Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
Viruses ; 16(5)2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38793665

RESUMEN

Epidemiologic studies have established that mpox (formerly known as monkeypox) outbreaks worldwide in 2022-2023, due to Clade IIb mpox virus (MPXV), disproportionately affected gay, bisexual, and other men who have sex with men. More than 35% and 40% of the mpox cases suffer from co-infection with HIV and sexually transmitted infections (STIs) (e.g., Chlamydia trachomatis, Neisseria gonorrhoeae, Treponema pallidum, and herpes simplex virus), respectively. Bacterial superinfection can also occur. Co-infection of MPXV and other infectious agents may enhance disease severity, deteriorate outcomes, elongate the recovery process, and potentially contribute to the morbidity and mortality of the ensuing diseases. However, the interplays between MPXV and HIV, bacteria, other STI pathogens and host cells are poorly studied. There are many open questions regarding the impact of co-infections with HIV, STIs, or bacterial superinfections on the diagnosis and treatment of MPXV infections, including clinical and laboratory-confirmed mpox diagnosis, suboptimal treatment effectiveness, and induction of antiviral drug resistance. In this review article, we will discuss the progress and knowledge gaps in MPXV biology, antiviral therapy, pathogenesis of human MPXV and its co-infection with HIV, STIs, or bacterial superinfections, and the impact of the co-infections on the diagnosis and treatment of mpox disease. This review not only sheds light on the MPXV infection and co-infection of other etiologies but also calls for more research on MPXV life cycles and the molecular mechanisms of pathogenesis of co-infection of MPXV and other infectious agents, as well as research and development of a novel multiplex molecular testing panel for the detection of MPXV and other STI co-infections.


Asunto(s)
Coinfección , Infecciones por VIH , Enfermedades de Transmisión Sexual , Humanos , Masculino , Coinfección/microbiología , Coinfección/virología , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , Monkeypox virus , Mpox/virología , Enfermedades de Transmisión Sexual/microbiología , Enfermedades de Transmisión Sexual/virología , Enfermedades de Transmisión Sexual/complicaciones , Sobreinfección/microbiología , Sobreinfección/virología , Femenino
2.
J Neuroinflammation ; 21(1): 127, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741181

RESUMEN

HIV-associated neurocognitive disorders (HAND) are a spectrum of cognitive impairments that continue to affect approximately half of all HIV-positive individuals despite effective viral suppression through antiretroviral therapy (ART). White matter pathologies have persisted in the ART era, and the degree of white matter damage correlates with the degree of neurocognitive impairment in patients with HAND. The HIV protein Nef has been implicated in HAND pathogenesis, but its effect on white matter damage has not been well characterized. Here, utilizing in vivo, ex vivo, and in vitro methods, we demonstrate that Nef-containing extracellular vesicles (Nef EVs) disrupt myelin sheaths and inflict damage upon oligodendrocytes within the murine central nervous system. Intracranial injection of Nef EVs leads to reduced myelin basic protein (MBP) staining and a decreased number of CC1 + oligodendrocytes in the corpus callosum. Moreover, cerebellar slice cultures treated with Nef EVs exhibit diminished MBP expression and increased presence of unmyelinated axons. Primary mixed brain cultures and enriched oligodendrocyte precursor cell cultures exposed to Nef EVs display a decreased number of O4 + cells, indicative of oligodendrocyte impairment. These findings underscore the potential contribution of Nef EV-mediated damage to oligodendrocytes and myelin maintenance in the pathogenesis of HAND.


Asunto(s)
Vesículas Extracelulares , VIH-1 , Ratones Endogámicos C57BL , Oligodendroglía , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Animales , Oligodendroglía/metabolismo , Oligodendroglía/patología , Oligodendroglía/virología , Ratones , Vesículas Extracelulares/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , VIH-1/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Células Cultivadas , Humanos , Masculino
4.
FASEB J ; 37(12): e23301, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37942865

RESUMEN

HIV-associated neurocognitive disorders (HAND) is a term describing a complex set of cognitive impairments accompanying HIV infection. Successful antiretroviral therapy (ART) reduces the most severe forms of HAND, but milder forms affect over 50% of people living with HIV (PLWH). Pathogenesis of HAND in the ART era remains unknown. A variety of pathogenic factors, such as persistent HIV replication in the brain reservoir, HIV proteins released from infected brain cells, HIV-induced neuroinflammation, and some components of ART, have been implicated in driving HAND pathogenesis in ART-treated individuals. Here, we propose another factor-impairment of cholesterol homeostasis and lipid rafts by HIV-1 protein Nef-as a possible contributor to HAND pathogenesis. These effects of Nef on cholesterol may also underlie the effects of other pathogenic factors that constitute the multifactorial nature of HAND pathogenesis. The proposed Nef- and cholesterol-focused mechanism may provide a long-sought unified explanation of HAND pathogenesis that takes into account all contributing factors. Evidence for the impairment by Nef of cellular cholesterol balance, potential effects of this impairment on brain cells, and opportunities to therapeutically target this element of HAND pathogenesis are discussed.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/patología , VIH-1/metabolismo , Encéfalo/metabolismo , Colesterol/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/uso terapéutico
6.
Cell Rep ; 41(8): 111674, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36417867

RESUMEN

A possible explanation for chronic inflammation in HIV-infected individuals treated with anti-retroviral therapy is hyperreactivity of myeloid cells due to a phenomenon called "trained immunity." Here, we demonstrate that human monocyte-derived macrophages originating from monocytes initially treated with extracellular vesicles containing HIV-1 protein Nef (exNef), but differentiating in the absence of exNef, release increased levels of pro-inflammatory cytokines after lipopolysaccharide stimulation. This effect is associated with chromatin changes at the genes involved in inflammation and cholesterol metabolism pathways and upregulation of the lipid rafts and is blocked by methyl-ß-cyclodextrin, statin, and an inhibitor of the lipid raft-associated receptor IGF1R. Bone-marrow-derived macrophages from exNef-injected mice, as well as from mice transplanted with bone marrow from exNef-injected animals, produce elevated levels of tumor necrosis factor α (TNF-α) upon stimulation. These phenomena are consistent with exNef-induced trained immunity that may contribute to persistent inflammation and associated co-morbidities in HIV-infected individuals with undetectable HIV load.


Asunto(s)
Vesículas Extracelulares , Infecciones por VIH , Seropositividad para VIH , VIH-1 , Humanos , Ratones , Animales , VIH-1/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Vesículas Extracelulares/metabolismo , Macrófagos/metabolismo , Inflamación/metabolismo
7.
Front Immunol ; 13: 903884, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35874772

RESUMEN

Findings that certain infections induce immunity not only against the causing agent, but also against an unrelated pathogen have intrigued investigators for many years. Recently, underlying mechanisms of this phenomenon have started to come to light. It was found that the key cells responsible for heterologous protection are innate immune cells such as natural killer cells (NKs), dendritic cells, and monocytes/macrophages. These cells are 'primed' by initial infection, allowing them to provide enhanced response to subsequent infection by the same or unrelated agent. This phenomenon of innate immune memory was termed 'trained immunity'. The proposed mechanism for trained immunity involves activation by the first stimulus of metabolic pathways that lead to epigenetic changes, which maintain the cell in a "trained" state, allowing enhanced responses to a subsequent stimulus. Innate immune memory can lead either to enhanced responses or to suppression of subsequent responses ('tolerance'), depending on the strength and length of the initial stimulation of the immune cells. In the context of HIV infection, innate memory induced by infection is not well understood. In this Hypothesis and Theory article, we discuss evidence for HIV-induced trained immunity in human monocytes, its possible mechanisms, and implications for HIV-associated co-morbidities.


Asunto(s)
Infecciones por VIH , Inmunidad Innata , Humanos , Tolerancia Inmunológica , Células Asesinas Naturales , Monocitos
9.
Mol Neurobiol ; 59(2): 1088-1097, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34843091

RESUMEN

HIV-associated neurocognitive disorders (HAND) is a term used to describe a variety of neurological impairments observed in HIV-infected individuals. The pathogenic mechanisms of HAND and of its connection to HIV infection remain unknown, but one of the considered hypotheses suggests that HIV infection accelerates the development of Alzheimer's disease. Previous studies suggested that HIV-1 Nef may contribute to HAND by inhibiting cholesterol efflux, increasing the abundance of lipid rafts, and affecting their functionality. Our comparative analysis of postmortem brain samples demonstrated a trend toward the decreased abundance of cholesterol transporter ABCA1 in samples from HIV-infected ART-treated individuals relative to samples from uninfected controls, and a reverse correlation between ABCA1 and flotillin 1, a marker for lipid rafts, in all analyzed samples. The brain samples from HIV-infected individuals, both with and without HAND, were characterized by the increased abundance of p-Tau217 peptide, which correlated with the abundance of flotillin 1. HIV-1 Nef was analyzed in samples from HAND-affected individuals by Western blot with 4 different antibodies and by LC-MS/MS, producing a Nef-positivity score. A significant correlation was found between this score and the abundance of flotillin 1, the abundance of p-Tau217, and the severity of HAND. These results highlight the contribution of Nef and Nef-dependent impairment of cholesterol efflux to HAND pathogenesis and support a connection between the pathogenesis of HAND and Alzheimer's disease.


Asunto(s)
Infecciones por VIH , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Proteínas tau , Encéfalo/metabolismo , Cromatografía Liquida , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Humanos , Trastornos Neurocognitivos/complicaciones , Espectrometría de Masas en Tándem , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas tau/metabolismo
10.
Comput Struct Biotechnol J ; 19: 3876-3884, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34584633

RESUMEN

HIV-1 infection impairs cellular cholesterol efflux by downmodulating the cholesterol transporter ABCA1, leading to metabolic co-morbidities like cardio-vascular disease. The main mechanism of this effect is impairment by the HIV-1 protein Nef of the ABCA1 interaction with the endoplasmic reticulum chaperone calnexin, which leads to a block in ABCA1 maturation followed by its degradation. However, ABCA1 is also downmodulated by Nef delivered with the extracellular vesicles, suggesting involvement of a direct Nef:ABCA1 interaction at the plasma membrane. Here, we present an optimized model of the Nef:ABCA1 interaction, which identifies interaction sites and provides an opportunity to perform a virtual screening for potential inhibitors. Interestingly, the predicted sites on Nef involved in the ABCA1 interaction overlap with those involved in the interaction with calnexin. The compounds previously shown to block Nef:calnexin interaction were among the top ranking ligands in docking simulations with ABCA1-interacting sites on Nef, suggesting the possibility that both interactions can be inhibited by the same chemical compounds. This study identifies a series of compounds for potential development as inhibitors of Nef-mediated co-morbidities of HIV infection.

11.
PLoS One ; 16(5): e0251599, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33984028

RESUMEN

Chronic HIV infection may exacerbate atherosclerotic vascular disease, which at advanced stages presents as necrotic plaques rich in crystalline cholesterol. Such lesions can catastrophically rupture precipitating myocardial infarct and stroke, now important causes of mortality in those living with HIV. However, in this population little is known about plaque structure relative to crystalline content and its chemical composition. Here, we first interrogated plaque crystal structure and composition in atherosclerotic SIV-infected macaques using non-linear optical microscopy. By stimulated Raman scattering and second harmonic generation approaches both amorphous and crystalline plaque lipid was detected and the crystal spectral profile indicated a cholesterol ester (CE) dominated composition. Versus controls, SIV+ samples had a greater number of cholesterol crystals (CCs), with the difference, in part, accounted for by crystals of a smaller length. Given the ester finding, we profiled HIV+ plaques and also observed a CE crystalline spectral signature. We further profiled plaques from Ldlr-/- mice fed a high fat diet, and likewise, found CE-dominate crystals. Finally, macrophage exposure to CCs or AcLDL induced auto-fluorescent puncta that co-stained with the LC3B autophagy sensor. In aggregate, we show that atheromatous plaques from mice, macaques and humans, display necrotic cores dominated by esterified CCs, and that plaque macrophages may induce autophagic vesicle formation upon encountering CCs. These findings help inform our knowledge of plaque core lipid evolution and how the process may incite systemic inflammation.


Asunto(s)
Ésteres del Colesterol/análisis , Infecciones por VIH/patología , Placa Aterosclerótica/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Animales , VIH/aislamiento & purificación , Infecciones por VIH/complicaciones , Macaca , Masculino , Ratones , Ratones Endogámicos C57BL , Imagen Óptica , Placa Aterosclerótica/complicaciones , Células RAW 264.7 , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación
12.
Front Immunol ; 11: 574508, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133090

RESUMEN

COVID-19 is a global pandemic currently in an acute phase of rapid expansion. While public health measures remain the most effective protection strategy at this stage, when the peak passes, it will leave in its wake important health problems. Historically, very few viruses have ever been eradicated. Instead, the virus may persist in communities causing recurrent local outbreaks of the acute infection as well as several chronic diseases that may arise from the presence of a "suppressed" virus or as a consequence of the initial exposure. An ideal solution would be an anti-viral medication that (i) targets multiple stages of the viral lifecycle, (ii) is insensitive to frequent changes of viral phenotype due to mutagenesis, (iii) has broad spectrum, (iv) is safe and (v) also targets co-morbidities of the infection. In this Perspective we discuss a therapeutic approach that owns these attributes, namely "lipid raft therapy." Lipid raft therapy is an approach aimed at reducing the abundance and structural modifications of host lipid rafts or at targeted delivery of therapeutics to the rafts. Lipid rafts are the sites of the initial binding, activation, internalization and cell-to-cell transmission of SARS-CoV-2. They also are key regulators of immune and inflammatory responses, dysregulation of which is characteristic to COVID-19 infection. Lipid raft therapy was successful in targeting many viral infections and inflammatory disorders, and can potentially be highly effective for treatment of COVID-19.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Microdominios de Membrana/efectos de los fármacos , Neumonía Viral/tratamiento farmacológico , Animales , COVID-19 , Comorbilidad , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/virología , Sistemas de Liberación de Medicamentos , Humanos , Microdominios de Membrana/virología , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/virología , Tratamiento Farmacológico de COVID-19
13.
Viruses ; 12(9)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32961937

RESUMEN

The C-promoter binding factor-1 (CBF-1) is a potent and specific inhibitor of the human immunodeficiency virus (HIV)-1 LTR promoter. Here, we demonstrate that the knockdown of endogenous CBF-1 in latently infected primary CD4+ T cells, using specific small hairpin RNAs (shRNA), resulted in the reactivation of latent HIV proviruses. Chromatin immunoprecipitation (ChIP) assays using latently infected primary T cells and Jurkat T-cell lines demonstrated that CBF-1 induces the establishment and maintenance of HIV latency by recruiting polycomb group (PcG/PRC) corepressor complexes or polycomb repressive complexes 1 and 2 (PRC1 and PRC2). Knockdown of CBF-1 resulted in the dissociation of PRCs corepressor complexes enhancing the recruitment of RNA polymerase II (RNAP II) at HIV LTR. Knockdown of certain components of PRC1 and PRC2 also led to the reactivation of latent proviruses. Similarly, the treatment of latently infected primary CD4+ T cells with the PRC2/EZH2 inhibitor, 3-deazaneplanocin A (DZNep), led to their reactivation.


Asunto(s)
VIH-1/fisiología , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Latencia del Virus/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Inmunoprecipitación de Cromatina , Epigenómica , Regulación Viral de la Expresión Génica , Técnicas de Silenciamiento del Gen , Infecciones por VIH/virología , Duplicado del Terminal Largo de VIH , VIH-1/genética , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Regiones Promotoras Genéticas , Provirus/genética , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño , Transcriptoma , Activación Viral , Latencia del Virus/genética
14.
PLoS Pathog ; 16(8): e1008679, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32790802

RESUMEN

Antiretroviral drugs that target various stages of the Human Immunodeficiency Virus (HIV) life cycle have been effective in curbing the AIDS epidemic. However, drug resistance, off-target effects of antiretroviral therapy (ART), and varying efficacy in prevention underscore the need to develop novel and alternative therapeutics. In this study, we investigated whether targeting the signaling molecule Sphingosine-1-phosphate (S1P) would inhibit HIV-1 infection and generation of the latent reservoir in primary CD4 T cells. We show that FTY720 (Fingolimod), an FDA-approved functional antagonist of S1P receptors, blocks cell-free and cell-to-cell transmission of HIV and consequently reduces detectable latent virus. Mechanistically, FTY720 impacts the HIV-1 life cycle at two levels. Firstly, FTY720 reduces the surface density of CD4, thereby inhibiting viral binding and fusion. Secondly, FTY720 decreases the phosphorylation of the innate HIV restriction factor SAMHD1 which is associated with reduced levels of total and integrated HIV, while reducing the expression of Cyclin D3. In conclusion, targeting the S1P pathway with FTY720 could be a novel strategy to inhibit HIV replication and reduce the seeding of the latent reservoir.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Infecciones por VIH/tratamiento farmacológico , VIH-1/crecimiento & desarrollo , Proteína 1 que Contiene Dominios SAM y HD/antagonistas & inhibidores , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Linfocitos T/inmunología , Replicación Viral , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/inmunología , Humanos , Lisofosfolípidos/metabolismo , Fosforilación , Proteína 1 que Contiene Dominios SAM y HD/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Linfocitos T/efectos de los fármacos , Latencia del Virus
15.
Arterioscler Thromb Vasc Biol ; 40(10): 2346-2359, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32787522

RESUMEN

OBJECTIVE: AIBP (apolipoprotein A-I binding protein) is an effective and selective regulator of lipid rafts modulating many metabolic pathways originating from the rafts, including inflammation. The mechanism of action was suggested to involve stimulation by AIBP of cholesterol efflux, depleting rafts of cholesterol, which is essential for lipid raft integrity. Here we describe a different mechanism contributing to the regulation of lipid rafts by AIBP. Approach and Results: We demonstrate that modulation of rafts by AIBP may not exclusively depend on the rate of cholesterol efflux or presence of the key regulator of the efflux, ABCA1 (ATP-binding cassette transporter A-I). AIBP interacted with phosphatidylinositol 3-phosphate, which was associated with increased abundance and activation of Cdc42 and rearrangement of the actin cytoskeleton. Cytoskeleton rearrangement was accompanied with reduction of the abundance of lipid rafts, without significant changes in the lipid composition of the rafts. The interaction of AIBP with phosphatidylinositol 3-phosphate was blocked by AIBP substrate, NADPH (nicotinamide adenine dinucleotide phosphate), and both NADPH and silencing of Cdc42 interfered with the ability of AIBP to regulate lipid rafts and cholesterol efflux. CONCLUSIONS: Our findings indicate that an underlying mechanism of regulation of lipid rafts by AIBP involves PIP-dependent rearrangement of the cytoskeleton.


Asunto(s)
Citoesqueleto de Actina/enzimología , Colesterol/metabolismo , Microdominios de Membrana/enzimología , Racemasas y Epimerasas/metabolismo , Transportador 1 de Casete de Unión a ATP/metabolismo , Citoesqueleto de Actina/genética , Animales , Células HeLa , Humanos , Microdominios de Membrana/genética , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Transducción de Señal , Células THP-1 , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
16.
J Biol Chem ; 295(38): 13377-13392, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32732283

RESUMEN

HIV-associated neurocognitive disorders (HANDs) are a frequent outcome of HIV infection. Effective treatment of HIV infection has reduced the rate of progression and severity but not the overall prevalence of HANDs, suggesting ongoing pathological process even when viral replication is suppressed. In this study, we investigated how HIV-1 protein Nef secreted in extracellular vesicles (exNef) impairs neuronal functionality. ExNef were rapidly taken up by neural cells in vitro, reducing the abundance of ABC transporter A1 (ABCA1) and thus cholesterol efflux and increasing the abundance and modifying lipid rafts in neuronal plasma membranes. ExNef caused a redistribution of amyloid precursor protein (APP) and Tau to lipid rafts and increased the abundance of these proteins, as well as of Aß42 ExNef further potentiated phosphorylation of Tau and activation of inflammatory pathways. These changes were accompanied by neuronal functional impairment. Disruption of lipid rafts with cyclodextrin reversed the phenotype. Short-term treatment of C57BL/6 mice with either purified recombinant Nef or exNef similarly resulted in reduced abundance of ABCA1 and elevated abundance of APP in brain tissue. The abundance of ABCA1 in brain tissue of HIV-infected human subjects diagnosed with HAND was lower, and the abundance of lipid rafts was higher compared with HIV-negative individuals. Levels of APP and Tau in brain tissue correlated with the abundance of Nef. Thus, modification of neuronal cholesterol trafficking and of lipid rafts by Nef may contribute to early stages of neurodegeneration and pathogenesis in HAND.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Microdominios de Membrana/metabolismo , Trastornos Neurocognitivos/metabolismo , Neuronas/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas tau/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Línea Celular Tumoral , Colesterol/genética , Colesterol/metabolismo , Infecciones por VIH/complicaciones , Infecciones por VIH/genética , Infecciones por VIH/patología , VIH-1/genética , Humanos , Microdominios de Membrana/genética , Ratones , Trastornos Neurocognitivos/etiología , Trastornos Neurocognitivos/genética , Trastornos Neurocognitivos/patología , Neuronas/patología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Proteínas tau/genética
17.
Vaccines (Basel) ; 8(3)2020 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-32731435

RESUMEN

The term "Exosomes" defines small extracellular vesicles, ranging from 30 to 150 nm in diameter, secreted by most eukaryotic cells into surrounding body fluids including blood, saliva, urine, bile and breast milk [...].

18.
FASEB J ; 34(6): 7253-7264, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32367579

RESUMEN

Drug repurposing is potentially the fastest available option in the race to identify safe and efficacious drugs that can be used to prevent and/or treat COVID-19. By describing the life cycle of the newly emergent coronavirus, SARS-CoV-2, in light of emerging data on the therapeutic efficacy of various repurposed antimicrobials undergoing testing against the virus, we highlight in this review a possible mechanistic convergence between some of these tested compounds. Specifically, we propose that the lysosomotropic effects of hydroxychloroquine and several other drugs undergoing testing may be responsible for their demonstrated in vitro antiviral activities against COVID-19. Moreover, we propose that Niemann-Pick disease type C (NPC), a lysosomal storage disorder, may provide new insights into potential future therapeutic targets for SARS-CoV-2, by highlighting key established features of the disorder that together result in an "unfavorable" host cellular environment that may interfere with viral propagation. Our reasoning evolves from previous biochemical and cell biology findings related to NPC, coupled with the rapidly evolving data on COVID-19. Our overall aim is to suggest that pharmacological interventions targeting lysosomal function in general, and those particularly capable of reversibly inducing transient NPC-like cellular and biochemical phenotypes, constitute plausible mechanisms that could be used to therapeutically target COVID-19.


Asunto(s)
Antivirales/farmacocinética , Betacoronavirus/fisiología , Infecciones por Coronavirus/tratamiento farmacológico , Reposicionamiento de Medicamentos , Endosomas/virología , Hidroxicloroquina/farmacología , Lisosomas/virología , Enfermedad de Niemann-Pick Tipo C/patología , Neumonía Viral/tratamiento farmacológico , Proteína ADAM17/fisiología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Antivirales/farmacología , Antivirales/uso terapéutico , Bencilisoquinolinas/farmacología , Bencilisoquinolinas/uso terapéutico , Transporte Biológico , COVID-19 , Catepsina L/fisiología , Endocitosis , Endosomas/efectos de los fármacos , Endosomas/fisiología , Glicopéptidos/farmacología , Glicopéptidos/uso terapéutico , Humanos , Hidroxicloroquina/farmacocinética , Hidroxicloroquina/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/fisiología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lípidos de la Membrana/metabolismo , Microdominios de Membrana/fisiología , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/metabolismo , Oxiesteroles/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Receptores Virales/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/fisiología , Triazoles/farmacología , Triazoles/uso terapéutico , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
19.
Oncotarget ; 11(7): 699-726, 2020 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-32133046

RESUMEN

Despite reductions in mortality from the use of highly active antiretroviral therapy (HAART), the presence of latent or transcriptionally silent proviruses prevents HIV cure/eradication. We have previously reported that DNA-dependent protein kinase (DNA-PK) facilitates HIV transcription by interacting with the RNA polymerase II (RNAP II) complex recruited at HIV LTR. In this study, using different cell lines and peripheral blood mononuclear cells (PBMCs) of HIV-infected patients, we found that DNA-PK stimulates HIV transcription at several stages, including initiation, pause-release and elongation. We are reporting for the first time that DNA-PK increases phosphorylation of RNAP II C-terminal domain (CTD) at serine 5 (Ser5) and serine 2 (Ser2) by directly catalyzing phosphorylation and by augmenting the recruitment of the positive transcription elongation factor (P-TEFb) at HIV LTR. Our findings suggest that DNA-PK expedites the establishment of euchromatin structure at HIV LTR. DNA-PK inhibition/knockdown leads to the severe impairment of HIV replication and reactivation of latent HIV provirus. DNA-PK promotes the recruitment of Tripartite motif-containing 28 (TRIM28) at LTR and assists the release of paused RNAP II through TRIM28 phosphorylation. These results provide the mechanisms through which DNA-PK controls the HIV gene expression and, likely, can be extended to cellular gene expression, including during cell malignancy, where the role of DNA-PK has been well-established.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...