Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Bull Am Meteorol Soc ; 101(3): E323-E340, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32483389

RESUMEN

Severe weather events including tornadoes, damaging winds, hail, and their combination produce changes in land surface vegetation and urban settings that are frequently observed through remote sensing. Capabilities continue to improve through a growing constellation of governmental and commercial assets, increasing the spatial resolution of visible, near to shortwave infrared, and thermal infrared remote sensing. Here, we highlight cases where visual interpretation of imagery benefitted severe weather damage assessments made within the NOAA/NWS Damage Assessment Toolkit. Examples demonstrate utility of imagery in assessing tracks and changes in remote areas where staffing limitations or access prevent a ground-based assessment.

2.
Cell Commun Signal ; 17(1): 76, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31315632

RESUMEN

BACKGROUND: Tumor suppressor WWOX physically binds p53 and TIAF1 and together induces apoptosis and tumor suppression. To understand the molecular action, here we investigated the formation of WWOX/TIAF1/p53 triad and its regulation of cancer cell migration, anchorage-independent growth, SMAD promoter activation, apoptosis, and potential role in neurodegeneration. METHODS: Time-lapse microscopy was used to measure the extent of cell migration. Protein/protein interactions were determined by co-immunoprecipitation, FRET microscopy, and yeast two-hybrid analysis. The WWOX/TIAF1/p53 triad-mediated cancer suppression was determined by measuring the extent of cell migration, anchorage-independent growth, SMAD promoter activation, and apoptosis. p53-deficient lung cancer cell growth in nude mice was carried out to assess the tumor suppressor function of ectopic p53 and/or WWOX. RESULTS: Wwox-deficient MEF cells exhibited constitutive Smad3 and p38 activation and migrated individually and much faster than wild type cells. TGF-ß increased the migration of wild type MEF cells, but significantly suppressed Wwox knockout cell migration. While each of the triad proteins is responsive to TGF-ß stimulation, ectopically expressed triad proteins suppressed cancer cell migration, anchorage-independent growth, and SMAD promoter activation, as well as caused apoptosis. The effects are due in part to TIAF1 polymerization and its retention of p53 and WWOX in the cytoplasm. p53 and TIAF1 were effective in suppressing anchorage-independent growth, and WWOX ineffective. p53 and TIAF1 blocked WWOX or Smad4-regulated SMAD promoter activation. WWOX suppressed lung cancer NCI-H1299 growth and inhibited splenomegaly by inflammatory immune response, and p53 blocked the event in nude mice. The p53/WWOX-cancer mice exhibited BACE upregulation, APP degradation, tau tangle formation, and amyloid ß generation in the brain and lung. CONCLUSION: The WWOX/TIAF1/p53 triad is potent in cancer suppression by blocking cancer cell migration, anchorage-independent growth and SMAD promoter activation, and causing apoptosis. Yet, p53 may functionally antagonize with WWOX. p53 blocks WWOX inhibition of inflammatory immune response induced by cancer, and this leads to protein aggregation in the brain as seen in the Alzheimer's disease and other neurodegeneration.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias de la Mama/terapia , Neoplasias Pulmonares/terapia , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Oxidorreductasa que Contiene Dominios WW/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Ratones Desnudos , Proteínas Nucleares/antagonistas & inhibidores , Agregado de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/deficiencia , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/deficiencia , Oxidorreductasa que Contiene Dominios WW/antagonistas & inhibidores , Oxidorreductasa que Contiene Dominios WW/deficiencia
3.
PLoS One ; 14(6): e0218883, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31251791

RESUMEN

A real-time understanding of the distribution and duration of power outages after a major disaster is a precursor to minimizing their harmful consequences. Here, we develop an approach for using daily satellite nighttime lights data to create spatially disaggregated power outage estimates, tracking electricity restoration efforts after disasters strike. In contrast to existing utility data, these estimates are independent, open, and publicly-available, consistently measured across regions that may be serviced by several different power companies, and inclusive of distributed power supply (off-grid systems). We apply the methodology in Puerto Rico following Hurricane Maria, which caused the longest blackout in US history. Within all of the island's settlements, we track outages and recovery times, and link these measures to census-based demographic characteristics of residents. Our results show an 80% decrease in lights, in total, immediately after Hurricane Maria. During the recovery, a disproportionate share of long-duration power failures (> 120 days) occurred in rural municipalities (41% of rural municipalities vs. 29% of urban municipalities), and in the northern and eastern districts. Unexpectedly, we also identify large disparities in electricity recovery between neighborhoods within the same urban area, based primarily on the density of housing. For many urban areas, poor residents, the most vulnerable to increased mortality and morbidity risks from power losses, shouldered the longest outages because they lived in less dense, detached housing where electricity restoration lagged. The approach developed in this study demonstrates the potential of satellite-based estimates of power recovery to improve the real-time monitoring of disaster impacts, globally, at a spatial resolution that is actionable for the disaster response community.


Asunto(s)
Tormentas Ciclónicas , Desastres , Electricidad , Nave Espacial , Humanos , Centrales Eléctricas , Puerto Rico
4.
Oncotarget ; 8(12): 19137-19155, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-27845895

RESUMEN

Malignant cancer cells frequently secrete significant amounts of transforming growth factor beta (TGF-ß), hyaluronan (HA) and hyaluronidases to facilitate metastasizing to target organs. In a non-canonical signaling, TGF-ß binds membrane hyaluronidase Hyal-2 for recruiting tumor suppressors WWOX and Smad4, and the resulting Hyal-2/WWOX/Smad4 complex is accumulated in the nucleus to enhance SMAD-promoter dependent transcriptional activity. Yeast two-hybrid analysis showed that WWOX acts as a bridge to bind both Hyal-2 and Smad4. When WWOX-expressing cells were stimulated with high molecular weight HA, an increased formation of endogenous Hyal-2/WWOX/Smad4 complex occurred rapidly, followed by relocating to the nuclei in 20-40 min. In WWOX-deficient cells, HA failed to induce Smad2/3/4 relocation to the nucleus. To prove the signaling event, we designed a real time tri-molecular FRET analysis and revealed that HA induces the signaling pathway from ectopic Smad4 to WWOX and finally to p53, as well as from Smad4 to Hyal-2 and then to WWOX. An increased binding of the Smad4/Hyal-2/WWOX complex occurs with time in the nucleus that leads to bubbling cell death. In contrast, HA increases the binding of Smad4/WWOX/p53, which causes membrane blebbing but without cell death. In traumatic brain injury-induced neuronal death, the Hyal-2/WWOX complex was accumulated in the apoptotic nuclei of neurons in the rat brains in 24 hr post injury, as determined by immunoelectron microscopy. Together, HA activates the Hyal-2/WWOX/Smad4 signaling and causes bubbling cell death when the signaling complex is overexpressed.


Asunto(s)
Muerte Celular/fisiología , Ácido Hialurónico/metabolismo , Neoplasias/patología , Oxidorreductasas/metabolismo , Transducción de Señal/fisiología , Proteína Smad4/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Transferencia Resonante de Energía de Fluorescencia , Humanos , Inmunoprecipitación , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Neoplasias/metabolismo , Ratas , Técnicas del Sistema de Dos Híbridos , Oxidorreductasa que Contiene Dominios WW
5.
PLoS One ; 4(6): e5755, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19484134

RESUMEN

BACKGROUND: Tissue exudates contain low levels of serum complement proteins, and their regulatory effects on prostate cancer progression are largely unknown. We examined specific serum complement components in coordinating the activation of tumor suppressors p53 and WWOX (also named FOR or WOX1) and kinases ERK, JNK1 and STAT3 in human prostate DU145 cells. METHODOLOGY/PRINCIPAL FINDINGS: DU145 cells were cultured overnight in 1% normal human serum, or in human serum depleted of an indicated complement protein. Under complement C1q- or C6-free conditions, WOX1 and ERK were mainly present in the cytoplasm without phosphorylation, whereas phosphorylated JNK1 was greatly accumulated in the nuclei. Exogenous C1q rapidly restored the WOX1 activation (with Tyr33 phosphorylation) in less than 2 hr. Without serum complement C9, p53 became activated, and hyaluronan (HA) reversed the effect. Under C6-free conditions, HA induced activation of STAT3, an enhancer of metastasis. Notably, exogenous C1q significantly induced apoptosis of WOX1-overexpressing DU145 cells, but not vehicle-expressing cells. A dominant negative and Y33R mutant of WOX1 blocked the apoptotic effect. C1q did not enhance p53-mediated apoptosis. By total internal reflection fluorescence (TIRF) microscopy, it was determined that C1q destabilized adherence of WOX1-expressing DU145 cells by partial detaching and inducing formation of clustered microvilli for focal adhesion particularly in between cells. These cells then underwent shrinkage, membrane blebbing and death. Remarkably, as determined by immunostaining, benign prostatic hyperplasia and prostate cancer were shown to have a significantly reduced expression of tissue C1q, compared to age-matched normal prostate tissues. CONCLUSIONS/SIGNIFICANCE: We conclude that complement C1q may induce apoptosis of prostate cancer cells by activating WOX1 and destabilizing cell adhesion. Downregulation of C1q enhances prostate hyperplasia and cancerous formation due to failure of WOX1 activation.


Asunto(s)
Apoptosis , Complemento C1q/fisiología , Regulación Neoplásica de la Expresión Génica , Oxidorreductasas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adhesión Celular , Línea Celular Tumoral , Complemento C1q/metabolismo , Regulación hacia Abajo , Genes Dominantes , Humanos , Masculino , Microscopía Fluorescente/métodos , Modelos Biológicos , Fosforilación , Proteína p53 Supresora de Tumor/metabolismo , Oxidorreductasa que Contiene Dominios WW
6.
J Biol Chem ; 284(23): 16049-59, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19366691

RESUMEN

Transforming growth factor beta (TGF-beta) initiates multiple signal pathways and activates many downstream kinases. Here, we determined that TGF-beta1 bound cell surface hyaluronidase Hyal-2 on microvilli in type II TGF-beta receptor-deficient HCT116 cells, as determined by immunoelectron microscopy. This binding resulted in recruitment of proapoptotic WOX1 (also named WWOX or FOR) and formation of Hyal-2.WOX1 complexes for relocation to the nuclei. TGF-beta1 strengthened the binding of the catalytic domain of Hyal-2 with the N-terminal Tyr-33-phosphorylated WW domain of WOX1, as determined by time lapse fluorescence resonance energy transfer analysis in live cells, co-immunoprecipitation, and yeast two-hybrid domain/domain mapping. In promoter activation assay, ectopic WOX1 or Hyal-2 alone increased the promoter activity driven by Smad. In combination, WOX1 and Hyal-2 dramatically enhanced the promoter activation (8-9-fold increases), which subsequently led to cell death (>95% of promoter-activated cells). TGF-beta1 supports L929 fibroblast growth. In contrast, transiently overexpressed WOX1 and Hyal-2 sensitized L929 to TGF-beta1-induced apoptosis. Together, TGF-beta1 invokes a novel signaling by engaging cell surface Hyal-2 and recruiting WOX1 for regulating the activation of Smad-driven promoter, thereby controlling cell growth and death.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Membrana Celular/fisiología , Hialuronoglucosaminidasa/fisiología , Oxidorreductasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Apoptosis , Moléculas de Adhesión Celular/genética , Neoplasias Colorrectales , Cartilla de ADN , Transferencia Resonante de Energía de Fluorescencia , Proteínas Ligadas a GPI , Regulación de la Expresión Génica , Células HCT116 , Humanos , Hialuronoglucosaminidasa/genética , Células L , Ratones , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/deficiencia , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Oxidorreductasa que Contiene Dominios WW
7.
Cell Signal ; 20(7): 1303-12, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18403180

RESUMEN

Zfra is a small size 31-amino-acid C2H2 zinc finger-like protein, which is known to interact with c-Jun N-terminal kinase 1 (JNK1), WW domain-containing oxidoreductase (WWOX, FOR or WOX1), TNF receptor-associated death domain protein (TRADD) and nuclear factor kappaB (NF-kappaB) during stress response. Here, we show that Zfra became phosphorylated at Ser8 (as determined by specific antibody) and translocated to the mitochondria in response to inducers of mitochondrial permeability transition (MPT) (e.g. staurosporine and betulinic acid). Overexpressed Zfra induced cell death. This event is associated, in part, with increased dissipation of mitochondrial membrane potential (MMP) and increased chromosomal DNA fragmentation. Intriguingly, Zfra significantly downregulated Bcl-2 and yet blocked cytochrome c release from the mitochondria. Overexpression of an S8G-Zfra mutant (Ser8 to Gly8 alteration) could not induce cell death, probably due to its failure of translocating to the mitochondria and causing MMP dissipation. Over-expressed proapoptotic WOX1 induced cytochrome c release from the mitochondria. Zfra bound and blocked the effect of WOX1. Taken together, Ser8 is essential for overexpressed Zfra to exert cell death via the mitochondrial pathway. Zfra downregulates Bcl-2 and induces MMP dissipation but causes no cytochrome c release, indicating a novel death pathway from the mitochondria.


Asunto(s)
Citocromos c/metabolismo , Proteínas de Unión al ADN/metabolismo , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Proteínas de Unión al ADN/química , Regulación hacia Abajo/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Datos de Secuencia Molecular , Oxidorreductasas/metabolismo , Triterpenos Pentacíclicos , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Serina/metabolismo , Treonina/metabolismo , Triterpenos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Ácido Betulínico
8.
BMC Mol Biol ; 8: 50, 2007 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-17567906

RESUMEN

BACKGROUND: Zfra is a 31-amino-acid zinc finger-like protein, which is known to regulate cell death by tumor necrosis factor (TNF) and overexpressed TNF receptor- or Fas-associated death domain proteins (TRADD and FADD). In addition, Zfra undergoes self-association and interacts with c-Jun N-terminal kinase 1 (JNK1) in response to stress stimuli. To further delineate the functional properties of Zfra, here we investigated Zfra regulation of the activation of p53, WOX1 (WWOX or FOR), NF-kappaB, and JNK1 under apoptotic stress. RESULTS: Transiently overexpressed Zfra caused growth suppression and apoptotic death of many but not all types of cells. Zfra either enhanced or blocked cell death caused by TRADD, FADD, or receptor-interacting protein (RIP) in a dose-related manner. This modulation is related with Zfra binding with TRADD, NF-kappaB, JNK1 and WOX1, as determined by GST pull-down analysis, co-immunoprecipitation, and mapping by yeast two-hybrid analysis. Functionally, transiently overexpressed Zfra sequestered NF-kappaB (p65), WOX1, p53 and phospho-ERK (extracellular signal-activated kinase) in the cytoplasm, and TNF or UV light could not effectively induce nuclear translocation of these proteins. Zfra counteracted the apoptotic functions of Tyr33-phosphorylated WOX1 and Ser46-phosphorylated p53. Alteration of Ser8 to Gly abolished the apoptotic function of Zfra and its regulation of WOX1 and p53. CONCLUSION: In response to TNF, Zfra is upregulated and modulates TNF-mediated cell death via interacting with TRADD, FADD and RIP (death-inducing signaling complex) at the receptor level, and downstream effectors NF-kappaB, p53, WOX1, and JNK1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Oxidorreductasas/metabolismo , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Muerte Celular/fisiología , Línea Celular , Chlorocebus aethiops , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , Proteína Quinasa 8 Activada por Mitógenos/genética , Mutagénesis Sitio-Dirigida , FN-kappa B/genética , Oxidorreductasas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Serina/metabolismo , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Técnicas del Sistema de Dos Híbridos , Tirosina/metabolismo , Rayos Ultravioleta , Oxidorreductasa que Contiene Dominios WW
9.
Int J Mol Med ; 19(1): 173-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17143562

RESUMEN

Linear non-sulfated hyaluronan (HA) does not bind complement proteins yet inhibits their hemolytic function. We have previously induced the complement inhibitory function of HA by heat treatment. However, heated HA readily loses its anti-complementary activity probably due to instantaneous interchain re-association. Here, HA solutions were heated and then freeze-dried. Compared to native HA, heated/freeze-dried HA stably restricted serum complement-mediated hemolysis via the classical pathway, in which serum C1 hemolytic function and C3 activation were blocked. Also, treated HA had a significantly increased binding of component C1q, C1r, C1s, C2, C5, C9, P, D and H. Further, when HA was gel-fractionated by electrophoresis and then freeze-dried, its anti-complementary activity was stably induced. Both native and heated/freeze-dried HA stimulated ERK phosphorylation in prostate DU145 cells. However, treated HA suppressed the expression of tumor suppressors WOX1 and WOX2. Together, HA with an altered conformation stabilizes its inhibition and binding of complement proteins. It may recognize cell surface receptors differently from native HA, thereby differentially regulating the expression of cellular proteins.


Asunto(s)
Vía Clásica del Complemento , Proteínas del Sistema Complemento/metabolismo , Ácido Hialurónico/química , Oxidorreductasas/metabolismo , Neoplasias de la Próstata/metabolismo , Línea Celular Tumoral , Activación de Complemento/efectos de los fármacos , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Liofilización , Geles/química , Hemólisis/efectos de los fármacos , Calor , Humanos , Ácido Hialurónico/farmacología , Masculino , Conformación Molecular , Fosforilación , Proteínas Supresoras de Tumor , Oxidorreductasa que Contiene Dominios WW
10.
J Biol Chem ; 280(52): 43100-8, 2005 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-16219768

RESUMEN

WW domain-containing oxidoreductase WOX1, also named WWOX or FOR, undergoes Tyr33 phosphorylation at its first N-terminal WW domain and subsequent nuclear translocation in response to sex steroid hormones and stress stimuli. The activated WOX1 binds tumor suppressor p53, and both proteins may induce apoptosis synergistically. Functional suppression of WOX1 by antisense mRNA or a dominant negative abolishes p53-mediated apoptosis. Here, we determined that UV light, anisomycin, etoposide, and hypoxic stress rapidly induced phosphorylation of p53 at Ser46 and WOX1 at Tyr33 (phospho-WOX1) and their binding interactions in several tested cancer cells. Mapping by yeast two-hybrid analysis and co-immunoprecipitation showed that phospho-WOX1 physically interacted with Ser46-phosphorylated p53. Knockdown of WOX1 protein expression by small interfering RNA resulted in L929 fibroblast resistance to apoptosis by tumor necrosis factor, staurosporine, UV light, and ectopic p53, indicating an essential role of WOX1 in stress stimuli-induced apoptosis. Notably, UV light could not induce p53 protein expression in these WOX1 knockdown cells, although p53 mRNA levels were not reduced. Suppression of WOX1 by dominant negative WOX1 (to block Tyr33 phosphorylation) also abolished UV light-induced p53 protein expression. Time course analysis showed that the stability of ectopic wild type p53, tagged with DsRed, was decreased in WOX1 knockdown cells. Inhibition of MDM2 by nutlin-3 increased the binding of p53 and WOX1 and stability of p53. Together, our data show that WOX1 plays a critical role in conferring cellular sensitivity to apoptotic stress and that Tyr33 phosphorylation in WOX1 is essential for binding and stabilizing Ser46-phosphorylated p53.


Asunto(s)
Oxidorreductasas/fisiología , Serina/química , Estaurosporina/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Tirosina/química , Transporte Activo de Núcleo Celular , Animales , Anisomicina/farmacología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Citosol/metabolismo , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Etopósido/farmacología , Fibroblastos/metabolismo , Genes Dominantes , Humanos , Hipoxia , Imidazoles/metabolismo , Inmunoprecipitación , Proteínas Luminiscentes/farmacología , Ratones , Microscopía Fluorescente , Modelos Biológicos , Fosforilación , Piperazinas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteínas Supresoras de Tumor , Técnicas del Sistema de Dos Híbridos , Células U937 , Rayos Ultravioleta , Oxidorreductasa que Contiene Dominios WW
11.
Biochem Biophys Res Commun ; 327(2): 415-23, 2005 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-15629131

RESUMEN

By cDNA library screening, here we isolated an unusual gene transcript encoding a 31-amino-acid zinc finger-like peptide that regulates apoptosis (named Zfra). Northern blotting and RT/PCR showed the transcript is abundant in spleen but absent in several prostate and breast cancer cells. When stably expressed in L929 fibroblasts, Zfra conferred resistance to the cytotoxic effects of TNF and FasL. In contrast, transiently expressed Zfra could enhance or inhibit the cytotoxicity of overexpressed death domain proteins TRADD, FADD, and RIP of the TNF signaling pathway. By GST pull-down assay and co-immunoprecipitation, TNF and UV light were shown to induce Zfra to rapidly self-associate and bind JNK1. While JNK1 is a downstream effector of the TNF signaling, Zfra regulation of the TNF cytotoxic function is likely due to its interaction, in part, with JNK1.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/toxicidad , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Apoptosis/efectos de los fármacos , Secuencia de Bases , Proteínas Portadoras , Línea Celular , Clonación Molecular , ADN Complementario/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteína Ligando Fas , Proteína de Dominio de Muerte Asociada a Fas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Focalización Isoeléctrica , Glicoproteínas de Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Peso Molecular , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia , Factores de Transcripción/química , Factores de Transcripción/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo
12.
Oncogene ; 24(4): 714-23, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15580310

RESUMEN

Human WWOX gene encodes a proapoptotic WW domain-containing oxidoreductase WOX1 (also named WWOX, FOR2 or WWOXv1). Apoptotic and stress stimuli activate WOX1 via Tyr33 phosphorylation and nuclear translocation. WOX1 possesses a tetrad NSYK motif in the C-terminal short-chain alcohol dehydrogenase/reductase (SDR) domain, which may bind estrogen and androgen. Here, we determined that 17beta-estradiol (E(2)) activated WOX1, p53 and ERK in COS7 fibroblasts, primary lung epithelial cells, and androgen receptor (AR)-negative prostate DU145 cells, but not in estrogen receptor (ER)-positive breast MCF7 cells. Androgen also activated WOX1 in the AR-negative DU145 cells. These observations suggest that sex hormone-mediated Tyr33 phosphorylation and nuclear translocation of WOX1 is independent of ER and AR. Stress stimuli increase physical binding of p53 with WOX1 in vivo. We determined here that E(2) increased the formation of p53/WOX1 complex and their nuclear translocation in COS7 cells; however, nuclear translocation of this complex could not occur in MCF7 cells. By immunohistochemistry, we determined that progression of prostate from normal to hyperplasia, cancerous and metastatic stages positively correlate with upregulation and activation of WOX1 and WOX2 (FOR1/WWOXv2). In contrast, breast cancer development to a premetastatic state is associated with upregulation and Tyr33 phosphorylation of cytosolic WOX1 and WOX2, followed by significant downregulation or absent expression during metastasis. These Tyr33-phosphorylated proteins are mostly located in the mitochondria without translocating to the nuclei, which is comparable to those findings in cultured breast cancer cells. Together, sex steroid hormone-induced activation of WOX1 and WOX2 is independent of ER and AR, and this activation positively correlates with cancerous progression of prostate and breast to a premetastatic state.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Estradiol/farmacología , Oxidorreductasas/metabolismo , Neoplasias de la Próstata/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Progresión de la Enfermedad , Humanos , Hiperplasia , Masculino , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Metástasis de la Neoplasia , Oxidorreductasas/genética , Fosforilación , Neoplasias de la Próstata/patología , Unión Proteica , Transporte de Proteínas , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor , Tirosina/genética , Tirosina/metabolismo , Oxidorreductasa que Contiene Dominios WW
13.
J Biol Chem ; 279(29): 30498-506, 2004 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-15126504

RESUMEN

Numerous enzymes hyperphosphorylate Tau in vivo, leading to the formation of neurofibrillary tangles (NFTs) in the neurons of Alzheimer's disease (AD). Compared with age-matched normal controls, we demonstrated here that the protein levels of WW domain-containing oxidoreductase WOX1 (also known as WWOX or FOR), its Tyr33-phosphorylated form, and WOX2 were significantly down-regulated in the neurons of AD hippocampi. Remarkably knock-down of WOX1 expression by small interfering RNA in neuroblastoma SK-N-SH cells spontaneously induced Tau phosphorylation at Thr212/Thr231 and Ser515/Ser516, enhanced phosphorylation of glycogen synthase kinase 3beta (GSK-3beta) and ERK, and enhanced NFT formation. Also an increased binding of phospho-GSK-3beta with phospho-Tau was observed in these WOX1 knock-down cells. In comparison, increased phosphorylation of Tau, GSK-3beta, and ERK, as well as NFT formation, was observed in the AD hippocampi. Activation of JNK1 by anisomycin further increased Tau phosphorylation, and SP600125 (a JNK inhibitor) and PD-98059 (an MEK1/2 inhibitor) blocked Tau phosphorylation and NFT formation in these WOX1 knock-down cells. Ectopic or endogenous WOX1 colocalized with Tau, JNK1, and GSK-3beta in neurons and cultured cells. 17Beta-estradiol, a neuronal protective hormone, increased the binding of WOX1 and GSK-3beta with Tau. Mapping analysis showed that WOX1 bound Tau via its COOH-terminal short-chain alcohol dehydrogenase/reductase domain. Together WOX1 binds Tau via its short-chain alcohol dehydrogenase/reductase domain and is likely to play a critical role in regulating Tau hyperphosphorylation and NFT formation in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Regulación hacia Abajo , Oxidorreductasas/biosíntesis , Oxidorreductasas/química , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Animales , Anisomicina/farmacología , Antracenos/farmacología , Encéfalo/patología , Células COS , Línea Celular , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Estradiol/metabolismo , Flavonoides/farmacología , Vectores Genéticos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Técnicas In Vitro , Ratones , Microscopía Fluorescente , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/metabolismo , Fosforilación , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Retroviridae/genética , Serina/química , Temperatura , Treonina/química , Factores de Tiempo , Proteínas Supresoras de Tumor , Técnicas del Sistema de Dos Híbridos , Tirosina/química , Oxidorreductasa que Contiene Dominios WW
14.
DNA Cell Biol ; 23(1): 67-74, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14965474

RESUMEN

TIAF1 is a TGF-beta 1-induced factor that protects L929 fibroblasts from TNF-mediated apoptosis. In contrast, overexpressed TIAF1 induces growth inhibition and apoptosis of monocytic U937 and various nonfibroblast cells. TIAF1-mediated apoptosis of U937 cells involves upregulation of p53, p21, and Smad2/4, but downregulation of ERK phosphorylation. To determine whether p53 and TIAF1 functionally interact in regulating cell death, ectopic TIAF1 and p53 were shown to induce apoptosis of U937 cells in both synergistic and antagonistic manners. At optimal levels both TIAF1 and p53 mediated apoptosis cooperatively. Also, both proteins suppressed adherence-independent growth of L929 cells. In contrast, initiation of apoptosis by overexpressed TIAF1 was blocked by low doses of p53, and vice versa. Furthermore, ectopic p53 blocked an ongoing apoptosis in U937 cells stably expressing TIAF1. Yeast two-hybrid analyses failed to demonstrate the binding of p53 with TIAF1, suggesting an unidentified protein that links the p53/TIFA1 interaction. Suppression of TIAF1 expression by siRNA could not inhibit Ser15 phosphorylation in p53 in response to UV and etoposide. However, nuclear translocation of these Ser15-phosphorylated p53 was significantly reduced in TIAF1-silenced cells. Taken together, TIAF1 and p53 functionally interact in regulating apoptosis, and TIAF1 is likely to participate in the nuclear translocation of activated p53.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Silenciador del Gen , Cadenas Pesadas de Miosina/metabolismo , Miosinas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , División Celular , Humanos , Células L , Ratones , Proteínas Nucleares , Fosforilación , Fosfoserina/metabolismo , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Células U937
15.
Biochem Pharmacol ; 66(8): 1347-54, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14555208

RESUMEN

Human WWOX gene encodes a putative tumor suppressor WW domain-containing oxidoreductase WOX1 (also known as WWOX or FOR). A high frequency of loss of heterozygosity (LOH) of this gene has been shown in prostate, lung, breast and other cancers. In addition, numerous aberrant WWOX mRNA transcripts have been found in cancer cells. WOX1 is a proapoptotic protein. In response to stress or apoptotic stimuli, WOX1 became phosphorylated at Tyr33, which enabled its complex formation with activated p53 and JNK1. The p53/WOX1 complex translocated to the mitochondria and further to the nuclei to mediate apoptosis. WOX1 mutants, which were inactivated for nuclear translocation or Tyr33 phosphorylation, failed to induce apoptosis, indicating that activation of WOX1 via Tyr33 phosphorylation, followed by nuclear translocation, is essential for inducing cell death. WOX1 induced apoptosis synergistically with p53. In contrast, transiently activated JNK1 induced anti-apoptotic response, and this protective activity inhibited WOX1-induced apoptosis. Taken together, WOX1 is involved in stress and apoptotic responses, and is likely to regulate the activation of both p53 and JNK1.


Asunto(s)
Apoptosis/fisiología , Oxidorreductasas/metabolismo , Estrés Fisiológico/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Transporte Biológico , Núcleo Celular/metabolismo , Insuficiencia Cardíaca , Humanos , Mitocondrias/metabolismo , Proteína Quinasa 8 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Factor de Necrosis Tumoral alfa/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Oxidorreductasa que Contiene Dominios WW
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...