearts from Gal KO mice. This was associated with a reduction in the extent of endothelial deposition of IgG, IgM, and complement C3b. CONCLUSIONS: These findings demonstrate that expression of human DAF in association with elimination of the Gal epitope provides added protection from complement-mediated injury in these models of HAR.


Assuntos
Antígenos CD55/biossíntese , Proteínas do Sistema Complemento/toxicidade , Galactosiltransferases/deficiência , Transplante Heterólogo , Animais , Antígenos CD55/genética , Sobrevivência Celular , Células Cultivadas , Complemento C3/metabolismo , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Epitopos , Galactosiltransferases/genética , Homozigoto , Humanos , Linfócitos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miocárdio , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Baço/imunologia , Suínos
16.
Transplantation ; 64(2): 197-204, 1997 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-9256173

RESUMO

BACKGROUND: Inactivation of the alpha1,3-galactosyltransferase (GalT) gene by homologous recombination (knockout [KO] mice) and competition for the enzyme's N-acetyllactosamine substrate by transgenically expressed alpha1,2-fucosyltransferase (H-transferase) are two genetic approaches to elimination of the Gal alpha1,3Gal (alphaGal) epitope, which is the major xenoantigen in pigs against which humans have preformed antibodies. Such genetic manipulations often have unpredictable results. METHODS: A panel of 19 selected lectins was used to characterize the changes in cell surface glycosylation in GalT KO and H-transferase transgenic mice, compared with nontransgenic littermate controls. RESULTS: GalT KO mice showed complete elimination of the alphaGal epitope, as reported previously. Surprisingly, however, this was associated with only a modest increase in N-acetyllactosamine residues and had little other effect on the pattern of lectin binding. In contrast, the pattern of lectin binding to H-transferase transgenic mouse cells was more profoundly disturbed and indicated, in addition to the expected expression of H substance and suppression of the alphaGal epitope, that there was a marked reduction in alpha2,3-sialylation and exposure of the normally cryptic antigens, sialylated Tn and Forssman antigens. Similar changes in lectin reactivity with porcine aortic endothelial cells were induced by neuraminidase treatment. CONCLUSIONS: Lectins were able to bind underlying carbohydrate structures (sialylated Tn and Forssman antigens) that are normally cryptic antigens on H-transferase transgenic mouse spleen and cardiac endothelial cells, probably as a consequence of the reduction in the electronegativity of the cell surface due to reduced sialylation. As humans have preformed anti-Tn and anti-Forssman antibodies, it is possible that these structures may become targets of the xenograft rejection process, including hyperacute rejection.


Assuntos
Fucosiltransferases/genética , Galactosiltransferases/genética , Camundongos Knockout/genética , Camundongos Knockout/metabolismo , Camundongos Transgênicos/genética , Camundongos Transgênicos/metabolismo , Transplante Heterólogo/imunologia , Adsorção , Animais , Aorta/metabolismo , Sangue/metabolismo , Sequência de Carboidratos , Membrana Celular/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Epitopos/genética , Galactosiltransferases/imunologia , Glicosilação , Humanos , Imuno-Histoquímica , Lectinas/metabolismo , Camundongos , Miocárdio/química , Miocárdio/citologia , Baço/química , Baço/citologia , Suínos
17.
J Immunol ; 150(10): 4395-406, 1993 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-7683317

RESUMO

The suspension pro granulocyte/macrophage (SPGM1) cell line was established from a transplantable mouse progranulocytic/promacrophage tumor. Surprisingly, SPGM1 cells expressed a typical CD5 pre-B cell phenotype, being positive for Ly-1 (CD5), PB76, B220 (CD45RA), and the pre-B Ig receptor complex (microH chains, lambda 5 and vpre-B surrogate L chains, and the IgM alpha (mb-1) and Ig beta (B29) co-receptor molecules). Southern Blot analysis revealed clonal rearrangement of the microH chain locus and germ-line L chain loci. SPGM1 formed blast cell-, macrophage-, and occasional granulocytic colonies in soft agar in the presence of murine IL-3. IL-3 also induced macrophage differentiation of SPGM1 cells in suspension cultures. The earliest changes were detectable at 24 h by Northern blot analysis. IL-3-treatment increased Mac1 mRNA, induced c-fms mRNA, and down-regulated mRNA for mu, lambda 5, vpre-B and mb-1. After 2 to 4 days the cells were larger, strongly adherent, expressed the macrophage markers Mac1 and F4/80, had lost microH chain and PB76 surface expression, and readily phagocytosed latex beads. Thus SPGM1 has all the characteristic features of a CD5+ pre-B cell line. However, IL-3 predominantly induced SPGM1 to switch its differentiation program from a pre-B cell to a macrophage. This inducible, rapid switch of virtually the entire population provides a unique model for the molecular analysis of mechanisms involved in cell-fate determination.


Assuntos
Antígenos CD/análise , Linfócitos B/citologia , Macrófagos/citologia , Animais , Antígenos de Superfície/genética , Linfócitos B/fisiologia , Antígenos CD5 , Diferenciação Celular , Linhagem Celular , Feminino , Expressão Gênica , Genes fms , Cadeias mu de Imunoglobulina/análise , Cadeias mu de Imunoglobulina/genética , Imunofenotipagem , Técnicas In Vitro , Interleucina-3/farmacologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C3H , RNA Mensageiro/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptores de Antígenos de Linfócitos B/análise , Receptores de Antígenos de Linfócitos B/genética , Células Tumorais Cultivadas
18.
Blood ; 80(1): 102-12, 1992 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-1377046

RESUMO

We describe the development of a human bone marrow (BM) culture system which allows study of the interaction of stromal cell lines (SCL) and highly purified hematopoietic progenitor cells. Normal BM stromal cells were electroporated with a plasmid containing the simian virus 40 (SV40) large T antigen (SV40 T Ag) under the control of a synthetic metallothionein promoter (MT4); this construct is designated MT4 SV40 T Ag. SCL in which the rate of proliferation could be controlled by altering the zinc (Zn) concentration were characterized, demonstrating that the SCL were heterogeneous with respect to G-CSF and GM-CSF production. Suppression of SCL proliferation on removal of Zn made it possible to use these lines in coculture with purified CD34+ progenitor cells from umbilical cord blood. The ability to control proliferation of SCL has allowed us to maintain the survival and expansion of colony-forming cells in culture for up to 2 months. These lines have enabled us to test for stromal cell characteristics at a clonal level and provided us with a tool to analyze the events leading to lineage commitment and hematopoietic differentiation, as demonstrated by suppression of hematopoiesis by an antibody directed against the c-kit molecule.


Assuntos
Células da Medula Óssea , Hematopoese , Células-Tronco Hematopoéticas/citologia , Antígenos CD/análise , Antígenos CD34 , Antígenos Transformantes de Poliomavirus/genética , Antígenos de Superfície/análise , Medula Óssea/imunologia , Divisão Celular/efeitos dos fármacos , Transformação Celular Viral , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Fatores de Crescimento de Células Hematopoéticas/biossíntese , Células-Tronco Hematopoéticas/imunologia , Humanos , Técnicas In Vitro , Interleucina-6/biossíntese , Metalotioneína/genética , Fator de Crescimento Derivado de Plaquetas/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Fator de Células-Tronco , Zinco/farmacologia
19.
Leuk Res ; 16(6-7): 655-63, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1378917

RESUMO

In this report we describe two newly isolated pre-B acute lymphoblastic leukaemia cell lines. Both cell lines lack EBV as detected by the EBNA-1 gene probed Southern-blots. Neither cell line expressed the B-cell-specific CD20 antigen on the cell membrane. However surface expression of CD20 was induced by phorbol ester (TPA) on both LiLa-1 and LK63 cell lines. Other pre-B and B-cell lines, such as Reh, Nalm-1, and BALL-1 did not exhibit these changes in phenotype. Previous immunoprecipitation studies have noted that a broad 50-55 kD band co-precipitates with the characteristic 33-37 kD CD20 protein. We demonstrate that, while the 33-37 kD CD20 species was undetectable on resting LiLa-1 and LK63 cells, in each case a 50-55 kD protein was immunoprecipitated by the CD20 antibody. However, the failure to detect any cell surface CD20-associated antigen on the control cells by immunophenotyping indicated that the CD20 epitope of the 50-55 kD molecule was not expressed on the cell surface. Following exposure to TPA the 50-55 kD species was reduced over 48-72 h while the level of the p33-37 CD20 protein was increased. Northern-blot analysis showed that the 50-55 kD protein was not a cryptic form of CD20 as the uninduced cells contained no detectable CD20 mRNA. The decrease of the 50-55 kD protein and the acquisition of the mature CD20 molecule were paralleled by a decline in proliferative activity in both cell lines. As expression of CD20 by normal pre-B cells also coincides with the cessation of cell division and maturation towards a mature B-cell phenotype, these cell lines appear to represent models for a discrete stage of B-cell differentiation which may be valuable in defining the signals regulating pre-B-cell proliferation.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Antígenos CD/metabolismo , Antígenos CD20 , Antígenos de Diferenciação de Linfócitos B/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Northern Blotting , Southern Blotting , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Imunofenotipagem , Proteínas de Membrana/metabolismo , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Fosforilação , Testes de Precipitina , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
20.
Proc Natl Acad Sci U S A ; 89(5): 1611-5, 1992 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-1311845

RESUMO

We describe the molecular cloning of a receptor tyrosine kinase from a cell line (LK63) derived from a case of human pre-B-cell leukemia. We have previously shown that a monoclonal antibody (IIIA4) raised against LK63 recognized a glycosylated, cell-surface 135-kDa molecule (HEK), which displayed tyrosine kinase activity in vitro. The HEK protein was purified by using a IIIA4 antibody column and both N-terminal and internal amino acid sequences were obtained. A 51-mer degenerate oligonucleotide based on the internal amino acid sequence was used to screen an LK63-derived lambda gt10 cDNA library under low-stringency hybridization conditions. One clone of 2.5 kilobases (kb) was isolated and characterized and used to rescreen the library under more-stringent hybridization conditions. A 4.5-kb clone containing the entire HEK coding region was isolated and its complete DNA sequence was determined. The 4.5-kb insert was subcloned into the expression vector CDM8 and transfected into COS cells. COS cells transfected with the sense HEK/CDM8 construct stained specifically with the IIIA4 antibody, thereby confirming that the antigen recognized by the IIIA4 antibody and the expressed protein product of the HEK cDNA clone were identical. DNA sequence analysis revealed that HEK is a newly discovered member of the EPH/ELK family of receptor tyrosine kinases. Northern blot analysis of a number of cell lines demonstrated the expression of 5.5- to 6.0-kb HEK transcripts in LK63 and the T-cell lines JM and HSB-2. Southern blot analysis of DNA from LK63 suggested that the HEK gene was neither amplified nor rearranged in the LK63 tumor.


Assuntos
Linfócitos/enzimologia , Proteínas de Neoplasias/genética , Proteínas Tirosina Quinases/genética , Receptores de Superfície Celular/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Southern Blotting , Chlorocebus aethiops , Clonagem Molecular , DNA/genética , Expressão Gênica , Genes , Técnicas In Vitro , Dados de Sequência Molecular , Família Multigênica , RNA Mensageiro/genética , Receptor EphA3 , Alinhamento de Sequência , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...