Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Virus Erad ; 9(3): 100341, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37663574

RESUMEN

Despite more than 20 years of combination antiretroviral therapy (cART), complete eradication of HIV remains a daunting task. While cART has been very effective in limiting new cycles of infection and keeping viral load below detectable levels with partial restoration of immune functions, it cannot provide a cure. Evidently, the interruption of cART leads to a quick rebound of the viral load within a few weeks. These consistent observations have revealed HIV ability to persist as an undetectable latent reservoir in a variety of tissues that remain insensitive to antiretroviral therapies. The 'Block-and-Lock' approach to drive latent cells into deep latency has emerged as a viable strategy to achieve a functional cure. It entails the development of latency-promoting agents with anti-HIV functions. Recent reports have suggested sulforaphane (SFN), an inducer of NRF-2 (nuclear erythroid 2-related factor 2)-mediated antioxidative signaling, to possess anti-HIV properties by restricting HIV replication at the early stages. However, the effect of SFN on the expression of integrated provirus remains unexplored. We have hypothesized that SFN may promote latency and prevent reactivation. Our results indicate that SFN can render latently infected monocytes and CD4+ T cells resistant to reactivation. SFN treatments antagonized the effects of known latency reactivating agents, tumor necrosis pactor (TNF-α), and phorbol 12-myristate 13-acetate (PMA), and caused a significant reduction in HIV transcription, viral RNA copies, and p24 levels. Furthermore, this block of reactivation was found to be mediated by SFN-induced NRF-2 signaling that specifically decreased the activation of NFκB signaling and thus restricted the HIV-1 promoter (5'LTR) activity. Overall, our study provides compelling evidence to highlight the latency-promoting potential of SFN which could be used in the 'Block-and-Lock' approach to achieve an HIV cure.

2.
Front Neurol ; 14: 1240300, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37719766

RESUMEN

Introduction: Due to advances in combined anti-retroviral treatment (cART), there is an increased burden of age-related cerebrovascular disease (CBVD), in people living with HIV (PWH). The underlying CNS injury can be assessed by measuring cerebral blood flow (CBF) and cerebrovascular reactivity (CVR). Methods: 35 treatment-naïve PWH and 53 HIV negative controls (HC) were enrolled in this study. Study participants underwent T1-weighted anatomical, pseudo-continuous arterial spin labeling, and resting-state functional MRI to obtain measures of CBF and CVR prior to starting cART treatment and at two-time points (12 weeks and 2 years) post-cART initiation. Controls were scanned at the baseline and 2-year visits. We also measured plasma levels of microparticles of endothelial and glial origin and well-known endothelial inflammation markers, ICAM-1 and VCAM-1, to assess HIV-associated endothelial inflammation and the interaction of these peripheral markers with brain neurovascular function. Results: HIV infection was found to be associated with reduced CVR and increased levels of endothelial and glial microparticles (MPs) prior to initiation of cART. Further, CVR correlated negatively with peripheral MP levels in PWH. Discussion: Our results suggest that while cART treatment has a beneficial effect on the neurovascular function after initiation, these benefits are suboptimal over time.

3.
J Neurovirol ; 28(2): 305-311, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35181862

RESUMEN

We previously showed that HIV-1 can alter the expression of tight junction proteins by downregulating Sonic hedgehog (Shh) signaling, thereby disrupting blood-brain barrier (BBB) integrity. In this study, we employed a conditional, CNS specific, Tat transgenic murine model to investigate if HIV-Tat exerts its neurotoxic effects by downregulating Shh signaling. Results indicate that Tat + mice exhibit significantly reduced expression of Shh and Gli1. HIV-Tat induced downregulation of Shh signaling correlated with disruption of BBB function and induced infiltration of peripheral leukocytes into the brain tissue. Further, our in vivo and in vitro experiments suggest that activation of Shh signaling can rescue detrimental effects of Tat on endothelial function by inducing the expression of junctional proteins and by decreasing the levels of inflammatory cytokines/chemokines.


Asunto(s)
Infecciones por VIH , VIH-1 , Productos del Gen tat del Virus de la Inmunodeficiencia Humana , Animales , Barrera Hematoencefálica/metabolismo , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , VIH-1/fisiología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Transducción de Señal , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
4.
J Leukoc Biol ; 109(4): 807-820, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32663904

RESUMEN

In addition to their role in hemostasis, platelets store numerous immunoregulatory molecules such as CD40L, TGFß, ß2-microglobulin, and IL-1ß and release them upon activation. Previous studies indicate that activated platelets form transient complexes with monocytes, especially in HIV infected individuals and induce a proinflammatory monocyte phenotype. Because monocytes can act as precursors of dendritic cells (DCs) during infection/inflammation as well as for generation of DC-based vaccine therapies, we evaluated the impact of activated platelets on monocyte differentiation into DCs. We observed that in vitro cultured DCs derived from platelet-monocyte complexes (PMCs) exhibit reduced levels of molecules critical to DC function (CD206, dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin, CD80, CD86, CCR7) and reduced antigen uptake capacity. DCs derived from PMCs also showed reduced ability to activate naïve CD4+ and CD8+ T cells, and secrete IL-12p70 in response to CD40L stimulation, resulting in decreased ability to promote type-1 immune responses to HIV antigens. Our results indicate that formation of complexes with activated platelets can suppress the development of functional DCs from such monocytes. Disruption of PMCs in vivo via antiplatelet drugs such as Clopidogrel/Prasugrel or the application of platelet-free monocytes for DCs generation in vitro, may be used to enhance immunization and augment the immune control of HIV.


Asunto(s)
Plaquetas/citología , Diferenciación Celular , Células Dendríticas/citología , Monocitos/citología , Adolescente , Adulto , Anciano , Movimiento Celular , Citocinas/metabolismo , Células Dendríticas/ultraestructura , Endotelio/metabolismo , Femenino , Infecciones por VIH/inmunología , Humanos , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Fenotipo , Linfocitos T/inmunología , Adulto Joven
5.
J Am Heart Assoc ; 9(17): e015998, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32819189

RESUMEN

Background Microvesicles are cell membrane-derived vesicles that have been shown to augment inflammation. Specifically, monocyte-derived microvesicles (MDMVs), which can express the coagulation protein tissue factor, contribute to thrombus formation and cardiovascular disease. People living with HIV experience higher prevalence of cardiovascular disease and also exhibit increased levels of plasma microvesicles. The process of microvesicle release has striking similarity to budding of enveloped viruses. The surface protein tetherin inhibits viral budding by physically tethering budding virus particles to cells. Hence, we investigated the role of tetherin in regulating the release of MDMVs during HIV infection. Methods and Results The plasma of aviremic HIV-infected individuals had increased levels of tissue factor + MDMVs, as measured by flow cytometry, and correlated to reduced tetherin expression on monocytes. Superresolution confocal and electron microscopy showed that tetherin localized at the site of budding MDMVs. Mechanistic studies revealed that the exposure of monocytes to HIV-encoded Tat triggered tetherin loss and subsequent rise in MDMV production. Overexpression of tetherin in monocytes led to morphologic changes in the pseudopodia directly underneath the MDMVs. Further, tetherin knockout mice demonstrated a higher number of circulating MDMVs and less time to bleeding cessation. Conclusions Our studies define a novel regulatory mechanism of MDMV release through tetherin and explore its contribution to the procoagulatory state that is frequently observed in people with HIV. Such insights could lead to improved therapies for individuals infected with HIV and also for those with cardiovascular disease.


Asunto(s)
Antivirales/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Micropartículas Derivadas de Células/genética , Infecciones por VIH/metabolismo , Adulto , Animales , Factores de Coagulación Sanguínea/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/farmacología , Antígeno 2 del Estroma de la Médula Ósea/ultraestructura , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/etiología , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/patología , Micropartículas Derivadas de Células/virología , Femenino , VIH/efectos de los fármacos , Infecciones por VIH/sangre , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , Humanos , Inmunohistoquímica/métodos , Inflamación/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Monocitos/metabolismo , Prevalencia , Proteínas Reguladoras y Accesorias Virales/metabolismo
6.
Sci Rep ; 9(1): 2781, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808933

RESUMEN

Neurotrophin signaling modulates the differentiation and function of mature blood cells. The expression of neurotrophin receptors and ligands by hematopoietic and stromal cells of the bone marrow indicates that neurotrophins have the potential to regulate hematopoietic cell fate decisions. This study investigates the role of neurotrophins and Tropomyosin receptor kinases (Trk) in the development of megakaryocytes (MKs) and their progeny cells, platelets. Results indicate that primary human MKs and MK cells lines, DAMI, Meg-01 and MO7e express TrkA, the primary receptor for Nerve Growth Factor (NGF) signaling. Activation of TrkA by NGF enhances the expansion of human MK progenitors (MKPs) and, to some extent, MKs. Whereas, inhibition of TrkA receptor by K252a leads to a 50% reduction in the number of both MKPs and MKs and is associated with a 3-fold increase in the production of platelets. In order to further confirm the role of TrkA signaling in platelet production, TrkA deficient DAMI cells were generated using CRISPR-Cas9 technology. Comparative analysis of wild-type and TrkA-deficient Dami cells revealed that loss of TrkA signaling induced apoptosis of MKs and increased platelet production. Overall, these findings support a novel role for TrkA signaling in platelet production and highlight its potential as therapeutic target for Thrombocytopenia.


Asunto(s)
Plaquetas/citología , Diferenciación Celular , Proliferación Celular , Megacariocitos/citología , Receptor trkA/metabolismo , Trombopoyesis , Apoptosis , Plaquetas/metabolismo , Puntos de Control del Ciclo Celular , Células Cultivadas , Humanos , Megacariocitos/metabolismo , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/genética , Transducción de Señal , Células del Estroma/citología , Células del Estroma/metabolismo
7.
Sci Rep ; 7(1): 9578, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28852071

RESUMEN

Infiltration of infected leukocytes culminates in establishment of a brain niche for Human Immunodeficiency Virus (HIV) during acute phase of infection, initiating an ongoing cascade of persistent viral replication and inflammation, that causes irreversible neuronal injury and HIV associated neurocognitive disease (HAND). In this study, humanized mice were treated with Smoothened Agonist (SAG), a Sonic Hedgehog (Shh) mimetic in order to fortify blood brain barrier (BBB) and dampen leukocyte extravasation into CNS during AHI. Results indicate that SAG treatment reduced viral burden in the CNS immediately after HIV transmission, but also conferred extended neuroprotection via increased BBB integrity (elevated levels of tight-junction protein, Claudin 5, and reduced S100B levels in periphery). These mice also showed healthier neurons with thick, uniform dendrites and reduced numbers of activated astrocytes. Additional in vitro experiments suggested SAG treatment was not associated with the establishment or reversal of latency in the target cells. Altogether, these findings validate neuroprotective role of Shh signaling and highlight the therapeutic potential of Shh mimetics against CNS complications associated with HIV infection. Further our results strongly demonstrate that pharmacological interventions to reduce leukocyte mobilization during early HIV infection, can provide prolonged neuroprotection, which might significantly delay the onset of HAND.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central/patología , Enfermedades Virales del Sistema Nervioso Central/virología , Quimiotaxis de Leucocito/efectos de los fármacos , Ciclohexilaminas/farmacología , Infecciones por VIH/patología , Infecciones por VIH/virología , Leucocitos/efectos de los fármacos , Leucocitos/patología , Imitación Molecular , Tiofenos/farmacología , Enfermedad Aguda , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Proteínas Hedgehog/metabolismo , Memoria Inmunológica , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Development ; 144(10): 1820-1830, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28428215

RESUMEN

The maternally methylated KvDMR1 ICR regulates imprinted expression of a cluster of maternally expressed genes on human chromosome 11p15.5. Disruption of imprinting leads to Beckwith-Wiedemann syndrome (BWS), an overgrowth and cancer predisposition condition. In the majority of individuals with BWS, maternal-specific methylation at KvDMR1 is absent and genes under its control are repressed. We analyzed a mouse model carrying a poly(A) truncation cassette inserted to prevent RNA transcripts from elongation through KvDMR1. Maternal inheritance of this mutation resulted in absence of DNA methylation at KvDMR1, which led to biallelic expression of Kcnq1ot1 and suppression of maternally expressed genes. This study provides further evidence that transcription is required for establishment of methylation at maternal gametic DMRs. More importantly, this mouse model recapitulates the molecular phenotypic characteristics of the most common form of BWS, including loss of methylation at KvDMR1 and biallelic repression of Cdkn1c, suggesting that deficiency of maternal transcription through KvDMR1 may be an underlying cause of some BWS cases.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Metilación de ADN , Silenciador del Gen , ARN Largo no Codificante/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , ARN Mensajero Almacenado/genética , Transcripción Genética
9.
J Neurovirol ; 23(3): 347-357, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28116673

RESUMEN

The neuroteratogenic nature of Zika Virus (ZIKV) infection has converted what would have been a tropical disease into a global threat. Zika is transmitted vertically via infected placental cells especially in the first and second trimesters. In the developing central nervous system (CNS), ZIKV can infect and induce apoptosis of neural progenitor cells subsequently causing microcephaly as well as other neuronal complications in infants. Its ability to infect multiple cell types (placental, dermal, and neural) and increased environmental stability as compared to other flaviviruses (FVs) has broadened the transmission routes for ZIKV infection from vector-mediated to transmitted via body fluids. To further complicate the matters, it is genetically similar (about 40%) with the four serotypes of dengue virus (DENV), so much so that it can almost be called a fifth DENV serotype. This homology poses the risk of causing cross-reactive immune responses and subsequent antibody-dependent enhancement (ADE) of infection in case of secondary infections or for immunized individuals. All of these factors complicate the development of a single preventive vaccine candidate or a pharmacological intervention that will completely eliminate or cure ZIKV infection. We discuss all of these factors in detail in this review and conclude that a combinatorial approach including immunization and treatment might prove to be the winning strategy.


Asunto(s)
Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Microcefalia/prevención & control , Complicaciones Infecciosas del Embarazo/prevención & control , Dengue Grave/prevención & control , Vacunas Virales/administración & dosificación , Infección por el Virus Zika/prevención & control , Virus Zika/patogenicidad , Antivirales/uso terapéutico , Bacteriocinas/uso terapéutico , Terapia Combinada , Ciclohexilaminas/uso terapéutico , Virus del Dengue/efectos de los fármacos , Virus del Dengue/patogenicidad , Virus del Dengue/fisiología , Femenino , Feto , Humanos , Microcefalia/inmunología , Microcefalia/virología , Péptidos/uso terapéutico , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , Dengue Grave/inmunología , Dengue Grave/transmisión , Dengue Grave/virología , Tiofenos/uso terapéutico , Vacunas Virales/biosíntesis , Virus Zika/efectos de los fármacos , Virus Zika/fisiología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología
10.
PLoS One ; 11(6): e0157115, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27270236

RESUMEN

Platelets play an essential role in hemostasis and wound healing by facilitating thrombus formation at sites of injury. Platelets also mediate inflammation and contain several pro-inflammatory molecules including cytokines and chemokines that mediate leukocyte recruitment and activation. Not surprisingly, platelet dysfunction is known to contribute to several inflammatory disorders. Antiplatelet therapies, such as aspirin, adenosine diphosphate (ADP) antagonists, glycoprotein IIb/IIIa (GPIIb/IIIa) inhibitors, and anticoagulants such as warfarin, dampen platelet activity at the risk of unwarranted bleeding. Thus, the development of drugs that reduce platelet-mediated inflammation without interfering with thrombus formation is of importance to combat platelet-associated disorders. We have shown here for the first time that the tetracycline antibiotic, minocycline, administered to HIV-infected individuals reduces plasma levels of soluble CD40L and platelet factor 4 levels, host molecules predominately released by platelets. Minocycline reduced the activation of isolated platelets in the presence of the potent platelet activator, thrombin, as measured by ELISA and flow cytometry. Platelet degranulation was reduced upon exposure to minocycline as shown by mepacrine retention and flow cytometry. However, minocycline had no effect on spreading, aggregation, GPIIb/IIIa activation, or in vivo thrombus formation. Lastly, immunoblot analysis suggests that the antiplatelet activity of minocycline is likely mediated by inhibition of mixed lineage kinase 3 (MLK3)-p38 MAPK signaling axis and loss of p38 activity. Our findings provide a better understanding of platelet biology and a novel repurposing of an established antibiotic, minocycline, to specifically reduce platelet granule release without affecting thrombosis, which may yield insights in generating novel, specific antiplatelet therapies.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Minociclina/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Regulación hacia Abajo/efectos de los fármacos , Humanos , Quinasas Quinasa Quinasa PAM/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
11.
Sci Rep ; 6: 26876, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27241024

RESUMEN

Human Immunodeficiency Virus type-1 (HIV)-associated neurocognitive disorder is characterized by recruitment of activated/infected leukocytes into the CNS via disrupted Blood Brain Barrier (BBB) that contributes to persistent neuro-inflammation. In this report, humanized NOD/scid-IL2Rγc(null) mice were used to establish that impaired Sonic hedgehog (Shh) signaling is associated with loss of BBB function and neurological damage, and that modulating Shh signaling can rescue these detrimental effects. Plasma viral load, p24 levels and CD4(+) T cells were measured as markers of productive HIV infection. These mice also showed impaired exclusion of Evans blue dye from the brain, increased plasma levels of S100B, an astrocytic protein, and down-regulation of tight junction proteins Occludin and Claudin5, collectively indicating BBB dysfunction. Further, brain tissue from HIV(+) mice indicated reduced synaptic density, neuronal atrophy, microglial activation, and astrocytosis. Importantly, reduced expression of Shh and Gli1 was also observed in these mice, demonstrating diminished Shh signaling. Administration of Shh mimetic, smoothened agonist (SAG) restored BBB integrity and also abated the neuropathology in infected mice. Together, our results suggest a neuroprotective role for Shh signaling in the context of HIV infection, underscoring the therapeutic potential of SAG in controlling HAND pathogenesis.


Asunto(s)
Complejo SIDA Demencia/tratamiento farmacológico , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Ciclohexilaminas/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Tiofenos/farmacología , Complejo SIDA Demencia/genética , Complejo SIDA Demencia/patología , Complejo SIDA Demencia/virología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Astrocitos/virología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Recuento de Células , Claudina-5/genética , Claudina-5/metabolismo , Regulación de la Expresión Génica , VIH-1/patogenicidad , VIH-1/fisiología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuroglía/patología , Neuroglía/virología , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , Ocludina/genética , Ocludina/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Transducción de Señal , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
12.
J Neurovirol ; 21(2): 186-98, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25636783

RESUMEN

Long-term persistence of human immunodeficiency virus type-1 (HIV) in the central nervous system (CNS) results in mild to severe neurocognitive impairment in a significant proportion of the HIV-infected population. These neurological deficits are known as HIV-associated neurocognitive disorders (HAND). Microglia are CNS-resident immune cells that are directly infected by HIV and consequently secrete proinflammatory molecules that contribute to HIV-induced neuroinflammation. Indeed, the number of activated macrophage and microglia in the brain is more highly correlated with cognitive impairment than the amount of neuronal apoptosis. Ankyrin-rich membrane spanning protein (ARMS/Kidins220) is a multidomain transmembrane protein that is involved with neurotrophin signaling in the CNS. We have previously established the role of ARMS in mediating neuronal survival via a neurotrophin-dependent mechanism. Recent reports also have suggested that ARMS is involved with cell signaling in multiple immune cell types. In this study, we aim to investigate the role of ARMS in HIV Tat-mediated microglial cell activation by employing in vitro methods. Following ARMS depletion by a lentivirus encoding ARMS-specific short hairpin RNA (shRNA), we observed a marked reduction in the HIV Tat-induced proinflammatory response, associated with loss of tumor necrosis factor alpha production and nuclear factor-kappa B (NF-κB) activation. Furthermore, co-immunoprecipitation studies suggested that ARMS physically interacts with inhibitory kappa B kinase subunits in order to facilitate NF-κB activation. Our results establish the role of ARMS in microglial activation by HIV Tat and warrant additional studies to better understand these molecular mechanisms, which may uncover novel therapeutic targets for the treatment of HAND.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Proteínas de la Membrana/metabolismo , Microglía/virología , Proteínas del Tejido Nervioso/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Complejo SIDA Demencia/inmunología , Animales , Línea Celular , Técnica del Anticuerpo Fluorescente , Células HEK293 , VIH-1 , Humanos , Immunoblotting , Inmunoprecipitación , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Microglía/inmunología , Microglía/metabolismo , Transfección
13.
J Immunol ; 192(10): 4674-84, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24729609

RESUMEN

HIV-1-associated neuroinflammation persists even with effective combined antiretroviral therapy, and it is associated with the presence of activated monocytes/macrophages within the CNS. To infiltrate the CNS, monocytes transmigrate across the selectively permeable blood-brain barrier, which is compromised during HIV-1 infection. Interestingly, platelet-derived excess soluble CD40 ligand found in the plasma and cerebrospinal fluid of HIV-1-infected individuals with cognitive impairment has previously been implicated in increased blood-brain barrier permeability. In this study we show that soluble CD40 ligand also promotes the formation of complexes between inflammatory monocytes and activated platelets (PMCs), which are detected by flow cytometry as monocytes that express excess of CD61, a platelet marker, and that these complexes are increased in individuals with HIV-1 infection. PMCs exhibit an enhanced ability to adhere to human brain microvascular endothelial cells as compared with monocytes alone, and they migrate across the transendothelial barrier. These complexes can be found marginalized in the lumen of postcapillary venules in postmortem brain tissue derived from cases of HIV-1-associated encephalitis. The extravasation of monocytes across the brain endothelium may exacerbate neuroinflammation, indicating that enhancing this event via platelet interaction may be a contributing factor in the development of cognitive impairment. Thus, dampening platelet activation, and in turn PMC formation, with antiplatelet agents may prove beneficial in developing adjunctive therapies for use in combination with combined antiretroviral therapy in an effort to reduce HIV-1-associated neurologic deficit.


Asunto(s)
Plaquetas/inmunología , Barrera Hematoencefálica/inmunología , Encefalitis/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Monocitos/inmunología , Adulto , Plaquetas/patología , Barrera Hematoencefálica/patología , Ligando de CD40/inmunología , Circulación Cerebrovascular/inmunología , Encefalitis/etiología , Encefalitis/patología , Células Endoteliales/inmunología , Células Endoteliales/patología , Femenino , Infecciones por VIH/complicaciones , Infecciones por VIH/patología , Humanos , Integrina beta3/inmunología , Masculino , Persona de Mediana Edad , Monocitos/patología
14.
Biochem Biophys Res Commun ; 379(3): 710-5, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19133234

RESUMEN

The heme-regulated eIF-2alpha kinase, also called the heme-regulated inhibitor (HRI), is activated under various cytoplasmic stresses in reticulocytes leading to inhibition of initiation of protein synthesis. Our previous studies indicated that the promoter activity and expression of the human HRI (hHRI) increase in human K562 cells during heat shock and lead exposure. Contrary to this, hemin chloride which inactivates the kinase, downregulates HRI expression. Here, we attempted to understand the mechanism of regulation of hHRI expression in the lead- and hemin-exposed cells. Our results demonstrate the involvement of two transcription factors, Elk-1 and MZF-1 in regulating HRI expression. Chromatin immunoprecipitation assays established further that Elk-1 is involved in upregulating HRI expression during stress along with a co-activator p300, while MZF-1 along with HDAC-1 is instrumental in its downregulation during hemin treatment. We also demonstrate the involvement of ERK pathway in activating Elk-1 during stress resulting in an over expression of hHRI.


Asunto(s)
Regulación de la Expresión Génica , Estrés Fisiológico/genética , eIF-2 Quinasa/genética , Proteína Elk-1 con Dominio ets/metabolismo , Secuencia de Bases , Inmunoprecipitación de Cromatina , Clonación Molecular , Hemina/toxicidad , Histona Desacetilasa 1 , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Plomo/toxicidad , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Fosforilación , Regiones Promotoras Genéticas , Factores de Transcripción p300-CBP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...