Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Extracell Vesicles ; 13(4): e12439, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38647111

RESUMEN

Our previous findings demonstrated that astrocytic HIF-1α plays a major role in HIV-1 Tat-mediated amyloidosis which can lead to Alzheimer's-like pathology-a comorbidity of HIV-Associated Neurocognitive Disorders (HAND). These amyloids can be shuttled in extracellular vesicles, and we sought to assess whether HIV-1 Tat stimulated astrocyte-derived EVs (ADEVs) containing the toxic amyloids could result in neuronal injury in vitro and in vivo. We thus hypothesized that blocking HIF-1α could likely mitigate HIV-1 Tat-ADEV-mediated neuronal injury. Rat hippocampal neurons when exposed to HIV-1 Tat-ADEVs carrying the toxic amyloids exhibited amyloid accumulation and synaptodendritic injury, leading to functional loss as evidenced by alterations in miniature excitatory post synaptic currents. The silencing of astrocytic HIF-1α not only reduced the biogenesis of ADEVs, as well as amyloid cargos, but also ameliorated neuronal synaptodegeneration. Next, we determined the effect of HIV-1 Tat-ADEVs carrying amyloids in the hippocampus of naive mice brains. Naive mice receiving the HIV-1 Tat-ADEVs, exhibited behavioural changes, and Alzheimer's 's-like pathology accompanied by synaptodegeneration. This impairment(s) was not observed in mice injected with HIF-1α silenced ADEVs. This is the first report demonstrating the role of amyloid-carrying ADEVs in mediating synaptodegeneration leading to behavioural changes associated with HAND and highlights the protective role of HIF-1α.


Asunto(s)
Astrocitos , Vesículas Extracelulares , VIH-1 , Hipocampo , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neuronas , Vesículas Extracelulares/metabolismo , Animales , Astrocitos/metabolismo , Ratones , Ratas , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , VIH-1/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Trastornos Neurocognitivos/metabolismo , Trastornos Neurocognitivos/etiología , Infecciones por VIH/metabolismo , Infecciones por VIH/complicaciones , Masculino , Complejo SIDA Demencia/metabolismo
2.
PLoS Biol ; 18(5): e3000660, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32453744

RESUMEN

Increased life expectancy of patients diagnosed with HIV in the current era of antiretroviral therapy is unfortunately accompanied with the prevalence of HIV-associated neurocognitive disorders (HANDs) and risk of comorbidities such as Alzheimer-like pathology. HIV-1 transactivator of transcription (Tat) protein has been shown to induce the production of toxic neuronal amyloid protein and also enhance neurotoxicity. The contribution of astrocytes in Tat-mediated amyloidosis remains an enigma. We report here, in simian immunodeficiency virus (SIV)+ rhesus macaques and patients diagnosed with HIV, brain region-specific up-regulation of amyloid precursor protein (APP) and Aß (40 and 42) in astrocytes. In addition, we find increased expression of ß-site cleaving enzyme (BACE1), APP, and Aß in human primary astrocytes (HPAs) exposed to Tat. Mechanisms involved up-regulation of hypoxia-inducible factor (HIF-1α), its translocation and binding to the long noncoding RNA (lncRNA) BACE1-antisense transcript (BACE1-AS), resulting, in turn, in the formation of the BACE1-AS/BACE1 RNA complex, subsequently leading to increased BACE1 protein, and activity and generation of Aß-42. Gene silencing approaches confirmed the regulatory role of HIF-1α in BACE1-AS/BACE1 in Tat-mediated amyloidosis. This is the first report implicating the role of the HIF-1α/lncRNABACE1-AS/BACE1 axis in Tat-mediated induction of astrocytic amyloidosis, which could be targeted as adjunctive therapies for HAND-associated Alzheimer-like comorbidity.


Asunto(s)
Amiloidosis/virología , Astrocitos/metabolismo , Infecciones por VIH/complicaciones , Trastornos Neurocognitivos/virología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Péptidos beta-Amiloides/metabolismo , Amiloidosis/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Infecciones por VIH/metabolismo , VIH-1 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macaca mulatta , Persona de Mediana Edad , Trastornos Neurocognitivos/metabolismo , Fragmentos de Péptidos/metabolismo , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba
3.
Autophagy ; 16(2): 289-312, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30990365

RESUMEN

Although cocaine exposure has been shown to potentiate neuroinflammation by upregulating glial activation in the brain, the role of mitophagy in this process remains an enigma. In the present study, we sought to examine the role of impaired mitophagy in cocaine-mediated activation of microglia and to determine the ameliorative potential of superoxide dismutase mimetics in this context. Our findings demonstrated that exposure of mouse primary microglial cells (mPMs) to cocaine resulted in decreased mitochondrial membrane potential, that was accompanied by increased expression of mitophagy markers, PINK1 and PRKN. Exposure of microglia to cocaine also resulted in increased expression of DNM1L and OPTN with a concomitant decrease in the rate of mitochondrial oxygen consumption as well as impaired mitochondrial functioning. Additionally, in the presence of cocaine, microglia also exhibited upregulated expression of autophagosome markers, BECN1, MAP1LC3B-II, and SQSTM1. Taken together, these findings suggested diminished mitophagy flux and accumulation of mitophagosomes in the presence of cocaine. These findings were further confirmed by imaging techniques such as transmission electron microscopy and confocal microscopy. Cocaine-mediated activation of microglia was further monitored by assessing the expression of the microglial marker (ITGAM) and the inflammatory cytokine (Tnf, Il1b, and Il6) mRNAs. Pharmacological, as well as gene-silencing approaches aimed at blocking both the autophagy/mitophagy and SIGMAR1 expression, underscored the role of impaired mitophagy in cocaine-mediated activation of microglia. Furthermore, superoxide dismutase mimetics such as TEMPOL and MitoTEMPO were shown to alleviate cocaine-mediated impaired mitophagy as well as microglial activation.Abbreviations: 3-MA: 3-methyladenine; Δψm: mitochondrial membrane potential; ACTB: actin, beta; AIF1: allograft inflammatory factor 1; ATP: adenosine triphosphate; BAF: bafilomycin A1; BECN1: beclin 1, autophagy related; CNS: central nervous system; DNM1L: dynamin 1 like; DMEM: Dulbecco modified Eagle medium; DAPI: 4,6-Diamidino-2-phenylindole; DRD2: dopamine receptor D2; ECAR: extracellular acidification rate; FBS: fetal bovine serum; FCCP: Trifluoromethoxy carbonylcyanide phenylhydrazone; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; IL1B: interleukin 1, beta; IL6: interleukin 6; ITGAM: integrin subunit alpha M; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; mPMs: mouse primary microglial cells; MRC: maximal respiratory capacity; NFKB: nuclear factor kappa B; NLRP3: NLR family pyrin domain containing 3; NTRK2: neurotrophic receptor tyrosine kinase 2; OCR: oxygen consumption rate; OPTN: optineurin; PBS: phosphate buffered saline; PINK1: PTEN induced putative kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; ROS: reactive oxygen species; siRNA: small interfering RNA; SQSTM1: sequestosome 1; TNF: tumor necrosis factor.


Asunto(s)
Cocaína/efectos adversos , Microglía/patología , Mitocondrias/patología , Mitofagia , Superóxido Dismutasa/metabolismo , Animales , Autofagia , Beclina-1/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Óxidos N-Cíclicos , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Silenciador del Gen , Mediadores de Inflamación/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Mitofagia/efectos de los fármacos , Modelos Biológicos , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/ultraestructura , Proteínas Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores sigma/metabolismo , Transducción de Señal , Regulación hacia Arriba/efectos de los fármacos
4.
J Clin Invest ; 128(12): 5428-5433, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30277472

RESUMEN

In the era of combined antiretroviral therapy (cART), lung diseases such as chronic bronchitis (CB) and chronic obstructive pulmonary disease (COPD) are common among persons living with HIV (PLWH), particularly smokers. Although smoking is highly prevalent among PLWH, HIV may be an independent risk factor for lung diseases; however, the role of HIV and cigarette smoke (CS) and their potential interaction in the development of chronic lung diseases among PLWH has not been delineated. To investigate this interaction, cynomolgus macaques were exposed to CS and/or simian-adapted human immunodeficiency virus (SHIV) and treated with cART. The development of CB and the lung functions were evaluated following CS±SHIV treatment. The results showed that in the lung, SHIV was a strong independent risk factor for goblet cell metaplasia/hyperplasia and mucus formation, MUC5AC synthesis, loss of tight junction proteins, and increased expression of Th2 cytokines/transcription factors. In addition, SHIV and CS synergistically reduced lung function and increased extrathoracic tracheal ring thickness. Interestingly, SHIV infection generated significant numbers of HIV-gp120+ epithelial cells (HGECs) in small airways and alveoli, and their numbers doubled in CS+SHIV-infected lungs. We conclude that even with cART, SHIV independently induces CB and pro-COPD changes in the lung, and the effects are exacerbated by CS.


Asunto(s)
Fumar Cigarrillos , Infecciones por VIH , VIH-1 , Pulmón , Alveolos Pulmonares , Enfermedad Pulmonar Obstructiva Crónica , Animales , Fumar Cigarrillos/efectos adversos , Fumar Cigarrillos/patología , Fumar Cigarrillos/fisiopatología , Infecciones por VIH/patología , Infecciones por VIH/fisiopatología , Pulmón/patología , Pulmón/fisiopatología , Pulmón/virología , Macaca fascicularis , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiopatología , Alveolos Pulmonares/virología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/virología
5.
Autophagy ; 14(9): 1596-1619, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29966509

RESUMEN

While the advent of combination antiretroviral therapy (cART) has dramatically increased the life expectancy of HIV-1 infected individuals, paradoxically, however, the prevalence of HIV-1-associated neurocognitive disorders is on the rise. Based on the premise that the cytotoxic HIV-1 protein, transactivator of transcription (TAT), a known activator of glial cells that is found to persist in the central nervous system (CNS) despite cART, we sought to explore the role of defective mitophagy in HIV-1 TAT-mediated microglial activation. Our results demonstrated that exposure of mouse primary microglia to HIV-1 TAT resulted in cellular activation involving altered mitochondrial membrane potential that was accompanied by accumulation of damaged mitochondria. Exposure of microglia to HIV-1 TAT resulted in increased expression of mitophagy signaling proteins, such as PINK1, PRKN, and DNM1L, with a concomitant increase in the formation of autophagosomes, as evidenced by increased expression of BECN1 and MAP1LC3B-II. Intriguingly, exposure of cells to HIV-1 TAT also resulted in increased expression of SQSTM1, signifying thereby a possible blockade of the mitophagy flux, leading, in turn, to the accumulation of mitophagosomes. Interestingly, HIV-1 TAT-mediated activation of microglia was associated with decreased rate of extracellular acidification and mitochondrial oxygen consumption and increased expression of proinflammatory cytokines, such as Tnf, Il1b, and Il6. HIV-1 TAT-mediated defective mitophagy leading to microglial activation was further validated in vivo in the brains of HIV-1 transgenic rats. In conclusion, HIV-1 TAT activates microglia by increasing mitochondrial damage via defective mitophagy. ABBREVIATIONS: 3-MA: 3-methyladenine; Δψm: mitochondrial membrane potential; ACTB: actin, beta; AIF1: allograft inflammatory factor 1; ATP: adenosine triphosphate; BAF: bafilomycin A1; BECN1: beclin 1, autophagy related; cART: combined antiretroviral therapy; CNS: central nervous system; DNM1L: dynamin 1 like; DMEM: Dulbecco modified Eagle medium; DAPI: 4,6-diamidino-2-phenylindole ; ECAR: extracellular acidification rate; FBS: fetal bovine serum; FCCP: trifluoromethoxy carbonylcyanide phenylhydrazone; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HAND: HIV-1-associated neurocognitive disorders; HIV-1 TAT: human immunodeficiency virus-1 transactivator of transcription; IL1B: interleukin 1, beta; IL6: interleukin 6; ITGAM: integrin subunit alpha M; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; mPMs: mouse primary microglial cells; MRC: maximal respiratory capacity; mt-CO1: mitochondrially encoded cytochrome c oxidase; mt-ND6: mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 6; NFKB1: nuclear factor kappa B subunit 1; NLRP3: NLR family pyrin domain containing 3; OCR: oxygen consumption rate; PBS: phosphate-buffered saline; PINK1: PTEN induced putative kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; ROS: reactive oxygen species; siRNA: small interfering RNA; SQSTM1: sequestosome 1; TNF: tumor necrosis factor.


Asunto(s)
Productos del Gen tat/farmacología , VIH-1/química , Microglía/metabolismo , Mitocondrias/patología , Mitofagia/efectos de los fármacos , Animales , Autofagia/efectos de los fármacos , Beclina-1/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Citocinas/metabolismo , Metabolismo Energético/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Modelos Biológicos , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/ultraestructura , Proteínas Quinasas/metabolismo , Ratas Transgénicas , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
6.
Oncotarget ; 9(26): 18648-18663, 2018 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-29719633

RESUMEN

LncRNAs are long non-coding regulatory RNAs that are longer than 200 nucleotides. One of the major functions of lncRNAs is the regulation of specific gene expression at multiple steps including, recruitment and expression of basal transcription machinery, post-transcriptional modifications and epigenetics [1]. Emerging evidence suggests that lncRNAs also play a critical role in maintaining tissue homeostasis during physiological and pathological conditions, lipid homeostasis, as well as epithelial and smooth muscle cell homeostasis, a topic that has been elegantly reviewed [2-5]. While aberrant expression of lncRNAs has been implicated in several disease conditions, there is paucity of information about their contribution to the etiology of diseases [6]. Several studies have compared the expression of lncRNAs under normal and cancerous conditions and found differential expression of several lncRNAs, suggesting thereby an involvement of lncRNAs in disease processes [7, 8]. Furthermore, the ability of lncRNAs to influence epigenetic changes also underlies their role in disease pathogenesis since epigenetic regulation is known to play a critical role in many human diseases [1]. LncRNAs thus are not only involved in homeostatic functioning but also play a vital role in the progression of many diseases, thereby underscoring their potential as novel therapeutic targets for the alleviation of a variety of human disease conditions.

7.
Theranostics ; 8(1): 256-276, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29290806

RESUMEN

RATIONALE: Long-acting slow effective release antiretroviral therapy (LASER ART) was developed to improve patient regimen adherence, prevent new infections, and facilitate drug delivery to human immunodeficiency virus cell and tissue reservoirs. In an effort to facilitate LASER ART development, "multimodal imaging theranostic nanoprobes" were created. These allow combined bioimaging, drug pharmacokinetics and tissue biodistribution tests in animal models. METHODS: Europium (Eu3+)- doped cobalt ferrite (CF) dolutegravir (DTG)- loaded (EuCF-DTG) nanoparticles were synthesized then fully characterized based on their size, shape and stability. These were then used as platforms for nanoformulated drug biodistribution. RESULTS: Folic acid (FA) decoration of EuCF-DTG (FA-EuCF-DTG) nanoparticles facilitated macrophage targeting and sped drug entry across cell barriers. Macrophage uptake was higher for FA-EuCF-DTG than EuCF-DTG nanoparticles with relaxivities of r2 = 546 mM-1s-1 and r2 = 564 mM-1s-1 in saline, and r2 = 850 mM-1s-1 and r2 = 876 mM-1s-1 in cells, respectively. The values were ten or more times higher than what was observed for ultrasmall superparamagnetic iron oxide particles (r2 = 31.15 mM-1s-1 in saline) using identical iron concentrations. Drug particles were detected in macrophage Rab compartments by dual fluorescence labeling. Replicate particles elicited sustained antiretroviral responses. After parenteral injection of FA-EuCF-DTG and EuCF-DTG into rats and rhesus macaques, drug, iron and cobalt levels, measured by LC-MS/MS, magnetic resonance imaging, and ICP-MS were coordinate. CONCLUSION: We posit that these theranostic nanoprobes can assess LASER ART drug delivery and be used as part of a precision nanomedicine therapeutic strategy.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Imagen Multimodal/métodos , Nanomedicina Teranóstica/métodos , Animales , Sistemas de Liberación de Medicamentos/métodos , Europio/química , Europio/farmacocinética , Ácido Fólico/química , Compuestos Heterocíclicos con 3 Anillos/química , Compuestos Heterocíclicos con 3 Anillos/farmacocinética , Macaca mulatta , Macrófagos/metabolismo , Microscopía Confocal , Nanopartículas/química , Oxazinas , Piperazinas , Piridonas
8.
Mol Neurobiol ; 55(8): 6713-6733, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29344928

RESUMEN

A recent study from our lab has revealed a link between morphine-mediated autophagy and synaptic impairment. The current study was aimed at investigating whether morphine-mediated activation of astrocytes involved the ER stress/autophagy axis. Our in vitro findings demonstrated upregulation of GFAP indicating astrocyte activation with a concomitant increase in the production of proinflammatory cytokines in morphine-exposed human astrocytes. Using both pharmacological and gene-silencing approaches, it was demonstrated that morphine-mediated defective autophagy involved upstream activation of ER stress with subsequent downstream astrocyte activation via the µ-opioid receptor (MOR). In vivo validation demonstrated preferential activation of ER stress/autophagy axis in the areas of the brain not associated with pain such as the basal ganglia, frontal cortex, occipital cortex, and the cerebellum of morphine-dependent rhesus macaques, and this correlated with increased astrocyte activation and neuroinflammation. Interventions aimed at blocking either the MOR or ER stress could thus likely be developed as promising therapeutic targets for abrogating morphine-mediated astrocytosis.


Asunto(s)
Autofagia , Encéfalo/patología , Estrés del Retículo Endoplásmico , Gliosis/patología , Morfina/efectos adversos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Citocinas/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Macaca mulatta , Modelos Biológicos , Receptores Opioides mu/metabolismo , Regulación hacia Arriba/efectos de los fármacos
9.
Cell Death Dis ; 7(11): e2481, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27882942

RESUMEN

Exosomes are membrane-enriched extracellular vesicles with a proposed diameter in the range of 30-100 nm. They are released during both normal homeostasis as well as under pathological conditions by most cell types. In recent years, there has been robust interest in the study of these vesicles as conduits for the delivery of information between cells in both analogous as well as disparate tissues. Their ability to transport specialized cargo including signaling mediators, proteins, messenger RNA and miRNAs characterizes these vesicles as primary facilitators of cell-to-cell communication and regulation. Exosomes have also been demonstrated to have important roles in the field of cancer biology and metastasis. More recently, their role in several neurodegenerative disorders has been gaining increased momentum as these particles have been shown to promote the spread of toxic factors such as amyloid beta and prions, adding further validity to their role as important regulators of disease pathogenesis. This review briefly summarizes current findings and thoughts on exosome biology in the context of neurodegenerative disorders and the manipulation of these particles for the development of potential therapeutic strategies.


Asunto(s)
Complejo SIDA Demencia/complicaciones , Complejo SIDA Demencia/patología , Vesículas Extracelulares/metabolismo , Enfermedades Neurodegenerativas/patología , Animales , Exosomas/metabolismo , Humanos
10.
Curr HIV Res ; 14(5): 412-416, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27009095

RESUMEN

BACKGROUND: While the advancement of highly active antiretroviral therapy (HAART) has transformed the course of HIV/AIDS from a death sentence to a manageable chronic condition, the prevalence of a constellation of neurological disorders collectively termed as HIV-associated neurocognitive disorders (HAND) continues to persist in these patients. HAND is characterized by cognitive dysfunction, depression, impaired memory and/or deficits in motor skills. The underlying factors leading to HAND have been the subject of extensive research and are thought to be associated with HIV infection in the CNS combined with robust immune activation of resident cells of the CNS. In addition, there is a strong correlation between chronic substance abuse and the manifestation of HAND. Among the various commonly abused drugs, cocaine has been extensively studied for its ability to exacerbate the neuropathogenesis of HAND. Ample evidence suggests that cocaine not only facilitates viral replication in macrophages and microglia, but also inflicts deleterious effects on various other cells of the CNS, thereby contributing to the potentiation of HAND. Cocaine has been shown to enhance the permeability of the blood-brain barrier (BBB) through various mechanisms including direct pro-apoptotic effects on brain endothelial cells, systemic induction of inflammatory factors which have been demonstrated to down-regulate tight junction proteins and via up-regulation of several endothelial adhesion molecules leading to accelerated breach of the BBB and increased influx of HIV-infected leukocytes into the CNS. Cocaine also enhances viral replication in CNS astrocytes and promotes astrogliosis via astrocyte activation and proliferation. Furthermore, cocaine also exacerbates neuroinflammatory responses by mediating microglial activation and migration. In addition to cellular injury mediated by inflammatory responses, cocaine also directly affects the brain reward system by disrupting the homeostasis of neurotransmitters such as dopamine and acetylcholine and works synergistically with viral proteins such as tat and gp120 to promote neuronal injury. CONCLUSION: This review highlights previous studies in the field on the role of cocaine in the progression of HAND and gives an overview of the major signaling pathways in the CNS that are involved in this process.


Asunto(s)
Complejo SIDA Demencia/fisiopatología , Cocaína/metabolismo , Inhibidores de Captación de Dopamina/metabolismo , Infecciones por VIH/complicaciones , Trastornos Relacionados con Sustancias/complicaciones , Apoptosis , Barrera Hematoencefálica/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , VIH/efectos de los fármacos , VIH/fisiología , Humanos , Macrófagos/virología , Microglía/virología , Permeabilidad/efectos de los fármacos , Replicación Viral/efectos de los fármacos
11.
J Neuroinflammation ; 13: 33, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26860188

RESUMEN

BACKGROUND: Neuroinflammation associated with advanced human immunodeficiency virus (HIV)-1 infection is often exacerbated by chronic cocaine abuse. Cocaine exposure has been demonstrated to mediate up-regulation of inflammatory mediators in in vitro cultures of microglia. The molecular mechanisms involved in this process, however, remain poorly understood. In this study, we sought to explore the underlying signaling pathways involved in cocaine-mediated activation of microglial cells. METHODS: BV2 microglial cells were exposed to cocaine and assessed for toll-like receptor (TLR2) expression by quantitative polymerase chain reaction (qPCR), western blot, flow cytometry, and immunofluorescence staining. The mRNA and protein levels of cytokines (TNFα, IL-6, MCP-1) were detected by qPCR and ELISA, respectively; level of reactive oxygen species (ROS) production was examined by the Image-iT LIVE Green ROS detection kit; activation of endoplasmic reticulum (ER)-stress pathways were detected by western blot. Chromatin immunoprecipitation (ChIP) assay was employed to discern the binding of activating transcription factor 4 (ATF4) with the TLR2 promoter. Immunoprecipitation followed by western blotting with tyrosine antibody was used to determine phosphorylation of TLR2. Cocaine-mediated up-regulation of TLR2 expression and microglial activation was validated in cocaine-injected mice. RESULTS: Exposure of microglial cells to cocaine resulted in increased expression of TLR2 with a concomitant induction of microglial activation. Furthermore, this effect was mediated by NADPH oxidase-mediated rapid accumulation of ROS with downstream activation of the ER-stress pathways as evidenced by the fact that cocaine exposure led to up-regulation of pPERK/peIF2α/ATF4 and TLR2. The novel role of ATF4 in the regulation of TLR2 expression was confirmed using genetic and pharmacological approaches. CONCLUSIONS: xThe current study demonstrates that cocaine-mediated activation of microglia involves up-regulation of TLR2 through the ROS-ER stress-ATF4-TLR2 axis. Understanding the mechanism(s) involved in cocaine-mediated up-regulation of ROS-ER stress/TLR2 expression and microglial activation could have implications for the development of potential therapeutic targets aimed at resolving neuroinflammation in cocaine abusers.


Asunto(s)
Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Microglía/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Animales Recién Nacidos , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
12.
Autophagy ; 11(7): 995-1009, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26043790

RESUMEN

Cocaine abuse leads to neuroinflammation, which, in turn, contributes to the pathogenesis of neurodegeneration associated with advanced HIV-1 infection. Autophagy plays important roles in both innate and adaptive immune responses. However, the possible functional link between cocaine and autophagy has not been explored before. Herein, we demonstrate that cocaine exposure induced autophagy in both BV-2 and primary rat microglial cells as demonstrated by a dose- and time-dependent induction of autophagy-signature proteins such as BECN1/Beclin 1, ATG5, and MAP1LC3B. These findings were validated wherein cocaine treatment of BV-2 cells resulted in increased formation of puncta in cells expressing either endogenous MAP1LC3B or overexpressing GFP-MAP1LC3B. Specificity of cocaine-induced autophagy was confirmed by treating cells with inhibitors of autophagy (3-MA and wortmannin). Intriguingly, cocaine-mediated induction of autophagy involved upstream activation of 2 ER stress pathways (EIF2AK3- and ERN1-dependent), as evidenced by the ability of the ER stress inhibitor salubrinal to ameliorate cocaine-induced autophagy. In vivo validation of these findings demonstrated increased expression of BECN1, ATG5, and MAP1LC3B-II proteins in cocaine-treated mouse brains compared to untreated animals. Increased autophagy contributes to cocaine-mediated activation of microglia since pretreatment of cells with wortmannin resulted in decreased expression and release of inflammatory factors (TNF, IL1B, IL6, and CCL2) in microglial cells. Taken together, our findings suggest that cocaine exposure results in induction of autophagy that is closely linked with neuroinflammation. Targeting autophagic proteins could thus be considered as a therapeutic strategy for the treatment of cocaine-related neuroinflammation diseases.


Asunto(s)
Autofagia/efectos de los fármacos , Cocaína/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Microglía/metabolismo , Transducción de Señal/efectos de los fármacos , Adenina/análogos & derivados , Adenina/farmacología , Androstadienos/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 5 Relacionada con la Autofagia , Beclina-1 , Biomarcadores/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/ultraestructura , Modelos Biológicos , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Wortmanina
13.
J Immunol ; 186(7): 4119-29, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21368226

RESUMEN

HIV-associated neurologic disorders (HAND) are estimated to affect almost 60% of HIV-infected individuals. HIV encephalitis, the pathologic correlate of the most severe form of HAND, is often characterized by glial activation, cytokine-chemokine dysregulation, and neuronal damage and loss. However, the severity of HIV encephalitis correlates better with glial activation rather than viral load. Using the macaque model, it has been demonstrated that SIV encephalitis correlates with increased expression of the mitogen platelet-derived growth factor (PDGF) B chain in the brain. The goal of this study was to explore the role of PDGF-B chain in HIV-associated activation and proliferation of astrocytes. Specifically, the data demonstrate that exposure of rat and human astrocytes to the HIV-1 protein Tat resulted in the induction of PDGF at both the mRNA and protein levels. Furthermore, PDGF-BB induction was regulated by activation of ERK1/2 and JNK signaling pathways and the downstream transcription factor early growth response 1. Chromatin immunoprecipitation assays demonstrated binding of Egr-1 to the PDGF-B promoter. Exposure of astrocytes to PDGF-BB in turn led to increased proliferation and the release of proinflammatory cytokines MCP-1 and IL-1ß. Because astrogliosis is linked to disease severity, understanding its regulation by PDGF-BB could aid in the development of therapeutic intervention strategies for HAND.


Asunto(s)
Astrocitos/inmunología , Astrocitos/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología , Animales , Astrocitos/patología , Becaplermina , Línea Celular Tumoral , Proliferación Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , VIH-1/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Proto-Oncogénicas c-sis , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
14.
J Neuroimmune Pharmacol ; 6(3): 399-408, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21274647

RESUMEN

The current study was aimed at investigating the effect of HIV-1 protein Tat on the retinal neurosensory cell line R28. Exposure of Tat resulted in induction of pro-inflammatory mediators such as CXCL10 and TNF-α in addition to the activation marker GFAP in these cells. Conditioned media from Tat-treated R28 cells was able to induce monocyte migration, an effect that was blocked by CXCR3 antagonist. Complementary studies in the HIV-1 Tat-transgenic mice, showed a complete absence of the nuclear layer and the outer photoreceptor segments of the retina with a concomitant increase in glial activation. These findings lend support to the observation in post-HAART era of increased incidence of immune response-mediated retinal degeneration. These findings have direct relevance to diseases such as immune response uveitis and patients recovering from CMV retinitis.


Asunto(s)
Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/virología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL10/biosíntesis , Inmunohistoquímica , Ratones , Ratones Transgénicos , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/biosíntesis , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología
15.
AIDS ; 24(14): 2161-70, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20706108

RESUMEN

OBJECTIVE: HIV-associated nephropathy (HIVAN) is characterized by the development of glomerulosclerosis and is associated with glomerular epithelial cell proliferation. It has recently been shown that activation of the Notch signaling pathway in podocytes results in glomerulosclerosis and podocyte proliferation. To determine whether Notch signaling is involved in renal disorder associated with HIVAN, we evaluated the expression of Notch receptors in HIVAN. DESIGN: We evaluated the expression of the Notch signaling pathway using an HIV-transgenic (HIV-Tg) rat model of HIVAN, and biopsy samples from HIVAN and normal controls. METHODS: Paraffin sections and kidney lysates were used for immunohistochemistry, immunofluorescence and western blot analysis. RESULTS: A collapsing variant of glomerulosclerosis and focal segmental sclerosis was observed in HIV-Tg rats. Glomeruli of HIV-Tg rats demonstrated activation of Notch1 and Notch4, as determined by the presence of the intracellular domains. In addition, we observed increased expression of the Notch target protein, hairy enhancer of split homolog-1 in glomeruli of these animals. The expression of the Groucho homolog transducin-like enhancer protein 4, a Notch effector protein, and the homeodomain protein cut homeobox 1 were also significantly increased in glomeruli of HIV-Tg rats, and this was associated with decreased expression of the cyclin kinase inhibitor p27. Intriguingly, renal biopsy samples from HIVAN patients also showed upregulation of cleaved Notch1 and Notch4 in the glomeruli compared with the expression in normal kidneys. CONCLUSION: Our results demonstrate activation of Notch signaling pathway in HIVAN, thereby underscoring its role in disease pathogenesis.


Asunto(s)
Nefropatía Asociada a SIDA/patología , Infecciones por VIH/patología , Glomérulos Renales/patología , Podocitos/metabolismo , Receptores Notch/metabolismo , Animales , Proliferación Celular , Infecciones por VIH/genética , Inmunohistoquímica , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética
16.
Toxicol Appl Pharmacol ; 240(2): 286-91, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19576918

RESUMEN

The human immunodeficiency virus (HIV-1) protein Tat has been implicated in mediating neuronal apoptosis, one of the hallmark features of HIV-associated dementia (HAD). Mitigation of the toxic effects of Tat could thus be a potential mechanism for reducing HIV toxicity in the brain. In this study we demonstrated that Tat-induced neurotoxicity was abolished by NMDA antagonist-MK801, suggesting the role of glutamate in this process. Furthermore, we also found that pretreatment of SH-SY5Y cells with PDGF exerted protection against Tat toxicity by decreasing extracellular glutamate levels. We also demonstrated that extracellular calcium chelator EGTA was able to abolish PDGF-mediated neuroprotection, thereby underscoring the role of calcium signaling in PDGF-mediated neuroprotection. We also showed that Erk signaling pathway was critical for PDGF-mediated protection of cells. Additionally, blocking calcium entry with EGTA resulted in suppression of PDGF-induced Erk activation. These findings thus underscore the role of PDGF-mediated calcium signaling and Erk phosphorylation in the protection of cells against HIV Tat toxicity.


Asunto(s)
Apoptosis , Señalización del Calcio , Ácido Glutámico/metabolismo , VIH-1/metabolismo , Neuronas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Apoptosis/efectos de los fármacos , Becaplermina , Señalización del Calcio/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Quelantes/farmacología , Citoprotección , Maleato de Dizocilpina/farmacología , Ácido Egtácico/farmacología , Activación Enzimática , Antagonistas de Aminoácidos Excitadores/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Fosforilación , Proteínas Proto-Oncogénicas c-sis , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Tiempo
17.
Glia ; 57(7): 734-43, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18985732

RESUMEN

HIV encephalitis (HIVE), the pathologic correlate of HIV-associated dementia (HAD) is characterized by astrogliosis, cytokine/chemokine dysregulation, and neuronal degeneration. Increasing evidence suggests that inflammation is actively involved in the pathogenesis of HAD. In fact, the severity of HAD/HIVE correlates more closely with the presence of activated glial cells than with the presence and amount of HIV-infected cells in the brain. Astrocytes, the most numerous cell type within the brain, provide an important reservoir for the generation of inflammatory mediators, including interferon-gamma inducible peptide-10 (CXCL10), a neurotoxin and a chemoattractant, implicated in the pathophysiology of HAD. Additionally, the proinflammatory cytokines, IFN-gamma and TNF-alpha, are also markedly increased in CNS tissues during HIV-1 infection. In this study, we hypothesized that the interplay of host cytokines and HIV-1 could lead to enhanced expression of the toxic chemokine, CXCL10. Our findings demonstrate a synergistic induction of CXCL10 mRNA and protein in human astrocytes exposed to HIV-1 and the proinflammatory cytokines. Signaling molecules, including JAK, STATs, MAPK (via activation of Erk1/2, AKT, and p38), and NF-kappaB were identified as instrumental in the synergistic induction of CXCL10. Understanding the mechanisms involved in HIV-1 and cytokine-mediated up-regulation of CXCL10 could aid in the development of therapeutic modalities for HAD.


Asunto(s)
Astrocitos/metabolismo , Quimiocina CXCL10/metabolismo , Citocinas/metabolismo , VIH-1/fisiología , Astrocitos/inmunología , Astrocitos/virología , Western Blotting , Línea Celular , Quimiocina CXCL10/genética , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Humanos , Inmunohistoquímica , Interferón gamma/metabolismo , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Neurovirol ; 14(3): 196-204, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18569454

RESUMEN

Synergistic interactions between viral proteins and soluble host factors released from infected mononuclear phagocytes play a critical role in the pathogenesis of human immunodeficiency virus (HIV)-associated dementia (HAD). The chemokine CXCL10 has been found to be closely associated with the progression of HIV-1-related central nervous system (CNS) disease and its related neuropsychiatric impairment. In this report the authors demonstrate that the HIV-1 protein Tat can interact with the proinflammatory cytokine interferon (IFN)-gamma to dramatically induce the expression of CXCL10 in macrophages. Synergistic induction of CXCL10 by both Tat and IFN-gamma was susceptible to inhibition by the MEK1/2 inhibitor U0126 and the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580. In addition, JAK/STAT pathway plays a major role in Tat/gamma-mediated CXCL10 induction in macrophages because pretreatment of stimulated macrophages with JAK inhibitor completely abrogated the synergistic induction of the chemokine. Functionality of the synergistically induced CXCL10 was further demonstrated by its chemotactic activity for peripheral blood lymphocytes. Taken together, these findings demonstrate that the cooperative interaction of Tat and IFN-gamma results in enhanced chemokine expression, which in turn can amplify the inflammatory responses within the CNS of HAD patients by recruiting more lymphocytes in the brain.


Asunto(s)
Quimiocina CXCL10/metabolismo , VIH-1/metabolismo , Interferón gamma/farmacología , Macrófagos/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/virología , Células Cultivadas , Humanos , Quinasas Janus/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Front Biosci ; 13: 3913-8, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508485

RESUMEN

The encephalopathy caused by HIV, known clinically as HIV-associated dementia (HAD) and pathologically as HIV encephalitis (HIVE), results from intense infiltration of mononuclear cells, productive replication of the virus in monocyte-derived macrophages/microglia, abortive replication in astrocytes and activation of macrophages/microglia and astrocytes leading to neuronal degeneration in the brains of infected persons. Recent findings have suggested that development of HAD is based more on the activation process than on direct evidence of virus replication in the brain. Since HAD is based on the encephalitic process, major studies have been directed to the mechanisms regulating the inflammatory process. Monocyte chemoattractant protein 1, MCP-1, is a chemokine that is implicated in this process and also in the development of activation in the brain. In this review, we have attempted to identify mechanisms that induce expression of MCP-1 in the brain and the role that it plays in recruitment of mononuclear cells from blood to brain and in the activation processes of inflammatory and neural cells that lead to development of degenerative changes in the neuronal population.


Asunto(s)
Complejo SIDA Demencia/fisiopatología , Astrocitos/fisiología , Quimiocina CCL2/fisiología , Macrófagos/fisiología , Macrófagos/virología , Complejo SIDA Demencia/epidemiología , Barrera Hematoencefálica , VIH/patogenicidad , Humanos , Prevalencia
20.
J Neurovirol ; 14(1): 62-72, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18300076

RESUMEN

The human immunodeficiency virus (HIV)-1 envelope glycoprotein gp120 has been implicated in mediating neuronal apoptosis, a hallmark feature of HIV-associated dementia (HAD). Mitigation of the toxic effects of gp120 could thus be a potential mechanism for reducing HIV toxicity in the brain. In this study the authors hypothesized that neurotrophic factor, such as platelet-derived growth factor (PDGF), could protect the neurons against gp120-mediated apoptosis. SH-SY5Y cells treated with gp120 exhibited increased cell death when measured by lactate dehydrogenase (LDH) and deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL) assay, with concomitant loss of neurites and increased cell rounding. Pretreatment with PDGF-BB, however, reduced gp120-associated neurotoxicity and rescued the neurite outgrowth. Additionally, gp120-mediated activation of caspase-3 was also significantly reduced in cells pretreated with PDGF-BB. Antiapoptotic effects of PDGF-BB were also confirmed by monitoring levels of anti- and proapoptotic genes, Bcl-xL and Bax, respectively. Furthermore, PDGF-mediated protection against gp120 involved the phosphoinositide (PI) 3-kinase/Akt pathway. Taken together these findings lead us to suggest that PDGF-BB could be considered as a therapeutic agent that can mitigate gp120-mediated neurotoxicity in HAD.


Asunto(s)
Efecto Citopatogénico Viral/efectos de los fármacos , Proteína gp120 de Envoltorio del VIH/toxicidad , Neuronas/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Apoptosis/efectos de los fármacos , Becaplermina , Caspasa 3/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Línea Celular Tumoral/ultraestructura , Forma de la Célula/efectos de los fármacos , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Neuroblastoma/patología , Neuronas/metabolismo , Neuronas/ultraestructura , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Proto-Oncogénicas c-sis , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Proteína X Asociada a bcl-2/biosíntesis , Proteína X Asociada a bcl-2/genética , Proteína bcl-X/biosíntesis , Proteína bcl-X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA