Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Transl Med ; 14: 80, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-27008379

RESUMEN

BACKGROUND: S100A13 and high mobility group A (HMGA1) are known to play essential roles in the carcinogenesis and progression of cancer. However, the correlation between S100A13 and HMGA1 during cancer progression is not yet well understood. In this study, we determined the effects of S100A13 on HMGA1 expression in thyroid cancer cells and examined the role of HMGA1 in thyroid cancer progression. METHODS: Stable ectopic S100A13 expression TT cellular proliferation was evaluated by nude mice xenografts assays. The effect of lentivirus-mediated S100A13 knockdown on thyroid cancer cellular oncogenic properties were evaluated by MTT, colony formation assays and transwell assays in TPC1 and SW579 cells. The effect of siRNA-mediated HMGA1 knockdown on thyroid cancer cellular proliferation and invasion were evaluated by MTT, colony formation assays and transwell assays. The tissue microarray was performed to investigate the correlation between S100A13 and HMGA1 expression in tumor tissues. RESULTS: The ectopic expression of S100A13 could increase tumor growth in a TT cell xenograft mouse model. Moreover, lentivirus-mediated S100A13 knockdown led to the inhibition of cellular oncogenic properties in thyroid cancer cells, and HMGA1 was found to be involved in the effect of S100A13 on thyroid cancer growth and invasion. Furthermore, siRNA-mediated HMGA1 knockdown was proved to inhibit the growth of TPC1 cells and invasive abilities of SW579 cells. Clinically, it was revealed that both S100A13 and HMGA1 showed a higher expression levels in thyroid cancer cases compared with those in matched normal thyroid cases (P = 0.007 and P = 0.000); S100A13 and HMGA1 expressions were identified to be positively correlated (P = 0.004, R = 0.316) when analyzed regardless of thyroid cancer types. CONCLUSIONS: This is the first report for the association between HMGA1 and S100A13 expression in the modulation of thyroid cancer growth and invasion. Those results would provide an essential insight into the effect of S100A13 on carcinogenesis of thyroid tumor, rending S100A13 to be potential biological marker for the diagnosis of thyroid cancer.


Asunto(s)
Proteína HMGA1a/metabolismo , Proteínas S100/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Lentivirus/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Fluorescente , Invasividad Neoplásica , ARN Interferente Pequeño/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Eur J Pharmacol ; 960: 176122, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-37863414

RESUMEN

Ulcerative colitis is an inflammatory bowel disease with increasing prevalence and incidence. Current treatments for ulcerative colitis are not generally applicative and are often accompanied by side effects. IGF2 is an endogenous protein that plays roles in anti-inflammation and stemness maintenance, but little is known about its mechanism and function in the progression of ulcerative colitis. In this study, mouse recombinant IGF2 was used in a mouse model of ulcerative colitis established by DSS. IGF2 expression was reduced in colon tissues but not plasma of DSS-induced colitis mice. IGF2R expression was also decreased in colitis colons, which was then elevated by recombinant IGF2. Recombinant IGF2 alleviated colon injury in colitis, which was evaluated by colon shortening, body weight loss and DAI score. IGF2 treatment also relieved the inflammatory response in colitis, which was assessed by the spleen weight index, MPO activity and proinflammatory cytokine expression and was also detected in LPS-stimulated RAW264.7 cells in vitro. Moreover, IGF2R was predicted and further verified to interact with the Sting protein, and the cGAS-Sting pathway as a key pathway for stemness regulation, was upregulated in colonic colons, which was blocked by IGF2 treatment. Additionally, IGF2 treatment can maintain colonic stemness and further repair colonic tight junction function in DSS-induced colitis. In conclusion, IGF2/IGF2R downregulated the cGAS-Sting pathway to sustain colonic stemness and barrier integrity to protect against ulcerative colitis induced by DSS.


Asunto(s)
Colitis Ulcerosa , Colitis , Ratones , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/metabolismo , Colon , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/metabolismo , Transducción de Señal , Nucleotidiltransferasas/metabolismo , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
3.
Biochem Biophys Res Commun ; 420(1): 17-23, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22390933

RESUMEN

It is well-known that sphingosine-1-phosphate (S1P), the phospholipid content of HDL, binding to S1P receptors can raise COX-2 expression and PGI(2) release through p38MAPK/CREB pathway. In the present study we assess the action of SR-B1 initiated PI3K-Akt-eNOS signaling in the regulation of COX-2 expression and PGI(2) production in response to HDL. We found that apoA1 could increase PGI(2) release and COX-2 expression in ECV 304 endothelial cells. Furthermore, SR-B1 was found to be involved in HDL induced up-regulation of COX-2 and PGI(2). Over-expressed SR-B1 did not significantly increase the expression of COX-2 and the PGI(2) levels, but knock-down of SR-B1 by siRNA could significantly attenuate COX-2 expression and PGI(2) release together with p38MAPK and CREB phosphorylation. Consistently, the declines of p-p38MAPK, p-CREB, COX-2 and PGI(2) were also observed after incubation with LY294002 (25µmol/L; PI3K special inhibitor) or L-NAME (50µmol/L; eNOS special inhibitor). In addition, we demonstrated the increases of PGI(2) release, COX-2 expression and p38MAPK phosphorylation, when nitric oxide level was raised through the incubation of L-arginine (10 or 20nmol/L) in endothelial cells. Taking together, our data support that SR-B1 mediated PI3K-Akt-eNOS signaling was involved in HDL-induced COX-2 expression and PGI(2) release in endothelial cells.


Asunto(s)
Células Endoteliales/metabolismo , Epoprostenol/biosíntesis , Lipoproteínas HDL/metabolismo , Receptores Depuradores de Clase B/metabolismo , Apolipoproteína A-I/metabolismo , Apolipoproteína A-I/farmacología , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclooxigenasa 2/biosíntesis , Células Endoteliales/efectos de los fármacos , Humanos , Lipoproteínas HDL/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Depuradores de Clase B/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Acta Pharmacol Sin ; 31(10): 1336-42, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20835266

RESUMEN

AIM: To explore the mechanisms involved in ox-LDL transcytosis across endothelial cells and the role of caveolae in this process. METHODS: An in vitro model was established to investigate the passage of oxidized low density lipoprotein (ox-LDL) through a tight monolayer of human umbilical vein endothelial cells (HUVEC) cultured on a collagen-coated filter. Passage of DiI-labeled ox-LDL through the monolayer was measured using a fluorescence spectrophotometer. The uptake and efflux of ox-LDL by HUVEC were determined using fluorescence microscopy and HPLC. RESULTS: Caveolae inhibitors - carrageenan (250 µg/mL), filipin (5 µg/mL), and nocodazole (33 µmol/L)-decreased the transport of ox-LDL across the monolayer by 48.9%, 72.4%, and 79.8% as compared to the control group. In addition, they effectively decreased ox-LDL uptake and inhibited the efflux of ox-LDL. Caveolin-1 and LOX-1 were up-regulated by ox-LDL in a time-dependent manner and decreased gradually after depletion of ox-LDL (P<0.05). After treatment HUVEC with ox-LDL and silencing caveolin-1, NF-κB translocation to the nucleus was blocked and LOX-1 expression decreased (P<0.05). CONCLUSION: Caveolae can be a carrier for ox-LDL and may be involved in the uptake and transcytosis of ox-LDL by HUVEC.


Asunto(s)
Caveolas/metabolismo , Caveolina 1/metabolismo , Endocitosis , Células Endoteliales/metabolismo , Lipoproteínas LDL/metabolismo , Venas Umbilicales/metabolismo , Células Cultivadas , Cromatografía Líquida de Alta Presión , Humanos , Microscopía Fluorescente , Receptores Depuradores de Clase E/biosíntesis , Espectrometría de Fluorescencia , Transcitosis
5.
Artículo en Inglés | MEDLINE | ID: mdl-31678514

RESUMEN

Atherosclerosis (AS) is characterized by lipids metabolism disorder and inflammatory response. Accumulating evidence has demonstrated that Wingless type 5a (Wnt5a) is implicated in cardiovascular diseases through non-canonical Wnt cascades. However, its precise role during the pathogenesis of AS is still unclear. Therefore, the present study aims to investigate the role and the underlying mechanism of Wnt5a/receptor tyrosine kinase-like orphan receptor 2 (Ror2) pathways in the promotion of AS process through affecting lipid accumulation and inflammation. In atherosclerotic clinical samples, Wnt5a levels were measured by using enzyme-linked immunosorbent assay (ELISA) assay. In vivo experiments were conducted by using apolipoprotein E knockout (apoE-/-) mice model. Vascular smooth muscle cells (VSMCs) were applied for in vitro studies. Wnt5a was highly expressed in both of atherosclerotic clinical samples and apoE-/- mice. The knockdown of Wnt5a significantly inhibited cholesterol accumulation and inflammatory response. Additionally, the lipopolysaccharide (LPS)-induced inflammation aggravated the cholesterol accumulation and decreased adenosine triphosphate (ATP)-binding cassette transporter A1 (ABCA1) expression in VSMCs. Depletion of intracellular cholesterol by ß-cyclodextrin (ß-CD) led to the upregulation of ABCA1 and the inhibition of inflammation. Conversely, the overexpression of Wnt5a inhibited ABCA1 expression, facilitated cholesterol accumulation, impared cholesterol efflux, promoted NF-κB nuclear translocation and the inflammatory cytokines secretion. Moreover, the knockdown of Ror2 increased ABCA1 expression and reduced Wnt5a-induced cholesterol accumulation and inflammatory responses. Furthermore, the knockdown of ABCA1 enhanced cholesterol accumulation and inflammatory response. Therefore, Wnt5a/Ror2 pathway was critical in regulating cholesterol homeostasis and inflammatory response, which might be a promising therapeutic target for AS therapy.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Inflamación/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Proteína Wnt-5a/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Aterosclerosis/sangre , Aterosclerosis/inmunología , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/sangre , Inflamación/inmunología , Masculino , Ratones , Ratones Noqueados para ApoE , Músculo Liso Vascular/citología , Miocitos del Músculo Liso , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/sangre , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Transducción de Señal/inmunología , Proteína Wnt-5a/sangre , Proteína Wnt-5a/genética
6.
J Breast Cancer ; 22(1): 15-28, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30941230

RESUMEN

PURPOSE: Pokemon, also known as ZBTB7A, belongs to the POZ and Krüppel (POK) family of transcription repressors and is implicated in tumor progression as a key proto-oncogene. This present study aimed at determining the mechanism by which Pokemon inhibits transforming growth factor ß (TGFß)-Smad4 pathway-dependent proliferation arrest of breast cancer cells via specificity protein 1 (SP1). METHODS: Over-expressing plasmid or small interfering RNA (siRNA) transfection was used to regulate Pokemon levels. The EdU incorporation assay, MTS assay, and clone formation were used to identify the inhibitory effect of Pokemon siRNA on cell proliferation. Quantitative real-time polymerase chain reaction assay confirmed that Pokemon deletion inhibited the expression of proliferation-associated genes. The dual-luciferase reporter assay, electrophoretic mobility shift assay, and co-immunoprecipitation assay were used to analyze binding between Pokemon, Smad4, and SP1. RESULTS: Pokemon deletion induced proliferation arrest of breast cancer cells and inhibited the expression of proliferation-associated genes, especially Smad4. Pokemon bound with SP1 to interdict Smad4 promoter activity. Information on clinical samples was obtained from The Cancer Genome Atlas data, in which the Pokemon mRNA levels showed a negative correlation with Smad4 levels in different subtypes of breast cancer in two independent datasets. CONCLUSION: We demonstrated that Pokemon binds to SP1 to down-regulate Smad4 expression, thereby promoting proliferation of breast cancer cells. This suggests that Pokemon is a potential TGFß-signaling participant in breast cancer progression.

7.
Cardiovasc Drugs Ther ; 22(6): 453-60, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18665442

RESUMEN

AIM: To investigate the effects of the sensitizer rosiglitazone on the proliferation of vascular smooth muscle cell (VSMC) induced by high glucose administration. METHODS: VSMCs were isolated from rat thoracic aortas and cultured in 10% fetal bovine serum (FBS). VSMC proliferation was evaluated by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay and cell counting. The cell cycle was examined by flow cytometry. The protein expressions of proliferating cell nuclear antigen (PCNA) and matrix metalloproteinases-2 (MMP-2) were evaluated by Western blotting. MMP-2 mRNA expression was analyzed by reverse transcription-polymerase chain reaction (RT-PCR) and gelatinolytic activity was determined by zymography. RESULTS: Promoted VSMC proliferation significantly increased the number of VSMCs in the S phase, the expressions of PCNA and MMP-2, and MMP-2 activity, as well as decreased the proportion of VSMCs in the G(0)/G(1) phase. Rosiglitazone at a concentration of 10 mumol/L markedly inhibited glucose-induced VSMC proliferation (1.869 +/- 0.22 vs 0.820 +/- 0.15, P < 0.01). Concomitantly, rosiglitazone inhibited PCNA expression (0.96 +/- 0.07 vs 0.75 +/- 0.06, P < 0.05) and cell cycle progression from G(0)/G(1) to S phase (the proportion of VSMCs in the G(0)/G(1) and S phase were 69.6 +/- 3.96% vs 84.3 +/- 1.73% and 25.2 +/- 1.73% vs 10.1 +/- 1.42% (P < 0.01), respectively). Furthermore, rosiglitazone significantly decreased MMP-2 mRNA expression (0.98 +/- 0.08 vs 0.71 +/- 0.05, P < 0.05), protein expression (0.80 +/- 0.04 vs 0.64 +/- 0.03, P < 0.05) and MMP-2 activity (320 +/- 25% vs 248 +/- 21%, P < 0.05). CONCLUSION: Rosiglitazone significantly inhibited VSMC proliferation, at least in part by inhibiting high glucose-induced G(1)-->S phase transition, PCNA expression and MMP-2 synthesis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glucosa/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Tiazolidinedionas/farmacología , Animales , Aorta Torácica/citología , Bovinos , Recuento de Células , Células Cultivadas , Dimetilsulfóxido/química , Relación Dosis-Respuesta a Droga , Citometría de Flujo/métodos , Glucosa/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Masculino , Manosa/farmacología , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rosiglitazona , Fase S/efectos de los fármacos , Piel/efectos de los fármacos , Piel/lesiones , Sales de Tetrazolio , Tiazoles , Azul de Tripano , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
8.
Acta Pharmacol Sin ; 29(10): 1233-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18817629

RESUMEN

AIM: To investigate the effect of diallyl disulfide (DADS), a component of garlic, on apoptosis in human mammary cancer cell line (MCF-7) and its mechanisms. METHODS: Cytotoxicity was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide assays. Morphology of apoptotic cells was detected by acridine orange and ethidium bromide staining. Apoptotic cells stained with propidium iodide were examined using flow cytometry. Protein levels were detected by Western blot analysis. RESULTS: DADS inhibited the proliferation of MCF-7 cells and induced the apoptotic ratio to increase rapidly. Cleavage of the caspase-3 and caspase-3 substrate poly(ADP-ribose) polymerase was observed in MCF-7 cells after 24 h of treatment with DADS. When the MCF-7 cells were treated with 200 micromol x L DADS, the stress-activated protein kinase extracellular signal-regulated kinase (ERK), a mitogen-activated protein kinase, was inhibited after 6 h; c-Jun N-terminal kinase (JNK), that is stress-activated protein kinase (SAPK), and p38 mitogen-activated protein kinase were activated after 6 h. CONCLUSION: These results suggest that DADS both inhibits the proliferation of MCF-7 cells and induces apoptosis of MCF-7 cells. The mechanisms may include the inhibition of ERK and the activation of the SAPK/JNK and p38 pathways.


Asunto(s)
Compuestos Alílicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Sulfuros/farmacología , Caspasa 3/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Femenino , Humanos , MAP Quinasa Quinasa 4/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Oncol Rep ; 38(2): 1303-1311, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28677794

RESUMEN

Our previous study reported several alternative splicing variants of arginine N-methyltransferase 2 (PRMT2), which lose different exons in the C-terminals of the wild-type PRMT2 gene. Particularly, due to frame-shifting, PRMT2ß encodes a novel amino acid sequence at the C-terminus of the protein, the function of which is not understood. In the present study, we determined the role of PRMT2ß in breast cancer cell proliferation, apoptosis and its effect on the Akt signaling pathway. Stable breast cancer MCF7 cell line with lentivirus-mediated PRMT2ß overexpression was obtained after selection by puromycin for 2 weeks. The effect of lentivirus-mediated PRMT2ß overexpression on breast cancer cellular oncogenic properties was evaluated by MTT, colony formation, cell cycle analysis and apoptosis assays in MCF7 cells. Luciferase activity assay and western blot analysis were performed to characterize the effects of PRMT2ß on cyclin D1 promoter activities and the Akt signaling pathway. Tissue microarray was performed to investigate the association of PRMT2ß with breast cancer progression. Lentivirus-mediated PRMT2ß overexpression suppressed the cell proliferation and colony formation of breast cancer MCF7 cells. PRMT2ß overexpression induced cell cycle arrest and apoptosis of MCF7 cells. Furthermore, PRMT2ß was revealed to suppress the transcription activity of the cyclin D1 promoter, and PRMT2ß was also found to inhibit cyclin D1 expression via the suppression of Akt/GSK-3ß signaling in breast cancer cells. Clinically, it was revealed that PRMT2ß expression was negatively correlated with human epidermal growth factor receptor 2 (HER2) (p=0.033) in breast tumors. Our results revealed that PRMT2ß, a novel splice variant of PRMT2, plays potential antitumor effect by suppressing cyclin D1 expression and inhibiting Akt signaling activity. This also opens a new avenue for treating breast cancer.


Asunto(s)
Empalme Alternativo , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína-Arginina N-Metiltransferasas/genética , Apoptosis , Neoplasias de la Mama/metabolismo , Estudios de Casos y Controles , Proliferación Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Pronóstico , Isoformas de Proteínas , Proteína-Arginina N-Metiltransferasas/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Células Tumorales Cultivadas
10.
Int J Oncol ; 50(5): 1567-1578, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28393241

RESUMEN

The role of transforming growth factor-ß1 (TGF-ß1) is complicated and plays a different role in the development of cancer. High mobility group A (HMGA1) participates in multiple cellular biology processes, and exerts important roles in the epithelial-mesenchymal transition (EMT). However, the correlation of TGF-ß1 and HMGA1 in cancer cells is not yet fully understood. In this study, we determined the effects of TGF-ß1 on HMGA1 expression in thyroid cancer cells and examined the role of HMGA1 in thyroid cancer progression. With real-time PCR and immunofluorescence staining, our study demonstrated that TGF-ß1 induced the expression of HMGA1 through phosphoinositide 3-kinase (PI3K) and the extracellular signal-related kinase (ERK) signaling in thyroid cancer cells. With luciferase reported assay, the HMGA1 promoter activity was activated by TGF-ß1 in the SW579 cells. Furthermore, lentivirus-mediated HMGA1 knockdown inhibits cellular oncogenic properties of thyroid cancer cells. Clinically, tissue microarray revealed that HMGA1 was expressed in thyroid carcinoma more than that in normal thyroid tissues (P<0.001); expression of HMGA1 and MMP-2 was identified to be positively correlated (P=0.017). The present study established the first link between HMGA1 and TGF-ß1 in the regulation of thyroid cancer proliferation and invasion, and provided evidence for the pivotal role of HMGA1 in the progression of thyroid cancer, indicating HMGA1 to be potential biological marker for the diagnosis of thyroid cancer.


Asunto(s)
Proteína HMGA1a/genética , Metaloproteinasa 2 de la Matriz/genética , Neoplasias de la Tiroides/genética , Factor de Crecimiento Transformador beta1/genética , Adulto , Anciano , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Proteína HMGA1a/biosíntesis , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Persona de Mediana Edad , Invasividad Neoplásica/genética , Neoplasias de la Tiroides/patología
11.
Asian Pac J Cancer Prev ; 13(10): 5201-5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23244135

RESUMEN

BACKGROUND: To evaluate the role of the X-ray repair cross complementing group 3 (XRCC3) T241M polymorphism in head and neck cancer susceptibility. MATERIALS AND METHODS: We performed a meta-analysis of all available studies, which included 3,191 cases and 5,090 controls. RESULTS: Overall, a significant risk effect of the T241M polymorphism was not found under homologous contrast (MM vs TT: OR=1.293, 95% CI=0.926-1.805; TM vs TT: OR=1.148 95% CI=0.930-1.418) and recessive models (MM vs TT+TM): OR=1.170, 95% CI=0.905- 1.512, but a significantly increased risk was observed under a dominant model (MM+TM vs TT): OR=1.243, 95% CI=1.001-1.544. In stratified analyses, there were no significant associations for Asians or Caucasians. CONCLUSION: Our meta-analysis suggested the XRCC3 241M allele (MM+TM) might act as a head and neck cancer risk factor among all subjects, and the effect of T241M polymorphism on head and neck susceptibility should be studied with a larger, stratified population.


Asunto(s)
Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad , Neoplasias de Cabeza y Cuello/etiología , Polimorfismo Genético/genética , Estudios de Casos y Controles , Humanos , Pronóstico , Factores de Riesgo
12.
Recent Pat Anticancer Drug Discov ; 7(2): 154-67, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22339355

RESUMEN

ATP citrate lyase (ACL or ACLY) is an extra-mitochondrial enzyme widely distributed in various human and animal tissues. ACL links glucose and lipid metabolism by catalyzing the formation of acetyl-CoA and oxaloacetate from citrate produced by glycolysis in the presence of ATP and CoA. ACL is aberrantly expressed in many immortalized cells and tumors, such as breast, liver, colon, lung and prostate cancers, and is correlated reversely with tumor stage and differentiation, serving as a negative prognostic marker. ACL is an upstream enzyme of the long chain fatty acid synthesis, providing acetyl-CoA as an essential component of the fatty acid synthesis. Therefore, ACL is a key enzyme of cellular lipogenesis and potent target for cancer therapy. As a hypolipidemic strategy of metabolic syndrome and cancer treatment, many small chemicals targeting ACL have been designed and developed. This review article provides an update for the research and development of ACL inhibitors with a focus on their patent status, offering a new insight into their potential application.


Asunto(s)
ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , Antineoplásicos/química , Diseño de Fármacos , Inhibidores Enzimáticos/química , Lipogénesis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , ATP Citrato (pro-S)-Liasa/química , ATP Citrato (pro-S)-Liasa/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ácido Cítrico/análogos & derivados , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ratones , Patentes como Asunto , Procesamiento Proteico-Postraduccional
13.
Eur J Pharmacol ; 676(1-3): 51-6, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22173123

RESUMEN

Numerous studies revealed that spinal inflammation and immune response play an important role in neuropathic pain. In this study, we investigated the effects of intrathecal injection of a Toll-like receptor (TLR4) inhibitor epigallocatechin gallate (EGCG) on neuropathic pain induced by chronic constriction injury of the sciatic nerve (CCI). A total of 120 rats were randomly assigned into 4 groups: sham-operated group, CCI group, CCI plus normal saline group and CCI plus EGCG group. CCI and sham surgeries were performed and both thermal hyperalgesia and mechanical allodynia were tested. Lumbar spinal cord was sampled and the mRNA and protein expressions of TLR4 and High Mobility Group 1 protein (HMGB1) were detected, the contents of tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1ß) and interleukin-10 (IL-10) were measured by ELISA, and immunohistochemistry for nuclear factor kappa B (NF-κB) was also carried out. When compared with the sham group, both mechanical and heat pain thresholds were significantly decreased, and the mRNA and protein expressions of TLR4 and HMGB1, the contents of TNF-α, IL-1ß and IL-10 in the spinal cords and NF-κB expression in the spinal dorsal horn were markedly increased in CCI rats (P<0.05). After intrathecal injection of EGCG (1mg/kg) once daily from 1day before to 3days after CCI surgery, the expressions of TLR4, NF-κB, HMGB1, TNF-α and IL-1ß were markedly decreased while the content of IL-10 in the spinal cord increased significantly accompanied by dramatical improvement of pain behaviors in CCI rats (P<0.05). These results show that the TLR4 signaling pathway plays an important role in the occurrence and development of neuropathic pain, and the therapy targeting TLR4 might be a novel strategy in the treatment of neuropathic pain.


Asunto(s)
Catequina/análogos & derivados , Inyecciones Espinales , Neuralgia/tratamiento farmacológico , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Catequina/administración & dosificación , Catequina/farmacología , Catequina/uso terapéutico , Enfermedad Crónica , Constricción , Regulación de la Expresión Génica/efectos de los fármacos , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Masculino , FN-kappa B/metabolismo , Neuralgia/genética , Neuralgia/metabolismo , Neuralgia/prevención & control , Umbral del Dolor/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Ciático/efectos de los fármacos , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Nervio Ciático/fisiopatología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
Cytotechnology ; 64(1): 43-51, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21938469

RESUMEN

Carvacrol is one of the members of monoterpene phenol and is present in the volatile oils of Thymus vulgaris, Carum copticum, origanum and oregano. It is a safe food additive commonly used in our daily life, and few studies have indicated that carvacrol has anti-hepatocarcinogenic activities. The rationale of the study was to examine whether carvacrol affects apoptosis of human hepatoma HepG2 cells. In this study, we showed that carvacrol inhibited HepG2 cell growth by inducing apoptosis as evidenced by Hoechst 33258 stain and Flow cytometric (FCM) analysis. Incubation of HepG2 cells with carvacrol for 24 h induced apoptosis by the activation of caspase-3, cleavage of PARP and decreased Bcl-2 gene expression. These results demonstrated that a significant fraction of carvacrol treated cells died by an apoptotic pathway in HepG2 cells. Moreover, carvacrol selectively altered the phosphorylation state of members of the MAPK superfamily, decreasing phosphorylation of ERK1/2 significantly in a dose-dependent manner, and activated phosphorylation of p38 but not affecting JNK MAPK phosphorylation. These results suggest that carvacrol may induce apoptosis by direct activation of the mitochondrial pathway, and the mitogen-activated protein kinase pathway may play an important role in the antitumor effect of carvacrol. These results have identified, for the first time, the biological activity of carvacrol in HepG2 cells and should lead to further development of carvacrol for liver disease therapy.

15.
FEBS J ; 279(2): 316-35, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22093364

RESUMEN

Protein N-arginine methyltransferases (PRMTs) participate in a number of cellular processes, including cell growth, nuclear/cytoplasmic protein shuttling, differentiation, RNA splicing and post-transcriptional regulation. PRMT2 (also known as HRMT1L1) is clearly involved in lung function, the inflammatory response, apoptosis promotion, Wnt signaling and leptin signaling regulation through different mechanisms. In this study, we report the molecular and cell biological characterization of three novel PRMT2 splice variants isolated from breast cancer cells and referred to as PRMT2α, PRMT2ß and PRMT2γ. Compared with the wild-type PRMT2, these variants lack different motifs and therefore generate distinct C-terminal domains. Confocal microscopy scanning revealed a distinct intracellular localization of PRMT2 variants, suggesting that the alternatively spliced C-terminus of PRMT2 can directly influence its subcellular localization. Our findings reveal that these variants are capable of binding to estrogen receptor alpha (ERα) both in vitro and in vivo, and the N-terminal regions of these variants contribute to ERα-PRMT2 interactions. Furthermore, these variants were proved to be able to enhance ERα-mediated transactivation activity. Luciferase reporter assays showed that PRMT2s could modulate promoter activities of the ERα-targeted genes of Snail and E-cadherin. In addition, PRMT2 silencing could enhance 17ß-estradiol-induced proliferation by regulating E2F1 expression and E2F1-responsive genes in ERα-positive breast cancer cells. Real-time PCR and immunohistochemistry showed that overall PRMT2 expression was upregulated in breast cancer tissues and significantly associated with ERα positivity status both in breast cancer cell lines and breast cancer tissues. We speculate that PRMT2 and its splice variants may directly modulate ERα signaling and play a role in the progression of breast cancer.


Asunto(s)
Empalme Alternativo , Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Neoplasias de la Mama/patología , Carcinoma/patología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genes Reporteros , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Activación Transcripcional
16.
Gene ; 487(1): 1-9, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21820040

RESUMEN

The arginine N-methyltransferase 2 protein (PRMT2, also known as HRMT1L1) is thought to act as a coactivator of ERα. The present results show the occurrence of a novel transcript by alternative polyadenylation in the human PRMT2 gene. We demonstrated that the newly identified intron-retaining PRMT2L2 transcript is functionally intact, efficiently translated into protein in vivo. PRMT2 and PRMT2L2 mRNA expression profiles overlap with the distribution of ERα, with the strongest abundance in estrogen target tissues. Transient co-transfection assays demonstrated that PRMT2L2 enhance ERα-mediated transactivation activity of ERE-Luc in a ligand-dependent manner. Confocal microscopy scanning revealed a distinct intra-cellular localization of their fusion proteins, suggesting that the C-terminal region absent in PRMT2L2 is critical for the localization. Statistical analysis further showed that both PRMT2 and PRMT2L2 mRNAexpressions were up-regulated in breast cancer tissues, and significantly associated with ERα positivity status. Thus, post-transcriptional processing mechanism as alternative polyadenylation and splicing may play a crucial role in regulating human PRMT2 gene expression.


Asunto(s)
Empalme Alternativo , Neoplasias de la Mama/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Poliadenilación/genética , Proteína-Arginina N-Metiltransferasas/genética , Secuencia de Aminoácidos , Secuencia de Bases , Northern Blotting , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isoenzimas/genética , Microscopía Confocal , Datos de Secuencia Molecular , Proteína-Arginina N-Metiltransferasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA