Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Virol ; 96(4): e0195321, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34878918

RESUMO

While combination antiretroviral therapy maintains undetectable viremia in people living with HIV (PLWH), a lifelong treatment is necessary to prevent viremic rebound after therapy cessation. This rebound seemed mainly caused by long-lived HIV-1 latently infected cells reverting to a viral productive status. Reversing latency and elimination of these cells by the so-called shock-and-kill strategy is one of the main investigated leads to achieve an HIV-1 cure. Small molecules referred to as latency reversal agents (LRAs) proved to efficiently reactivate latent CD4+ T cells. However, the LRA impact on de novo infection or HIV-1 production in productively infected macrophages remains elusive. Nontoxic doses of bryostatin-1, JQ1, and romidepsin were investigated in human monocyte-derived macrophages (MDMs). Treatment with bryostatin-1 or romidepsin resulted in a downregulation of CD4 and CCR5 receptors, respectively, accompanied by a reduction of R5 tropic virus infection. HIV-1 replication was mainly regulated by receptor modulation for bryostatin-1, while romidepsin effects rely on upregulation of SAMHD1 activity. LRA stimulation of chronically infected cells did not enhance HIV-1 production or gene expression. Surprisingly, bryostatin-1 caused a major decrease in viral production. This effect was not viral strain specific but appears to occur only in myeloid cells. Bryostatin-1 treatment of infected MDMs led to decreased amounts of capsid and matrix mature proteins with little to no modulation of precursors. Our observations revealed that bryostatin-1-treated myeloid and CD4+ T cells respond differently upon HIV-1 infection. Therefore, additional studies are warranted to more fully assess the efficiency of HIV-1 eradicating strategies. IMPORTANCE HIV-1 persists in a cellular latent form despite therapy that quickly propagates infection upon treatment interruption. Reversing latency would contribute to eradicate these cells, closing the gap to a cure. Macrophages are an acknowledged HIV-1 reservoir during therapy and are suspected to harbor latency establishment in vivo. However, the impact of latency reversal agents (LRAs) on HIV-1 infection and viral production in human macrophages is poorly known but nonetheless crucial to probe the safety of this strategy. In this in vitro study, we discovered encouraging antireplicative features of distinct LRAs in human macrophages. We also described a new viral production inhibition mechanism by protein kinase C agonists that is specific to myeloid cells. This study provides new insights into HIV-1 propagation restriction potentials by LRAs in human macrophages and underline the importance of assessing latency reversal strategy on all HIV-1-targeted cells.


Assuntos
Fármacos Anti-HIV/farmacologia , Briostatinas/farmacologia , HIV-1/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Antígenos CD4/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Depsipeptídeos/farmacologia , Diterpenos/farmacologia , Proteína do Núcleo p24 do HIV/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/virologia , Receptores CCR5/metabolismo , Proteína 1 com Domínio SAM e Domínio HD/metabolismo , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
2.
J Virol ; 95(20): e0118821, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34379507

RESUMO

Several host factors influence HIV-1 infection and replication. The p53-mediated antiviral role in monocyte-derived macrophages (MDMs) was previously highlighted. Indeed, an increase in p53 level results in a stronger restriction against HIV-1 early replication steps through SAMHD1 activity. In this study, we investigated the potential role of some p53 isoforms in HIV-1 infection. Transfection of isoform-specific small interfering RNA (siRNA) induced distinctive effects on the virus life cycle. For example, in contrast to an siRNA targeting all isoforms, a knockdown of Δ133p53 transcripts reduced virus replication in MDMs that was correlated with a decrease in phosphorylated inactive SAMHD1. Combination of Δ133p53 knockdown and nutlin-3, a pharmacological inhibitor of MDM2 that stabilizes p53, further reduced susceptibility of MDMs to HIV-1 infection, thus suggesting an inhibitory role of Δ133p53 toward p53 antiviral activity. In contrast, p53ß knockdown in MDMs increased the viral production independently of SAMHD1. Moreover, experiments with a Nef-deficient virus showed that this viral protein plays a protective role against the antiviral environment mediated by p53. Finally, HIV-1 infection affected the expression pattern of p53 isoforms by increasing p53ß and p53γ mRNA levels while stabilizing the protein level of p53α and some isoforms from the p53ß subclass. The balance between the various p53 isoforms is therefore an important factor in the overall susceptibility of macrophages to HIV-1 infection, fine-tuning the p53 response against HIV-1. This study brings a new understanding of the complex role of p53 in virus replication processes in myeloid cells. IMPORTANCE As of today, HIV-1 infection is still considered a global pandemic without a functional cure, partly because of the presence of stable viral reservoirs. Macrophages constitute one of these cell reservoirs, contributing to the viral persistence. Studies investigating the host factors involved in cell susceptibility to HIV-1 infection might lead to a better understanding of reservoir formation and will eventually allow the development of an efficient cure. Our team previously showed the antiviral role of p53 in macrophages, which acts by compromising the early steps of HIV-1 replication. In this study, we demonstrate the involvement of p53 isoforms, which regulate p53 activity and define the cellular environment influencing viral replication. In addition, the results concerning the potential role of p53 in antiviral innate immunity could be transposed to other fields of virology and suggest that knowledge in oncology can be applied to HIV-1 research.


Assuntos
Infecções por HIV/metabolismo , Macrófagos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/metabolismo , HIV-1/patogenicidade , HIV-1/fisiologia , Humanos , Imunidade Inata/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Macrófagos/virologia , Isoformas de Proteínas/genética , Proteína Supressora de Tumor p53/genética , Ligação Viral , Replicação Viral
3.
Glia ; 69(2): 255-280, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32910482

RESUMO

Human immunodeficiency virus type-1 (HIV-1) causes a spectrum of neurological impairments, termed HIV-associated neurocognitive disorder (HAND), following the infiltration of infected cells into the brain. Even though the implementation of antiretroviral therapy reduced the systemic viral load, the prevalence of HAND remains unchanged and infected patients develop persisting neurological disturbances affecting their quality of life. As a result, HAND have gained importance in basic and clinical researches, warranting the need of developing new adjunctive treatments. Nonetheless, a better understanding of the molecular and cellular mechanisms remains necessary. Several studies consolidated their efforts into elucidating the neurotoxic signaling leading to HAND including the deleterious actions of HIV-1 viral proteins and inflammatory mediators. However, the scope of these studies is not sufficient to address all the complexity related to HAND development. Fewer studies focused on an altered neuroprotective capacity of the brain to respond to HIV-1 infection. Neurotrophic factors are endogenous polyproteins involved in neuronal survival, synaptic plasticity, and neurogenesis. Any defects in the processing or production of these crucial factors might compose a risk factor rendering the brain more vulnerable to neuronal damages. Due to their essential roles, they have been investigated for their diverse interplays with HIV-1 infection. In this review, we present a complete description of the neurotrophic factors involved in HAND. We discuss emerging concepts for their therapeutic applications and summarize the complex mechanisms that down-regulate their production in favor of a neurotoxic environment. For certain factors, we finally address opposing roles that rather lead to increased inflammation.


Assuntos
Infecções por HIV , HIV-1 , Complexo AIDS Demência , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Humanos , Fatores de Crescimento Neural , Neuroproteção , Qualidade de Vida
4.
J Neurovirol ; 27(2): 279-301, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33646495

RESUMO

HIV-1 infection in the central nervous system (CNS) causes the release of neurotoxic products from infected cells which trigger extensive neuronal loss. Clinically, this results in HIV-1-associated neurocognitive disorders (HAND). However, the effects on neuroprotective factors in the brain remain poorly understood and understudied in this situation. HAND is a multifactorial process involving several players, and the complex cellular mechanisms have not been fully elucidated yet. In this study, we reported that HIV-1 infection of astrocytes limits their potential to express the protective chemokine fractalkine in response to an inflammatory environment. We next confirmed that this effect was not due to a default in its shedding from the cell surface. We then investigated the biological mechanism responsible for this reduced fractalkine expression and found that HIV-1 infection specifically blocks the interaction of transcription factor NF-κB on its promoter with no effect on other cytokines. Moreover, we demonstrated that fractalkine production in astrocytes is regulated in response to immune factors secreted by infected/activated microglia and macrophages. In contrast, we observed that conditioned media from these infected cells also trigger neuronal apoptosis. At last, we demonstrated a strong neuroprotective action of fractalkine on human neurons by reducing neuronal damages. Taken together, our results indicate new relevant interactions between HIV-1 and fractalkine signaling in the CNS. This study provides new information to broaden the understanding of HAND and possibly foresee new therapeutic strategies. Considering its neuro-protective functions, reducing its production from astrocytes could have important outcomes in chronic neuroinflammation and in HIV-1 neuropathogenesis.


Assuntos
Complexo AIDS Demência/metabolismo , Astrócitos/virologia , Quimiocina CX3CL1/biossíntese , Astrócitos/imunologia , Astrócitos/metabolismo , Células Cultivadas , HIV-1 , Humanos
5.
Glia ; 68(11): 2212-2227, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32250524

RESUMO

Since the introduction of the combined antiretroviral therapy, HIV-1 infection has become a manageable chronic disease in which patients display a life expectancy almost identical to the general population. Nevertheless, various age-related pathologies such as neurocognitive disorders have emerged as serious complications. A "shock and kill" strategy using latency-reversing agents (LRA) to reactivate HIV-1 has been proposed to eliminate the viral reservoir in such chronically infected patients. However, the impact of LRA on the central nervous system remains elusive. Given that an increased amyloid beta (Aß) deposition is a feature of HIV-1-infected brains, we investigated the consequences of HIV-1 infection and treatment with two LRA (bryostatin-1 and JQ1) on the capacity of human astrocytes to engulf and clear Aß. We show here that HIV-1-infected astrocytes accumulate a very high amount of Aß compared to uninfected cells, but the engulfed peptide in degraded very slowly. The LRA bryostatin-1 induces a reduction in Aß endocytosis, whereas JQ1 treatment results in a very slow degradation of the ingested material associated with a reduced expression of the endopeptidase neprilysin. An exposure to JQ1 also induces a sustained release of Aß-loaded microvesicles. Thus, both HIV-1 infection and treatment with some LRA could contribute to the reported Aß accumulation in the brain of HIV-1-infected persons.


Assuntos
Infecções por HIV , HIV-1 , Peptídeos beta-Amiloides , Astrócitos , Azepinas , Briostatinas/farmacologia , Infecções por HIV/tratamento farmacológico , Homeostase , Humanos , Triazóis , Ativação Viral , Latência Viral
6.
J Immunol ; 198(3): 1229-1241, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27994072

RESUMO

A shock-and-kill approach involving the simultaneous treatment of HIV-1-infected patients with latency-reversing agents (LRAs) and combination antiretroviral therapy was proposed as a means to eradicate viral reservoirs. Currently available LRAs cannot discriminate between HIV-1-infected and uninfected cells. Therefore, the risks and benefits of using broad-spectrum LRAs need to be carefully evaluated, particularly in the CNS, where inflammation and leukocyte transmigration must be tightly regulated. We used a real-time impedance-sensing system to dynamically record the impact of different classes of LRAs on the integrity of tight monolayers of the immortalized human cerebral microvascular endothelial cell line hCMEC/D3. Results show that prostratin and bryostatin-1 can significantly damage the integrity of an endothelial monolayer. Moreover, prostratin and bryostatin-1 induce secretion of some proinflammatory cytokines and an increase of ICAM-1 expression. Additional studies demonstrated that prostratin and bryostatin-1 also affect adhesion and transmigration of CD4+ and CD8+ T cells as well as monocytes in an in vitro human blood-brain barrier (BBB) model. Prostratin and bryostatin-1 could thus be considered as potent regulators of BBB permeability and inflammation that influence leukocyte transport across the BBB. Altogether, these findings contribute to a better understanding of the potential risks and benefits of using a shock-and-kill approach with LRAs on the normal physiological functions of the BBB.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Briostatinas/farmacologia , HIV-1/fisiologia , Leucócitos/efeitos dos fármacos , Ésteres de Forbol/farmacologia , Latência Viral/efeitos dos fármacos , Acetamidas/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Azepinas/farmacologia , Briostatinas/efeitos adversos , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/análise , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/fisiologia , Citocinas/metabolismo , Decitabina , Humanos , Inflamação/induzido quimicamente , Molécula 1 de Adesão Intercelular/análise , Leucócitos/fisiologia , Ésteres de Forbol/efeitos adversos , Quinazolinas/farmacologia , Receptores de Superfície Celular/análise
7.
Glia ; 66(7): 1363-1381, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29464785

RESUMO

The "shock and kill" HIV-1 cure strategy proposes eradication of stable cellular reservoirs by clinical treatment with latency-reversing agents (LRAs). Although resting CD4+ T cells latently infected with HIV-1 constitute the main reservoir that is targeted by these approaches, their consequences on other reservoirs such as the central nervous system are still unknown and should be taken into consideration. We performed experiments aimed at defining the possible role of astrocytes in HIV-1 persistence in the brain and the effect of LRA treatments on this viral sanctuary. We first demonstrate that the diminished HIV-1 production in a proliferating astrocyte culture is due to a reduced proliferative capacity of virus-infected cells compared with uninfected astrocytes. In contrast, infection of non-proliferating astrocytes led to a robust HIV-1 infection that was sustained for over 60 days. To identify astrocytes latently infected with HIV-1, we designed a new dual-color reporter virus called NL4.3 eGFP-IRES-Crimson that is fully infectious and encodes for all viral proteins. Although we detected a small fraction of astrocytes carrying silent HIV-1 proviruses, we did not observe any reactivation using various LRAs and even strong inducers such as tumor necrosis factor, thus suggesting that these proviruses were either not transcriptionally competent or in a state of deep latency. Our findings imply that astrocytes might not constitute a latent reservoir per se but that relentless virus production by this brain cell population could contribute to the neurological disorders seen in HIV-1-infected persons subjected to combination antiretroviral therapy.


Assuntos
Astrócitos/fisiologia , Astrócitos/virologia , Infecções por HIV/fisiopatologia , HIV-1/fisiologia , Astrócitos/patologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD4-Positivos/virologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Células HEK293 , HIV-1/genética , Humanos , Latência Viral
8.
J Virol ; 91(16)2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28539453

RESUMO

In this study, we investigated the effect of acetate, the most concentrated short-chain fatty acid (SCFA) in the gut and bloodstream, on the susceptibility of primary human CD4+ T cells to HIV-1 infection. We report that HIV-1 replication is increased in CD3/CD28-costimulated CD4+ T cells upon acetate treatment. This enhancing effect correlates with increased expression of the early activation marker CD69 and impaired class I/II histone deacetylase (HDAC) activity. In addition, acetate enhances acetylation of histones H3 and H4 and augments HIV-1 integration into the genome of CD4+ T cells. Thus, we propose that upon antigen presentation, acetate influences class I/II HDAC activity that transforms condensed chromatin into a more relaxed structure. This event leads to a higher level of viral integration and enhanced HIV-1 production. In line with previous studies showing reactivation of latent HIV-1 by SCFAs, we provide evidence that acetate can also increase the susceptibility of primary human CD4+ T cells to productive HIV-1 infection.IMPORTANCE Alterations in the fecal microbiota and intestinal epithelial damage involved in the gastrointestinal disorder associated with HIV-1 infection result in microbial translocation that leads to disease progression and virus-related comorbidities. Indeed, notably via production of short-chain fatty acids, bacteria migrating from the lumen to the intestinal mucosa could influence HIV-1 replication by epigenetic regulatory mechanisms, such as histone acetylation. We demonstrate that acetate enhances virus production in primary human CD4+ T cells. Moreover, we report that acetate impairs class I/II histone deacetylase activity and increases integration of HIV-1 DNA into the host genome. Therefore, it can be postulated that bacterial metabolites such as acetate modulate HIV-1-mediated disease progression.


Assuntos
Acetatos/metabolismo , Linfócitos T CD4-Positivos/virologia , HIV-1/fisiologia , Inibidores de Histona Desacetilases/metabolismo , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , Integração Viral , Acetilação , Antígenos CD/análise , Antígenos de Diferenciação de Linfócitos T/análise , Linfócitos T CD4-Positivos/química , Linfócitos T CD4-Positivos/efeitos dos fármacos , Células Cultivadas , Humanos , Lectinas Tipo C/análise , Replicação Viral
9.
Proc Natl Acad Sci U S A ; 112(26): E3392-401, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26056259

RESUMO

Target of Egr1 (TOE1) is a nuclear protein localized primarily in nucleoli and Cajal bodies that was identified as a downstream target of the immediate early gene Egr1. TOE1 displays a functional deadenylation domain and has been shown to participate in spliceosome assembly. We report here that TOE1 can function as an inhibitor of HIV-1 replication and show evidence that supports a direct interaction of TOE1 with the viral specific transactivator response element as part of the inhibitory mechanism. In addition, we show that TOE1 can be secreted by activated CD8(+) T lymphocytes and can be cleaved by the serine protease granzyme B, one of the main components of cytotoxic granules. Both full-length and cleaved TOE1 can spontaneously cross the plasma membrane and penetrate cells in culture, retaining HIV-1 inhibitory activity. Antiviral potency of TOE1 and its cell-penetrating capability have been identified to lie within a 35-amino-acid region containing the nuclear localization sequence.


Assuntos
HIV-1/fisiologia , Fusão de Membrana/fisiologia , Proteínas Nucleares/fisiologia , Replicação Viral/fisiologia , Células HEK293 , Repetição Terminal Longa de HIV/genética , HIV-1/genética , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteólise , Ativação Transcricional
10.
J Neuroinflammation ; 14(1): 242, 2017 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-29228979

RESUMO

BACKGROUND: Despite effectiveness of the combined antiretroviral therapy, HIV-1 persists in long-lived latently infected cells. Consequently, new therapeutic approaches aimed at eliminating this latent reservoir are currently being developed. A "shock and kill" strategy using latency-reversing agents (LRA) to reactivate HIV-1 has been proposed. However, the impact of LRA on the central nervous system (CNS) remains elusive. METHODS: We used human fetal astrocytes and investigated the effects of several LRA on their functional and secretory activities. Astrocytes were infected with VSV-G-pseudotyped HIV-1 before treatment with various blood-brain barrier (BBB)-permeable LRA at subcytotoxic doses, which allow HIV-1 reactivation based on previous in vitro and clinical studies. Cells and supernatants were then used to evaluate effects of infection and LRA on (i) viability and metabolic activity of astrocytes using a colorimetric MTS assay; (ii) chemokines and proinflammatory cytokines secretion and gene expression by astrocytes using ELISA and RT-qPCR, respectively; (iii) expression of complement component 3 (C3), a proxy for astrogliosis, by RT-qPCR; (iv) glutamate uptake capacity by a fluorometric assay; and (v) modulation of neutrophil transmigration across an in vitro BBB model. RESULTS: We demonstrate that bryostatin-1 induces secretion of chemokines CCL2 and IL-8 and proinflammatory cytokines IL-6 and GM-CSF, whereas their production is repressed by JQ1. Bryostatin-1 also increases expression of complement component 3 and perturbs astrocyte glutamate homeostasis. Lastly, bryostatin-1 enhances transmigration of neutrophils across an in vitro blood-brain barrier model and induces formation of neutrophil extracellular traps. CONCLUSIONS: These observations highlight the need to carefully assess the potential harmful effect to the CNS when selecting LRA for HIV-1 reactivation strategies.


Assuntos
Adjuvantes Imunológicos/toxicidade , Astrócitos/efeitos dos fármacos , Azepinas/toxicidade , Encéfalo/efeitos dos fármacos , Briostatinas/toxicidade , Quimiotaxia de Leucócito/efeitos dos fármacos , Triazóis/toxicidade , Encéfalo/patologia , HIV-1/fisiologia , Humanos , Inflamação/patologia , Neutrófilos/efeitos dos fármacos , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos
11.
Immun Inflamm Dis ; 11(1): e590, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36480653

RESUMO

INTRODUCTION: HIV-1 eradication is hindered by the presence of inducible long-lived reservoirs of latently infected cells which rapidly disseminate viral particles upon treatment interruption. Eliminating these reservoirs by the so-called shock and kill strategy represents a crucial concept toward an HIV-1 cure. Several molecules called latency-reversing agents (LRAs) are under intensive investigations to reactivate virus gene expression. These studies are mainly conducted on CD4+ T cells where LRAs are well tolerated and did not induce global cellular activation. However, despite their broad spectrum, the putative impact of LRAs on other cellular reservoirs such as macrophages is still ill-defined. METHODS: We investigated the impact of the protein kinase C (PKC) activator bryostatin-1, bromodomain inhibitor JQ1 and histone deacetylase inhibitor romidepsin used either alone or in combination on human primary monocyte-derived macrophages (MDMs). RESULTS: We demonstrate that bryostatin-1, JQ1, and romidepsin or their combinations are not toxic at nanomolar concentrations but induce metabolic and morphologic alterations of MDMs. Bryostatin-1 triggered the secretion of pro-inflammatory cytokines, while JQ-1 decreased it. Phagocytosis and endocytosis were modestly impaired upon bryostatin-1 treatment whereas efferocytosis was markedly downregulated by romidepsin. Despite its pro-inflammatory profile, bryostatin-1 did not induce classically activated macrophage markers. Finally, we reveal that conditioned medium from bryostatin-1-treated macrophages did not potentiate its reactivation feature. CONCLUSIONS: Our study reveals that LRAs can diversely impact basic physiologic features of human primary macrophages and could potentially decrease reactivation of nearby CD4+ T cells latently infected with HIV-1. Our observations further stress the need to include different cell populations when assessing HIV-1 cure strategies.


Assuntos
Infecções por HIV , Soropositividade para HIV , Humanos , Ativação Viral , Latência Viral , Briostatinas/farmacologia , Briostatinas/uso terapêutico , Linfócitos T CD4-Positivos , Infecções por HIV/tratamento farmacológico , Macrófagos
12.
Retrovirology ; 9: 4, 2012 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-22236409

RESUMO

In HIV-1-infected individuals, there is often discordance between viremia in peripheral blood and viral load found in the central nervous system (CNS). Although the viral burden is often lower in the CNS compartment than in the plasma, neuroinflammation is present in most infected individuals, albeit attenuated by the current combined antiretroviral therapy. The HIV-1-associated neurological complications are thought to result not only from direct viral replication, but also from the subsequent neuroinflammatory processes. The eicosanoids - prostanoids and leukotrienes - are known as potent inflammatory lipid mediators. They are often present in neuroinflammatory diseases, notably HIV-1 infection. Their exact modulatory role in HIV-1 infection is, however, still poorly understood, especially in the CNS compartment. Nonetheless, a handful of studies have provided evidence as to how these lipid mediators can modulate HIV-1 infection. This review summarizes findings indicating how eicosanoids may influence the progression of neuroAIDS.


Assuntos
Infecções por HIV/imunologia , Infecções por HIV/patologia , HIV-1/imunologia , HIV-1/patogenicidade , Mediadores da Inflamação/metabolismo , Leucotrienos/metabolismo , Prostaglandinas/metabolismo , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Humanos
13.
J Neuroinflammation ; 9: 55, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22424294

RESUMO

BACKGROUND: Microglia are one of the main cell types to be productively infected by HIV-1 in the central nervous system (CNS). Leukotriene B4 (LTB4) and cysteinyl-leukotrienes such as LTC4 are some of the proinflammatory molecules produced in infected individuals that contribute to neuroinflammation. We therefore sought to investigate the role of leukotrienes (LTs) in HIV-1 infection of microglial cells. METHODS: To evaluate the role of LTs on HIV-1 infection in the CNS, monocyte-derived microglial-like cells (MDMis) were utilized in this study. Leukotriene-treated MDMis were infected with either fully replicative brain-derived HIV-1 isolates (YU2) or R5-tropic luciferase-encoding particles in order to assess viral production and expression. The efficacy of various steps of the replication cycle was evaluated by means of p24 quantification by ELISA, luciferase activity determination and quantitative real-time polymerase chain reaction (RT-PCR). RESULTS: We report in this study that virus replication is reduced upon treatment of MDMis with LTB4 and LTC4. Additional experiments indicate that these proinflammatory molecules alter the pH-independent entry and early post-fusion events of the viral life cycle. Indeed, LT treatment induced a diminution in integrated proviral DNA while reverse-transcribed viral products remained unaffected. Furthermore, decreased C-C chemokine receptor type 5 (CCR5) surface expression was observed in LT-treated MDMis. Finally, the effect of LTs on HIV-1 infection in MDMis appears to be mediated partly via a signal transduction pathway involving protein kinase C. CONCLUSIONS: These data show for the first time that LTs influence microglial cell infection by HIV-1, and may be a factor in the control of viral load in the CNS.


Assuntos
Regulação Viral da Expressão Gênica/efeitos dos fármacos , Infecções por HIV/virologia , Leucotrieno B4/farmacologia , Microglia/efeitos dos fármacos , Microglia/virologia , Encéfalo/citologia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Infecções por HIV/patologia , HIV-1/metabolismo , Humanos , Microglia/metabolismo , Substância P/metabolismo , Transfecção
14.
J Immunol ; 184(6): 2899-907, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20147629

RESUMO

Coinfection of HIV-1 patients with Plasmodium falciparum, the etiological agent of malaria, results in a raise of viral load and an acceleration of disease progression. The primary objective of this study was to investigate whether the malarial pigment hemozoin (HZ), a heme by-product of hemoglobin digestion by malaria parasites, can affect HIV-1 transmission by monocytes-derived dendritic cells (DCs) to CD4(+) T cells when HZ is initially internalized in monocytes before their differentiation in DCs. We demonstrate in this study that HZ treatment during the differentiation process induces an intermediate maturation phenotype when compared with immature and fully mature DCs. Furthermore, the DC-mediated transfer of HIV-1 is enhanced in presence of HZ, a phenomenon that may be linked with the capacity of HZ-loaded cells to interact and activate CD4(+) T cells. Altogether our findings suggest a new mechanism that could partially explain the increased HIV-1 virus production during a coinfection with P. falciparum. Understanding the multifaceted interactions between P. falciparum and HIV-1 is an important challenge that could lead to the development of new treatment strategies.


Assuntos
Linfócitos T CD4-Positivos/virologia , Diferenciação Celular/efeitos dos fármacos , HIV-1/efeitos dos fármacos , Hemeproteínas/farmacologia , Monócitos/virologia , Replicação Viral/efeitos dos fármacos , Antimaláricos/farmacologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/imunologia , Diferenciação Celular/imunologia , Linhagem Celular , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Células Dendríticas/virologia , HIV-1/imunologia , Humanos , Imunofenotipagem , Malária Falciparum/imunologia , Malária Falciparum/patologia , Malária Falciparum/virologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/patologia , Pigmentos Biológicos/farmacologia , Replicação Viral/imunologia
15.
Cell Microbiol ; 12(5): 615-25, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20082614

RESUMO

Together, Plasmodium falciparum (P. falciparum) and HIV-1 infections cause more than four million deaths a year. There is still limited information about the putative impact of the malaria pigment hemozoin (HZ) on the dissemination of HIV-1. As so, we propose a premise where HZ present in human dendritic cells (DCs) could modulate HIV-1 transfer to CD4(+) T cells. We report here that HZ promotes transmission of HIV-1 by immature monocyte-derived DCs (iMDDCs). Moreover, we noted that in the presence of HZ, iMDDCs were less permissive to productive HIV-1 infection. The HZ-dependent modulation of the interaction between iMDDCs and HIV-1 seems to be partly due to a decreased expression of CCR5 and also to the induction of a more mature phenotype as proven by microscopy and flow cytometry analyses. Therefore, exposure of iMDDCs to malaria pigments provokes their maturation rendering them more potent to trans-infect CD4(+) T cells with HIV-1.


Assuntos
Células Dendríticas/virologia , HIV-1/crescimento & desenvolvimento , Hemeproteínas/fisiologia , Monócitos/virologia , Plasmodium falciparum/patogenicidade , Fatores de Virulência/fisiologia , Animais , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Células Cultivadas , Células Dendríticas/parasitologia , Regulação para Baixo , Humanos , Monócitos/parasitologia , Receptores CCR5/biossíntese , Receptores de HIV/biossíntese
16.
Virology ; 561: 47-57, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34146963

RESUMO

Thymidylate synthase (TS) is a key enzyme in nucleotide biosynthesis. A study performed by our group on human monocyte-derived macrophages (MDMs) infected with HIV-1 showed that many enzymes related to the folate cycle pathway, such as TS, are upregulated in productively infected cells. Here, we suggest that TS is essential for an effective HIV-1 infection in MDMs. Indeed, a TS specific small interfering RNA (siRNA) as well as the TS specific inhibitor Raltitrexed (RTX) caused a reduction in productively infected cells. Quantitative PCR analysis showed that this treatment decreased the efficacy of the early steps of the viral cycle. The RTX inhibitory effect was counteracted by dNTP addition. These results suggest that TS is essential for the early stages of HIV-1 infection by providing optimal dNTP concentrations in MDMs. TS and its related pathway may thus be considered as a potential therapeutic target for HIV-1 treatment.


Assuntos
HIV-1/fisiologia , Macrófagos/enzimologia , Macrófagos/virologia , Timidilato Sintase/metabolismo , Replicação Viral , Células Cultivadas , Inibidores Enzimáticos , Humanos , Quinazolinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Proteína 1 com Domínio SAM e Domínio HD/metabolismo , Tiofenos/farmacologia , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/genética , Nucleotídeos de Timina/metabolismo , Replicação Viral/efeitos dos fármacos
17.
Tissue Eng Part C Methods ; 27(3): 152-166, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33573474

RESUMO

The lack of appropriate experimental models often limits our ability to investigate the establishment of infections in specific tissues. To reproduce the structural and spatial organization of vaginal mucosae to study human immunodeficiency virus type-1 (HIV-1) infection, we used the self-assembly technique to bioengineer tridimensional vaginal mucosae using human cells extracted from HIV-1-negative healthy pre- and postmenopausal donors. We produced a stroma, free of exogenous material, that can be adapted to generate near-to-native vaginal tissue with the best complexity obtained with seeded epithelial cells on the organ-specific stroma. The autologous engineered tissues had mechanical properties close to native mucosa and shared similar glycogen production, which declined in reconstructed tissues of the postmenopausal donor. The in vitro-engineered tissues were also rendered immune competent by adding human monocyte-derived macrophages (MDMs) on the epithelium or in the stroma layers. The model was infected with HIV-1, and viral replication and transcytosis were observed when immunocompetent reconstructed vaginal mucosa tissue has incorporated MDMs into the stroma and infected with free HIV-1 green fluorescent protein (GFP) viral particles. These data illustrate a natural permissiveness of immunocompetent untransformed human vaginal mucosae to HIV-1 infection. This model offers a physiological tool to explore viral load, HIV-1 transmission in an environment that may contribute to the virus propagation, and new antiviral treatments in vitro. Impact statement This study introduces an innovative immunocompetent three-dimensional human organ-specific vaginal mucosa free of exogenous material for in vitro modeling of human immunodeficiency virus type-1 (HIV-1) infection. The proposed model is histologically close to native tissue, especially by presenting glycogen accumulation in the epithelium's superficial cells, responsive to estrogen, and able to sustain a monocyte-derived macrophage population infected or not by HIV-1 during ∼2 months.


Assuntos
Infecções por HIV , HIV-1 , Feminino , Hormônios , Humanos , Mucosa , Vagina
18.
J Virol ; 83(6): 2778-82, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19109380

RESUMO

Dendritic cells (DC) are considered to be important contributors to human immunodeficiency virus type 1 (HIV-1) transmission and pathogenesis. As the first target cells in mucosal tissues, they can be become productively infected and can also capture virions and transfer them efficiently to CD4(+) T cells located within lymphoid tissues. Resting CD4(+) T cells appear to be another major target of HIV-1 in vivo, yet several blocks restrict replication in such cells. We report here that physical contact between virus-infected quiescent CD4(+) T cells and uninfected autologous immature DC in the absence of any foreign antigen relieves these restrictions, allowing a highly productive HIV-1 replication.


Assuntos
Linfócitos T CD4-Positivos/virologia , Células Dendríticas/imunologia , HIV-1/imunologia , HIV-1/fisiologia , Replicação Viral , Técnicas de Cocultura , Citometria de Fluxo , Genes Reporter , Proteína do Núcleo p24 do HIV/biossíntese , Humanos , Luciferases/metabolismo , RNA Viral/biossíntese
19.
Nucleic Acids Res ; 36(7): 2353-65, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18299284

RESUMO

The interaction between human immunodeficiency virus type 1 (HIV-1) and RNA silencing pathways is complex and multifaceted. Essential for efficient viral transcription and supporting Tat-mediated transactivation of viral gene expression, the trans-activation responsive (TAR) element is a structured RNA located at the 5' end of all transcripts derived from HIV-1. Here, we report that this element is a source of microRNAs (miRNAs) in cultured HIV-1-infected cell lines and in HIV-1-infected human CD4+ T lymphocytes. Using primer extension and ribonuclease (RNase) protection assays, we delineated both strands of the TAR miRNA duplex deriving from a model HIV-1 transcript, namely miR-TAR-5p and miR-TAR-3p. In vitro RNase assays indicate that the lack of a free 3' extremity at the base of TAR may contribute to its low processing reactivity in vivo. Both miR-TAR-5p and miR-TAR-3p down-regulated TAR miRNA sensor activity in a process that required an integral miRNA-guided RNA silencing machinery. miR-TAR-3p exerted superior gene downregulatory effects, probably due to its preferential release from HIV-1 TAR RNA by the RNase III Dicer. Our study suggests that the TAR element of HIV-1 transcripts releases functionally competent miRNAs upon asymmetrical processing by Dicer, thereby providing novel insights into viral miRNA biogenesis.


Assuntos
Regulação Viral da Expressão Gênica , Repetição Terminal Longa de HIV , HIV-1/genética , MicroRNAs/metabolismo , Processamento Pós-Transcricional do RNA , Animais , Sequência de Bases , Linhagem Celular , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , RNA Viral/química , RNA Viral/metabolismo , Ribonuclease III/metabolismo
20.
Methods Mol Biol ; 487: 415-33, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19301659

RESUMO

Recent experimental evidences support the existence of an increasingly complex and multifaceted interaction between viruses and the microRNA-guided RNA silencing machinery of human cells. The discovery of small interfering RNAs (siRNAs), which are designed to mediate cleavage of specific messenger RNAs (mRNAs), prompted virologists to establish therapeutic strategies based on siRNAs with the aim to suppress replication of several viruses, including human immunodeficiency virus type 1 (HIV-1). It has been appreciated only recently that viral RNAs can also be processed endogenously by the microRNA-generating enzyme Dicer or recognized by cellular miRNAs, in processes that could be viewed as an adapted antiviral defense mechanism. Known to repress mRNA translation through recognition of specific binding sites usually located in their 3' untranslated region, miRNAs of host or viral origin may exert regulatory effects towards host and/or viral genes and influence viral replication and/or the host response to viral infection. This article summarizes our current state of knowledge on the relationship between HIV-1 and miRNA-guided RNA silencing, and discusses the different aspects of their interaction.


Assuntos
Infecções por HIV/genética , HIV-1/genética , MicroRNAs/fisiologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA