Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 538
Filtrar
1.
J Neurovirol ; 30(3): 303-315, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38943022

RESUMEN

Although previous studies have suggested that subtype B HIV-1 proviruses in the brain are associated with physiological changes and immune activation accompanied with microgliosis and astrogliosis, and indicated that both HIV-1 subtype variation and geographical location might influence the neuropathogenicity of HIV-1 in the brain. The natural course of neuropathogenesis of the most widespread subtype C HIV-1 has not been adequately investigated, especially for people living with HIV (PLWH) in sub-Saharan Africa. To characterize the natural neuropathology of subtype C HIV-1, postmortem frontal lobe and basal ganglia tissues were collected from nine ART-naïve individuals who died of late-stage AIDS with subtype C HIV-1 infection, and eight uninfected deceased individuals as controls. Histological staining was performed on all brain tissues to assess brain pathologies. Immunohistochemistry (IHC) against CD4, p24, Iba-1, GFAP, and CD8 in all brain tissues was conducted to evaluate potential viral production and immune activation. Histological results showed mild perivascular cuffs of lymphocytes only in a minority of the infected individuals. Viral capsid p24 protein was only detected in circulating immune cells of one infected individual, suggesting a lack of productive HIV-1 infection of the brain even at the late-stage of AIDS. Notably, similar levels of Iba-1 or GFAP between HIV + and HIV- brain tissues indicated a lack of microgliosis and astrogliosis, respectively. Similar levels of CD8 + cytotoxic T lymphocyte (CTL) infiltration between HIV + and HIV- brain tissues indicated CTL were not likely to be involved within subtype C HIV-1 infected participants of this cohort. Results from this subtype C HIV-1 study suggest that there is a lack of productive infection and limited neuropathogenesis by subtype C HIV-1 even at late-stage disease, which is in contrast to what was reported for subtype B HIV-1 by other investigators.


Asunto(s)
Proteína Ácida Fibrilar de la Glía , VIH-1 , Humanos , VIH-1/inmunología , VIH-1/patogenicidad , Masculino , Femenino , Adulto , Persona de Mediana Edad , África del Sur del Sahara , Proteína Ácida Fibrilar de la Glía/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Infecciones por VIH/patología , Ganglios Basales/inmunología , Ganglios Basales/patología , Ganglios Basales/virología , Proteínas de Unión al Calcio/inmunología , Proteínas de Unión al Calcio/genética , Lóbulo Frontal/inmunología , Lóbulo Frontal/patología , Lóbulo Frontal/virología , Proteína p24 del Núcleo del VIH/inmunología , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/patología , Complejo SIDA Demencia/virología , Antígenos CD4/inmunología , Linfocitos T CD8-positivos/inmunología , Gliosis/inmunología , Gliosis/patología , Gliosis/virología , Astrocitos/inmunología , Astrocitos/patología , Astrocitos/virología , Encéfalo/patología , Encéfalo/inmunología , Encéfalo/virología , Proteínas de Microfilamentos
2.
J Acquir Immune Defic Syndr ; 90(1): 106-114, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35090158

RESUMEN

BACKGROUND: We hypothesized that the induction of monocyte activation biomarkers, especially soluble urokinase-type plasminogen activator receptor (suPAR) and interferon γ-inducible protein 10 (IP-10), is lower in HIV-1C than HIV-1B, owing to a defective Tat cysteine dimotif (C30S). METHODS: A total of 68 paired cerebrospinal fluid (CSF) and blood samples from people with HIV (PWH), free of CNS opportunistic infections, from a Southern Brazil outpatient HIV clinic were evaluated such as HIV-1B subtype (n = 27), HIV-1C (n = 26), other (n = 15), and 19 HIV-negative controls. The levels of suPAR, IP-10, neopterin, and ß2 microglobulin (ß2m) in the CSF and serum were quantified using different immunoassays. RESULTS: Overall, in PWH, increases in CSF suPAR, CSF/serum suPAR, and CSF/serum ß2m correlated with worse working memory deficits (r = 0.303, 0.353, and 0.289, respectively, all P < 0.05). The medians of IP-10, suPAR, neopterin, and ß2m in CSF and serum and the CSF/serum ratio and suPAR index were comparable between the HIV-1B and HIV-1C subtypes. CSF IP-10 and neopterin and serum IP-10 and suPAR levels were higher in PWH than the HIV-negative controls (P = 0.015, P = 0.001, P < 0.0001, and P < 0.001, respectively). The serum ß2m level was higher in HIV-associated dementia than neuropsychologically normal or asymptomatic (P = 0.024). DISCUSSION: We observed that higher levels of CSF suPAR and the suPAR quotient correlated with worse working memory deficit. Elevated levels of monocyte activation were similar in both HIV-1 B and C subtypes, providing no evidence of reduced neuropathogenicity of HIV-1 subtype C Tat compared with subtype B.


Asunto(s)
Complejo SIDA Demencia , Quimiocina CXCL10 , Infecciones por VIH , Trastornos de la Memoria , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Complejo SIDA Demencia/líquido cefalorraquídeo , Complejo SIDA Demencia/virología , Biomarcadores/líquido cefalorraquídeo , Estudios de Casos y Controles , Quimiocina CXCL10/líquido cefalorraquídeo , Infecciones por VIH/líquido cefalorraquídeo , Infecciones por VIH/virología , VIH-1 , Humanos , Trastornos de la Memoria/líquido cefalorraquídeo , Trastornos de la Memoria/virología , Neopterin , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo
3.
Mediators Inflamm ; 2021: 1267041, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34483726

RESUMEN

HIV-1 can incite activation of chemokine receptors, inflammatory mediators, and glutamate receptor-mediated excitotoxicity. The mechanisms associated with such immune activation can disrupt neuronal and glial functions. HIV-associated neurocognitive disorder (HAND) is being observed since the beginning of the AIDS epidemic due to a change in the functional integrity of cells from the central nervous system (CNS). Even with the presence of antiretroviral therapy, there is a decline in the functioning of the brain especially movement skills, noticeable swings in mood, and routine performance activities. Under the umbrella of HAND, various symptomatic and asymptomatic conditions are categorized and are on a rise despite the use of newer antiretroviral agents. Due to the use of long-lasting antiretroviral agents, this deadly disease is becoming a manageable chronic condition with the occurrence of asymptomatic neurocognitive impairment (ANI), symptomatic mild neurocognitive disorder, or HIV-associated dementia. In-depth research in the pathogenesis of HIV has focused on various mechanisms involved in neuronal dysfunction and associated toxicities ultimately showcasing the involvement of various pathways. Increasing evidence-based studies have emphasized a need to focus and explore the specific pathways in inflammation-associated neurodegenerative disorders. In the current review, we have highlighted the association of various HIV proteins and neuronal cells with their involvement in various pathways responsible for the development of neurotoxicity.


Asunto(s)
Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/virología , Sistema Nervioso Central/virología , VIH-1/metabolismo , Proteínas Virales/metabolismo , Complejo SIDA Demencia/fisiopatología , Antirretrovirales/uso terapéutico , Astrocitos/virología , Sistema Nervioso Central/fisiopatología , Genoma , Proteína gp120 de Envoltorio del VIH/metabolismo , Proteína gp41 de Envoltorio del VIH/metabolismo , Infecciones por VIH/complicaciones , Infecciones por VIH/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Inflamación , Quinurenina/metabolismo , Macrófagos/virología , Microglía/virología , Neuronas/virología , Oligodendroglía/virología , Receptores de N-Metil-D-Aspartato/metabolismo , Carga Viral , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Viroporinas/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo
4.
Mol Neurobiol ; 58(11): 5421-5436, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33983546

RESUMEN

Maintaining an intact pool of neural progenitor cells (NPCs) is crucial for generating new and functionally active neurons. Methamphetamine (METH) can exacerbate the HIV-induced deficit of adult neurogenesis; however, potential mechanisms of this influence are still poorly understood. In the present study, we present evidence that chronic exposure to METH combined with brain infection by EcoHIV results in enhanced proliferation of NPCs in the subventricular zone (SVZ) in mice. This effect was long-lasting as it was preserved ex vivo in NPCs isolated from the exposed mice over several passages in the absence of additional treatments. Increased proliferation in response to METH plus HIV was associated with dysregulation of cyclin B1 and cyclin D. Transcriptomic studies indicated that 27 out of the top 30 differentially expressed genes in response to METH plus EcoHIV were targets of the forkhead box O transcriptional factor (FOXO) and primarily FOXO3. Additional ex vivo studies and in vitro experiments using human NPCs exposed to METH and infected with HIV revealed upregulation of the CXCL12-CXCR4 axis, leading to activation of downstream pAkt and pErk, the pathways that can phosphorylate FOXO3 and force its exports from the nuclei into the cytoplasm. Indeed, nuclear expulsion of FOXO3 was demonstrated both in mice exposed to METH and infected with EcoHIV and in cell cultures of human NPCs. These results provide novel information that exposure to METH combined with HIV infection can induce aberrant proliferation of SVZ-derived NPCs and identifies CXCL12-CXCR4-Akt-1-mediated phosphorylation of FOXO3 as the mechanism responsible for this effect.


Asunto(s)
Proteína Forkhead Box O3/fisiología , VIH-1/fisiología , Metanfetamina/toxicidad , Células-Madre Neurales/efectos de los fármacos , Complejo SIDA Demencia/complicaciones , Complejo SIDA Demencia/virología , Animales , Encéfalo/patología , Encéfalo/virología , Ciclo Celular , División Celular , Células Cultivadas , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/genética , Cromonas/farmacología , Modelos Animales de Enfermedad , Regulación Viral de la Expresión Génica , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Humanos , Ventrículos Laterales/patología , Masculino , Metanfetamina/farmacología , Ratones , Ratones Endogámicos C57BL , Morfolinas/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Células-Madre Neurales/virología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Trastornos Relacionados con Sustancias/complicaciones
5.
AIDS Care ; 33(3): 389-397, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32279542

RESUMEN

BACKGROUND: The screening strategy for HIV-Associated Neurocognitive Disorders (HAND) is challenging. The French Expert Report recommend the use of the Cognitive Complaints Questionnaire (QPC) and the Montreal Cognitive assessment. However, the QPC has never been studied in People Living with HIV (PLWH). This study aims to determine the degree of agreement between QPC and the presence of HAND according to Frascati criteria, established by a battery of neuropsychological tests. METHODS: Data from patients who performed both a QPC and a battery of neuropsychological tests over a six-month follow-up period were evaluated retrospectively. RESULTS: A total of 121 patients were selected, with a median age of 53.1 years old. Among participants, 92.6% had an undetectable plasma viral load, 49.6% had a nadir CD4 less than 200/mm3 and 23.1% had a CDC stage C. Median CD4 cell count was 686/mm3. Prevalence of HAND was 57%, including 28.9% of Asymptomatic Neurocognitive Impairment, 24.8% of Mild Neurocognitive Disorder and 3.3% of HIV-associated Dementia. This analyze shows no agreement between QPC and HIV-associated neurocognitive disorders (kappa = -0.007). CONCLUSIONS: The QPC is not relevant in the screening for HAND. Thus, it urges to develop a specific tool to assess cognitive complaints among PLWH.


Asunto(s)
Complejo SIDA Demencia/diagnóstico , Infecciones por VIH/complicaciones , Tamizaje Masivo/métodos , Trastornos Neurocognitivos/diagnóstico , Complejo SIDA Demencia/epidemiología , Complejo SIDA Demencia/psicología , Complejo SIDA Demencia/virología , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/uso terapéutico , Terapia Antirretroviral Altamente Activa , Cognición/fisiología , Estudios Transversales , Femenino , Infecciones por VIH/tratamiento farmacológico , Humanos , Masculino , Persona de Mediana Edad , Trastornos Neurocognitivos/epidemiología , Trastornos Neurocognitivos/etiología , Pruebas Neuropsicológicas , Estudios Retrospectivos
6.
J Leukoc Biol ; 109(3): 675-681, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32578908

RESUMEN

Approximately 15-40% of people living with HIV develop HIV-associated neurocognitive disorders, HAND, despite successful antiretroviral therapy. There are no therapies to treat these disorders. HIV enters the CNS early after infection, in part by transmigration of infected monocytes. Currently, there is a major opioid epidemic in the United States. Opioid use disorder in the context of HIV infection is important because studies show that opioids exacerbate HIV-mediated neuroinflammation that may contribute to more severe cognitive deficits. Buprenorphine is an opioid derivate commonly prescribed for opiate agonist treatment. We used the EcoHIV mouse model to study the effects of buprenorphine on cognitive impairment and to correlate these with monocyte migration into the CNS. We show that buprenorphine treatment prior to mouse EcoHIV infection prevents the development of cognitive impairment, in part, by decreased accumulation of monocytes in the brain. We propose that buprenorphine has a novel therapeutic benefit of limiting the development of neurocognitive impairment in HIV-infected opioid abusers as well as in nonabusers, in addition to decreasing the use of harmful opioids. Buprenorphine may also be used in combination with HIV prevention strategies such as pre-exposure prophylaxis because of its safety profile.


Asunto(s)
Complejo SIDA Demencia/prevención & control , Buprenorfina/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Complejo SIDA Demencia/complicaciones , Complejo SIDA Demencia/virología , Animales , Antígenos Ly/metabolismo , Encéfalo/patología , Buprenorfina/farmacología , Enfermedad Crónica , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/prevención & control , Disfunción Cognitiva/virología , Modelos Animales de Enfermedad , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Fenotipo , Carga Viral/efectos de los fármacos
7.
J Neuroimmune Pharmacol ; 16(2): 238-250, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33123948

RESUMEN

Despite adherence to treatment, individuals living with HIV have an increased risk for developing cognitive impairments, referred to as HIV-associated neurological disorders (HAND). Due to continued growth in the HIV population, particularly amongst the aging cohort, the neurobiological mechanisms of HAND are increasingly relevant. Similar to other viral proteins (e.g. Tat, Gp120, Vpr), the Negative Factor (Nef) is associated with numerous adverse effects in the CNS as well as cognitive impairments. In particular, emerging data indicate the consequences of Nef may be facilitated by the modulation of cellular autophagy as well as its inclusion into extracellular vesicles (EVs). The present review examines evidence for the molecular mechanisms by which Nef might contribute to neuronal dysfunction underlying HAND, with a specific focus on autophagy and EVs. Based on the these data, we propose an integrated model by which Nef may contribute to underlying neuronal dysfunction in HAND and highlight potentially novel therapeutic targets for HAND. Graphical abstract.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/virología , Modelos Neurológicos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Autofagia/fisiología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/virología , Humanos , Neuronas/virología
8.
Retrovirology ; 17(1): 35, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33213476

RESUMEN

BACKGROUND: HIV associated neurocognitive disorders cause significant morbidity and mortality despite the advent of highly active antiretroviral therapy. A deeper understanding of fundamental mechanisms underlying HIV infection and pathogenesis in the central nervous system is warranted. Microglia are resident myeloid cells of the brain that are readily infected by HIV and may constitute a CNS reservoir. We evaluated two microglial model cell lines (C20, HMC3) and two sources of primary cell-derived microglia (monocyte-derived microglia [MMG] and induced pluripotent stem cell-derived microglia [iPSC-MG]) as potential model systems for studying HIV-microglia interactions. RESULTS: All four microglial model cells expressed typical myeloid markers with the exception of low or absent CD45 and CD11b expression by C20 and HMC3, and all four expressed the microglia-specific markers P2RY12 and TMEM119. Marked differences were observed upon gene expression profiling, however, indicating that MMG and iPSC-MG cluster closely together with primary human microglial cells, while C20 and HMC3 were similar to each other but very different from primary microglia. Expression of HIV-relevant genes also revealed important differences, with iPSC-MG and MMG expressing relevant genes at levels more closely resembling primary microglia. iPSC-MG and MMG were readily infected with R5-tropic HIV, while C20 and HMC3 lack CD4 and require pseudotyping for infection. Despite many similarities, HIV replication dynamics and HIV-1 particle capture by Siglec-1 differed markedly between the MMG and iPSC-MG. CONCLUSIONS: MMG and iPSC-MG appear to be viable microglial models that are susceptible to HIV infection and bear more similarities to authentic microglia than two transformed microglia cell lines. The observed differences in HIV replication and particle capture between MMG and iPSC-MG warrant further study.


Asunto(s)
VIH-1/fisiología , VIH-1/patogenicidad , Microglía/virología , Modelos Biológicos , Complejo SIDA Demencia/virología , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular Transformada , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Células Madre Pluripotentes Inducidas/citología , Microglía/citología , Microglía/metabolismo , Monocitos/citología , Virión/metabolismo , Replicación Viral/genética
9.
J Neurovirol ; 26(6): 809-823, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32880873

RESUMEN

The virology of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and the human immune response to the virus are under vigorous investigation. There are now several reports describing neurological symptoms in individuals who develop coronavirus disease 2019 (COVID-19), the syndrome associated with SARS-CoV-2 infection. The prevalence, incidence, and clinical course of these symptoms will become clearer in the coming months and years through epidemiological studies. However, the long-term neurological and cognitive consequence of SARS-CoV-2 infection will remain conjectural for some time and will likely require the creation of cohort studies that include uninfected individuals. Considering the early evidence for neurological involvement in COVID-19 it may prove helpful to compare SARS-CoV-2 with another endemic and neurovirulent virus, human immunodeficiency virus-1 (HIV-1), when designing such cohort studies and when making predictions about neuropsychological outcomes. In this paper, similarities and differences between SARS-CoV-2 and HIV-1 are reviewed, including routes of neuroinvasion, putative mechanisms of neurovirulence, and factors involved in possible long-term neuropsychological sequelae. Application of the knowledge gained from over three decades of neuroHIV research is discussed, with a focus on alerting researchers and clinicians to the challenges in determining the cause of neurocognitive deficits among long-term survivors.


Asunto(s)
Complejo SIDA Demencia/virología , COVID-19/complicaciones , COVID-19/virología , Enfermedades del Sistema Nervioso/virología , SARS-CoV-2 , Humanos
11.
J Neuroinflammation ; 16(1): 182, 2019 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-31561750

RESUMEN

BACKGROUND: Since HIV-associated neurocognitive disorders (HANDs) occur in up to half of HIV-positive individuals, even with combined antiretroviral therapy (cART), adjunctive therapies are needed. Chronic CNS inflammation contributes to HAND and HIV encephalitis (HIVE). Baricitinib is a JAK 1/2 inhibitor approved in the USA, EU, and Japan for rheumatoid arthritis, demonstrating potent inhibition of IL-6, D-dimer, CRP, TNF-α, IFN-α/ß, and other pro-inflammatory cytokines. METHODS: Our modified murine HAND model was used to evaluate the ability of baricitinib to cross the blood-brain barrier (BBB) and modulate monocyte/macrophage-driven HAND. Severity of HAND was measured by assessing cognitive performance of low- and high-dose baricitinib treated versus untreated HAND mice. The severity of brain neuroinflammation was evaluated in these mouse groups after flow cytometric analyses. We also assessed the ability of baricitinib to block events in myeloid and lymphoid cells in vitro that may undergird the persistence of HIV in the central nervous system (CNS) in primary human macrophages (Mϕ) and lymphocytes including HIV replication, HIV-induced activation, reservoir expansion, and reservoir maintenance. RESULTS: In vivo, both doses of 10 and 50 mg/kg qd baricitinib crossed the BBB and reversed behavioral abnormalities conferred by HIV infection. Moreover, baricitinib significantly reduced HIV-induced neuroinflammation marked by glial activation: activated microglia (MHCII+/CD45+) and astrogliosis (GFAP). Baricitinib also significantly reduced the percentage of p24+ human macrophages in mouse brains (p < 0.05 versus HAND mice; t test). In vitro, baricitinib significantly reduced markers of persistence, reservoir size, and reseeding in Mϕ. CONCLUSION: These results show that blocking the JAK/STAT pathway reverses cognitive deficits and curtails inflammatory markers in HAND in mice. Our group recently reported safety and tolerability of ruxolitinib in HIV-infected individuals (Marconi et al., Safety, tolerability and immunologic activity of ruxolitinib added to suppressive ART, 2019), underscoring potential safety and utility of JAK inhibitors for additional human trials. The data reported herein coupled with our recent human trial with JAK inhibitors provide compelling preclinical data and impetus for considering a trial of baricitinib in HAND individuals treated with cART to reverse cognitive deficits and key events driving viral persistence.


Asunto(s)
Complejo SIDA Demencia/patología , Complejo SIDA Demencia/virología , Azetidinas/farmacología , Sulfonamidas/farmacología , Activación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones SCID , Purinas , Pirazoles , Latencia del Virus/efectos de los fármacos
12.
Int J STD AIDS ; 30(6): 617-619, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30722753

RESUMEN

We present the case of a 52-year-old man with human immunodeficiency virus (HIV) encephalitis resulting from cerebrospinal fluid (CSF) viral escape, to illustrate therapeutic challenges in patients with emergent CSF genotypic HIV drug resistance. This case report highlights the usefulness of CSF HIV-resistance testing to guide antiretroviral therapy and treatment optimizing decisions.


Asunto(s)
Complejo SIDA Demencia/tratamiento farmacológico , Fármacos Anti-VIH/uso terapéutico , Líquido Cefalorraquídeo/virología , Infecciones por VIH/tratamiento farmacológico , VIH-1/aislamiento & purificación , Plasma/virología , Carga Viral/efectos de los fármacos , Complejo SIDA Demencia/virología , Fármacos Anti-VIH/efectos adversos , Terapia Antirretroviral Altamente Activa , Farmacorresistencia Viral , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/genética , Humanos , Masculino , Persona de Mediana Edad , ARN Viral/sangre , ARN Viral/líquido cefalorraquídeo , Resultado del Tratamiento
14.
J Neuroimmune Pharmacol ; 14(1): 44-51, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30617749

RESUMEN

Mixed lineage kinases (MLKs) are a group of serine-threonine kinases that evolved in part to respond to endogenous and exogenous insults that result in oxidative stress and pro-inflammatory responses from innate immune cells. Human immunodeficiency virus type 1 (HIV-1) thrives in these conditions and is associated with the development of associated neurocognitive disorders (HAND). As part of a drug discovery program to identify new therapeutic strategies for HAND, we created a library of broad spectrum MLK inhibitors with drug-like properties. Serendipitously, the lead compound, URMC-099 has proved useful not only in reversing damage to synaptic architecture in models of HAND, but also serves to restore autophagy as a protective response when given in concert with nanoformulated antiretroviral therapy (nanoART) in persistently infected macrophages. These findings are reviewed in the context of MLK3 biology and cellular signaling pathways relevant to new HIV-1 therapies. Graphical abstract.


Asunto(s)
Complejo SIDA Demencia/virología , VIH-1/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Macrófagos/virología , Animales , VIH-1/fisiología , Humanos , Macrófagos/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirroles/farmacología , Latencia del Virus/efectos de los fármacos , Latencia del Virus/fisiología , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
15.
PLoS One ; 14(1): e0203566, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30653517

RESUMEN

Internal RNA modifications have been known for decades, however their roles in mRNA regulation have only recently started to be elucidated. Here we investigated the most abundant mRNA modification, N6-methyladenosine (m6A) in transcripts from the hippocampus of HIV transgenic (Tg) rats. The distribution of m6A peaks within HIV transcripts in HIV Tg rats largely corresponded to the ones observed for HIV transcripts in cell lines and T cells. Host transcripts were found to be differentially m6A methylated in HIV Tg rats. The functional roles of the differentially m6A methylated pathways in HIV Tg rats is consistent with a key role of RNA methylation in the regulation of the brain transcriptome in chronic HIV disease. In particular, host transcripts show significant differential m6A methylation of genes involved in several pathways related to neural function, suggestive of synaptodendritic injury and neurodegeneration, inflammation and immune response, as well as RNA processing and metabolism, such as splicing. Changes in m6A methylation were usually positively correlated with differential expression, while differential m6A methylation of pathways involved in RNA processing were more likely to be negatively correlated with gene expression changes. Thus, sets of differentially m6A methylated, functionally-related transcripts appear to be involved in coordinated transcriptional responses in the context of chronic HIV. Altogether, our results support that m6A methylation represents an additional layer of regulation of HIV and host gene expression in vivo that contributes significantly to the transcriptional effects of chronic HIV.


Asunto(s)
Complejo SIDA Demencia/genética , VIH-1/genética , Hipocampo/patología , ARN Mensajero/genética , Transcriptoma/genética , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/patología , Complejo SIDA Demencia/virología , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Modelos Animales de Enfermedad , Epigénesis Genética/genética , Epigénesis Genética/inmunología , Regulación de la Expresión Génica/inmunología , VIH-1/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Masculino , Metilación , Empalme del ARN/genética , ARN Mensajero/aislamiento & purificación , ARN Viral/genética , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Análisis de Secuencia de ARN , Transcriptoma/inmunología
16.
Sci Rep ; 9(1): 827, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696863

RESUMEN

The marked increase in life expectancy for HIV-1 seropositive individuals, following the great success of combination antiretroviral therapy (cART), heralds an examination of the progression of HIV-1 associated neurocognitive disorders (HAND). However, since the seminal call for animal models of HIV-1/AIDS in 1988, there has been no extant in vivo animal model system available to provide a truly longitudinal study of HAND. Here, we demonstrate that the HIV-1 transgenic (Tg) rat, resembling HIV-1 seropositive individuals on lifelong cART, exhibits age-related, progressive neurocognitive impairments (NCI), including alterations in learning, sustained attention, flexibility, and inhibition; deficits commonly observed in HIV-1 seropositive individuals. Pyramidal neurons from layers II-III of the medial prefrontal cortex (mPFC) displayed profound synaptic dysfunction in HIV-1 Tg animals relative to controls; dysfunction that was characterized by alterations in dendritic branching complexity, synaptic connectivity, and dendritic spine morphology. NCI and synaptic dysfunction in pyramidal neurons from layers II-III of the mPFC independently identified the presence of the HIV-1 transgene with at least 78.5% accuracy. Thus, even in the absence of sensory or motor system deficits and comorbidities, HAND is a neurodegenerative disease characterized by age-related disease progression; impairments which may be due, at least partly, to synaptic dysfunction in the mPFC. Further, the progression of HAND with age in the HIV-1 Tg rat and associated synaptic dysfunction affords an instrumental model system for the development of therapeutics and functional cure strategies.


Asunto(s)
Complejo SIDA Demencia/patología , Enfermedades Neurodegenerativas/patología , Células Piramidales/patología , Complejo SIDA Demencia/virología , Animales , Modelos Animales de Enfermedad , Femenino , VIH-1 , Masculino , Enfermedades Neurodegenerativas/virología , Corteza Prefrontal/citología , Corteza Prefrontal/patología , Corteza Prefrontal/virología , Células Piramidales/virología , Ratas , Ratas Transgénicas
17.
J Acquir Immune Defic Syndr ; 80(4): e95-e102, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30531306

RESUMEN

BACKGROUND: Mitochondrial DNA (mtDNA) copy number varies by cell type and energy demands. Blood mtDNA copy number has been associated with neurocognitive function in persons without HIV. Low mtDNA copy number may indicate disordered mtDNA replication; high copy number may reflect a response to mitochondrial dysfunction. We hypothesized that blood mtDNA copy number estimated from genome-wide genotyping data is related to neurocognitive impairment (NCI) in persons with HIV. METHODS: In the CNS HIV Antiretroviral Therapy Effects Research (CHARTER) study, peripheral blood mtDNA copy number was obtained from genome-wide genotyping data as a ratio of mtDNA single-nucleotide polymorphism probe intensities relative to nuclear DNA single-nucleotide polymorphisms. In a multivariable regression model, associations between mtDNA copy number and demographics, blood cell counts, and HIV disease and treatment characteristics were tested. Associations of mtDNA copy number with the global deficit score (GDS), GDS-defined NCI (GDS ≥ 0.5), and HIV-associated neurocognitive disorder (HAND) diagnosis were tested by logistic regression, adjusting for potential confounders. RESULTS: Among 1010 CHARTER participants, lower mtDNA copy number was associated with longer antiretroviral therapy duration (P < 0.001), but not with d-drug exposure (P = 0.85). mtDNA copy number was also associated with GDS (P = 0.007), GDS-defined NCI (P < 0.001), and HAND (P = 0.002). In all analyses, higher mtDNA copy number was associated with poorer cognitive performance. CONCLUSIONS: Higher mtDNA copy number estimated from peripheral blood genotyping was associated with worse neurocognitive performance in adults with HIV. These results suggest a connection between peripheral blood mtDNA and NCI, and may represent increased mtDNA replication in response to mitochondrial dysfunction.


Asunto(s)
Complejo SIDA Demencia/genética , Variaciones en el Número de Copia de ADN/genética , ADN Mitocondrial/genética , Infecciones por VIH/psicología , Mitocondrias/genética , Trastornos Neurocognitivos/genética , Complejo SIDA Demencia/virología , Adulto , Antirretrovirales/uso terapéutico , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/patología , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Trastornos Neurocognitivos/virología , Pruebas Neuropsicológicas
18.
J Neurovirol ; 25(1): 72-84, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30519968

RESUMEN

The measurement and determinants of HIV-associated neurocognitive disorders (HAND) are under intense debate. We used latent profile analysis (LPA) and machine learning to define neurocognitive performance profiles and identify their associated risk factors in HIV patients receiving antiretroviral therapy (ART). Neurocognitive performance was assessed by a multidomain neuropsychological test battery. LPA was used to define individual neurocognitive profiles. Random forest analyses (RFA) identified the most important factors distinguishing each profile. Three profiles emerged from the LPA: profile 1 (P1, n = 159) achieved the highest performance, while profile 2 (P2, n = 163) had lowered executive functions and verbal memory, and profile 3 (P3, n = 59) was globally impaired. RFA achieved good prediction (area under the curve ≥ 0.80) only for global impairment (P3). Non-North American descent was the dominant predictor of P3, followed by factors coinciding with non-North American descent (female sex and toxoplasma seropositivity). Additional predictors included unemployment, current depressive symptoms, lower nadir CD4, and longstanding HIV. Restricting analyses to North Americans pointed to the additional importance of ART achieving high CSF levels and older age in prediction of P3. HAND diagnoses were most common in the globally impaired profile (P3 = 89.8%), followed by the group with reduced higher-order neurocognitive performance (P2 = 16.6%). Thus, implementation of LPA and RFA empirically distinguished three distinct neurocognitive performance profiles in this HIV-infected cohort while also highlighting potential risk factors and their relative importance to neurocognitive impairment. These data-driven analytical methods pointed to discernible demographic, HIV- and treatment-related risk factor constellations in patients born outside and within North America that might influence diagnostic and therapeutic decisions.


Asunto(s)
Complejo SIDA Demencia/diagnóstico , Fármacos Anti-VIH/uso terapéutico , Cognición/fisiología , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/fisiopatología , Complejo SIDA Demencia/tratamiento farmacológico , Complejo SIDA Demencia/fisiopatología , Complejo SIDA Demencia/virología , Adulto , Terapia Antirretroviral Altamente Activa , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Encéfalo/virología , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/virología , Cognición/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/virología , Estudios de Cohortes , Depresión/fisiopatología , Depresión/psicología , Función Ejecutiva/efectos de los fármacos , Función Ejecutiva/fisiología , Femenino , Humanos , Aprendizaje Automático , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Persona de Mediana Edad , Pruebas Neuropsicológicas , Factores de Riesgo , Índice de Severidad de la Enfermedad , Desempleo/psicología
19.
J Neuroimmune Pharmacol ; 14(1): 120-133, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29981000

RESUMEN

HIV-associated neurocognitive disorders (HAND) occur in ~50% of HIV infected individuals despite combined antiretroviral therapy. Transmigration into the CNS of CD14+CD16+ monocytes, particularly those that are HIV infected and express increased surface chemokine receptor CCR2, contributes to neuroinflammation and HAND. To examine whether in HIV infected individuals CCR2 on CD14+CD16+ monocytes serves as a potential peripheral blood biomarker of HAND, we examined a cohort of 45 HIV infected people. We correlated CCR2 on CD14+CD16+ monocytes with cognitive status, proton magnetic resonance spectroscopy (1H-MRS) measured neurometabolite levels, and peripheral blood mononuclear cell (PBMC) HIV DNA copies. We determined that CCR2 was increased specifically on CD14+CD16+ monocytes from people with HAND (median [interquartile range (IQR)]) (63.3 [51.6, 79.0]), compared to those who were not cognitively impaired (38.8 [26.7, 56.4]) or those with neuropsychological impairment due to causes other than HIV (39.8 [30.2, 46.5]). CCR2 was associated with neuronal damage, based on the inverse correlation of CCR2 on CD14+CD16+ monocytes with total N-Acetyl Aspartate (tNAA)/total Creatine (tCr) (r2 = 0.348, p = 0.01) and Glutamine-Glutamate (Glx)/tCr (r2 = 0.356, p = 0.01) in the right and left caudate nucleus, respectively. CCR2 on CD14+CD16+ monocytes also correlated with PBMC HIV DNA copies (ρ = 0.618, p = 0.02) that has previously been associated with HAND. These findings suggest that CCR2 on CD14+CD16+ monocytes may be a peripheral blood biomarker of HAND, indicative of increased HIV infected CD14+CD16+ monocyte entry into the CNS that possibly increases the macrophage viral reservoir and contributes to HAND.


Asunto(s)
Complejo SIDA Demencia/sangre , Complejo SIDA Demencia/virología , Biomarcadores/sangre , Monocitos/virología , Receptores CCR2/sangre , Adulto , Anciano , ADN Viral/análisis , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Masculino , Persona de Mediana Edad , Receptores de IgG/metabolismo
20.
J Neuroimmune Pharmacol ; 14(1): 110-119, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30194646

RESUMEN

The question of whether the human brain is an anatomical site of persistent HIV-1 infection during suppressive antiretroviral therapy (ART) is critical, but remains unanswered. The presence of virus in the brains of HIV patients whose viral load is effectively suppressed would demonstrate not only the potential for CNS to act as an anatomical HIV reservoir, but also the urgent need to understand the factors contributing to persistent HIV behind the blood-brain barrier. Here, we investigated for the first time the presence of cells harboring HIV DNA and RNA in the brains from subjects with undetectable plasma viral load and sustained viral suppression, as identified by the National NeuroAIDS Tissue Consortium. Using new, highly sensitive in situ hybridization techniques, RNAscope and DNAscope, in combination with immunohistochemistry, we were able to detect HIV-1 in the brains of all virally suppressed cases and found that brain macrophages and microglia, but not astrocytes, were the cells harboring HIV DNA in the brain. This study demonstrated that HIV reservoirs persist in brain macrophages/microglia during suppressive ART, which cure/treatment strategies will need to focus on targeting.


Asunto(s)
Encéfalo/virología , ADN Viral/análisis , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Macrófagos/virología , Complejo SIDA Demencia/virología , Adulto , Fármacos Anti-VIH/uso terapéutico , Astrocitos , Femenino , VIH-1/fisiología , Humanos , Masculino , Persona de Mediana Edad , Latencia del Virus/fisiología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...