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.
Mucosal Immunol ; 11(6): 1694-1704, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30076393

RESUMEN

Intestinal macrophages in healthy human mucosa are profoundly down-regulated for inflammatory responses (inflammation anergy) due to stromal TGF-ß inactivation of NF-κB. Paradoxically, in cytomegalovirus (CMV) intestinal inflammatory disease, one of the most common manifestations of opportunistic CMV infection, intestinal macrophages mediate severe mucosal inflammation. Here we investigated the mechanism whereby CMV infection promotes macrophage-mediated mucosal inflammation. CMV infected primary intestinal macrophages but did not replicate in the cells or reverse established inflammation anergy. However, CMV infection of precursor blood monocytes, the source of human intestinal macrophages in adults, prevented stromal TGF-ß-induced differentiation of monocytes into inflammation anergic macrophages. Mechanistically, CMV up-regulated monocyte expression of the TGF-ß antagonist Smad7, blocking the ability of stromal TGF-ß to inactivate NF-κB, thereby enabling MyD88 and NF-κB-dependent cytokine production. Smad7 expression also was markedly elevated in mucosal tissue from subjects with CMV colitis and declined after antiviral ganciclovir therapy. Confirming these findings, transfection of Smad7 antisense oligonucleotide into CMV-infected monocytes restored monocyte susceptibility to stromal TGF-ß-induced inflammation anergy. Thus, CMV-infected monocytes that recruit to the mucosa, not resident macrophages, are the source of inflammatory macrophages in CMV mucosal disease and implicate Smad7 as a key regulator of, and potential therapeutic target for, CMV mucosal disease.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/fisiología , Inflamación/inmunología , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Proteína smad7/metabolismo , Células Cultivadas , Anergia Clonal , Humanos , Macrófagos/virología , Monocitos/virología , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , Proteína smad7/genética , Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven
2.
Eur J Neurosci ; 46(7): 2241-2252, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28612962

RESUMEN

A greater understanding of neural mechanisms contributing to anxiety is needed in order to develop better therapeutic interventions. This study interrogates a novel molecular mechanism that shapes anxiety-like behaviour, demonstrating that the microRNA miR-101a-3p and its target, enhancer of zeste homolog 2 (Ezh2) in the amygdala, contribute to rodent anxiety-like behaviour. We utilized rats that were selectively bred for differences in emotionality and stress reactivity, showing that high-novelty-responding (HR) rats, which display low trait anxiety, have lower miR-101a-3p levels in the amygdala compared to low-novelty-responding (LR) rats that characteristically display high trait anxiety. To determine whether there is a causal relationship between amygdalar miR-101a-3p and anxiety behaviour, we used a viral approach to overexpress miR-101a-3p in the amygdala of HR rats and test whether it would increase their typically low levels of anxiety-like behaviour. We found that increasing miR-101a-3p in the amygdala increased HRs' anxiety-like behaviour in the open-field test and elevated plus maze. Viral-mediated miR-101a-3p overexpression also reduced expression of the histone methyltransferase Ezh2, which mediates gene silencing via trimethylation of histone 3 at lysine 27 (H3K27me3). Knockdown of Ezh2 with short-interfering RNA (siRNA) also increased HRs' anxiety-like behaviour, but to a lesser degree than miR-101a-3p overexpression. Overall, our data demonstrate that increasing miR-101a-3p expression in the amygdala increases anxiety-like behaviour and that this effect is at least partially mediated via repression of Ezh2. This work adds to the growing body of evidence implicating miRNAs and epigenetic regulation as molecular mediators of anxiety behaviour.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , MicroARNs/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Masculino , Aprendizaje por Laberinto , MicroARNs/genética , Ratas
3.
J Clin Oncol ; 34(27): 3276-83, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27507879

RESUMEN

PURPOSE: The biology of HIV-associated cancers may differ depending on immunologic and virologic context during development. Therefore, an understanding of the burden of Kaposi's sarcoma (KS) and non-Hodgkin lymphoma (NHL) relative to antiretroviral therapy (ART), virologic suppression, and CD4 count is important. PATIENTS AND METHODS: KS and NHL diagnoses during 1996 to 2011 were identified among patients with HIV infection in eight clinical cohorts in the United States. Among patients in routine HIV clinical care, the proportion of cases in categories of ART use, HIV RNA, and CD4 count at diagnosis were described across calendar time. Person-time and incidence rates were calculated for each category. RESULTS: We identified 466 patients with KS and 258 with NHL. In recent years, KS was more frequently diagnosed after ART initiation (55% in 1996 to 2001 v 76% in 2007 to 2011; P-trend = .02). The proportion of patients with NHL who received ART was higher but stable over time (83% overall; P-trend = .81). An increasing proportion of KS and NHL occurred at higher CD4 counts (P < .05 for KS and NHL) and with undetectable HIV RNA (P < .05 for KS and NHL). In recent years, more person-time was contributed by patients who received ART, had high CD4 counts and had undetectable HIV RNA, whereas incidence rates in these same categories remained stable or declined. CONCLUSION: Over time, KS and NHL occurred at higher CD4 counts and lower HIV RNA values, and KS occurred more frequently after ART initiation. These changes were driven by an increasing proportion of patients with HIV who received effective ART, had higher CD4 counts, and had suppressed HIV RNA and not by increases in cancer risk within these subgroups. An improved understanding of HIV-associated cancer pathogenesis and outcomes in the context of successful ART is therefore important.


Asunto(s)
Infecciones por VIH/epidemiología , Linfoma no Hodgkin/epidemiología , Sarcoma de Kaposi/epidemiología , Adulto , Estudios de Cohortes , Femenino , Infecciones por VIH/patología , Humanos , Incidencia , Linfoma de Células B Grandes Difuso/epidemiología , Linfoma de Células B Grandes Difuso/virología , Linfoma no Hodgkin/patología , Linfoma no Hodgkin/virología , Masculino , Persona de Mediana Edad , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/virología , Estados Unidos/epidemiología
4.
Oncotarget ; 6(20): 17895-910, 2015 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-26255626

RESUMEN

Aberrations in the mTOR (mechanistic target of rapamycin) axis are frequently reported in cancer. Using publicly available tumor genome sequencing data, we identified several point mutations in MTOR and its upstream regulator RHEB (Ras homolog enriched in brain) in patients with clear cell renal cell carcinoma (ccRCC), the most common histology of kidney cancer. Interestingly, we found a prominent cluster of hyperactivating mutations in the FAT (FRAP-ATM-TTRAP) domain of mTOR in renal cell carcinoma that led to an increase in both mTORC1 and mTORC2 activities and led to an increased proliferation of cells. Several of the FAT domain mutants demonstrated a decreased binding of DEPTOR (DEP domain containing mTOR-interacting protein), while a subset of these mutations showed altered binding of the negative regulator PRAS40 (proline rich AKT substrate 40). We also identified a recurrent mutation in RHEB in ccRCC patients that leads to an increase in mTORC1 activity. In vitro characterization of this RHEB mutation revealed that this mutant showed considerable resistance to TSC2 (Tuberous Sclerosis 2) GAP (GTPase activating protein) activity, though its interaction with TSC2 remained unaltered. Mutations in the FAT domain of MTOR and in RHEB remained sensitive to rapamycin, though several of these mutations demonstrated residual mTOR kinase activity after treatment with rapamycin at clinically relevant doses. Overall, our data suggests that point mutations in the mTOR pathway may lead to downstream mTOR hyperactivation through multiple different mechanisms to confer a proliferative advantage to a tumor cell.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Proteínas de Unión al GTP Monoméricas/genética , Neuropéptidos/genética , Mutación Puntual , Serina-Treonina Quinasas TOR/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Proliferación Celular/efectos de los fármacos , Análisis Mutacional de ADN , Bases de Datos Genéticas , Resistencia a Antineoplásicos/genética , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Proteína Homóloga de Ras Enriquecida en el Cerebro , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Transfección , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
5.
Monoclon Antib Immunodiagn Immunother ; 33(6): 420-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25513981

RESUMEN

Although antibodies are commercially available to allow investigation into the biology of the age-regulating protein Klotho, problems with antibody specificity and application functionality are significant barriers to progress. Chief among these limitations is the inability of current tools to allow in vivo validation of binding partners originally identified through transfection of tagged proteins. To overcome this barrier, we generated a series of hybridoma cell lines by immunizing rats with a GST-KL1 fusion protein. Purified antibodies generated from these cell lines differentially detect human or mouse Klotho protein via Western blot, immunocyto/histochemistry, and immunoprecipitation. Specificity of antibody binding to Klotho was confirmed by mass spectrometry following immunoprecipitation. With this confidence in antibody specificity, co-immunoprecipitation was utilized to validate the interaction of Klotho/FGFR and Klotho/wnt7a in mouse kidney lysates.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Glucuronidasa/inmunología , Hibridomas/inmunología , Animales , Anticuerpos Monoclonales/genética , Western Blotting , Cartilla de ADN/genética , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Proteínas Klotho , Espectrometría de Masas , Ratones , Plásmidos/genética , Ratas
6.
PLoS One ; 9(1): e86338, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24466036

