Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Semin Immunol ; 51: 101472, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33648815

RESUMO

The relevance of monocyte and macrophage reservoirs in virally suppressed people with HIV (vsPWH) has previously been debatable. Macrophages were assumed to have a moderate life span and lack self-renewing potential. However, recent studies have challenged this dogma and now suggest an important role of these cell as long-lived HIV reservoirs. Lentiviruses have a long-documented association with macrophages and abundant evidence exists that macrophages are important target cells for HIV in vivo. A critical understanding of HIV infection, replication, and latency in macrophages is needed in order to determine the appropriate method of measuring and eliminating this cellular reservoir. This review provides a brief discussion of the biology and acute and chronic infection of monocytes and macrophages, with a more substantial focus on replication, latency and measurement of the reservoir in cells of myeloid origin.


Assuntos
Infecções por HIV , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Linfócitos T CD4-Positivos , Humanos , Macrófagos , Monócitos , Replicação Viral
2.
Curr HIV/AIDS Rep ; 21(2): 62-74, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38411842

RESUMO

PURPOSE OF REVIEW: HIV reservoirs are the main barrier to cure. CD4+ T cells have been extensively studied as the primary HIV-1 reservoir. However, there is substantial evidence that HIV-1-infected myeloid cells (monocytes/macrophages) also contribute to viral persistence and pathogenesis. RECENT FINDINGS: Recent studies in animal models and people with HIV-1 demonstrate that myeloid cells are cellular reservoirs of HIV-1. HIV-1 genomes and viral RNA have been reported in circulating monocytes and tissue-resident macrophages from the brain, urethra, gut, liver, and spleen. Importantly, viral outgrowth assays have quantified persistent infectious virus from monocyte-derived macrophages and tissue-resident macrophages. The myeloid cell compartment represents an important target of HIV-1 infection. While myeloid reservoirs may be more difficult to measure than CD4+ T cell reservoirs, they are long-lived, contribute to viral persistence, and, unless specifically targeted, will prevent an HIV-1 cure.


Assuntos
Infecções por HIV , Soropositividade para HIV , HIV-1 , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Humanos , Infecções por HIV/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Macrófagos , Linfócitos T CD4-Positivos , Latência Viral , Carga Viral
3.
Psychosom Med ; 84(8): 966-975, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36162063

RESUMO

OBJECTIVE: Simian immunodeficiency virus (SIV) infection of macaques recapitulates many aspects of HIV pathogenesis and is similarly affected by both genetic and environmental factors. Psychosocial stress is associated with immune system dysregulation and worse clinical outcomes in people with HIV. This study assessed the impact of single housing, as a model of psychosocial stress, on innate immune responses of pigtailed macaques ( Macaca nemestrina ) during acute SIV infection. METHODS: A retrospective analysis of acute SIV infection of 2- to si6-year-old male pigtailed macaques was performed to compare the innate immune responses of socially ( n = 41) and singly ( n = 35) housed animals. Measures included absolute monocyte count and subsets, and in a subset ( n ≤ 18) platelet counts and activation data. RESULTS: SIV infection resulted in the expected innate immune parameter changes with a modulating effect from housing condition. Monocyte number increased after infection for both groups, driven by classical monocytes (CD14 + CD16 - ), with a greater increase in socially housed animals (227%, p < .001, by day 14 compared with preinoculation time points). Platelet numbers recovered more quickly in the socially housed animals. Platelet activation (P-selectin) increased by 65% ( p = .004) and major histocompatibility complex class I surface expression by 40% ( p = .009) from preinoculation only in socially housed animals, whereas no change in these measures occurred in singly housed animals. CONCLUSIONS: Chronic psychosocial stress produced by single housing may play an immunomodulatory role in the innate immune response to acute retroviral infection. Dysregulated innate immunity could be one of the pathways by which psychosocial stress contributes to immune suppression and increased disease severity in people with HIV.


Assuntos
Infecções por HIV , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Habitação , Imunidade Inata , Macaca nemestrina , Masculino , Selectina-P/farmacologia , Estudos Retrospectivos , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/genética , Estresse Psicológico
4.
J Infect Dis ; 224(12): 2113-2121, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33970274

RESUMO

BACKGROUND: Although social distancing is a key public health response during viral pandemics, psychosocial stressors, such as social isolation, have been implicated in adverse health outcomes in general [1] and in the context of infectious disease, such as human immunodeficiency virus (HIV) [2, 3]. A comprehensive understanding of the direct pathophysiologic effects of psychosocial stress on viral pathogenesis is needed to provide strategic and comprehensive care to patients with viral infection. METHODS: To determine the effect of psychosocial stress on HIV pathogenesis during acute viral infection without sociobehavioral confounders inherent in human cohorts, we compared commonly measured parameters of HIV progression between singly (n = 35) and socially (n = 41) housed simian immunodeficiency virus (SIV)-infected pigtailed macaques (Macaca nemestrina). RESULTS: Singly housed macaques had a higher viral load in the plasma and cerebrospinal fluid and demonstrated greater CD4 T-cell declines and more CD4 and CD8 T-cell activation compared with socially housed macaques throughout acute SIV infection. CONCLUSIONS: These data demonstrate that psychosocial stress directly impacts the pathogenesis of acute SIV infection and imply that it may act as an integral variable in the progression of HIV infection and potentially of other viral infections.


Assuntos
Infecções por HIV , HIV/patogenicidade , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Estresse Psicológico , Animais , Linfócitos T CD4-Positivos/imunologia , Humanos , Ativação Linfocitária , Macaca nemestrina , Síndrome de Imunodeficiência Adquirida dos Símios/psicologia , Carga Viral
5.
J Neurochem ; 158(2): 500-521, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33899944

RESUMO

In healthy conditions, pannexin-1 (Panx-1) channels are in a close state, but in several pathological conditions, including human immunodeficiency virus-1 (HIV) and NeuroHIV, the channel becomes open. However, the mechanism or contribution of Panx-1 channels to the HIV pathogenesis and NeuroHIV is unknown. To determine the contribution of Panx-1 channels to the pathogenesis of NeuroHIV, we used a well-established model of simian immunodeficiency virus (SIV) infection in macaques (Macaca mulatta) in the presence of and absence of a Panx-1 blocker to later examine the synaptic/axonal compromise induced for the virus. Using Golgi's staining, we demonstrated that SIV infection compromised synaptic and axonal structures, especially in the white matter. Blocking Panx-1 channels after SIV infection prevented the synaptic and axonal compromise induced by the virus, especially by maintaining the more complex synapses. Our data demonstrated that targeting Panx-1 channels can prevent and maybe revert brain synaptic compromise induced by SIV infection.


Assuntos
Conexinas/metabolismo , Infecções por HIV/metabolismo , HIV-1 , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Sinapses/patologia , Animais , Axônios/patologia , Conexinas/antagonistas & inibidores , Espinhas Dendríticas/patologia , Substância Cinzenta/patologia , Humanos , Macaca mulatta , Proteínas do Tecido Nervoso/antagonistas & inibidores , Replicação Viral , Substância Branca/patologia
6.
J Immunol ; 202(1): 105-118, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30504423

RESUMO

An integral component of the antiviral response, type I IFNs require regulation to modulate immune activation. We identify ß-arrestin 2 as a key modulator of type I IFN in primary human macrophages, an essential component of the innate immune response. ß-Arrestin 2 was selectively activated by CCL2/CCR2 signaling, which induced a decrease in IFN-α, but not IFN-ß expression. Small interfering RNA knockdown of ß-arrestin 2 demonstrated its role in IFNAR1 internalization, as well as STAT1 and IRF3 activation. As a result, cytokine responses were not propagated following HIV infection and TLR3 activation. However, remnants of IFN signaling remained intact, despite ß-arrestin 2 activation, as IFN-ß, IFN-γ, IFN-λ1, IRF7, TRAIL, and MxA expression were sustained. Similar effects of ß-arrestin 2 on IFN signaling occurred in hepatocytes, suggesting that arrestins may broadly modulate IFN responses in multiple cell types. In summary, we identify a novel role of ß-arrestin 2 as an integral regulator of type I IFN through its internalization of IFNAR1 and a subsequent selective loss of downstream IFN signaling.


Assuntos
Infecções por HIV/imunologia , HIV-1/fisiologia , Hepatócitos/fisiologia , Interferon-alfa/metabolismo , Macrófagos/imunologia , Receptor de Interferon alfa e beta/metabolismo , beta-Arrestina 2/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Cultura Primária de Células , RNA Interferente Pequeno/genética , Receptores CCR2/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Ativação Transcricional , beta-Arrestina 2/genética
7.
J Virol ; 93(15)2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31118264

RESUMO

Understanding the cellular and anatomical sites of latent virus that contribute to human immunodeficiency virus (HIV) rebound is essential for eradication. In HIV-positive patients, CD4+ T lymphocytes comprise a well-defined functional latent reservoir, defined as cells containing transcriptionally silent genomes able to produce infectious virus once reactivated. However, the persistence of infectious latent virus in CD4+ T cells in compartments other than blood and lymph nodes is unclear. Macrophages (Mϕ) are infected by HIV/simian immunodeficiency virus (SIV) and are likely to carry latent viral genomes during antiretroviral therapy (ART), contributing to the reservoir. Currently, the gold standard assay used to measure reservoirs containing replication-competent virus is the quantitative viral outgrowth assay (QVOA). Using an SIV-macaque model, the CD4+ T cell and Mϕ functional latent reservoirs were measured in various tissues using cell-specific QVOAs. Our results showed that blood, spleen, and lung in the majority of suppressed animals contain latently infected Mϕs. Surprisingly, the numbers of CD4+ T cells, monocytes, and Mϕs carrying infectious genomes in blood and spleen were at comparable frequencies (∼1 infected cell per million). We also demonstrate that ex vivo viruses produced in the Mϕ QVOA are capable of infecting activated CD4+ T cells. These results strongly suggest that latently infected tissue Mϕs can reestablish productive infection upon treatment interruption. This study provides the first comparison of CD4+ T cell and Mϕ functional reservoirs in a macaque model. It is the first confirmation of the persistence of latent genomes in monocytes in blood and Mϕs in the spleen and lung of SIV-infected ART-suppressed macaques. Our results demonstrate that transcriptionally silent genomes in Mϕs can contribute to viral rebound after ART interruption and should be considered in future HIV cure strategies.IMPORTANCE This study suggests that CD4+ T cells found throughout tissues in the body can contain replication-competent SIV and contribute to rebound of the virus after treatment interruption. In addition, this study demonstrates that macrophages in tissues are another cellular reservoir for SIV and may contribute to viral rebound after treatment interruption. This new insight into the size and location of the SIV reservoir could have great implications for HIV-infected individuals and should be taken into consideration for the development of future HIV cure strategies.


Assuntos
Antirretrovirais/administração & dosagem , Linfócitos T CD4-Positivos/virologia , Macrófagos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Latência Viral , Animais , Células Sanguíneas/virologia , Células Cultivadas , Pulmão/virologia , Macaca , Vírus da Imunodeficiência Símia/isolamento & purificação , Baço/virologia
8.
Am J Pathol ; 188(1): 125-134, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29229308

RESUMO

A retrospective neuropathologic review of 30 SIV-infected pigtailed macaques receiving combination antiretroviral therapy (cART) was conducted. Seventeen animals with lymphocyte-dominant inflammation in the brain and/or meninges that clearly was morphologically distinct from prototypic SIV encephalitis and human immunodeficiency virus encephalitis were identified. Central nervous system (CNS) infiltrates in cART-treated macaques primarily comprised CD20+ B cells and CD3+ T cells with fewer CD68+ macrophages. Inflammation was associated with low levels of SIV RNA in the brain as shown by in situ hybridization, and generally was observed in animals with episodes of cerebrospinal fluid (CSF) viral rebound or sustained plasma and CSF viremia during treatment. Although the lymphocytic CNS inflammation in these macaques shared morphologic characteristics with uncommon immune-mediated neurologic disorders reported in treated HIV patients, including CNS immune reconstitution inflammatory syndrome and neurosymptomatic CSF escape, the high prevalence of CNS lesions in macaques suggests that persistent adaptive immune responses in the CNS also may develop in neuroasymptomatic or mildly impaired HIV patients yet remain unrecognized given the lack of access to CNS tissue for histopathologic evaluation. Continued investigation into the mechanisms and outcomes of CNS inflammation in cART-treated, SIV-infected macaques will advance our understanding of the consequences of residual CNS HIV replication in patients on cART, including the possible contribution of adaptive immune responses to HIV-associated neurocognitive disorders.


Assuntos
Antirretrovirais/uso terapêutico , Encéfalo/patologia , Encefalite/patologia , Linfócitos/patologia , Meningite/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Animais , Encefalite/complicações , Inflamação/patologia , Macaca nemestrina , Masculino , Meningite/complicações , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vírus da Imunodeficiência Símia , Carga Viral
9.
Curr Top Microbiol Immunol ; 417: 111-130, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29770863

RESUMO

Lentiviruses infect myeloid cells, leading to acute infection followed by persistent/latent infections not cleared by the host immune system. HIV and SIV are lentiviruses that infect CD4+ lymphocytes in addition to myeloid cells in blood and tissues. HIV infection of myeloid cells in brain, lung, and heart causes tissue-specific diseases that are mostly observed during severe immunosuppression, when the number of circulating CD4+ T cells declines to exceeding low levels. Antiretroviral therapy (ART) controls viral replication but does not successfully eliminate latent virus, which leads to viral rebound once ART is interrupted. HIV latency in CD4+ lymphocytes is the main focus of research and concern when HIV eradication efforts are considered. However, myeloid cells in tissues are long-lived and have not been routinely examined as a potential reservoir. Based on a quantitative viral outgrowth assay (QVOA) designed to evaluate latently infected CD4+ lymphocytes, a similar protocol was developed for the assessment of latently infected myeloid cells in blood and tissues. Using an SIV ART model, it was demonstrated that myeloid cells in blood and brain harbor latent SIV that can be reactivated and produce infectious virus in vitro, demonstrating that myeloid cells have the potential to be an additional latent reservoir of HIV that should be considered during HIV eradication strategies.


Assuntos
Sistema Nervoso Central/virologia , Modelos Animais de Doenças , Macaca mulatta/virologia , Macrófagos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Latência Viral , Animais , Linfócitos T CD4-Positivos/virologia , Infecções por HIV/virologia , Humanos , Carga Viral
10.
Curr HIV/AIDS Rep ; 16(1): 96-104, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30734905

RESUMO

PURPOSE OF REVIEW: In addition to preventive protocols and antiretroviral therapy, HIV-1 eradication has been considered as an additional strategy to help fight the AIDS epidemic. With the support of multiple funding agencies, research groups worldwide have been developing protocols to achieve either a sterilizing or a functional cure for HIV-infection. RECENT FINDINGS: Most of the studies focus on the elimination or suppression of circulating CD4+ T cells, the best characterized HIV-1 latent reservoir. The role of the central nervous system (CNS) as a latent reservoir is still controversial. Although brain macrophages and astrocytes are susceptible to HIV-1 infection, it has not been ascertained whether the CNS carries latent HIV-1 during cART and, if so, whether the virus can be reactivated and spread to other compartments after ART interruption. Here, we examine the implications of HIV-1 eradication strategies on the CNS, regardless of whether it is a true latent reservoir and, if so, whether it is present in all patients.


Assuntos
Encéfalo/virologia , Linfócitos T CD4-Positivos/virologia , Erradicação de Doenças/métodos , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , Latência Viral/fisiologia , Astrócitos/virologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Reservatórios de Doenças/virologia , HIV-1/efeitos dos fármacos , Humanos , Macrófagos/virologia , Latência Viral/efeitos dos fármacos
11.
Am J Pathol ; 187(1): 91-109, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27993242

RESUMO

Mounting evidence implicates antiretroviral (ARV) drugs as potential contributors to the persistence and evolution of clinical and pathological presentation of HIV-associated neurocognitive disorders in the post-ARV era. Based on their ability to induce endoplasmic reticulum (ER) stress in various cell types, we hypothesized that ARV-mediated ER stress in the central nervous system resulted in chronic dysregulation of the unfolded protein response and altered amyloid precursor protein (APP) processing. We used in vitro and in vivo models to show that HIV protease inhibitor (PI) class ARVs induced neuronal damage and ER stress, leading to PKR-like ER kinase-dependent phosphorylation of the eukaryotic translation initiation factor 2α and enhanced translation of ß-site APP cleaving enzyme-1 (BACE1). In addition, PIs induced ß-amyloid production, indicative of increased BACE1-mediated APP processing, in rodent neuroglial cultures and human APP-expressing Chinese hamster ovary cells. Inhibition of BACE1 activity protected against neuronal damage. Finally, ARVs administered to mice and SIV-infected macaques resulted in neuronal damage and BACE1 up-regulation in the central nervous system. These findings implicate a subset of PIs as potential mediators of neurodegeneration in HIV-associated neurocognitive disorders.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Inibidores da Protease de HIV/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Células Cultivadas , Macaca , Masculino , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Estabilidade Proteica/efeitos dos fármacos , Ratos , Ritonavir/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , eIF-2 Quinase/metabolismo
12.
J Neurovirol ; 24(2): 204-212, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28975505

RESUMO

Simian immunodeficiency virus (SIV) infection of pigtailed macaques is a highly representative and well-characterized animal model for HIV neuropathogenesis studies that provides an excellent opportunity to study and develop prognostic markers of HIV-associated neurocognitive disorders (HAND) for HIV-infected individuals. SIV studies can be performed in a controlled setting that enhances reproducibility and offers high-translational value. Similar to observations in HIV-infected patients receiving antiretroviral therapy (ART), ongoing neurodegeneration and inflammation are present in SIV-infected pigtailed macaques treated with suppressive ART. By developing quantitative viral outgrowth assays that measure both CD4+ T cells and macrophages harboring replication competent SIV as well as a highly sensitive mouse-based viral outgrowth assay, we have positioned the SIV/pigtailed macaque model to advance our understanding of latent cellular reservoirs, including potential CNS reservoirs, to promote HIV cure. In addition to contributing to our understanding of the pathogenesis of HAND, the SIV/pigtailed macaque model also provides an excellent opportunity to test innovative approaches to eliminate the latent HIV reservoir in the brain.


Assuntos
Antivirais/farmacologia , Sistema Nervoso Central/efeitos dos fármacos , Disfunção Cognitiva/tratamento farmacológico , Modelos Animais de Doenças , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Latência Viral/efeitos dos fármacos , Complexo AIDS Demência/tratamento farmacológico , Complexo AIDS Demência/imunologia , Complexo AIDS Demência/fisiopatologia , Complexo AIDS Demência/virologia , Animais , Terapia Antirretroviral de Alta Atividade , Sistema Nervoso Central/virologia , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/virologia , Humanos , Macaca nemestrina , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/fisiopatologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/patogenicidade , Vírus da Imunodeficiência Símia/fisiologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/virologia , Carga Viral/efeitos dos fármacos , Latência Viral/fisiologia
13.
J Virol ; 90(12): 5643-5656, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27030272

RESUMO

UNLABELLED: Despite the success of combined antiretroviral therapy (ART), human immunodeficiency virus (HIV) infection remains a lifelong infection because of latent viral reservoirs in infected patients. The contribution of CD4(+) T cells to infection and disease progression has been extensively studied. However, during early HIV infection, macrophages in brain and other tissues are infected and contribute to tissue-specific diseases, such as encephalitis and dementia in brain and pneumonia in lung. The extent of infection of monocytes and macrophages has not been rigorously assessed with assays comparable to those used to study infection of CD4(+) T cells and to evaluate the number of CD4(+) T cells that harbor infectious viral genomes. To assess the contribution of productively infected monocytes and macrophages to HIV- and simian immunodeficiency virus (SIV)-infected cells in vivo, we developed a quantitative virus outgrowth assay (QVOA) based on similar assays used to quantitate CD4(+) T cell latent reservoirs in HIV- and SIV-infected individuals in whom the infection is suppressed by ART. Myeloid cells expressing CD11b were serially diluted and cocultured with susceptible cells to amplify virus. T cell receptor ß RNA was measured as a control to assess the potential contribution of CD4(+) T cells in the assay. Virus production in the supernatant was quantitated by quantitative reverse transcription-PCR. Productively infected myeloid cells were detected in blood, bronchoalveolar lavage fluid, lungs, spleen, and brain, demonstrating that these cells persist throughout SIV infection and have the potential to contribute to the viral reservoir during ART. IMPORTANCE: Infection of CD4(+) T cells and their role as latent reservoirs have been rigorously assessed; however, the frequency of productively infected monocytes and macrophages in vivo has not been similarly studied. Myeloid cells, unlike lymphocytes, are resistant to the cytopathic effects of HIV. Moreover, tissue-resident macrophages have the ability to self-renew and persist in the body for months to years. Thus, tissue macrophages, once infected, have the characteristics of a potentially stable viral reservoir. A better understanding of the number of productively infected macrophages is crucial to further evaluate the role of infected myeloid cells as a potential viral reservoir. In the study described here we compared the frequency of productively infected CD4(+) T cells and macrophages in an SIV-infected macaque model. We developed a critical assay that will allow us to quantitate myeloid cells containing viral genomes that lead to productive infection in SIV-infected macaques and assess the role of macrophages as potential reservoirs.


Assuntos
Linfócitos T CD4-Positivos/virologia , Genoma Viral , Macrófagos/virologia , Monócitos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/isolamento & purificação , Carga Viral , Animais , Antígeno CD11b/análise , Modelos Animais de Doenças , Reservatórios de Doenças/virologia , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Infecções por HIV/virologia , Humanos , Macaca mulatta , Reação em Cadeia da Polimerase em Tempo Real , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Replicação Viral
14.
Am J Pathol ; 186(8): 2068-2087, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27322772

RESUMO

The effects of HIV infection on spleen and its cellular subsets have not been fully characterized, particularly for macrophages in which diverse populations exist. We used an accelerated SIV-infected macaque model to examine longitudinal effects on T-cell and macrophage populations and their susceptibilities to infection. Substantial lymphoid depletion occurred, characterized by follicular burn out and a loss of CD3 T lymphocytes, which was associated with cellular activation and transient dysregulations in CD4/CD8 ratios and memory effector populations. In contrast, the loss of CD68 and CD163(+)CD68(+) macrophages and increase in CD163 cells was irreversible, which began during acute infection and persisted until terminal disease. Mac387 macrophages and monocytes were transiently recruited into spleen, but were not sufficient to mitigate the changes in macrophage subsets. Type I interferon, M2 polarizing genes, and chemokine-chemokine receptor signaling were up-regulated in spleen and drove macrophage alterations. SIV-infected T cells were numerous within the white pulp during acute infection, but were rarely observed thereafter. CD68, CD163, and Mac387 macrophages were highly infected, which primarily occurred in the red pulp independent of T cells. Few macrophages underwent apoptosis, indicating that they are a long-lasting target for HIV/SIV. Our results identify macrophages as an important contributor to HIV/SIV infection in spleen and in promoting morphologic changes through the loss of specific macrophage subsets that mediate splenic organization.


Assuntos
Macrófagos/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Baço/imunologia , Baço/patologia , Linfócitos T/imunologia , Animais , Imuno-Histoquímica , Hibridização In Situ , Macaca nemestrina , Análise de Sequência com Séries de Oligonucleotídeos , Síndrome de Imunodeficiência Adquirida dos Símios/patologia
15.
J Infect Dis ; 214(10): 1520-1530, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27613775

RESUMO

BACKGROUND: Lung CD4+ T-cell depletion and dysfunction, CD8+ T-cell alveolitis, smoking, and poor control of human immunodeficiency virus (HIV) are features of HIV-associated chronic obstructive pulmonary disease (COPD), but these changes have not been evaluated in smokers at risk for COPD. We evaluated the impact of viral suppression following initiation of antiretroviral therapy (ART) on HIV-specific immunity and the balance of the CD4+ T-cell to CD8+ T-cell ratio in the lung. METHODS: Using flow cytometry, we assessed the T-cell immune response in lung and blood specimens obtained from 12 actively smoking HIV-positive patients before ART initiation and after ART-associated viral suppression. RESULTS: HIV suppression resulted in enhanced lung and systemic HIV-specific CD4+ T-cell immune responses without significant changes in CD8+ T-cell responses. We observed an increase in lung ratios of CD4+ T cells to CD8+ T cells and CD4+ T-cell frequencies, decreased CD8+ T-cell numbers, and resolution of CD8+ T-cell alveolitis after ART in 9 of 12 individuals. Viral suppression reduced Fas receptor and programmed death 1 expression in lung CD4+ T cells, correlating with enhanced effector function and reduced susceptibility to apoptosis. HIV suppression rescued peripheral but not lung HIV-specific CD4+ T-cell proliferation, resulting in augmented effector multifunction. DISCUSSION: Together, our results demonstrate that HIV suppression restores lung mucosal HIV-specific CD4+ T-cell multifunctional immunity and balance in the ratio of CD4+ T cells to CD8+ T cells, often resolving CD8+ T-cell alveolitis in active smokers. Peripheral expansion and redistribution of CD4+ T cells and increased resistance to apoptosis are 2 mechanisms contributing to immunologic improvement following viral suppression in patients at risk for HIV-associated COPD.


Assuntos
Antirretrovirais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/tratamento farmacológico , Pulmão/patologia , Doença Pulmonar Obstrutiva Crônica/prevenção & controle , Fumar/efeitos adversos , Adulto , Relação CD4-CD8 , Feminino , Citometria de Fluxo , Infecções por HIV/complicações , Humanos , Pulmão/imunologia , Masculino , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/patologia , Resultado do Tratamento
16.
J Infect Dis ; 212(9): 1387-96, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25883388

RESUMO

BACKGROUND: Sensitive assays are needed for detection of residual human immunodeficiency virus (HIV) in patients with undetectable plasma viral loads to determine whether eradication strategies are effective. The gold standard quantitative viral outgrowth assay (QVOA) underestimates the magnitude of the viral reservoir. We sought to determine whether xenograft of leukocytes from HIV type 1 (HIV)-infected patients with undetectable plasma viral loads into immunocompromised mice would result in viral amplification. METHODS: Peripheral blood mononuclear cells or purified CD4(+) T cells from HIV or simian immunodeficiency virus (SIV)-infected subjects with undetectable plasma viral loads were adoptively transferred into NOD.Cg-Prkdc(scid)Il2rg(tm1Wjl)/SzJ (NSG) mice. The mice were monitored for viremia following depletion of human CD8(+) T cells to minimize antiviral activity. In some cases, humanized mice were also treated with activating anti-CD3 antibody. RESULTS: With this murine viral outgrowth assay (MVOA), we successfully amplified replication-competent HIV or SIV from all subjects tested, including 5 HIV-positive patients receiving suppressive antiretroviral therapy (ART) and 6 elite controllers or suppressors who were maintaining undetectable viral loads without ART, including an elite suppressor from whom we were unable to recover virus by QVOA. CONCLUSIONS: Our results suggest that the MVOA has the potential to serve as a powerful tool to identify residual HIV in patients with undetectable viral loads.


Assuntos
Infecções por HIV/diagnóstico , HIV-1/isolamento & purificação , Carga Viral , Animais , Terapia Antirretroviral de Alta Atividade , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Modelos Animais de Doenças , Infecções por HIV/tratamento farmacológico , HIV-1/crescimento & desenvolvimento , Humanos , Interleucina-2/sangue , Leucócitos Mononucleares/virologia , Macaca , Masculino , Camundongos , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Vírus da Imunodeficiência Símia/isolamento & purificação , Viremia/veterinária
17.
J Immunol ; 191(8): 4280-7, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24043899

RESUMO

Expressed on tissue-resident macrophages, the receptor tyrosine kinase, recepteur d'orgine nantais (RON), functions to maintain inflammation homeostasis by activating genes that promote wound repair and resolve inflammation while repressing genes that perpetuate tissue damage and cell death. Chronic HIV-1 infection is associated with dysregulated inflammation, and we hypothesize that diminished RON expression contributes to the development of end organ diseases such as HIV-1-associated CNS disease. To explore RON function in vivo, we used CNS tissue from a well-characterized SIV macaque model and examined the temporal regulation of RON in the brain during the course of infection. Following prolonged SIV infection, RON expression was inversely correlated with the development of CNS disease; RON was maintained in animals that did not develop CNS lesions and was reduced in SIV-infected macaques that demonstrated moderate to severe inflammatory lesions. Arginase-1 expression was reduced in the brain during late infection, whereas expression of the inflammatory genes, IL-12p40 and TNF-α, was elevated. To validate a role for RON in regulating HIV-1 in primary cells, we used human tissue-resident macrophages isolated from tonsil as a tractable cell model. RON signaling in tissue-resident macrophages, both ligand dependent and independent, limited HIV-1 replication. Furthermore, prolonged HIV-1 infection in vitro resulted in downregulation of RON. We propose a model in which, following chronic HIV-1 infection in the brain, RON expression is decreased, genes that quell inflammation are repressed, and inflammatory mediators are induced to promote tissue inflammation.


Assuntos
Doenças do Sistema Nervoso Central/imunologia , Receptores Proteína Tirosina Quinases/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Animais , Arginase/biossíntese , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Doenças do Sistema Nervoso Central/virologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , Humanos , Inflamação , Subunidade p40 da Interleucina-12/biossíntese , Macaca nemestrina , Macrófagos/imunologia , Macrófagos/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores Proteína Tirosina Quinases/genética , Vírus da Imunodeficiência Símia/imunologia , Fator de Necrose Tumoral alfa/biossíntese
18.
Am J Respir Crit Care Med ; 190(7): 744-55, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25137293

RESUMO

RATIONALE: As overall survival improves, individuals with HIV infection become susceptible to other chronic diseases, including accelerated chronic obstructive pulmonary disease (COPD). OBJECTIVES: To determine whether individuals with HIV-associated COPD exhibit dysregulated lung mucosal T-cell immunity compared with control subjects. METHODS: Using flow cytometry, we evaluated peripheral blood and lung mucosal T-cell immunity in 14 HIV(+)COPD(+), 13 HIV(+)COPD(-), and 7 HIV(-)COPD(+) individuals. MEASUREMENTS AND MAIN RESULTS: HIV(+)COPD(+) individuals demonstrated profound CD4(+) T-cell depletion with reduced CD4/CD8 T-cell ratios in bronchoalveolar lavage-derived lung mononuclear cells, not observed in peripheral blood mononuclear cells, and diminished CD4(+) T cell absolute numbers, compared with control subjects. Furthermore, HIV(+)COPD(+) individuals demonstrated decreased pulmonary HIV-specific and staphylococcal enterotoxin B-reactive CD4(+) memory responses, including loss of multifunctionality, compared with HIV(+)COPD(-) control subjects. In contrast, lung mucosal HIV-specific CD8(+) T-cell responses were preserved. Lung CD4(+) T cells from HIV(+)COPD(+) individuals expressed increased surface Fas death receptor (CD95) and programmed death-1, but similar bronchoalveolar lavage viral loads as control subjects. However, programmed death-1 expression inversely correlated with HIV-specific lung CD4(+)IFN-γ(+) T-cell responses, suggesting functional exhaustion. Moreover, lung CD4(+) T cells from HIV(+)COPD(+) patients demonstrated increased basal and HIV antigen-induced expression of the early apoptosis marker annexin V compared with control subjects, which was significantly attenuated with anti-Fas blockade. Lastly, lung mucosal, but not blood, CD4(+)/CD8(+) ratios from HIV(+) patients significantly correlated with the FEV1, but not in HIV(-)COPD(+) patients. CONCLUSIONS: Together, our results provide evidence for profound lung mucosal CD4(+) T-cell depletion via a Fas-dependent activation-induced cell death mechanism, along with impaired HIV-specific CD4(+) immunity as immunologic features of HIV-associated COPD.


Assuntos
Apoptose/imunologia , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/imunologia , Pulmão/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Linfócitos T CD8-Positivos/imunologia , Morte Celular , Estudos de Coortes , Feminino , Citometria de Fluxo/métodos , Infecções por HIV/complicações , Humanos , Imunidade nas Mucosas/imunologia , Leucócitos Mononucleares/imunologia , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/complicações , Carga Viral/imunologia , Receptor fas/imunologia
19.
J Neurovirol ; 20(1): 39-53, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24420448

RESUMO

HIV-associated neurocognitive disorder (HAND), characterized by a wide spectrum of behavioral, cognitive, and motor dysfunctions, continues to affect approximately 50 % of HIV(+) patients despite the success of combination antiretroviral drug therapy (cART) in the periphery. Of note, potential toxicity of antiretroviral drugs in the central nervous system (CNS) remains remarkably underexplored and may contribute to the persistence of HAND in the cART era. Previous studies have shown antiretrovirals (ARVs) to be neurotoxic in the peripheral nervous system in vivo and in peripheral neurons in vitro. Alterations in lipid and protein metabolism, mitochondrial damage, and oxidative stress all play a role in peripheral ARV neurotoxicity. We hypothesized that ARVs also induce cellular stresses in the CNS, ultimately leading to neuronal damage and contributing to the changing clinical and pathological picture seen in HIV-positive patients in the cART era. In this report, we show that ARVs are neurotoxic in the CNS in both pigtail macaques and rats in vivo. Furthermore, in vitro, ARVs lead to accumulation of reactive oxygen species (ROS), and ultimately induction of neuronal damage and death. Whereas ARVs alone caused some activation of the endogenous antioxidant response in vitro, augmentation of this response by a fumaric acid ester, monomethyl fumarate (MMF), blocked ARV-induced ROS generation, and neuronal damage/death. These findings implicate oxidative stress as a contributor to the underlying mechanisms of ARV-induced neurotoxicity and will provide an access point for adjunctive therapies to complement ARV therapy and reduce neurotoxicity in this patient population.


Assuntos
Complexo AIDS Demência/patologia , Antirretrovirais/toxicidade , Encéfalo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Western Blotting , Encéfalo/patologia , Encéfalo/virologia , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Macaca , Masculino , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Immunol ; 188(8): 3876-85, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22407919

RESUMO

HIV-associated neurologic disorders are a mounting problem despite the advent of highly active antiretroviral therapy. To address mechanisms of HIV-associated neurologic disorders, we used an SIV pigtailed macaque model to study innate immune responses in brain that suppress viral replication during acute infection. We previously reported that during acute infection in brain, noncanonical type I IFN signaling occurs, where IFN-ß mRNA is induced while IFN-α is simultaneously suppressed. Two downstream IFN-stimulated genes, MxA and TRAIL, also show differential expression patterns. In this study, we show that differential signaling is due to interactions between macrophages and astrocytes. Astrocytes produce high levels of CCL2 upon SIV infection, which binds to CCR2 receptors on macrophages, leading to a selective suppression of IFN-α and the IFN-stimulated gene TRAIL while simultaneously inducing IFN-ß and MxA. The interactions between chemokine and cytokine pathways are a novel finding that may specifically occur in the CNS.


Assuntos
Astrócitos/imunologia , Macrófagos/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Astrócitos/metabolismo , Astrócitos/virologia , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/virologia , Comunicação Celular , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/imunologia , Regulação da Expressão Gênica , Imunidade Inata , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Macaca mulatta , Macrófagos/metabolismo , Macrófagos/virologia , Proteínas de Resistência a Myxovirus , Cultura Primária de Células , RNA Mensageiro/biossíntese , RNA Mensageiro/imunologia , Receptores CCR2/genética , Receptores CCR2/imunologia , Transdução de Sinais , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA