Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Front Endocrinol (Lausanne) ; 12: 745984, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34630335

RESUMO

Although spermatogenic dysfunction is widely found in patients with human immunodeficiency virus (HIV), the underlying reasons remain unclear. Thus far, potential hypotheses involving viral reservoirs, testicular inflammation, hormone imbalance, and cachexia show inconsistent correlation with spermatogenic dysfunction. Here, northern pig-tailed macaques (NPMs) exhibited marked spermatogenic dysfunction after long-term infection with simian immunodeficiency virus (SIVmac239), with significant decreases in Johnsen scores, differentiated spermatogonial stem cells, and testicular proliferating cells. The above hypotheses were also evaluated. Results showed no differences between SIV- and SIV+ NPMs, except for an increase in follicle stimulating hormone (FSH) during SIV infection, which had no direct effect on the testes. However, long-term SIVmac239 infection undermined pancreatic islet ß cell function, partly represented by significant reductions in cellular counts and autophagy levels. Pancreatic islet ß cell dysfunction led to glucose metabolism disorder at the whole-body level, which inhibited lactate production by Sertoli cells in testicular tissue. As lactate is the main energy substrate for developing germ cells, its decrease was strongly correlated with spermatogenic dysfunction. Therefore, glucose metabolism disorder appears to be a primary cause of spermatogenic dysfunction in NPMs with long-term SIVmac239 infection.


Assuntos
Transtornos do Metabolismo de Glucose/complicações , Macaca nemestrina , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Espermatogênese/fisiologia , Animais , Glucose/metabolismo , Transtornos do Metabolismo de Glucose/fisiopatologia , Transtornos do Metabolismo de Glucose/veterinária , Infertilidade Masculina/etiologia , Infertilidade Masculina/metabolismo , Infertilidade Masculina/fisiopatologia , Infertilidade Masculina/veterinária , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/virologia , Macaca nemestrina/metabolismo , Macaca nemestrina/fisiologia , Macaca nemestrina/virologia , Masculino , Análise do Sêmen/veterinária , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/fisiopatologia , Vírus da Imunodeficiência Símia/fisiologia
2.
J Neurovirol ; 26(4): 511-519, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32488843

RESUMO

HIV-associated neuroinflammation is primarily driven by CNS macrophages including microglia. Regulation of these immune responses, however, remains to be characterized in detail. Using the SIV/macaque model of HIV, we evaluated CNS expression of triggering receptor expressed on myeloid cells 2 (TREM2) which is constitutively expressed by microglia and contributes to cell survival, proliferation, and differentiation. Loss-of-function mutations in TREM2 are recognized risk factors for neurodegenerative diseases including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Nasu-Hakola disease (NHD); recent reports have also indicated a role for TREM2 in HIV-associated neuroinflammation. Using in situ hybridization (ISH) and qRT-PCR, TREM2 mRNA levels were found to be significantly elevated in frontal cortex of macaques with SIV encephalitis compared with uninfected controls (P = 0.02). TREM2 protein levels were also elevated as measured by ELISA of frontal cortex tissue homogenates in these animals. Previously, we characterized the expression of CSF1R (colony-stimulating factor 1 receptor) in this model; the TREM2 and CSF1R promoters both contain a PU.1 binding site. While TREM2 and CSF1R mRNA levels in the frontal cortex were highly correlated (Spearman R = 0.79, P < 0.001), protein levels were not well correlated. In SIV-infected macaques released from ART to study viral rebound, neither TREM2 nor CSF1R mRNA increased with rebound viremia. However, CSF1R protein levels remained significantly elevated unlike TREM2 (P = 0.02). This differential expression suggests that TREM2 and CSF1R play unique, distinct roles in the pathogenesis of HIV CNS disease.


Assuntos
Encefalite Viral/genética , Macaca nemestrina/imunologia , Macrófagos/imunologia , Glicoproteínas de Membrana/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Vírus da Imunodeficiência Símia/imunologia , Animais , Terapia Antirretroviral de Alta Atividade/métodos , Antivirais/farmacologia , Esquema de Medicação , Encefalite Viral/tratamento farmacológico , Encefalite Viral/imunologia , Encefalite Viral/virologia , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/imunologia , Lobo Frontal/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Macaca nemestrina/genética , Macaca nemestrina/virologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Masculino , Glicoproteínas de Membrana/imunologia , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/virologia , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/imunologia , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Transativadores/genética , Transativadores/imunologia
3.
PLoS Pathog ; 15(7): e1007925, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31260493

RESUMO

HIV-1 does not persistently infect macaques due in part to restriction by several macaque host factors. This has been partially circumvented by generating chimeric SIV/HIV-1 viruses (SHIVs) that encode SIV antagonist of known restriction factors. However, most SHIVs replicate poorly in macaques unless they are further adapted in culture and/or macaques (adapted SHIVs). Therefore, development of SHIVs encoding HIV-1 sequences derived directly from infected humans without adaptation (unadapted SHIVs) has been challenging. In contrast to the adapted SHIVs, the unadapted SHIVs have lower replication kinetics in macaque lymphocytes and are sensitive to type-1 interferon (IFN). The HIV-1 Envelope (Env) in the chimeric virus determines both the reduced replication and the IFN-sensitivity differences. There is limited information on macaque restriction factors that specifically limit replication of the more biologically relevant, unadapted SHIV variants. In order to identify the IFN-induced host factor(s) that could contribute to the inhibition of SHIVs in macaque lymphocytes, we measured IFN-induced gene expression in immortalized pig-tailed macaque (Ptm) lymphocytes using RNA-Seq. We found 147 genes that were significantly upregulated upon IFN treatment in Ptm lymphocytes and 31/147 were identified as genes that encode transmembrane helices and thus are likely present in membranes where interaction with viral Env is plausible. Within this group of upregulated genes with putative membrane-localized proteins, we identified several interferon-induced transmembrane protein (IFITM) genes, including several previously uncharacterized Ptm IFITM3-related genes. An evolutionary genomic analysis of these genes suggests the genes are IFITM3 duplications not found in humans that are both within the IFITM locus and also dispersed elsewhere in the Ptm genome. We observed that Ptm IFITMs are generally packaged at higher levels in unadapted SHIVs when compared to adapted SHIVs. CRISPR/Cas9-mediated knockout of Ptm IFITMs showed that depletion of IFITMs partially rescues the IFN sensitivity of unadapted SHIV. Moreover, we found that the depletion of IFITMs also increased replication of unadapted SHIV in the absence of IFN treatment, suggesting that Ptm IFITMs are likely important host factors that limit replication of unadapted SHIVs. In conclusion, this study shows that Ptm IFITMs selectively restrict replication of unadapted SHIVs. These findings suggest that restriction factors including IFITMs vary in their potency against different SHIV variants and may play a role in selecting for viruses that adapt to species-specific restriction factors.


Assuntos
HIV-1/fisiologia , HIV-1/patogenicidade , Vírus da Imunodeficiência Símia/fisiologia , Vírus da Imunodeficiência Símia/patogenicidade , Produtos do Gene env do Vírus da Imunodeficiência Humana/fisiologia , Adaptação Fisiológica , Animais , Genes env , HIV-1/genética , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Especificidade de Hospedeiro , Humanos , Interferon-alfa/metabolismo , Macaca nemestrina/genética , Macaca nemestrina/imunologia , Macaca nemestrina/virologia , Processamento de Proteína Pós-Traducional , Vírus Reordenados/genética , Vírus Reordenados/patogenicidade , Vírus Reordenados/fisiologia , Vírus da Imunodeficiência Símia/genética , Replicação Viral
4.
Sci Rep ; 9(1): 5661, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30952887

RESUMO

A HIV vaccine that provides mucosal immunity is urgently needed. We evaluated an intranasal recombinant Fowlpox virus (rFPV) priming vaccine followed by intramuscular Modified Vaccinia Ankara (rMVA) booster vaccine, both expressing SIV antigens. The vaccination generated mucosal and systemic SIV-specific CD4+ T cell mediated immunity and was associated with partial protection against high-dose intrarectal SIVmac251 challenge in outbred pigtail macaques. Three of 12 vaccinees were completely protected and these animals elicited sustained Gag-specific poly-functional, cytotoxic mucosal CD4+ T cells, complemented by systemic poly-functional CD4+ and CD8+ T cell immunity. Humoral immune responses, albeit absent in completely protected macaques, were associated with partial control of viremia in animals with relatively weaker mucosal/systemic T cell responses. Co-expression of an IL-4R antagonist by the rFPV vaccine further enhanced the breadth and cytotoxicity/poly-functionality of mucosal vaccine-specific CD4+ T cells. Moreover, a single FPV-gag/pol/env prime was able to induce rapid anamnestic gp140 antibody response upon SIV encounter. Collectively, our data indicated that nasal vaccination was effective at inducing robust cervico-vaginal and rectal immunity, although cytotoxic CD4+ T cell mediated mucosal and systemic immunity correlated strongly with 'complete protection', the different degrees of protection observed was multi-factorial.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vírus da Varíola das Aves Domésticas/imunologia , Macaca nemestrina/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vacinas Sintéticas/imunologia , Vacinas contra a AIDS/imunologia , Administração Intranasal/métodos , Animais , Linfócitos T CD8-Positivos/imunologia , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Imunidade nas Mucosas/imunologia , Imunização Secundária/métodos , Memória Imunológica/imunologia , Injeções Intramusculares/métodos , Macaca nemestrina/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vacinação/métodos , Vacínia/imunologia , Vaccinia virus/imunologia
5.
Nat Commun ; 9(1): 3371, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30135445

RESUMO

The immunological and virological events that contribute to the establishment of Zika virus (ZIKV) infection in humans are unclear. Here, we show that robust cellular innate immune responses arising early in the blood and tissues in response to ZIKV infection are significantly stronger in males and correlate with increased viral persistence. In particular, early peripheral blood recruitment of plasmacytoid dendritic cells and higher production of monocyte chemoattractant protein (MCP-1) correspond with greater viral persistence and tissue dissemination. We also identify non-classical monocytes as primary in vivo targets of ZIKV infection in the blood and peripheral lymph node. These results demonstrate the potential differences in ZIKV pathogenesis between males and females and a key role for early cellular innate immune responses in the blood in viral dissemination and ZIKV pathogenesis.


Assuntos
Imunidade Inata/fisiologia , Macaca nemestrina/imunologia , Macaca nemestrina/virologia , Zika virus/imunologia , Animais , Quimiocina CCL2/metabolismo , Macaca nemestrina/metabolismo , Infecção por Zika virus/imunologia , Infecção por Zika virus/metabolismo
6.
Nat Med ; 24(3): 368-374, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29400709

RESUMO

Zika virus (ZIKV) is a flavivirus with teratogenic effects on fetal brain, but the spectrum of ZIKV-induced brain injury is unknown, particularly when ultrasound imaging is normal. In a pregnant pigtail macaque (Macaca nemestrina) model of ZIKV infection, we demonstrate that ZIKV-induced injury to fetal brain is substantial, even in the absence of microcephaly, and may be challenging to detect in a clinical setting. A common and subtle injury pattern was identified, including (i) periventricular T2-hyperintense foci and loss of fetal noncortical brain volume, (ii) injury to the ependymal epithelium with underlying gliosis and (iii) loss of late fetal neuronal progenitor cells in the subventricular zone (temporal cortex) and subgranular zone (dentate gyrus, hippocampus) with dysmorphic granule neuron patterning. Attenuation of fetal neurogenic output demonstrates potentially considerable teratogenic effects of congenital ZIKV infection even without microcephaly. Our findings suggest that all children exposed to ZIKV in utero should receive long-term monitoring for neurocognitive deficits, regardless of head size at birth.


Assuntos
Feto/virologia , Complicações Infecciosas na Gravidez/fisiopatologia , Infecção por Zika virus/virologia , Zika virus/patogenicidade , Animais , Modelos Animais de Doenças , Feminino , Feto/fisiopatologia , Humanos , Macaca nemestrina/virologia , Microcefalia/diagnóstico por imagem , Microcefalia/fisiopatologia , Microcefalia/virologia , Neurogênese/genética , Gravidez , Complicações Infecciosas na Gravidez/diagnóstico por imagem , Complicações Infecciosas na Gravidez/virologia , Zika virus/genética , Infecção por Zika virus/genética , Infecção por Zika virus/fisiopatologia
7.
J Med Primatol ; 46(4): 158-161, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28748668

RESUMO

Simian betaretroviruses include the well-known exogenous simian retroviruses (SRV-1 through SRV-8), and some closely related simian endogenous retroviruses (SERV). Here, we characterized two new viral genomes, which appear to represent novel SERVs but have characteristics of both SRV and SERV highlighting the need to develop new assays providing molecular and serologic differentiation of SERV and SRV to avoid false positives.


Assuntos
Retrovirus Endógenos/isolamento & purificação , Genoma Viral , Macaca nemestrina/virologia , Infecções por Retroviridae/diagnóstico , Retrovirus dos Símios/isolamento & purificação , Infecções Tumorais por Vírus/diagnóstico , Animais , Retrovirus Endógenos/genética , Indonésia , Retrovirus dos Símios/genética
8.
PLoS One ; 11(7): e0159281, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27415779

RESUMO

In the 20th century, thirteen distinct human immunodeficiency viruses emerged following independent cross-species transmission events involving simian immunodeficiency viruses (SIV) from African primates. In the late 1900s, pathogenic SIV strains also emerged in the United Sates among captive Asian macaque species following their unintentional infection with SIV from African sooty mangabeys (SIVsmm). Since their discovery in the 1980s, SIVs from rhesus macaques (SIVmac) and pig-tailed macaques (SIVmne) have become invaluable models for studying HIV pathogenesis, vaccine design and the emergence of viruses. SIV isolates from captive crab-eating macaques (SIVmfa) were initially described but lost prior to any detailed molecular and genetic characterization. In order to infer the origins of the lost SIVmfa lineage, we located archived material and colony records, recovered its genomic sequence by PCR, and assessed its phylogenetic relationship to other SIV strains. We conclude that SIVmfa is the product of two cross-species transmission events. The first was the established transmission of SIVsmm to rhesus macaques, which occurred at the California National Primate Research Center in the late 1960s and the virus later emerged as SIVmac. In a second event, SIVmac was transmitted to crab-eating macaques, likely at the Laboratory for Experimental Medicine and Surgery in Primates in the early 1970s, and it was later spread to the New England Primate Research Center colony in 1973 and eventually isolated in 1986. Our analysis suggests that SIVmac had already emerged by the early 1970s and had begun to diverge into distinct lineages. Furthermore, our findings suggest that pathogenic SIV strains may have been more widely distributed than previously appreciated, raising the possibility that additional isolates may await discovery.


Assuntos
Macaca fascicularis/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Animais , Cercocebus atys/virologia , Modelos Animais de Doenças , Genoma Viral/genética , Macaca nemestrina/virologia , Filogenia
9.
J Neurovirol ; 22(4): 498-507, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26727909

RESUMO

In the fourth decade of the HIV epidemic, the relationship between host immunity and HIV central nervous system (CNS) disease remains incompletely understood. Using a simian immunodeficiency virus (SIV)/macaque model, we examined CNS outcomes in pigtailed macaques expressing the MHC class I allele Mane-A1*084:01 which confers resistance to SIV-induced CNS disease and induces the prototypic viral escape mutation Gag K165R. Insertion of gag K165R into the neurovirulent clone SIV/17E-Fr reduced viral replication in vitro compared to SIV/17E-Fr. We also found lower cerebrospinal fluid (CSF), but not plasma, viral loads in macaques inoculated with SIV/17E-Fr K165R versus those inoculated with wildtype. Although escape mutation K165R was genotypically stable in plasma, it rapidly reverted to wildtype Gag KP9 in both CSF and in microglia cultures. We induced robust Gag KP9-specific CTL tetramer responses by vaccinating Mane-A*084:01-positive pigtailed macaques with a Gag KP9 virus-like particle (VLP) vaccine. Upon SIV/17E-Fr challenge, vaccinated animals had lower SIV RNA in CSF compared to unvaccinated controls, but showed no difference in plasma viral loads. These data clearly demonstrate that viral fitness in the CNS is distinct from the periphery and underscores the necessity of understanding the consequences of viral escape in CNS disease with the advent of new therapeutic vaccination strategies.


Assuntos
Produtos do Gene gag/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , RNA Viral/líquido cefalorraquidiano , Vacinas contra a SAIDS/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Alelos , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Expressão Gênica , Produtos do Gene gag/genética , Antígenos de Histocompatibilidade Classe I/genética , Evasão da Resposta Imune/genética , Macaca nemestrina/imunologia , Macaca nemestrina/virologia , Masculino , Microglia/imunologia , Microglia/virologia , Mutação , Cultura Primária de Células , RNA Viral/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/patogenicidade , Vacinação , Carga Viral/imunologia , Replicação Viral/imunologia
10.
Comp Med ; 65(5): 448-54, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26473350

RESUMO

Measles is a highly contagious viral disease in NHP. The infection can range from asymptomatic to rapidly fatal, resulting in significant morbidity and mortality in captive populations. In addition to appropriate quarantine practices, restricted access, the immunization of all personnel in contact with NHP, and the wearing of protective clothing including face masks, measles immunization further reduces the infection risk. Commercially available measles vaccines are effective for use in NHP, but interruptions in their availability have prevented the implementation of ongoing, consistent vaccination programs. This need for a readily available vaccine led us to perform a broad, multicenter safety and immunogenicity study of another candidate vaccine, MVac (Serum Institute of India), a monovalent measles vaccine derived from live Edmonston-Zagreb strain virus that had been attenuated after 22 passages on human diploid cells.


Assuntos
Macaca mulatta/virologia , Macaca nemestrina/virologia , Vacina contra Sarampo/administração & dosagem , Vírus do Sarampo/imunologia , Sarampo/veterinária , Doenças dos Macacos/prevenção & controle , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Feminino , Esquemas de Imunização , Macaca mulatta/imunologia , Macaca nemestrina/imunologia , Masculino , Sarampo/sangue , Sarampo/imunologia , Sarampo/prevenção & controle , Sarampo/virologia , Vacina contra Sarampo/efeitos adversos , Vacina contra Sarampo/imunologia , Vírus do Sarampo/patogenicidade , Doenças dos Macacos/sangue , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Testes Sorológicos/veterinária , Fatores de Tempo , Estados Unidos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia
11.
J Virol ; 89(20): 10156-75, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26223646

RESUMO

UNLABELLED: Deletion of Gly-720 and Tyr-721 from a highly conserved GYxxØ trafficking signal in the SIVmac239 envelope glycoprotein cytoplasmic domain, producing a virus termed ΔGY, leads to a striking perturbation in pathogenesis in rhesus macaques (Macaca mulatta). Infected macaques develop immune activation and progress to AIDS, but with only limited and transient infection of intestinal CD4(+) T cells and an absence of microbial translocation. Here we evaluated ΔGY in pig-tailed macaques (Macaca nemestrina), a species in which SIVmac239 infection typically leads to increased immune activation and more rapid progression to AIDS than in rhesus macaques. In pig-tailed macaques, ΔGY also replicated acutely to high peak plasma RNA levels identical to those for SIVmac239 and caused only transient infection of CD4(+) T cells in the gut lamina propria and no microbial translocation. However, in marked contrast to rhesus macaques, 19 of 21 pig-tailed macaques controlled ΔGY replication with plasma viral loads of <15 to 50 RNA copies/ml. CD4(+) T cells were preserved in blood and gut for up to 100 weeks with no immune activation or disease progression. Robust antiviral CD4(+) T cell responses were seen, particularly in the gut. Anti-CD8 antibody depletion demonstrated CD8(+) cellular control of viral replication. Two pig-tailed macaques progressed to disease with persisting viremia and possible compensatory mutations in the cytoplasmic tail. These studies demonstrate a marked perturbation in pathogenesis caused by ΔGY's ablation of the GYxxØ trafficking motif and reveal, paradoxically, that viral control is enhanced in a macaque species typically predisposed to more pathogenic manifestations of simian immunodeficiency virus (SIV) infection. IMPORTANCE: The pathogenesis of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) reflects a balance between viral replication, host innate and adaptive antiviral immune responses, and sustained immune activation that in humans and Asian macaques is associated with persistent viremia, immune escape, and AIDS. Among nonhuman primates, pig-tailed macaques following SIV infection are predisposed to more rapid disease progression than are rhesus macaques. Here, we show that disruption of a conserved tyrosine-based cellular trafficking motif in the viral transmembrane envelope glycoprotein cytoplasmic tail leads in pig-tailed macaques to a unique phenotype in which high levels of acute viral replication are followed by elite control, robust cellular responses in mucosal tissues, and no disease. Paradoxically, control of this virus in rhesus macaques is only partial, and progression to AIDS occurs. This novel model should provide a powerful tool to help identify host-specific determinants for viral control with potential relevance for vaccine development.


Assuntos
Motivos de Aminoácidos , Linfócitos T CD4-Positivos/imunologia , Imunidade nas Mucosas , Macaca nemestrina/virologia , Deleção de Sequência , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Linfócitos T CD4-Positivos/virologia , Progressão da Doença , Feminino , Expressão Gênica , Intestinos/imunologia , Intestinos/virologia , Macaca mulatta/virologia , Masculino , Dados de Sequência Molecular , Mucosa/imunologia , Mucosa/virologia , Sinais Direcionadores de Proteínas , Estrutura Terciária de Proteína , Transporte Proteico , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Especificidade da Espécie , Proteínas do Envelope Viral/deficiência , Proteínas do Envelope Viral/genética , Carga Viral/genética , Carga Viral/imunologia , Viremia/imunologia , Viremia/patologia , Replicação Viral/genética , Replicação Viral/imunologia
12.
PLoS One ; 10(3): e0121666, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25822622

RESUMO

TRIMCyp is a fusion protein consisting of the TRIM5 gene product and retrotransposed Cyclophilin A (CypA). Two primate TRIMCyp fusion proteins with varying anti-HIV-1 activities independently evolved in owl monkeys and Old World monkeys. In addition, Old World monkey TRIMCyps lack exon7, which encodes amino acids in the Linker2 region. Previous studies on TRIM5α indicated that this region affects anti-retroviral activity, cytoplasmic body formation, and multimerization. The effects of exon7 deletion on the functions of the TRIMCyp are unclear. In this study, we found that the cytoplasmic bodies and multimers of owl monkey TRIMCyp (omTRIMCyp) are different from those of northern pig-tailed macaque TRIMCyp (npmTRIMCyp). In addition, we demonstrated that exon7 deletion affected cytoplasmic body formation and multimerization. Moreover, we unexpectedly found two chimeric proteins of omTRIMCyp and npmTRIMCyp that failed to block HIV-1 replication, despite the presence of CypA in omTRIMCyp. Further studies indicated that the cytoplasmic bodies and spontaneous multimerization were not responsible for TRIMCyp anti-HIV-1 activity. Moreover, potent viral restriction is associated with higher amounts of monomeric TRIMCyp when the CypA domain is able to recognize and bind to the HIV-1 capsid. Our results suggested that the deletion of exon7 during the evolution of TRIMCyp affected its function.


Assuntos
Proteínas de Transporte/genética , Ciclofilina A/genética , HIV-1/fisiologia , Animais , Aotidae/genética , Aotidae/virologia , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Gatos , Linhagem Celular , Ciclofilina A/química , Ciclofilina A/fisiologia , Citoplasma/genética , Citoplasma/virologia , Evolução Molecular , Éxons , Células HEK293 , HIV-1/patogenicidade , Especificidade de Hospedeiro , Humanos , Macaca nemestrina/genética , Macaca nemestrina/virologia , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Deleção de Sequência , Virulência , Montagem de Vírus , Replicação Viral
13.
J Neurovirol ; 21(2): 148-58, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25672885

RESUMO

Immune pressure exerted by MHC class I-restricted cytotoxic T cells drives the development of viral escape mutations, thereby regulating HIV disease progression. Nonetheless, the relationship between host immunity and HIV central nervous system (CNS) disease remains poorly understood. The simian immunodeficiency virus (SIV) macaque model recapitulates key features of HIV infection including development of AIDS and CNS disease. To investigate cell-mediated immunity regulating SIV CNS disease progression, we compared the incidence of SIV encephalitis and the influence of MHC class I allele expression on the development of CNS disease in rhesus macaques (Macaca mulatta) versus pigtailed macaques (Macaca nemestrina). After inoculation with the immunosuppressive swarm SIV/DeltaB670 and the neurovirulent molecular clone SIV/17E-Fr, pigtailed macaques progressed more rapidly to AIDS, had higher plasma and cerebrospinal fluid (CSF) viral loads, and were more likely to progress to SIV-associated encephalitis (SIVE) compared to rhesus macaques. In addition, MHC class I alleles were neuroprotective in both species (Mamu-A*001 in rhesus macaques and Mane-A1*084:01:01 in pigtailed macaques); animals expressing these alleles were less likely to develop SIV encephalitis and correspondingly had lower viral replication in the brain. Species-specific differences in susceptibility to SIV disease demonstrated that cell mediated immune responses are critical to SIV CNS disease progression.


Assuntos
Suscetibilidade a Doenças/imunologia , Macaca mulatta/virologia , Macaca nemestrina/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Complexo AIDS Demência/imunologia , Animais , Progressão da Doença , Antígenos de Histocompatibilidade Classe I/imunologia , Macaca mulatta/imunologia , Macaca nemestrina/imunologia , Vírus da Imunodeficiência Símia
14.
J Leukoc Biol ; 97(3): 599-609, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25605872

RESUMO

The testis is a site of immune privilege in rodents, and there is evidence that T cell responses are also suppressed in the primate testis. Local immunosuppression is a potential mechanism for HIV persistence in tissue reservoirs that few studies have examined. The response of the pig-tailed macaque testis to SIVmac239 infection was characterized to test this possibility. Testes were surgically removed during early-chronic (10 wk) and late-chronic (24-30 wk) SIV infection in 4 animals and compared with those from 7 uninfected animals. SIV infection caused only minor disruption to the seminiferous epithelium without marked evidence of inflammation or consistent changes in total intratesticular leukocyte numbers. Infection also led to an increase in the relative proportion of testicular effector memory CD8(+) T cell numbers and a corresponding reduction in central memory CD4(+) T cells. A decrease in the relative proportion of resident-type CD163(+) macrophages and DCs was also observed. SIV-specific CD8(+) T cells were detectable in the testis, 10-11 wk after infection by staining with SIV Gag-specific or Tat-specific MHC-I tetramers. However, testicular CD8(+) T cells from the infected animals had suppressed cytokine responses to mitogen activation. These results support the possibility that local immunosuppression in the testis may be restricting the ability of T cells to respond to SIV or HIV infection. Local immunosuppression in the testis may be an underexplored mechanism allowing HIV persistence.


Assuntos
Imunidade , Macaca nemestrina/imunologia , Macaca nemestrina/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/imunologia , Testículo/imunologia , Animais , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Células Dendríticas/patologia , Granulócitos/patologia , Células HEK293 , Humanos , Células Matadoras Naturais/patologia , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/patologia , Masculino , Fenótipo , Túbulos Seminíferos/imunologia , Túbulos Seminíferos/patologia , Túbulos Seminíferos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Testículo/patologia , Testículo/virologia
15.
J Virol ; 89(7): 3888-909, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25609822

RESUMO

UNLABELLED: Two rhadinovirus lineages have been identified in Old World primates. The rhadinovirus 1 (RV1) lineage consists of human herpesvirus 8, Kaposi's sarcoma-associated herpesvirus (KSHV), and closely related rhadinoviruses of chimpanzees, gorillas, macaques and other Old World primates. The RV2 rhadinovirus lineage is distinct and consists of closely related viruses from the same Old World primate species. Rhesus macaque rhadinovirus (RRV) is the RV2 prototype, and two RRV isolates, 26-95 and 17577, were sequenced. We determined that the pig-tailed macaque RV2 rhadinovirus, MneRV2, is highly associated with lymphomas in macaques with simian AIDS. To further study the role of rhadinoviruses in the development of lymphoma, we sequenced the complete genome of MneRV2 and identified 87 protein coding genes and 17 candidate microRNAs (miRNAs). A strong genome colinearity and sequence homology were observed between MneRV2 and RRV26-95, although the open reading frame (ORF) encoding the KSHV ORFK15 homolog was disrupted in RRV26-95. Comparison with MneRV2 revealed several genomic anomalies in RRV17577 that were not present in other rhadinovirus genomes, including an N-terminal duplication in ORF4 and a recombinative exchange of more distantly related homologs of the ORF22/ORF47 interacting glycoprotein genes. The comparison with MneRV2 has revealed novel genes and important conservation of protein coding domains and transcription initiation, termination, and splicing signals, which have added to our knowledge of RV2 rhadinovirus genetics. Further comparisons with KSHV and other RV1 rhadinoviruses will provide important avenues for dissecting the biology, evolution, and pathology of these closely related tumor-inducing viruses in humans and other Old World primates. IMPORTANCE: This work provides the sequence characterization of MneRV2, the pig-tailed macaque homolog of rhesus rhadinovirus (RRV). MneRV2 and RRV belong to the rhadinovirus 2 (RV2) rhadinovirus lineage of Old World primates and are distinct but related to Kaposi's sarcoma-associated herpesvirus (KSHV), the etiologic agent of Kaposi's sarcoma. Pig-tailed macaques provide important models of human disease, and our previous studies have indicated that MneRV2 plays a causal role in AIDS-related lymphomas in macaques. Delineation of the MneRV2 sequence has allowed a detailed characterization of the genome structure, and evolutionary comparisons with RRV and KSHV have identified conserved promoters, splice junctions, and novel genes. This comparison provides insight into RV2 rhadinovirus biology and sets the groundwork for more intensive next-generation (Next-Gen) transcript and genetic analysis of this class of tumor-inducing herpesvirus. This study supports the use of MneRV2 in pig-tailed macaques as an important model for studying rhadinovirus biology, transmission and pathology.


Assuntos
DNA Viral/química , DNA Viral/genética , Genoma Viral , Herpesvirus Humano 8/genética , Filogenia , Rhadinovirus/genética , Animais , Macaca nemestrina/virologia , MicroRNAs/genética , Dados de Sequência Molecular , Rhadinovirus/isolamento & purificação , Análise de Sequência de DNA , Homologia de Sequência , Sintenia , Proteínas Virais/genética
16.
Science ; 344(6190): 1401-5, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24948736

RESUMO

Primate lentiviruses exhibit narrow host tropism, reducing the occurrence of zoonoses but also impairing the development of optimal animal models of AIDS. To delineate the factors limiting cross-species HIV-1 transmission, we passaged a modified HIV-1 in pigtailed macaques that were transiently depleted of CD8(+) cells during acute infection. During adaptation over four passages in macaques, HIV-1 acquired the ability to antagonize the macaque restriction factor tetherin, replicated at progressively higher levels, and ultimately caused marked CD4(+) T cell depletion and AIDS-defining conditions. Transient treatment with an antibody to CD8 during acute HIV-1 infection caused rapid progression to AIDS, whereas untreated animals exhibited an elite controller phenotype. Thus, an adapted HIV-1 can cause AIDS in macaques, and stark differences in outcome can be determined by immunological perturbations during early infection.


Assuntos
Síndrome da Imunodeficiência Adquirida/virologia , Modelos Animais de Doenças , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Macaca nemestrina/virologia , Síndrome da Imunodeficiência Adquirida/imunologia , Síndrome da Imunodeficiência Adquirida/transmissão , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Antígenos CD8/imunologia , HIV-1/genética , Proteínas do Vírus da Imunodeficiência Humana/química , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Depleção Linfocítica , Macaca nemestrina/imunologia , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Replicação Viral
17.
J Infect Dis ; 210(6): 904-12, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24688074

RESUMO

We recently demonstrated direct evidence of increased monoamine oxidase (MAO) activity in the brain of a simian immunodeficiency virus (SIV) model of human immunodeficiency virus (HIV)-associated central nervous system (CNS) disease, consistent with previously reported dopamine deficits in both SIV and HIV infection. In this study, we explored potential mechanisms behind this elevated activity. MAO B messenger RNA was highest in macaques with the most severe SIV-associated CNS lesions and was positively correlated with levels of CD68 and GFAP transcripts in the striatum. MAO B messenger RNA also correlated with viral loads in the CNS of SIV-infected macaques and with oxidative stress. Furthermore, in humans, striatal MAO activity was elevated in individuals with HIV encephalitis, compared with activity in HIV-seronegative controls. These data suggest that the neuroinflammation and oxidative stress caused by SIV infection in the CNS may provide the impetus for increased transcription of MAO B and that MAO, and more broadly, oxidative stress, have significant potential as therapeutic targets in CNS disease due to HIV.


Assuntos
Complexo AIDS Demência/enzimologia , Encéfalo/enzimologia , Monoaminoxidase/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/enzimologia , Adulto , Animais , Química Encefálica , Corpo Estriado/enzimologia , Feminino , Perfilação da Expressão Gênica , Glutationa/análise , Humanos , Macaca nemestrina/virologia , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Reação em Cadeia da Polimerase em Tempo Real , Carga Viral
18.
PLoS One ; 9(4): e94375, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24732038

RESUMO

HIV immune pathogenesis is postulated to involve two major mechanisms: 1) chronic innate immune responses that drive T cell activation and apoptosis and 2) induction of immune regulators that suppress T cell function and proliferation. Both arms are elevated chronically in lymphoid tissues of non-natural hosts, which ultimately develop AIDS. However, these mechanisms are not elevated chronically in natural hosts of SIV infection that avert immune pathogenesis despite similarly high viral loads. In this study we investigated whether minocycline could modulate these pathogenic antiviral responses in non-natural hosts of HIV and SIV. We found that minocycline attenuated in vitro induction of type I interferon (IFN) and the IFN-stimulated genes indoleamine 2,3-dioxygenase (IDO1) and TNF-related apoptosis inducing ligand (TRAIL) in human plasmacytoid dendritic cells and PBMCs exposed to aldrithiol-2 inactivated HIV or infectious influenza virus. Activation-induced TRAIL and expression of cytotoxic T-lymphocyte antigen 4 (CTLA-4) in isolated CD4+ T cells were also reduced by minocycline. Translation of these in vitro findings to in vivo effects, however, were mixed as minocycline significantly reduced markers of activation and activation-induced cell death (CD25, Fas, caspase-3) but did not affect expression of IFNß or the IFN-stimulated genes IDO1, FasL, or Mx in the spleens of chronically SIV-infected pigtailed macaques. TRAIL expression, reflecting the mixed effects of minocycline on activation and type I IFN stimuli, was reduced by half, but this change was not significant. These results show that minocycline administered after infection may protect against aspects of activation-induced cell death during HIV/SIV immune disease, but that in vitro effects of minocycline on type I IFN responses are not recapitulated in a rapid progressor model in vivo.


Assuntos
Infecções por HIV/imunologia , HIV-1/imunologia , Imunidade , Minociclina/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Tetraciclina/química , Animais , Antirretrovirais/farmacologia , Antirretrovirais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Antígeno CTLA-4/metabolismo , Separação Celular , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Quimioterapia Combinada , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Humanos , Imunidade/efeitos dos fármacos , Imunidade/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Influenza Humana/imunologia , Influenza Humana/virologia , Interferon Tipo I/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Macaca nemestrina/imunologia , Macaca nemestrina/virologia , Minociclina/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Tetraciclina/farmacologia
19.
J Virol ; 87(24): 13676-93, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109218

RESUMO

The complete sequence of retroperitoneal fibromatosis-associated herpesvirus Macaca nemestrina (RFHVMn), the pig-tailed macaque homolog of Kaposi's sarcoma-associated herpesvirus (KSHV), was determined by next-generation sequence analysis of a Kaposi's sarcoma (KS)-like macaque tumor. Colinearity of genes was observed with the KSHV genome, and the core herpesvirus genes had strong sequence homology to the corresponding KSHV genes. RFHVMn lacked homologs of open reading frame 11 (ORF11) and KSHV ORFs K5 and K6, which appear to have been generated by duplication of ORFs K3 and K4 after the divergence of KSHV and RFHV. RFHVMn contained positional homologs of all other unique KSHV genes, although some showed limited sequence similarity. RFHVMn contained a number of candidate microRNA genes. Although there was little sequence similarity with KSHV microRNAs, one candidate contained the same seed sequence as the positional homolog, kshv-miR-K12-10a, suggesting functional overlap. RNA transcript splicing was highly conserved between RFHVMn and KSHV, and strong sequence conservation was noted in specific promoters and putative origins of replication, predicting important functional similarities. Sequence comparisons indicated that RFHVMn and KSHV developed in long-term synchrony with the evolution of their hosts, and both viruses phylogenetically group within the RV1 lineage of Old World primate rhadinoviruses. RFHVMn is the closest homolog of KSHV to be completely sequenced and the first sequenced RV1 rhadinovirus homolog of KSHV from a nonhuman Old World primate. The strong genetic and sequence similarity between RFHVMn and KSHV, coupled with similarities in biology and pathology, demonstrate that RFHVMn infection in macaques offers an important and relevant model for the study of KSHV in humans.


Assuntos
Genoma Viral , Herpesvirus Humano 8/genética , Macaca nemestrina/virologia , Doenças dos Primatas/virologia , Sarcoma de Kaposi/veterinária , Sequência de Aminoácidos , Animais , Sequência de Bases , Feminino , Herpesvirus Humano 8/química , Herpesvirus Humano 8/classificação , Herpesvirus Humano 8/isolamento & purificação , Sequenciamento de Nucleotídeos em Larga Escala , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Rhadinovirus/química , Rhadinovirus/classificação , Rhadinovirus/genética , Sarcoma de Kaposi/virologia , Alinhamento de Sequência , Proteínas Virais/química , Proteínas Virais/genética
20.
J Virol ; 87(12): 6678-92, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23552412

RESUMO

Human immunodeficiency virus type 1 (HIV-1) antagonizes innate restriction factors in order to infect and persistently replicate in a host. In a previous study, we demonstrated that HIV-1 NL4-3 with a simian immunodeficiency virus mne (SIVmne) vif gene substitution (HSIV-vif-NL4-3) could infect and replicate in pig-tailed macaques (PTM), indicating that APOBEC3 proteins are primary barriers to transmission. Because viral replication was persistent but low, we hypothesized that HSIV-vif-NL4-3 may be suppressed by type I interferons (IFN-I), which are known to upregulate the expression of innate restriction factors. Here, we demonstrate that IFN-α more potently suppresses HSIV-vif-NL4-3 in PTM CD4(+) T cells than it does pathogenic SIVmne027. Importantly, we identify a variant (HSIV-vif-Yu2) that is resistant to IFN-α, indicating that the IFN-α-induced barrier can be overcome by HSIV-vif chimeras in PTM CD4(+) T cells. Interestingly, HSIV-vif-Yu2 and HSIV-vif-NL4-3 are similarly restricted by PTM BST2/Tetherin, and neither virus downregulates it from the surface of infected PTM CD4(+) T cells. Resistance to IFN-α-induced restriction appears to be conferred by a determinant in HSIV-vif-Yu2 that includes env su. Finally, we show that the Yu-2 env su allele may overcome an IFN-α-induced barrier to entry. Together, our data demonstrate that the prototype macaque-tropic HIV-1 clones based on NL4-3 may not sufficiently antagonize innate restriction in PTM cells. However, variants with resistance to IFN-α-induced restriction factors in PTM CD4(+) T cells may enhance viral replication by overcoming a barrier early in the viral replication cycle.


Assuntos
Linfócitos T CD4-Positivos/virologia , HIV-1/genética , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Interferon-alfa/imunologia , Macaca nemestrina/virologia , Animais , Contagem de Linfócito CD4 , Farmacorresistência Viral/imunologia , Células HEK293 , Células HeLa , Humanos , Interferon-alfa/farmacologia , Macaca nemestrina/imunologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...