Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mediators Inflamm ; 2017: 5958429, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28676732

RESUMEN

There are increasing evidences of proinflammatory cytokine involvement in cancer development. Here, we found that two cytokines, IL-6 and TNF-α, activated colorectal cancer cells to be more invasive and stem-like. Combined treatment of IL-6 and TNF-α phosphorylated transcription factors STAT3 in a synergistic manner. STAT3, STAT1, and NF-κB physically interacted upon the cytokine stimulation. STAT3 was bound to the promoter region of human telomerase reverse transcriptase (hTERT). IL-6 and TNF-α stimulation further enhanced STAT3 binding affinity. Stem cell marker Oct-4 was upregulated in colorectal cancer cells upon IL-6 and TNF-α stimulation. Withaferin A, an anti-inflammatory steroidal lactone, inhibited the IL-6- and TNF-α-induced cancer cell invasion and decreased colonosphere formation. Notably, withaferin A inhibited STAT3 phosphorylation and abolished the STAT3, STAT1, and NF-κB interactions. Oct-4 expression was also downregulated by withaferin A inhibition. The binding of STAT3 to the hTERT promoter region and telomerase activity showed reduction with withaferin A treatments. Proinflammatory cytokine-induced cancer cell invasiveness is mediated by a STAT3-regulated mechanism in colorectal cancer cells. Our data suggest that withaferin A could be a promising anticancer agent that effectively inhibits the progression of colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Interleucina-6/farmacología , FN-kappa B/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Telomerasa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Witanólidos/farmacología , Western Blotting , Inmunoprecipitación de Cromatina , Células HT29 , Humanos , Inmunoprecipitación , FN-kappa B/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Telomerasa/genética
2.
Cell Biol Int ; 38(4): 531-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24375948

RESUMEN

Human amniotic fluid stem cells (HAFSCs) have a high proliferative capacity and a good differentiation potential, and may thus be suitable for regenerative medicine. To date, urothelial differentiation mechanisms of HAFSCs are poorly understood. We have investigated the urothelial differentiation potential of HAFSCs so that they can be therapeutically applied to cure defective diseases of bladder. To induce the stem cell differentiation, HAFSCs were cultured in a bladder cancer-derived conditioned medium. After 2 weeks of culture, HAFSCs began to express the urothelial lineage-specific markers (UPII, CK8 and FGF10). Meanwhile, pluripotency markers (Oct-4, Sox-2 and Nanog) were downregulated at both RNA and protein levels in the differentiated HAFSCs. Immunocytochemistry data revealed that differentiated HAFSCs expressed urothelial markers of UPII and CK8. We have screened the receptor tyrosine kinase arrays with the differentiated HAFSCs. The screening showed that MuSK, Tie-1 and EphA4 receptor tyrosine kinases were upregulated, whereas EphA7 and FGF R1 kinases were downregulated in HAFSCs. The data suggest that HAFSCs can be an important urothelium cell source, which can be used for urinary tract engineering.


Asunto(s)
Líquido Amniótico/citología , Medios de Cultivo Condicionados/farmacología , Células Madre/efectos de los fármacos , Urotelio/citología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Queratina-8/metabolismo , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Células Madre/citología , Células Madre/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Uroplaquina II/metabolismo , Urotelio/metabolismo
3.
Oncotarget ; 10(44): 4516-4531, 2019 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-31360301

RESUMEN

Curcumin is a biologically active polyphenol that exists in Indian spice turmeric. It has been reported that curcumin exerted anti-inflammatory, anti-oxidant and anti-cancer effects in numerous in vitro and in vivo studies. However, it is not well-understood the molecular mechanism of curcumin for the cancer stem cells and telomerase in colorectal cancer. In this study, compound 19, a nitrogen-containing curcumin analog, was used to treat human colorectal cancer cells. Compound 19 showed a greater anti-proliferative activity than curcumin while displayed no significant toxicity toward normal human colon epithelial cells. Compound 19 exerted anti-inflammatory activities by deactivating STAT3 and NF-κB. In cancer stem cell populations, CD44, Oct-4 and ALDHA1 expressions were abolished upon treating with compound 19. Cancer stem cell biomarkers CD51 and CD133 positive populations were reduced and telomerase activities were decreased with the reduced STAT3 binding to hTERT promoters. This means compound 19 dually inhibits canonical and non-canonical functions of telomerase. Furthermore, compound 19 treatments induced cell cycle arrest at G1 phase and apoptosis. Human apoptosis-related array screening revealed that activated caspase 3, catalase, clusterin and cytochrome C led to apoptosis. Taken together, our data suggest that compound 19 can be a novel therapeutic agent for metastatic colorectal cancer by concurrently targeting STAT3 and NF-κB signaling pathways.

4.
Oncotarget ; 9(68): 32943-32957, 2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-30250641

RESUMEN

Colorectal cancer is one of the leading causes for mortalities worldwide. The most common cause of colorectal cancer mortality is hepatic metastasis. There has been a limited advancement in the targeted-therapies for metastatic colorectal cancer. Conventional chemotherapeutic agent 5-fluorouracil has been used for various cancer treatments including colorectal cancer. Development of drug resistance and severe toxicity are major hurdles for its use in clinical setting. Resveratrol is a natural polyphenolic compound which has protective effects against aging-related diseases. In this study, we have tested whether combined treatments of resveratrol and 5-FU enhanced inhibitory effects against colorectal cancer cell growth. We herein showed that resveratrol and 5-FU combination treatments caused the anti-cancer activities by simultaneously inhibiting STAT3 and Akt signaling pathways. Resveratrol treatment induced S-phase specific cell cycle arrest, and when combined with 5-FU, it showed further increase in colorectal cancer cell apoptosis. Combined treatments of resveratrol and 5-FU inhibited epithelial-mesenchymal transition. Notably, resveratrol showed anti-inflammatory effects by downregulating inflammatory biomarkers, pSTAT3 and pNFκB. Resveratrol and 5-FU treatments inhibited STAT3 phosphorylation and its binding to the promoter region of human telomerase reverse transcriptase (hTERT). Our data provide the first evidence that resveratrol can enhance anti-telomeric and pro-apoptotic potentials of 5-FU in colorectal cancer, hence lead to re-sensitization to chemotherapy.

5.
Artículo en Inglés | MEDLINE | ID: mdl-29932172

RESUMEN

Cancer Drug resistance is a medical concern that requires extensive research and a thorough understanding in order to overcome. Remarkable achievements related to this field have been accomplished and further work is needed in order to optimize the cure for cancer and serve as the basis for precise medicine with few or no side effects.

6.
Anticancer Res ; 37(2): 445-453, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28179289

RESUMEN

BACKGROUND: Colorectal cancer is the third leading cause of cancer-related mortality in most developed countries. This mortality is mainly due to the metastatic progression to the liver with frequent recurrence. Colorectal cancer remains a therapeutic challenge and this has intensified the search for new drug targets. In an effort to establish a novel targeted-therapy, we studied the molecular mechanisms of cancer stem cell inhibitor salinomycin. MATERIALS AND METHODS: Co-immunoprecipitation was performed to examine STAT3-STAT1 protein interactions. Telomerase activity was measured by polymerase chain reaction (PCR) and ELISA assays. Apoptosis and cell stress arrays were analyzed to identify key proteins responding to salinomycin treatments. RESULTS: IL-6 and TNF-α induced STAT3 and STAT1 interactions, however the interactions were abolished by salinomycin challenge. Salinomycin reduced cancer stem cell phenotype and decreased telomerase activity of colorectal cancer cells. CONCLUSION: Our work uncovers a new mechanism through which salinomycin inhibits cancer stemness suggesting a novel targeted-therapy for metastatic colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Piranos/farmacología , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Telomerasa/antagonistas & inhibidores , Benzamidas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ensayo de Inmunoadsorción Enzimática , Transición Epitelial-Mesenquimal , Células HT29 , Humanos , Inmunoprecipitación , Interleucina-6/farmacología , Fosforilación , Reacción en Cadena de la Polimerasa , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Telomerasa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , beta Catenina/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis
7.
Int J Oncol ; 49(2): 487-98, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27279256

RESUMEN

Colorectal cancer (CRC) is one of the most commonly diagnosed cancers worldwide. The malignant CRC that undergoes metastasis in the advanced stage is usually refractory to existing chemotherapy and shows a poor prognosis. However, to date, efficient targeted-therapy for metastatic CRC is ill-defined. We tested the hypothesis that combined treatment of flavonoid morin and telomerase inhibitor MST­312 may reduce the cancer stem cell (CSC) traits. To characterize CSC phenotype, we performed the CD133/CD44 subpopulation profiling, tumorsphere formation assay, cell invasion assay and wound healing assay. We have examined the augmenting effects of the combined treatment of morin and MST­312 for 5-FU (5-fluorouracil) efficacy in human colorectal cancer. Morin and MST­312 combined treatment reduced CD133 (+) and CD44 (+) subpopulations in human colorectal and breast cancer cells, respectively. Tumorsphere formation and cell invasiveness were decreased with the morin and MST­312 combination treatment. Consistent with these data, morin and MST­312 treatment decreased the wound healing capacity of human breast cancer cells. Stress and apoptosis antibody arrays revealed that there were specific upregulated and downregulated proteins resulting from different treatments. Phosphorylation levels of BAD, p53 and Chk1 were enhanced upon morin/MST­312 treatments in HT-29 cells, whereas caspase-3 cleavage level and expression of IκBα were downregulated by combined morin/MST­312 treatment in SW620 cells. Finally, morin and MST­312 co-treatment further augmented the 5-FU efficacy, chemosensitizing the 5-FU resistant human colorectal cancer cells. Taken together, our study suggests that novel targeted-therapy can be implemented by using flavonoid morin and telomerase inhibitor MST­312 for improved cancer prognosis.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Benzamidas/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Flavonoides/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Telomerasa/antagonistas & inhibidores , Animales , Benzamidas/administración & dosificación , Línea Celular Tumoral , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Flavonoides/administración & dosificación , Fluorouracilo/administración & dosificación , Fluorouracilo/farmacología , Células HT29 , Humanos , Ratones , Células 3T3 NIH , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosforilación/efectos de los fármacos , Telomerasa/metabolismo
8.
Anticancer Res ; 35(1): 39-46, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25550533

RESUMEN

BACKGROUND/AIM: The cancer stem cell (CSC) model postulates the existence of a small proportion of cancer cells capable of sustaining tumor formation, self-renewal and differentiation. Signal Transducer and Activator of Transcription 3 (STAT3) signaling is known to be selectively activated in breast CSC populations. However, it is yet to be determined which molecular mechanisms regulate STAT3 signaling in CSCs and what chemopreventive agents are effective for suppressing CSC growth. The aim of this study was to examine the potential efficacy of curcumin and epigallocatechin gallate (EGCG) against CSC and to uncover the molecular mechanisms of their anticancer effects. MATERIALS AND METHODS: To suppress the CSC phenotype, two breast cancer cell lines (MDA-MB-231 cells and MCF7 cells transfected with HER2) were treated with curcumin (10 µM) with or without EGCG (10 µM) for 48 h. We used tumor-sphere formation and wound-healing assays to determine CSC phenotype. To quantify CSC populations, Fluorescence-activated cell sorting profiling was monitored. STAT3 phosphorylation and interaction with Nuclear Factor-kB (NFkB) were analyzed by performing western blot and immunoprecipitation assays. RESULTS: Combined curcumin and EGCG treatment reduced the cancer stem-like Cluster of differentiation 44 (CD44)-positive cell population. Western blot and immunoprecipitation analyses revealed that curcumin and EGCG specifically inhibited STAT3 phosphorylation and STAT3-NFkB interaction was retained. CONCLUSION: This study suggests that curcumin and EGCG function as antitumor agents for suppressing breast CSCs. STAT3 and NFκB signaling pathways could serve as targets for reducing CSCs leading to novel targeted-therapy for treating breast cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Catequina/análogos & derivados , Curcumina/farmacología , FN-kappa B/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Transcripción STAT3/metabolismo , Catequina/farmacología , Movimiento Celular , Regulación hacia Abajo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Receptores de Hialuranos/metabolismo , Células MCF-7 , Células Madre Neoplásicas/efectos de los fármacos , Fenotipo , Fosforilación , Procesamiento Proteico-Postraduccional , Transducción de Señal
9.
Int J Oncol ; 46(4): 1461-72, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25652255

RESUMEN

Hormone therapy targeting estrogen receptor α (ERα) is the most effective treatment for breast cancer. However, this treatment eventually fails as the tumor develops resistance. Although reduced expression of ER-α is a known contributing factor to endocrine resistance, the mechanism of ER-α downregulation in endocrine resistance is still not fully understood. The present study shows that Slug has an inverse relationship with ERα in breast and prostate cancer patient samples. Also the inhibition of Slug blocks mammary stem cell activity in primary mammary epithelial cells. We hypothesize that Slug may be a key transcription factor in the regulation of ERα expression. To understand the Slug-ERα signaling pathway, we employed resistant cell line MCF-TAMR (ERα relatively negative) derived from its parental MCF-7 (ERα positive) cell line and assessed changes in cell phenotype, activity and response to therapy. Conversely, we performed knockdown of Slug in the high-Slug expressing cell line MDA-MB-231 and assessed reversal of the mesenchymal phenotype. Microarray analysis showed that Slug is overexpressed in high grade breast and prostate cancer tissues. Additionally, Slug overexpression leads to drug resistance. Furthermore, we demonstrated that Slug binds directly to ERα promoter E-boxes and represses ERα expression. This resulted in decrease in epithelial-to-mesenchymal transition in cancer cells. These findings demonstrate that Slug, by regulation of ERα expression, contributes to tumor progression and could serve as an important target for cancer therapy.


Asunto(s)
Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/genética , Neoplasias de la Próstata/patología , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Células MCF-7 , Masculino , Neoplasias Mamarias Experimentales , Ratones , Ratones Desnudos , Neoplasias de la Próstata/metabolismo , Factores de Transcripción de la Familia Snail , Tamoxifeno/farmacología
10.
Int J Oncol ; 44(2): 403-11, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24297508

RESUMEN

Clinically, HER2 proto-oncogene amplification is found in about 25-30% of human breast cancers, where it is correlated to a poor prognosis. Constitutive STAT3 activation is found in about 50-60% of the breast tumors and associated with tumorigenesis and drug resistance. In this study, we showed that STAT3 was phosphorylated in HER2-overexpressing, ER-positive human breast tumors and, furthermore, phosphorylated STAT3 promoted the stem-like cell phenotype. We examined the dysregulation of the stem cell markers (Oct-4, Sox-2 and CD44) and the tumorsphere formation in HER2-overexpressing human breast cancer cell lines. We demonstrated that the STAT3 inhibitor, Stattic, treatment abolished the cancer stem cell phenotype in HER2-positive breast cancers. Combined treatment of Herceptin and Stattic showed the synergistic effect on the cancer cell growth in vitro. In addition, when the STAT3 gene was knocked down, the expression of the stem cell markers Oct-4, Sox-2 and CD44 were downregulated and tumorsphere formation was abolished. HER2-elicited STAT3 signaling may provide a potential model for drug resistance induced by stem-like cell characteristics. This mechanism may be responsible for acquiring resistance to Herceptin in the treatment of HER2-overexpressing breast tumors. Based on our findings, targeting pSTAT3 could overcome Herceptin-induced resistance in HER2-overexpressing breast tumors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transición Epitelial-Mesenquimal , Células Madre Neoplásicas/patología , Receptor ErbB-2/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Biomarcadores de Tumor , Western Blotting , Neoplasias de la Mama/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Óxidos S-Cíclicos/farmacología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Femenino , Humanos , Inmunoprecipitación , Ratones , Células 3T3 NIH , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fosforilación/efectos de los fármacos , Proto-Oncogenes Mas , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos , Análisis de Matrices Tisulares , Trastuzumab , Células Tumorales Cultivadas
11.
In Vitro Cell Dev Biol Anim ; 49(10): 746-51, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23949743

RESUMEN

Fibroblast growth factor 10 (FGF10) is required for embryonic epidermal morphogenesis including brain development, lung morphogenesis, and initiation of limb bud formation. In this study, we investigated the role of FGF10 as a lead induction factor for stem cell differentiation toward urothelial cell. To this end, human multipotent stem cell in vitro system was employed. Human amniotic fluid stem cells were co-cultured with immortalized bladder cancer lines to induce directed differentiation into urothelial cells. Urothelial markers, uroplakin II, III, and cytokeratin 8, were monitored by RT-PCR, immunocytochemistry, and Western blot analysis. Co-cultured stem cells began to express uroplakin II, III, and cytokeratin 8. Targeted FGF10 gene knockdown from bladder cancer cells abolished the directed differentiation. In addition, when FGF10 downstream signaling was blocked with the Mek inhibitor, the co-culture system lost the capacity to induce urothelial differentiation. Exogenous addition of recombinant FGF10 protein promoted stem cell differentiation into urothelium cell lineage. Together, this report suggests that paracrine FGF10 signaling stimulates the differentiation of human stem cell into urothelial cells. Current study provides insight into the potential role of FGF10 as a lead growth factor for bladder regeneration and its therapeutic application for bladder transplantation.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Células Madre/citología , Neoplasias de la Vejiga Urinaria/fisiopatología , Urotelio/citología , Líquido Amniótico/citología , Western Blotting , Línea Celular Tumoral , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/fisiología
12.
PLoS One ; 8(12): e83971, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24386318

RESUMEN

Mounting clinical data suggest that high telomerase activity is tightly associated with cancer progression and poor outcomes. Constitutively activated STAT3 is found in ∼60% of human malignancies and shows a dismal prognosis. We previously reported that activated STAT3 promoted epithelial-mesenchymal transition (EMT) and cancer stem cell phenotype in human breast cancer. However, little is known how STAT3 is regulated in the cancer stem cell and by which mechanisms STAT3 contributes to poor prognosis in aggressive breast cancer. Here we demonstrate that STAT3 physically interacts with CD44 and NF-kB and activates the catalytic subunit of telomerase (hTERT) in human breast cancer stem cells. STAT3 plays a role as a signal transducing molecule between CD44 and NF-kB. In addition to functioning as a catalytic subunit of telomerase, hTERT has been reported to function as a transcription co-factor which drives EMT and cancer stem cell phenotype in human cancer. We observed that activated hTERT increases CD44 (+) subpopulation, whereas targeted knock-down of hTERT abolished cancer stem cell phenotype. Targeted STAT3 knock-down cells also down-regulated hTERT and decreased CD44 subpopulation. Finally, CD44 knock-down resulted in the abrogation of cancer stem cell phenotype and concurrent down-regulation of pSTAT3 and hTERT. Our study delineates the signaling pathway where STAT3 functions as a modulator for CD44 and hTERT, promoting a cancer stem cell phenotype. The constitutive activation of STAT3 signaling that leads to regulation of hTERT pathway may provide novel therapeutic targets for human breast cancer stem cells.


Asunto(s)
Neoplasias de la Mama/patología , Células Madre Neoplásicas/patología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Telomerasa/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Receptores de Hialuranos/metabolismo , FN-kappa B/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Fenotipo , Fosforilación , Regiones Promotoras Genéticas/genética , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA