Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
3.
FEBS Lett ; 595(13): 1782-1796, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33960419

RESUMEN

Sorcin is a calcium-binding protein involved in maintaining endoplasmic reticulum (ER) Ca2+ stores. We have previously shown that overexpressing sorcin under the rat insulin promoter was protective against high-fat diet-induced pancreatic beta-cell dysfunction in vivo. Activating transcription factor 6 (ATF6) is a key mediator of the unfolded protein response (UPR) that provides cellular protection during the progression of ER stress. Here, using nonexcitable HEK293 cells, we show that sorcin overexpression increased ATF6 signalling, whereas sorcin knock out caused a reduction in ATF6 transcriptional activity and increased ER stress. Altogether, our data suggest that sorcin downregulation during lipotoxic stress may prevent full ATF6 activation and a normal UPR during the progression of obesity and insulin resistance.


Asunto(s)
Factor de Transcripción Activador 6/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Técnicas de Inactivación de Genes/métodos , Obesidad/genética , Palmitatos/efectos adversos , Animales , Calcio/metabolismo , Células Cultivadas , Progresión de la Enfermedad , Regulación hacia Abajo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Humanos , Resistencia a la Insulina , Ratones , Obesidad/metabolismo , Transducción de Señal , Activación Transcripcional/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos
4.
Cell Death Dis ; 11(6): 445, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32522979

RESUMEN

SHQ1 was reported to control the biogenesis and assembly of H/ACA ribonucleoprotein particles (RNPs). It was independently isolated as a growth suppressor, GRIM1, in a genetic screen. Recent studies have indicated that SHQ1 inhibits prostate cancer growth and metastasis. SHQ1 facilitates MYC RNA splicing to promote T-acute lymphoblastic leukemia (T-ALL) development. Thus, the mechanisms of SHQ1 in cancers remain largely unknown. We report here that SHQ1 promotes tumor apoptosis and chemo-sensitivity in hepatocellular carcinoma (HCC) cells. In HCC tissues from patients, expression of SHQ1 was significantly decreased in the tumor compared to adjacent tissues. Experiments with HCC xenograft models revealed that restoring SHQ1 levels enhanced the anti-tumor activity of the endoplasmic reticulum (ER) stress inducer tunicamycin (TM) and common chemotherapy drug paclitaxel (PTX). Mechanistically, SHQ1 is an ER-stress response gene which is regulated by p50ATF6 and XBP1s through an ER stress response like element located on the SHQ1 promoter. SHQ1 interacts with the ER chaperone GRP78 to release ER sensors PERK/IRE1α/ATF6 from GRP78/ER-sensor complexes, leading to hyper-activation of unfolded protein response (UPR). In the persistent ER stress conditions of a HepG2 xenograft tumor model, SHQ1-mediated hyper-activation of ER-sensor signaling induces apoptosis. Our study thus demonstrates a SHQ1-mediated ER-stress response feedback loop that promotes tumor sensitivity to chemotherapeutics.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Estrés del Retículo Endoplásmico/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Unión Competitiva/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Proteínas de Choque Térmico/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Paclitaxel/farmacología , Unión Proteica/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Interferon Cytokine Res ; 38(9): 378-387, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30230984

RESUMEN

Melanoma is one of the most lethal skin malignancies in the world. Interferons (IFNs) have been also demonstrated in response to tumor cell and IFNs such as IFN-α have been used for melanoma treatment. The long chain double-stranded RNA (dsRNA) (from a variety of nonviral sources) is a potent activator of the IFN system and an inducer of cell apoptosis. Panaxadiolsaponins (PDS) is a major Panax ginseng-derived active component with known antitumor activity and immune modulation. Here, we investigated a hypothesis that the combination of PDS and total natural dsRNA (as opposed to the synthetic dsRNA) will suppress tumor growth better than the individual agents. We have evaluated the antitumor and immunostimulatory effects of the combination of natural long chain dsRNA (derived from yeast) and PDS on melanoma cell line B16 and mice xenograft model. The underlying mechanisms of growth suppression were investigated by analyzing dsRNA-activated pathways, apoptosis, and cell cycle. Natural dsRNA and PDS exert superior anticancer effects than either agent alone. Natural dsRNA and PDS combination might be a promising strategy for treating malignancies, including melanoma.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Melanoma Experimental/tratamiento farmacológico , ARN Bicatenario/farmacología , Sapogeninas/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Melanoma Experimental/patología , Ratones , Panax/química , Células Tumorales Cultivadas
6.
Oncotarget ; 8(40): 68191-68207, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978108

RESUMEN

Studies suggest that tunicamycin may work as a therapeutic drug to cancer cells by inducing stress in the endoplasmic reticulum (ER) through unfolded protein response (UPR) and thereby promoting apoptosis. However, mechanisms of the prolonged activation of the UPR under sustained ER stress in the regulation of cell apoptosis are largely unknown. To delineate the role of candidate genes in the apoptotic process under ER stress and to search for new therapeutic strategies to treat metastatic castration resistant prostate cancer, we performed whole genome expression microarray analysis in tunicamycin treated metastatic androgen-insensitive prostate cancer cells, PC-3. Among several induced genes, the expression of eNOS (NOS3) gene was remarkably high. The increased expression of eNOS activates mTORC1 through RagC. This results into an accumulation of p62 (SQSTM1) which facilitates aggregation of ubiquitinated protein thus compromising clearance of misfolded toxic protein aggregates. Lastly, association of p62 proteins and misfolded proteins promote reactive oxygen species (ROS) mediated mitochondrial apoptosis. Overall, our data demonstrate that tunicamycin induced ER stress promotes prostate cancer cell death by activating mTORC1 through eNOS-RagC pathway.

7.
Mol Carcinog ; 56(7): 1687-1702, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28218424

RESUMEN

p16INK4A and p53 are two important tumor suppressor proteins that play essential roles during cell proliferation and aging through regulating the expression of several genes. Here, we report that p16INK4A and p53 co-regulate a plethora of transcripts. Furthermore, both proteins colocalize in the nucleus of human primary skin fibroblasts and breast luminal cells, and form a heteromer whose level increases in response to genotoxic stress as well as aging of human fibroblasts and various mouse organs. CDK4 is also present in this heteromeric complex, which is formed only in the presence of DNA both in vitro using pure recombinant proteins and in vivo. We have also shown that p16INK4A enhances the binding efficiency of p53 to its cognate sequence presents in the CDKN1A promoter in vitro, and both proteins are present at the promoters of CDKN1A and BAX in vivo. Importantly, the fourth ankyrin repeat of p16INK4A and the C-terminal domain of p53 were necessary for the physical association between these two proteins. The physiologic importance of this association was revealed by the inability of cancer-associated p16INK4A mutants to interact with p53 and to transactivate the expression of its major targets CDKN1A and BAX in the p16-defective U2OS cells expressing either wild-type or mutated p16INK4A . Furthermore, the association between p16INK4A and p53 was capital for their nuclear colocalization, the X-ray-dependent induction of p21 and Bax proteins as well as the induction of apoptosis in various types of cells. Together, these results show DNA-dependent physical interaction between p16INK4A and p53.


Asunto(s)
Apoptosis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , ADN/metabolismo , Regulación de la Expresión Génica , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , ADN/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/metabolismo
8.
J Biol Chem ; 291(42): 22030-22042, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27590344

RESUMEN

Expression of DAPK1, a critical regulator of autophagy and apoptosis, is lost in a wide variety of tumors, although the mechanisms are unclear. A transcription factor complex consisting of ATF6 (an endoplasmic reticulum-resident factor) and C/EBP-ß is required for the IFN-γ-induced expression of DAPK1 IFN-γ-induced proteolytic processing of ATF6 and phosphorylation of C/EBP-ß are obligatory for the formation of this transcriptional complex. We report that defects in this pathway fail to control growth of chronic lymphocytic leukemia (CLL). Consistent with these observations, IFN-γ and chemotherapeutics failed to activate autophagy in CLL patient samples lacking ATF6 and/or C/EBP-ß. Together, these results identify a molecular basis for the loss of DAPK1 expression in CLL.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Autofagia , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Leucemia Linfocítica Crónica de Células B/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Transcripción Activador 6/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular Transformada , Proteínas Quinasas Asociadas a Muerte Celular/genética , Femenino , Humanos , Interferón gamma/farmacología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Masculino , Proteínas de Neoplasias/genética
9.
Pathog Dis ; 73(9): ftv080, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26424808

RESUMEN

Bacillus anthracis, the causative agent of anthrax, is acquired by mammalian hosts from the environment, as quiescent endospores. These endospores must germinate inside host cells, forming vegetative bacilli, before they can express the virulence factors that enable them to evade host defenses and disseminate throughout the body. While the role of macrophages and dendritic cells in this initial interaction has been established, the role of polymorphonuclear leukocytes (PMNs) has not been adequately defined. We discovered that while B. anthracis 34F2 Sterne endospores germinate poorly within non-activated human PMNs, these phagocytes exhibit rapid microbicidal activity toward the outgrown vegetative bacilli, independent of superoxide and nitric oxide. These findings suggest that a non-free radical pathway kills B. anthracis bacilli. We also find in PMNs an autophagic mechanism of bacterial killing based on the rapid induction of LC-3 conversion, beclin-1 expression, sequestosome 1 (SQSTM1) degradation and inhibition of bactericidal activity by the inhibitor, 3-methyladenine. These findings extend to PMNs an autophagic bactericidal mechanism previously described for other phagocytes.


Asunto(s)
Carbunco/inmunología , Carbunco/patología , Autofagia , Bacillus anthracis/inmunología , Viabilidad Microbiana , Neutrófilos/inmunología , Neutrófilos/microbiología , Animales , Western Blotting , Células Cultivadas , Humanos , Ratones , Fagocitosis , Especies de Nitrógeno Reactivo/análisis , Especies Reactivas de Oxígeno/análisis
10.
Mol Cell Biol ; 34(21): 4033-48, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25135476

RESUMEN

The death-associated protein kinase 1 (DAPK1) is an important regulator of cell death and autophagy. Recently, we have identified that ATF6, an endoplasmic reticulum-resident transcription factor, in association with the transcription factor CEBP-ß, regulates the gamma interferon (IFN-γ)-induced expression of Dapk1 (P. Gade et al., Proc. Natl. Acad. Sci. U. S. A. 109:10316-10321, 2012, doi.org/10.1073/pnas.1119273109). IFN-γ-induced proteolytic processing of ATF6 and phosphorylation of C/EBP-ß were essential for the formation of a novel transcriptional complex that regulates DAPK1. Here, we report that IFN-γ activates the ASK1-MKK3/MKK6-p38 mitogen-activated protein kinase (MAPK) pathway for controlling the activity of ATF6. The terminal enzyme in this pathway, p38 MAPK, phosphorylates a critical threonine residue in ATF6 upstream of its DNA binding domain. ATF6 mutants defective for p38 MAPK phosphorylation fail to undergo proteolytic processing in the Golgi apparatus and drive IFN-γ-induced gene expression and autophagy. We also show that mice lacking Ask1 are highly susceptible to lethal bacterial infection owing to defective autophagy. Together, these results identify a novel host defense pathway controlled by IFN-γ signaling.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Autofagia , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Interferón gamma/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas , Factor de Transcripción Activador 6/genética , Animales , Bacillus anthracis/patogenicidad , Sitios de Unión , Línea Celular , Proteínas Quinasas Asociadas a Muerte Celular/genética , Técnicas de Silenciamiento del Gen , Humanos , Hígado/inmunología , Hígado/microbiología , Ratones , Mutación , Fosforilación , Células Sf9 , Bazo/inmunología , Bazo/microbiología
11.
Breast Cancer Res Treat ; 145(1): 5-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24668500

RESUMEN

Nicotine, a main addictive compound in tobacco smoke, has been linked to promotion and progression of lung, head and neck, pancreatic, and breast cancers, but the detailed mechanisms of cancer progression remain elusive. Here, we show that nicotine induces the expression of galectin-3 (an anti-apoptotic ß-galactoside-binding lectin) in breast cancer cell line and in primary tumors from breast cancer patients. Nicotine-induced up regulation of galectin-3 is due to an increased expression of α9 isoform of nicotinic acetylcholine receptor (α9nAChR), which activates transcription factor STAT3 that in turn, physically binds to galectin-3 (LGALS3) promoter and induces transcription of galectin-3. Intracellular galectin-3 increased mitochondrial integrity and suppressed chemotherapeutic-induced apoptosis of breast cancer cell. Moreover, nicotine-induced enrichment of side population cells with cancer stem cell-like properties was modulated by galectin-3 expression and could be significantly reduced by transient knock down of LGALS3 and its upstream signaling molecules STAT3 and α9nAChR. Thus, galectin-3 or its upstream signaling molecule STAT3 or α9nAChR could be a potential target to prevent nicotine-induced chemoresistance in breast cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Células Madre Neoplásicas/metabolismo , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Transducción de Señal/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Fragmentación del ADN , Galectina 3/biosíntesis , Humanos , Inmunohistoquímica , Células Madre Neoplásicas/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Nicotínicos/metabolismo , Factor de Transcripción STAT3/metabolismo , Células de Población Lateral , Transfección
12.
J Biol Chem ; 288(49): 35511-25, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24163379

RESUMEN

p16(INK4a) is a tumor suppressor protein involved in several stress-related cellular responses, including apoptosis. Recent lines of evidence indicate that p16(INK4a) is also a modulator of gene expression. However, the molecular mechanisms underlying this novel function are still obscure. Here, we present clear evidence that p16(INK4a) modulates the levels of various microRNAs, with marked positive effect on miR-141 and miR-146b-5p. This effect is mediated through the formation of the p16-CDK4-Sp1 heterocomplex, which binds to Sp1 consensus-binding motifs present in the promoters of miR-141 and miR-146b-5p, and it enables their transcription. In addition, we have shown that p16(INK4a) interacts with Sp1 through the fourth ankyrin repeat, which is crucial for Sp1 binding to the miR-141 and miR-146b-5p promoters and their transcriptional activation. The physiological importance of this association was revealed by the inability of cancer-related p16(INK4a) mutants to interact with Sp1. Moreover, we have shown p16-CDK4-Sp1-dependent up-regulation of miR-141 and miR-146b-5p following UV light-induced DNA damage and the role of these two microRNAs in mediating p16-related induction of apoptosis in response to this genotoxic stress. Together, these results indicate that p16(INK4a) associates with CDK4 not only to inhibit the cell cycle but also to enable the transcription of two important onco-microRNAs, which act as downstream effectors.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Factor de Transcripción Sp1/metabolismo , Secuencia de Bases , Ciclo Celular , Línea Celular , Quinasa 4 Dependiente de la Ciclina/química , Inhibidor p16 de la Quinasa Dependiente de Ciclina/química , Daño del ADN , Humanos , Datos de Secuencia Molecular , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Estabilidad del ARN , Factor de Transcripción Sp1/química , Activación Transcripcional/efectos de la radiación , Rayos Ultravioleta/efectos adversos
13.
Proc Natl Acad Sci U S A ; 110(45): E4213-22, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24145455

RESUMEN

Gene-associated with retinoid-interferon induced mortality-19 (GRIM-19), a STAT3-inhibitory protein, was isolated as a growth-suppressive gene product using a genome-wide expression knockdown screen. We and others have shown a loss of expression and occurrence of mutations in the GRIM-19 gene in a variety of primary human cancers, indicating its potential role as tumor suppressor. To help investigate its role in tumor development in vivo, we generated a genetically modified mouse in which Grim-19 can be conditionally inactivated. Deletion of Grim-19 in the skin significantly increased the susceptibility of mice to chemical carcinogenesis, resulting in development of squamous cell carcinomas. These tumors had high Stat3 activity and an increased expression of Stat3-responsive genes. Loss of Grim-19 also caused mitochondrial electron transport dysfunction resulting from failure to assemble electron transport chain complexes and altered the expression of several cellular genes involved in glycolysis. Surprisingly, the deletion of a single copy of the Grim-19 gene was sufficient to promote carcinogenesis and formation of invasive squamous cell carcinomas. These observations highlight the critical role of GRIM-19 as a tumor suppressor.


Asunto(s)
Carcinogénesis/genética , Carcinoma de Células Escamosas/genética , NADH NADPH Oxidorreductasas/genética , Animales , Cartilla de ADN/genética , Componentes del Gen , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Vectores Genéticos/genética , Inmunohistoquímica , Ratones , Ratones Noqueados , NADH NADPH Oxidorreductasas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/metabolismo , Análisis de Secuencia de ARN
14.
Oxid Med Cell Longev ; 2013: 798401, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23738042

RESUMEN

Lung epithelial and endothelial cell death caused by pro-oxidant insults is a cardinal feature of acute lung injury/acute respiratory distress syndrome (ALI/ARDS) patients. The NF-E2-related factor 2 (NRF2) activation in response to oxidant exposure is crucial to the induction of several antioxidative and cytoprotective enzymes that mitigate cellular stress. Since prolonged exposure to hyperoxia causes cell death, we hypothesized that chronic hyperoxia impairs NRF2 activation, resulting in cell death. To test this hypothesis, we exposed nonmalignant small airway epithelial cells (AECs) to acute (1-12 h) and chronic (36-48 h) hyperoxia and evaluated cell death, NRF2 nuclear accumulation and target gene expression, and NRF2 recruitment to the endogenous HMOX1 and NQO1 promoters. As expected, hyperoxia gradually induced death in AECs, noticeably and significantly by 36 h; ~60% of cells were dead by 48 h. However, we unexpectedly found increased expression levels of NRF2-regulated antioxidative genes and nuclear NRF2 in AECs exposed to chronic hyperoxia as compared to acute hyperoxia. Chromatin Immunoprecipitation (ChIP) assays revealed an increased recruitment of NRF2 to the endogenous HMOX1 and NQO1 promoters in AECs exposed to acute or chronic hyperoxia. Thus, our findings demonstrate that NRF2 activation and antioxidant gene expression are functional during hyperoxia-induced lung epithelial cell death and that chronic hyperoxia does not impair NRF2 signaling overall.


Asunto(s)
Antioxidantes/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Hiperoxia/metabolismo , Hiperoxia/patología , Pulmón/patología , Factor 2 Relacionado con NF-E2/metabolismo , Muerte Celular/genética , Núcleo Celular/metabolismo , Supervivencia Celular/genética , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/genética , Humanos , NAD(P)H Deshidrogenasa (Quinona)/genética , Regiones Promotoras Genéticas/genética , Unión Proteica/genética
15.
J Interferon Cytokine Res ; 33(4): 189-98, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23570385

RESUMEN

The interferon (IFN) family of cytokines regulates many cellular processes, such as transcription, translation, post-translational modifications, and protein degradation. IFNs induce growth inhibition and/or cell death, depending on the cell type, by employing different proteins. This review describes a novel growth-suppressive pathway employed by IFNs that affects rRNA levels. Maturation of rRNA involves numerous noncoding small regulatory RNA-guided processes. These regulatory RNAs, called small nucleolar RNA (snoRNAs), function as a ribonucleoprotein particle (RNP) in the nucleolus. The biogenesis of snoRNPs is dependent on core protein and assembly factors. Our laboratory recently isolated a growth-suppressive protein gene associated with retinoid-IFN-induced mortality (GRIM)-1 using a genetic screen. IFN-inducible GRIM-1 (SHQ1) is an assembly factor that controls one arm of the snoRNP machinery. GRIM-1 inhibits sno/scaRNP formation to induce growth suppression via reduction in mature rRNA levels. Loss of GRIM-1 observed in certain cancers implicates it to be a novel tumor suppressor. Certain snoRNAs have been reported to act as either oncogenes or tumor suppressors in vitro. Recent studies have shown that certain sno/scaRNAs are further processed into micro RNA-like molecules to control translation of protein-coding RNAs. We present a model as to how these small regulatory RNAs influence cell growth and a potential role for GRIM-1 in this process.


Asunto(s)
Interferones/metabolismo , Neoplasias/metabolismo , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Portadoras/metabolismo , Humanos , Interferones/genética , Péptidos y Proteínas de Señalización Intracelular , Neoplasias/genética , Región Organizadora del Nucléolo/genética , Región Organizadora del Nucléolo/metabolismo , ARN Ribosómico/biosíntesis
16.
PLoS One ; 8(3): e57855, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23472114

RESUMEN

SerpinB2 or plasminogen activator inhibitor type 2 (PAI-2) is highly induced in macrophages in response to inflammatory stimuli and is linked to the modulation of innate immunity, macrophage survival, and inhibition of plasminogen activators. Lipopolysaccharide (LPS), a potent bacterial endotoxin, can induce SerpinB2 expression via the toll-like receptor 4 (TLR4) by ∼1000-fold over a period of 24 hrs in murine macrophages. To map the LPS-regulated SerpinB2 promoter regions, we transfected reporter constructs driven by the ∼5 kb 5'-flanking region of the murine SerpinB2 gene and several deletion mutants into murine macrophages. In addition, we compared the DNA sequence of the murine 5' flanking sequence with the sequence of the human gene for homologous functional regulatory elements and identified several regulatory cis-acting elements in the human SERPINB2 promoter conserved in the mouse. Mutation analyses revealed that a CCAAT enhancer binding (C/EBP) element, a cyclic AMP response element (CRE) and two activator protein 1 (AP-1) response elements in the murine SerpinB2 proximal promoter are essential for optimal LPS-inducibility. Electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays demonstrated that LPS induces the formation of C/EBP-ß containing complexes with the SerpinB2 promoter. Importantly, both constitutive and LPS-induced SerpinB2 expression was severely abrogated in C/EBP-ß-null mouse embryonic fibroblasts (MEFs) and primary C/EBP-ß-deficient peritoneal macrophages. Together, these data provide new insight into C/EBP-ß-dependent regulation of inflammation-associated SerpinB2 expression.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica , Lipopolisacáridos/metabolismo , Inhibidor 2 de Activador Plasminogénico/metabolismo , Animales , Secuencia de Bases , Línea Celular , Secuencia Conservada , Análisis Mutacional de ADN , Fibroblastos/citología , Humanos , Macrófagos/metabolismo , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas
17.
Proc Natl Acad Sci U S A ; 110(13): 5052-7, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23479624

RESUMEN

Cancer metastasis and immune suppression are critical issues in cancer therapy. Here, we show that a ß-galactoside-binding lectin [galectin-3 (gal3)] that recognizes the Thomsen-Friedenreich disaccharide (TFD, Galß1,3GalNAc) present on the surface of most cancer cells is involved in promoting angiogenesis, tumor-endothelial cell adhesion, and metastasis of prostate cancer cells, as well as evading immune surveillance through killing of activated T cells. To block gal3-mediated interactions, we purified a glycopeptide from cod (designated TFD100) that binds gal3 with picomolar affinity. TFD100 blocks gal3-mediated angiogenesis, tumor-endothelial cell interactions, and metastasis of prostate cancer cells in mice at nanomolar levels. Moreover, apoptosis of activated T cells induced by either recombinant gal3 or prostate cancer patient serum-associated gal3 was inhibited at nanomolar concentration of TFD100. Because the gal3-TFD interaction is a key factor driving metastasis in most epithelial cancers, this high-affinity TFD100 should be a promising antimetastatic agent for the treatment of various cancers, including prostate adenocarcinoma.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Proteínas Anticongelantes/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas de Peces/farmacología , Gadus morhua , Galectina 3/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Linfocitos T/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Proteínas Anticongelantes/química , Proteínas Anticongelantes/aislamiento & purificación , Antígenos de Carbohidratos Asociados a Tumores/química , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Proteínas de Peces/química , Proteínas de Peces/aislamiento & purificación , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Jurkat , Masculino , Ratones , Metástasis de la Neoplasia , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
18.
J Cancer Res Clin Oncol ; 139(6): 971-80, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23463096

RESUMEN

OBJECTIVES: To investigate the therapeutic utility of an attenuated bacterium carrying a plasmid that co-expresses Endostatin, an inhibitor of tumor neovasculogenesis, and a shRNA that targets Stat3 to suppress prostate cancer growth. METHODS: Plasmid pEndo-Si-Stat3 was constructed and introduced into an attenuated strain of Salmonella enterica serovar typhimurium. The resultant recombinant bacterium was used as a vector to deliver the plasmid to tumor cells growing in vivo. Tumor-associated gene and protein expression changes were measured by using RT-PCR and Western blot analyses. Expression of Endostatin in tumor tissue was detected by ELISA. The presence of vector bacteria in tissues was monitored and tumor destruction was assessed by using TUNEL and H&E staining assays. RESULTS: Bacterially delivered pEndo-Si-Stat3 decreased Stat3 levels and increased Endostatin expression in mouse tumors, resulting in a significant suppression of tumor growth (P < 0.01). Expression of Bcl-2 and PCNA was down-regulated and Caspase3 expression was up-regulated to promote apoptosis of tumor cells. CONCLUSIONS: Successful delivery by attenuated Salmonella of the combination therapeutic plasmid simultaneously knocked down the expression of Stat3 and resulted in over-expression of Endostatin, which synergistically inhibited prostate cancer growth.


Asunto(s)
Endostatinas/genética , Técnicas de Transferencia de Gen , Neoplasias de la Próstata/terapia , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/genética , Animales , Antígenos CD34/metabolismo , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Terapia Combinada , Endostatinas/metabolismo , Expresión Génica , Terapia Genética , Vectores Genéticos/genética , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Plásmidos/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Salmonella typhimurium/genética , Factores de Tiempo , Carga Tumoral/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
J Biol Chem ; 288(11): 7930-7941, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23386605

RESUMEN

The signal transducer and activator of transcription 3 (STAT3) protein is critical for multiple cytokine and growth factor-induced biological responses in vivo. Its transcriptional activity is controlled by a transient phosphorylation of a critical tyrosine. Constitutive activation of STAT3 imparts resistance to apoptosis, promotes cell proliferation, and induces de novo micro-angiogenesis, three of the six cardinal hallmarks of a typical cancer cell. Earlier we reported the isolation of GRIM-19 as a growth suppressor using a genome-wide expression knockdown strategy. GRIM-19 binds to STAT3 and suppresses its transcriptional activity. To understand the pathological relevance of GRIM-19, we screened a set of primary head and neck tumors and identified three somatic mutations in GRIM-19. Wild-type GRIM-19 suppressed cellular transformation by a constitutively active form of STAT3, whereas tumor-derived mutants L71P, L91P and A95T significantly lost their ability to associate with STAT3, block gene expression, and suppress cellular transformation and tumor growth in vivo. Additionally, these mutants lost their capacity to prevent metastasis. These mutations define a mechanism by which STAT3 activity is deregulated in certain human head and neck tumors.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Regulación Neoplásica de la Expresión Génica , Chaperonas Moleculares/metabolismo , Mutación , NADH NADPH Oxidorreductasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Neoplasias de la Boca/metabolismo , Metástasis de la Neoplasia , Ratas , Transcripción Genética
20.
Cancer Lett ; 335(1): 242-50, 2013 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23435374

RESUMEN

The E6 protein of the oncogenic HPV-16 functions by interfering with the normal cell cycle control mechanisms, particularly those controlled by p53. In this study, we developed a dual expression plasmid that coexpressed-E6-specific siRNA and wild type p53, and to evaluate its effects on cervical cancer growth. We found that simultaneous expression of pSi-E6-P53 caused a robust suppression of tumor growth when compared to the controls either E6-specific siRNA or p53 alone. In conclusion, our findings demonstrate that a combined strategy of co-expressed E6-specific siRNA and p53 synergistically and more effectively suppressed cervical tumor growth when compared with single treatment.


Asunto(s)
Proliferación Celular , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/terapia , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/terapia , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Terapia Genética/métodos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología , Plásmidos/genética , Proteínas Represoras/metabolismo , Salmonella typhimurium/genética , Transformación Bacteriana , Carga Tumoral , Proteína p53 Supresora de Tumor/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA