Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 88(4): 2011-24, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24307581

RESUMO

As an alternative to targeting human immunodeficiency virus (HIV), we have developed vaccines targeting CCR5, a self-protein critically involved in HIV replication and pathogenesis. By displaying peptides derived from CCR5 at high density on the surface of virus-like particles, we can efficiently induce high-titer IgG antibodies against this self-molecule. Here, we investigated whether prophylactic immunization of rhesus macaques with a particle-based vaccine targeting two regions of macaque CCR5 could prevent or suppress vaginal infection with highly virulent SIVmac251. Twelve macaques were vaccinated with a bacteriophage Qß-based vaccine targeting macaque CCR5 (Qß.CCR5). Six control animals were immunized with the Qß platform alone. All animals immunized with Qß.CCR5 developed high-titer anti-CCR5 antibody responses. Macaques were vaginally challenged with a high dose of SIVmac251. The mean peak viral RNA levels in the vaccinated groups were 30-fold lower than in the control group (10(6.8) versus 10(8.3) copies/ml plasma). Three of the 12 vaccinated macaques dramatically suppressed simian immunodeficiency virus (SIV) replication: peak viral loads were low (10(3) to 10(4) RNA copies/ml), and SIV RNA became undetectable from 6 weeks onward. No viral RNA or DNA could be detected in colon and lymph node biopsy specimens collected 13 months after challenge. In vivo depletion of CD8(+) cells failed to induce a viral rebound. However, once anti-CCR5 antibody responses had waned, the 3 animals became infected after intravaginal and/or intravenous rechallenge. In conclusion, vaccination against CCR5 was associated with dramatic suppression of virus replication in a subset (25%) of macaques. These data support further research of vaccination against CCR5 to combat HIV infection.


Assuntos
Imunoglobulina G/imunologia , Receptores CCR5/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vacinas Virais/imunologia , Administração Intravaginal , Allolevivirus , Sequência de Aminoácidos , Animais , Macaca mulatta/virologia , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/imunologia , RNA Viral/sangue , Carga Viral , Replicação Viral/fisiologia
2.
J Virol ; 87(17): 9523-37, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23785209

RESUMO

Human immunodeficiency virus (HIV) is characterized by immune activation, while chronic malaria is associated with elevated interleukin-10 (IL-10) levels. How these apparently antagonizing forces interact in the coinfected host is poorly understood. Using a rhesus macaque model of simian immunodeficiency virus (SIV)-Plasmodium fragile coinfection, we evaluated how innate immune effector cells affect the balance between immune activation and regulation. In vitro Toll-like receptor (TLR) responses of peripheral blood myeloid dendritic cells (mDC) and monocytes were temporarily associated with acute parasitemic episodes and elevated plasma IL-10 levels. Prolonged infection resulted in a decline of mDC function. Monocytes maintained TLR responsiveness but, in addition to IL-12 and tumor necrosis factor alpha, also produced IL-10. Consistent with the role of spleen in the clearance of parasite-infected red blood cells, coinfected animals also had increased splenic IL-10 mRNA levels. The main cellular source of IL-10 in the spleens of coinfected animals, however, was not splenic macrophages but T cells, suggesting an impairment of adaptive immunity. In contrast to those in spleen, IL-10-positive cells in axillary lymph nodes of coinfected animals were predominantly mDC, reminiscent of the immunosuppressive phenotype of peripheral blood mDC. Concurrent with IL-10 induction, however, SIV infection promoted elevated systemic IL-12 levels. The continuously increasing ratio of plasma IL-12 to IL-10 suggested that the overall host response in SIV-P. fragile-coinfected animals was shifted toward immune activation versus immune regulation. Therefore, SIV-P. fragile coinfection might be characterized by earlier manifestation of immune dysfunction and exhaustion than that of single-pathogen infections. This could translate into increased morbidity in HIV-malaria-coinfected individuals.


Assuntos
Coinfecção/imunologia , Interações Hospedeiro-Patógeno/imunologia , Malária/complicações , Malária/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia , Animais , Coinfecção/genética , Células Dendríticas/imunologia , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Interações Hospedeiro-Patógeno/genética , Humanos , Imunidade Inata/genética , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Tecido Linfoide/imunologia , Macaca mulatta , Malária/genética , Masculino , Monócitos/imunologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
3.
Retrovirology ; 9: 57, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22805180

RESUMO

BACKGROUND: We reported previously that while prolonged tenofovir monotherapy of macaques infected with virulent simian immunodeficiency virus (SIV) resulted invariably in the emergence of viral mutants with reduced in vitro drug susceptibility and a K65R mutation in reverse transcriptase, some animals controlled virus replication for years. Transient CD8+ cell depletion or short-term tenofovir interruption within 1 to 5 years of treatment demonstrated that a combination of CD8+ cell-mediated immune responses and continued tenofovir therapy was required for sustained suppression of viremia. We report here follow-up data on 5 such animals that received tenofovir for 8 to 14 years. RESULTS: Although one animal had a gradual increase in viremia from 3 years onwards, the other 4 tenofovir-treated animals maintained undetectable viremia with occasional viral blips (≤ 300 RNA copies/ml plasma). When tenofovir was withdrawn after 8 to 10 years from three animals with undetectable viremia, the pattern of occasional episodes of low viremia (≤ 3600 RNA/ml plasma) continued throughout the 10-month follow-up period. These animals had low virus levels in lymphoid tissues, and evidence of multiple SIV-specific immune responses. CONCLUSION: Under certain conditions (i.e., prolonged antiviral therapy initiated early after infection; viral mutants with reduced drug susceptibility) a virus-host balance characterized by strong immunologic control of virus replication can be achieved. Although further research is needed to translate these findings into clinical applications, these observations provide hope for a functional cure of HIV infection via immunotherapeutic strategies that boost antiviral immunity and reduce the need for continuous antiretroviral therapy.


Assuntos
Adenina/análogos & derivados , Organofosfonatos/farmacologia , DNA Polimerase Dirigida por RNA/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia/patogenicidade , Replicação Viral , Adenina/imunologia , Adenina/farmacologia , Alelos , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Formação de Anticorpos , Antivirais/imunologia , Antivirais/farmacologia , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Genes MHC Classe I , Técnicas de Genotipagem , Imunidade Celular , Ativação Linfocitária , Macaca mulatta , Testes de Neutralização , Organofosfonatos/imunologia , RNA Viral/sangue , DNA Polimerase Dirigida por RNA/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/enzimologia , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Tenofovir , Fatores de Tempo , Resultado do Tratamento , Viremia/patologia , Viremia/virologia
4.
Antimicrob Agents Chemother ; 56(9): 4980-4, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22777046

RESUMO

To compare tissue-based pharmacokinetics and efficacy of oral tenofovir disoproxyl fumarate (TDF) versus subcutaneous tenofovir (TFV), macaques were treated for 2 weeks starting 1 week after simian immunodeficiency virus inoculation. Despite lower plasma TFV levels in the oral TDF arm, similar TFV diphosphate levels and antiviral activities were measured in lymphoid cells of most tissues. In intestinal tissues, however, oral TDF resulted in higher active drug levels, associated with lower virus levels and better immune preservation.


Assuntos
Adenina/análogos & derivados , Antivirais/farmacocinética , Organofosfonatos/farmacocinética , RNA Viral/antagonistas & inibidores , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Adenina/administração & dosagem , Adenina/farmacocinética , Administração Oral , Animais , Antivirais/administração & dosagem , Esquema de Medicação , Injeções Subcutâneas , Intestinos/química , Intestinos/virologia , Linfócitos/química , Linfócitos/virologia , Macaca mulatta , Masculino , Organofosfonatos/administração & dosagem , RNA Viral/biossíntese , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Tenofovir , Resultado do Tratamento
5.
J Virol ; 85(22): 11655-63, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21917966

RESUMO

In sub-Saharan Africa, HIV-1 infection frequently occurs in the context of other coinfecting pathogens, most importantly, Mycobacterium tuberculosis and malaria parasites. The consequences are often devastating, resulting in enhanced morbidity and mortality. Due to the large number of confounding factors influencing pathogenesis in coinfected people, we sought to develop a nonhuman primate model of simian immunodeficiency virus (SIV)-malaria coinfection. In sub-Saharan Africa, Plasmodium falciparum is the most common malaria parasite and is responsible for most malaria-induced deaths. The simian malaria parasite Plasmodium fragile can induce clinical symptoms, including cerebral malaria in rhesus macaques, that resemble those of P. falciparum infection in humans. Thus, based on the well-characterized rhesus macaque model of SIV infection, this study reports the development of a novel rhesus macaque SIV-P. fragile coinfection model to study human HIV-P. falciparum coinfection. Using this model, we show that coinfection is associated with an increased, although transient, risk of both HIV and malaria transmission. Specifically, SIV-P. fragile coinfected macaques experienced an increase in SIV viremia that was temporarily associated with an increase in potential SIV target cells and systemic immune activation during acute parasitemia. Conversely, primary parasitemia in SIV-P. fragile coinfected animals resulted in higher gametocytemia that subsequently translated into higher oocyst development in mosquitoes. To our knowledge, this is the first animal model able to recapitulate the increased transmission risk of both HIV and malaria in coinfected humans. Therefore, this model could serve as an essential tool to elucidate distinct immunological, virological, and/or parasitological parameters underlying disease exacerbation in HIV-malaria coinfected people.


Assuntos
Malária/complicações , Malária/transmissão , Plasmodium falciparum/patogenicidade , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Modelos Animais de Doenças , Humanos , Macaca mulatta , Masculino , Parasitemia/complicações , Plasmodium falciparum/isolamento & purificação , Doenças dos Primatas/transmissão , Vírus da Imunodeficiência Símia/isolamento & purificação , Viremia/complicações
6.
J Virol ; 85(6): 2878-90, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191005

RESUMO

The use of animal models of human cytomegalovirus (HCMV) infection is critical to refine HCMV vaccine candidates. Previous reports have demonstrated that immunization of rhesus monkeys against rhesus cytomegalovirus (RhCMV) can reduce both local and systemic replication of RhCMV following experimental RhCMV challenge. These studies used prime/boost combinations of DNA expression plasmids alone or DNA priming and boosting with either inactivated virion particles or modified vaccinia virus Ankara (MVA) expressing the same antigens. Viral outcomes included reduced RhCMV replication at the site of subcutaneous inoculation and RhCMV viremia following intravenous inoculation. Since shedding of cytomegalovirus from mucosal surfaces is critical for horizontal transmission of the virus, DNA priming/MVA boosting was evaluated for the ability to reduce oral shedding of RhCMV following subcutaneous challenge. Of six rhesus monkeys vaccinated exclusively against RhCMV glycoprotein B (gB), phosphoprotein 65 (pp65), and immediate-early 1 (IE1), half showed viral loads in saliva that were lower than those of control monkeys by 1 to 3 orders of magnitude. Further, there was a strong association of memory pp65 T cell responses postchallenge in animals exhibiting the greatest reduction in oral shedding. These results highlight the fact that a DNA/MVA vaccination regimen can achieve a notable reduction in a critical parameter of viral replication postchallenge. The recently completed clinical trial of a gB subunit vaccine in which the rate of HCMV infection was reduced by 50% in the individuals receiving the vaccine is consistent with the results of this study suggesting that additional immunogens are likely essential for maximum protection in an outbred human population.


Assuntos
Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/imunologia , Doenças dos Primatas/prevenção & controle , Vacinas de DNA/imunologia , Eliminação de Partículas Virais , Animais , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Vacinas contra Citomegalovirus/administração & dosagem , Modelos Animais de Doenças , Feminino , Imunização Secundária/métodos , Macaca mulatta , Masculino , Mucosa Bucal/virologia , Doenças dos Primatas/imunologia , Doenças dos Primatas/virologia , Saliva/virologia , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Carga Viral
7.
J Med Primatol ; 41(3): 225-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22620273

RESUMO

BACKGROUND: Congenital human cytomegalovirus (HCMV) infection can result in lifelong neurological deficits. Seronegative pregnant woman often acquire primary HCMV from clinically asymptomatic, but HCMV-shedding children. METHODS: Potential age-related differences in viral and immune parameters of primary RhCMV infection were examined in an oral rhesus CMV infection model in specific pathogen free macaques. RhCMV shedding was measured by real time PCR in plasma, saliva and urine. Immune parameters, including neutralizing and binding antibodies and RhCMV-specific T cell responses, were assessed in longitudinally collected blood samples. RESULTS: The oral RhCMV infection model in infant SPF rhesus macaques demonstrated that (i) the susceptibility to oral RhCMV infection declines with age, and (ii) infant macaques shed RhCMV more persistently and at higher titers compared to adult macaques. (iii) CONCLUSIONS: The oral infant RhCMV infection model appears to reflect viral pathogenesis in human HCMV-infected children. Larger studies are needed to define immune parameters associated with better control of RhCMV in adult compared to young animals.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus , Modelos Animais de Doenças , Organismos Livres de Patógenos Específicos , Envelhecimento , Animais , Criança , Feminino , Humanos , Gravidez , Eliminação de Partículas Virais
8.
Toxicol Appl Pharmacol ; 236(1): 39-48, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19371618

RESUMO

The epidemiologic link between air pollutant exposure and asthma has been supported by experimental findings, but the mechanisms are not understood. In this study, we evaluated the impact of combined ozone and house dust mite (HDM) exposure on the immunophenotype of peripheral blood and airway lymphocytes from rhesus macaque monkeys during the postnatal period of development. Starting at 30 days of age, monkeys were exposed to 11 cycles of filtered air, ozone, HDM aerosol, or ozone+HDM aerosol. Each cycle consisted of ozone delivered at 0.5 ppm for 5 days (8 h/day), followed by 9 days of filtered air; animals received HDM aerosol during the last 3 days of each ozone exposure period. Between 2-3 months of age, animals co-exposed to ozone+HDM exhibited a decline in total circulating leukocyte numbers and increased total circulating lymphocyte frequency. At 3 months of age, blood CD4+/CD25+ lymphocytes were increased with ozone+HDM. At 6 months of age, CD4+/CD25+ and CD8+/CD25+ lymphocyte populations increased in both blood and lavage of ozone+HDM animals. Overall volume of CD25+ cells within airway mucosa increased with HDM exposure. Ozone did not have an additive effect on volume of mucosal CD25+ cells in HDM-exposed animals, but did alter the anatomical distribution of this cell type throughout the proximal and distal airways. We conclude that a window of postnatal development is sensitive to air pollutant and allergen exposure, resulting in immunomodulation of peripheral blood and airway lymphocyte frequency and trafficking.


Assuntos
Poluentes Atmosféricos/imunologia , Antígenos de Dermatophagoides/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Pulmão/imunologia , Ozônio/imunologia , Mucosa Respiratória/imunologia , Aerossóis , Fatores Etários , Animais , Animais Recém-Nascidos , Líquido da Lavagem Broncoalveolar/imunologia , Quimiotaxia de Leucócito , Imunofenotipagem , Exposição por Inalação , Macaca mulatta , Masculino
9.
Hum Vaccin ; 5(3): 141-50, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18690014

RESUMO

This pilot study tested the immunogenicity of a novel cationic liposome-DNA complex (CLDC) immunomodulatory vaccine adjuvant. Combined with a specific antigen, CLDC enhanced anti-SIV immune responses induced by various SIV vaccine candidates. Rhesus macaques immunized in the presence of CLDC developed stronger SIV-specific T and B cell responses compared to animals immunized without CLDC. These differences persisted and resulted in better memory responses after an in vivo boost of the animals several months later with whole AT-2 inactivated SIVmac239. Thus, CLDC should be explored further as a potential immunomodulatory adjuvant in HIV vaccine design.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Lipossomos/administração & dosagem , Oligodesoxirribonucleotídeos/administração & dosagem , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Animais , Linfócitos B/imunologia , Memória Imunológica , Macaca mulatta , Masculino , Projetos Piloto , Linfócitos T/imunologia
10.
Antimicrob Agents Chemother ; 52(9): 3144-60, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18573931

RESUMO

The reverse transcriptase (RT) inhibitor tenofovir (TFV) is highly effective in the simian immunodeficiency virus (SIV) macaque model of human immunodeficiency virus infection. The current report describes extended safety and efficacy data on 32 animals that received prolonged (>or=1- to 13-year) daily subcutaneous TFV regimens. The likelihood of renal toxicity (proximal renal tubular dysfunction [PRTD]) correlated with plasma drug concentrations, which depended on the dosage regimen and age-related changes in drug clearance. Below a threshold area under the concentration-time curve for TFV in plasma of approximately 10 microg x h/ml, an exposure severalfold higher than that observed in humans treated orally with 300 mg TFV disoproxil fumarate (TDF), prolonged TFV administration was not associated with PRTD based on urinalysis, serum chemistry analyses, bone mineral density, and clinical observations. At low-dose maintenance regimens, plasma TFV concentrations and intracellular TFV diphosphate concentrations were similar to or slightly higher than those observed in TDF-treated humans. No new toxicities were identified. The available evidence does not suggest teratogenic effects of prolonged low-dose TFV treatment; by the age of 10 years, one macaque, on TFV treatment since birth, had produced three offspring that were healthy by all criteria up to the age of 5 years. Despite the presence of viral variants with a lysine-to-arginine substitution at codon 65 (K65R) of RT in all 28 SIV-infected animals, 6 animals suppressed viremia to undetectable levels for as long as 12 years of TFV monotherapy. In conclusion, these findings illustrate the safety and sustained benefits of prolonged TFV-containing regimens throughout development from infancy to adulthood, including pregnancy.


Assuntos
Fármacos Anti-HIV , Modelos Animais de Doenças , Complicações Infecciosas na Gravidez , Inibidores da Transcriptase Reversa , Síndrome de Imunodeficiência Adquirida dos Símios , Adenina/administração & dosagem , Adenina/efeitos adversos , Adenina/análogos & derivados , Adenina/farmacocinética , Adenina/uso terapêutico , Fatores Etários , Animais , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/efeitos adversos , Fármacos Anti-HIV/farmacocinética , Fármacos Anti-HIV/uso terapêutico , Feminino , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Humanos , Macaca mulatta , Organofosfonatos/administração & dosagem , Organofosfonatos/efeitos adversos , Organofosfonatos/farmacocinética , Organofosfonatos/uso terapêutico , Gravidez , Complicações Infecciosas na Gravidez/tratamento farmacológico , Complicações Infecciosas na Gravidez/virologia , Inibidores da Transcriptase Reversa/administração & dosagem , Inibidores da Transcriptase Reversa/farmacocinética , Inibidores da Transcriptase Reversa/farmacologia , Inibidores da Transcriptase Reversa/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/fisiologia , Tenofovir , Fatores de Tempo , Resultado do Tratamento
11.
Brain Behav Immun ; 22(5): 676-89, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17719201

RESUMO

From the beginning of the AIDS epidemic, stress has been a suspected contributor to the wide variation seen in disease progression, and some evidence supports this idea. Not all individuals respond to a stressor in the same way, however, and little is known about the biological mechanisms by which variations in individuals' responses to their environment affect disease-relevant immunologic processes. Using the simian immunodeficiency virus/rhesus macaque model of AIDS, we explored how personality (Sociability) and genotype (serotonin transporter promoter) independently interact with social context (Stable or Unstable social conditions) to influence behavioral expression, plasma cortisol concentrations, SIV-specific IgG, and expression of genes associated with Type I interferon early in infection. SIV viral RNA set-point was strongly and negatively correlated with survival as expected. Set-point was also associated with expression of interferon-stimulated genes, with CXCR3 expression, and with SIV-specific IgG titers. Poorer immune responses, in turn, were associated with display of sustained aggression and submission. Personality and genotype acted independently as well as in interaction with social condition to affect behavioral responses. Together, the data support an "interactionist" perspective [Eysenck, H.J., 1991. Personality, stress and disease: an interactionist perspective. Psychol. Inquiry 2, 221-232] on disease. Given that an important goal of HIV treatment is to maintain viral set-point as low as possible, our data suggest that supplementing anti-retroviral therapy with behavioral or pharmacologic modulation of other aspects of an organism's functioning might prolong survival, particularly among individuals living under conditions of threat or uncertainty.


Assuntos
Imunidade/fisiologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Síndrome de Imunodeficiência Adquirida dos Símios/psicologia , Comportamento Social , Agressão/fisiologia , Animais , Anticorpos Antivirais/sangue , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Genótipo , Hidrocortisona/sangue , Imunoglobulina G/sangue , Interferon Tipo I/genética , Macaca mulatta , Masculino , Reação em Cadeia da Polimerase , RNA Viral/sangue , Receptores CXCR3/genética , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Estresse Psicológico/imunologia , Estresse Psicológico/fisiopatologia , Carga Viral , Replicação Viral
12.
J Immunol Methods ; 312(1-2): 45-53, 2006 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-16647080

RESUMO

Simian immunodeficiency virus (SIV)-infection in macaques provides an important animal model for human immunodeficiency virus-1 (HIV-1) infection. The involvement of perforin (PFN), released by cytotoxic cells to mediate killing of virus-infected cells, has been difficult to assess in this experimental model due to a lack of reagents. We therefore evaluated monoclonal antibodies (mAbs) Pf-80, Pf-164 and Pf-344, previously raised against human PFN, for cross-reactivity with macaque PFN. Mabs Pf-164 and Pf-344 reacted with intracellular PFN in peripheral blood mononuclear cells (PBMC) from cynomolgus and rhesus macaques by flow cytometry and stained PFN in rhesus lymphoid tissue by immunohistochemistry (IHC). Moreover, PFN capture enzyme-linked immunosorbent (ELISA) and enzyme-linked immunospot (ELISpot) assays utilizing mAbs Pf-164/Pf-80 for capture and mAb Pf-344 for detection were used to quantify PFN release by mitogen-stimulated cynomolgus and rhesus PBMC. The PFN ELISpot was further used to quantify antigen-specific CD8+ T cells by ex vivo stimulation of PBMC from cynomolgus macaques immunized against SIV/HIV-1. These macaque PFN-reactive mAbs and immunoassays will be valuable new tools for investigation of cytotoxic T lymphocyte (CTL) responses in non-human primate models of infectious diseases as well as for vaccine development.


Assuntos
Anticorpos Monoclonais/imunologia , Modelos Animais de Doenças , Macaca/virologia , Glicoproteínas de Membrana/análise , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Macaca/imunologia , Glicoproteínas de Membrana/metabolismo , Perforina , Proteínas Citotóxicas Formadoras de Poros , Vacinas contra a SAIDS/imunologia , Baço/química , Baço/imunologia , Linfócitos T Citotóxicos/imunologia
13.
Neuroreport ; 25(13): 973-8, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25102373

RESUMO

Pediatric HIV infection remains a global health crisis with a worldwide infection rate of 2.5 million (WHO, Geneva Switzerland, 2009). Children are much more susceptible to HIV-1 neurological impairments compared with adults, which is exacerbated by coinfections. A major obstacle in pediatric HIV research is sample access. The proposed studies take advantage of ongoing pediatric simian immunodeficiency virus (SIV) pathogenesis and vaccine studies to test the hypothesis that pediatric SIV infection diminishes neuronal populations and neurogenesis in the hippocampus. Newborn rhesus macaques (Macaca mulatta) that received intravenous inoculation of highly virulent SIVmac251 (n=3) or vehicle (control n=4) were used in this study. After a 6-18-week survival time, the animals were euthanized and the brains prepared for quantitative histopathological analysis. Systematic sections through the hippocampus were either Nissl stained or immunostained for doublecortin (DCX+), a putative marker of immature neurons. Using design-based stereology, we report a 42% reduction in the pyramidal neuron population of the CA1, CA2, and CA3 fields of the hippocampus (P<0.05) in SIV-infected infants. The DCX+ neuronal population was also significantly reduced within the dentate gyrus of the hippocampus. The loss of hippocampal neurons and neurogenic capacity may contribute to the rapid neurocognitive decline associated with pediatric HIV infection. These data suggest that pediatric SIV infection, which leads to significant neuronal loss in the hippocampus within 3 months, closely models a subset of pediatric HIV infections with rapid progression.


Assuntos
Hipocampo/patologia , Neurônios/patologia , Células Piramidais/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia , Animais , Animais Recém-Nascidos , Proteínas do Domínio Duplacortina , Hipocampo/metabolismo , Imuno-Histoquímica , Macaca mulatta , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Células Piramidais/metabolismo , Distribuição Aleatória , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo
14.
Trials Vaccinol ; 2: 53-63, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24454591

RESUMO

Mother-to-child-transmission of HIV by breast-feeding remains a major obstacle in the eradication of HIV infection. Compared to adults, HIV-infected infants have more rapid disease and show higher susceptibility to co-infections like tuberculosis (TB). Although the Bacille Calmette-Guérin vaccine can be administered at birth to protect against TB, BCG can disseminate in HIV-infected infants and increase mortality. Thus, a pediatric combination vaccine to stop both HIV and TB infection in infants is urgently needed. Towards the goal of developing a pediatric combination HIV-TB vaccine to prevent both oral HIV acquisition by breast-feeding and TB infection, we tested and optimized an immunization regimen using a novel live attenuated Mycobacterium tuberculosis vaccine engineered to express simian immunodeficiency (SIV) antigens followed by heterologous MVA-SIV boosting in the infant macaque model. A single oral dose of the attenuated Mtb-SIV vaccine strain mc26435 during the first week of life was sufficient to induce persistent TB-specific immune responses. SIV-specific immunity was induced at low but comparable magnitudes after oral or intradermal priming, and was enhanced following MVA-SIV boosts. T cell responses were most pronounced in intestinal tissues and oral lymph nodes. Importantly, in addition to plasma SIV-specific IgG and IgA antibodies, infant macaques developed mucosal SIV-specific IgA in saliva and intestinal IgA and IgG. While future SIV and Mtb challenge studies will be needed to determine the protective efficacy of the Mtb-SIV / MVA-SIV vaccine, infants at high risk for oral HIV acquisition by breast-feeding and TB infection could profoundly benefit from an effective combination vaccine.

15.
Sci Transl Med ; 4(136): 136ra69, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649090

RESUMO

The mechanisms by which some HIV-infected subjects resist disease progression, whereas others progress rapidly, are incompletely understood. Viral and host genetic factors, such as nef deletions and major histocompatibility complex alleles, explain a portion of the observed variability. However, it has been difficult to identify host immune functions that may be present before infection and that allow resistance to lentiviral disease progression. Here, we show that simian immunodeficiency virus replication in the infected rhesus macaque is limited by the size of the preexisting T helper 17 (T(H)17) cell compartment: Animals with a high representation of such cells in blood and intestinal tissue before infection experienced peak and set-point viral loads about one log unit lower than those with a lower representation of T(H)17 cells. Reciprocally, treatment of macaques with interleukin-2 and granulocyte colony-stimulating factor before infection led to depletion of T(H)17 cells, reduction of the ratio between T(H)17 cells and CD3(+)CD4(+)CD25(+)CD127(low) regulatory T cells, and higher viral loads for 6 months after infection. These results demonstrate that the composition of the host immune system before infection has an influence on the course of disease after infection. Furthermore, to the extent that this influence shapes and interacts with T cell-mediated responses to virus, our findings provide a new framework for understanding interindividual variation in responses to therapies and vaccines against HIV.


Assuntos
Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Células Th17/metabolismo , Replicação Viral/fisiologia , Animais , Macaca mulatta
16.
Clin Vaccine Immunol ; 19(8): 1170-81, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22695156

RESUMO

Many resource-poor countries are faced with concurrent epidemics of AIDS and tuberculosis (TB) caused by human immunodeficiency virus (HIV) and Mycobacterium tuberculosis, respectively. Dual infections with HIV and M. tuberculosis are especially severe in infants. There is, however, no effective HIV vaccine, and the only licensed TB vaccine, the Mycobacterium bovis bacillus Calmette-Guérin (BCG) vaccine, can cause disseminated mycobacterial disease in HIV-infected children. Thus, a pediatric vaccine to prevent HIV and M. tuberculosis infections is urgently needed. We hypothesized that a highly attenuated M. tuberculosis strain containing HIV antigens could be safely administered at birth and induce mucosal and systemic immune responses to protect against HIV and TB infections, and we rationalized that vaccine safety could be most rigorously assessed in immunocompromised hosts. Of three vaccine candidates tested, the recombinant attenuated M. tuberculosis strain mc(2)6435 carrying a simian immunodeficiency virus (SIV) Gag expression plasmid and harboring attenuations of genes critical for replication (panCD and leuCD) and immune evasion (secA2), was found to be safe for oral or intradermal administration to non-SIV-infected and SIV-infected infant macaques. Safety was defined as the absence of clinical symptoms, a lack of histopathological changes indicative of M. tuberculosis infection, and a lack of mycobacterial dissemination. These data represent an important step in the development of novel TB vaccines and suggest that a combination recombinant attenuated M. tuberculosis-HIV vaccine could be a safe alternative to BCG for the pediatric population as a whole and, more importantly, for the extreme at-risk group of HIV-infected infants.


Assuntos
Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Vacinas contra a Tuberculose/efeitos adversos , Administração Oral , Animais , Animais Recém-Nascidos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Injeções Intradérmicas , Macaca , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/efeitos adversos , Vacinas contra a SAIDS/genética , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/patogenicidade , Vacinas contra a Tuberculose/genética , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética
17.
Vaccine ; 29(17): 3124-37, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21377510

RESUMO

Despite antiretroviral medications, the rate of pediatric HIV-1 infections through breast-milk transmission has been staggering in developing countries. Therefore, the development of a vaccine to protect vulnerable infant populations should be actively pursued. We previously demonstrated that oral immunization of newborn macaques with vesicular stomatitis virus expressing simian immunodeficiency virus genes (VSV-SIV) followed 2 weeks later by an intramuscular boost with modified vaccinia ankara virus expressing SIV (MVA-SIV) successfully induced SIV-specific T and B cell responses in multiple lymphoid tissues, including the tonsil and intestine [13]. In the current study, we tested the oral VSV-SIV prime/systemic MVA-SIV boost vaccine for efficacy against multiple oral SIVmac251 challenges starting two weeks after the booster vaccination. The vaccine did not prevent SIV infection. However, in vaccinated infants, the level of SIV-specific plasma IgA (but not IgG) at the time of challenge was inversely correlated with peak viremia. In addition, the levels of SIV-specific IgA in saliva and plasma were inversely correlated with viral load at euthanasia. Animals with tonsils that contained higher frequencies of SIV-specific TNF-α- or IFN-γ-producing CD8(+) T cells and central memory T cells at euthanasia also had lower viremia. Interestingly, a marked depletion of CD25(+)FoxP3(+)CD4(+) T cells was observed in the tonsils as well as the intestine of these animals, implying that T regulatory cells may be a major target of SIV infection in infant macaques. Overall, the data suggest that, in infant macaques orally infected with SIV, the co-induction of local antiviral cytotoxic T cells and T regulatory cells that promote the development of IgA responses may result in better control of viral replication. Thus, future vaccination efforts should be directed towards induction of IgA and mucosal T cell responses to prevent or reduce virus replication in infants.


Assuntos
Portadores de Fármacos , Vetores Genéticos , Vacinas contra a SAIDS/imunologia , Vacinação/métodos , Vaccinia virus/genética , Vesiculovirus/genética , Administração Oral , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/sangue , Sangue/imunologia , Sangue/virologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Feminino , Imunoglobulina A/análise , Imunoglobulina A/sangue , Injeções Intramusculares , Macaca mulatta , Masculino , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/genética , Saliva/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Linfócitos T Reguladores/imunologia
18.
PLoS One ; 6(4): e19323, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21559444

RESUMO

BACKGROUND: Pathogenic versus protective outcomes to Dengue virus (DENV) infection are associated with innate immune function. This study aimed to determine the role of increased TLR3- and TLR7/8-mediated innate signaling after Dengue infection of rhesus macaques in vivo to evaluate its impact on disease and anti-DENV immune responses. METHODOLOGY/PRINCIPAL FINDINGS: TLR3 and TLR7/8 agonists (emulsified in Montanide) were administered subcutaneously to rhesus macaques at 48 hours and 7 days after DENV infection. The Frequency and activation of myeloid dendritic cells, plasmacytoid dendritic cells, and B cells were measured by flow cytometry while the serum levels of 14 different cytokines and chemokines were quantified. Adaptive immune responses were measured by DENV-specific antibody subtype measurements. Results showed that the combined TLR agonists reduced viral replication and induced the development of a proinflammatory reaction, otherwise absent in Dengue infection alone, without any clear signs of exacerbated disease. Specifically, the TLR-induced response was characterized by activation changes in mDC subsets concurrent with higher serum levels of CXCL-10 and IL-1Ra. TLR stimulation also induced higher titers of anti-DENV antibodies and acted to increase the IgG2/IgG1 ratio of anti-DENV to favor the subtype associated with DENV control. We also observed an effect of DENV-mediated suppression of mDC activation consistent with prior in vitro studies. CONCLUSIONS/SIGNIFICANCE: These data show that concurrent TLR3/7/8 activation of the innate immune response after DENV infection in vivo acts to increase antiviral mechanisms via increased inflammatory and humoral responses in rhesus macaques, resulting in decreased viremia and melioration of the infection. These findings underscore an in vivo protective rather than a pathogenic role for combined TLR3/7/8-mediated activation in Dengue infection of rhesus macaques. Our study provides definitive proof-of-concept into the mechanism by which DENV evades immune recognition and activation in vivo.


Assuntos
Vírus da Dengue/genética , Dengue/imunologia , Receptor 3 Toll-Like/agonistas , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Replicação Viral , Animais , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Imunidade Inata , Inflamação , Leucócitos Mononucleares/citologia , Macaca mulatta , Masculino
19.
J Acquir Immune Defic Syndr ; 55(1): 14-28, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20616742

RESUMO

Type I interferons play an important role in the early defense against viral and other pathogens. These innate responses are also critically important in shaping the subsequent adaptive response. Thus, a more thorough knowledge of innate response types and mechanisms will improve our understanding of pathogenesis and guide the development of new therapeutics. Interferon alpha (IFN-alpha) is used clinically in the treatment of HIV and hepatitis C infections. The majority of IFA-alpha therapy is based on a single IFN-alpha subtype, IFN-alpha2. However, IFN-alpha comprises a family of multiple subtypes. The biologic functions of the distinct subtypes and how they relate to disease are poorly understood. The current study developed the tools to distinguish and measure multiple IFN-alpha subtypes on the mRNA level in rhesus macaques that are used widely as an important animal model for human diseases. We were able to identify and measure nine distinct rhesus IFN-alpha subtypes. Furthermore, we could demonstrate that in response to oral pathogenic SIV infection, several IFN-alpha subtypes are rapidly induced in lymphoid but not at oral and gastrointestinal mucosal surfaces. Although each IFN-alpha subtype was induced at distinct levels, their relative expression patterns were identical in all lymphoid tissues examined.


Assuntos
Interferon-alfa/biossíntese , Interferon-alfa/classificação , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Animais Recém-Nascidos , Perfilação da Expressão Gênica , Interferon-alfa/genética , Mucosa Intestinal/imunologia , Tecido Linfoide/imunologia , Macaca mulatta , Mucosa Bucal/imunologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
20.
Vaccine ; 28(6): 1481-92, 2010 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-19995539

RESUMO

In a previously developed infant macaque model mimicking HIV infection by breast-feeding, we demonstrated that intramuscular immunization with recombinant poxvirus vaccines expressing simian immunodeficiency virus (SIV) structural proteins provided partial protection against infection following oral inoculation with virulent SIV. In an attempt to further increase systemic but also local antiviral immune responses at the site of viral entry, we tested the immunogenicity of different orally administered, replicating vaccines. One group of newborn macaques received an oral prime immunization with a recombinant vesicular stomatitis virus expressing SIVmac239 gag, pol and env (VSV-SIVgpe), followed 2 weeks later by an intramuscular boost immunization with MVA-SIV. Another group received two immunizations with live-attenuated SIVmac1A11, administered each time both orally and intravenously. Control animals received mock immunizations or non-SIV VSV and MVA control vectors. Analysis of SIV-specific immune responses in blood and lymphoid tissues at 4 weeks of age demonstrated that both vaccine regimens induced systemic antibody responses and both systemic and local cell-mediated immune responses. The safety and immunogenicity of the VSV-SIVgpe+MVA-SIV immunization regimen described in this report provide the scientific incentive to explore the efficacy of this vaccine regimen against virulent SIV exposure in the infant macaque model.


Assuntos
Vetores Genéticos , Imunização Secundária/métodos , Vacinas contra a SAIDS/imunologia , Vacinação/métodos , Vaccinia virus/genética , Vesiculovirus/genética , Administração Oral , Animais , Animais Recém-Nascidos , Anticorpos Anti-HIV/sangue , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Macaca mulatta , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/efeitos adversos , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA