Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Xenotransplantation ; 22(6): 413-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26490547

RESUMO

BACKGROUND: Survival of vascularized xenografts is dependent on pre-emptive inhibition of the xenoantibody response against galactosyltransferase knockout (GTKO) porcine organs. Our analysis in multiple GTKO pig-to-primate models of xenotransplantation has demonstrated that the anti-non-gal-α-1,3-gal (anti-non-Gal) xenoantibody response displays limited structural diversity. This allowed our group to identify an experimental compound which selectively inhibited induced anti-non-Gal IgM xenoantibodies. However, because this compound had an unknown safety profile, we extended this line of research to include screening small molecules with known safety profiles allowing rapid advancement to large animal models. METHODS: The NIH clinical collections of small molecules were screened by ELISA for their ability to inhibit xenoantibody binding to GTKO pig endothelial cells. Serum collected from non-immunosuppressed rhesus monkeys at day 14 post-injection with GTKO pig endothelial cells was utilized as a source of elicited xenoantibody for initial screening. Virtual small molecule screening based on xenoantibody structure was used to assess the likelihood that the identified small molecules bound xenoantibody directly. As a proxy for selectivity, ELISAs against tetanus toxoid and the natural antigens laminin, thyroglobulin, and single-stranded DNA (ssDNA) were utilized to assess the ability of the identified reagents to inhibit additional antibody responses. The identified inhibitory small molecules were further tested for their ability to inhibit xenoantibody elicited in multiple settings, including rhesus monkeys pre-treated with an anti-non-Gal selective anti-idiotypic antibody, non-immunosuppressed rhesus monkeys immunized with wild-type fetal pig isletlike cell clusters, and non-immunosuppressed baboons transplanted with GTKO multiple transgenic pig kidneys. RESULTS: Four clinically relevant small molecules inhibited anti-non-Gal IgM binding to GTKO pig endothelial cells in vitro. Three of these drugs displayed a limited region of structural similarity suggesting they may inhibit xenoantibody by a similar mechanism. One of these, the anti-hypertensive agent clonidine, displayed only minimal inhibition of antibodies elicited by vaccination against tetanus toxoid or pre-existing natural antibodies against laminin, thyroglobulin, or ssDNA. Furthermore, clonidine inhibited elicited anti-non-Gal IgM from all animals that demonstrated a xenoantibody response in each experimental setting. CONCLUSIONS: Clinically relevant small molecule drugs with known safety profiles can inhibit xenoantibody elicited against non-Gal antigens in diverse experimental xenotransplantation settings. These molecules are ready to be tested in large animal models. However, it will first be necessary to optimize the timing and dosing required to inhibit xenoantibodies in vivo.


Assuntos
Anticorpos Heterófilos/sangue , Clonidina/farmacologia , Xenoenxertos/imunologia , Papio/imunologia , Animais , Técnicas de Inativação de Genes , Imunoglobulina M/imunologia , Macaca mulatta , Modelos Animais , Sus scrofa , Suínos , Transplante Heterólogo/métodos
2.
Cell ; 159(6): 1277-89, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25480293

RESUMO

Glycosylation processes are under high natural selection pressure, presumably because these can modulate resistance to infection. Here, we asked whether inactivation of the UDP-galactose:ß-galactoside-α1-3-galactosyltransferase (α1,3GT) gene, which ablated the expression of the Galα1-3Galß1-4GlcNAc-R (α-gal) glycan and allowed for the production of anti-α-gal antibodies (Abs) in humans, confers protection against Plasmodium spp. infection, the causative agent of malaria and a major driving force in human evolution. We demonstrate that both Plasmodium spp. and the human gut pathobiont E. coli O86:B7 express α-gal and that anti-α-gal Abs are associated with protection against malaria transmission in humans as well as in α1,3GT-deficient mice, which produce protective anti-α-gal Abs when colonized by E. coli O86:B7. Anti-α-gal Abs target Plasmodium sporozoites for complement-mediated cytotoxicity in the skin, immediately after inoculation by Anopheles mosquitoes. Vaccination against α-gal confers sterile protection against malaria in mice, suggesting that a similar approach may reduce malaria transmission in humans.


Assuntos
Escherichia coli/fisiologia , Imunoglobulina M/imunologia , Malária Falciparum/imunologia , Malária Falciparum/transmissão , Plasmodium/fisiologia , Polissacarídeos/imunologia , Adulto , Animais , Anopheles/parasitologia , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Autoantígenos/imunologia , Linhagem Celular Tumoral , Criança , Escherichia coli/classificação , Escherichia coli/imunologia , Feminino , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Trato Gastrointestinal/microbiologia , Vida Livre de Germes , Humanos , Imunoglobulina M/sangue , Malária Falciparum/microbiologia , Malária Falciparum/parasitologia , Camundongos , Plasmodium/classificação , Plasmodium/crescimento & desenvolvimento , Plasmodium/imunologia , Plasmodium falciparum/imunologia , Plasmodium falciparum/fisiologia , Esporozoítos/imunologia , Receptor Toll-Like 9/agonistas
4.
Xenotransplantation ; 21(4): 341-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24806998

RESUMO

BACKGROUND: Xenotransplantation of porcine organs holds promise of solving the human organ donor shortage. The use of α-1,3-galactosyltransferase knockout (GTKO) pig donors mitigates hyperacute rejection, while delayed rejection is currently precipitated by potent immune and hemostatic complications. Previous analysis by our laboratory suggests that clotting factor VIII (FVIII) inhibitors might be elicited by the structurally restricted xenoantibody response which occurs after transplantation of either pig GTKO/hCD55/hCD59/hHT transgenic neonatal islet cell clusters or GTKO endothelial cells. METHODS: A recombinant xenoantibody was generated using sequences from baboons demonstrating an active xenoantibody response at day 28 after GTKO/hCD55/hCD59/hHT transgenic pig neonatal islet cell cluster transplantation. Rhesus monkeys were immunized with GTKO pig endothelial cells to stimulate an anti-non-Gal xenoantibody response. Serum was collected at days 0 and 7 after immunization. A two-stage chromogenic assay was used to measure FVIII cofactor activity and identify antibodies which inhibit FVIII function. Molecular modeling and molecular dynamics simulations were used to predict antibody structure and the residues which contribute to antibody-FVIII interactions. Competition ELISA was used to verify predictions at the domain structural level. RESULTS: Antibodies that inhibit recombinant human FVIII function are elicited after non-human primates are transplanted with either GTKO pig neonatal islet cell clusters or endothelial cells. There is an apparent increase in inhibitor titer by 15 Bethesda units (Bu) after transplant, where an increase greater than 5 Bu can indicate pathology in humans. Furthermore, competition ELISA verifies the computer modeled prediction that the recombinant xenoantibody, H66K12, binds the C1 domain of FVIII. CONCLUSIONS: The development of FVIII inhibitors is a novel illustration of the potential impact the humoral immune response can have on coagulative dysfunction in xenotransplantation. However, the contribution of these antibodies to rejection pathology requires further evaluation because "normal" coagulation parameters after successful xenotransplantation are not fully understood.


Assuntos
Fator VIII/antagonistas & inibidores , Transplante das Ilhotas Pancreáticas/efeitos adversos , Macaca mulatta/imunologia , Papio/imunologia , Transplante Heterólogo/efeitos adversos , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Anticorpos Heterófilos/biossíntese , Anticorpos Heterófilos/química , Anticorpos Heterófilos/genética , Simulação por Computador , Células Endoteliais/imunologia , Células Endoteliais/transplante , Fator VIII/química , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Técnicas de Inativação de Genes , Humanos , Transplante das Ilhotas Pancreáticas/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homologia de Sequência de Aminoácidos , Sus scrofa
5.
Xenotransplantation ; 21(3): 244-53, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24645827

RESUMO

BACKGROUND: Promising developments in porcine islet xenotransplantation could resolve the donor pancreas shortage for patients with type 1 diabetes. Using α1,3-galactosyltransferase gene knockout (GTKO) donor pigs with multiple transgenes should extend xenoislet survival via reducing complement activation, thrombus formation, and the requirement for exogenous immune suppression. Studying the xenoantibody response to GTKO/hCD55/hCD59/hHT islets in the pig-to-baboon model, and comparing it with previously analyzed responses, would allow the development of inhibitory reagents capable of targeting conserved idiotypic regions. METHODS: We generated IgM heavy and light chain gene libraries from 10 untreated baboons and three baboons at 28 days following transplantation of GTKO/hCD55/hCD59/hHT pig neonatal islet cell clusters with immunosuppression. Flow cytometry was used to confirm the induction of a xenoantibody response. IgM germline gene usage was compared pre- and post-transplant. Homology modeling was used to compare the structure of xenoantibodies elicited after transplantation of GTKO/hCD55/hCD59/hHT pig islets with those induced by GTKO and wild-type pig endothelial cells without further genetic modification. RESULTS: IgM xenoantibodies that bind to GTKO pig cells and wild-type pig cells were induced after transplantation. These anti-non-Gal antibodies were encoded by the IGHV3-66*02 (Δ28%) and IGKV1-12*02 (Δ25%) alleles, for the immunoglobulin heavy and light chains, respectively. IGHV3-66 is 86.7% similar to IGHV3-21 which was elicited by rhesus monkeys in response to GTKO endothelial cells. Heavy chain genes most similar to IGHV3-66 were found to utilize the IGHJ4 gene in 85% of V-D regions analyzed. However, unlike the wild-type response, a consensus complementary determining region 3 was not identified. CONCLUSIONS: Additional genetic modifications in transgenic GTKO pigs do not substantially modify the structure of the restricted group of anti-non-Gal xenoantibodies that mediate induced xenoantibody responses with or without immunosuppression. The use of this information to develop new therapeutic agents to target this restricted response will likely be beneficial for long-term islet cell survival and for developing targeted immunosuppressive regimens with less toxicity.


Assuntos
Animais Geneticamente Modificados , Anticorpos Heterófilos/metabolismo , Rejeição de Enxerto/imunologia , Imunoglobulina M/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Suínos/genética , Transplante Heterólogo/métodos , Sequência de Aminoácidos , Animais , Sequência de Bases , Biomarcadores/metabolismo , Antígenos CD55/genética , Antígenos CD55/metabolismo , Antígenos CD59/genética , Antígenos CD59/metabolismo , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Técnicas de Inativação de Genes , Marcadores Genéticos , Rejeição de Enxerto/prevenção & controle , Imunoglobulina M/genética , Dados de Sequência Molecular , Papio
6.
Xenotransplantation ; 21(3): 254-66, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24635144

RESUMO

BACKGROUND: B-cell depletion significantly extends survival of α-1,3-galactosyltranferase knockout (GTKO) porcine organs in pig-to-primate models. Our previous work demonstrated that the anti-non-Gal xenoantibody response is structurally restricted. Selective inhibition of xenoantigen/xenoantibody interactions could prolong xenograft survival while preserving B-cell-mediated immune surveillance. METHODS: The anti-idiotypic antibody, B4N190, was selected from a synthetic human phage display library after enrichment against a recombinant anti-non-Gal xenoantibody followed by functional testing in vitro. The inhibitory small molecule, JMS022, was selected from the NCI diversity set III using virtual screening based on predicted xenoantibody structure. Three rhesus monkeys were pre-treated with anti-non-Gal-specific single-chain anti-idiotypic antibody, B4N190. A total of five monkeys, including two untreated controls, were then immunized with GTKO porcine endothelial cells to initiate an anti-non-α-1,3-Gal (non-Gal) xenoantibody response. The efficacy of the inhibitory small molecule specific for anti-non-Gal xenoantibody, JMS022, was tested in vitro. RESULTS: After the combination of in vivo anti-id and in vitro small molecule treatments, IgM xenoantibody binding to GTKO cells was reduced to pre-immunization levels in two-thirds of animals; however, some xenoantibodies remained in the third animal. Furthermore, when treated with anti-id alone, all three experimental animals displayed a lower anti-non-Gal IgG xenoantibody response compared with controls. Treatment with anti-idiotypic antibody alone reduced IgM xenoantibody response intensity in only one of three monkeys injected with GTKO pig endothelial cells. In the one experimental animal, which displayed reduced IgM and IgG responses, select B-cell subsets were also reduced by anti-id therapy alone. Furthermore, natural antibody responses, including anti-laminin, anti-ssDNA, and anti-thyroglobulin antibodies were intact despite targeted depletion of anti-non-Gal xenoantibodies in vivo indicating that selective reduction of xenoantibodies can be accomplished without total B-cell depletion. CONCLUSIONS: This preliminary study demonstrates the strength of approaches designed to selectively inhibit anti-non-Gal xenoantibody. Both anti-non-Gal-specific anti-idiotypic antibody and small molecules can be used to selectively limit xenoantibody responses.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Heterófilos/imunologia , Rejeição de Enxerto/prevenção & controle , Imunoglobulina M/imunologia , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Anticorpos Anti-Idiotípicos/metabolismo , Anticorpos Heterófilos/metabolismo , Linfócitos B/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Técnicas de Inativação de Genes , Marcadores Genéticos , Rejeição de Enxerto/imunologia , Imunoglobulina M/metabolismo , Macaca mulatta , Suínos/genética
7.
Kidney Int ; 85(2): 265-75, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24088952

RESUMO

Xenotransplantation using pigs as donors offers the possibility of eliminating the chronic shortage of donor kidneys, but there are several obstacles to be overcome before this goal can be achieved. Preclinical studies have shown that, while porcine renal xenografts are broadly compatible physiologically, they provoke a complex rejection process involving preformed and elicited antibodies, heightened innate immune cell reactivity, dysregulated coagulation, and a strong T cell-mediated adaptive response. Furthermore, the susceptibility of the xenograft to proinflammatory and procoagulant stimuli is probably increased by cross-species molecular defects in regulatory pathways. To balance these disadvantages, xenotransplantation has at its disposal a unique tool to address particular rejection mechanisms and incompatibilities: genetic modification of the donor. This review focuses on the pathophysiology of porcine renal xenograft rejection, and on the significant genetic, pharmacological, and technical progress that has been made to prolong xenograft survival.


Assuntos
Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Transplante de Rim/efeitos adversos , Imunidade Adaptativa , Animais , Animais Geneticamente Modificados , Genótipo , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Imunidade Inata , Imunossupressores/uso terapêutico , Fenótipo , Especificidade da Espécie , Suínos , Tolerância ao Transplante , Transplante Heterólogo , Resultado do Tratamento
8.
Xenotransplantation ; 20(4): 252-61, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23865597

RESUMO

BACKGROUND: Glutaraldehyde fixation does not guarantee complete tissue biocompatibility in current clinical bioprosthetic heart valves (BHVs). Particularly, circulating anti-αGal human antibodies increase significantly from just 10 days after a BHV implantation. The inactivation of such epitope should be mandatory to meet the requirements for a perspectively safe clinical application; nevertheless, its quantitative assessment in commercially available BHVs has never been carried out. METHODS: In this investigation, seven different models of BHVs were tested. The number of epitopes was determined with reference to a standard αGal source by an ELISA test. The presence of xenoantigen was subsequently confirmed by immunofluorescence analysis. Porcine tissue, knockout for the αGal epitopes, was used as negative control. RESULTS: Epic™ valve was the only model among those tested, in which the αGal antigen appeared to be completely shielded. Composite Trifecta™ valve exhibited conflicting results: cusps of bovine pericardial tissue were devoid of reactive αGal epitopes, while the stent cover strip of porcine pericardium still maintained 30% of active antigens originally present in native tissue. All other tested BHVs express an αGal amount not significantly different from that exhibited by porcine Mosaic(®) valve (5.2 ± 0.6 × 10(10) each 10 mg of tissue). CONCLUSIONS: For the first time, the quantitative evaluation of the αGal epitope in heart valve bioprostheses, already in clinical practice for about 40 yrs, was finally determined. Such quantification might provide indications of biocompatibility relevant for the selection of bioprosthetic devices and an increase in the confidence of the patient. It might become a major quality control tool in the production and redirection of future investigation in the quest for αGal-free long-lasting substitutes.


Assuntos
Epitopos/imunologia , Galactosiltransferases/imunologia , Glutaral/farmacologia , Próteses Valvulares Cardíacas , Valvas Cardíacas/efeitos dos fármacos , Valvas Cardíacas/imunologia , Transplante Heterólogo/métodos , Animais , Anticorpos Anti-Idiotípicos/imunologia , Antígenos/imunologia , Bovinos , Ensaio de Imunoadsorção Enzimática/métodos , Epitopos/genética , Galactosiltransferases/genética , Técnicas de Inativação de Genes , Rejeição de Enxerto/imunologia , Humanos , Masculino , Teste de Materiais , Modelos Animais , Suínos
9.
Xenotransplantation ; 20(2): 100-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23442186

RESUMO

BACKGROUND: We investigated whether graft produced anti-human CD2, mediated by adenovirus (Adv) transduction of pig neonatal islet cell clusters (pNICC), would protect xenografts in a humanized mouse model from immune attack and whether such immunosuppression would remain local. METHODS: A mouse anti-human CD2 Ab (CD2hb11) previously generated by us was genetically engineered to produce chimeric and humanized versions. The three forms of CD2hb11 were named dilimomab (mouse), diliximab (chimeric) and dilizumab (humanized). All 3 forms of CD2hb11 Ab were tested for their ability to bind CD3(+) human T cells and to inhibit a human anti-pig xenogeneic mixed lymphocyte reaction (MLR). They were administered systemically in a humanized mouse model in order to test their ability to deplete human CD3(+) T cells and whether they induced a cytokine storm. An adenoviral vector expressing diliximab was generated for transduction of pNICC. Humanized mice were transplanted with either control-transduced pNICC or diliximab-transduced pNICC and human T cells within grafts and spleens were enumerated by flow cytometry. RESULTS: Dilimomab and diliximab inhibited a human anti-pig xenogeneic response but dilizumab did not. All 3 forms of CD2hb11 Ab bound human T cells in vitro though dilimomab and diliximab exhibited 300-fold higher avidity than dilizumab. All 3 anti-CD2 Abs could deplete human CD3(+) T cells in vivo in a humanized mouse model without inducing upregulation of activation markers or significant release of cytokines. Humanized mice transplanted with diliximab-transduced pNICC afforded depletion of CD3(+) T cells at the graft site leaving the peripheral immune system intact. CONCLUSIONS: Local production of a single Ab against T cells can reduce graft infiltration at the xenograft site and may reduce the need for conventional, systemic immunosuppression.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD2/imunologia , Imunossupressores/farmacologia , Transplante das Ilhotas Pancreáticas/imunologia , Linfócitos T/imunologia , Transplante Heterólogo/imunologia , Adenoviridae/genética , Animais , Anticorpos Heterófilos/imunologia , Anticorpos Heterófilos/farmacologia , Anticorpos Monoclonais/imunologia , Antígenos Heterófilos/genética , Antígenos Heterófilos/imunologia , Antígenos CD2/genética , Quimera , Citometria de Fluxo , Rejeição de Enxerto/imunologia , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Especificidade da Espécie
10.
Diabetes ; 62(6): 2026-35, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23364452

RESUMO

Islet allograft survival limits the long-term success of islet transplantation as a potential curative therapy for type 1 diabetes. A number of factors compromise islet survival, including recurrent diabetes. We investigated whether CD39, an ectonucleotidase that promotes the generation of extracellular adenosine, would mitigate diabetes in the T cell-mediated multiple low-dose streptozotocin (MLDS) model. Mice null for CD39 (CD39KO), wild-type mice (WT), and mice overexpressing CD39 (CD39TG) were subjected to MLDS. Adoptive transfer experiments were performed to delineate the efficacy of tissue-restricted overexpression of CD39. The role of adenosine signaling was examined using mutant mice and pharmacological inhibition. The susceptibility to MLDS-induced diabetes was influenced by the level of expression of CD39. CD39KO mice developed diabetes more rapidly and with higher frequency than WT mice. In contrast, CD39TG mice were protected. CD39 overexpression conferred protection through the activation of adenosine 2A receptor and adenosine 2B receptor. Adoptive transfer experiments indicated that tissue-restricted overexpression of CD39 conferred robust protection, suggesting that this may be a useful strategy to protect islet grafts from T cell-mediated injury.


Assuntos
Antígenos CD/metabolismo , Apirase/metabolismo , Diabetes Mellitus Experimental/metabolismo , Animais , Antígenos CD/genética , Apirase/genética , Diabetes Mellitus Experimental/genética , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo
11.
Hepatology ; 57(4): 1597-606, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22829222

RESUMO

UNLABELLED: Ischemia-reperfusion injury (IRI) is a major limiting event for successful liver transplantation, and CD4+ T cells and invariant natural killer T (iNKT) cells have been implicated in promoting IRI. We hypothesized that hepatic overexpression of CD39, an ectonucleotidase with antiinflammatory functions, will protect liver grafts after prolonged cold ischemia. CD39-transgenic (CD39tg) and wildtype (WT) mouse livers were transplanted into WT recipients after 18 hours cold storage and pathological analysis was performed 6 hours after transplantation. Serum levels of alanine aminotransferase and interleukin (IL)-6 were significantly reduced in recipients of CD39tg livers compared to recipients of WT livers. Furthermore, less severe histopathological injury was demonstrated in the CD39tg grafts. Immune analysis revealed that CD4+ T cells and iNKT cells were significantly decreased in number in the livers of untreated CD39tg mice. This was associated with a peripheral CD4+ T cell lymphopenia due to defective thymocyte maturation. To assess the relative importance of liver-resident CD4+ T cells and iNKT cells in mediating liver injury following extended cold preservation and transplantation, WT mice depleted of CD4+ T cells or mice genetically deficient in iNKT cells were used as donors. The absence of CD4+ T cells, but not iNKT cells, protected liver grafts from early IRI. CONCLUSION: Hepatic CD4+ T cells, but not iNKT cells, play a critical role in early IRI following extended cold preservation in a liver transplant model.


Assuntos
Antígenos CD/metabolismo , Apirase/metabolismo , Linfócitos T CD4-Positivos/patologia , Transplante de Fígado/patologia , Linfopenia/patologia , Traumatismo por Reperfusão/prevenção & controle , Regulação para Cima , Alanina Transaminase/sangue , Animais , Antígenos CD/genética , Apirase/genética , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Interleucina-6/sangue , Células Matadoras Naturais/patologia , Transplante de Fígado/imunologia , Linfopenia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Linfócitos T Reguladores/patologia
12.
Am J Pathol ; 181(1): 322-33, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22613024

RESUMO

Modulation of purinergic signaling, which is critical for vascular homeostasis and the response to vascular injury, is regulated by hydrolysis of proinflammatory ATP and/or ADP by ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD-1; CD39) to AMP, which then is hydrolyzed by ecto-5'-nucleotidase (CD73) to adenosine. We report here that compared with littermate controls (wild type), transgenic mice expressing human ENTPDase-1 were resistant to the formation of an occlusive thrombus after FeCl(3)-induced carotid artery injury. Treatment of mice with the nonhydrolyzable ADP analog, adenosine-5'-0-(2-thiodiphosphate) trilithium salt, Ado-5'-PP[S], negated the protection from thrombosis, consistent with a role for ADP in platelet recruitment and thrombus formation. ENTPD-1 expression decreased whole-blood aggregation after stimulation by ADP, an effect negated by adenosine-5'-0-(2-thiodiphosphate) trilithium salt, Ado-5'-PP[S] stimulation, and limited the ability to maintain the platelet fibrinogen receptor, glycoprotein α(IIb)/ß(3), in a fully activated state, which is critical for thrombus formation. In vivo treatment with a CD73 antagonist, a nonselective adenosine-receptor antagonist, or a selective A(2A) or A(2B) adenosine-receptor antagonist, negated the resistance to thrombosis in transgenic mice expressing human ENTPD-1, suggesting a role for adenosine generation and engagement of adenosine receptors in conferring in vivo resistance to occlusive thrombosis in this model. In summary, our findings identify ENTPDase-1 modulation of purinergic signaling as a key determinant of the formation of an occlusive thrombus after vascular injury.


Assuntos
Antígenos CD/fisiologia , Apirase/fisiologia , Trombose das Artérias Carótidas/prevenção & controle , Adenosina/fisiologia , Animais , Antígenos CD/metabolismo , Apirase/metabolismo , Trombose das Artérias Carótidas/induzido quimicamente , Trombose das Artérias Carótidas/patologia , Células Cultivadas , Cloretos , Compostos Férricos , Camundongos , Camundongos Transgênicos , Ativação Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Receptores Purinérgicos P2/fisiologia , Transdução de Sinais/fisiologia
13.
J Mol Cell Cardiol ; 52(5): 958-61, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22269791

RESUMO

UNLABELLED: CD39 (ectonucleoside triphosphate diphosphohydrolase-1; ENTPD-1) rapidly hydrolyzes ATP and ADP to AMP; AMP is hydrolyzed by ecto-5'-nucleotidase (CD73) to adenosine, an anti-thrombotic and cardiovascular protective mediator. While expression of human CD39 in a murine model of myocardial ischemia/reperfusion (I/R) injury confers cardiac protection, the translational therapeutic potential of these findings requires further testing in a large animal model. To determine if transgenic expression of CD39 reduces infarct size in a swine model of myocardial ischemia/reperfusion injury, transgenic pigs expressing human CD39 (hCD39) were generated via somatic cell nuclear transfer and characterized. Expression of hC39 in cardiac tissue was confirmed by immunoblot and immunohistochemistry. Myocardial I/R injury was induced by intracoronary balloon inflation in the left anterior descending (LAD) artery for 60 min followed by 3 hours of reperfusion. The ischemic area was delineated by perfusion with 5% phthalo blue and the myocardial infarct size was determined by triphenyl tetrazolium chloride (TTC) staining. During ischemia, the rate-pressure product was significantly lower in control versus hCD39-Tg swine. Following reperfusion, compared to littermate control swine, hCD39-Tg animals displayed a significant reduction in infarct size (hCD39-Tg: 17.2 ± 4.3% vs. CONTROL: 44.7 ± 5.2%, P=0.0025). Our findings demonstrate for the first time that the findings in transgenic mouse models translate to large animal transgenic models and validate the potential to translate CD39 into the clinical arena to attenuate human myocardial ischemia/reperfusion injury.


Assuntos
Antígenos CD/biossíntese , Apirase/biossíntese , Traumatismo por Reperfusão Miocárdica/metabolismo , Suínos/genética , Animais , Animais Geneticamente Modificados , Antígenos CD/genética , Apirase/genética , Pressão Sanguínea , Vasos Coronários/patologia , Frequência Cardíaca , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Humanos , Isquemia/metabolismo , Isquemia/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Regiões Promotoras Genéticas , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
14.
J Mol Cell Cardiol ; 51(6): 927-35, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21939667

RESUMO

Modulation of purinergic signaling is critical to myocardial homeostasis. Ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD-1; CD39) which converts the proinflammatory molecules ATP or ADP to AMP is a key regulator of purinergic modulation. However, the salutary effects of transgenic over expression of ENTPD-1 on myocardial response to ischemic injury have not been tested to date. Therefore we hypothesized that ENTPD-1 over expression affords myocardial protection from ischemia-reperfusion injury via specific cell signaling pathways. ENTPD-1 transgenic mice, which over express human ENTPDase-1, and wild-type (WT) littermates were subjected to either ex vivo or in vivo ischemia-reperfusion injury. Infarct size, inflammatory cell infiltrate and intracellular signaling molecule activation were evaluated. Infarct size was significantly reduced in ENTPD-1 versus WT hearts in both ex vivo and in vivo studies. Following ischemia-reperfusion injury, ENTPD-1 cardiac tissues demonstrated an increase in the phosphorylation of the cellular signaling molecule extracellular signal-regulated kinases 1/2 (ERK 1/2) and glycogen synthase kinase-3ß (GSK-3ß). Resistance to myocardial injury was abrogated by treatment with a non-selective adenosine receptor antagonist, 8-SPT or the more selective A(2B) adenosine receptor antagonist, MRS 1754, but not the A(1) selective antagonists, DPCPX. Additionally, treatment with the ERK 1/2 inhibitor PD98059 or the mitochondrial permeability transition pore opener, atractyloside, abrogated the cardiac protection provided by ENTPDase-1 expression. These results suggest that transgenic ENTPDase-1 expression preferentially conveys myocardial protection from ischemic injury via adenosine A(2B) receptor engagement and associated phosphorylation of the cellular protective signaling molecules, Akt, ERK 1/2 and GSK-3ß that prevents detrimental opening of the mitochondrial permeability transition pore.


Assuntos
Antígenos CD/genética , Antígenos CD/metabolismo , Apirase/genética , Apirase/metabolismo , Expressão Gênica , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/prevenção & controle , Fosforilação , Receptor A2B de Adenosina/metabolismo , Transdução de Sinais
15.
Xenotransplantation ; 18(2): 121-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21496119

RESUMO

BACKGROUND: Expression of multiple graft-protective proteins targeted to different locations (i.e., intracellular, cell surface, and secreted) has become an increasingly important goal in xenotransplantation. The 2A "ribosome skip" signal is used as a linker to enable transgene co-expression, but some studies have shown that post-translational modification and trafficking of 2A-linked proteins may be adversely affected depending on their position relative to 2A. We tested whether several relevant proteins, subject to a range of processing and localization mechanisms, could be efficiently co-expressed using the 2A system. METHODS: Six expression cassettes were constructed, each containing up to four 2A-linked open reading frames, encoding combinations of human CD55, thrombomodulin (TBM), CD39, CTLA4-Ig and hygromycin resistance. Each linker incorporated a furin cleavage site to remove the carboxy-terminal extension that remains on upstream proteins after 2A processing. The cassettes were used to produce vectors for transfection, adenoviral transduction and transgenesis. Expression was detected by flow cytometry and/or Western blotting. RESULTS: All proteins were expressed in the appropriate location following transient transfection of COS-7 cells, irrespective of the number of linked genes. The percentage of stable transfectants expressing a linked gene was increased 10-fold (from 4-5% to 58-67%) by incorporating the hygromycin resistance gene into the cassette. Stable transfection of transgenic GalT KO pig fibroblasts with a hygromycin- TBM-CD39 construct resulted in surface expression of both TBM and CD39 by the majority of hygromycin-resistant cells. Expression was maintained after flow cytometric sorting and expansion. Adenoviral transduction of NIT-1 mouse insulinoma cells with a TBM-CD39 construct resulted in strong expression of both genes on the cell surface. Mice transgenic for 3-gene (CD55- TBM-CD39) or 4-gene (CD55- TBM-CTLA4Ig-CD39) constructs expressed all genes except CD55. CONCLUSIONS: These results confirm the versatility of the 2A system, and demonstrate that careful construct design can minimize potential problems with post-translational modification and trafficking. In addition, incorporation of a selection marker into the 2A-linked chain can dramatically increase the proportion of stable transfectants expressing proteins of interest. This provides a powerful method for the rapid modification of existing genetically modified pigs.


Assuntos
Antígenos CD/genética , Apirase/genética , Antígenos CD55/genética , Elementos de DNA Transponíveis/genética , Sobrevivência de Enxerto/genética , Imunoconjugados/genética , Trombomodulina/genética , Transplante Heterólogo/métodos , Abatacepte , Adenoviridae/genética , Animais , Animais Geneticamente Modificados , Antígenos CD/metabolismo , Apirase/metabolismo , Sequência de Bases , Antígenos CD55/metabolismo , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Fibroblastos/metabolismo , Fibroblastos/patologia , Galactosiltransferases/genética , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Imunoconjugados/metabolismo , Insulinoma/metabolismo , Insulinoma/patologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Processamento de Proteína Pós-Traducional , Suínos , Trombomodulina/metabolismo , Transfecção
16.
Curr Opin Organ Transplant ; 16(2): 214-21, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21415824

RESUMO

PURPOSE OF REVIEW: Deletion of the α1,3-galactosyltransferase (GalT) gene in pigs has removed a major xenoantigen but has not eliminated the problem of dysregulated coagulation and vascular injury. Rejecting GalT knockout organ xenografts almost invariably show evidence of thrombosis and platelet sequestration, and primate recipients frequently develop consumptive coagulopathy. This review examines recent findings that illuminate potential mechanisms of this current barrier to successful xenotransplantation. RECENT FINDINGS: The coagulation response to xenotransplantation differs depending on the type of organ and quite likely the distinct vasculatures. Renal xenografts appear more likely to initiate consumptive coagulopathy than cardiac xenografts, possibly reflecting differential transcriptional responses. Liver xenografts induce rapid and profound thrombocytopenia resulting in recipient death within days due to bleeding; ex-vivo data suggest that liver endothelial cells and hepatocytes are responsible for platelet consumption by a coagulation-independent process.It has been proposed that expression of recipient tissue factor on platelets and monocytes is an important trigger of consumptive coagulopathy. Finally, pigs transgenic for human anticoagulants and antithrombotics are slowly but surely coming on line, but have not yet been rigorously tested to date. SUMMARY: Successful control of coagulation dysregulation in xenotransplantation may require different combinatorial pharmacological and genetic strategies for different organs.


Assuntos
Transtornos da Coagulação Sanguínea/etiologia , Coagulação Sanguínea , Rejeição de Enxerto/etiologia , Transplante Heterólogo/efeitos adversos , Animais , Coagulação Sanguínea/genética , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/genética , Transtornos da Coagulação Sanguínea/imunologia , Transtornos da Coagulação Sanguínea/prevenção & controle , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Deleção de Genes , Rejeição de Enxerto/sangue , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Humanos , Imunidade Inata , Suínos , Tolerância ao Transplante , Transplante Heterólogo/imunologia
17.
Transplant Rev (Orlando) ; 25(1): 9-20, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21126659

RESUMO

Xenotransplantation of solid organs will only ever become a clinical reality with genetic modification of the pig, which is now widely accepted as the most likely donor species for humans. The understanding of the barriers to xenotransplantation has required advances in genetic technologies to resolve these problems. Hyperacute rejection has been overcome by overexpression of complement regulatory proteins or targeted disruption of the enzyme associated with the major carbohydrate xenoantigen. The subsequent barriers of disordered coagulation, induced antibody, and cell-mediated rejection remain challenging. The mechanisms for these incompatibilities are being deciphered, and multiple genetic manipulations to resolve these issues are currently in progress. Moreover, new technologies offer help to producing sizeable numbers of modified pigs in a timely manner. This article retraces the basis and foreshadows progress of the genetically modified pig for xenotransplantation as it advances toward the clinic.


Assuntos
Rejeição de Enxerto/genética , Organismos Geneticamente Modificados , Suínos/genética , Transplante Heterólogo/tendências , Animais , Rejeição de Enxerto/imunologia , Humanos
18.
Transplantation ; 90(12): 1260-4, 2010 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-21063242

RESUMO

BACKGROUND: Adenosine agonists are protective in numerous models of ischemia-reperfusion injury (IRI). Pericellular adenosine is generated by the hydrolysis of extracellular adenosine triphosphate and adenosine diphosphate by the ectonucleotidase CD39 and the subsequent hydrolysis of adenosine monophosphate (AMP) by the ectonucleotidase CD73. CD39 activity is protective in kidney IRI, whereas the role of CD73 remains unclear. METHODS: Wild-type (WT), CD73-deficient (CD73KO), CD39-transgenic (CD39tg), and hybrid CD39tg.CD73KO mice underwent right nephrectomy and unilateral renal ischemia (18-min ischemia by microvascular pedicle clamp). Renal function (serum creatinine [SCr], micromolar per liter) and histologic renal injury (score 0-9) were assessed after 24-hr reperfusion. Treatments included a CD73 inhibitor and soluble CD73. RESULTS: Compared with WT mice (n=33, SCr 81.0, score 4.1), (1) CD73KO mice were protected (n=17, SCr 48.9, score 2.0, P<0.05), (2) CD39tg mice were protected (n=11, SCr 45.6, score 1.3, P<0.05), (3) WT mice treated with CD73 inhibitor were protected (n=9, SCr 43.3, score 1.2, P<0.05), (4) CD73KO mice reconstituted with soluble CD73 lost their protection (n=10, SCr 63.8, score 3.1, P=ns), (5) WT mice treated with soluble CD73 were not protected (n=7, SCr 78.0, score 4.1), and (6) CD39tg.CD73KO mice were protected (n=8, SCr 55.5, score 0.7, P<0.05). CONCLUSIONS: Deficiency or inhibition of CD73 protects in kidney IRI, and CD39-mediated protection does not seem to be dependent on adenosine generation. These findings suggest that AMP may play a direct protective role in kidney IRI, which could be used in therapeutic development and organ preservation. Investigating the mechanisms by which AMP mediates protection may lead to new targets for research in kidney IRI.


Assuntos
5'-Nucleotidase/antagonistas & inibidores , Antígenos CD/fisiologia , Apirase/fisiologia , Nefropatias/prevenção & controle , Traumatismo por Reperfusão/prevenção & controle , 5'-Nucleotidase/deficiência , 5'-Nucleotidase/fisiologia , Monofosfato de Adenosina/metabolismo , Monofosfato de Adenosina/fisiologia , Animais , Antígenos CD/genética , Apirase/genética , Creatinina/sangue , Modelos Animais de Doenças , Nefropatias/fisiopatologia , Testes de Função Renal , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Traumatismo por Reperfusão/fisiopatologia
19.
Transplantation ; 90(9): 951-7, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20808263

RESUMO

BACKGROUND: Immunosuppression focused at or near the graft site would reduce the need for systemic immunosuppression thus educing fewer side effects. We investigated whether locally produced CTLA4Ig, mediated by adenovirus (Adv) transduction of mouse islets, would protect allografts and whether such immunosuppression would remain localized. METHODS: Adv-CTLA4Ig- or Adv-control-transduced islets were grafted under the kidney capsule of fully allogeneic diabetic mice. CTLA4Ig secreted from the grafted islets was detected by enzyme immunoassay of blood or immunohistochemistry of graft sections. Graft survival was monitored by blood glucose measurement. Histologic scores of graft sections stained with Gomori aldehyde fuchsin to detect insulin granules or hematoxylin-eosin to detect inflammation were used to compare grafts placed at different sites within the same mouse. RESULTS: Adv-CTLA4Ig-transduced islet grafts secreted CTLA4Ig that was detected transiently in the circulation but persistently at the graft site. Survival of these grafts was significantly enhanced compared with control Adv-transduced and untransduced grafts. The kidney graft site availed elucidation of the site of action of CTLA4Ig. Histologic scores indicated that CTLA4Ig-producing grafts were protected by comparison with control grafts on the contralateral kidney. Hence, graft protection was not attributable to general systemic immunosuppression. Indeed, survival of CTLA4Ig-producing grafts was enhanced over control grafts placed at the opposite pole of the same kidney, indicating that graft protection was not solely due to inhibition of priming in the common draining lymph nodes. CONCLUSIONS: Islet allografts are protected by locally produced CTLA4Ig-disrupting immune interactions at the effector site.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Rejeição de Enxerto/prevenção & controle , Imunoconjugados/uso terapêutico , Imunossupressores/uso terapêutico , Transplante das Ilhotas Pancreáticas/imunologia , Abatacepte , Animais , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto , Imunoconjugados/sangue , Terapia de Imunossupressão/métodos , Transplante das Ilhotas Pancreáticas/patologia , Transplante das Ilhotas Pancreáticas/fisiologia , Camundongos , Transplante Homólogo/imunologia
20.
Structure ; 18(2): 228-38, 2010 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-20159467

RESUMO

Many pathogenic bacteria have sophisticated mechanisms to interfere with the mammalian immune response. These include the disruption of host extracellular ATP levels that, in humans, is tightly regulated by the nucleoside triphosphate diphosphohydrolase family (NTPDases). NTPDases are found almost exclusively in eukaryotes, the notable exception being their presence in some pathogenic prokaryotes. To address the function of bacterial NTPDases, we describe the structures of an NTPDase from the pathogen Legionella pneumophila (Lpg1905/Lp1NTPDase) in its apo state and in complex with the ATP analog AMPPNP and the subtype-specific NTPDase inhibitor ARL 67156. Lp1NTPDase is structurally and catalytically related to eukaryotic NTPDases and the structure provides a basis for NTPDase-specific inhibition. Furthermore, we demonstrate that the activity of Lp1NTPDase correlates directly with intracellular replication of Legionella within macrophages. Collectively, these findings provide insight into the mechanism of this enzyme and highlight its role in host-pathogen interactions.


Assuntos
Proteínas de Bactérias/química , Eucariotos/enzimologia , Legionella pneumophila/enzimologia , Estrutura Terciária de Proteína , Pirofosfatases/química , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Adenilil Imidodifosfato/química , Adenilil Imidodifosfato/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Pirofosfatases/genética , Pirofosfatases/metabolismo , Ratos , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA