Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Gut ; 68(11): 1994-2006, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30826748

RESUMEN

BACKGROUND AND AIMS: Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related death worldwide. Neurotransmitter-initiated signalling pathway is profoundly implicated in tumour initiation and progression. Here, we investigated whether dysregulated neurotransmitter receptors play a role during pancreatic tumourigenesis. METHODS: The Cancer Genome Atlas and Gene Expression Omnibus datasets were used to identify differentially expressed neurotransmitter receptors. The expression pattern of gamma-aminobutyric acid type A receptor pi subunit (GABRP) in human and mouse PDAC tissues and cells was studied by immunohistochemistry and western blot analysis. The in vivo implications of GABRP in PDAC were tested by subcutaneous xenograft model and lung metastasis model. Bioinformatics analysis, transwell experiment and orthotopic xenograft model were used to identify the in vitro and in vivo effects of GABRP on macrophages in PDAC. ELISA, co-immunoprecipitation, proximity ligation assay, electrophysiology, promoter luciferase activity and quantitative real-time PCR analyses were used to identify molecular mechanism. RESULTS: GABRP expression was remarkably increased in PDAC tissues and associated with poor prognosis, contributed to tumour growth and metastasis. GABRP was correlated with macrophage infiltration in PDAC and pharmacological deletion of macrophages largely abrogated the oncogenic functions of GABRP in PDAC. Mechanistically, GABRP interacted with KCNN4 to induce Ca2+ entry, which leads to activation of nuclear factor κB signalling and ultimately facilitates macrophage infiltration by inducing CXCL5 and CCL20 expression. CONCLUSIONS: Overexpressed GABRP exhibits an immunomodulatory role in PDAC in a neurotransmitter-independent manner. Targeting GABRP or its interaction partner KCNN4 may be an effective therapeutic strategy for PDAC.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Macrófagos/fisiología , Ratones , Transducción de Señal/fisiología
2.
Biochem Biophys Res Commun ; 514(3): 632-638, 2019 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-31076106

RESUMEN

Acetyl-CoA synthetase 2 (ACSS2) generates acetyl-CoA from acetate is important for histone acetylation and gene expression. ACSS2 fulfills distinct functions depending on its cellular location in tumor cells. The role and cellular localization of ACSS2 in hepatocellular carcinoma (HCC) remains to be studied. Herein, we identified that the alternative transcription start site selection of ACSS2 was significantly different between HCC and corresponding adjacent tissues. Alternative transcription start site selection produced two different ACSS2 transcripts, ACSS2-S1 and ACSS2-S2. The two isoforms of ACSS2 had different subcellular localization and different functions. Overexpression of ACSS2-S2 promoted cell proliferation and invasion, but ACSS2-S1 did not. The ACSS2-S1 was mainly present in cytoplasm, and ACSS2-S2 was distributed in both nucleus and cytoplasm. Finally, we demonstrated that alternative transcription start site selection of ACSS2 correlates ribosome biogenesis in HCC. Our findings reveal an oncogenic role of ACSS2-S2 in HCC progression via increase of ribosome biogenesis, and suggest ACSS2-S2 might be a potential therapeutic target against the HCC.


Asunto(s)
Acetato CoA Ligasa/genética , Carcinoma Hepatocelular/metabolismo , Núcleo Celular/metabolismo , Neoplasias Hepáticas/metabolismo , Ribosomas/metabolismo , Sitio de Iniciación de la Transcripción , Acetato CoA Ligasa/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Pronóstico , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ribosomas/genética
3.
Brain Behav Immun ; 77: 150-160, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30590110

RESUMEN

The maturation of natural killer (NK) cells is critical for the acquisition of robust effector functions and the immune response to tumors. However, the influence of psychological stress on NK-cell maturation remains unknown. In this study, we investigated the alteration of NK-cell maturation in response to enriched environment (EE) exposure, which induced eustress, or positive stress, in mice. Analysis of markers representing distinct mature stages revealed that EE promoted the terminal maturation of NK cells both centrally in the bone marrow and peripherally in the spleen and blood. Additionally, EE increased CD27+ immature and intermediate-mature NK cell proliferation in the bone marrow. Furthermore, EE exposure brought about a similar promoting effect on NK-cell maturation in tumor-bearing mice. In tumor-bearing mice, EE substantially enhanced the proliferative potential of splenic CD27+ NK cells compared to those in the bone marrow. EE-housed mice displayed a tumor-resistant phenotype and an increased proportion of intratumoral NK cells, especially CD11b+ CD27- mature NK cells, while splenectomy abolished the tumor-retardant effect caused by EE and EE-induced NK-cell infiltration into tumors. Given that our previous study demonstrated an important role for NK cells in EE-induced tumor inhibition, the findings of this study further indicate that the enhanced maturation and proliferation of splenic NK cells may contribute to EE-induced tumor inhibition to some extent. Taken together, the results of this study suggest a positive modulating effect of environment-induced eustress on NK-cell maturation, with potential implications for understanding how eustress boosts NK-cell antitumor immunity.


Asunto(s)
Ambiente , Células Asesinas Naturales/inmunología , Estrés Psicológico/inmunología , Animales , Médula Ósea , Células de la Médula Ósea/inmunología , Diferenciación Celular/inmunología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Citotoxicidad Inmunológica/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/fisiología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Bazo/inmunología
4.
PLoS Genet ; 12(4): e1005895, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27093186

RESUMEN

Small cell lung cancer (SCLC) is an aggressive disease with poor survival. A few sequencing studies performed on limited number of samples have revealed potential disease-driving genes in SCLC, however, much still remains unknown, particularly in the Asian patient population. Here we conducted whole exome sequencing (WES) and transcriptomic sequencing of primary tumors from 99 Chinese SCLC patients. Dysregulation of tumor suppressor genes TP53 and RB1 was observed in 82% and 62% of SCLC patients, respectively, and more than half of the SCLC patients (62%) harbored TP53 and RB1 mutation and/or copy number loss. Additionally, Serine/Arginine Splicing Factor 1 (SRSF1) DNA copy number gain and mRNA over-expression was strongly associated with poor survival using both discovery and validation patient cohorts. Functional studies in vitro and in vivo demonstrate that SRSF1 is important for tumorigenicity of SCLC and may play a key role in DNA repair and chemo-sensitivity. These results strongly support SRSF1 as a prognostic biomarker in SCLC and provide a rationale for personalized therapy in SCLC.


Asunto(s)
Carcinoma de Células Pequeñas/genética , Neoplasias Pulmonares/genética , Proteínas Oncogénicas/genética , Factores de Empalme Serina-Arginina/genética , Adulto , Anciano , Variaciones en el Número de Copia de ADN , Daño del ADN , Femenino , Silenciador del Gen , Humanos , Masculino , Persona de Mediana Edad , Mutación
5.
Gastroenterology ; 153(3): 799-811.e33, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28583823

RESUMEN

BACKGROUND & AIMS: Individuals with Down syndrome have a low risk for many solid tumors, prompting the search for tumor suppressor genes on human chromosome 21 (HSA21). We aimed to identify and explore potential mechanisms of tumor suppressors on HSA21 in hepatocellular carcinoma (HCC). METHODS: We compared expression of HSA21 genes in 14 pairs of primary HCC and adjacent noncancer liver tissues using the Affymetrix HG-U133 Plus 2.0 array (Affymetrix, Santa Clara, CA). HCC tissues and adjacent normal liver tissues were collected from 108 patients at a hospital in China for real-time polymerase chain reaction and immunohistochemical analyses; expression levels of regulator of calcineurin 1 (RCAN1) isoform 4 (RCAN1.4) were associated with clinical features. We overexpressed RCAN1.4 from lentiviral vectors in MHCC97H and HCCLM3 cells and knocked expression down using small interfering RNAs in SMMC7721 and Huh7 cells. Cells were analyzed in proliferation, migration, and invasion assays. HCC cells that overexpressed RCAN1.4 or with RCAN1.4 knockdown were injected into livers or tail veins of nude mice; tumor growth and numbers of lung metastases were quantified. We performed bisulfite pyrosequencing and methylation-specific polymerase chain reaction analyses to analyze CpG island methylation. We measured phosphatase activity of calcineurin in HCC cells. RESULTS: RCAN1.4 mRNA and protein levels were significantly decreased in primary HCC compared with adjacent noncancer liver tissues. Reduced levels of RCAN1.4 mRNA were significantly associated with advanced tumor stages, poor differentiation, larger tumor size, and vascular invasion. Kaplan-Meier survival analysis showed that patients with HCCs with lower levels of RCAN1.4 mRNA had shorter time of overall survival and time to recurrence than patients whose tumors had high levels of RCAN1.4 mRNA. In HCC cell lines, expression of RCAN1.4 significantly reduced proliferation, migration, and invasive activity. HCC cells that overexpressed RCAN1.4 formed smaller xenograft tumors, with fewer metastases and blood vessels, than control HCC cells. In HCC cells, RCAN1.4 inhibited expression of insulin-like growth factor 1 and vascular endothelial growth factor A by reducing calcineurin activity and blocking nuclear translocation of nuclear factor of activated T cells (NFAT1). HCC cells incubated with the calcineurin inhibitor cyclosporin A had decreased nuclear level of NFAT1. HCC cells had hypermethylation of a CpG island in the 5' regulatory region of RCAN1.4, which reduced its expression. CONCLUSIONS: RCAN1.4 is down-regulated in HCC tissues, compared with non-tumor liver tissues. RCAN1.4 prevents cell proliferation, migration, and invasion in vitro; overexpressed RCAN1.4 in HCC cells prevents growth, angiogenesis, and metastases of xenograft tumors by inhibiting calcineurin activity and nuclear translocation of NFAT1.


Asunto(s)
Calcineurina/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas Musculares/genética , Factores de Transcripción NFATC/metabolismo , ARN Mensajero/análisis , Adulto , Anciano , Animales , Carcinoma Hepatocelular/química , Carcinoma Hepatocelular/secundario , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Cromosomas Humanos Par 21 , Islas de CpG/genética , Metilación de ADN , Proteínas de Unión al ADN , Supervivencia sin Enfermedad , Regulación hacia Abajo , Femenino , Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/análisis , Hígado/química , Neoplasias Hepáticas/química , Masculino , Ratones , Persona de Mediana Edad , Proteínas Musculares/análisis , Factores de Transcripción NFATC/genética , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Trasplante de Neoplasias , Isoformas de Proteínas/genética , Transporte de Proteínas/efectos de los fármacos , Secuencias Reguladoras de Ácidos Nucleicos , Transducción de Señal , Tasa de Supervivencia , Carga Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Gastroenterology ; 153(1): 277-291.e19, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28315323

RESUMEN

BACKGROUND & AIMS: Desmoplasia and poor vascularity cause severe metabolic stress in pancreatic ductal adenocarcinomas (PDACs). Serotonin (5-HT) is a neuromodulator with neurotransmitter and neuroendocrine functions that contributes to tumorigenesis. We investigated the role of 5-HT signaling in the growth of pancreatic tumors. METHODS: We measured the levels of proteins that regulate 5-HT synthesis, packaging, and degradation in pancreata from KrasG12D/+/Trp53R172H/+/Pdx1-Cre (KPC) mice, which develop pancreatic tumors, as well as in PDAC cell lines and a tissue microarray containing 81 human PDAC samples. We also analyzed expression levels of proteins involved in 5-HT synthesis and degradation by immunohistochemical analysis of a tissue microarray containing 311 PDAC specimens, and associated expression levels with patient survival times. 5-HT level in 14 matched PDAC tumor and non-tumor tissues were analyzed by ELISA. PDAC cell lines were incubated with 5-HT and cell survival and apoptosis were measured. We analyzed expression of the 5-HT receptor HTR2B in PDAC cells and effects of receptor agonists and antagonists, as well as HTR2B knockdown with small hairpin RNAs. We determined the effects of 5-HT stimulation on gene expression profiles of BxPC-3 cells. Regulation of glycolysis by 5-HT signaling via HTR2B was assessed by immunofluorescence and immunoprecipitation analyses, as well as by determination of the extracellular acid ratio, glucose consumption, and lactate production. Primary PDACs, with or without exposure to SB204741 (a selective antagonist of HTR2B), were grown as xenograft tumors in mice, and SB204741 was administered to tumor-bearing KPC mice; tumor growth and metabolism were measured by imaging analyses. RESULTS: In immunohistochemical analysis of a tissue microarray of PDAC specimens, increased levels of TPH1 and decreased level of MAOA, which regulate 5-HT synthesis and degradation, correlated with stage and size of PDACs and shorter patient survival time. We found levels of 5-HT to be increased in human PDAC tissues compared with non-tumor pancreatic tissues, and PDAC cell lines compared with non-transformed pancreatic cells. Incubation of PDAC cell lines with 5-HT increased proliferation and prevented apoptosis. Agonists of HTR2B, but not other 5-HT receptors, promoted proliferation and prevented apoptosis of PDAC cells. Knockdown of HTR2B in PDAC cells, or incubation of cells with HTR2B inhibitors, reduced their growth as xenograft tumors in mice. We observed a correlation between 5-HT and glycolytic flux in PDAC cells; levels of metabolic enzymes involved in glycolysis, the phosphate pentose pathway, and hexosamine biosynthesis pathway increased significantly in PDAC cells following 5-HT stimulation. 5-HT stimulation led to formation of the HTR2B-LYN-p85 complex, which increased PI3K-Akt-mTOR signaling and the Warburg effect by increasing protein levels of MYC and HIF1A. Administration of SB204741 to KPC mice slowed growth and metabolism of established pancreatic tumors and prolonged survival of the mice. CONCLUSIONS: Human PDACs have increased levels of 5-HT, and PDAC cells increase expression of its receptor, HTR2B. These increases allow for tumor glycolysis under metabolic stress and promote growth of pancreatic tumors and PDAC xenograft tumors in mice.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor de Serotonina 5-HT2B/metabolismo , Serotonina/metabolismo , Anciano , Animales , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/química , Carcinoma Ductal Pancreático/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Silenciador del Gen , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Indoles/uso terapéutico , Ácido Láctico/biosíntesis , Masculino , Ratones , Persona de Mediana Edad , Monoaminooxidasa/análisis , Trasplante de Neoplasias , Páncreas/química , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptor de Serotonina 5-HT2B/genética , Serotonina/análisis , Serotonina/farmacología , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/uso terapéutico , Transducción de Señal , Estrés Fisiológico , Serina-Treonina Quinasas TOR/metabolismo , Análisis de Matrices Tisulares , Transcriptoma , Triptófano Hidroxilasa/análisis , Urea/análogos & derivados , Urea/uso terapéutico , Familia-src Quinasas/metabolismo
7.
Cell Physiol Biochem ; 46(1): 226-237, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29587298

RESUMEN

BACKGROUND/AIMS: Epidermal growth factor receptor variant III (EGFRvIII), the most frequent EGFR variant, is constitutively activated without binding to EGF and is correlated with a poor prognosis. CH12, a human-mouse chimeric monoclonal antibody, has been developed in our laboratory and selectively binds to overexpressed EGFR and EGFRvIII. A previous study had reported that EGFR could influence autophagic activity, and autophagy is closely related to tumor development and the response to drug therapy. In this study, we aimed to elucidate the effect of CH12 on autophagy and efficacy of combining CH12 with an autophagy inhibitor against EGFRvIII-positive tumors. METHODS: EGFRvIII was overexpressed in liver cancer, glioblastoma and breast cancer, and the change in the autophagy-relevant protein levels was analyzed by western blot assays, LC3 punctate aggregation was analyzed by immunofluorescence. The interaction of Beclin-1 and Rubicon was assessed by co-immunoprecipitation (Co-IP) after CH12 treatment. The efficacy of ATG7 or Beclin-1 siRNA in combination with CH12 in Huh-7-EGFRvIII cells was assessed by CCK-8 assays. The autophagy and apoptosis signaling events in Huh-7-EGFRvIII cells upon treatment with control, CH12, siRNA or combination for 48 h were assessed by western blot assays. RESULTS: Our results showed that, in cancer cell lines overexpressing EGFRvIII, only the liver cancer cell lines Huh-7 and PLC/PRF/5 suggested autophagy activation. We then investigated the mechanism of autophagy activation after EGFRvIII overexpression. The results showed that EGFRvIII interacted with Rubicon, an autophagy inhibition protein, and released Beclin-1 to form the inducer complex, thus contributing to autophagy. In addition, CH12, via inhibiting the phosphorylation of EGFRvIII, promoted the interaction of EGFRvIII with Rubicon, further inducing autophagy. In vitro assays suggested that knocking down the expression of the key proteins ATG7 or Beclin-1 in the autophagy pathway with siRNA inhibits tumor cell proliferation. Combining autophagy-related proteins 7 (ATG7) or Beclin-1 siRNA with CH12 in Huh-7-EGFRvIII cells showed better inhibition of cell proliferation. CONCLUSION: EGFRvIII could induce autophagy, and CH12 treatment could improve autophagy activity in EGFRvIII-positive liver cancer cells. The combination of CH12 with an autophagy inhibitor or siRNA against key proteins in the autophagy pathway displayed more significant efficacy on EGFRvIII-positive tumor cells than monotherapy, and induced cell apoptosis.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Autofagia/efectos de los fármacos , Receptores ErbB/inmunología , Anticuerpos Monoclonales/inmunología , Proteína 7 Relacionada con la Autofagia/antagonistas & inhibidores , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia , Beclina-1/antagonistas & inhibidores , Beclina-1/genética , Beclina-1/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/genética , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Células MCF-7 , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Fosforilación/efectos de los fármacos , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Sequestosoma-1/metabolismo
8.
Mol Ther ; 25(10): 2270-2279, 2017 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-28757080

RESUMEN

The incorporation of an endogenous safety switch represents a rational strategy for the control of toxicities following the administration of adoptive T cell therapies. An ideal safety switch should be capable of depleting the transferred T cells with minimal injury to normal tissues. We generated a fusion receptor by engineering a cryptic 806 epitope of human epidermal growth factor receptor (EGFR) into the N terminus of the full-length human folate receptor 1 (FOLR1), designated as FR806. The expression of FR806 allows transduced T cells to be targeted with CH12, a monoclonal antibody recognizing the 806 epitope, but not wild-type EGFR in healthy tissues. FR806, therefore, constitutes a specific cell-surface marker for the elimination of transduced T cells. We demonstrate that the antibody-drug conjugate (ADC) CH12-MMAF is efficiently internalized by FR806-expressing T cells and has the potential to eliminate them. Transfected T cells could, furthermore, be efficiently detected and purified using CH12 antibodies. In immuno-compromised mice, CH12-MMAF eliminated the majority of transferred T cells expressing FR806 and anti-CD19 chimeric antigen receptor (CAR). The selectivity for the 806 epitope and internalization capacity of FOLR1 makes FR806 an efficient safety switch, which may additionally be used as a detection and purification biomarker for human T cell immunotherapies.


Asunto(s)
Traslado Adoptivo/métodos , Biomarcadores/sangre , Linfocitos T/inmunología , Animales , Línea Celular , Humanos , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Ratones , Ratones SCID , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Biochem Biophys Res Commun ; 494(1-2): 113-119, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-29050937

RESUMEN

Dysregulated potassium (K+) channels have previously been shown to promote the development and progression of many types of cancers. Meanwhile, K+ channels are particularly important in regulating the endocrine and exocrine functions of pancreas. However, the expression pattern and prognostic significance of K+ channels in pancreatic ductal adenocarcinoma (PDAC) remain unknown. In this study, by screening a GEO dataset containing 36 microdissected PDAC and matching normal pancreatic tissue samples, four differentially expressed K+ channels (KCNJ5, KCNJ16, KCNN4 and KCNK1) were identified in PDAC. by immunohistochemical analysis of pancreatic tissue sections from Pdx1-Cre; LSL-KrasG12D/+ mice (KC), Pdx1-Cre; LSL-KrasG12D/+; LSL-Trp53R172H/+ mice (KPC) and human PDAC tissue microarrays, we found that Ca2+-activated K+ channel KCNN4 was significantly elevated in pancreatic intraepithelial neoplasia (PanIN) and PDAC epithelia compared with untransformed pancreas tissues. Higher epithelial KCNN4 expression was closely correlated with advanced TNM stages and predicted a poor prognosis in patients with PDAC. Elevated KCNN4 expression was significantly associated with shorter survival in univariable and multivariable analyses. Collectively, the identification of expression pattern of K+ channels in PDAC and its precursor PanIN demonstrates the importance of KCNN4 channel during the malignant transformation of PDAC. On the basis of the prognostic signals from two independent cohorts, KCNN4 should be considered as a promising therapeutic target.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Anciano , Animales , Biomarcadores de Tumor/metabolismo , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patología , Carcinoma Ductal Pancreático/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Neoplasias Pancreáticas/patología , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Pronóstico
10.
J Med Genet ; 53(9): 643-6, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27287394

RESUMEN

BACKGROUND: The transformation of healthy gastric tissue into intestinal metaplasia (IM) is thought to be a critical premalignant step in the development of intestinal-type gastric adenocarcinoma (GA). How such premalignancies contribute to the development of GA is, however, poorly understood. METHODS: In this study, the extent and clonal complexity in IM tissue from patients without gastric cancer were analysed by measuring variations of multiple microsatellite (MS) markers. RESULTS: Even though these tissues are generally regarded as clinically benign, we found extensive MS length heterogeneity between and within individual IM glands, indicating that complex genome diversity is already pervasive in these tissues. Based on a clonal relationship analysis, we found that there exist multiple clones within individual IM glands and that MS alterations can accumulate in these clones. Moreover, we found spatially distant IM glands with the same MS phenotype, suggesting that these MS alterations were progressed by gland fission. CONCLUSIONS: These results provide evidence that genetic instability is an early event, present within metaplastic tissues of otherwise non-cancer patients, and such frequent genetic alterations can be part of the pathophysiological rationale for the requirement of this phase during gastric carcinogenesis.


Asunto(s)
Metaplasia/genética , Mutación/genética , Neoplasias Gástricas/genética , Adenocarcinoma/genética , Adulto , Femenino , Humanos , Masculino , Repeticiones de Microsatélite/genética , Persona de Mediana Edad , Mosaicismo , Lesiones Precancerosas/genética
11.
Biochem Biophys Res Commun ; 473(2): 593-9, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-27033603

RESUMEN

Living in an enriched housing environment is an established model of eustress and has been consistently shown to reduce the growth of transplanted tumors, including pancreatic cancer. Here, we further investigate the influence of an enriched environment (EE) on the efficacy of chemotherapy in pancreatic cancer. Male C57BL/6 mice were housed in EE or standard environment (SE) conditions and transplanted with syngeneic Panc02 pancreatic cancer cells. Tumor-bearing mice were treated with 5-fluorouracil (5-FU) or gemcitabine (GEM) to examine their sensitivities to chemotherapy. The results showed that both 5-FU and GEM exerted the dose dependent inhibition of tumor growth. The tumor inhibition rates of low-dose 5-FU and GEM were improved from 17.7% and 23.6% to 46.3% and 49.9% by EE housing. Importantly, tumor cells isolated from the pancreatic cancer xenografts of EE mice had significantly enhanced sensitivities to both 5-FU and GEM (IC50 for 5-FU: 2.8 µM versus 27.3 µM; IC50 for GEM: 0.8 µM versus 5.0 µM). Furthermore, using microarray analyses, we identified the "ATP-binding cassette (ABC) transporter" that was overrepresented among EE-induced down-regulated genes in pancreatic cancer. Particularly, the tumoral expression of ABC transporter A8b (ABCA8b) was confirmed to be significantly decreased by EE. Over-expression of ABCA8b in mouse pancreatic cancer cells led to a marked decrease in the sensitivity to chemotherapeutic drugs both in vitro and in vivo. In conclusion, our data indicate that benign stressful stimulation can synergistically boost the efficiency of chemotherapeutics in pancreatic cancer, which suggests a novel strategy for adjuvant cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Desoxicitidina/análogos & derivados , Fluorouracilo/uso terapéutico , Páncreas/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Transportadoras de Casetes de Unión a ATP/análisis , Animales , Desoxicitidina/uso terapéutico , Vivienda para Animales , Masculino , Ratones Endogámicos C57BL , Páncreas/patología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/psicología , Estrés Psicológico , Gemcitabina
12.
Hepatology ; 62(4): 1145-59, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26082033

RESUMEN

UNLABELLED: Hormones and their corresponding receptors are vital in controlling metabolism under normal physiologic and pathologic conditions, but less is known about their roles in the metabolism of cancer. Using a small interfering RNA screening approach, we examined the effects of silencing 20 well-known hormone receptors on the Warburg effect, specifically by measuring the production of lactate in four established hepatocellular carcinoma (HCC) cell lines. We found that silencing a variety of hormone receptors had effects on the production of this metabolite. Unexpectedly silencing of mineralocorticoid receptor (MR) significantly increased lactate production in all these HCC cell lines. Subsequent in vitro and in vivo studies showed that gain- and loss-of-function of MR significantly influenced HCC cellular proliferation, cell cycle distribution, and apoptosis. Furthermore, mechanistic studies revealed that MR as a transcriptional factor directly regulated the expression of miR-338-3p, suppressing the Warburg effects of HCC cells by targeting a key enzyme of glycolysis: pyruvate kinase, liver and red blood cells. Moreover, MR expression was significantly down-regulated in 81% of HCC patient tissues, caused by both chromosome deletion and histone deacetylation. Low expression of MR in tumor tissues was associated with poor patient prognosis. The expression level of miR-338-3p was found to positively correlate with the expression of MR in HCC tissues and to inversely correlate with expression of the enzyme pyruvate kinase, liver and red blood cells. CONCLUSION: MR affects HCC development by modulating the miR-338-3p/pyruvate kinase, liver and red blood cells axis with an ability to suppress the Warburg effect.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Glucólisis , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MicroARNs/fisiología , Piruvato Quinasa/fisiología , Receptores de Mineralocorticoides/fisiología , Carcinoma Hepatocelular/patología , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Células Tumorales Cultivadas
13.
Hepatology ; 62(4): 1132-44, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26054020

RESUMEN

UNLABELLED: Cancer cells possess a unique metabolic phenotype that allows them to preferentially utilize glucose through aerobic glycolysis. This phenomenon is referred to as the "Warburg effect." Accumulating evidence suggests that microRNAs (miRNAs), a class of small noncoding regulatory RNAs, interact with oncogenes/tumor suppressors and induce such metabolic reprograming in cancer cells. To systematically study the metabolic roles of miRNAs in cancer cells, we developed a gain-of-function miRNA screen in HeLa cells. Subsequent investigation of the characterized miRNAs indicated that miR-199a-5p acts as a suppressor for glucose metabolism. Furthermore, miR-199a-5p is often down-regulated in human liver cancer, and its low expression level was correlated with a low survival rate, large tumor size, poor tumor differentiation status, high tumor-node-metastasis stage and the presence of tumor thrombus of patients. MicroRNA-199a-5p directly targets the 3'-untranslated region of hexokinase 2 (HK2), an enzyme that catalyzes the irreversible first step of glycolysis, thereby suppressing glucose consumption, lactate production, cellular glucose-6-phosphate and adenosine triphosphate levels, cell proliferation, and tumorigenesis of liver cancer cells. Moreover, HK2 is frequently up-regulated in liver cancer tissues and associated with poor patient outcomes. The up-regulation of hypoxia-inducible factor-1α under hypoxic conditions suppresses the expression of miR-199a-5p and promotes glycolysis, whereas reintroduction of miR-199a-5p interferes with the expression of HK2, abrogating hypoxia-enhanced glycolysis. CONCLUSION: miR-199a-5p/HK2 reprograms the metabolic process in liver cancer cells and provides potential prognostic predictors for liver cancer patients.


Asunto(s)
Glucólisis , Hexoquinasa/metabolismo , Ácido Láctico/biosíntesis , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MicroARNs/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células Tumorales Cultivadas
14.
Carcinogenesis ; 36(10): 1201-12, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26249360

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading causes for cancer related mortality worldwide. Poor prognosis of HCC patients is mainly due to frequent metastasis and recurrence. Deregulation of metastasis suppressors in malignant cells plays critical roles during cancer metastasis. Thus, novel metastasis suppressors are urgently needed to be uncovered to shed new light on molecular mechanisms driving HCC metastasis. In the present study, decreased expression of leukemia inhibitory factor receptor (LIFR) was demonstrated in HCC, and its expression levels were even lower in HCC with metastasis. Downregulated LIFR expression predicted poor prognosis in HCC patients. LIFR was an independent and significant risk factor for their recurrence and survival. Silencing LIFR resulted in forced metastasis of HCC cells, whereas ectopic overexpression of LIFR attenuated migration and invasion of HCC cells in vitro and in vivo. Moreover, LIFR knockdown could activate phosphoinositide 3-kinase/V-akt Murine Thymoma Viral Oncogene Homolog (PI3K/AKT) signaling through enhancing phosphorylation of Janus kinase 1 (JAK1), which successively promoted matrix metalloproteinase 13 (MMP13) expression and HCC metastasis. Combination of LIFR and p-AKT or MMP13 was a more powerful predictor of poor prognosis for HCC patients. Together, these findings conclude that LIFR functions as a novel metastasis suppressor in HCC and may serve as a prognostic biomarker for HCC patients.


Asunto(s)
Carcinoma Hepatocelular/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/biosíntesis , Neoplasias Hepáticas/genética , Metaloproteinasa 13 de la Matriz/biosíntesis , Proteína Oncogénica v-akt/genética , Adulto , Anciano , Biomarcadores de Tumor , Carcinoma Hepatocelular/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Janus Quinasa 1/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Pronóstico , Transducción de Señal
15.
Int J Cancer ; 136(12): 2973-9, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25403721

RESUMEN

Intestinal metaplasia (IM) has been recognized as the first irreversible precancerous stage of intestinal-type gastric cancer at which genetic instabilities, such as microsatellite (MS) instability and loss of heterozygosity, can already be detected. However, the extent and clonal relationship of these genetic lesions in the precancerous tissues are not fully appreciated. In this work, we have used well established MS markers to analyze the relatedness of spatially separated individual metaplastic glands as well as subsegments within single glands from the same patients. We found that individual IM glands frequently show different marker lengths even for closely apposed IM glands, suggesting that these tissues have already gained the ability to independently evolve their genome regardless of whether or not they share a common origin. Furthermore, within individual IM glands, there is also significant intra-gland diversity in the MS markers. Since most of these cells are not dividing and only have a limited lifespan, this result indicates that in each IM gland, a single dominant clone is rare and new clones are constantly created by either progenitor cells or stem cells. This greatly enhanced ability to create de novo genetic alterations may underlie the importance of this stage in the eventual progression toward cancer. Given the widely observed phenotype switch in the early stages of many solid tumors, whether this associated genetic stability is also an intrinsic property of metaplastic transformation should be extensively characterized to further our understanding of cancer initiation.


Asunto(s)
Mucosa Intestinal/metabolismo , Repeticiones de Microsatélite/genética , Mosaicismo , Lesiones Precancerosas/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Células Clonales/metabolismo , Células Clonales/patología , Progresión de la Enfermedad , Electroforesis Capilar , Femenino , Heterogeneidad Genética , Humanos , Mucosa Intestinal/patología , Pérdida de Heterocigocidad , Masculino , Metaplasia/genética , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Lesiones Precancerosas/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
16.
J Transl Med ; 13: 379, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-26653219

RESUMEN

BACKGROUND AND AIMS: Orthotopic liver transplantation (OLT) can be an effective treatment option for certain patients with early stage hepatocellular carcinoma (HCC) meeting Milan, UCSF, or Hangzhou criteria. However, HCC recurrence rates post-OLT range from 20 to 40 %, with limited follow-up options. Elucidating genetic drivers common to primary and post-OLT recurrent tumors may further our understanding and help identify predictive biomarkers of recurrence-both to ultimately help manage clinical decisions for patients undergoing OLT. METHODS: Whole exome and RNA sequencing in matched primary and recurrent tumors, normal adjacent tissues, and blood from four Chinese HCC patients was conducted. SiRNA knockdown and both qRT-PCR and Western assays were performed on PLCPRF5, SNU449 and HEPG2 cell lines; immunohistochemistry and RNA Sequencing were conducted on the primary tumors of Chinese HCC patients who experienced tumor recurrence post-OLT (n = 9) or did not experience tumor recurrence (n = 12). RESULTS: In three independent HCC studies of patients undergoing transplantation (n = 21) or surgical resection (n = 242, n = 44) of primary tumors (total n = 307), HERC5 mRNA under-expression correlated with shorter: time to tumor recurrence (p = 0.007 and 0.02) and overall survival (p = 0.0063 and 0.023), even after adjustment for relevant clinical variables. HERC5 loss drives CCL20 mRNA and protein over-expression and associates with regulatory T cell infiltration as measured by FOXP3 expression. Further, matched primary and recurrent tumors from the 4 HCC patients indicated clonal selection advantage of Wnt signaling activation and CDKN2A inactivation. CONCLUSIONS: HERC5 plays a crucial role in HCC immune evasion and has clinical relevance as a reproducible prognostic marker for risk of tumor recurrence and survival in patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/cirugía , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/cirugía , Trasplante de Hígado , Variaciones en el Número de Copia de ADN , Humanos , Pronóstico , Recurrencia
18.
BMC Genomics ; 15 Suppl 1: S13, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24564407

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most highly malignant and lethal cancers of the world. Its pathogenesis has been reported to be multi-factorial, and the molecular carcinogenesis of HCC can not be attributed to just a few individual genes. Based on the microRNA and mRNA expression profiling of normal liver tissues, pericancerous hepatocellular tissues and hepatocellular carcinoma tissues, we attempted to find prognosis related gene sets for HCC patients. RESULTS: We identified differentially expressed genes (DEG) from three comparisons: Cancer/Normal, Cancer/Pericancerous and Pericancerous/Normal. GSEA (gene set enrichment analysis) were performed. Based on the enriched gene sets of GO terms, pathways and transcription factor targets, it was found that the genome instability and cell proliferation increased while the metabolism and differentiation decreased in HCC tissues. The expression profile of DEGs in each enriched gene set was used to correlate to the postoperative survival time of HCC patients. Nine gene sets were found to prognostic correlation. Furthermore, after substituting DEG-targeting-microRNA for DEG members of each gene set, two gene sets with the microRNA expression profiles were obtained that had prognostic potential. CONCLUSIONS: The malignancy of HCC could be represented by gene sets, and pericancerous liver exhibits important characteristics of liver cancer. The expression level of gene sets not only in HCC but also in the pericancerous liver showed potential for prognosis implying an option for HCC prognosis at an early stage. Additionally, the gene-targeting-microRNA expression profiles also showed prognostic potential, demonstrating that the multi-factorial molecular pathogenesis of HCC is contributed by various genes and microRNAs.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Hígado/metabolismo , MicroARNs/genética , ARN Mensajero/genética , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/cirugía , Diferenciación Celular , Cromosomas Humanos Par 1 , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/cirugía , Metabolismo , Pronóstico
19.
Mol Cancer ; 13: 226, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25273699

RESUMEN

BACKGROUND: A remolded microenvironment in hepatocellular carcinoma (HCC) caused by abnormally expressed matricellular proteins could promote HCC progression. The cell-matrix interactions mediated by integrins play an important role in tumor microenvironment. Epidermal Growth Factor-like repeats and Discoidin I-Like Domains 3 (EDIL3), an extracellular matrix (ECM) protein with angiogenic and anti-inflammatory effects, is abnormally highly expressed in HCC. Here we aim to analyze its expression in liver and HCC tissues, investigate the underlined mechanisms accounted for HCC progression. METHODS: EDIL3 expression level is examined in normal liver, cirrhotic liver and HCC at both mRNA and protein level. The association between EDIL3 and clinical outcomes is analyzed. The pattern of EDIL3 expression and location is examined using Immunofluorescence and ELISA. Overexpression or knock-down of EDIL3 in a panel of cell lines are subjected to assays related to proliferation, invasion, and anoikis to investigate the mechanisms of this matrix protein in HCC progression. Recombinant EDIL3 treatment is applied to confirm the results. RESULTS: Compared with normal liver and cirrhotic liver, EDIL3 is elevated in HCC. High level of EDIL3 protein is much more commonly in patients with larger tumor or portal vein tumor thrombus (PVTT) formation, associated with poor prognosis. EDIL3 is abundantly expressed in HCC cells and secreted by cancer cells. In vitro and in vivo studies indicate that EDIL3, probably in an autocrine manner, inhibits anoikis and promotes anchorage-independent growth of HCC cells. Further mechanistic studies suggest integrin ligation by EDIL3 and thus that the sustained activation of the FAK-Src-AKT signal is responsible for the anoikis resistance and anchorage independence. Both the administration of cilengitide, a RGD-containing integrin antagonist, and silencing of integrin αV, an important RGD-binding integrin, results in the blockade of anoikis-resistance induced by EDIL3. CONCLUSION: Our study suggests that high levels of autocrine EDIL3 may contribute to a receptive microenvironment for the survival of detached HCC cells and may involve in cancer cell spreading. We also highlight the importance of interaction between EDIL3 and integrin αV and suggest disrupting the ligation of EDIL3 to integrins via RGD-blocking in selected patients may bear potential therapeutic value.


Asunto(s)
Anoicis , Comunicación Autocrina , Carcinoma Hepatocelular/patología , Proteínas Portadoras/metabolismo , Integrina alfaV/metabolismo , Neoplasias Hepáticas/patología , Oligopéptidos/metabolismo , Animales , Anoicis/genética , Comunicación Autocrina/genética , Proteínas de Unión al Calcio , Carcinogénesis/patología , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proteínas Portadoras/genética , Adhesión Celular , Moléculas de Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones Desnudos , Persona de Mediana Edad , Vena Porta/patología , Pronóstico , Reproducibilidad de los Resultados , Transducción de Señal , Trombosis/patología , Regulación hacia Arriba/genética
20.
J Hepatol ; 60(6): 1225-34, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24607627

RESUMEN

BACKGROUND & AIMS: Monoamine oxidase A (MAOA), a catecholamine neurotransmitter degrading enzyme, is closely associated with neurological and psychiatric disorders. However, its role in cancer progression remains unknown. METHODS: Hepatocellular carcinoma (HCC) tissue arrays (n=254) were used to investigate the correlation between MAOA expression and clinicopathological findings. In vitro invasion and anoikis assays, and in vivo intrahepatic and lung metastasis models were used to determine the role of MAOA in HCC metastasis. Quantitative real-time PCR, western blotting, immunohistochemical staining and HPLC analysis were performed to uncover the mechanism of MAOA in HCC. RESULTS: We found that MAOA expression was significantly downregulated in 254 clinical HCC samples and was closely correlated with cancer vasoinvasion, metastasis, and poor prognoses. We then demonstrated that MAOA suppressed norepinephrine/epinephrine (NE/E)-induced HCC invasion and anoikis inhibition, and uncovered that the effects of NE/E on HCC behaviors were primarily mediated through alpha 1A (ADRA1A) and beta 2 adrenergic receptors (ADRB2). In addition to the canonical signaling pathway, which is mediated via adrenergic receptors (ADRs), we found that ADR-mediated EGFR transactivation was also involved in NE-induced HCC invasion and anoikis inhibition. Notably, we found that MAOA could synergize with EGFR inhibitors or ADR antagonists to abrogate NE-induced HCC behaviors. CONCLUSIONS: Taken together, the results of our study may provide insights into the application of MAOA as a novel predictor of clinical outcomes and indicate that increasing MAOA expression or enzyme activity may be a new approach that can be used for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Inhibidores de la Monoaminooxidasa/farmacología , Monoaminooxidasa/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Anoicis/efectos de los fármacos , Anoicis/fisiología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/secundario , Línea Celular Transformada , Línea Celular Tumoral , Epinefrina/metabolismo , Receptores ErbB/metabolismo , Femenino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Monoaminooxidasa/genética , Norepinefrina/metabolismo , Valor Predictivo de las Pruebas , Pronóstico , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA