Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 11(1): e1004610, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25569288

RESUMO

Lentiviral Envelope (Env) antigenic variation and related immune evasion present major hurdles to effective vaccine development. Centralized Env immunogens that minimize the genetic distance between vaccine proteins and circulating viral isolates are an area of increasing study in HIV vaccinology. To date, the efficacy of centralized immunogens has not been evaluated in the context of an animal model that could provide both immunogenicity and protective efficacy data. We previously reported on a live-attenuated (attenuated) equine infectious anemia (EIAV) virus vaccine, which provides 100% protection from disease after virulent, homologous, virus challenge. Further, protective efficacy demonstrated a significant, inverse, linear relationship between EIAV Env divergence and protection from disease when vaccinates were challenged with viral strains of increasing Env divergence from the vaccine strain Env. Here, we sought to comprehensively examine the protective efficacy of centralized immunogens in our attenuated vaccine platform. We developed, constructed, and extensively tested a consensus Env, which in a virulent proviral backbone generated a fully replication-competent pathogenic virus, and compared this consensus Env to an ancestral Env in our attenuated proviral backbone. A polyvalent attenuated vaccine was established for comparison to the centralized vaccines. Additionally, an engineered quasispecies challenge model was created for rigorous assessment of protective efficacy. Twenty-four EIAV-naïve animals were vaccinated and challenged along with six-control animals six months post-second inoculation. Pre-challenge data indicated the consensus Env was more broadly immunogenic than the Env of the other attenuated vaccines. However, challenge data demonstrated a significant increase in protective efficacy of the polyvalent vaccine. These findings reveal, for the first time, a consensus Env immunogen that generated a fully-functional, replication-competent lentivirus, which when experimentally evaluated, demonstrated broader immunogenicity that does not equate to higher protective efficacy.


Assuntos
Anemia Infecciosa Equina/prevenção & controle , Cavalos/imunologia , Vírus da Anemia Infecciosa Equina/imunologia , Vacinas Atenuadas/uso terapêutico , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Variação Antigênica/imunologia , Sequência de Bases , Variação Genética , Vírus da Anemia Infecciosa Equina/genética , Dados de Sequência Molecular , Filogenia , Resultado do Tratamento , Proteínas do Envelope Viral/genética , Vacinas Virais/uso terapêutico
2.
PLoS One ; 8(6): e66093, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23785473

RESUMO

Lentiviral envelope (Env) antigenic variation and associated immune evasion present major obstacles to vaccine development. The concept that Env is a critical determinant for vaccine efficacy is well accepted, however defined correlates of protection associated with Env variation have yet to be determined. We reported an attenuated equine infectious anemia virus (EIAV) vaccine study that directly examined the effect of lentiviral Env sequence variation on vaccine efficacy. The study identified a significant, inverse, linear correlation between vaccine efficacy and increasing divergence of the challenge virus Env gp90 protein compared to the vaccine virus gp90. The report demonstrated approximately 100% protection of immunized ponies from disease after challenge by virus with a homologous gp90 (EV0), and roughly 40% protection against challenge by virus (EV13) with a gp90 13% divergent from the vaccine strain. In the current study we examine whether the protection observed when challenging with the EV0 strain could be conferred to animals via chimeric challenge viruses between the EV0 and EV13 strains, allowing for mapping of protection to specific Env sequences. Viruses containing the EV13 proviral backbone and selected domains of the EV0 gp90 were constructed and in vitro and in vivo infectivity examined. Vaccine efficacy studies indicated that homology between the vaccine strain gp90 and the N-terminus of the challenge strain gp90 was capable of inducing immunity that resulted in significantly lower levels of post-challenge virus and significantly delayed the onset of disease. However, a homologous N-terminal region alone inserted in the EV13 backbone could not impart the 100% protection observed with the EV0 strain. Data presented here denote the complicated and potentially contradictory relationship between in vitro virulence and in vivo pathogenicity. The study highlights the importance of structural conformation for immunogens and emphasizes the need for antibody binding, not neutralizing, assays that correlate with vaccine protection.


Assuntos
Anemia Infecciosa Equina/prevenção & controle , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Linhagem Celular , Anemia Infecciosa Equina/imunologia , Anemia Infecciosa Equina/virologia , Ordem dos Genes , Genoma Viral , Cavalos , Imunidade Celular , Imunidade Humoral , Vírus da Anemia Infecciosa Equina/patogenicidade , Provírus/genética , Recombinação Genética , Carga Viral , Vacinas Virais/genética , Virulência/genética
3.
Xenotransplantation ; 19(5): 311-6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22970807

RESUMO

BACKGROUND: Thrombin formation is a key feature in the activation of coagulation in pig xenograft recipients. As thrombin is known to activate endothelial and immune cells, we explored whether thrombin activation of pig endothelial cells (EC) was associated with an increased human T-cell response. METHODS: α1,3-galactosyltransferase gene-knockout (GTKO) pig aortic EC (pAEC) were activated by porcine interferon-gamma (pIFNγ), human (h)IFN-γ, or thrombin. Swine leukocyte antigen (SLA) class I and class II expression were measured. Human peripheral blood mononuclear cells (PBMC) and CD4+ T-cell proliferation in response to activated pAEC, the effect of thrombin on pig CD80/CD86 mRNA, and the effect of thrombin inhibition by hirudin were evaluated. RESULTS: After pAEC activation, SLA I expression did not change, and only pIFNγ upregulated SLA II expression. PBMC proliferation to pIFNγ- and thrombin-activated pAEC was significantly higher (P < 0.001 and P < 0.01) than to non-activated pAEC. CD4+ T-cell proliferation to pIFNγ- and thrombin-activated pAEC was significantly higher (P < 0.001 and P < 0.01) than to non-activated pAEC. Thrombin inhibition by hirudin reduced thrombin-induced upregulation of pAEC CD86 mRNA, and significantly reduced human PBMC proliferation to pAEC in comparison with thrombin alone (P < 0.05). CONCLUSIONS: Thrombin upregulates CD86 mRNA on pAEC, which is associated with increased human T-cell proliferation against pAEC. Hirudin reduces CD86 mRNA in thrombin-activated pAEC and is associated with downregulation of the human T-cell proliferative response. The transplantation of organs from GTKO pigs transgenic for human thrombomodulin, and/or endothelial protein C receptor, in addition to therapeutic regulation of thrombin activation may reduce the cellular response to a pig xenograft and thus reduce the need for intensive immunosuppressive therapy.


Assuntos
Células Endoteliais/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Antígenos Heterófilos/genética , Antígeno B7-1/genética , Antígeno B7-2/genética , Sequência de Bases , Proliferação de Células , DNA Complementar/genética , Células Endoteliais/efeitos dos fármacos , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Técnicas de Inativação de Genes , Hirudinas/farmacologia , Antígenos de Histocompatibilidade Classe I , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Interferon gama/farmacologia , Ativação Linfocitária , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sus scrofa/genética , Sus scrofa/imunologia , Trombina/farmacologia , Transplante Heterólogo
4.
Xenotransplantation ; 19(4): 221-32, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22909135

RESUMO

BACKGROUND: CD154 blockade-based immunosuppression successfully prevents both humoral and cellular adaptive immune responses in baboons receiving α1,3-galactosyltransferase gene-knockout (GTKO) pig organs. Using a GTKO pig artery transplantation model in baboons, we evaluated the efficacy of CD28/B7 costimulatory pathway blockade in comparison with CD154 blockade. METHODS: Baboons received artery patch grafts from GTKO pigs, with no (Group1), anti-CD154mAb-based (Group2), or CTLA4-Ig-based (Group3) immunosuppressive therapy. Anti-pig IgM and IgG antibody and cellular responses were monitored. Xenografts were immunohistologically evaluated for antibody and complement deposition, and cellular infiltration. RESULTS: Group1 baboons developed increased IgM and IgG antibody and cellular responses against GTKO antigens. In Group2, anti-CD154mAb alone prevented the development of both IgM and IgG antibody and cellular responses,but not cellular infiltration of the graft. In the single baboon that received anti-thymocyte globulin (ATG) + mycophenolate mofetil (MMF) + anti-CD154mAb, cellular infiltration of the graft was not seen. In Group3, CTLA4-Ig with ATG + MMF inhibited the cellular proliferative response to pig antigens but did not prevent the IgG response or cellular infiltration. CONCLUSIONS: (i) Artery patch transplantation is a simple model to monitor the adaptive immune response to xenografts; (ii) anti-CD154mAb prevents sensitization but not cellular infiltration (but, without anticoagulation, may result in early thrombosis of a pig xenograft); (iii) although in only one baboon, the addition of ATG and MMF prevents cellular infiltration and (iv) replacement of anti-CD154mAb by CTLA4-Ig (at the doses used), even in combination with ATG and MMF, prevents the cellular proliferative response to GTKO pig antigens but is insufficient to prevent the development of anti-pig antibodies.


Assuntos
Modelos Imunológicos , Transplante Heterólogo/imunologia , Imunidade Adaptativa , Animais , Antígenos Heterófilos/imunologia , Artérias/transplante , Ligante de CD40/antagonistas & inibidores , Ligante de CD40/imunologia , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Técnicas de Inativação de Genes , Imunidade Inata , Imunossupressores/administração & dosagem , Modelos Animais , Papio/imunologia , Suínos/genética , Suínos/imunologia , Transplante Heterólogo/efeitos adversos , Transplante Heterólogo/patologia
5.
J Virol Methods ; 185(2): 221-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22820072

RESUMO

A faster semi-automated 96-well microtiter plate assay to determine viral infectivity titers, or viral focal units (vfu), of equine infectious anemia virus (EIAV) stocks is described. Optimization of the existing method modernizes a classic virological technique for viral titer determination by quantitating EIAV in experimentally infected cells via a cell-based ELISA. To allow for automation, multiple parameters of the current assay procedures were modified resulting in an assay that required only one quarter the original amount of virus and/or serum for infectivity or neutralization assays, respectively. Equivalent reductions in the required volumes of tissue culture, cell processing, and protein detection reagents were also achieved. Additionally, the new assay decreased the time required from start to finish from 10 days to 6 days (viral titer) or 7 days (viral neutralization), while increasing the number of samples that can be processed concurrently by transition to a 96-well microtiter plate format and by automated counting.


Assuntos
Automação Laboratorial/métodos , Ensaio de Imunoadsorção Enzimática/veterinária , Ensaios de Triagem em Larga Escala/veterinária , Vírus da Anemia Infecciosa Equina/isolamento & purificação , Animais , Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Linhagem Celular , Ensaio de Imunoadsorção Enzimática/métodos , Ensaios de Triagem em Larga Escala/métodos , Indicadores e Reagentes/química , Vírus da Anemia Infecciosa Equina/química , Testes de Neutralização/métodos , Testes de Neutralização/veterinária , Sensibilidade e Especificidade , Fatores de Tempo , Ensaio de Placa Viral/métodos , Ensaio de Placa Viral/veterinária , Proteínas Virais/química , Proteínas Virais/isolamento & purificação
6.
Transplantation ; 93(8): 769-76, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22441321

RESUMO

BACKGROUND: We set out to determine whether B-cell tolerance to A/B-incompatible alloantigens and pig xenoantigens could be achieved in infant baboons. METHODS: Artery patch grafts were implanted in the abdominal aorta in 3-month-old baboons using A/B-incompatible (AB-I) allografts or wild-type pig xenografts (pig). Group 1 (Gp1) (controls, n=6) received no immunosuppressive therapy (IS) and no graft. Gp2 (n=2) received an AB-I or pig graft but no IS. Gp3 received AB-I grafts+IS (Gp3A: n=2) or pig grafts+IS (Gp3B: n=2). IS consisted of ATG, anti-CD154mAb, and mycophenolate mofetil until age 8 to 12 months. Gp4 (n=2) received IS only but no graft. RESULTS: In Gp1, anti-A/B and cytotoxic anti-pig immunoglobulin-M increased steadily during the first year. Gp2 became sensitized to donor-specific AB-I or pig antigens within 2 weeks. Gp3 and Gp4 infants that received anti-CD154mAb made no or minimal anti-A/B and anti-pig antibodies while receiving IS. DISCUSSION: The production of natural anti-A/B and anti-pig antibodies was inhibited by IS with anti-CD154mAb, even in the absence of an allograft or xenograft, suggesting that natural antibodies may not be entirely T-cell independent. These data are in contrast to clinical experience with AB-I allotransplantation in infants, who cease producing only donor-specific antibodies.


Assuntos
Anticorpos/imunologia , Soro Antilinfocitário/uso terapêutico , Imunossupressores/uso terapêutico , Ácido Micofenólico/análogos & derivados , Linfócitos T/imunologia , Animais , Animais Recém-Nascidos , Antígenos/imunologia , Soro Antilinfocitário/imunologia , Aorta Abdominal/imunologia , Aorta Abdominal/cirurgia , Artérias/imunologia , Artérias/transplante , Ligante de CD40/imunologia , Tolerância Imunológica/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Imunossupressores/imunologia , Ácido Micofenólico/imunologia , Ácido Micofenólico/uso terapêutico , Papio , Suínos , Transplante Heterólogo
7.
Xenotransplantation ; 19(1): 56-63, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22360754

RESUMO

BACKGROUND: Lack of Gal expression on pig cells is associated with a reduced primate humoral immune response as well as a reduction in cytokine production by human cells in vitro. We investigated whether lack of Gal expression is associated with reduced human T-cell response in vitro. METHODS: Peripheral blood mononuclear cells (PBMCs) were obtained from healthy humans and naïve baboons. Human CD4+ and CD8+ T cells were isolated. Porcine aortic endothelial cells (pAECs) were isolated from wild-type (WT) and α1,3-galactosyltransferase gene-knockout (GTKO) pigs. WT pAECs were treated with α-galactosidase, reducing Gal expression. Swine leukocyte antigen (SLA) class I and II expression on pAECs was measured, as was T-cell proliferation and cytokine production in response to pAECs. RESULTS: Reduced Gal expression on WT pAECs after α-galactosidase treatment was associated with reduced human PBMC proliferation (P<0.005). SLA class I and II expression on WT and GTKO pAECs was comparable. Human CD4+ and CD8+ T-cell proliferation was less against GTKO pAECs before (P<0.001) and after (P<0.01 and P<0.05, respectively) activation. Human and baboon PBMC proliferation was less against GTKO pAECs before (P<0.05) and after (P<0.01 and P<0.05, respectively) activation. Human PBMCs produced a comparable cytokine/chemokine response to WT and GTKO pAECs. However, there was less production of IFN-γ/TNF-α by CD4+ and IFN-γ/granzyme B/IP-10 by CD8+ T cells in response to GTKO pAECs. CONCLUSIONS: The absence of Gal on pig cells is associated with reduced human T-cell proliferation (and possibly selected cytokine production). Adaptive primate T-cell responses are likely to be reduced in GTKO xenograft recipients.


Assuntos
Dissacarídeos/imunologia , Leucócitos Mononucleares/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Transplante Heterólogo/imunologia , Animais , Animais Geneticamente Modificados , Proliferação de Células , Dissacarídeos/genética , Endotélio Vascular/fisiologia , Epitopos/imunologia , Antígenos de Histocompatibilidade Classe I , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunidade Humoral/imunologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Masculino , Papio , Suínos , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , alfa-Galactosidase/metabolismo
8.
Xenotransplantation ; 18(3): 183-95, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21696448

RESUMO

BACKGROUND: Mesenchymal stromal cells (MSC) are being investigated as immunomodulatory therapy in the field of transplantation, particularly islet transplantation. While MSC can regenerate across species barriers, the immunoregulatory influence of genetically modified pig MSC (pMSC) on the human and non-human primate T-cell responses has not been studied. METHODS: Mesenchymal stromal cells from wild-type (WT), α1,3-galactosyltransferase gene knockout (GTKO) and GTKO pigs transgenic for the human complement-regulatory protein CD46 (GTKO/CD46) were isolated and tested for differentiation. Antibody binding and T-cell responses to WT and GTKO pMSC in comparison with GTKO pig aortic endothelial cells (pAEC) were investigated. The expression of swine leukocyte antigen (SLA) class II (SLA II) was tested. Costimulatory molecules CD80 and CD86 mRNA levels were measured. Human T-cell proliferation and the production of pro-inflammatory cytokines in response to GTKO and GTKO/CD46 pMSC in comparison with human MSC (hMSC) were evaluated. RESULTS: α1,3-galactosyltransferase gene knockout and GTKO/CD46 pMSC isolation and differentiation were achieved in vitro. Binding of human antibodies and T-cell responses were lower to GTKO than those to WT pMSC. Human and baboon (naïve and sensitized) antibody binding were significantly lower to GTKO pMSC than to GTKO pAEC. Before activation, <1% of GTKO pMSC expressed SLA II, compared with 2.5% of GTKO pAEC. After pig interferon-gamma (pIFN-γ) activation, 99% of GTKO pAEC upregulated SLA II expression, compared with 49% of GTKO pMSC. Only 3% of GTKO pMSC expressed CD80 compared with 80% of GTKO pAEC without activation. After pIFN-γ activation, GTKO pAEC upregulated CD86 mRNA level stronger than GTKO pMSC. The human CD4(+) T-cell response to GTKO pMSC was significantly weaker than that to GTKO pAEC, even after pIFN-γ activation. More than 99% of GTKO/CD46 pMSC expressed hCD46. Human peripheral blood mononuclear cells and CD4(+) T-cell responses to GTKO and GTKO/CD46 pMSC were comparable with those to hMSC, and all were significantly lower than to GTKO pAEC. GTKO/CD46 pMSC downregulated human T-cell proliferation as efficiently as hMSC. The level of proinflammatory cytokines IL-2, IFN-γ, TNF-α, and sCD40L correlated with the downregulation of T-cell proliferation by all types of MSC. CONCLUSION: Genetically modified pMSC is significantly less immunogenic than WT pMSC. GTKO/CD46 pMSC downregulates the human T-cell responses to pig antigens as efficiently as human MSC, which can be advantageous for therapeutic cell xenotransplantation.


Assuntos
Animais Geneticamente Modificados , Linfócitos T CD4-Positivos/imunologia , Células-Tronco Mesenquimais/imunologia , Células Estromais/imunologia , Animais , Ligante de CD40/imunologia , Diferenciação Celular , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Antígenos de Histocompatibilidade Classe I , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunoglobulinas/imunologia , Interferon gama/imunologia , Interleucina-2/imunologia , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Papio/imunologia , Células Estromais/citologia , Células Estromais/fisiologia , Suínos , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...