Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Intervalo de año de publicación
1.
Am J Pathol ; 187(3): 570-580, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28087162

RESUMEN

Menin (MEN1) is a tumor-suppressor protein in neuroendocrine tissue. Therefore, we tested the novel hypothesis that menin regulates cholangiocarcinoma proliferation. Menin and miR-24 expression levels were measured in the following intrahepatic and extrahepatic cholangiocarcinoma (CCA) cell lines, Mz-ChA-1, TFK-1, SG231, CCLP, HuCCT-1, and HuH-28, as well as the nonmalignant human intrahepatic biliary line, H69. miR-24 miRNA and menin protein levels were manipulated in vitro in Mz-ChA-1 cell lines. Markers of proliferation and angiogenesis (Ki-67, vascular endothelial growth factors A/C, vascular endothelial growth factor receptors 2/3, angiopoietin 1/2, and angiopoietin receptors 1/2) were evaluated. Mz-ChA-1 cells were injected into the flanks of nude mice and treated with miR-24 inhibitor or inhibitor scramble. Menin expression was decreased in advanced CCA specimens, whereas miR-24 expression was increased in CCA. Menin overexpression decreased proliferation, angiogenesis, migration, and invasion. Inhibition of miR-24 increased menin protein expression while decreasing proliferation, angiogenesis, migration, and invasion. miR-24 was shown to negatively regulate menin expression by luciferase assay. Tumor burden and expression of proliferative and angiogenic markers was decreased in the miR-24 inhibited tumor group compared to controls. Interestingly, treated tumors were more fibrotic than the control group. miR-24-dependent expression of menin may be important in the regulation of nonmalignant and CCA proliferation and may be an additional therapeutic tool for managing CCA progression.


Asunto(s)
Colangiocarcinoma/genética , Colangiocarcinoma/patología , MicroARNs/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Anciano , Inductores de la Angiogénesis/metabolismo , Animales , Neoplasias de los Conductos Biliares/patología , Conductos Biliares/patología , Biopsia , Línea Celular Tumoral , Proliferación Celular/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Hepatol ; 64(6): 1295-304, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26923637

RESUMEN

BACKGROUND & AIMS: Disruption of circadian rhythm is associated with cancer development and progression. MicroRNAs (miRNAs) are a class of small non-coding RNAs that trigger mRNA translation inhibition. We aimed to evaluate the role of Per1 and related miRNAs in cholangiocarcinoma growth. METHODS: The expression of clock genes was evaluated in human cholangiocarcinoma tissue arrays and cholangiocarcinoma lines. The rhythmic expression of clock genes was evaluated in cholangiocarcinoma cells and H69 (non-malignant cholangiocytes) by qPCR. We measured cell proliferation, cell cycle and apoptosis in Mz-ChA-1 cells after Per1 overexpression. We examined tumor growth in vivo after injection of Per1 overexpressing cells. We verified miRNAs that targets Per1. The circadian rhythm of miR-34a was evaluated in cholangiocarcinoma and H69 cells. We evaluated cell proliferation, apoptosis and invasion after inhibition of miR-34a in vitro, and the potential molecular mechanisms by mRNA profiling after overexpression of Per1. RESULTS: Expression of Per1 was decreased in cholangiocarcinoma. The circadian rhythm of Per1 expression was lost in cholangiocarcinoma cells. Decreased cell proliferation, lower G2/M arrest, and enhanced apoptosis were shown in Per1 overexpressing cells. An in vivo study revealed decreased tumor growth, decreased proliferation, angiogenesis and metastasis after overexpressing Per1. Per1 was verified as a target of miR-34a. miR-34a was rhythmically expressed in cholangiocarcinoma cells and H69. The inhibition of miR-34a decreased proliferation, migration and invasion in cholangiocarcinoma cells. mRNA profiling has shown that overexpression of Per1 inhibits cell growth through regulation of multiple cancer-related pathways, such as cell cycle, cell growth and apoptosis pathways. CONCLUSIONS: Disruption of circadian rhythms of clock genes contribute to the malignant phenotypes of human cholangiocarcinoma. LAY SUMMARY: The current study is about how biological clock and its regulators affect the bile duct tumor growth. The disruption of biological clock has a negative impact in different cancers. Per1 is a gene that is involved in maintaining the biological clock and show 24h oscillation. Reduced levels of Per1 and disruption of 24h circadian rhythm was found in bile duct cancer cells. Therefore, a genetic modified bile duct cancer cells was created. It has a higher level of Per1 expression and partially recovered circadian rhythm. Those genetic modified cells also displayed slower cell growth or higher rate of cell death. We also used mice model that lack of immune system to show that our genetic modified bile duct cells form smaller tumor. In addition, we tried to see how Per1 is communicating with other genes in regarding of controlling the tumor growth. We found Per1 is regulated by microRNA-34a, a small non-coding RNA that directly binds to genes and inhibit gene expression. Decreased level of miR-34a has also significantly reduced tumor growth through controlling the cell growth and cell death balance. Therefore bile duct cancer patients may be treated with miR-34a inhibitor or Per1 stimulator in the future.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/patología , MicroARNs/fisiología , Proteínas Circadianas Period/fisiología , Animales , Proteínas CLOCK/genética , Línea Celular Tumoral , Proliferación Celular , Ritmo Circadiano , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica
3.
Dig Liver Dis ; 47(11): 964-72, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26277684

RESUMEN

BACKGROUND: Since limited in vitro tools exist for evaluating the pathophysiology of extrahepatic bile ducts, we aim to develop an extrahepatic cholangiocyte culture system from normal rats. METHODS: Extrahepatic ducts were dissected from rats, cut in half length-wise and cultured on collagen-I coated plates. Transepithelial electrical resistance was measured. At ∼85% confluence, in extrahepatic cholangiocytes we measured: (i) cell size and distribution, and expression for cytokeratin-19, secretin, secretin receptor and somatostatin receptor type II (SSTR2), cystic fibrosis transmembrane conductance regulator (CFTR), chloride bicarbonate anion exchanger 2 (AE2), vascular endothelial growth factor-A (VEGF-A) and nerve growth factor (NGF); and (ii) the effect of secretin and/or somatostatin on 3'-5'-cyclic adenosine monophosphate (cAMP) levels and proliferation. RESULTS: Cytokeratin-positive extrahepatic cholangiocytes were cultured for 6 passages to form a cell monolayer. Cholangiocytes proliferated to confluence over a 2-week period. The size of extrahepatic cholangiocytes averaged ∼16 µm. Extrahepatic ducts and cholangiocytes were positive for secretin, secretin receptor and SSTR2, CFTR, AE2, VEGF-A and NGF. In extrahepatic cholangiocyte cultures, secretin increased cAMP (prevented by somatostatin), chloride efflux and proliferation. CONCLUSIONS: Extrahepatic cholangiocyte cultures may be important for studying diseases targeting extrahepatic cholangiocytes such as biliary atresia.


Asunto(s)
Conductos Biliares Extrahepáticos/citología , Antiportadores de Cloruro-Bicarbonato/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Queratina-19/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de la Hormona Gastrointestinal/metabolismo , Receptores de Somatostatina/metabolismo , Secretina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Conductos Biliares Extrahepáticos/efectos de los fármacos , Conductos Biliares Extrahepáticos/fisiología , Técnicas de Cultivo de Célula , Línea Celular , Proliferación Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Impedancia Eléctrica , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Ratas , Secretina/farmacología , Somatostatina/farmacología
4.
J Neuroinflammation ; 11: 121, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25012628

RESUMEN

BACKGROUND: Acute liver failure leads to systemic complications with one of the most dangerous being a decline in neurological function, termed hepatic encephalopathy. Neurological dysfunction is exacerbated by an increase of toxic metabolites in the brain that lead to neuroinflammation. Following various liver diseases, hepatic and circulating chemokines, such as chemokine ligand 2 (CCL2), are elevated, though their effects on the brain following acute liver injury and subsequent hepatic encephalopathy are unknown. CCL2 is known to activate microglia in other neuropathies, leading to a proinflammatory response. However, the effects of CCL2 on microglia activation and the pathogenesis of hepatic encephalopathy following acute liver injury remain to be determined. METHODS: Hepatic encephalopathy was induced in mice via injection of azoxymethane (AOM) in the presence or absence of INCB 3284 dimesylate (INCB), a chemokine receptor 2 inhibitor, or C 021 dihydrochloride (C021), a chemokine receptor 4 inhibitor. Mice were monitored for neurological decline and time to coma (loss of all reflexes) was recorded. Tissue was collected at coma and used for real-time PCR, immunoblots, ELISA, or immunostaining analyses to assess the activation of microglia and consequences on pro-inflammatory cytokine expression. RESULTS: Following AOM administration, microglia activation was significantly increased in AOM-treated mice compared to controls. Concentrations of CCL2 in the liver, serum, and cortex were significantly elevated in AOM-treated mice compared to controls. Systemic administration of INCB or C021 reduced liver damage as assessed by serum liver enzyme biochemistry. Administration of INCB or C021 significantly improved the neurological outcomes of AOM-treated mice, reduced microglia activation, reduced phosphorylation of ERK1/2, and alleviated AOM-induced cytokine upregulation. CONCLUSIONS: These findings suggest that CCL2 is elevated systemically following acute liver injury and that CCL2 is involved in both the microglia activation and neurological decline associated with hepatic encephalopathy. Methods used to modulate CCL2 levels and/or reduce CCR2/CCR4 activity may be potential therapeutic targets for the management of hepatic encephalopathy due to acute liver injury.


Asunto(s)
Quimiocina CCL2/metabolismo , Encefalopatía Hepática/complicaciones , Encefalopatía Hepática/patología , Microglía/metabolismo , Enfermedades del Sistema Nervioso/etiología , Regulación hacia Arriba/efectos de los fármacos , Animales , Azoximetano/toxicidad , Benzamidas/administración & dosificación , Benzamidas/uso terapéutico , Temperatura Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Carcinógenos/toxicidad , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/genética , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Glicina/administración & dosificación , Glicina/análogos & derivados , Glicina/uso terapéutico , Encefalopatía Hepática/inducido químicamente , Encefalopatía Hepática/prevención & control , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
5.
Am J Physiol Gastrointest Liver Physiol ; 306(9): G759-68, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24603459

RESUMEN

Substance P (SP) promotes cholangiocyte growth during cholestasis by activating its receptor, NK1R. SP is a proteolytic product of tachykinin (Tac1) and is deactivated by membrane metalloendopeptidase (MME). This study aimed to evaluate the functional role of SP in the regulation of cholangiocarcinoma (CCA) growth. NK1R, Tac1, and MME expression and SP secretion were assessed in human CCA cells and nonmalignant cholangiocytes. The proliferative effects of SP (in the absence/presence of the NK1R inhibitor, L-733,060) and of L-733,060 were evaluated. In vivo, the effect of L-733,060 treatment or MME overexpression on tumor growth was evaluated by using a xenograft model of CCA in nu/nu nude mice. The expression of Tac1, MME, NK1R, PCNA, CK-19, and VEGF-A was analyzed in the resulting tumors. Human CCA cell lines had increased expression of Tac1 and NK1R, along with reduced levels of MME compared with nonmalignant cholangiocytes, resulting in a subsequent increase in SP secretion. SP treatment increased CCA cell proliferation in vitro, which was blocked by L-733,060. Treatment with L-733,060 alone inhibited CCA proliferation in vitro and in vivo. Xenograft tumors derived from MME-overexpressed human Mz-ChA-1 CCA cells had a slower growth rate than those derived from control cells. Expression of PCNA, CK-19, and VEGF-A decreased, whereas MME expression increased in the xenograft tumors treated with L-733,060 or MME-overexpressed xenograft tumors compared with controls. The study suggests that SP secreted by CCA promotes CCA growth via autocrine pathway. Blockade of SP secretion and NK1R signaling may be important for the management of CCA.


Asunto(s)
Neoplasias de los Conductos Biliares/enzimología , Conductos Biliares Intrahepáticos/enzimología , Proliferación Celular , Colangiocarcinoma/enzimología , Neprilisina/metabolismo , Sustancia P/metabolismo , Animales , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/efectos de los fármacos , Conductos Biliares Intrahepáticos/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Queratina-19/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neprilisina/genética , Antagonistas del Receptor de Neuroquinina-1/farmacología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Receptores de Neuroquinina-1/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Gastroenterology ; 146(7): 1795-808.e12, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24583060

RESUMEN

BACKGROUND & AIMS: Proliferating cholangiocytes secrete and respond to neuroendocrine hormones, including secretin. We investigated whether secretin secreted by S cells and cholangiocytes stimulates biliary proliferation in mice. METHODS: Cholestasis was induced in secretin knockout (Sct(-/-)) and wild-type (control) mice by bile duct ligation (BDL). At days 3 and 7 after BDL, control and Sct(-/-) mice received tail-vein injections of morpholinos against microRNA 125b or let7a. One week later, liver tissues and cholangiocytes were collected. Immunohistochemical, immunoblot, luciferase reporter, and real-time polymerase chain reaction assays were performed. Intrahepatic bile duct mass (IBDM) and proliferation were measured. Secretin secretion was measured in conditioned media from cholangiocytes and S cells and in serum and bile. RESULTS: Secretin secretion was increased in supernatants from cholangiocytes and S cells and in serum and bile after BDL in control mice. BDL Sct(-/-) mice had lower IBDM, reduced proliferation, and reduced production of vascular endothelial growth factor (VEGF) A and nerve growth factor (NGF) compared with BDL control. BDL and control mice given morpholinos against microRNA 125b or let7a had increased IBDM. Livers of mice given morpholinos against microRNA 125b had increased expression of VEGFA, and those treated with morpholinos against microRNA let7a had increased expression of NGF. Secretin regulated VEGF and NGF expression that negatively correlated with microRNA 125b and let7a levels in liver tissue. CONCLUSIONS: After liver injury, secretin produced by cholangiocytes and S cells reduces microRNA 125b and let7a levels, resulting in up-regulation of VEGF and NGF. Modulation of cholangiocyte expression of secretin could be a therapeutic approach for biliary diseases.


Asunto(s)
Conductos Biliares/metabolismo , Proliferación Celular , Colestasis/metabolismo , Hígado/metabolismo , MicroARNs/metabolismo , Secretina/metabolismo , Animales , Apoptosis , Bilis/metabolismo , Conductos Biliares/patología , Células Cultivadas , Colestasis/genética , Colestasis/patología , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Células Enteroendocrinas/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Morfolinos/administración & dosificación , Factor de Crecimiento Nervioso/metabolismo , Secretina/sangre , Secretina/deficiencia , Secretina/genética , Transducción de Señal , Factores de Tiempo , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA