Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Cells ; 9(6)2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32526847

RESUMEN

A significant number of people living with HIV (PLWH) develop HIV-associated neurocognitive disorders (HAND) despite highly effective antiretroviral therapy (ART). Dysregulated macroautophagy (autophagy) is implicated in HAND pathogenesis. The viral protein Nef, expressed even with suppressive ART, and certain antiretrovirals affect autophagy in non-CNS cells. Astrocytes, vital for CNS microenvironment homeostasis and neuronal health, require autophagy for their own homeostasis. We hypothesized that extracellular Nef and/or ART impact astrocyte autophagy, thus contributing to HAND. We studied in-bulk and selective autophagic flux in primary human astrocytes treated with extracellular Nef and/or a combination of tenofovir+emtricitabine+raltegravir (ART) using Western blotting, a tandem fluorescent LC3 reporter, and transmission electron microscopy/morphometry. We show that after 24 h treatment, Nef and ART decrease autophagosomes through different mechanisms. While Nef accelerates autophagosome degradation without inducing autophagosome formation, ART inhibits autophagosome formation. Combination Nef+ART further depletes autophagosomes by inducing both abnormalities. Additionally, extracellular Nef and/or ART inhibit lysosomal degradation of p62, indicating Nef and/or ART affect in-bulk and selective autophagy differently. Dysregulation of both autophagic processes is maintained after 7 days of Nef and/or ART treatment. Persistent autophagy dysregulation due to chronic Nef and/or ART exposure may ultimately result in astrocyte and neuronal dysfunction, contributing to HAND.


Asunto(s)
Antirretrovirales/uso terapéutico , Astrocitos/efectos de los fármacos , Autofagia/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Trastornos Neurocognitivos/inducido químicamente , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/uso terapéutico , Antirretrovirales/farmacología , Infecciones por VIH/genética , Humanos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
3.
Protein Pept Lett ; 27(11): 1151-1158, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32364062

RESUMEN

BACKGROUND: There is no effective and safe preventive/therapeutics vaccine against HIV-1 worldwide. Different viral proteins such as Nef, and two regions of Env including; variable loop of gp120 (V3) and membrane proximal external region of gp41 (MPER) are particularly important for vaccine development in different strategies and they are also the primary targets of cellular and humoral immune responses. On the other side, LDP12 is a new cell-penetrating peptide (CPP) which is capable of therapeutic application and cargoes delivery across the cellular membrane. OBJECTIVE: In current study, we designed and produced Nef-MPER-V3 fusion protein harboring LDP12 that has the capability of being used in future vaccine studies. METHODS: The CPP-protein was expressed in E. coli Rosseta (DE3) strain and purified through Ni-NTA column. Characterization of cellular delivery and toxicity of the recombinant protein were evaluated by western blotting and MTT assay. RESULTS: Our results showed that the CPP-protein was successfully expressed and purified with high yield of 5 mg/L. Furthermore, non-cytotoxic effect was observed and specific band (~ 37 KDa) in western blotting indicated the capability of LDP12 to improve the rate of penetration into HEK-293T cells in comparison with a control sample. CONCLUSION: Altogether, the data indicated that LDP12 CPP could be utilized to internalize HIV-1 Nef-MPER-V3 protein into eukaryotic cell lines without any toxicity and represented a valuable potential vaccine candidate and this guarantees the further evaluation towards the assessment of its immunogenicity in mice, which is currently under process.


Asunto(s)
Vacunas contra el SIDA , Péptidos de Penetración Celular , Portadores de Fármacos , Proteína gp41 de Envoltorio del VIH , VIH-1 , Oligopéptidos , Proteínas Recombinantes de Fusión , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Vacunas contra el SIDA/química , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/farmacocinética , Vacunas contra el SIDA/farmacología , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/inmunología , Péptidos de Penetración Celular/farmacocinética , Péptidos de Penetración Celular/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Células HEK293 , Proteína gp41 de Envoltorio del VIH/química , Proteína gp41 de Envoltorio del VIH/inmunología , Proteína gp41 de Envoltorio del VIH/farmacocinética , Proteína gp41 de Envoltorio del VIH/farmacología , VIH-1/química , VIH-1/inmunología , Humanos , Oligopéptidos/química , Oligopéptidos/inmunología , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/química , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacocinética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
4.
J Neuroinflammation ; 15(1): 303, 2018 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-30382871

RESUMEN

BACKGROUND: HIV-associated neurocognitive disorder (HAND) is a neurodegenerative disease associated with persistent neuroinflammation and subsequent neuron damage. Pro-inflammatory factors and neurotoxins from activated astrocytes by HIV-1 itself and its encoded proteins, including the negative factor (Nef), are involved in the pathogenesis of HAND. This study was designed to find potential lncRNAs that regulate astrocyte functions and inflammation process. METHODS: We performed microarray analysis of lncRNAs from primary mouse astrocytes treated with Nef protein. Top ten lncRNAs were validated through real-time PCR analysis. Gene ontology (GO) and KEGG pathway analysis were applied to explore the potential functions of lncRNAs. RIP and ChIP assays were performed to demonstrate the mechanism of lncRNA regulating gene expression. RESULTS: There were 638 co-upregulated lncRNAs and 372 co-downregulated lncRNAs in primary astrocytes treated with Nef protein for both 6 h and 12 h. GO and KEGG pathway analysis showed that the biological functions of top differential-expressed mRNAs were associated with inflammatory cytokines and chemokine. Knockdown of lncRNA AK006025, not AK138360, inhibited significantly CXCL9, CXCL10 (IP-10), and CXCL11 expression in astrocytes treated with Nef protein. Mechanism study showed that AK006025 associated with CBP/P300 was enriched in the promoter of CXCL9, CXCL10, and CXCL11 genes. CONCLUSIONS: Our findings uncovered the expression profiles of lncRNAs and mRNAs in vitro, which might help to understand the pathways that regulate astrocyte activation during the process of HAND.


Asunto(s)
Astrocitos/efectos de los fármacos , Quimiocina CXCL1/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , ARN Largo no Codificante/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Transfección , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
5.
J Infect Dis ; 216(11): 1415-1424, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-28968792

RESUMEN

HLA-B*52:01-C*12:02, which is found in approximately 20% of all Japanese persons, is well known to be associated with ulcerative colitis and Takayasu arteritis. This haplotype is also known to be protective in individuals infected with human immunodeficiency virus (HIV) type 1. Recent studies showed that HLA-B*52:01-restricted HIV-1-specific T cells suppress HIV-1 and that HLA-C*12:02 together with KIR2DL2 play an important role in natural killer cell-mediated control of HIV-1. However, the role of HLA-C*12:02-restricted cytotoxic T lymphocytes (CTLs) in suppressing HIV-1 replication remains unknown. In the present study, we demonstrated that HLA-C*12:02-restricted CTLs specific for 2 immunodominant epitopes, Pol IY11 and Nef MY9, contributed to the suppression of HIV-1 replication in HIV-1-infected individuals. Further analysis demonstrated that these 2 HLA-C*12:02-restricted CTLs together with 4 HLA-B*52:01-restricted ones effectively suppressed HIV-1 in individuals with the HLA-B*52:01-C*12:02 haplotype. Thus, both HLA-C*12:02 and HLA-B*52:01 alleles contribute to HIV-1 suppression via both HIV-1-specific CTLs and natural killer cells in individuals with this haplotype.


Asunto(s)
VIH-1/efectos de los fármacos , Antígenos HLA-B/farmacología , Antígeno HLA-B52/farmacología , Antígenos HLA-C/farmacología , Haplotipos/inmunología , Alelos , Línea Celular , Cromo/análisis , Citocinas/análisis , Epítopos de Linfocito T , Infecciones por VIH/dietoterapia , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Antígenos HLA-B/inmunología , Antígeno HLA-B52/inmunología , Antígenos HLA-C/inmunología , Antígenos HLA-C/aislamiento & purificación , Interacciones Huésped-Patógeno , Humanos , Epítopos Inmunodominantes/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Receptores KIR2DL2/fisiología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Replicación Viral/efectos de los fármacos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Productos del Gen pol del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen pol del Virus de la Inmunodeficiencia Humana/farmacología
6.
Oncotarget ; 8(7): 11302-11315, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-28076321

RESUMEN

PURPOSE: Discovery and development of a novel anticancer PEG-SMR-Clu peptide to prevent breast cancer metastasis. How breast cancer cells and primary mammary epithelial cells interact and communicate with each other to promote tumorigenesis and how to prevent tumor metastasis has long been a concern of researchers. Cancer cells secrete exosomes containing proteins and RNA. These factors can influence tumor development by directly targeting cancer cells and tumor stroma. In this study, we determined the effects of a peptide as an inhibitor of exosome secretion on breast tumors. We developed a peptide derived from the Secretion Modification Region (SMR) of HIV-1 Nef protein that was modified with PEG on the N-terminus and with a Clusterin (Clu)-binding peptide on the C-terminus. Attachment of PEG to the SMR peptide, termed PEGylation, offers improved water solubility and stability as well as reduced clearance through the kidneys, leading to a longer circulation time. The 12-mer Clu-binding peptide plays multiple roles in tumor development and metastasis. The Clu peptide can be detected by antibody in vivo, thus it has the potential to be used to monitor tumor status and treatment efficacy in animal studies and eventually in cancer patients. RESULTS: PEG-SMRwt-Clu and PEG-SMRwt peptides inhibited the growth of both of MCF-7 (estrogen responsive, ER+) and MDA-MD-231 (estrogen non-responsive, ER-) human breast cancer cells in a dose and time-dependent manner, without inducing cytotoxic effects. The SMRwt peptide, combined with paclitaxel, induced G2/M phase cell cycle arrest on MCF-7 and MDA-MB-231 cells but did not promote apoptosis. PEG-SMRwt-Clu peptide treatment blocked exosome release from both MCF-7 and MDA-MB-231 cells. This effect was blocked by knockdown of the chaperone protein mortalin by either antibody or siRNA. MATERIALS AND METHODS: MCF-7 and MDA-MB-231 breast tumor cells were treated with PEG-SMR-Clu peptide alone and in combination with paclitaxel and cisplatin. Cell proliferation and viabilty were determined via cell cycle analysis using Cellometer imaging cytometry, Annexin V and MTT assays. The effects of the PEG-SMR-Clu peptide on tumor exosome release were determined by testing isolated exosome fractions, for (i) expression of CD63 and Alix proteins by Western blotting, (ii) NanoSight nanoparticle tracking analysis (NTA 10) to measure exosomes size and concentration, and (iii) measurement of acetylcholinesterase (AchE) for exosome specific enzyme activity. CONCLUSIONS: PEG-SMRwt-CLU peptides inhibited the growth of human breast cancer cells and blocked tumor exosome release in vitro. The peptide alone did not cause increased cytotoxicity or apoptosis induction, but did cause cell cycle G2/M phase arrest in both estrogen responsive and non-responsive breast cancer cells. These data suggest a potential therapeutic value of SMR to prevent breast cancer metastasis and as an adjuvant for the chemotherapeutic treatment of human breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Clusterina/farmacología , Exosomas/metabolismo , Péptidos/farmacología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Neoplasias de la Mama/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Polietilenglicoles/farmacología
7.
J Neurovirol ; 22(2): 129-39, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26631079

RESUMEN

The negative factor (Nef) of human immunodeficiency virus (HIV) is an accessory protein that is thought to be integral to HIV-associated immune- and neuroimmune pathogenesis. Here, we show that nef-transfected microglia-released Nef+ exosome (exNef) disrupts the apical blood-brain barrier (BBB) and that only nef-transfected microglia release Nef in exosomes. nef-gfp-transduced neurons and astrocytes release exosomes but did not release exNef in the extracellular space. Apical administration of exNef derived from nef-transfected 293T cells reduced transendothelial electrical resistance (TEER) and increased permeability of the BBB. Microglia-derived exNef applied to either the apical/basal BBB significantly reduced expression of the tight junction protein, ZO-1, suggesting a mechanism of exNef-mediated neuropathogenesis. Microglia exposed to exNef release elevated levels of Toll-like receptor-induced cytokines and chemokines IL-12, IL-8, IL-6, RANTES, and IL-17A. Magnetic nanoparticle delivery of Nef peptides containing the Nef myrisolation site across an in vitro BBB ultimately reduced nef-transfected microglia release of Nef exosomes and prevented the loss of BBB integrity and permeability as measured by TEER and dextran-FITC transport studies, respectively. Overall, we show that exNef is released from nef-gfp-transfected microglia; exNef disrupts integrity and permeability, and tight junctions of the BBB, and induces microglial cytokine/chemokine secretion. These exNef-mediated effects were significantly restricted by Nef peptides. Taken together, this study provides preliminary evidence of the role of exNef in HIV neuroimmune pathogenesis and the feasibility of a nanomedicine-based therapeutics targeting exNef to treat HIV-associated neuropathogenesis.


Asunto(s)
Portadores de Fármacos/farmacología , Exosomas/metabolismo , Microglía/efectos de los fármacos , Péptidos/farmacología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Línea Celular , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , VIH-1/química , Humanos , Interleucinas/genética , Interleucinas/inmunología , Nanopartículas de Magnetita/química , Microglía/citología , Microglía/metabolismo , Modelos Biológicos , Péptidos/síntesis química , Transducción de Señal , Transfección , Transgenes , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
8.
Aging Cell ; 14(4): 534-46, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25847297

RESUMEN

To maintain bone mass turnover and bone mineral density (BMD), bone marrow (BM) mesenchymal stem cells (MSCs) are constantly recruited and subsequently differentiated into osteoblasts. HIV-infected patients present lower BMD than non-HIV infected individuals and a higher prevalence of osteopenia/osteoporosis. In antiretroviral treatment (ART)-naive patients, encoded HIV proteins represent pathogenic candidates. They are released by infected cells within BM and can impact on neighbouring cells. In this study, we tested whether HIV proteins Tat and/or Nef could induce senescence of human BM-MSCs and reduce their capacity to differentiate into osteoblasts. When compared to nontreated cells, MSCs chronically treated with Tat and/or Nef up to 30 days reduced their proliferative activity and underwent early senescence, associated with increased oxidative stress and mitochondrial dysfunction. The antioxidant molecule N-acetyl- cysteine had no or minimal effects on Tat- or Nef-induced senescence. Tat but not Nef induced an early increase in NF-κB activity and cytokine/chemokine secretion. Tat-induced effects were prevented by the NF-κB inhibitor parthenolide, indicating that Tat triggered senescence via NF-κB activation leading to oxidative stress. Otherwise, Nef- but not Tat-treated cells displayed early inhibition of autophagy. Rapamycin, an autophagy inducer, reversed Nef-induced senescence and oxidative stress. Moreover, Tat+Nef had cumulative effects. Finally, Tat and/or Nef decreased the MSC potential of osteoblastic differentiation. In conclusion, our in vitro data show that Tat and Nef could reduce the number of available precursors by inducing MSC senescence, through either enhanced inflammation or reduced autophagy. These results offer new insights into the pathophysiological mechanisms of decreased BMD in HIV-infected patients.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Acetilcisteína/farmacología , Antioxidantes/farmacología , Autofagia/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Osteocalcina/genética , Osteocalcina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Cultivo Primario de Células , Proteínas Recombinantes/farmacología , Sesquiterpenos/farmacología , Transducción de Señal , Sirolimus/farmacología
9.
PLoS One ; 9(4): e93699, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24705461

RESUMEN

Monocytes and macrophages utilize the class A and B scavenger receptors to recognize and perform phagocytosis of invading microbes before a pathogen-specific immune response is generated. HIV-1 Nef protein affects the innate immune system impairing oxidative burst response and phagocytic capacity of macrophages. Our data show that exogenous recombinant myristoylated Nef protein induces a marked CD36 downregulation in monocytes from Peripheral Blood Mononuclear Cells, in Monocyte-Derived Macrophages (MDMs) differentiated by cytokines and in MDMs contained in a mixed culture obtained expanding PBMCs under Human Erythroid Massive Amplification condition. Under the latter culture condition we identify three main populations after 6 days of expansion: lymphocytes (37.8 ± 14.7%), erythroblasts (46.7±6.1%) and MDMs (15.7 ± 7.5%). The Nef addition to the cell culture significantly downregulates CD36 expression in MDMs, but not in erythroid cells. Furthermore, CD36 inhibition is highly specific since it does not modify the expression levels of other MDM markers such as CD14, CD11c, CD86, CD68, CD206, Toll-like Receptor 2 and Toll-like Receptor 4. Similar results were obtained in MDMs infected with VSV-G pseudotyped HIV-1-expressing Nef. The reduced CD36 membrane expression is associated with decrease of correspondent CD36 mRNA transcript. Furthermore, Nef-induced CD36 downregulation is linked to both impaired scavenger activity with reduced capability to take up oxidized lipoproteins and to significant decreased phagocytosis of fluorescent beads and GFP-expressing Salmonella tiphymurium. In addition we observed that Nef induces TNF-α release in MDMs. Although these data suggest a possible involvement of TNF-α in mediating Nef activity, our results exclude a possible relationship between Nef-induced TNF-α release and Nef-mediated CD36 downregulation. The present work shows that HIV-1 Nef protein may have a role in the strategies elaborated by HIV-1 to alter pathogen disease outcomes, by modulating CD36 expression in macrophages, favoring the onset of opportunistic infections in HIV-1 infected people.


Asunto(s)
Antígenos CD36/metabolismo , Regulación hacia Abajo/fisiología , VIH-1/genética , Inmunidad Innata/inmunología , Macrófagos/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/inmunología , Cartilla de ADN/genética , Regulación hacia Abajo/efectos de los fármacos , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , VIH-1/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Salmonella typhimurium/metabolismo , Estadísticas no Paramétricas , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
10.
Cell Death Dis ; 5: e1080, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24556695

RESUMEN

HIV-1 Nef protein has key roles at almost all stages of the viral life cycle. We assessed the role of Nef and of the translation elongation factor eEF1A in primary human macrophages. Nuclear retention experiments and inhibition of the exportin-t (Exp-t) pathway suggested that cytoplasmic relocalization of eEF1A, mediated by Exp-t occurs in Nef-treated monocyte-derived macrophages (MDMs). We observed the presence of tRNA in the Nef/eEF1A complexes. Nucleocytoplasmic relocalization of the Nef/eEF1A complexes prevented stress-induced apoptosis of MDMs treated with brefeldin A. Blockade of stress-induced apoptosis of MDMs treated with HIV-1 Nef resulted from enhanced nucleocytoplasmic transport of eEF1A with decreased release of mitochondrial cytochrome c, and from increased tRNA binding to cytochrome c, ultimately leading to an inhibition of caspase activation. Our results indicate that HIV-1 Nef, through the nucleocytoplasmic relocalization of eEF1A and tRNAs, enhances resistance to stress-induced apoptosis in primary human macrophages.


Asunto(s)
Apoptosis/efectos de los fármacos , Brefeldino A/farmacología , Macrófagos/efectos de los fármacos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Caspasas/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Resistencia a Medicamentos , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Mitocondrias/metabolismo , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/metabolismo , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Interferencia de ARN , ARN de Transferencia/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Transfección , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
11.
PLoS One ; 9(1): e86998, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24489825

RESUMEN

Human and Simian Immunodeficiency virus (HIV-1, HIV-2, and SIV) encode an accessory protein, Nef, which is a pathogenesis and virulence factor. Nef is a multivalent adapter that dysregulates the trafficking of many immune cell receptors, including chemokine receptors (CKRs). Physiological endocytic itinerary of agonist occupied CXCR4 involves ubiquitinylation of the phosphorylated receptor at three critical lysine residues and dynamin-dependent trafficking through the ESCRT pathway into lysosomes for degradation. Likewise, Nef induced CXCR4 degradation was critically dependent on the three lysines in the C-terminal -SSLKILSKGK- motif. Nef directly recruits the HECT domain E3 ligases AIP4 or NEDD4 to CXCR4 in the resting state. This mechanism was confirmed by ternary interactions of Nef, CXCR4 and AIP4 or NEDD4; by reversal of Nef effect by expression of catalytically inactive AIP4-C830A mutant; and siRNA knockdown of AIP4, NEDD4 or some ESCRT-0 adapters. However, ubiquitinylation dependent lysosomal degradation was not the only mechanism by which Nef downregulated CKRs. Agonist and Nef mediated CXCR2 (and CXCR1) degradation was ubiquitinylation independent. Nef also profoundly downregulated the naturally truncated CXCR4 associated with WHIM syndrome and engineered variants of CXCR4 that resist CXCL12 induced internalization via an ubiquitinylation independent mechanism.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , VIH-1/química , Monocitos/metabolismo , Receptores CXCR4/genética , Ubiquitina/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Secuencias de Aminoácidos , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Endocitosis , Complejos de Clasificación Endosomal Requeridos para el Transporte/antagonistas & inhibidores , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , VIH-1/genética , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , Datos de Secuencia Molecular , Monocitos/efectos de los fármacos , Monocitos/virología , Ubiquitina-Proteína Ligasas Nedd4 , Cultivo Primario de Células , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores CXCR4/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
12.
PLoS One ; 8(1): e54295, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23372701

RESUMEN

HIV-1 Nef protein down-regulates several cell surface receptors through its interference with the cell sorting and trafficking machinery. Here we demonstrate for the first time the ability of Nef to down-regulate cell surface expression of the negative immune modulator CTLA-4. Down-regulation of CTLA-4 required the Nef motifs DD175, EE155 and LL165, all known to be involved in vesicle trafficking. Disruption of the lysosomal functions by pH-neutralizing agents prevented CTLA-4 down-regulation by Nef, demonstrating the implication of the endosomal/lysosomal compartments in this process. Confocal microscopy experiments visualized the co-localization between Nef and CTLA-4 in the early and recycling endosomes but not at the cell surface. Overall, our results provide a novel mechanism by which HIV-1 Nef interferes with the surface expression of the negative regulator of T cell activation CTLA-4. Down-regulation of CTLA-4 may contribute to the mechanisms by which HIV-1 sustains T cell activation, a critical step in viral replication and dissemination.


Asunto(s)
Antígeno CTLA-4/genética , VIH-1/genética , Proteínas Recombinantes de Fusión/farmacología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Secuencias de Aminoácidos , Antígenos CD4/genética , Antígenos CD4/metabolismo , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Células HEK293 , VIH-1/química , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
13.
J Immunol ; 188(8): 3620-7, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22407921

RESUMEN

HIV-1 proteins, including Tat, gp120, and Nef, activate macrophages (MΦ), which is consistent with the fact that HIV-1 infection is characterized by sustained immune activation. Meanwhile, MΦ are functionally classified into two types: proinflammatory M1-MΦ and anti-inflammatory M2-MΦ. We show that HIV-1 proteins, particularly Nef, preferentially activate M2-MΦ. Extracellular Tat, gp120, and Nef activated MAPK and NF-κB pathways in human peripheral blood monocyte-derived MΦ. However, the activation was marked in M-CSF-derived M2-MΦ but not GM-CSF-derived M1-MΦ. Nef was the most potent activator, and its signaling activation was comparable to that by TNF-α. Indeed, Nef was internalized more rapidly by M2-MΦ than by M1-MΦ. The myristoylation and proline-rich motif of Nef were responsible for the observed signaling activation. Consistent with the activation of MAPK/NF-κB pathways, Nef stimulated the production of a number of proinflammatory cytokines/chemokines by M2-MΦ. However, Nef reduced the expression of CD163 and phagocytosis, the characteristic markers of M2-MΦ, indicating that Nef drives an M2-like to M1-like phenotypic shift. Because the differentiation of most tissue MΦ depends on M-CSF and its receptor, which is the essential axis for the anti-inflammatory M2-MΦ phenotype, the current study reveals an efficient mechanism by which HIV-1 proteins, such as Nef, induce the proinflammatory MΦ.


Asunto(s)
Proteína gp120 de Envoltorio del VIH/fisiología , VIH-1/inmunología , Macrófagos/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología , Biomarcadores/metabolismo , Diferenciación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Proteína gp120 de Envoltorio del VIH/farmacología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Inflamación/inmunología , Inflamación/patología , Activación de Macrófagos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Especificidad de Órganos , Fenotipo , Cultivo Primario de Células , Transducción de Señal , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología
14.
Cell Death Dis ; 2: e246, 2011 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-22190005

RESUMEN

Mitochondrial dysfunction is implicated in a majority of neurodegenerative disorders and much study of neurodegenerative disease is done on cultured neurons. In traditional tissue culture, the oxygen level that cells experience is dramatically higher (21%) than in vivo conditions (1-11%). These differences can alter experimental results, especially, pertaining to mitochondria and oxidative metabolism. Our results show that primary neurons cultured at physiological oxygen levels found in the brain showed higher polarization, lower rates of ROS production, larger mitochondrial networks, greater cytoplasmic fractions of mitochondria and larger mitochondrial perimeters than those cultured at higher oxygen levels. Although neurons cultured in either physiological oxygen or atmospheric oxygen exhibit significant increases in mitochondrial reactive oxygen species (ROS) production when treated with the human immunodeficiency virus (HIV) virotoxin trans-activator of transcription, mitochondria of neurons cultured at physiological oxygen underwent depolarization with dramatically increased cell death, whereas those cultured at atmospheric oxygen became hyperpolarized with no increase in cell death. Studies with a second HIV virotoxin, negative regulation factor (Nef), revealed that Nef treatment also increased mitochondrial ROS production for both the oxygen conditions, but resulted in mitochondrial depolarization and increased death only in neurons cultured in physiological oxygen. These results indicate a role for oxidative metabolism in a mechanism of neurotoxicity during HIV infection and demonstrate the importance of choosing the correct, physiological, culture oxygen in mitochondrial studies performed in neurons.


Asunto(s)
VIH/metabolismo , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxígeno/metabolismo , Péptidos Cíclicos/farmacología , Adenosina Trifosfato/metabolismo , Apoptosis , Células Cultivadas , Productos del Gen tat/farmacología , Humanos , Microscopía Confocal , Mitocondrias/química , Neuronas/citología , Especies Reactivas de Oxígeno/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
15.
PLoS One ; 6(8): e23442, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21858117

RESUMEN

BACKGROUND: The mucosal tissues play a central role in the transmission of HIV-1 infection as well as in the pathogenesis of AIDS. Despite several clinical studies reported intestinal dysfunction during HIV infection, the mechanisms underlying HIV-induced impairments of mucosal epithelial barrier are still unclear. It has been postulated that HIV-1 alters enterocytic function and HIV-1 proteins have been detected in several cell types of the intestinal mucosa. In the present study, we analyzed the effect of the accessory HIV-1 Nef protein on human epithelial cell line. METHODOLOGY/PRINCIPAL FINDINGS: We used unstimulated or IFN-γ-stimulated Caco-2 cells, as a model for homeostatic and inflamed gastrointestinal tracts, respectively. We investigated the effect of exogenous recombinant Nef on monolayer integrity analyzing its uptake, transepithelial electrical resistance, permeability to FITC-dextran and the expression of tight junction proteins. Moreover, we measured the induction of proinflammatory mediators. Exogenous Nef was taken up by Caco-2 cells, increased intestinal epithelial permeability and upset the IFN-γ-induced reduction of transepithelial resistance, interfering with tight junction protein expression. Moreover, Nef inhibited IFN-γ-induced apoptosis and up-regulated TNF-α, IL-6 and MIP-3α production by Caco-2 cells while down-regulated IL-10 production. The simultaneous exposure of Caco-2 cells to Nef and IFN-γ did not affect cytokine secretion respect to untreated cells. Finally, we found that Nef counteracted the IFN-γ induced arachidonic acid cascade. CONCLUSION/SIGNIFICANCE: Our findings suggest that exogenous Nef, perturbing the IFN-γ-induced impairment of intestinal epithelial cells, could prolong cell survival, thus allowing for accumulation of viral particles. Our results may improve the understanding of AIDS pathogenesis, supporting the discovery of new therapeutic interventions.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Interferón gamma/farmacología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Apoptosis/efectos de los fármacos , Ácido Araquidónico/metabolismo , Transporte Biológico/efectos de los fármacos , Western Blotting , Células CACO-2 , Quimiocinas/metabolismo , Ciclooxigenasa 1 , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Dextranos/farmacocinética , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Células HEK293 , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Recombinantes/farmacología , Proteína de la Zonula Occludens-1 , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
16.
J Infect Dis ; 203(12): 1824-31, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21606541

RESUMEN

Human immunodeficiency virus (HIV) infection is associated with insulin resistance. HIV type 1 Nef downregulates cell surface protein expression, alters signal transduction, and interacts with the cytoskeleton and proteins involved in actin polymerization. These functions are required for glucose uptake by insulin-stimulated adipocytes. We sought to determine whether Nef alters adipocyte glucose homeostasis. Using radiolabeled glucose, we found that adipocytes exposed to recombinant Nef took in 42% less glucose after insulin stimulation than did control cells. This reduction resulted from a Nef-dependent inhibition of glucose transporter 4 (GLUT4) trafficking, as assessed by means of immunofluorescence microscopy. Immunoblot analysis revealed a decrease in phosphorylation of signal transducing proteins after Nef treatment, and fluorescence microscopy showed a dramatic alteration in cortical actin organization. We conclude that Nef interferes with insulin-stimulated processes in adipocytes. We have identified HIV Nef, which is detectable and antigenic in serum samples from HIV-infected people, as a novel contributor to the development of insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Glucosa/metabolismo , Infecciones por VIH/complicaciones , VIH-1 , Resistencia a la Insulina/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Células 3T3-L1 , Actinas/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/virología , Animales , Western Blotting , Membrana Celular/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Proteínas Activadoras de GTPasa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Insulina/farmacología , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/sangre , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
17.
J Biol Chem ; 285(17): 12629-37, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20068037

RESUMEN

Human immunodeficiency virus-1 (HIV-1) impairs tumor necrosis factor-alpha (TNF-alpha)-mediated macrophage apoptosis induced by Mycobacterium tuberculosis (Mtb). HIV Nef protein plays an important role in the pathogenesis of AIDS. We have tested the hypothesis that exogenous Nef is a factor that inhibits TNF-alpha production/apoptosis in macrophages infected with Mtb. We demonstrate that Mtb and Nef individually trigger TNF-alpha production in macrophages. However, TNF-alpha production is dampened when the two are present simultaneously, probably through cross-regulation of the individual signaling pathways leading to activation of the TNF-alpha promoter. Mtb-induced TNF-alpha production is abrogated upon mutation of the Ets, Egr, Sp1, CRE, or AP1 binding sites on the TNF-alpha promoter, whereas Nef-mediated promoter activation depends only on the CRE and AP1 binding sites, pointing to differences in the mechanisms of activation of the promoter. Mtb-dependent promoter activation depends on the mitogen-activated kinase (MAPK) kinase kinase ASK1 and on MEK/ERK signaling. Nef inhibits ASK1/p38 MAPK-dependent Mtb-induced TNF-alpha production probably by inhibiting binding of ATF2 to the TNF-alpha promoter. It also inhibits MEK/ERK-dependent Mtb-induced binding of FosB to the promoter. Nef-driven TNF-alpha production occurs in an ASK1-independent, Rac1/PAK1/p38 MAPK-dependent, and MEK/ERK-independent manner. The signaling pathways used by Mtb and Nef to trigger TNF-alpha production are therefore distinctly different. In addition to attenuating Mtb-dependent TNF-alpha promoter activation, Nef also reduces Mtb-dependent TNF-alpha mRNA stability probably through its ability to inhibit ASK1/p38 MAPK signaling. These results provide new insight into how HIV Nef probably exacerbates tuberculosis infection by virtue of its ability to dampen Mtb-induced TNF-alpha production.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/metabolismo , Apoptosis , VIH-1/metabolismo , Macrófagos/metabolismo , Mycobacterium tuberculosis/metabolismo , Tuberculosis/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Síndrome de Inmunodeficiencia Adquirida/microbiología , Factor de Transcripción Activador 2/metabolismo , Línea Celular , VIH-1/patogenicidad , Humanos , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/microbiología , Macrófagos/virología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Elementos de Respuesta , Tuberculosis/virología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Quinasas p21 Activadas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rac1/metabolismo
18.
J Surg Res ; 156(2): 257-64, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19540523

RESUMEN

BACKGROUND: Infection of human immunodeficiency virus (HIV) has been associated with several chronic diseases, including pulmonary artery hypertension and atherosclerosis. However, the underlying mechanisms of these vascular complications are largely unknown. The objective of this study was to test a novel hypothesis that HIV Nef, an accessory HIV protein, may directly affect endothelial functions and gene expression in pulmonary arteries. METHODS: Fresh porcine pulmonary artery rings and human pulmonary artery endothelial cells (HPAECs) were treated with HIV Nef for 24 h. With a myograph device, vasomotor function was determined with thromboxane A2 analog, U46619, for contraction, bradykinin, and sodium nitroprusside for relaxation. The expression of endothelial nitric oxide synthase (eNOS) was determined with real-time PCR and immunohistochemistry. Nitric oxide (NO) production was determined by Calorimetric Nitric Oxide Assay kit. Superoxide anion levels were detected with lucigenin-enhanced chemiluminescence assay and dihydroethidium (DHE) staining. RESULTS: The endothelium-dependent vasorelaxation in response to bradykinin was significantly reduced in HIV Nef-treated porcine pulmonary artery rings in a concentration-dependent manner. In response to bradykinin (10(-8) mol/L), HIV Nef (10 ng/mL) significantly reduced vasorelaxation by 32% compared with untreated controls (P < 0.05). In addition, HIV Nef significantly decreased eNOS expression in the vessels and HPAECs. HIV Nef at 10 ng/mL significantly decreased NO production in HPAECs by 21% compared with controls (P < 0.05). Furthermore, HIV Nef significantly increased superoxide anion production in porcine pulmonary arteries and HPAECs compared with controls (P < 0.05). Consequently, Mn (III) tetrakis porphyrin, a superoxide dismutase mimic, effectively blocked HIV Nef-induced vasomotor dysfunction and superoxide anion production. The specificity of HIV Nef action was confirmed by anti-Nef antibody blocking and Nef heat inactivation. CONCLUSIONS: HIV Nef protein significantly decreases endothelium-dependent vasorelaxation in porcine pulmonary arteries. It also reduces eNOS expression and induces oxidative stress in both porcine pulmonary arteries and HPAECs. This study demonstrates a new mechanism of HIV Nef, which causes endothelial dysfunction and may contribute to the human pulmonary artery disease in HIV-infected patients.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Enfermedades Vasculares/fisiopatología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Expresión Génica , Humanos , Modelos Animales , Óxido Nítrico Sintasa/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Superóxidos/metabolismo , Porcinos , Vasodilatación/efectos de los fármacos , Sistema Vasomotor/efectos de los fármacos
19.
J Biol Chem ; 283(48): 33191-8, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18826950

RESUMEN

Alveolar macrophages represent critical effector cells of innate immunity to infectious challenge in the lungs and recognize bacterial pathogens through pattern recognition receptors such as Toll-like receptors (TLRs). Phosphatidylinositol 3-kinase (PI3K) regulates TLR-mediated cytokine release, but whether HIV infection influences PI3K signaling pathway and alters TLR4-mediated macrophage response has not been investigated. In the current study, surface TLR4 expression were similar but TLR4 activation (lipid A, 10 microg/ml) resulted in lower TNF-alpha release by HIV+ human macrophages compared with healthy cells. Pharmacological inhibition of PI3K (LY294002) normalized TNF-alpha release in HIV+ macrophages and augments ERK1/2 mitogen-activated protein kinase phosphorylation in response to lipid A. Importantly, HIV+ macrophages demonstrated increased constitutive phosphatidylinositol 3,4,5-trisphosphate formation, increased phosphorylation of downstream signaling molecules Akt and glycogen synthase kinase-3beta (GSK-3beta) at Ser9, and reduced PTEN protein expression. As a functional assessment of GSK-3beta phosphorylation, TLR4-mediated interleukin-10 release was significantly higher in HIV+ human macrophages compared with healthy cells. Incubation of human macrophages with exogenous HIV Nef protein induced phosphorylation of Akt and GSK-3beta (whereas phosphorylation was reduced by PI3K inhibition) and promoted interleukin-10 release. Taken together, these data demonstrate increased constitutive activation of the PI3K signaling pathway in HIV+ macrophages and support the concept that PI3K activation (by HIV proteins such as Nef) may contribute to reduced TLR4-mediated TNF-alpha release in HIV+ human macrophages and impair host cell response to infectious challenge.


Asunto(s)
Infecciones por VIH/metabolismo , VIH-1 , Sistema de Señalización de MAP Quinasas , Macrófagos Alveolares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Cromonas/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/inmunología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Infecciones por VIH/inmunología , VIH-1/inmunología , Humanos , Interleucina-10/inmunología , Interleucina-10/metabolismo , Lípido A/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/virología , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfolinas/farmacología , Proteína Oncogénica v-akt/inmunología , Proteína Oncogénica v-akt/metabolismo , Fosfohidrolasa PTEN/inmunología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatos de Fosfatidilinositol/inmunología , Fosfatos de Fosfatidilinositol/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Fosforilación/inmunología , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Células U937 , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
20.
Virol J ; 5: 60, 2008 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-18489774

RESUMEN

BACKGROUND: HIV-1 mediated perturbation of the plasma membrane can produce an alteration in the transmembrane gradients of cations and other small molecules leading to cell death. Several HIV-1 proteins have been shown to perturb membrane permeability and ion transport. Xenopus laevis oocytes have few functional endogenous ion channels, and have proven useful as a system to examine direct effects of exogenously added proteins on ion transport. RESULTS: HIV-1 Nef induces alterations in the intracellular potassium concentration in CD4+ T-lymphoblastoid cells, but not intracellular pH. Two electrode voltage-clamp recording was used to determine that Nef did not form ion channel-like pores in Xenopus oocytes. CONCLUSION: These results suggest that HIV-1 Nef regulates intracellular ion concentrations indirectly, and may interact with membrane proteins such as ion channels to modify their electrical properties.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Líquido Intracelular/metabolismo , Potasio/metabolismo , Proteínas Reguladoras y Accesorias Virales/farmacología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Línea Celular Tumoral , Humanos , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Líquido Intracelular/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Proteínas Recombinantes/farmacología , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...