Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Med Mol Morphol ; 52(4): 198-208, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30805710

RESUMEN

Cancer cells surviving in ascites exhibit cancer stem cell (CSC)-like features. This study analyzed the expression of the CSC marker CD133 in the ascites-derived exosomes obtained from patients with unresectable pancreatic cancer. In addition, inverse correlation of CD133 expression with prognosis was examined. Of the 133 consecutive patients, 19 patients were enrolled in the study. Exosomes derived from the malignant ascites demonstrated higher density and wider variation in size than those from non-malignant ascites. Western blot revealed enhanced expression of CD133 in exosomes obtained from patients with pancreatic cancer compared to those obtained from patients with gastric cancer or liver cirrhosis. A xenograft mouse model with malignant ascites was established by intraperitoneal inoculation of human pancreatic cancer cells in nude mice. Results obtained from the human study were reproduced in the mouse model. Statistically significant equilateral correlation was identified between the band intensity of CD133 in western blot and overall survival of patients. Lectin microarray analyses revealed glycosylation of CD133 by sialic acids as the major glycosylation among diverse others responsible for the glycosylation of exosomal CD133. These findings suggest that highly glycosylated CD133 in ascites-derived exosomes as a potential biomarker for better prognosis of patients with advanced pancreatic cancer.


Asunto(s)
Antígeno AC133/metabolismo , Ascitis/metabolismo , Biomarcadores de Tumor/metabolismo , Exosomas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Línea Celular Tumoral , Glicosilación , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células PC-3 , Pronóstico , Neoplasias Gástricas/metabolismo
2.
Cancer Sci ; 109(9): 2801-2810, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29981246

RESUMEN

CD44v9 is expressed in cancer stem cells (CSC) and stabilizes the glutamate-cystine transporter xCT on the cytoplasmic membrane, thereby decreasing intracellular levels of reactive oxygen species (ROS). This mechanism confers ROS resistance to CSC and CD44v9-expressing cancer cells. The aims of the present study were to assess: (i) expression status of CD44v9 and xCT in hepatocellular carcinoma (HCC) tissues, including those derived from patients treated with hepatic arterial infusion chemoembolization (HAIC) therapy with cisplatin (CDDP); and (ii) whether combination of CDDP with sulfasalazine (SASP), an inhibitor of xCT, was more effective on tumor cells than CDDP alone by inducing ROS-mediated apoptosis. Twenty non-pretreated HCC tissues and 7 HCC tissues administered HAIC therapy with CDDP before surgical resection were subjected to immunohistochemistry analysis of CD44v9 and xCT expression. Human HCC cell lines HAK-1A and HAK-1B were used in this study; the latter was also used for xenograft experiments in nude mice to assess in vivo efficacy of combination treatment. CD44v9 positivity was significantly higher in HAIC-treated tissues (5/7) than in non-pretreated tissues (2/30), suggesting the involvement of CD44v9 in the resistance to HAIC. xCT was significantly expressed in poorly differentiated HCC tissues. Combination treatment effectively killed the CD44v9-harboring HAK-1B cells through ROS-mediated apoptosis and significantly decreased xenografted tumor growth. In conclusion, the xCT inhibitor SASP augmented ROS-mediated apoptosis in CDDP-treated HCC cells, in which the CD44v9-xCT system functioned. As CD44v9 is typically expressed in HAIC-resistant HCC cells, combination treatment with SASP with CDDP may overcome such drug resistance.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/fisiología , Carcinoma Hepatocelular/tratamiento farmacológico , Receptores de Hialuranos/fisiología , Neoplasias Hepáticas/tratamiento farmacológico , Sulfasalazina/farmacología , Anciano , Anciano de 80 o más Años , Sistema de Transporte de Aminoácidos y+/análisis , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/química , Cisplatino/farmacología , Resistencia a Antineoplásicos , Femenino , Células Hep G2 , Humanos , Receptores de Hialuranos/análisis , Neoplasias Hepáticas/química , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo
3.
Dig Dis Sci ; 62(6): 1527-1536, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28365916

RESUMEN

BACKGROUND AND AIMS: Pigment epithelium-derived factor (PEDF) has been shown to be a potent inhibitor of inflammation through its anti-oxidative property. Since oxidative response is considered to play the pivotal role of the development and progression of nonalcoholic steatohepatitis (NASH), it is conceivable that PEDF may play a protective role against NASH. In this study, we examined whether administration of PEDF slowed the progression of NASH in mice models. METHODS: Mice were fed methionine- and choline-deficient (MCD) diet with or without intramuscular administration of adenovirus-expressing PEDF (Ad-PEDF). Effects of PEDF administration on NASH were histologically and biochemically evaluated. RESULTS: Administration of Ad-PEDF significantly decreased hepatic fat storage as well as serum levels of ALT in MCD diet-fed mice. Dihydroethidium staining showed that MCD diet-triggered oxidative stress was reduced in the liver of Ad-PEDF-administered mice compared to that of PBS- or Ad-LacZ-administered mice. Activation of Kupffer cells and hepatic fibrosis was also inhibited by Ad-PEDF administration. Quantitative real-time RT-PCR revealed that MCD diet up-regulated expressions of TNF-α, IL-1ß, IL-6, TGF-ß, collagen-1, and collagen-3 mRNA, which were also attenuated with Ad-PEDF administration, whereas MCD diet-induced down-regulation of expressions of PPAR-γ mRNA was restored with Ad-PEDF administration. Furthermore, immunoblotting analysis showed that MCD diet-induced up-regulation of NADPH oxidase components was significantly decreased in Ad-PEDF-administered mice. CONCLUSIONS: The present results demonstrated for the first time that PEDF could slow the development and progression of steatohepatitis through the suppression of steatosis and inflammatory response in MCD diet-fed mice. Our study suggests that PEDF supplementation may be a novel therapeutic strategy for the treatment of NASH.


Asunto(s)
Tejido Adiposo/patología , Proteínas del Ojo/farmacología , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Factores de Crecimiento Nervioso/farmacología , Inhibidores de Proteasas/farmacología , ARN Mensajero/metabolismo , Serpinas/farmacología , Adenoviridae/genética , Alanina Transaminasa/sangre , Animales , Deficiencia de Colina/complicaciones , Colágeno Tipo I/genética , Colágeno Tipo III/genética , Dieta , Modelos Animales de Enfermedad , Regulación hacia Abajo , Proteínas del Ojo/genética , Hígado Graso/genética , Hígado Graso/patología , Inyecciones Intramusculares , Interleucina-1beta/genética , Interleucina-6/genética , Macrófagos del Hígado , Cirrosis Hepática/prevención & control , Masculino , Metionina/administración & dosificación , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/metabolismo , Factores de Crecimiento Nervioso/genética , Estrés Oxidativo , PPAR gamma/genética , Serpinas/genética , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba
4.
J Gastroenterol Hepatol ; 29(10): 1830-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24731186

RESUMEN

BACKGROUND AND AIM: Preclinical studies in rodent models of chronic liver fibrosis have shown that transplantation of peripheral blood (PB) CD34(+) cells leads to hepatic regeneration and a reduction of liver fibrosis by suppressing hepatic stellate cell activity and increasing matrix metalloproteinase activity. The aim of this study was to examine the safety and clinical efficacy of intrahepatic transplantation of autologous granulocyte colony-stimulating factor (G-CSF)-mobilized PB-CD34(+) cells in patients with decompensated liver cirrhosis. METHODS: PB-CD34(+) cells were isolated from G-CSF-mobilized apheresis products. Ten patients were treated with G-CSF-mobilized PB-CD34(+) cells (treatment group) and seven patients were treated with standard medical therapy. For mobilization, patients in the treatment group received subcutaneous injections of 10 µg G-CSF/kg/day for 5 days. The cells were then injected at three different doses (5 × 10(5) , 1 × 10(6) and 2 × 10(6) cells/kg) through the hepatic artery. Thereafter, all patients were followed up for 24 months. RESULTS: G-CSF treatment and leukapheresis were well tolerated, and no serious adverse events were observed. Patients in the treatment group had a significant but transient splenomegaly. After 24 weeks, serum albumin was significantly increased in patients who had received middle or high doses of CD34(+) cells compared with baseline. Doppler ultrasound showed a significant increase in hepatic blood flow velocity and blood flow volume after CD34(+) cell therapy. The hepatic vein pressure gradient decreased in two patients who received high-dose CD34(+) cells at week 16. CONCLUSIONS: CD34(+) cell therapy is feasible, safe and effective in slowing the decline of hepatic reserve function.


Asunto(s)
Antígenos CD34 , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Cirrosis Hepática/terapia , Trasplante de Células Madre de Sangre Periférica/métodos , Anciano , Autoinjertos , Estudios de Factibilidad , Femenino , Estudios de Seguimiento , Factor Estimulante de Colonias de Granulocitos/farmacología , Arteria Hepática , Células Estrelladas Hepáticas/parasitología , Venas Hepáticas/fisiopatología , Humanos , Inyecciones Subcutáneas , Circulación Hepática , Cirrosis Hepática/enzimología , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Regeneración Hepática , Masculino , Metaloproteinasas de la Matriz/metabolismo , Persona de Mediana Edad , Estudios Prospectivos , Terapéutica , Factores de Tiempo , Presión Venosa
5.
J Gastroenterol Hepatol ; 28(1): 168-78, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22849788

RESUMEN

BACKGROUND AND AIM: In cirrhosis, sinusoidal endothelial cell injury results in increased endothelin-1 (ET-1) and decreased nitric oxide synthase (NOS) activity, leading to portal hypertension. However, the effects of transplanted endothelial progenitor cells (EPCs) on the cirrhotic liver have not yet been clarified. We investigated whether EPC transplantation reduces portal hypertension. METHODS: Cirrhotic rats were created by the administration of carbon tetrachloride (CCl(4) ) twice weekly for 10 weeks. From week 7, rat bone marrow-derived EPCs were injected via the tail vein in this model once a week for 4 weeks. Endothelial NOS (eNOS), vascular endothelial growth factor (VEGF) and caveolin expressions were examined by Western blots. Hepatic tissue ET-1 was measured by a radioimmunoassay (RIA). Portal venous pressure, mean aortic pressure, and hepatic blood flow were measured. RESULTS: Endothelial progenitor cell transplantation reduced liver fibrosis, α-smooth muscle actin-positive cells, caveolin expression, ET-1 concentration and portal venous pressure. EPC transplantation increased hepatic blood flow, protein levels of eNOS and VEGF. Immunohistochemical analyses of eNOS and isolectin B4 demonstrated that the livers of EPC-transplanted animals had markedly increased vascular density, suggesting reconstitution of sinusoidal blood vessels with endothelium. CONCLUSIONS: Transplantation of EPCs ameliorates vascular dysfunction and portal hypertension, suggesting this treatment may provide a new approach in the therapy of portal hypertension with liver cirrhosis.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Células Endoteliales/trasplante , Endotelio/metabolismo , Hipertensión Portal/fisiopatología , Hipertensión Portal/terapia , Cirrosis Hepática Experimental/fisiopatología , Trasplante de Células Madre , Animales , Tetracloruro de Carbono , Caveolinas/metabolismo , Proliferación Celular , Células Endoteliales/fisiología , Endotelina-1/metabolismo , Endotelio/enzimología , Hipertensión Portal/etiología , Circulación Hepática , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/complicaciones , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Presión Portal , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Cell Physiol ; 227(4): 1538-52, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21678408

RESUMEN

We investigated whether transplantation of purified human peripheral blood CD34(+) cells could reduce established liver fibrosis and up-regulate therapeutic regeneration. Human peripheral blood CD34(+) cells were isolated from total mononuclear cells of healthy volunteers by magnetic cell sorting. Recipient nude rats were injected intraperitoneally with carbon tetrachloride (CCl(4)) twice weekly for 3 weeks before single administration of CD34(+) cells. CCl(4) was then re-administered twice weekly for 3 more weeks, and the nude rats were sacrificed. Saline (control group), 1 × 10(5) (low-dose group), 5 × 10(5) (middle-dose group), or 2 × 10(6) (high-dose group) CD34(+) cells/kg body weight were intrasplenically transplanted after CCl(4) treatment for 3 weeks. Reverse transcriptase-polymerase chain reaction analysis of the freshly isolated CD34(+) cells revealed the expression of CD31, keratin19, α-smooth muscle actin (α-SMA), and epithelial growth factor, but not other liver related markers. The transplanted cells differentiated into vascular and sinusoidal endothelial cells, and vascular smooth muscle cells. CD34(+) cell transplantation reduced liver fibrosis in a dose-dependent fashion, with decreased collagen type-I and α-SMA-positive cells after 6 weeks of CCl(4) treatment by Mallory's Azan and immunohistochemical staining. Gelatin zymography showed that the expression levels of active matrix metalloproteinase-2 and -9 in CD34(+) cell transplanted livers were significantly stronger than those in saline-infused livers. In recipients of high-doses of CD34(+) cells, the number of PCNA-positive hepatocyte increased 6 weeks after CCl(4) treatment compared with saline-infused livers. We conclude that human peripheral blood CD34(+) cell transplantation halts established liver fibrosis and promotes hepatic regeneration in CCl(4)-induced chronic liver injury.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/terapia , Regeneración Hepática , Trasplante de Células Madre de Sangre Periférica , Animales , Antígenos CD34/metabolismo , Secuencia de Bases , Tetracloruro de Carbono/toxicidad , Diferenciación Celular , Supervivencia Celular , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/patología , Cartilla de ADN/genética , Células Endoteliales/patología , Expresión Génica , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Cirrosis Hepática/terapia , Regeneración Hepática/genética , Regeneración Hepática/fisiología , Masculino , Miocitos del Músculo Liso/patología , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Trasplante Heterólogo
7.
Cancer Sci ; 103(3): 542-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22151662

RESUMEN

The drug delivery system to tumors is a critical factor in upregulating the effect of anticancer drugs and reducing adverse events. Recent studies indicated selective migration of bone marrow-derived endothelial progenitor cells (EPC) into tumor tissues. Cytosine deaminase (CD) transforms nontoxic 5-fluorocytosine (5-FC) into the highly toxic 5-fluorouracil (5-FU). We investigated the antitumor effect of a new CD/5-FC system with CD cDNA transfected EPC for hepatocellular carcinoma (HCC) in mice. We used human hepatoma cell lines (HuH-7, HLF, HAK1-B, KYN-2, KIM-1) and a rat EPC cell line (TR-BME-2). Escherichia coli CD cDNA was transfected into TR-BME-2 (CD-TR-BME). The inhibitory effect of 5-FU on the proliferation of hepatoma cell lines and the inhibitory effect of 5-FU secreted by CD-TR-BME and 5-FC on the proliferation of co-cultured hepatoma cells were evaluated by a tetrazolium-based assay. In mouse subcutaneous xenograft models of KYN-2 and HuH-7, CD-TR-BME was transplanted intravenously followed by 5-FC injection intraperitoneally. HuH-7 cells were the most sensitive to 5-FU and KYN-2 cells were the most resistant. CD-TR-BME secreted 5-FU and inhibited HuH-7 proliferation in a 5-FC dose-dependent manner. CD-TR-BME were recruited into the tumor tissues and some were incorporated into tumor vessels. Tumor growth of HuH-7 was significantly suppressed during 5-FC administration. No bodyweight loss, ALT abnormality or bone marrow suppression was observed. These findings suggest that our new CD/5-FC system with CD cDNA transfected EPC could be an effective and safe treatment for suppression of 5-FU-sensitive HCC growth.


Asunto(s)
Antineoplásicos/administración & dosificación , Citosina Desaminasa/metabolismo , Células Endoteliales/trasplante , Flucitosina/administración & dosificación , Terapia Genética/métodos , Neoplasias Hepáticas/terapia , Animales , Línea Celular Tumoral , Movimiento Celular , Citosina Desaminasa/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Fluorouracilo/farmacología , Humanos , Masculino , Ratones , Ratones Desnudos , Microscopía Confocal , Profármacos/administración & dosificación , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos , Células Madre/enzimología , Transfección
8.
Eur J Clin Invest ; 42(7): 717-28, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22224757

RESUMEN

BACKGROUND: Using the dimethylnitrosamine (DMN) rat model of induced fibrosis, we investigated whether transfer of in vitro-expanded endothelial progenitor cells (EPCs) could reconstitute liver tissue and protect against liver fibrosis. MATERIALS AND METHODS: Low-density, adherent, rat bone marrow-derived mononuclear cells were cultured for one week in medium supporting the growth of chemokine (C-X-C motif) receptor 4 (CXCR4)-positive EPCs that were used for transplantation. Test rats were treated with weekly intraperitoneal injections of DMN over a period of 4 weeks. During that period, the rats were also transplanted weekly with in vivo-expanded EPCs. RESULTS: Transplanted CXCR4-positive expanded EPCs entered around the portal tracts, fibrous septa and hepatic sinusoids, locations at which stromal cell-derived factor 1 (SDF-1), a ligand attracting CXCR4-positive cells, was expressed nearby. In EPC-transplanted rats, we observed suppression of liver fibrogenesis, reduced deposition of type I collagen and fibronectin, fewer α-smooth muscle actin-positive cells and lower expression of transforming growth factor (TGF)-ß. The expression of growth factors promoting hepatic regeneration (hepatocyte growth factor, transforming growth factor-α (TGF-α), epidermal growth factor and vascular endothelial growth factor) was significantly increased in EPC-transplanted rats, resulting in hepatocyte proliferation. Immunohistochemical analyses of eNOS and isolectin B4 demonstrated that the livers of EPC-transplanted animals had markedly increased vascular density, suggesting reconstitution of sinusoidal blood vessels with endothelium. Liver function tests of transaminase, total bilirubin, total protein and albumin demonstrated that normal levels were maintained in EPC-transplanted rats. CONCLUSIONS: EPC transplantation effectively promotes the remodelling of tissues damaged by liver fibrosis; it can also reconstitute sinusoids in chronic liver injury.


Asunto(s)
Dimetilnitrosamina/toxicidad , Células Endoteliales/trasplante , Cirrosis Hepática Experimental/prevención & control , Regeneración Hepática/fisiología , Trasplante de Células Madre , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/citología , Tratamiento Basado en Trasplante de Células y Tejidos , Cirrosis Hepática Experimental/metabolismo , Masculino , Modelos Animales , Ratas , Ratas Sprague-Dawley
9.
Am J Pathol ; 176(1): 168-76, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19948828

RESUMEN

Pigment epithelium-derived factor (PEDF) has several biological actions on tumor cells, but its effects are cell-type dependent. The aim of this study was to examine the pathophysiological role of PEDF in hepatocellular carcinoma (HCC). PEDF expression was examined in various hepatoma cell lines and human HCC tissues, and was seen in various hepatoma cell lines including HepG2 cells. In human HCC tissues, PEDF expression was higher than in adjacent non-HCC tissues. In addition, serum PEDF levels were higher in HCC patients than in non-HCC patients, and curative treatment of HCC caused significant reductions in serum PEDF levels compared with pretreatment levels. In vitro experiments, camptothecin (CPT) was used to induce apoptosis and the effect of PEDF was investigated by knockdown of the PEDF gene in CPT-treated HepG2 cells. Knockdown of the PEDF gene enhanced CPT-induced apoptosis, simultaneously down-regulating Bcl-xL expression in HepG2 cells. Expression of apoptosis-related molecules and effects of bafilomycin A1 on CPT-induced apoptosis were also examined in PEDF gene knockdown HepG2 cells. Treatment with bafilomycin A1 suppressed CPT-induced decreases in Bcl-xL expression and increases in apoptosis in PEDF gene knockdown HepG2 cells. PEDF may, therefore, exert anti-apoptotic effects through inhibition of lysosomal degradation of Bcl-xL in CPT-treated HepG2 cells.


Asunto(s)
Apoptosis , Proteínas del Ojo/metabolismo , Lisosomas/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Procesamiento Proteico-Postraduccional , Serpinas/metabolismo , Proteína bcl-X/metabolismo , Apoptosis/efectos de los fármacos , Camptotecina/farmacología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Ciclo Celular/efectos de los fármacos , Ciclofilinas/genética , Ciclofilinas/metabolismo , Densitometría , Proteínas del Ojo/sangre , Proteínas del Ojo/genética , Proteínas del Ojo/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Leupeptinas/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Lisosomas/efectos de los fármacos , Macrólidos/farmacología , Factores de Crecimiento Nervioso/sangre , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Serpinas/sangre , Serpinas/genética , Serpinas/farmacología , Proteína bcl-X/genética
10.
Hepatogastroenterology ; 58(105): 103-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21510295

RESUMEN

BACKGROUND/AIMS: Nocturnal hypoglycemia is an aggravating factor for liver cirrhosis. However, in patients with compensated liver cirrhosis, a clinical parameter associated with nocturnal hypoglycemia remains unclear. The aim of this study is to investigate a clinical parameter associated with nocturnal hypoglycemia in patients with hepatitis C virus (HCV)-related compensated liver cirrhosis. METHODOLOGY: Twenty patients with HCV-related compensated liver cirrhosis were enrolled in this study. Nocturnal glucose profile was examined by the Continuous Glucose Monitoring System. According to the glucose levels between 21:00 to 6:00, patients were classified into a normoglycemia group (glucose level >70 mg/dL, n=10) or a nocturnal hypoglycemia group (glucose level <70 mg/dL, n=10). Differences in body compositions and biochemical parameters were examined between the two groups. RESULTS: Fifty percent of compensated cirrhotic patients showed nocturnal hypoglycemia. The serum level of free fatty acids, but not any other parameters, was significantly higher in the nocturnal hypoglycemia group compared to that in the normoglycemia group (553 +/- 209 vs. 367 +/- 131 mEq/L; p < 0.05). CONCLUSIONS: Nocturnal hypoglycemia occurred even in compensated cirrhotic patients. Higher serum level of free fatty acids may suggest the presence of nocturnal hypoglycemia in HCV-related compensated cirrhotic patients.


Asunto(s)
Ácidos Grasos no Esterificados/sangre , Hepatitis C Crónica/complicaciones , Hipoglucemia/sangre , Cirrosis Hepática/sangre , Cirrosis Hepática/virología , Anciano , Composición Corporal , Ritmo Circadiano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estadísticas no Paramétricas
11.
Clin J Gastroenterol ; 14(1): 330-335, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33038000

RESUMEN

We report a case of primary sclerosing cholangitis (PSC) with autoimmune hemolytic anemia (AIHA). A 47-year-old woman was diagnosed with PSC. One year later, she was admitted to our hospital for jaundice and fatigue. Magnetic resonance cholangiopancreatography (MRCP) showed worsening of the biliary stricture, and rapidly progressive anemia developed simultaneously. Based on the various laboratory findings, she was diagnosed with AIHA. The administration of prednisolone improved not only the anemia but also the biliary stricture. This case is impactful, as there are few case reports of PSC with AIHA. In addition, we were able to observe the changes in imaging findings using MRCP over time.


Asunto(s)
Anemia Hemolítica Autoinmune , Colangitis Esclerosante , Anemia Hemolítica Autoinmune/complicaciones , Anemia Hemolítica Autoinmune/tratamiento farmacológico , Pancreatocolangiografía por Resonancia Magnética , Colangitis Esclerosante/complicaciones , Colangitis Esclerosante/tratamiento farmacológico , Femenino , Humanos , Persona de Mediana Edad , Prednisolona/uso terapéutico
12.
Int J Mol Med ; 46(1): 427-438, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32377696

RESUMEN

The cytokine transforming growth factor­ß (TGF­ß) serves a key role in hepatic fibrosis and has cytostatic effects on hepatocytes. The present study investigated the anti­fibrogenic and regenerative effects of the TGF­ß receptor type I kinase inhibitor galunisertib (LY2157299) in mice with carbon tetrachloride (CCl4)­induced liver cirrhosis and in vitro. Mice were intraperitoneally treated with CCl4 for 8 weeks. At week 5, the mice were divided randomly into four treatment groups: Vehicle­treated; and treated with low­; middle­; and high­dose galunisertib, which was administered from weeks 5­8. The mice were sacrificed after 8 weeks of CCl4 treatment. Liver fibrosis, as evaluated by histology and determination of hydroxyproline content, progressed during week 4­8 of CCl4 treatment in the vehicle­treated mice. Galunisertib treatment dose­dependently prevented liver fibrosis, as demonstrated by the direct inhibition of α­smooth muscle actin­positive activated hepatic stellate cells (HSCs) after 8 weeks of CCl4 treatment. The levels of active matrix metalloproteinase (MMP)­9 in galunisertib­treated livers were significantly increased compared with the vehicle­treated livers. In the high­dose group, the number of PCNA­positive hepatocytes and endothelial cells markedly increased compared with the vehicle group. Reverse transcription­quantitative PCR analysis verified that interleukin­6 and epiregulin expression levels were significantly increased in livers from the group treated with high­dose galunisertib compared with the vehicle­treated group. Galunisertib inhibited the proliferation of activated HSCs and collagen synthesis in addition to restoring MMP activity. Moreover, galunisertib promoted liver remodeling by proliferating hepatocytes and vascular endothelial cells, while significantly increasing liver weight. These results are consistent with the cytostatic action of TGF­ß that negatively regulates liver regeneration, and demonstrated that galunisertib inhibited TGF­ß signaling, halted liver fibrosis progression and promoted hepatic regeneration. The results of the present study suggest that galunisertib may be an effective treatment for liver cirrhosis.


Asunto(s)
Tetracloruro de Carbono/toxicidad , Regeneración Hepática/efectos de los fármacos , Pirazoles/uso terapéutico , Quinolinas/uso terapéutico , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Animales , Línea Celular , Células Hep G2 , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones
13.
J Exp Med ; 215(2): 611-626, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29305395

RESUMEN

Angiogenesis plays an instrumental role in the modulation of adipose tissue mass and metabolism. Targeting adipose vasculature provides an outstanding opportunity for treatment of obesity and metabolic disorders. Here, we report the physiological functions of VEGFR1 in the modulation of adipose angiogenesis, obesity, and global metabolism. Pharmacological inhibition and genetic deletion of endothelial VEGFR1 augmented adipose angiogenesis and browning of subcutaneous white adipose tissue, leading to elevated thermogenesis. In a diet-induced obesity model, endothelial-VEGFR1 deficiency demonstrated a potent anti-obesity effect by improving global metabolism. Along with metabolic changes, fatty liver and insulin sensitivity were also markedly improved in VEGFR1-deficient high fat diet (HFD)-fed mice. Together, our data indicate that targeting of VEGFR1 provides an exciting new opportunity for treatment of obesity and metabolic diseases, such as liver steatosis and type 2 diabetes.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/metabolismo , Endotelio Vascular/metabolismo , Enfermedades Metabólicas/terapia , Receptor 1 de Factores de Crecimiento Endotelial Vascular/deficiencia , Tejido Adiposo Pardo/irrigación sanguínea , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/irrigación sanguínea , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica , Obesidad/etiología , Obesidad/metabolismo , Obesidad/terapia , Termogénesis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Cell Metab ; 28(1): 104-117.e5, 2018 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-29861385

RESUMEN

Intrinsic and evasive antiangiogenic drug (AAD) resistance is frequently developed in cancer patients, and molecular mechanisms underlying AAD resistance remain largely unknown. Here we describe AAD-triggered, lipid-dependent metabolic reprogramming as an alternative mechanism of AAD resistance. Unexpectedly, tumor angiogenesis in adipose and non-adipose environments is equally sensitive to AAD treatment. AAD-treated tumors in adipose environment show accelerated growth rates in the presence of a minimal number of microvessels. Mechanistically, AAD-induced tumor hypoxia initiates the fatty acid oxidation metabolic reprogramming and increases uptake of free fatty acid (FFA) that stimulates cancer cell proliferation. Inhibition of carnitine palmitoyl transferase 1A (CPT1) significantly compromises the FFA-induced cell proliferation. Genetic and pharmacological loss of CPT1 function sensitizes AAD therapeutic efficacy and enhances its anti-tumor effects. Together, we propose an effective cancer therapy concept by combining drugs that target angiogenesis and lipid metabolism.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Carnitina O-Palmitoiltransferasa/metabolismo , Resistencia a Antineoplásicos , Ácidos Grasos/metabolismo , Neoplasias , Neovascularización Patológica/metabolismo , Tejido Adiposo/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Carnitina O-Palmitoiltransferasa/genética , Línea Celular Tumoral , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico , Neoplasias/patología
15.
Mol Cancer Res ; 15(6): 744-752, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28179411

RESUMEN

Doublecortin-like kinase 1 (DCLK1), a marker for intestinal and pancreatic cancer stem cells, is highly expressed in neuroblastomas. This study was conducted to assess DCLK1 expression levels in pancreatic neuroendocrine tumor (PNET) tissues and to explore the roles of this molecule in clinical tissue from multiple PNET patients, cells (BON1, QGP1, and CM) and tumor xenografts. Immunohistochemically, all PNET tissues highly and diffusely expressed DCLK1 as a full-length isoform, identical to that detected in primary liver NETs. A DCLK1-overexpressing PNET cell line (QGP1-DCLK1) exhibited epithelial-mesenchymal transition (EMT)-related gene signatures, and robust upregulation of Slug (SNAI2), N-Cadherin (CDH2), and Vimentin (VIM) was validated by real-time PCR and immunoblotting. QGP1-DCLK1 cells had increased cell migration in a wound-healing assay and formed significantly larger xenograft tumors in nude mice. The factors involved in the formation of the fast-growing tumors included p-FAK (on Tyr925), p-ERK1/2, p-AKT, Paxillin, and Cyclin D1, which upon knockdown or pharmacologic inhibition of DCLK1 abolished the expression of these molecules. In conclusion, robust and ubiquitous expression of DCLK1 was first demonstrated here in human PNET tissue specimens and cells. DCLK1 characterized the PNET cell behavior, inducing p-FAK/SLUG-mediated EMT. These findings suggest the possibility of developing novel therapeutic strategies against PNETs by targeting DCLK1.Implications: Evidence here reveals that human PNETs diffusely and robustly express the cancer stem cell marker DCLK1, which drives SLUG-mediated EMT, and suggests that NETs share biological features for druggable targets with other tumors, including neuroblastoma that also highly expresses DCLK1. Mol Cancer Res; 15(6); 744-52. ©2017 AACR.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Quinasas Similares a Doblecortina , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Tirosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
World J Gastroenterol ; 23(37): 6833-6844, 2017 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-29085226

RESUMEN

AIM: To elucidate the role of STAT3 in hepatocarcinogenesis and biliary ductular proliferation following chronic liver injury. METHODS: We investigated thioacetamide (TAA)-induced liver injury, compensatory hepatocyte proliferation, and hepatocellular carcinoma (HCC) development in hepatic STAT3-deficient mice. In addition, we evaluated TAA-induced biliary ductular proliferation and analyzed the activation of sex determining region Y-box9 (SOX9) and Yes-associated protein (YAP), which regulate the transdifferentiation of hepatocytes to cholangiocytes. RESULTS: Both compensatory hepatocyte proliferation and HCC formation were significantly decreased in hepatic STAT3-deficient mice as compared with control mice. STAT3 deficiency resulted in augmentation of hepatic necrosis and fibrosis. On the other hand, biliary ductular proliferation increased in hepatic STAT3-deficient livers as compared with control livers. SOX9 and YAP were upregulated in hepatic STAT3-deficient hepatocytes. CONCLUSION: STAT3 may regulate hepatocyte proliferation as well as transdifferentiation into cholangiocytes and serve as a therapeutic target for HCC inhibition and biliary regeneration.


Asunto(s)
Sistema Biliar/fisiología , Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas/patología , Regeneración , Factor de Transcripción STAT3/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Sistema Biliar/citología , Carcinoma Hepatocelular/inducido químicamente , Proteínas de Ciclo Celular , Proliferación Celular , Transdiferenciación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Hepatocitos/fisiología , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas Experimentales/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfoproteínas/metabolismo , Fosforilación , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción STAT3/genética , Tioacetamida/toxicidad , Regulación hacia Arriba , Proteínas Señalizadoras YAP
17.
Neoplasia ; 18(7): 413-24, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27435924

RESUMEN

BACKGROUND & AIM: Aflibercept known as ziv-aflibercept in the United States is a soluble decoy receptor of both vascular endothelial growth factor (VEGF) receptor-1 and -2 known to inhibit the binding of VEGF and placental growth factor (PlGF) to VEGF receptor-1 and -2. Here, we analyzed the mechanisms of the antitumor effects of aflibercept in mouse hepatoma models. METHODS: In in vitro studies, we determined the effects of aflibercept on human umbilical vein cell (HUVEC) proliferation and bone marrow (BM) cell differentiation to endothelial progenitor cells (EPCs). In in vivo experiments, aflibercept was injected intraperitoneally in hepatoma cell tumor-bearing mice, and its inhibitory effects on tumor growth and BM cell migration to tumor tissues were evaluated. RESULTS: Aflibercept suppressed phosphorylation of VEGF receptor-1 and -2 in HUVEC and dose-dependently inhibited VEGF-induced HUVEC proliferation. It suppressed the differentiation of BM cells to EPCs and migration of BM cells to tumor tissues. It also suppressed tumor growth and prolonged survival time of tumor-bearing mice without side effects. In tumor tissues, aflibercept upregulated the expression of hypoxia inducible factor1-α, VEGF, PlGF, fibroblast growth factor-2, platelet derived growth factor-BB, and transforming growth factor-α and reduced microvascular density. It also reduced sinusoidal density in noncancerous liver tissues. CONCLUSIONS: Our results demonstrated potent antitumor activity for aflibercept in a mouse model of hepatocellular carcinoma. These effects were mediated through inhibition of neovascularization, caused by inhibition of endothelial cell proliferation, EPC differentiation, and BM cell migration to tumor tissues.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células de la Médula Ósea/citología , Carcinoma Hepatocelular/patología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/citología , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Receptores de Factores de Crecimiento Endotelial Vascular
18.
Mol Ther Methods Clin Dev ; 3: 16025, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27162932

RESUMEN

Ex vivo expansion of autologous cells is indispensable for cell transplantation therapy of patients with liver cirrhosis. The aim of this study was to investigate the efficacy of human ex vivo-expanded CD34(+) cells for treatment of cirrhotic rat liver. Recipient rats were intraperitoneally injected with CCl4 twice weekly for 3 weeks before administration of CD34(+) cells. CCl4 was then re-administered twice weekly for 3 more weeks, and the rats were sacrificed. Saline, nonexpanded or expanded CD34(+) cells were injected via the spleen. After 7 days, CD34(+) cells were effectively expanded in a serum-free culture medium. Expanded CD34(+) cells were also increasingly positive for cell surface markers of VE-cadherin, VEGF receptor-2, and Tie-2. The expression of proangiogenic growth factors and adhesion molecules in expanded CD34(+) cells increased compared with nonexpanded CD34(+) cells. Expanded CD34(+) cell transplantation reduced liver fibrosis, with a decrease of αSMA(+) cells. Assessments of hepatocyte and sinusoidal endothelial cell proliferative activity indicated the superior potency of expanded CD34(+) cells over non-expanded CD34(+) cells. The inhibition of integrin αvß3 and αvß5 disturbed the engraftment of transplanted CD34(+) cells and aggravated liver fibrosis. These findings suggest that expanded CD34(+) cells enhanced the preventive efficacy of cell transplantation in a cirrhotic model.

19.
Nat Commun ; 7: 12680, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27580750

RESUMEN

The impact of discontinuation of anti-VEGF cancer therapy in promoting cancer metastasis is unknown. Here we show discontinuation of anti-VEGF treatment creates a time-window of profound structural changes of liver sinusoidal vasculatures, exhibiting hyper-permeability and enlarged open-pore sizes of the fenestrated endothelium and loss of VE-cadherin. The drug cessation caused highly leaky hepatic vasculatures permit tumour cell intravasation and extravasation. Discontinuation of an anti-VEGF antibody-based drug and sunitinib markedly promotes liver metastasis. Mechanistically, host hepatocyte, but not tumour cell-derived vascular endothelial growth factor (VEGF), is responsible for cancer metastasis. Deletion of hepatocyte VEGF markedly ablates the 'off-drug'-induced metastasis. These findings provide mechanistic insights on anti-VEGF cessation-induced metastasis and raise a new challenge for uninterrupted and sustained antiangiogenic therapy for treatment of human cancers.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Indoles/uso terapéutico , Hígado/irrigación sanguínea , Hígado/patología , Neoplasias/patología , Pirroles/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Femenino , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia/patología , Neoplasias/tratamiento farmacológico , Sunitinib , Factor A de Crecimiento Endotelial Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA