Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Oncogene ; 41(47): 5092-5106, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36245058

RESUMEN

Gasdermin D (GSDMD) has recently been identified as a cytoplasmic effector protein that plays a central role in pyroptosis of immune cells. However, GSDMD is a universally expressed protein, and its function beyond pyroptosis, especially in cancer cells, has not been well characterized. Here, we report that predominant localization of GSDMD in the nucleoplasm in vivo indicates favorable clinical outcomes in colorectal cancer, while a lack of nuclear localization of GSDMD is associated with poor outcomes. Nuclear GSDMD, rather than cytoplasmic GSDMD, inhibits cell growth and promotes apoptosis in colorectal cancer. Hypoxia in the tumor microenvironment accounts for mild or moderate nuclear translocation of GSDMD in vivo. Under the stimulation of chemotherapy drugs, nuclear GSDMD promotes apoptosis via regulation of its subcellular distribution rather than pyroptosis-related cleavage. After nuclear translocation, GSDMD interacts with PARP-1 to dramatically inhibit its DNA damage repair-related function by functioning like the PARP inhibitor olaparib, thus forming a "hypoxia/chemotherapy-GSDMD nuclear translocation-PARP-1 blockade-DNA damage and apoptosis" axis. This study redefines the pyroptosis-independent function of GSDMD and suggests that the subcellular localization of GSDMD may serve as a molecular indicator of clinical outcomes and a promising therapeutic target in colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Piroptosis , Humanos , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Proteínas de Unión a Fosfato/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hipoxia , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Microambiente Tumoral
2.
Cell Death Dis ; 8(1): e2542, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28079886

RESUMEN

Human immunodeficiency virus-associated neurological disorders (HANDs) affect the majority of AIDS patients and are a significant problem among HIV-1-infected individuals who live longer because of combined anti-retroviral therapies. HIV-1 utilizes a number of viral proteins and subsequent cytokine inductions to unleash its toxicity on neurons. Among HIV-1 viral proteins, Nef is a small protein expressed abundantly in astrocytes of HIV-1-infected brains and has been suggested to have a role in the pathogenesis of HAND. In order to explore its effect in the central nervous system, HIV-1 Nef was expressed in primary human fetal astrocytes (PHFAs) using an adenovirus. Our results revealed that HIV-1 Nef is released in extracellular vesicles (EVs) derived from PHFA cells expressing the protein. Interestingly, HIV-1 Nef release in EVs was enriched significantly when the cells were treated with autophagy activators perifosine, tomaxifen, MG-132, and autophagy inhibitors LY294002 and wortmannin suggesting a novel role of autophagy signaling in HIV-1 Nef release from astrocytes. Next, Nef-carrying EVs were purified from astrocyte cultures and neurotoxic effects on neurons were analyzed. We observed that HIV-1 Nef-containing EVs were readily taken up by neurons as demonstrated by immunocytochemistry and immunoblotting. Furthermore, treatment of neurons with Nef-carrying EVs induced oxidative stress as evidenced by a decrease in glutathione levels. To further investigate its neurotoxic effects, we expressed HIV-1 Nef in primary neurons by adenoviral transduction. Intracellular expression of HIV-1 Nef caused axonal and neurite degeneration of neurons. Furthermore, expression of HIV-1 Nef decreased the levels of phospho-tau while enhancing total tau in primary neurons. In addition, treatment of primary neurons with Nef-carrying EVs suppressed functional neuronal action potential assessed by multielectrode array studies. Collectively, these data suggested that HIV-1 Nef can be a formidable contributor to neurotoxicity along with other factors, which leads to HAND in HIV-1-infected AIDS patients.


Asunto(s)
Vesículas Extracelulares/genética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Síndromes de Neurotoxicidad/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/virología , Autofagia/efectos de los fármacos , Autofagia/genética , Vesículas Extracelulares/virología , Feto/virología , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/virología , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/virología , Cultivo Primario de Células , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/administración & dosificación
3.
J Cell Physiol ; 232(3): 517-525, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27249540

