Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
RNA Biol ; 20(1): 136-139, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-37016725

RESUMEN

The Keystone Symposium 'Small Regulatory RNAs: From Bench to Bedside' was held in Santa Fe, New Mexico from May 1-4, 2022. The symposium was organized by Frank J. Slack, Jörg Vogel, Ivan Martinez and Karyn Schmidt, and brought together scientists working in noncoding RNA biology, therapeutics, and technologies to address mechanistic questions about small regulatory RNAs and facilitate translation of these findings into clinical applications. The conference addressed four specific aims: Aim 1. Focus on the exciting biology of small regulatory RNAs, highlighting the best current research into the role that small RNAs play in fundamental biological processes; Aim 2. Focus on the latest efforts to harness the power of these RNAs as agents in the fight against disease and provide the basic understanding that will drive the invention of powerful clinical tools; Aim 3. Attract leaders from both academia and industry working in small RNAs to one place for critical discussions that will advance the field and accelerate the bench to bedside use of this technology; Aim 4. Provide a stimulating environment where students, postdoctoral researchers and junior investigators, along with scientists from Biotechnology and Pharmaceutical companies specializing in small regulatory RNAs, can present and discuss their research with the best minds in the field.


Asunto(s)
ARN no Traducido , Humanos , ARN no Traducido/genética , Congresos como Asunto
2.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34725152

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) causes the endothelial tumor KS, a leading cause of morbidity and mortality in sub-Saharan Africa. KSHV-encoded microRNAs (miRNAs) are known to play an important role in viral oncogenesis; however, the role of host miRNAs in KS tumorigenesis remains largely unknown. Here, high-throughput small-RNA sequencing of the cellular transcriptome in a KS xenograft model revealed miR-127-3p as one of the most significantly down-regulated miRNAs, which we validated in KS patient tissues. We show that restoration of miR-127-3p suppresses KSHV-driven cellular transformation and proliferation and induces G1 cell cycle arrest by directly targeting the oncogene SKP2. This miR-127-3p-induced G1 arrest is rescued by disrupting the miR-127-3p target site in SKP2 messenger RNA (mRNA) using gene editing. Mechanistically, miR-127-3p-mediated SKP2 repression elevates cyclin-dependent kinase (CDK) inhibitor p21Cip1 and down-regulates cyclin E, cyclin A, and CDK2, leading to activation of the RB protein tumor suppressor pathway and suppression of the transcriptional activities of E2F and Myc, key oncoprotein transcription factors crucial for KSHV tumorigenesis. Consequently, metabolomics analysis during miR-127-3p-induced cell cycle arrest revealed significant depletion of dNTP pools, consistent with RB-mediated repression of key dNTP biosynthesis enzymes. Furthermore, miR-127-3p reconstitution in a KS xenograft mouse model suppresses KSHV-positive tumor growth by targeting SKP2 in vivo. These findings identify a previously unrecognized tumor suppressor function for miR-127-3p in KS and demonstrate that the miR-127-3p/SKP2 axis is a viable therapeutic strategy for KS.


Asunto(s)
Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Sarcoma de Kaposi/metabolismo , Animales , Carcinogénesis , Femenino , Herpesvirus Humano 8/fisiología , Humanos , Ratones Desnudos , Sarcoma de Kaposi/virología
3.
Cell Rep ; 27(4): 1151-1164.e5, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31018130

RESUMEN

Common fragile sites (CFSs) are genomic regions that display gaps and breaks in human metaphase chromosomes under replication stress and are often deleted in cancer cells. We studied an ∼300-bp subregion (Flex1) of human CFS FRA16D in yeast and found that it recapitulates characteristics of CFS fragility in human cells. Flex1 fragility is dependent on the ability of a variable-length AT repeat to form a cruciform structure that stalls replication. Fragility at Flex1 is initiated by structure-specific endonuclease Mus81-Mms4 acting together with the Slx1-4/Rad1-10 complex, whereas Yen1 protects Flex1 against breakage. Sae2 is required for healing of Flex1 after breakage. Our study shows that breakage within a CFS can be initiated by nuclease cleavage at forks stalled at DNA structures. Furthermore, our results suggest that CFSs are not just prone to breakage but also are impaired in their ability to heal, and this deleterious combination accounts for their fragility.


Asunto(s)
Rotura Cromosómica , Sitios Frágiles del Cromosoma/genética , Cromosomas Humanos Par 16/genética , Replicación del ADN , Endonucleasas/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , ADN Polimerasa III/genética , ADN Polimerasa III/metabolismo , Humanos , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Secuencias Repetidas en Tándem
4.
Cell Host Microbe ; 21(3): 367-375, 2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28238624

RESUMEN

Ebola virus (EBOV) infection is characterized by sporadic outbreaks caused by zoonotic transmission. Fixed changes in amino acid sequence, such as A82V in the EBOV glycoprotein (GP) that occurred early in the 2013-16 epidemic, are suspected to confer a selective advantage to the virus. We used biochemical assays of GP function to show that A82V, as well as a polymorphism in residue 544 identified in other outbreaks, enhances infection by decreasing the threshold for activation of membrane fusion activity triggered by the host factors cathepsin B and Niemann-Pick C1. Importantly, the increase in infectivity comes with the cost of decreased virus stability. Thus, emergence of a virus GP with altered properties that can affect transmission and virulence may have contributed to the severity and scope of the 2013-16 EBOV epidemic.


Asunto(s)
Sustitución de Aminoácidos , Ebolavirus/patogenicidad , Epidemias , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/virología , Proteínas Mutantes/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Portadoras/metabolismo , Catepsina B/metabolismo , Ebolavirus/genética , Ebolavirus/fisiología , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/metabolismo , Proteínas Mutantes/genética , Proteína Niemann-Pick C1 , Selección Genética , Proteínas del Envoltorio Viral/genética , Virulencia , Internalización del Virus
5.
PLoS Pathog ; 12(11): e1006020, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27880818

RESUMEN

Merkel cell polyomavirus (MCPyV) is an etiological agent of Merkel cell carcinoma (MCC), a highly aggressive skin cancer. The MCPyV small tumor antigen (ST) is required for maintenance of MCC and can transform normal cells. To gain insight into cellular perturbations induced by MCPyV ST, we performed transcriptome analysis of normal human fibroblasts with inducible expression of ST. MCPyV ST dynamically alters the cellular transcriptome with increased levels of glycolytic genes, including the monocarboxylate lactate transporter SLC16A1 (MCT1). Extracellular flux analysis revealed increased lactate export reflecting elevated aerobic glycolysis in ST expressing cells. Inhibition of MCT1 activity suppressed the growth of MCC cell lines and impaired MCPyV-dependent transformation of IMR90 cells. Both NF-κB and MYC have been shown to regulate MCT1 expression. While MYC was required for MCT1 induction, MCPyV-induced MCT1 levels decreased following knockdown of the NF-κB subunit RelA, supporting a synergistic activity between MCPyV and MYC in regulating MCT1 levels. Several MCC lines had high levels of MYCL and MYCN but not MYC. Increased levels of MYCL was more effective than MYC or MYCN in increasing extracellular acidification in MCC cells. Our results demonstrate the effects of MCPyV ST on the cellular transcriptome and reveal that transformation is dependent, at least in part, on elevated aerobic glycolysis.


Asunto(s)
Antígenos Transformadores de Poliomavirus/metabolismo , Carcinoma de Células de Merkel/virología , Transformación Celular Viral/fisiología , Glucólisis/genética , Neoplasias Cutáneas/virología , Carcinoma de Células de Merkel/metabolismo , Línea Celular , Biología Computacional , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Immunoblotting , Poliomavirus de Células de Merkel , Metabolómica , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias Cutáneas/metabolismo , Transfección
6.
J Cancer Res Clin Oncol ; 141(11): 2037-45, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25989942

RESUMEN

PURPOSE: A previous study showed that flavopiridol increased doxorubicin sensitivity in hypoxic hepatocellular carcinoma (HCC) cells by increasing apoptosis through suppressing hypoxia-inducible N-myc downstream-regulated gene-1 (NDRG1) expression. However, this has not been investigated in an in vivo HCC model. Therefore, we aimed to elucidate whether the combination of doxorubicin and flavopiridol has a synergistic anti-tumor effect in an in vivo HCC model. METHODS: An HCC mouse model was established by implanting C3H/He mouse with MH134 cells. Then, doxorubicin with or without flavopiridol was injected. The anti-tumor efficacy was assessed by evaluating tumor volumes, and the underlying mechanism was investigated by quantifying apoptotic cells, the Ki-67 proliferation index, and microvessel densities (MVDs). Immunohistochemistry of NDRG1 was performed to determine the underlying mechanism. RESULTS: Tumor growth was significantly suppressed in the doxorubicin + flavopiridol combination group compared to the other three groups. The percentage of apoptotic cells was significantly higher, and Ki-67-positive proliferating cells were significantly lower in the combination group compared to the other groups; however, MVDs were not significantly different across the groups. Increased apoptosis by flavopiridol occurred by suppressing hypoxia-inducible NDRG1 expression. CONCLUSIONS: These results show that a combination of doxorubicin and flavopiridol has a synergistic anti-tumor effect in an in vivo HCC model. This synergistic effect of combination therapy was attributed to increased apoptosis and decreased proliferation of tumor cells rather than decreased angiogenesis. These findings suggest that flavopiridol might be an effective adjuvant therapy to doxorubicin-resistant HCC cells by inducing apoptosis through suppression of NDRG1 expression.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proteínas de Ciclo Celular/biosíntesis , Doxorrubicina/uso terapéutico , Flavonoides/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Neoplasias Hepáticas/tratamiento farmacológico , Piperidinas/uso terapéutico , Animales , Antibióticos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Sinergismo Farmacológico , Hexoquinasa/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C3H , Trasplante de Neoplasias , Inhibidores de Proteínas Quinasas/uso terapéutico , Carga Tumoral/efectos de los fármacos
7.
J Cancer Res Clin Oncol ; 140(1): 133-44, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24281334

RESUMEN

PURPOSE: The hydrophobic bile acid, deoxycholic acid (DC), can induce apoptosis in hepatocytes. The roles of DC and its transporter are not yet established in hepatocellular carcinoma (HCC) cells. We investigated DC-induced alterations in HCC cell growth, with a particular focus on the effect of the expression of bile acid (BA)-transporting Na(+)-dependent taurocholic cotransporting polypeptides (NTCPs). METHODS: We determined NTCP expression in four human HCC cell lines: Huh-BAT, Huh-7, SNU-761, and SNU-475. NTCP expression and apoptotic signaling cascades were examined by immunoblot analyses. Cell viability was assessed using the 3,4-(5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium salt assay. Wound healing and invasion assays were performed to evaluate cell migration and invasion abilities. Real-time polymerase chain reaction was performed to measure IL-8 expression levels. Nuclear factor kappa B (NF-κB) activity was evaluated by enzyme-linked immunosorbent assay. RESULTS: The HCC cell lines revealed varying NTCP expression levels, and DC treatment had dual effects, depending on NTCP expression. DC induced apoptosis in NTCP-positive HCC cells, especially under hypoxic conditions. In NTCP-negative HCC cells, simultaneous treatment with DC and cyclooxygenase inhibitor markedly decreased aggressive cellular behaviors via the inhibition of NF-κB/COX-2/IL-8 pathways. CONCLUSION: Hydrophobic bile acid offers therapeutic potential for patients with advanced HCC via different mechanisms depending on NTCP expression levels within the tumor.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Ácido Desoxicólico/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/biosíntesis , Simportadores/biosíntesis , Apoptosis/fisiología , Carcinoma Hepatocelular/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Ciclooxigenasa 2/biosíntesis , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Inducción Enzimática/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Humanos , Interleucina-8/biosíntesis , Neoplasias Hepáticas/patología , FN-kappa B/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Simportadores/genética , Simportadores/metabolismo , Transfección
8.
Oncol Rep ; 25(6): 1739-46, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21455586

RESUMEN

Ursodeoxycholic acid (UDCA), a hydrophilic bile acid, has been shown to inhibit mitogenic signaling and suppressing cell proliferation in colonic tumorigenesis. The transcription of DLC1 (deleted in liver cancer), a tumor suppressor gene, is frequently silenced in various types of human cancer. In this study, we postulated that UDCA may inhibit DLC1 protein degradation in hepatocellular carcinoma (HCC) cells, and increased DLC1 expression may suppress HCC cell growth. Human HCC cell lines were used in this study. The methylation status was measured by methylation-specific PCR following sodium bisulfite treatment. Cell proliferation was assessed using an MTS assay. Kinase signaling cascades were evaluated by immunoblot analysis. For assessing ubiquitination, immunoprecipitation analysis was used. To inhibit cellular protein, specific small interfering RNAs (siRNAs) were transfected into cells. DLC1 protein levels increased over time following UDCA treatment. Specifically, UDCA increased the half-life of the DLC1 protein by inhibiting proteasomal degradation of DLC1 without affecting ubiquitination of the DLC1 protein. In addition, HCC cell growth was suppressed following UDCA treatment and this growth suppression was significantly reversed following transfection with DLC1-siRNA. Inhibition of DLC1 increased cellular proliferation; this was reduced after Rho-inhibitor treatment. Finally, RhoA activity was reduced following UDCA treatment; this result was reversed and thus increased following DLC1-siRNA transfection. In conclusion, these results demonstrate that UDCA induces DLC1 protein expression by inhibiting proteasomal DLC1 degradation in a ubiquitin-independent manner, and that DLC1 induction participates in UDCA-induced suppression of HCC cell growth. These observations implicate UDCA as an anti-proliferative agent in HCC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ácido Ursodesoxicólico/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Inmunoprecipitación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Neurogastroenterol Motil ; 16(1): 71-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20535329

RESUMEN

INTRODUCTION: Cholecystokinin (CCK) belongs to a group of endogenous molecules known as brain-gut neuropeptides and functions as a neuropeptide as well as a gut hormone. It remains unclear whether genetic variation of the CCK receptor plays a role in irritable bowel syndrome (IBS). The aim of this study was to determine and compare the allele and genotype frequencies of the CCK(1) receptor polymorphisms between healthy controls and patients with IBS. METHODS: Genotyping of 80 patients with IBS (who met the Rome III criteria) and 76 healthy controls was performed. We performed PCR amplification for the CCK(1) receptor intron 1 779 T > C and Exon 1 G > A. We confirmed polymorphisms by direct sequencing method. RESULTS: There was a significantly different trend for genotypic distributions of the CCK(1) receptor polymorphism between patients with IBS and healthy controls (p for trend = 0.048). The CCK(1) receptor intron 1 779 T >C polymorphic type was more common in patients with 'IBS-constipation predominant (IBS-C) and IBS-mixed (IBS-M) forms' (19/31, 61.3%) than healthy controls 32/76, 42.1% adjusted odd ratio 2.43, 95% Confidence interval 1.01-5.86). The genotypic distributions of the CCK(1) receptor exon 1 polymorphism were not significantly different between the two groups (p for trend = 0.223). CONCLUSIONS: CCK(1) receptor polymorphisms were associated with IBS. In particular, the CCK(1) receptor intron 1 779 T > C polymorphic type was associated with 'IBS-C and IBS-M'. Further studies are needed in larger number of patients with an even distribution of IBS subtypes.

10.
Am J Physiol Gastrointest Liver Physiol ; 298(1): G126-32, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19910526

RESUMEN

Statin has antifibrotic efficacy in human fibrosing diseases, such as pulmonary and renal fibrosis, and is therefore implicated in hepatic fibrosis. However, statin can also activate protein kinase C (PKC), which augments hepatic fibrogenesis and is thereby likely to reduce the antifibrotic efficacy of statin. This study was designed to explore the hypothesis that simultaneous treatment with statin and PKC inhibitor may synergistically enhance antifibrotic efficacy in hepatic fibrosis. Hepatic fibrosis models were established in BALB/c mice by intraperitoneal injection of carbon tetrachloride or thioacetamide for 6 wk. Pravastatin and enzastaurin (PKC inhibitor) were administered by gavage for 5 wk. Cellular apoptosis was explored using 4',6-diamidino-2-phenylindole or terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate biotin nick end-labeling (TUNEL) staining and immunoblot analysis. Hepatic fibrosis and hepatic stellate cell (HSC) activation were assessed by morphometric analysis of histological findings and immunohistochemistry for alpha-smooth muscle actin. In vitro, the addition of PKC inhibitor significantly increased statin-induced LX-2 cell apoptosis by enhancing the activation of mitochondrial apoptotic signals. TUNEL-positive HSCs were significantly increased in mice treated with statin + PKC inhibitor compared with those in control or single compound-treated mice. The percentage of area occupied by activated HSCs and the extent of collagen deposition were significantly decreased in mice treated with statin + PKC inhibitor compared with those in control or statin-treated mice. In conclusion, simultaneous treatment with statin and PKC inhibitor synergistically enhanced the antifibrotic efficacy in both in vitro and in vivo models of hepatic fibrosis and may therefore have therapeutic implication for reducing hepatic fibrosis.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Indoles/farmacología , Cirrosis Hepática/tratamiento farmacológico , Pravastatina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Apoptosis/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Tetracloruro de Carbono/toxicidad , Línea Celular Transformada , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Tioacetamida/toxicidad
11.
Oncol Rep ; 23(1): 113-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19956871

RESUMEN

MicroRNAs (miRNA) have recently been implicated in carcinogenesis and tumor progression. Hepatocellular carcinoma (HCC) is well known for frequent relapses following curative resection. We attempted to identify the miRNAs associated with HCC recurrence. We analyzed miRNA expression profiles in 25 pairs of HCC and adjacent non-tumor liver tissues from HCC patients using miRNA microarray. Out of 449 miRNAs assayed, we identified seven miRNAs associated with HCC recurrence. In particular, the highest ranked miR-15b was negatively correlated with recurrence. MiR-15b inhibitor transfection increased HCC cell proliferation and inhibited TRAIL-induced apoptosis, while miR-15b precursor transfection decreased proliferation and enhanced apoptosis. Bcl-w was identified as a target molecule regulated by miR-15b. These results indicate that miR-15b expression in HCC tissues may predict a low risk of HCC recurrence. In addition, the modulation of miR-15b expression may be useful as an apoptosis-sensitizing strategy for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/cirugía , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/cirugía , MicroARNs/biosíntesis , Adulto , Anciano , Apoptosis , Proteínas Reguladoras de la Apoptosis/biosíntesis , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
12.
Cancer Lett ; 274(2): 259-65, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-18986761

RESUMEN

Hypoxia induces survival signals in hepatocellular carcinoma (HCC). Hypoxia and retinoic acid (RA) may also induce interleukin-2 (IL-2) receptor expression, and thus we evaluated if RA and IL-2 receptor-targeted therapy are indicated in hypoxic HCCs. RA induced IL-2 receptor expression (R alpha, R beta, R gamma) in HCC cells, whereas hypoxia specifically induced IL-2 R gamma expression. IL-2 stimulated hypoxic HCC cell growth via p42/44 MAPK activation. Combination of denileukin diftitox and RA significantly suppressed hypoxic HCC cell growth compared to single agent-treated or normoxic cells. Therefore, denileukin diftitox and RA may be therapeutically implicated in infiltrative HCCs which are exposed to hypoxic environments.


Asunto(s)
Carcinoma Hepatocelular/patología , División Celular/efectos de los fármacos , Toxina Diftérica/farmacología , Interleucina-2/farmacología , Neoplasias Hepáticas/patología , Tretinoina/farmacología , Western Blotting , Carcinoma Hepatocelular/enzimología , Hipoxia de la Célula , Línea Celular Tumoral , Toxina Diftérica/química , Activación Enzimática , Humanos , Interleucina-2/química , Neoplasias Hepáticas/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo
13.
Cancer Lett ; 271(2): 314-22, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18657357

RESUMEN

Adrenomedullin is implicated in tumor progression and induced by hypoxia. We evaluated if adrenomedullin signaling is active in hepatocellular carcinoma (HCC), especially under hypoxic conditions, and to analyze its prognostic implication in HCC patients. HCC cells expressed adrenomedullin and its receptor, and hypoxia induced adrenomedullin expression. Adrenomedullin stimulated HCC cell growth via Akt activation, which was prevented by adrenomedullin peptide inhibitor. Clinico-pathological analysis revealed adrenomedullin extent was related to vascular invasion and N-cadherin intensity, which were reported to indicate a poor prognosis. In conclusion, adrenomedullin signaling is hypoxia-inducible and functionally active in HCCs, and its expression may be a prognostic factor.


Asunto(s)
Adrenomedulina/fisiología , Carcinoma Hepatocelular/patología , División Celular/fisiología , Hipoxia/patología , Neoplasias Hepáticas/patología , Adulto , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
14.
Biochem Biophys Res Commun ; 361(1): 156-61, 2007 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-17659258

RESUMEN

TGR5 is a novel G protein-coupled cell-surface bile acid receptor. In cholestasis, bile acids induce hepatocyte apoptosis by primarily activating death receptor-mediated signaling. We examined if bile acid-induced TGR5 activation is participating in bile acid-induced hepatocyte apoptosis. TGR5 expression and its responsiveness to bile acid were confirmed in human hepatocytes. TGR5 inhibition attenuated bile acid-induced caspase 8 activation, which resulted from reduced bile acid-induced caspase 8 recruited to a death-inducing signaling complex (DISC). Bile acid-induced c-Jun-N terminal kinase (JNK) activation was dependent on bile acid activation of TGR5. JNK formed complexes with caspase 8, which were reduced following bile acid treatment, but this reduction was prevented when TGR5 or JNK was inhibited. In conclusion, bile acids activate TGR5, which leads to JNK activation and reduced complex formation of JNK with caspase 8, thus facilitating caspase 8 recruitment to DISC. These observations suggest therapeutic applications for TGR5 signaling blockage in cholestasis.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Caspasa 8/metabolismo , Hepatocitos/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Apoptosis , Línea Celular Tumoral , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Activación Enzimática , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos
15.
FEBS Lett ; 581(16): 2954-8, 2007 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-17544413

RESUMEN

Wnt signaling was implicated in pulmonary and renal fibrosis. Since Wnt activity is enhanced in liver cirrhosis, Wnt signaling may also participate in hepatic fibrogenesis. Thus, we determined if Wnt signaling modulates hepatic stellate cell (HSC) activation and survival. Wnt3A treatment significantly activated human HSCs, while this was inhibited in secreted frizzled-related protein 1 (sFRP1) overexpressing cells. Wnt3A treatment significantly suppressed TRAIL-induced apoptosis in control HSCs versus sFRP1 over-expressing cells. Particularly, caspase 3 was more activated in sFRP1 over-expressing cells following TRAIL and Wnt3A treatment. These observations imply that Wnt signaling promotes hepatic fibrosis by enhancing HSC activation and survival.


Asunto(s)
Hepatocitos/citología , Cirrosis Hepática/etiología , Hígado/citología , Hígado/metabolismo , Proteínas Wnt/fisiología , Secuencia de Aminoácidos , Apoptosis/genética , Supervivencia Celular , Células Cultivadas , Hepatocitos/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Transducción de Señal/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Transfección , Proteínas Wnt/metabolismo , Proteínas Wnt/farmacología , Proteína Wnt3 , Proteína Wnt3A
16.
J Gastroenterol ; 41(8): 772-8, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16988766

RESUMEN

BACKGROUND: In cases of cholestasis, bile acids induce hepatocyte apoptosis by activating death receptor-mediated apoptotic signaling cascades. Eupatilin (5,7-dihydroxy-3,4,6-trimethoxyflavone) is a pharmacologically active ingredient found in Artemisia asiatica and exhibits cytoprotective effects against experimentally induced gastrointestinal, pancreatic, and hepatic damage. This study was undertaken to examine if eupatilin modulates bile acid-induced hepatocyte apoptosis. METHODS: Huh-BAT cells, a human hepatocellular carcinoma cell line stably transfected with a bile acid transporter, were used in this study. Apoptosis was quantified using 4',6-diamidino-2-phenylindole dihydrochloride staining, and its signaling cascades were explored by immunoblot analysis. Kinase signaling was evaluated by immunoblotting and by using selective inhibitors. Eupatilin's in vivo effect on bile acid-induced hepatocyte apoptosis was explored in bile duct-ligated rats. RESULTS: Eupatilin significantly reduced bile acid-mediated hepatocyte apoptosis by attenuating bile acid-induced caspase 8 cleavage. Eupatilin diminished the bile acid-induced activation of mitogen-activated protein kinases, including p38 mitogen-activated protein kinase and c-Jun N-terminal kinase. In particular, the eupatilin-mediated inhibition of bile acid-induced c-Jun N-terminal kinase activation was found to be responsible for attenuating caspase 8 cleavage. Moreover, eupatilin diminished hepatocyte apoptosis in bile duct-ligated rats. CONCLUSIONS: Eupatilin attenuates bile acid-induced hepatocyte apoptosis by suppressing bile acid-induced kinase activation. Therefore, eupatilin might be therapeutically efficacious in a variety of human liver diseases associated with cholestasis.


Asunto(s)
Apoptosis/efectos de los fármacos , Flavonoides/farmacología , Hepatocitos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Animales , Caspasa 8/metabolismo , Línea Celular Tumoral , Colestasis/tratamiento farmacológico , Ácido Desoxicólico/farmacología , Humanos , Immunoblotting , Etiquetado Corte-Fin in Situ , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Microscopía Fluorescente , Modelos Animales , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Oncol Rep ; 15(5): 1117-23, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16596172

RESUMEN

N-cadherin signaling has recently been implicated in the progression of certain epithelial tumors by promoting invasion and dissemination of cancer cells. N-cadherin has also been reported to exert an anti-apoptotic effect. In this study, we attempted to evaluate the participation of this adhesion molecule in the progression of human hepatocellular carcinomas (HCCs) by analyzing its anti-apoptotic signaling as well as its prognostic implication in HCC patients. N-cadherin was found to be expressed in human HCCs. We established a stable human HCC cell line expressing a truncated N-cadherin, NCaddeltaC, with a dominant-negative action. NCaddeltaC-expressing cells were more susceptible to bile acid-induced apoptosis than control cells. N-cadherin was found to complex with procaspase-8, and this association was diminished in NCaddeltaC-expressing cells, leading to enhanced procaspase-8 recruitment to death-inducing signaling complex following bile acid treatment. A clinicopathological analysis in patients who had undergone surgical resection for HCC revealed that tumoral N-cadherin up-regulation was significantly related to poor recurrence-free and overall survival. Our findings implicate N-cadherin signaling as contributing to HCC progression by exerting anti-apoptotic effects. Thus, we suggest that the selective interruption of this signaling may have therapeutic potential.


Asunto(s)
Apoptosis , Cadherinas/metabolismo , Carcinoma Hepatocelular/diagnóstico , Neoplasias Hepáticas/diagnóstico , Ácidos y Sales Biliares/farmacología , Biomarcadores de Tumor , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/cirugía , Caspasa 8 , Caspasas/metabolismo , Progresión de la Enfermedad , Femenino , Genes Dominantes , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/cirugía , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/diagnóstico , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/cirugía , Estadificación de Neoplasias , Pronóstico , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...