RESUMEN

Malignant melanoma is responsible for approximately 75% of skin cancer-related deaths. BRAF plays an important role in regulating the mitogen-activated protein kinase (MAPK) signaling cascade in melanoma with activating mutations in the serine/threonine kinase BRAF occurring in 60-70% of malignant melanomas. The BRAF-MEK-ERK (MAPK) pathway is a key regulator of melanoma cell invasion. In addition, activation of NFκB via the MAPK pathway is regulated through MEK-induced activation of IKK. These pathways are potential targets for prevention and treatment of melanoma. In this study, we investigated the effect of fisetin, a phytochemical present in fruits and vegetables, on melanoma cell invasion and epithelial-mesenchymal transition, and delineated the underlying molecular mechanism. Treatment of multiple human malignant melanoma cell lines with fisetin (5-20 µM) resulted in inhibition of cell invasion. BRAF mutated melanoma cells were more sensitive to fisetin treatment, and this was associated with a decrease in the phosphorylation of MEK1/2 and ERK1/2. In addition, fisetin inhibited the activation of IKK leading to a reduction in the activation of the NFκB signaling pathway. Treatment of cells with an inhibitor of MEK1/2 (PD98059) or of NFκB (caffeic acid phenethyl ester) also reduced melanoma cell invasion. Furthermore, treatment of fisetin promoted mesenchymal to epithelial transition in melanoma cells, which was associated with a decrease in mesenchymal markers (N-cadherin, vimentin, snail and fibronectin) and an increase in epithelial markers (E-cadherin and desmoglein). Employing three dimensional skin equivalents consisting of A375 cells admixed with normal human keratinocytes embedded onto a collagen-constricted fibroblast matrix, we found that treatment of fisetin reduced the invasive potential of melanoma cells into the dermis and increased the expression of E-cadherin with a concomitant decrease in vimentin. These results indicate that fisetin inhibits melanoma cell invasion through promotion of mesenchymal to epithelial transition and by targeting MAPK and NFκB signaling pathways.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Transdiferenciación Celular/efectos de los fármacos , Flavonoides/farmacología , Sistema de Señalización de MAP Quinasas , Antígenos CD , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Núcleo Celular/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Flavonoles , Humanos , Melanoma , FN-kappa B/metabolismo , Invasividad Neoplásica , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Vimentina/metabolismo
7.
J Neurochem ; 128(6): 950-61, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24138030

RESUMEN

Parkinson's disease is a neurodegenerative movement disorder. The histopathology of Parkinson's disease comprises proteinaceous inclusions known as Lewy bodies, which contains aggregated α-synuclein. Cathepsin D (CD) is a lysosomal protease previously demonstrated to cleave α-synuclein and decrease its toxicity in both cell lines and mouse brains in vivo. Here, we show that pharmacological inhibition of CD, or introduction of catalytically inactive mutant CD, resulted in decreased CD activity and increased cathepsin B activity, suggesting a possible compensatory response to inhibition of CD activity. However, this increased cathepsin B activity was not sufficient to maintain α-synuclein degradation, as evidenced by the accumulation of endogenous α-synuclein. Interestingly, the levels of LC3, LAMP1, and LAMP2, proteins involved in autophagy-lysosomal activities, as well as total lysosomal mass as assessed by LysoTracker flow cytometry, were unchanged. Neither autophagic flux nor proteasomal activities differs between cells over-expressing wild-type versus mutant CD. These observations point to a critical regulatory role for that endogenous CD activity in dopaminergic cells in α-synuclein homeostasis which cannot be compensated for by increased Cathepsin B. These data support the potential need to enhance CD function in order to attenuate α-synuclein accumulation as a therapeutic strategy against development of synucleinopathy.


Asunto(s)
Catepsina B/metabolismo , Catepsina D/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , alfa-Sinucleína/metabolismo , Autofagia/efectos de los fármacos , Autofagia/fisiología , Caspasas/metabolismo , Catepsina D/metabolismo , Línea Celular Tumoral , Expresión Génica/fisiología , Humanos , Lentivirus/genética , Lisosomas/metabolismo , Neuroblastoma , Neuronas/citología , Neuronas/efectos de los fármacos , Pepstatinas/farmacología , Inhibidores de Proteasas/farmacología
8.
Toxicol Appl Pharmacol ; 272(3): 879-87, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23954561

RESUMEN

Arsenic exposure is known to disrupt innate immune functions in humans and in experimental animals. In this study, we provide a mechanism by which arsenic trioxide (ATO) disrupts macrophage functions. ATO treatment of murine macrophage cells diminished internalization of FITC-labeled latex beads, impaired clearance of phagocytosed fluorescent bacteria and reduced secretion of pro-inflammatory cytokines. These impairments in macrophage functions are associated with ATO-induced unfolded protein response (UPR) signaling pathway characterized by the enhancement in proteins such as GRP78, p-PERK, p-eIF2α, ATF4 and CHOP. The expression of these proteins is altered both at transcriptional and translational levels. Pretreatment with chemical chaperon, 4-phenylbutyric acid (PBA) attenuated the ATO-induced activation in UPR signaling and afforded protection against ATO-induced disruption of macrophage functions. This treatment also reduced ATO-mediated reactive oxygen species (ROS) generation. Interestingly, treatment with antioxidant N-acetylcysteine (NAC) prior to ATO exposure, not only reduced ROS production and UPR signaling but also improved macrophage functions. These data demonstrate that UPR signaling and ROS generation are interdependent and are involved in the arsenic-induced pathobiology of macrophage. These data also provide a novel strategy to block the ATO-dependent impairment in innate immune responses.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Macrófagos/inmunología , Óxidos/toxicidad , Transducción de Señal/inmunología , Respuesta de Proteína Desplegada/inmunología , Animales , Trióxido de Arsénico , Arsenicales , Línea Celular , Chaperón BiP del Retículo Endoplásmico , Inmunidad Innata/inmunología , Macrófagos/efectos de los fármacos , Ratones , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos
9.
Am J Physiol Cell Physiol ; 305(2): C228-37, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23703526

RESUMEN

Endothelial adhesion molecules are critical effectors of inflammation ensuring coordinated interactions that allow leukocytes to home to sites of injury. These adhesion molecules are often extensively modified posttranslationaly by the addition of N-glycans, but if, or how, these modifications contribute to the protein function remains poorly understood. Herein we show that activated endothelial cells express two distinct N-glycoforms of intercellular adhesion molecule 1 (ICAM-1) that comprise a complex N-glycoform with α-2,6 sialic acid present at relatively high levels and a second, less abundant and previously undescribed high-mannose glycoform (HM-ICAM-1). This novel HM-ICAM-1 glycoform was also detected in human coronary artery specimens and moreover appeared to be the dominant glycoform in vivo. Production of exclusively HM-ICAM-1 in cells by α-mannosidase inhibition increased monocyte rolling and adhesion compared with mature ICAM-1 consistent with high-mannose epitopes providing leukocyte ligands. Cross-linking of ICAM-1 transmits outside-in signals that affect endothelial permeability and survival. Interestingly, cell signaling (assessed using ERK, VE-cadherin, and Akt phosphorylation) was maintained after cross-linking of HM-ICAM-1 compared with mature ICAM-1; however, interactions with the actin cytoskeleton were lost with HM-ICAM-1. These findings suggest that specific ICAM-1 N-glycoforms modulate distinct aspects of the inflammatory response and identify HM-ICAM-1 as a new therapeutic target for controlling leukocyte trafficking and endothelial inflammation.


Asunto(s)
Células Endoteliales/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Monocitos/fisiología , Transducción de Señal/fisiología , Animales , Células COS , Adhesión Celular/fisiología , Línea Celular , Chlorocebus aethiops , Citoesqueleto/fisiología , Células Endoteliales/citología , Glicosilación , Células Endoteliales de la Vena Umbilical Humana , Humanos , Molécula 1 de Adhesión Intercelular/química , Molécula 1 de Adhesión Intercelular/genética , Isoformas de Proteínas
10.
Toxicol Appl Pharmacol ; 266(2): 233-44, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23147569

RESUMEN

Histone deacetylase (HDAC) inhibitors are potent anticancer agents and show efficacy against various human neoplasms. Vorinostat is a potent HDAC inhibitor and has shown potential to inhibit growth of human xenograft tumors. However, its effect on the growth of skin neoplasm remains undefined. In this study, we show that vorinostat (2 µM) reduced expression of HDAC1, 2, 3, and 7 in epidermoid carcinoma A431 cells. Consistently, it increased acetylation of histone H3 and p53. Vorinostat (100mg/kg body weight, IP) treatment reduced human xenograft tumor growth in highly immunosuppressed nu/nu mice. Histologically, the vorinostat-treated tumor showed features of well-differentiation with large necrotic areas. Based on proliferating cell nuclear antigen (PCNA) staining and expression of cyclins D1, D2, E, and A, vorinostat seems to impair proliferation by down-regulating the expression of these proteins. However, it also induced apoptosis. The mechanism by which vorinostat blocks proliferation and makes tumor cells prone to apoptosis, involved inhibition of mTOR signaling which was accompanied by reduction in cell survival AKT and extracellular-signal regulated kinase (ERK) signaling pathways. Our data provide a novel mechanism-based therapeutic intervention for cutaneous squamous cell carcinoma (SCC). Vorinostat may be utilized to cure skin neoplasms in organ transplant recipient (OTR). These patients have high morbidity and surgical removal of these lesions which frequently develop in these patients, is difficult.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclinas/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Femenino , Inhibidores de Histona Desacetilasas/administración & dosificación , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/administración & dosificación , Huésped Inmunocomprometido , Ratones , Ratones Desnudos , Antígeno Nuclear de Célula en Proliferación/análisis , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/patología , Serina-Treonina Quinasas TOR/metabolismo , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
11.
PLoS One ; 7(6): e39586, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761832

RESUMEN

Endoplasmic reticulum (ER) stress-induced apoptosis has been implicated in various neurodegenerative diseases including Parkinson Disease, Alzheimer Disease and Huntington Disease. PUMA (p53 upregulated modulator of apoptosis) and BIM (BCL2 interacting mediator of cell death), pro-apoptotic BH3 domain-only, BCL2 family members, have previously been shown to regulate ER stress-induced cell death, but the upstream signaling pathways that regulate this response in neuronal cells are incompletely defined. Consistent with previous studies, we show that both PUMA and BIM are induced in response to ER stress in neuronal cells and that transcriptional induction of PUMA regulates ER stress-induced cell death, independent of p53. CHOP (C/EBP homologous protein also known as GADD153; gene name Ddit3), a critical initiator of ER stress-induced apoptosis, was found to regulate both PUMA and BIM expression in response to ER stress. We further show that CHOP knockdown prevents perturbations in the AKT (protein kinase B)/FOXO3a (forkhead box, class O, 3a) pathway in response to ER stress. CHOP co-immunoprecipitated with FOXO3a in tunicamycin treated cells, suggesting that CHOP may also regulate other pro-apoptotic signaling cascades culminating in PUMA and BIM activation and cell death. In summary, CHOP regulates the expression of multiple pro-apoptotic BH3-only molecules through multiple mechanisms, making CHOP an important therapeutic target relevant to a number of neurodegenerative conditions.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Factores de Transcripción Forkhead/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción CHOP/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Proteínas de la Membrana/genética , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Telencéfalo/citología , Telencéfalo/efectos de los fármacos , Telencéfalo/metabolismo , Factor de Transcripción CHOP/genética , Proteínas Supresoras de Tumor/genética , Tunicamicina/farmacología
12.
Future Microbiol ; 6(12): 1399-413, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22122438

RESUMEN

Latency-associated nuclear antigen (LANA) is encoded by the Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) open reading frame 73. LANA is expressed during latent KSHV infection of cells, including tumor cells, such as primary effusion lymphoma, KS and multicentric Castleman's disease. Latently infected cells have multiple extrachromosomal copies of covalently closed circular KSHV genomes (episomes) that are stably maintained in proliferating cells. LANA's best characterized function is that of mediating episome persistence. It does so by binding terminal repeat sequences to the chromosomal matrix, thus ensuring episome replication with each cell division and efficient DNA segregation to daughter nuclei after mitosis. To achieve these functions, LANA associates with different host cell proteins, including chromatin-associated proteins and proteins involved in DNA replication. In addition to episome maintenance, LANA has transcriptional regulatory effects and affects cell growth. LANA exerts these functions through interactions with different cell proteins.


Asunto(s)
Antígenos Virales/metabolismo , Herpesvirus Humano 8/patogenicidad , Proteínas Nucleares/metabolismo , Factores de Virulencia/metabolismo , Latencia del Virus , División Celular , Segregación Cromosómica , Replicación del ADN , ADN Viral/genética , Herpesvirus Humano 8/genética , Interacciones Huésped-Patógeno , Humanos , Plásmidos/genética , Replicación Viral
13.
J Virol ; 84(7): 3162-77, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20089649

RESUMEN

Human cytomegalovirus (HCMV) virion assembly takes place in the nucleus and cytoplasm of infected cells. The HCMV virion tegument protein pp150 (ppUL32) is an essential protein of HCMV and has been suggested to play a role in the cytoplasmic phase of HCMV assembly. To further define its role in viral assembly and to identify host cell proteins that interact with pp150 during viral assembly, we utilized yeast two-hybrid analyses to detect an interaction between pp150 and Bicaudal D1 (BicD1), a protein thought to play a role in trafficking within the secretory pathway. BicD1 is known to interact with the dynein motor complex and the Rab6 GTPase. The interaction between pp150 and BicD1 was confirmed by coimmunoprecipitation and fluorescence resonance energy transfer. Depletion of BicD1 with short hairpin RNA (shRNA) caused decreased virus yield and a defect in trafficking of pp150 to the cytoplasmic viral assembly compartment (AC), without altering trafficking to the AC of another essential tegument protein, pp28, or the viral glycoprotein complex gM/gN. The C terminus of BicD1 has been previously shown to interact with the GTPase Rab6, suggesting a potential role for Rab6-mediated vesicular trafficking in HCMV assembly. Finally, overexpression of the N terminus of truncated BicD1 acts in a dominant-negative manner and leads to disruption of the AC and a decrease in the assembly of infectious virus. This phenotype was similar to that observed following overexpression of dynamitin (p50) and provided additional evidence that morphogenesis of the AC and virus assembly were dynein dependent.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas del Citoesqueleto/fisiología , Fosfoproteínas/metabolismo , Proteínas de la Matriz Viral/metabolismo , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Proteínas del Citoesqueleto/análisis , Proteínas del Citoesqueleto/química , Dineínas/fisiología , Transferencia Resonante de Energía de Fluorescencia , Humanos , Microtúbulos/fisiología , Fosfoproteínas/análisis , Estructura Terciaria de Proteína , Transporte de Proteínas , Técnicas del Sistema de Dos Híbridos , Proteínas de la Matriz Viral/análisis , Ensamble de Virus
14.
J Neuropathol Exp Neurol ; 68(12): 1326-38, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19915483

RESUMEN

Perinatal hypoxia-ischemia may result in long-term neurological deficits. In addition to producing neuron death, HI causes death of neural precursor cells (NPCs) in the developing brain. To characterize the molecular pathways that regulate hypoxia-induced death of NPCs, we treated a mouse neural stem cell line (C17.2 cells) and fibroblastic growth factor II-expanded primary NPCs derived from wild-type or gene-disrupted mice, with oxygen glucose deprivation or the hypoxia mimetics desferrioxamine or cobalt chloride. Neural precursor cells undergoing hypoxia exhibited time- and concentration-dependent caspase-3 activation and cell death, which was significantly reduced by treatment with a broad caspase inhibitor or protein synthesis inhibition. Bax/Bak-deficient NPCs were protected from desferrioxamine-induced death and exhibited minimal caspase-3 activation. Oxygen glucose deprivation or hypoxia-mimetic exposure also resulted in increased hypoxia-inducible factor alpha and bcl-2/adenovirus E1B 19-kd interacting protein 3 (BNIP3) expression. BNIP3 shRNA treatment failed to affect hypoxia-induced caspase-3 activation but inhibited cell death and nuclear translocation of apoptosis-inducing factor, indicating that BNIP3 is an important regulator of caspase-independent NPC death after hypoxia. These studies demonstrate that hypoxia activates both caspase-dependent and -independent NPC death pathways that are critically regulated by multiple Bcl-2 family members.


Asunto(s)
Apoptosis/fisiología , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Neuronas/efectos de los fármacos , Neuronas/patología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos , Células Madre/patología
15.
J Virol ; 81(8): 4348-56, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17287261

RESUMEN

Kaposi's sarcoma-associated herpesvirus latency-associated nuclear antigen (LANA) tethers viral terminal repeat (TR) DNA to mitotic chromosomes to mediate episome persistence. The 1,162-amino-acid LANA protein contains both N- and C-terminal chromosome attachment regions. The LANA C-terminal domain self-associates to specifically bind TR DNA and mitotic chromosomes. Here, we used alanine scanning substitutions spanning residues 1023 to 1145 to investigate LANA self-association, DNA binding, and C-terminal chromosome association. No residues were essential for LANA oligomerization, as assayed by coimmunoprecipitation experiments, consistent with redundant roles for amino acids in self-association. Different subsets of amino acids were important for DNA binding, as assayed by electrophoretic mobility shift assay, and mitotic chromosome association, indicating that distinct C-terminal LANA subdomains effect DNA and chromosome binding. The DNA binding domains of LANA and EBNA1 are predicted to be structurally homologous; certain LANA residues important for DNA binding correspond to those with roles in EBNA1 DNA binding, providing genetic support for at least partial structural homology. In contrast to the essential role of N-terminal LANA chromosome targeting residues in DNA replication, deficient C-terminal chromosome association did not reduce LANA-mediated DNA replication.


Asunto(s)
Antígenos Virales/metabolismo , Cromosomas Humanos/metabolismo , ADN Viral/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Antígenos Virales/química , Antígenos Virales/genética , Células COS , Chlorocebus aethiops , Replicación del ADN/fisiología , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inmunoprecipitación , Mitosis , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/química , Proteínas Nucleares/genética , Unión Proteica , Estructura Terciaria de Proteína , Replicación Viral/fisiología
16.
Biochim Biophys Acta ; 1769(1): 29-40, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17239456

RESUMEN

RNA interference (RNAi) is implicated in maintaining tandem DNA arrays as constitutive heterochromatin. We used chromatin immunoprecipitation with antibodies to RNA polymerase II (RNAPol-ChIP) to test for transcription of the following repeat arrays in human cells: subtelomeric D4Z4, pericentromeric satellite 2, and centromeric satellite alpha. D4Z4 has a promoter-like sequence upstream of an ORF in its 3.3-kb repeat unit. A short D4Z4 array at 4q35 is linked to facioscapulohumeral muscular dystrophy (FSHD). By RNAPol-ChIP and RT-PCR, little or no transcription of D4Z4 was detected in FSHD and normal myoblasts; lymphoblasts from an FSHD patient, a control, and a patient with D4Z4 hypomethylation due to mutation of DNMT3B (ICF syndrome); and normal or cancer tissues. However, RNAPol-ChIP assays indicated transcription of D4Z4 in a chromosome 4-containing human-mouse somatic cell hybrid. ChIP and RT-PCR showed satellite DNA transcription in some cancers and lymphoblastoid cell lines, although only at a low level. Given the evidence for the involvement of RNAi in satellite DNA heterochromatinization, it is surprising that, at most, a very small fraction of satellite DNA was associated with RNA Pol II. In addition, our results do not support the previously hypothesized disease-linked differential transcription of D4Z4 sequences in short, FSHD-linked arrays.


Asunto(s)
ADN Satélite , Distrofia Muscular Facioescapulohumeral/genética , Secuencias Repetidas en Tándem , Transcripción Genética , Animales , Células Cultivadas , Humanos , Células Híbridas , Ratones , Mioblastos , ARN Polimerasa II
17.
Virology ; 357(2): 149-57, 2007 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16979209

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA) tethers KSHV terminal repeat (TR) DNA to mitotic chromosomes to efficiently segregate episomes to progeny nuclei. LANA contains N- and C-terminal chromosome binding regions. We now show that C-terminal LANA preferentially concentrates to paired dots at pericentromeric and peri-telomeric regions of a subset of mitotic chromosomes through residues 996-1139. Deletions within C-terminal LANA abolished both self-association and chromosome binding, consistent with a requirement for self-association to bind chromosomes. A deletion abolishing TR DNA binding did not affect chromosome targeting, indicating LANA's localization is not due to binding its recognition sequence in chromosomal DNA. LANA distributed similarly on human and non-human mitotic chromosomes. These results are consistent with C-terminal LANA interacting with a cell factor that concentrates at pericentromeric and peri-telomeric regions of mitotic chromosomes.


Asunto(s)
Antígenos Virales/metabolismo , Centrómero/metabolismo , Cromosomas/virología , Herpesvirus Humano 8/genética , Mitosis/genética , Proteínas Nucleares/metabolismo , Antígenos Virales/genética , Línea Celular , Cromosomas/metabolismo , ADN Viral/química , ADN Viral/metabolismo , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 8/fisiología , Humanos , Proteínas Nucleares/genética
18.
J Virol ; 80(18): 8909-19, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16940503

RESUMEN

The latency-associated nuclear antigen (LANA) of Kaposi's sarcoma-associated herpesvirus (KSHV) is required for viral episome maintenance in host cells during latent infection. Two regions of the protein have been implicated in tethering LANA/viral episomes to the host mitotic chromosomes, and LANA chromosome-binding sites are subjects of high interest. Because previous studies had identified bromodomain protein Brd4 as the mitotic chromosome anchor for the bovine papillomavirus E2 protein, which tethers the viral episomes to host mitotic chromosomes (J. You, J. L. Croyle, A. Nishimura, K. Ozato, and P. M. Howley, Cell 117:349-360, 2004, and J. You, M. R. Schweiger, and P. M. Howley, J. Virol. 79:14956-14961, 2005), we examined whether KSHV LANA interacts with Brd4. We found that LANA binds Brd4 in vivo and in vitro and that the binding is mediated by a direct protein-protein interaction between the ET (extraterminal) domain of Brd4 and a carboxyl-terminal region of LANA previously implicated in chromosome binding. Brd4 associates with mitotic chromosomes throughout mitosis and demonstrates a strong colocalization with LANA and the KSHV episomes on host mitotic chromosomes. Although another bromodomain protein, RING3/Brd2, binds to LANA in a similar fashion in vitro, it is largely excluded from the mitotic chromosomes in KSHV-uninfected cells and is partially recruited to the chromosomes in KSHV-infected cells. These data identify Brd4 as an interacting protein for the carboxyl terminus of LANA on mitotic chromosomes and suggest distinct functional roles for the two bromodomain proteins RING3/Brd2 and Brd4 in LANA binding. Additionally, because Brd4 has recently been shown to have a role in transcription, we examined whether Brd4 can regulate the CDK2 promoter, which can be transactivated by LANA.


Asunto(s)
Antígenos Virales/fisiología , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/fisiología , Proteínas de Fusión Oncogénica/química , Sitios de Unión , Proteínas de Ciclo Celular , Línea Celular , Cromosomas/metabolismo , Genoma , Genoma Viral , Infecciones por VIH/complicaciones , Humanos , Mitosis , Unión Proteica , Estructura Terciaria de Proteína , Factores de Transcripción
19.
J Virol ; 78(24): 14033-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15564510

RESUMEN

In latent infection, Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen 1 (LANA1)-specific binding to KSHV terminal repeat DNA mediates multicopy episome persistence. We now use electrophoretic mobility shift assays to investigate LANA1 binding to its 20-bp cognate sequence. Mutations at positions 6, 7, and 8 ((6)CCC(8)) severely reduced LANA1 binding, whereas mutations at other positions only modestly reduced binding. Since (6)CCC(8) is in the 5' half of an inverted repeat sequence, these results are consistent with an asymmetric role for the inverted repeat in LANA1 binding.


Asunto(s)
ADN Viral/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/química , Secuencias Repetidas Terminales/genética , Antígenos Virales , Sitios de Unión , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiología , Humanos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
20.
Virology ; 319(2): 225-36, 2004 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-14980483

RESUMEN

Latency-associated nuclear antigen 1 (LANA1) binds to Kaposi's sarcoma-associated herpesvirus (KSHV) terminal repeat (TR) DNA to mediate episome replication and persistence. LANA1 concentrates at sites of TR DNA along mitotic chromosomes, consistent with tethering KSHV DNA to chromosomes for efficient segregation of episomes to progeny nuclei. We now investigate LANA1 C-terminus self-association and DNA binding. The TR DNA binding domain was localized to LANA1 residues 996-1139. Scanning deletions within this region ablated both LANA1 oligomerization and DNA binding, consistent with a requirement for oligomerization to bind DNA. Furthermore, LANA1 bound TR DNA as an oligomer. Deletion of amino acids 1007-1021, N-terminal to the LANA1 oligomerization domain, ablated DNA binding, DNA replication, and episome persistence, implicating these residues in contacting DNA. Indeed, LANA1 residues 1007-1021 correspond to EBNA1 residues that contact the cognate sequence. Like EBNA1, the LANA1 DNA-binding domain has oligomerization activity and critical residues essential for recognizing DNA.


Asunto(s)
Antígenos Virales/metabolismo , Replicación del ADN , ADN Viral/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas Nucleares/metabolismo , Animales , Antígenos Virales/genética , Proteínas Nucleares/genética , Plásmidos , Secuencias Repetidas Terminales , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...