RESUMEN

Identification of cellular proteins, in addition to already known transcription factors such as NF-κB, Sp1, C-EBPß, NFAT, ATF/CREB, and LEF-1, which interact with the HIV-1 LTR, is critical in understanding the mechanism of HIV-1 replication in monocytes/macrophages. Our studies demonstrate upregulation of pyruvate kinase isoform M2 (PKM2) expression during HIV-1SF162 infection of monocyte/macrophages and reactivation of HIV-1 in U1 cells, a macrophage model of latency. We observed that HIV-1SF162 infection of monocyte/macrophages and reactivation of HIV-1 in U1 cells by PMA resulted in increased levels of nuclear PKM2 compared to PMA-induced U937 cells. Furthermore, there was a significant increase in the nuclear dimeric form of PKM2 in the PMA-induced U1 cells in comparison to PMA-induced U937 cells. We focused on understanding the potential role of PKM2 in HIV-1 LTR transactivation. Chromatin immunoprecipitation (ChIP) analysis in PMA-activated U1 and TZM-bl cells demonstrated the interaction of PKM2 with the HIV-1 LTR. Our studies show that overexpression of PKM2 results in transactivation of HIV-1 LTR-luciferase reporter in U937, U-87 MG, and TZM-bl cells. Using various truncated constructs of the HIV-1 LTR, we mapped the region spanning -120 bp to -80 bp to be essential for PKM2-mediated transactivation. This region contains the NF-κB binding site and deletion of this site attenuated PKM2-mediated activation of HIV-1 LTR. Immunoprecipitation experiments using U1 cell lysates demonstrated a physical interaction between PKM2 and the p65 subunit of NF-κB. These observations demonstrate for the first time that PKM2 is a transcriptional co-activator of HIV-1 LTR. J. Cell. Physiol. 232: 517-525, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Portadoras/metabolismo , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Proteínas de la Membrana/metabolismo , Hormonas Tiroideas/metabolismo , Sitios de Unión , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , VIH-1/efectos de los fármacos , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Modelos Biológicos , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Transporte de Proteínas/efectos de los fármacos , Eliminación de Secuencia , Acetato de Tetradecanoilforbol/farmacología , Factor de Transcripción ReIA/metabolismo , Células U937 , Replicación Viral/efectos de los fármacos , Proteínas de Unión a Hormona Tiroide
4.
Cell Cycle ; 14(7): 980-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25602755

RESUMEN

Viruses have developed various strategies to protect infected cells from apoptosis. HIV-1 infected macrophages are long-lived and considered reservoirs for HIV-1. One significant deciding factor between cell survival and cell death is glucose metabolism. We hypothesized that HIV-1 protects infected macrophages from apoptosis in part by modulating the host glycolytic pathway specifically by regulating hexokinase-1 (HK-1) an enzyme that converts glucose to glucose-6-phosphate. Therefore, we analyzed the regulation of HK-1 in HIV-1 infected PBMCs, and in a chronically HIV-1 infected monocyte-like cell line, U1. Our results demonstrate that HIV-1 induces a robust increase in HK-1 expression. Surprisingly, hexokinase enzymatic activity was significantly inhibited in HIV-1 infected PBMCs and in PMA differentiated U1 cells. Interestingly, we observed increased levels of mitochondria-bound HK-1 in PMA induced U1 cells and in the HIV-1 accessory protein, viral protein R (Vpr) transduced U937 cell derived macrophages. Dissociation of HK-1 from mitochondria in U1 cells using a pharmacological agent, clotrimazole (CTZ) induced mitochondrial membrane depolarization and caspase-3/7 mediated apoptosis. Dissociation of HK-1 from mitochondria in Vpr transduced U937 also activated caspase-3/7 activity. These observations indicate that HK-1 plays a non-metabolic role in HIV-1 infected macrophages by binding to mitochondria thereby maintaining mitochondrial integrity. These results suggest that targeting the interaction of HK-1 with the mitochondria to induce apoptosis in persistently infected macrophages may prove beneficial in purging the macrophage HIV reservoir.


Asunto(s)
Infecciones por VIH/enzimología , VIH-1/fisiología , Hexoquinasa/fisiología , Macrófagos/enzimología , Línea Celular Tumoral , Supervivencia Celular , Infecciones por VIH/virología , Humanos , Macrófagos/virología , Potencial de la Membrana Mitocondrial , Mitocondrias/enzimología , Monocitos/enzimología , Transporte de Proteínas
5.
PLoS One ; 9(5): e98304, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24847939

RESUMEN

Transcription from the HIV-1 promoter is controlled by a series of ubiquitous and inducible cellular proteins with the ability to enter the nucleus and interact with the promoter. A DNA sequence spanning nucleotides -120 to -80, which supports the association of the inducible NF-κB transcription factor, has received much attention. Here we demonstrate that the interplay between Rad51, a key regulator of the homologous recombination pathway of DNA repair and whose level is induced upon HIV-1 infection, with the NF-κB pathway, augments transcription of the viral promoter. Evidently, stimulation of the NF-κB pathway by PMA and/or TSA promotes association of Rad51 with the LTR DNA sequence and that the p65 subunit of NF-κB is important for this event. Our results also demonstrate that, similar to p65, Rad51 utilizes the NF-κB pathway to position itself in the nucleus as ectopic expression of an IκB mutant impedes its nuclear appearance and transcriptional activity upon the HIV-1 LTR. Treatment of peripheral blood mononuclear cells with small molecules that inhibit Rad51 activity results in greater than 50% decrease in the HIV-1 infection of cells. These observations provide evidence for the involvement of DNA repair factors in control of HIV-1 gene activation and offer a new avenue for the development of anti-viral therapeutics that affect viral gene transcription in latently infected cells.


Asunto(s)
VIH-1/fisiología , Recombinasa Rad51/metabolismo , Factor de Transcripción ReIA/metabolismo , Astrocitos/citología , Astrocitos/virología , Encéfalo/embriología , Núcleo Celular/metabolismo , Células Cultivadas , Regulación Viral de la Expresión Génica , Infecciones por VIH/metabolismo , Duplicado del Terminal Largo de VIH , Humanos , Leucocitos Mononucleares/citología , Mutación , Regiones Promotoras Genéticas , Recombinasa Rad51/genética , Recombinación Genética , Activación Transcripcional
6.
J Cell Physiol ; 229(8): 1039-46, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24648009

RESUMEN

The insulin-like growth factor-1 (IGF-1) signaling pathway plays an important role in neuronal cell differentiation. Recent studies have shown that IGF-1 has the capacity to counteract the retraction of neuronal processes in response to inflammatory cytokines such as TNF-α, which is a known factor for neuronal injury in the central nervous system. This event is thought to be mediated via interference of TNF-α-induced interaction of ß1-integrin with insulin receptor substrate-1 (IRS-1). Here, we demonstrate the interaction of IRS-1 with disintegrin and metalloproteinase ADAM10 through the N-terminal domain of IRS-1 and that this is involved in the regulation of neurite extension and retraction by IGF-1 and TNF-α, respectively. PC12 cells expressing the N-terminal domain show enhanced neurite extension after IGF-1 treatment and reduced neurite depletion relative to control cells after TNF-α treatment. The level of ADAM10 was found to be increased in immunohistochemical studies of HIV encephalitis clinical samples and is present with TNF-α and TNFR1 in both astrocytes and neurons. Altogether, these observations suggest a role for ADAM10 in the mechanism for IGF1/IRS-1 signaling pathway in sustaining the stability of neuronal processes.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/fisiología , Proteínas ADAM/genética , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Infecciones por VIH/complicaciones , Infecciones por VIH/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Integrina beta1/genética , Integrina beta1/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratas , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
7.
PLoS One ; 8(8): e69623, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936341

RESUMEN

Independent research groups reported that DING protein homologues isolated from bacterial, plant and human cells demonstrate the anti-HIV-1 activity. This might indicate that diverse organisms utilize a DING-mediated broad-range protective innate immunity response to pathogen invasion, and that this mechanism is effective also against HIV-1. We performed structural analyses and evaluated the anti-HIV-1 activity for four DING protein homologues isolated from different species. Our data show that bacterial PfluDING, plant p38SJ (pDING), human phosphate binding protein (HPBP) and human extracellular DING from CD4 T cells (X-DING-CD4) share high degrees of structure and sequence homology. According to earlier reports on the anti-HIV-1 activity of pDING and X-DING-CD4, other members of this protein family from bacteria and humans were able to block transcription of HIV-1 and replication of virus in cell based assays. The efficacy studies for DING-mediated HIV-1 LTR and HIV-1 replication blocking activity showed that the LTR transcription inhibitory concentration 50 (IC50) values ranged from 0.052-0.449 ng/ml; and the HIV-1 replication IC50 values ranged from 0.075-0.311 ng/ml. Treatment of cells with DING protein alters the interaction between p65-NF-κB and HIV-1 LTR. Our data suggest that DING proteins may be part of an innate immunity defense against pathogen invasion; the conserved structure and activity makes them appealing candidates for development of a novel therapeutics targeting HIV-1 transcription.


Asunto(s)
VIH-1/genética , Filogenia , Complejo Represivo Polycomb 1/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Homología de Secuencia de Aminoácido , Secuencias Repetidas Terminales/genética , Transcripción Genética/efectos de los fármacos , Secuencia de Aminoácidos , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Línea Celular Tumoral , Secuencia Conservada , VIH-1/efectos de los fármacos , VIH-1/fisiología , Humanos , Hypericum , Concentración 50 Inhibidora , Modelos Moleculares , Datos de Secuencia Molecular , Subunidad p50 de NF-kappa B/química , Complejo Represivo Polycomb 1/química , Regiones Promotoras Genéticas/genética , Multimerización de Proteína/efectos de los fármacos , Estructura Cuaternaria de Proteína , Pseudomonas fluorescens , Factor de Transcripción ReIA/química , Replicación Viral/efectos de los fármacos
8.
PLoS One ; 8(7): e68376, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874603

RESUMEN

Human immunodeficiency virus type 1 encoded viral protein Vpr is essential for infection of macrophages by HIV-1. Furthermore, these macrophages are resistant to cell death and are viral reservoir. However, the impact of Vpr on the macrophage proteome is yet to be comprehended. The goal of the present study was to use a stable-isotope labeling by amino acids in cell culture (SILAC) coupled with mass spectrometry-based proteomics approach to characterize the Vpr response in macrophages. Cultured human monocytic cells, U937, were differentiated into macrophages and transduced with adenovirus construct harboring the Vpr gene. More than 600 proteins were quantified in SILAC coupled with LC-MS/MS approach, among which 136 were significantly altered upon Vpr overexpression in macrophages. Quantified proteins were selected and clustered by biological functions, pathway and network analysis using Ingenuity computational pathway analysis. The proteomic data illustrating increase in abundance of enzymes in the glycolytic pathway (pentose phosphate and pyruvate metabolism) was further validated by western blot analysis. In addition, the proteomic data demonstrate down regulation of some key mitochondrial enzymes such as glutamate dehydrogenase 2 (GLUD2), adenylate kinase 2 (AK2) and transketolase (TKT). Based on these observations we postulate that HIV-1 hijacks the macrophage glucose metabolism pathway via the Vpr-hypoxia inducible factor 1 alpha (HIF-1 alpha) axis to induce expression of hexokinase (HK), glucose-6-phosphate dehyrogenase (G6PD) and pyruvate kinase muscle type 2 (PKM2) that facilitates viral replication and biogenesis, and long-term survival of macrophages. Furthermore, dysregulation of mitochondrial glutamate metabolism in macrophages can contribute to neurodegeneration via neuroexcitotoxic mechanisms in the context of NeuroAIDS.


Asunto(s)
Marcaje Isotópico/métodos , Macrófagos/metabolismo , Redes y Vías Metabólicas , Proteómica/métodos , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Western Blotting , Biología Computacional , Glucólisis , Humanos , Modelos Biológicos , Mapas de Interacción de Proteínas , Proteoma/metabolismo , Reproducibilidad de los Resultados , Transducción Genética , Células U937
9.
PLoS One ; 8(6): e65947, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23805194

RESUMEN

JC virus (JCV), a common human polyomavirus, is the etiological agent of the demyelinating disease, progressive multifocal leukoencephalopathy (PML). In addition to its role in PML, studies have demonstrated the transforming ability of the JCV early protein, T-antigen, and its association with some human cancers. JCV infection occurs in childhood and latent virus is thought to be maintained within the bone marrow, which harbors cells of hematopoietic and non-hematopoietic lineages. Here we show that non-hematopoietic mesenchymal stem cells (MSCs) isolated from the bone marrow of JCV T-antigen transgenic mice give rise to JCV T-antigen positive cells when cultured under neural conditions. JCV T-antigen positive cells exhibited neural crest characteristics and demonstrated p75, SOX-10 and nestin positivity. When cultured in conditions typical for mesenchymal cells, a population of T-antigen negative cells, which did not express neural crest markers arose from the MSCs. JCV T-antigen positive cells could be cultured long-term while maintaining their neural crest characteristics. When these cells were induced to differentiate into neural crest derivatives, JCV T-antigen was downregulated in cells differentiating into bone and maintained in glial cells expressing GFAP and S100. We conclude that JCV T-antigen can be stably expressed within a fraction of bone marrow cells differentiating along the neural crest/glial lineage when cultured in vitro. These findings identify a cell population within the bone marrow permissible for JCV early gene expression suggesting the possibility that these cells could support persistent viral infection and thus provide clues toward understanding the role of the bone marrow in JCV latency and reactivation. Further, our data provides an excellent experimental model system for studying the cell-type specificity of JCV T-antigen expression, the role of bone marrow-derived stem cells in the pathogenesis of JCV-related diseases and the opportunities for the use of this model in development of therapeutic strategies.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Virus JC/genética , Cresta Neural/metabolismo , Animales , Antígenos Virales de Tumores/genética , Células de la Médula Ósea/citología , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Transgénicos , Nestina/metabolismo , Cresta Neural/citología , Neuroglía/citología , Neuroglía/metabolismo , Osteogénesis , Proteínas S100/metabolismo , Factores de Transcripción SOXE/metabolismo
10.
J Neurooncol ; 107(2): 247-56, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22052333

RESUMEN

Malignant gliomas are a highly aggressive type of brain tumor with extremely poor prognosis. These tumors are highly invasive and are often surgically incurable and resistant to chemotherapeutics and radiotherapy. Thus, novel therapies that target pathways involved in growth and survival of the tumor cells are required for the treatment of this class of brain tumors. Previous studies revealed that epidermal growth factor receptor and extracellular-signal-regulated kinases (ERKs), which are involved in the induction of cell proliferation, are activated in the most aggressive type of glioma, i.e. glioblastoma multiforme (GBM). In fact, GBMs with increased levels of ERK activity exhibit a more aggressive phenotype than the others with moderate ERK activity, pointing to the importance of ERK and its kinase activity in the development and progression of these tumors. In this study, we have evaluated the effect of p38SJ, a novel member of the DING family of proteins, derived from Hypericum perforatum calluses, on the growth of malignant glioma cell lines, T98G and U-87MG by focusing on cell cycle and signaling pathways controlled by phosphorylation of various regulatory proteins including ERK. p38SJ, which exhibits profound phosphatase activity, shows the capacity to affect the phosphorylation status of several important kinases modulating signaling pathways, and cell growth and proliferation. Our results demonstrate that p38SJ reduces glioma cell viability and arrests cell cycle progression at G0/G1. The observed growth inhibitory effect of p38SJ is likely mediated by the downregulation of several cell cycle gatekeeper proteins, including cyclin E, Cdc2, and E2F-1. These results suggest that p38SJ may serve as a potential candidate for development of a therapeutic agent for the direct treatment of malignant gliomas and/or as a potential radiosensitizer.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/fisiopatología , Ubiquitina-Proteína Ligasas/farmacología , Animales , Proteína Quinasa CDC2 , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Transformada , Línea Celular Tumoral , Ciclina B/metabolismo , Ciclina E/metabolismo , Quinasas Ciclina-Dependientes , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F/metabolismo , Endopeptidasa K/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Glioblastoma/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hypericum/química , Ratones , Preparaciones de Plantas/farmacología , Complejo Represivo Polycomb 1 , Sales de Tetrazolio , Tiazoles , Factores de Tiempo , Transfección , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
11.
J Cell Physiol ; 227(8): 3119-27, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22034072

RESUMEN

An indispensable role for oligodendrocytes in the protection of axon function and promotion of neuronal survival is strongly supported by the finding of progressive neuron/axon degeneration in human neurological diseases that affect oligodendrocytes. Imaging and pathological studies of the CNS have shown the presence of neuroaxonal injury in progressive multifocal leukoencephalopathy (PML), a demyelinating disease of the CNS, resulting from destruction of oligodendrocytes upon productive replication of the pathogenic neurotropic polyomavirus JC. Here, we examined the extracellular factors involved in communication between oligodendrocytes and neurons. Culturing cortical neurons with conditioned medium (CM) from rat CG4 oligodendrocytic cells that express the JCV agnoprotein showed that CXCL5/LIX, which is a chemokine closely related to the human CXCL5/ENA78 and CXCL6/GCP-2 chemokines, is essential for neuronal cell survival. We found that in CM from agnoprotein-producing CG-4 cells level of CXC5/LIX is decreased compared to control cells. We also demonstrated that a reduced expression of CXCL5/LIX by CG4 GFP-Agno cells triggered a cascade of signaling events in cortical neurons. Analysis of mitogen-activated protein kinases (MAPK) and glycogen synthase kinase (GSK3) pathways showed that they are involved in mechanisms of neuronal apoptosis in response to the depletion of CXCL5/LIX signaling. These data suggest that agnoprotein-induced dysregulation of chemokine production by oligodendrocytes may contribute to neuronal/axonal injury in the pathogenesis of PML lesions.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL5/metabolismo , Leucoencefalopatía Multifocal Progresiva/metabolismo , Neuronas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Células Cultivadas , Medios de Cultivo Condicionados , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Virus JC/metabolismo , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva/patología , Leucoencefalopatía Multifocal Progresiva/virología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neuronas/citología , Oligodendroglía/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores
12.
J Neurosci Res ; 89(2): 183-98, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21162126

RESUMEN

In HIV patients, antiretroviral medications trigger metabolic abnormalities, including insulin resistance. In addition, the inflammatory cytokine tumor necrosis factor-α (TNFα), which is elevated in human immunodeficiency virus encephalitis (HIVE), also induces insulin resistance and inflicts neuronal damage in vitro. In differentiated PC12 cells and rat cortical neurons, high glucose (HG; 25 mM) triggers reactive oxygen species (ROS) accumulation, contributing to the retraction of neuronal processes, with only a minimal involvement of neuronal apoptosis. In the presence of TNFα, HG-treated neurons undergo massive apoptosis. Because mammalian homolog of the Forkhead family of transcription factors, Forkhead box O transcription factor 3a (FOXO3a), controls ROS metabolism, we asked whether FOXO3a could affect the fate of differentiated neurons in the paradigm of HIVE. We observed FOXO3a nuclear translocation in HG-treated neuronal cultures, accompanied by partial loss of mitochondrial potential and gradual retraction of neuronal processes. Addition of TNFα to HG-treated neurons increased expression of the FOXO-dependent proapoptotic gene Bim, which resulted in extensive apoptotic death. Insulin-like growth factor-I (IGF-I) significantly lowered intracellular ROS, which was accompanied by IGF-I-mediated FOXO3a nuclear export and decrease in its transcriptional activity. The clinical relevance of these findings is supported by detection of nuclear FOXO3a in TUNEL-positive cortical neurons from HIVE, especially in brain areas characterized by elevated TNFα.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Factores de Transcripción Forkhead/metabolismo , Resistencia a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neuronas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Western Blotting , Técnica del Anticuerpo Fluorescente , Proteína Forkhead Box O3 , Glucosa/metabolismo , Etiquetado Corte-Fin in Situ , Potencial de la Membrana Mitocondrial/fisiología , Proteínas de la Membrana/metabolismo , Neuronas/patología , Células PC12 , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Cell Biochem ; 112(1): 225-32, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21117063

RESUMEN

HIV-1 gene transcription is controlled by the cooperation of viral and host factors which bind to specific DNA sequences within the viral promoter spanning the long terminal repeat (LTR). Previously we showed that the St. John's Wort DING phosphatase, p27SJ, suppresses HIV-1 gene transcription by binding to the viral protein Tat and preventing its nuclear import. Here, we describe the inhibitory effect of p27SJ on the phosphorylation of the C-terminal domain (CTD) of RNA polymerase II (RNAPII). This inhibition leads to the suppression of the association of RNAPII with the LTR. Inhibition of binding of RNAPII to LTR by p27SJ resulted in the suppression of LTR transcription elongation and a decrease in LTR transcriptional activity. Another form of the St. John's Wort DING phosphatase, p38SJ, also suppressed binding of RNAPII to the LTR, reduced transcription elongation and was even more powerful than p27SJ in inhibiting the transcriptional activity of the LTR. Our data suggest a possible mechanism by which the p27SJ/p38SJ DING phosphatase can regulate HIV-1 LTR expression by inhibiting phosphorylation of the CTD of RNAPII and suppressing LTR transcription elongation.


Asunto(s)
VIH-1/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas de Plantas/metabolismo , Transcripción Genética , Glioblastoma/metabolismo , Duplicado del Terminal Largo de VIH/genética , VIH-1/metabolismo , Humanos , Monoéster Fosfórico Hidrolasas/genética , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo
14.
Cell Cycle ; 9(20): 4164-73, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20948313

RESUMEN

Purα is a nucleic acid-binding protein with DNA-unwinding activity, which has recently been shown to have a role in the cellular response to DNA damage. We have investigated the function of Purα in Ultraviolet-C (UVC) radiation-induced DNA damage and nucleotide excision repair (NER). Mouse embryo fibroblasts from PURA(-/-) knockout mice, which lack Purα, showed enhanced sensitivity to UVC irradiation as assessed by assays for cell viability and clonogenicity compared to Purα positive control cultures. In reporter plasmid reactivation assays to measure the removal of DNA adducts induced in vitro by UVC, the Purα-negative cells were less efficient in DNA damage repair. Purα-negative cells were also more sensitive to UVC-induced DNA damage measured by Comet assay and showed a decreased ability to remove UVC-induced cyclobutane pyrimidine dimers. In wild-type mouse fibroblasts, expression of Purα is induced following S-phase checkpoint activation by UVC in a similar manner to the NER factor TFIIH. Moreover, co-immunoprecipitation experiments showed that Purα physically associates with TFIIH. Thus, Purα has a role in NER and the repair of UVC-induced DNA damage.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Fibroblastos/efectos de la radiación , Proteínas del Tejido Nervioso/metabolismo , Rayos Ultravioleta , Animales , Células Cultivadas , Ensayo Cometa , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Dímeros de Pirimidina/genética , Dímeros de Pirimidina/metabolismo , Dímeros de Pirimidina/efectos de la radiación
15.
Cell Cycle ; 9(18): 3715-22, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20890127

RESUMEN

Infection with HIV-1 induces a variety of biological alterations to the host that are beneficial to the life cycle of the virus but may have adverse effects on the host cell. Here we demonstrate that expression of Rad51, a major component of the homologous recombination-directed DNA repair (HRR) pathway, is induced upon HIV-1 infection of microglial cells. Activation of Rad51 expression positively impacts on HIV-1 LTR transcription through a region of the viral promoter known for binding the inducible transcription factor NFκB. Rad51 showed the ability to form a complex with the p65 subunit of NFκB and regulate the level of p65 interaction with LTR DNA encompassing the κB motif. This study provides evidence for reciprocal interaction of HIV-1 and a host DNA repair protein that impacts on expression of the viral genome. These results also point to the ability of HIV-1 to recruit proteins involved in DNA repair that are necessary for retroviral DNA integration, efficient replication and prevention of viral-induced cell death.


Asunto(s)
Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Microglía/virología , Recombinasa Rad51/metabolismo , Enzimas Reparadoras del ADN/metabolismo , VIH-1/metabolismo , Humanos , Microglía/citología , Microglía/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Recombinasa Rad51/análisis , Recombinasa Rad51/genética , Factor de Transcripción ReIA/análisis , Factor de Transcripción ReIA/metabolismo
16.
J Interferon Cytokine Res ; 29(6): 313-26, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19441883

RESUMEN

Chemokines constitute a family of chemoattractant cytokines and are subdivided into four families on the basis of the number and spacing of the conserved cysteine residues in the N-terminus of the protein. Chemokines play a major role in selectively recruiting monocytes, neutrophils, and lymphocytes, as well as in inducing chemotaxis through the activation of G-protein-coupled receptors. Monocyte chemoattractant protein-1 (MCP-1/CCL2) is one of the key chemokines that regulate migration and infiltration of monocytes/macrophages. Both CCL2 and its receptor CCR2 have been demonstrated to be induced and involved in various diseases. Migration of monocytes from the blood stream across the vascular endothelium is required for routine immunological surveillance of tissues, as well as in response to inflammation. This review will discuss these biological processes and the structure and function of CCL2.


Asunto(s)
Quimiocina CCL2/metabolismo , Infecciones por VIH/inmunología , VIH-1/inmunología , Inmunoterapia , Monocitos/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Quimiocina CCL2/química , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiotaxis/inmunología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Infecciones por VIH/metabolismo , VIH-1/patogenicidad , Humanos , Monocitos/inmunología , Monocitos/patología , Polimorfismo Genético , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores CCR2/metabolismo , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/metabolismo , Virulencia
17.
J Cell Biochem ; 107(3): 400-7, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19343785

RESUMEN

p27SJ, a novel protein isolated from St John's wort (Hypericum perforatum), belongs to an emerging family of DING proteins that are related to a prokaryotic phosphate-binding protein superfamily. Here we demonstrate that p27SJ exhibits phosphatase activity and that its expression in cells decreases the level of phosphorylated Erk1/2, a key protein of several signaling pathways. Treatment of p27SJ-expressing cells with phosphatase inhibitors including okadaic acid, maintained Erk1/2 in its phosphorylated form, suggesting that dephosphorylation of Erk1/2 is mediated by p27SJ. Further, expression of p27SJ affects Erk1/2 downstream regulatory targets such as STAT3 and CREB. Moreover, the level of expression of cyclin A that associates with active ERK1/2 and is regulated by CREB, was modestly reduced in p27SJ-expressing cells. Accordingly, results from in vitro kinase assays revealed a noticeable decrease in the activity of cyclin A in cells expressing p27SJ. Cell cycle analysis demonstrated dysregulation at S and G2/M phases in cells expressing p27SJ, supporting the notion that a decline in cyclin A activity by p27SJ has a biological impact on cell growth. These observations provide evidence that p27SJ alters the state of Erk1/2 phosphorylation, and impacts several biological events associated with cell growth and function.


Asunto(s)
Ciclo Celular , Hypericum/enzimología , Fosfoproteínas Fosfatasas/metabolismo , Proteínas de Plantas/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Humanos , Hypericum/química , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Modelos Biológicos , Fosforilación , Conformación Proteica
18.
Brain Res ; 1275: 81-6, 2009 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-19328187

RESUMEN

The lack of productive infection of neurons by HIV-1 suggests that the neuronal damage seen in AIDS patients with cognitive disorders is caused indirectly via viral and cellular proteins with neurotoxic activity. Among HIV-1 proteins, Vpr has been shown to deregulate expression of various important cytokines and inflammatory proteins in infected and uninfected cells. However, the mechanisms underlying these changes remain unclear. Here, we demonstrate that neurons can take up Vpr that is released into the supernatant of HIV-infected microglia. We also found that administration of recombinant Vpr (rVpr) to human neurons resulted in a slow but sustained elevation of intracellular calcium [Ca(2+)]i. Interestingly, our data also show that [Ca(2+)]i elevation by Vpr leads to ROS production and impairs glutamate signaling in neuronal cells. Vpr disturbs calcium homeostasis through downregulation of endogenous PMCA. Finally, we found that the permeability of the plasma membrane increases in neurons treated with Vpr. Therefore, we conclude that soluble Vpr is a major viral factor that causes a disturbance in neuronal communication leading to neuronal dysfunction. The outcome of these studies will advance the understanding of HIV-1 pathogenesis and will help in the development of new therapeutic approaches.


Asunto(s)
Calcio/fisiología , VIH-1 , Neuronas/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/fisiología , Calcio/antagonistas & inhibidores , Comunicación Celular/fisiología , Línea Celular Tumoral , Células Cultivadas , Regulación hacia Abajo/fisiología , Humanos , Microglía/metabolismo , Microglía/virología , Neuronas/virología , Células U937 , Regulación hacia Arriba/fisiología
19.
Front Biosci (Landmark Ed) ; 14(1): 352-75, 2009 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-19273072

RESUMEN

The IGF-IR is a multifunctional tyrosine kinase receptor involved in several biological processes including cell proliferation, differentiation, DNA repair, and cell survival. In the brain IGF-I plays a critical role during embryonic and early postnatal development. In the mature brain, IGF-I binding sites have been found in different regions of the brain, and multiple reports confirmed a strong neuroprotective action of the IGF-IR against different pro-apoptotic insults. When the IGF-IR signaling system is insufficiently deployed, either by low level of expression in elderly individuals, or by the inhibition associated with inflammatory cytokines, neuronal function and survival could be compromised. The examples of such CNS pathologies include HIV associated dementia, diabetic neuropathies, and Alzheimer's disease. On the other hand, elevated expression activity of the IGF-IR may support uncontrolled cell proliferation and protection from apoptosis. Probably the best example of the IGF-IR involvement in brain tumors is medulloblastomas in which functional cooperation between viral oncoprotein, JC virus large T-antigen, and IGF-IR has been recently established. Therefore, better understanding of the beneficial and potentially harmful aspects of the IGF-IR can be critical for the development of new clinical regimens against neurodegenerative disorders and brain tumors.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Receptor IGF Tipo 1/fisiología , Neoplasias Encefálicas/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Receptor IGF Tipo 1/metabolismo
20.
J Interferon Cytokine Res ; 29(1): 1-7, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19014341

RESUMEN

The transforming growth factor-beta1 (TGF-beta1) is a cytokine involved in many biological events inlcuding immunosuppression, angiogenesis, cell growth, and apoptosis. Expression of TGF-beta1 at the transcriptional level is controlled by a series of ubiquitous and specialized factors whose activities can be modulated by a variety of signaling events. Here we demonstrate that activity of the TGF-beta1 promoter is increased by C/EBPbeta, a DNA-binding transcription factor whose activity can be influenced by several immunomodulators, in astrocytes and microglial cells. Interestingly, expression of Smad3 and Smad4, the downstream regulators of the TGF-beta1-signaling pathway, impairs the activity of C/EBPbeta on the TGF-beta1 promoter. Further, we demonstrate that MH2, a common domain among Smads that has protein-binding activities, interacts with C/EBPbeta and decreases its association with a region of the TGF-beta1 promoter that is responsive to C/EBPbeta activation. Interestingly, the p65 subunit of nuclear factor-kappaB (NF-kappaB), which also interacts with C/EBPbeta, cooperates with MH2 and decreased DNA-binding and transcriptional activities of C/EBPbeta on the TGF-beta1 promoter. These observations indicate that an autoregulatory mechanism, involving the MH2 domain of Smads, modulates activation of the TGF-beta1 promoter by C/EBPbeta. Further, our results show that the interplay between NF-kappaB and C/EBPbeta has an impact on the ability of C/EBPbeta to stimulate TGF-beta1 transcription, hence, suggesting that the cross-communication of signaling pathways that modulate NF-kappaB and C/EBPbeta may dictate the level of TGF-beta1 promoter activity.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , FN-kappa B/metabolismo , Proteína smad3/metabolismo , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta1/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular Tumoral , Humanos , FN-kappa B/genética , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína/genética , Transducción de Señal/genética , Proteína smad3/genética , Proteína Smad4/genética , Transcripción Genética , Transfección , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA