Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 510
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731913

RESUMEN

Despite combined antiretroviral therapy (cART) limiting HIV replication to undetectable levels in the blood, people living with HIV continue to experience HIV-associated neurocognitive disorder (HAND). HAND is associated with neurocognitive impairment, including motor impairment, and memory loss. HIV has been detected in the brain within 8 days of estimated exposure and the mechanisms for this early entry are being actively studied. Once having entered into the central nervous system (CNS), HIV degrades the blood-brain barrier through the production of its gp120 and Tat proteins. These proteins are directly toxic to endothelial cells and neurons, and propagate inflammatory cytokines by the activation of immune cells and dysregulation of tight junction proteins. The BBB breakdown is associated with the progression of neurocognitive disease. One of the main hurdles for treatment for HAND is the latent pool of cells, which are insensitive to cART and prolong inflammation by harboring the provirus in long-lived cells that can reactivate, causing damage. Multiple strategies are being studied to combat the latent pool and HAND; however, clinically, these approaches have been insufficient and require further revisions. The goal of this paper is to aggregate the known mechanisms and challenges associated with HAND.


Asunto(s)
Barrera Hematoencefálica , Humanos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , Infecciones por VIH/patología , Infecciones por VIH/metabolismo , Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/patología , VIH-1 , Trastornos Neurocognitivos/etiología , Trastornos Neurocognitivos/metabolismo , Trastornos Neurocognitivos/patología , Animales
2.
Ann Neurol ; 96(2): 306-320, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38752697

RESUMEN

OBJECTIVE: Although accumulating evidence implicating altered gut microbiota in human immunodeficiency virus (HIV) infection and neurodegenerative disorders; however, the association between dysbiosis of the gut microbiota and metabolites in the pathogenesis of HIV-associated neurocognitive disorder (HAND) remains unclear. METHODS: Fecal and plasma samples were obtained from 3 cohorts (HAND, HIV-non-HAND, and healthy controls), metagenomic analysis and metabolomic profiling were performed to investigate alterations in the gut microbial composition and circulating metabolites in HAND. RESULTS: The gut microbiota of people living with HIV (PLWH) had an increased relative abundance of Prevotella and a decreased relative abundance of Bacteroides. In contrast, Prevotella and Megamonas were substantially decreased, and Bacteroides and Phocaeicola were increased in HAND patients. Moreover, untargeted metabolomics identified several neurotransmitters and certain amino acids associated with neuromodulation, and the differential metabolic pathways of amino acids associated with neurocognition were depleted in HAND patients. Notably, most neuromodulatory metabolites are associated with an altered abundance of specific gut bacteria. INTERPRETATION: Our findings provide new insights into the intricate interplay between the gut and microbiome-brain axis in the pathogenesis of HAND, highlighting the potential for developing novel therapeutic strategies that specifically target the gut microbiota. ANN NEUROL 2024;96:306-320.


Asunto(s)
Aminoácidos , Microbioma Gastrointestinal , Metabolómica , Metagenómica , Humanos , Microbioma Gastrointestinal/fisiología , Masculino , Persona de Mediana Edad , Femenino , Metabolómica/métodos , Aminoácidos/metabolismo , Aminoácidos/sangre , Adulto , Infecciones por VIH/complicaciones , Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/microbiología , Heces/microbiología , Disbiosis
3.
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
4.
Sci Rep ; 13(1): 18418, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37891420

RESUMEN

Human immunodeficiency virus encephalitis (HIVE) is a severe neurological complication after HIV infection. Evidence shows that genetic factors play an important role in HIVE. The aim of the present study was to identify new potential therapeutic targets for HIVE. Differentially expressed gene (DEG), functional annotation and pathway, and protein-protein interaction analyses were performed to identify the hub genes associated with HIVE. Gene co-expression analysis was carried out to confirm the association between the hub genes and HIVE. Finally, the role of the hub genes in HIVE therapy was evaluated by conducting drug-gene interaction analysis. A total of 20 overlapping DEGs closely related to HIVE were identified. Functional annotation and pathway enrichment analysis indicated that the markedly enriched DEG terms included ion transport, type II interferon signaling, and synaptic signaling. Moreover, protein-protein interaction analysis revealed that 10 key HIVE-related genes were hub genes, including SCN8A, CDK5R2, GRM5, SCN2B, IFI44L, STAT1, SLC17A7, ISG15, FGF12, and FGF13. Furthermore, six hub genes were co-expressed with HIVE-associated host genes in human brain tissue. Finally, three hub genes (STAT1, ISG15, and SCN2B) interacted with several inflammation-associated drugs. These findings suggested that SCN8A, CDK5R2, GRM5, SCN2B, IFI44L, STAT1, SLC17A7, ISG15, FGF12, and FGF13 may be new targets for diagnosis and therapy of HIVE.


Asunto(s)
Complejo SIDA Demencia , Encefalitis , Infecciones por VIH , Humanos , Infecciones por VIH/complicaciones , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Encefalitis/metabolismo , Encéfalo/metabolismo , Biomarcadores/metabolismo , Complejo SIDA Demencia/metabolismo , Perfilación de la Expresión Génica , Biología Computacional , Factores de Crecimiento de Fibroblastos/metabolismo
5.
Pharmacol Ther ; 234: 108047, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34848202

RESUMEN

As our understanding of changes to the neurological system has improved, it has become clear that patients who have contracted human immunodeficiency virus type 1 (HIV-1) can potentially suffer from a cascade of neurological issues, including neuropathy, dementia, and declining cognitive function. The progression from mild to severe symptoms tends to affect motor function, followed by cognitive changes. Central nervous system deficits that are observed as the disease progresses have been reported as most severe in later-stage HIV infection. Examining the full spectrum of neuronal damage, generalized cortical atrophy is a common hallmark, resulting in the death of multiple classes of neurons. With antiretroviral therapy (ART), we can partially control disease progression, slowing the onset of the most severe symptoms such as, reducing viral load in the brain, and developing HIV-associated dementia (HAD). HAD is a severe and debilitating outcome from HIV-related neuropathologies. HIV neurotoxicity can be direct (action directly on the neuron) or indirect (actions off-site that affect normal neuronal function). There are two critical HIV-associated proteins, Tat and gp120, which bear responsibility for many of the neuropathologies associated with HAD and HIV-associated neurocognitive disorder (HAND). A cascade of systems is involved in HIV-related neurotoxicity, and determining a critical point where therapeutic strategies can be employed is of the utmost importance. This review will provide an overview of the existing hypotheses on HIV-neurotoxicity and the potential for the development of therapeutics to aid in the treatment of HIV-related nervous system dysfunction.


Asunto(s)
Complejo SIDA Demencia , Disfunción Cognitiva , Infecciones por VIH , VIH-1 , Síndromes de Neurotoxicidad , Complejo SIDA Demencia/tratamiento farmacológico , Complejo SIDA Demencia/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , VIH-1/metabolismo , Humanos , Neuronas/metabolismo , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/etiología
6.
Brain ; 144(11): 3355-3370, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34196664

RESUMEN

HIV-associated neurocognitive disorders (HAND) in the era of combination antiretroviral therapy are primarily manifested as impaired behaviours, glial activation/neuroinflammation and compromised neuronal integrity, for which there are no effective treatments currently available. In the current study, we used doxycycline-inducible astrocyte-specific HIV Tat transgenic mice (iTat), a surrogate HAND model, and determined effects of PNU-125096, a positive allosteric modulator of α7 nicotinic acetylcholine receptor (α7 nAChR) on Tat-induced behavioural impairments and neuropathologies. We showed that PNU-125096 treatment significantly improved locomotor, learning and memory deficits of iTat mice while inhibited glial activation and increased PSD-95 expression in the cortex and hippocampus of iTat mice. Using α7 nAChR knockout mice, we showed that α7 nAChR knockout eliminated the protective effects of PNU-125096 on iTat mice. In addition, we showed that inhibition of p38 phosphorylation by SB239063, a p38 MAPK-specific inhibitor exacerbated Tat neurotoxicity in iTat mice. Last, we used primary mouse cortical individual cultures and neuron-astrocytes co-cultures and in vivo staining of iTat mouse brain tissues and showed that glial activation was directly involved in the interplay among Tat neurotoxicity, α7 nAChR activation and the p38 MAPK signalling pathway. Taken together, these findings demonstrated for the first time that α7 nAChR activation led to protection against HAND and suggested that α7 nAChR modulator PNU-125096 holds significant promise for development of therapeutics for HAND.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/toxicidad
7.
J Neuroimmunol ; 355: 577542, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33845284

RESUMEN

We hypothesized that humoral immunity stimulation in the CNS in HIV-1C patients would be lower than that in HIV-1B due to a defective Tat chemokine dimotif (C30C31) that might influence cellular trafficking and CNS inflammation. Sixty-eight paired CSF and blood samples from people with HIV (PWH), free of CNS opportunistic infections, were included, HIV-1B (n = 27), HIV-1C (n = 26), and HIV negative (n = 25). IgG intrathecal synthesis was assayed using quantitative and qualitative methods. IgG oligoclonal bands (OCB) in CSF were observed in 51% of PWH, comparable between HIV-1B and HIV-1C, as well as the medians of IgG intrathecal synthesis formulas. The group with HIV infection aviremic in CSF and blood showed 75% of OCB. There was a poor positive correlation between the IgG quotient and GDS. The impact of HIV-1 on IgG intrathecal production was not subtype dependent. Low-grade CNS intrathecal IgG production persists in HIV CNS infection even in PWH with CSF and blood HIV RNA controlled.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Inmunoglobulina G/líquido cefalorraquídeo , ARN Viral/metabolismo , Complejo SIDA Demencia/diagnóstico , Adulto , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Estudios Transversales , Femenino , Infecciones por VIH/diagnóstico , Humanos , Inmunoglobulina G/biosíntesis , Masculino , Persona de Mediana Edad , Punción Espinal
8.
Autophagy ; 17(7): 1768-1782, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33890542

RESUMEN

Despite the promising therapeutic effects of combinatory antiretroviral therapy (cART), 20% to 30% of HIV/AIDS patients living with long term infection still exhibit related cognitive and motor disorders. Clinical studies in HIV-infected patients revealed evidence of basal ganglia dysfunction, tremors, fine motor movement deficits, gait, balance, and increased risk of falls. Among older HIV+ adults, the frequency of cases with SNCA/α-synuclein staining is higher than in older healthy persons and may predict an increased risk of developing a neurodegenerative disease. The accumulation of SNCA aggregates known as Lewy Bodies is widely described to be directly linked to motor dysfunction. These aggregates are naturally removed by Macroautophagy/autophagy, a cellular housekeeping mechanism, that can be disturbed by HIV-1. The molecular mechanisms involved in linking HIV-1 proteins and autophagy remain mostly unclear and necessitates further exploration. We showed that HIV-1 Vpr protein triggers the accumulation of SNCA in neurons after decreasing lysosomal acidification, deregulating lysosome positioning, and the expression levels of several proteins involved in lysosomal maturation. Viruses and retroviruses such as HIV-1 are known to manipulate autophagy in order to use it for their replication while blocking the degradative final step, which could destroy the virus itself. Our study highlights how the suppression of neuronal autophagy by HIV-1 Vpr is a mechanism leading to toxic protein aggregation and neurodegeneration.Abbreviations: BLOC1: Biogenesis of Lysosome-related Organelles Complex 1; CART: combinatory antiretroviral therapy; CVB: coxsackievirus; DAPI: 4',6-diamidino-2-phenylindole; DENV: dengue virus; GFP: green fluorescent protein; HCV: hepatitis C virus; HCMV: human cytomegalovirus; HIV: human immunodeficiency virus; Env: HIV-1 envelope glycoproteins; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; VSV: Indiana vesiculovirus; LTR: Long Terminal Repeat; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MLBs: multilamellar bodies; RIPA: Radioimmunoprecipitation assay buffer; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; Tat: transactivator of TAR; TEM: transmission electron microscope; Vpr: Viral protein R.


Asunto(s)
Complejo SIDA Demencia/etiología , Lisosomas/virología , Neuronas/virología , alfa-Sinucleína/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/patología , Animales , Autofagosomas/virología , Western Blotting , Encéfalo/patología , Encéfalo/virología , Técnica del Anticuerpo Fluorescente , VIH-1 , Humanos , Lisosomas/fisiología , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Neuronas/metabolismo , Neuronas/fisiología
9.
J Neurovirol ; 27(2): 279-301, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33646495

RESUMEN

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.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Astrocitos/virología , Quimiocina CX3CL1/biosíntesis , Astrocitos/inmunología , Astrocitos/metabolismo , Células Cultivadas , VIH-1 , Humanos
10.
Neurosci Lett ; 750: 135717, 2021 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-33587986

RESUMEN

In the era of combined antiretroviral therapy (cART), human immunodeficiency virus type 1 (HIV-1) is considered a chronic disease with an inflammatory component that specifically targets the brain and causes a high prevalence of HIV-1-associated neurocognitive disorders (HAND). The endocannabinoid (eCB) system has attracted interest as a target for treatment of neurodegenerative disorders, due to the potential anti-inflammatory and neuroprotective properties of cannabinoids, including its potential therapeutic use in HIV-1 neuropathogenesis. In this review, we summarize what is currently known about the structural and functional changes of the eCB system under conditions of HAND. This will be followed by summarizing the current clinical and preclinical findings on the effects of cannabis use and cannabinoids in the context of HIV-1 infection, with specifically focusing on viral load, cognition, inflammation, and neuroprotection. Lastly, we present some potential future directions to better understand the involvement of the eCB system and the role that cannabis use and cannabinoids play in neuroHIV.


Asunto(s)
Complejo SIDA Demencia/tratamiento farmacológico , Cannabinoides/uso terapéutico , Complejo SIDA Demencia/metabolismo , Animales , Cannabinoides/metabolismo , Humanos , Receptor Cannabinoide CB2/metabolismo
11.
Neuromolecular Med ; 23(1): 25-46, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33547562

RESUMEN

Neuro-inflammation accompanies numerous neurological disorders and conditions where it can be associated with a progressive neurodegenerative pathology. In a similar manner, alterations in sphingolipid metabolism often accompany or are causative features in degenerative neurological conditions. These include dementias, motor disorders, autoimmune conditions, inherited metabolic disorders, viral infection, traumatic brain and spinal cord injury, psychiatric conditions, and more. Sphingolipids are major regulators of cellular fate and function in addition to being important structural components of membranes. Their metabolism and signaling pathways can also be regulated by inflammatory mediators. Therefore, as certain sphingolipids exert distinct and opposing cellular roles, alterations in their metabolism can have major consequences. Recently, regulation of bioactive sphingolipids by neuro-inflammatory mediators has been shown to activate a neuronal NADPH oxidase 2 (NOX2) that can provoke damaging oxidation. Therefore, the sphingolipid-regulated neuronal NOX2 serves as a mechanistic link between neuro-inflammation and neurodegeneration. Moreover, therapeutics directed at sphingolipid metabolism or the sphingolipid-regulated NOX2 have the potential to alleviate neurodegeneration arising out of neuro-inflammation.


Asunto(s)
NADPH Oxidasa 2/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Esfingolípidos/fisiología , Complejo SIDA Demencia/metabolismo , Animales , Productos Biológicos/uso terapéutico , Encefalopatías Metabólicas Innatas/genética , Encefalopatías Metabólicas Innatas/metabolismo , Encefalopatías Metabólicas Innatas/terapia , Descubrimiento de Drogas , Encefalitis Viral/metabolismo , Activación Enzimática , Terapia de Reemplazo Enzimático , Humanos , Inflamación , Naftalenos/uso terapéutico , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/terapia , Neuronas/metabolismo , Oxidación-Reducción , Pirimidinonas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Infección por el Virus Zika/metabolismo
12.
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
13.
J Acquir Immune Defic Syndr ; 86(4): 496-499, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33273212

RESUMEN

BACKGROUND: Systemic immune activation and inflammation in chronic HIV infection are driving factors of non-AIDS-related events, including neurocognitive impairment. The role of inflammasome in monocytes from patients with HIV infection has been extensively studied, but its association with the extent of neurocognitive dysfunction has been poorly investigated. METHODS: We enrolled 79 HIV-positive patients; 44 with varying levels of HIV-associated neurocognitive disorder (HAND) and 35 without and 8 healthy donors. HAND subtypes included asymptomatic neurocognitive impairment (asymptomatic neurocognitive impairment; n = 19), mild neurocognitive disorder (MND; n = 17), and HIV-associated dementia (n = 8). We quantified plasmatic concentrations of proinflammatory cytokines (TNF-α, IL-6, IL-17A, IL-1ß, and IFN-γ) for all HIV patients, and the mRNA expression of genes involved in the inflammasome activity (NLRP3, PYCARD, NAIP, AIM2, IL-1ß, and IL-18) in monocytes of a subgroup of 28 HIV patients and 8 healthy donors. RESULTS: HIV patients' plasma concentrations of IFN-γ, IL-1ß, and IL-17A were undetectable. Levels of TNF-α and IL-6 were similar among the HIV patient groups. A trend toward an increased expression of inflammasome genes according to neurocognitive disorder severity was observed. Of note, the NLRP3 mRNA relative expression was higher in MND compared with other groups, and IL-1ß was lower in MND than HIV-associated dementia patients. CONCLUSIONS: Changes in inflammasome components in circulating monocytes according to different HAND severity suggest that NLRP3 may be a possible biomarker or target to better understand and treat the link between systemic inflammation and neurocognitive impairment in HIV infection.


Asunto(s)
Infecciones por VIH/complicaciones , VIH-1 , Interleucina-1beta/metabolismo , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Trastornos Neurocognitivos/metabolismo , Complejo SIDA Demencia/metabolismo , Adulto , Fármacos Anti-VIH/uso terapéutico , Femenino , Regulación de la Expresión Génica , Infecciones por VIH/tratamiento farmacológico , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Masculino , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Carga Viral
14.
Neurochem Int ; 141: 104880, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33065212

RESUMEN

The dysregulation of lipid homeostasis is emerging as a hallmark of many CNS diseases. As aberrant protein regulation is suggested to be a shared pathological feature amongst many neurodegenerative conditions, such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), disruptions in neuronal lipid processing may contribute to disease progression in the CNS. Specifically, given the endoplasmic reticulum (ER) dual role in lipid homeostasis as well as protein quality control (PQC) via unfolded protein response (UPR), lipid dysregulation in the CNS may converge on ER functioning and constitute a crucial mechanism underlying aberrant protein aggregation. In the current review, we discuss the diverse roles of lipid species as essential components of the CNS. Moreover, given the importance of both lipid dysregulation and protein aggregation in pathology of CNS diseases, we attempt to assess the potential downstream cross-talk between lipid dysregulation and ER dependent PQC mechanisms, with special focus on HIV-associated neurodegenerative disorders (HAND).


Asunto(s)
Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/fisiopatología , Estrés del Retículo Endoplásmico , Infecciones por VIH/fisiopatología , Metabolismo de los Lípidos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Animales , VIH-1 , Humanos , Respuesta de Proteína Desplegada
15.
J Neuroinflammation ; 17(1): 226, 2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32727588

RESUMEN

BACKGROUND: HIV-1 infection remains a major public health concern despite effective combination antiretroviral therapy (cART). The virus enters the central nervous system (CNS) early in infection and continues to cause HIV-associated neurocognitive disorders (HAND). The pathogenic mechanisms of HIV-associated brain injury remain incompletely understood. Since HIV-1 activates the type I interferon system, which signals via interferon-α receptor (IFNAR) 1 and 2, this study investigated the potential role of IFNAR1 in HIV-induced neurotoxicity. METHODS: We cross-bred HIVgp120-transgenic (tg) and IFNAR1 knockout (IFNAR1KO) mice. At 11-14 months of age, we performed a behavioral assessment and subsequently analyzed neuropathological alterations using deconvolution and quantitative immunofluorescence microscopy, quantitative RT-PCR, and bioinformatics. Western blotting of brain lysates and an in vitro neurotoxicity assay were employed for analysis of cellular signaling pathways. RESULTS: We show that IFNAR1KO results in partial, sex-dependent protection from neuronal injury and behavioral deficits in a transgenic model of HIV-induced brain injury. The IFNAR1KO rescues spatial memory and ameliorates loss of presynaptic terminals preferentially in female HIVgp120tg mice. Similarly, expression of genes involved in neurotransmission reveals sex-dependent effects of IFNAR1KO and HIVgp120. In contrast, IFNAR1-deficiency, independent of sex, limits damage to neuronal dendrites, microgliosis, and activation of p38 MAPK and restores ERK activity in the HIVgp120tg brain. In vitro, inhibition of p38 MAPK abrogates neurotoxicity caused similarly by blockade of ERK kinase and HIVgp120. CONCLUSION: Our findings indicate that IFNAR1 plays a pivotal role in both sex-dependent and independent processes of neuronal injury and behavioral impairment triggered by HIV-1.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/patología , Encéfalo/patología , Neuronas/patología , Receptor de Interferón alfa y beta/metabolismo , Animales , Encéfalo/metabolismo , Femenino , Proteína gp120 de Envoltorio del VIH , VIH-1 , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo
16.
J Neuroimmune Pharmacol ; 15(3): 387-389, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32696265

RESUMEN

Extracellular vesicles (EVs) are known to perform important biological functions and have been implicated in multiple disease pathogeneses, including HIV and drugs of abuse. EVs can carry biological molecules via biofluids such as plasma and cerebrospinal fluids (CSF) from healthy or disease organs to distant organs and deliver biomolecules to recipient cells that subsequently alter the physiology of the recipient organs. As biocarriers, EVs have the potential to be developed as non-invasive biomarkers for disease pathogenesis and drug abuse, as the level of specific EV components can be altered under disease/drug abuse conditions. Since many drugs don't cross the blood-brain barrier, EVs have shown the potential to encapsulate small drug molecules, including nucleotides, and carry these drugs to brain cells and enhance brain drug bioavailability. Through this special issue, we have covered several studies related to the role of EVs in altering biological functions via cell-cell interactions in healthy, HIV, and drug of abuse conditions. We have also included studies on the role of EVs as potential biomarkers for HIV pathogenesis and drugs of abuse. Further, the potential role of EVs in drug delivery in the CNS for diseases, including HIV-associated neurocognitive disorders and other neurological disorders, are covered in this issue.


Asunto(s)
Vesículas Extracelulares/metabolismo , Infecciones por VIH/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/patología , Biomarcadores , Barrera Hematoencefálica , Comunicación Celular , Sistemas de Liberación de Medicamentos , Vesículas Extracelulares/patología , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Humanos , Trastornos Relacionados con Sustancias/patología
17.
Brain Behav Immun ; 89: 480-490, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32717399

RESUMEN

The incidence of infectious diseases affecting the central nervous system (CNS) has been increasing over the last several years. Among the reasons for the expansion of these diseases and the appearance of new neuropathogens are globalization, global warming, and the increased proximity between humans and wild animals due to human activities such as deforestation. Neurotropism affecting normal brain function is shared by organisms such as viruses, bacteria, fungi, and parasites. Neuroinfections caused by these agents activate immune responses, inducing neuroinflammation, excitotoxicity, and neurodegeneration. Purinergic signaling is an evolutionarily conserved signaling pathway associated with these neuropathologies. During neuroinfections, host cells release ATP as an extracellular danger signal with pro-inflammatory activities. ATP is metabolized to its derivatives by ectonucleotidases such as CD39 and CD73; ATP and its metabolites modulate neuronal and immune mechanisms through P1 and P2 purinergic receptors that are involved in pathophysiological mechanisms of neuroinfections. In this review we discuss the beneficial or deleterious effects of various components of the purinergic signaling pathway in infectious diseases that affect the CNS, including human immunodeficiency virus (HIV-1) infection, herpes simplex virus type 1 (HSV-1) infection, bacterial meningitis, sepsis, cryptococcosis, toxoplasmosis, and malaria. We also provide a description of this signaling pathway in emerging viral infections with neurological implications such as Zika and SARS-CoV-2.


Asunto(s)
Infecciones del Sistema Nervioso Central/metabolismo , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2X/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Complejo SIDA Demencia/metabolismo , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/metabolismo , Encefalitis por Herpes Simple/metabolismo , Humanos , Malaria/metabolismo , Meningitis Bacterianas/metabolismo , Meningitis Criptocócica/metabolismo , Pandemias , Neumonía Viral/metabolismo , SARS-CoV-2 , Sepsis/metabolismo , Transducción de Señal , Toxoplasmosis Cerebral/metabolismo , Infección por el Virus Zika/metabolismo
18.
J Neuroimmune Pharmacol ; 15(4): 715-728, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32533296

RESUMEN

The fronto-striatal circuitry, involving the nucleus accumbens, ventral tegmental area, and prefrontal cortex, mediates goal-directed behavior and is targeted by both drugs of abuse and HIV-1 infection. Acutely, both drugs and HIV-1 provoke increased dopamine activity within the circuit. However, chronic exposure to drugs or HIV-1 leads to dysregulation of the dopamine system as a result of fronto-striatal adaptations to oppose the effects of repeated instances of transiently increased dopamine. Specifically, chronic drug use leads to reduced dopaminergic tone, upregulation of dopamine transporters, and altered circuit connectivity, sending users into an allosteric state in which goal-directed behaviors are dysregulated (i.e., addiction). Similarly, chronic exposure to HIV-1, even with combination antiretroviral therapy (cART), dysregulates dopamine and dopamine transporter function and alters connectivity of the fronto-striatal circuit, contributing to apathy and clinical symptoms of HIV-1 associated neurocognitive disorders (HAND). Thus, in a drug user also exposed to HIV-1, dysregulation of the fronto-striatal dopamine circuit advances at an exacerbated rate and appears to be driven by mechanisms unique from those seen with chronic drug use or HIV-1 exposure alone. We posit that the effects of drug use and HIV-1 infection on microglia interact to drive the progression of motivational dysfunction at an accelerated rate. The current review will therefore explore how the fronto-striatal circuit adapts to drug use (using cocaine as an example), HIV-1 infection, and both together; emphasizing proper methods and providing future directions to develop treatments for pathologies disrupting goal-directed behaviors and improve clinical outcomes for affected patients. Graphical Abstract Drug use and HIV-1 in the fronto-striatal circuit. Drugs of abuse and HIV-1 infection both target the fronto-striatal circuit which mediates goal-directed behavior. Acutely, drugs and HIV-1 increase dopamine activity; in contrast chronic exposure produces circuit adaptions leading to dysregulation, addiction and/or apathy. Comorbid drug use and HIV-1 infection may interact with microglia to exacerbate motivational dysregulation.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Cuerpo Estriado/metabolismo , Corteza Prefrontal/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Complejo SIDA Demencia/epidemiología , Complejo SIDA Demencia/psicología , Analgésicos Opioides/efectos adversos , Animales , Conducta Adictiva/epidemiología , Conducta Adictiva/metabolismo , Conducta Adictiva/psicología , Enfermedad Crónica , Cuerpo Estriado/efectos de los fármacos , Dopamina/metabolismo , Infecciones por VIH/epidemiología , Infecciones por VIH/metabolismo , Infecciones por VIH/psicología , Humanos , Red Nerviosa/efectos de los fármacos , Red Nerviosa/metabolismo , Trastornos Neurocognitivos/epidemiología , Trastornos Neurocognitivos/metabolismo , Trastornos Neurocognitivos/psicología , Corteza Prefrontal/efectos de los fármacos , Trastornos Relacionados con Sustancias/epidemiología , Trastornos Relacionados con Sustancias/psicología
19.
J Neuroimmune Pharmacol ; 15(4): 729-742, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32506353

RESUMEN

Antiretroviral therapy (ART) has transformed HIV into a chronic condition, lengthening and improving the lives of individuals living with this virus. Despite successful suppression of HIV replication, people living with HIV (PLWH) are susceptible to a growing number of comorbidities, including neuroHIV that results from infection of the central nervous system (CNS). Alterations in the dopaminergic system have long been associated with HIV infection of the CNS. Studies indicate that changes in dopamine concentrations not only alter neurotransmission, but also significantly impact the function of immune cells, contributing to neuroinflammation and neuronal dysfunction. Monocytes/macrophages, which are a major target for HIV in the CNS, are responsive to dopamine. Therefore, defining more precisely the mechanisms by which dopamine acts on these cells, and the changes in cellular function elicited by this neurotransmitter are necessary to develop therapeutic strategies to treat neuroHIV. This is especially important for vulnerable populations of PLWH with chemically altered dopamine concentrations, such as individuals with substance use disorder (SUD), or aging individuals using dopamine-altering medications. The specific neuropathologic and neurocognitive consequences of increased CNS dopamine remain unclear. This is due to the complex nature of HIV neuropathogenesis, and logistical and technical challenges that contribute to inconsistencies among cohort studies, animal models and in vitro studies, as well as lack of demographic data and access to human CNS samples and cells. This review summarizes current understanding of the impact of dopamine on HIV neuropathogenesis, and proposes new experimental approaches to examine the role of dopamine in CNS HIV infection. Graphical abstract HIV Neuropathogenesis in the Presence of a Disrupted Dopamine System. Both substance abuse disorders and the use of dopaminergic medications for age-related diseases are associated with changes in CNS dopamine concentrations and dopaminergic neurotransmission. These changes can lead to aberrant immune function, particularly in myeloid cells, which contributes to the neuroinflammation, neuropathology and dysfunctional neurotransmission observed in dopamine-rich regions in HIV+ individuals. These changes, which are seen despite the use antiretroviral therapy (ART), in turn lead to further dysregulation of the dopamine system. Thus, in individuals with elevated dopamine, the bi-directional interaction between aberrant dopaminergic neurotransmission and HIV infection creates a feedback loop contributing to HIV associated neurocognitive dysfunction and neuroHIV. However, the distinct contributions and interactions made by HIV infection, inflammatory mediators, ART, drugs of abuse, and age-related therapeutics are poorly understood. Defining more precisely the mechanisms by which these factors influence the development of neurological disease is critical to addressing the continued presence of neuroHIV in vulnerable populations, such as HIV-infected older adults or drug abusers. Due to the complexity of this system, understanding these effects will require a combination of novel experimental modalities in the context of ART. These will include more rigorous epidemiological studies, relevant animal models, and in vitro cellular and molecular mechanistic analysis.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Antirretrovirales/metabolismo , Dopamina/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Complejo SIDA Demencia/tratamiento farmacológico , Complejo SIDA Demencia/epidemiología , Animales , Antirretrovirales/farmacología , Antirretrovirales/uso terapéutico , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/epidemiología , Infecciones por VIH/metabolismo , Humanos , Trastornos Relacionados con Sustancias/epidemiología
20.
J Neuroimmune Pharmacol ; 15(4): 780-793, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32409991

RESUMEN

HIV infection affects an estimated 38 million people. Approximately 50% of HIV patients exhibit neurocognitive dysfunction termed HIV-Associated Neurocognitive Disorder (HAND). HAND is a consequence of chronic low-level neuroinflammation due to HIV entry into the brain. Initially, monocytes become activated in circulation and traffic to the brain. Monocytes, when activated, become susceptible to infection by HIV and can then carry the virus across the blood brain barrier. Once in the brain, activated monocytes secrete chemokines, which recruit virus-specific CD8+ T cells into the brain to further promote neuroinflammation. HAND is closely linked to systemic inflammation driven, in part, by HIV but is also due to persistent translocation of microorganisms across the GI tract. Persistent anti-viral responses in the GI tract compromise microbial barrier integrity. Indeed, HIV patients can exhibit remarkably high levels of activated (CD16+) monocytes in circulation. Recent studies, including our own, show that HIV patients using medical marijuana exhibit lower levels of circulating CD16+ monocytes than non-cannabis using HIV patients. Cannabis is a known immune modulator, including anti-inflammatory properties, mediated, in part, by ∆9-tetrahydrocannabinol (THC), as well as less characterized minor cannabinoids, such as cannabidiol (CBD), terpenes and presumably other cannabis constituents. The immune modulating activity of THC is largely mediated through cannabinoid receptors (CB) 1 and 2, with CB1 also responsible for the psychotropic properties of cannabis. Here we discuss the anti-inflammatory properties of cannabinoids in the context of HIV and propose CB2 as a putative therapeutic target for the treatment of neuroinflammation. Graphical Abstract HIV-associated neurocognitive disorder is a systemic inflammatory disease leading to activation of plasmacytoid dendritic cells, monocytes and T cells. Monocyte and CD8 T cell migration across the BBB and interaction with astrocytes promotes neurotoxic inflammatory mediators release. CB2 ligands are proposed as therapeutics capable of suppressing systemic and localized inflammation.


Asunto(s)
Complejo SIDA Demencia/tratamiento farmacológico , Cannabinoides/administración & dosificación , Sistemas de Liberación de Medicamentos/tendencias , Mediadores de Inflamación/antagonistas & inhibidores , Leucocitos/efectos de los fármacos , Receptor Cannabinoide CB2/agonistas , Complejo SIDA Demencia/metabolismo , Animales , Antiinflamatorios/administración & dosificación , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Dronabinol/administración & dosificación , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Leucocitos/metabolismo , Receptor Cannabinoide CB2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...