Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Hepatology ; 53(1): 106-15, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20967826

RESUMEN

UNLABELLED: Nonalcoholic steatohepatitis (NASH) is a leading cause of cirrhosis. Recently, we showed that NASH-related cirrhosis is associated with Hedgehog (Hh) pathway activation. The gene encoding osteopontin (OPN), a profibrogenic extracellular matrix protein and cytokine, is a direct transcriptional target of the Hh pathway. Thus, we hypothesize that Hh signaling induces OPN to promote liver fibrosis in NASH. Hepatic OPN expression and liver fibrosis were analyzed in wild-type (WT) mice, Patched-deficient (Ptc(+/-) ) (overly active Hh signaling) mice, and OPN-deficient mice before and after feeding methionine and choline-deficient (MCD) diets to induce NASH-related fibrosis. Hepatic OPN was also quantified in human NASH and nondiseased livers. Hh signaling was manipulated in cultured liver cells to assess direct effects on OPN expression, and hepatic stellate cells (HSCs) were cultured in medium with different OPN activities to determine effects on HSC phenotype. When fed MCD diets, Ptc(+/-) mice expressed more OPN and developed worse liver fibrosis (P < 0.05) than WT mice, whereas OPN-deficient mice exhibited reduced fibrosis (P < 0.05). In NASH patients, OPN was significantly up-regulated and correlated with Hh pathway activity and fibrosis stage. During NASH, ductular cells strongly expressed OPN. In cultured HSCs, SAG (an Hh agonist) up-regulated, whereas cyclopamine (an Hh antagonist) repressed OPN expression (P < 0.005). Cholangiocyte-derived OPN and recombinant OPN promoted fibrogenic responses in HSCs (P < 0.05); neutralizing OPN with RNA aptamers attenuated this (P < 0.05). CONCLUSION: OPN is Hh-regulated and directly promotes profibrogenic responses. OPN induction correlates with Hh pathway activity and fibrosis stage. Therefore, OPN inhibition may be beneficial in NASH.


Asunto(s)
Proteínas Hedgehog/fisiología , Cirrosis Hepática/etiología , Osteopontina/genética , Animales , Línea Celular , Deficiencia de Colina , Dieta , Hígado Graso/fisiopatología , Células Estrelladas Hepáticas , Humanos , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Osteopontina/biosíntesis , Osteopontina/deficiencia , Regulación hacia Arriba , Alcaloides de Veratrum/farmacología
2.
Hepatology ; 52(1): 278-90, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20578145

RESUMEN

UNLABELLED: Hepatic accumulation of myofibroblastic hepatic stellate cells (MF-HSCs) is pivotal in the pathogenesis of cirrhosis. Two events are necessary for MF-HSCs to accumulate in damaged livers: transition of resident, quiescent hepatic stellate cells (Q-HSCs) to MF-HSCs and expansion of MF-HSC numbers through increased proliferation and/or reduced apoptosis. In this study, we identified two novel mediators of MF-HSC accumulation: Ras-related C3 botulinum toxin substrate 1 (Rac1) and Hedgehog (Hh). It is unclear whether Rac1 and Hh interact to regulate the accumulation of MF-HSCs. We evaluated the hypothesis that Rac1 promotes activation of the Hh pathway, thereby stimulating signals that promote transition of Q-HSCs into MF-HSCs and enhance the viability of MF-HSCs. Using both in vitro and in vivo model systems, Rac1 activity was manipulated through adenoviral vector-mediated delivery of constitutively active or dominant-negative rac1. Rac1-transgenic mice with targeted myofibroblast expression of a mutated human rac1 transgene that produces constitutively active Rac1 were also examined. Results in all models demonstrated that activating Rac1 in HSC enhanced Hh signaling, promoted acquisition/maintenance of the MF-HSC phenotype, increased MF-HSC viability, and exacerbated fibrogenesis. Conversely, inhibiting Rac1 with dominant-negative rac1 reversed these effects in all systems examined. Pharmacologic manipulation of Hh signaling demonstrated that profibrogenic actions of Rac1 were mediated by its ability to activate Hh pathway-dependent mechanisms that stimulated myofibroblastic transition of HSCs and enhanced MF-HSC viability. CONCLUSION: These findings demonstrate that interactions between Rac1 and the Hh pathway control the size of MF-HSC populations and have important implications for the pathogenesis of cirrhosis.


Asunto(s)
Fibroblastos/patología , Proteínas Hedgehog/metabolismo , Células Estrelladas Hepáticas/patología , Cirrosis Hepática/patología , Proteína de Unión al GTP rac1/metabolismo , Animales , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Masculino , Fenotipo , Ratas , Ratas Sprague-Dawley , Proteína de Unión al GTP rac1/agonistas , Proteína de Unión al GTP rac1/genética
3.
Hepatology ; 51(6): 1998-2007, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20512988

RESUMEN

UNLABELLED: Liver inflammation is greater in nonalcoholic steatohepatitis (NASH) than steatosis, suggesting that immune responses contribute to nonalcoholic fatty liver disease (NAFLD) progression. Livers normally contain many natural killer T (NKT) cells that produce factors that modulate inflammatory and fibrogenic responses. Such cells are relatively depleted in steatosis, but their status in more advanced NAFLD is uncertain. We hypothesized that NKT cells accumulate and promote fibrosis progression in NASH. We aimed to determine if livers become enriched with NKT cells during NASH-related fibrosis; identify responsible mechanisms; and assess if NKT cells stimulate fibrogenesis. NKT cells were analyzed in wildtype mice and Patched-deficient (Ptc(+/-)) mice with an overly active Hedgehog (Hh) pathway, before and after feeding methionine choline-deficient (MCD) diets to induce NASH-related fibrosis. Effects of NKT cell-derived factors on hepatic stellate cells (HSC) were examined and fibrogenesis was evaluated in CD1d-deficient mice that lack NKT cells. NKT cells were quantified in human cirrhotic and nondiseased livers. During NASH-related fibrogenesis in wildtype mice, Hh pathway activation occurred, leading to induction of factors that promoted NKT cell recruitment, retention, and viability, plus liver enrichment with NKT cells. Ptc(+/-) mice accumulated more NKT cells and developed worse liver fibrosis; CD1d-deficient mice that lack NKT cells were protected from fibrosis. NKT cell-conditioned medium stimulated HSC to become myofibroblastic. Liver explants were 2-fold enriched with NKT cells in patients with non-NASH cirrhosis, and 4-fold enriched in patients with NASH cirrhosis. CONCLUSION: Hh pathway activation leads to hepatic enrichment with NKT cells that contribute to fibrosis progression in NASH.


Asunto(s)
Hígado Graso/inmunología , Hígado/inmunología , Células T Asesinas Naturales/fisiología , Animales , Deficiencia de Colina/complicaciones , Hígado Graso/metabolismo , Hígado Graso/patología , Fibrosis , Proteínas Hedgehog/metabolismo , Células Estrelladas Hepáticas/fisiología , Humanos , Hígado/patología , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
4.
Int J Cancer ; 126(6): 1378-89, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19642140

RESUMEN

We previously found that the gene encoding the Myelin and Lymphocyte protein, MAL, was among the most highly expressed genes in serous ovarian cancers from short-term survivors (<3 years) relative to those of long-term survivors (>7 years). In the present study, we have found that this difference in expression is partially attributable to differences in DNA methylation at a specific region within the MAL promoter CpG island. While MAL was largely unmethylated at the transcription start site (Region 1; -48 to +73 bp) in primary serous ovarian cancers, methylation of an upstream region (Region 2; -452 to -266 bp) was inversely correlated with MAL transcription in the primary cancers (R = -0.463) and ovarian cancer cell lines (R = -0.444). Following treatment of the OVCA432 cell line with 5-azacytidine, methylation of Region 2 decreased from 73.3% to 34.7% (p = 0.007) while Region 1 was unaffected. This was accompanied by a 10-fold increase in MAL expression. Since MAL transcripts are elevated in tumors from short-term survivors, all of whom were treated with platinum-based therapy, MAL may have a role in cisplatin response. We therefore determined the 50% growth inhibitory dose of cisplatin in 30 ovarian cancer cell lines and compared this to MAL expression. MAL transcript levels were higher in the resistant ovarian cell lines (p = 0.04). MAL methylation status may therefore serve as a marker of platinum sensitivity while MAL protein may be a target for development of novel therapies aimed at enhancing sensitivity to platinum-based drugs in ovarian cancer.


Asunto(s)
Metilación de ADN , Proteínas de Transporte de Membrana/genética , Proteínas de la Mielina/genética , Neoplasias Ováricas/genética , Regiones Promotoras Genéticas/genética , Proteolípidos/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Azacitidina/farmacología , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Células Epiteliales/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN/métodos , Sobrevivientes
5.
Gastroenterology ; 137(4): 1478-1488.e8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19577569

RESUMEN

BACKGROUND & AIMS: Repair responses define the ultimate outcomes of liver disease. This study evaluated the hypothesis that fibrogenic repair in nonalcoholic fatty liver disease (NAFLD) is mediated by Hedgehog (Hh) pathway activation and consequent induction of epithelial-to-mesenchymal transitions (EMT) in ductular-type progenitors. METHODS: Immature ductular cells were exposed to Sonic hedgehog (Shh) in the presence or absence of the Hh inhibitor cyclopamine to determine whether Hh-pathway activation directly modulates EMT in liver progenitors. Potential biologic correlates of progenitor cell EMT were assessed using mice fed methionine-choline-deficient + ethionine (MCDE) diets with or without cyclopamine. The effects of increased Hh signaling on EMT and fibrogenic repair during diet-induced NAFLD were also compared in wild-type (WT) and Patched haplo-insufficient (Ptc(+/-)) mice. Finally, evidence of Hh-pathway activation and EMT was examined in liver sections from patients with NAFLD. RESULTS: In cultured progenitors, Shh repressed expression of epithelial genes and EMT inhibitors but induced genes that are expressed by myofibroblasts. Cyclopamine reversed these effects. In mouse NAFLD models, Hh-pathway activation, EMT, expansion of myofibroblastic populations, and liver fibrosis occurred. Cyclopamine inhibited Hh-pathway activation and induction of EMT. Ptc(+/-) mice, which have an overactive Hh pathway, exhibited sustained overinduction of Hh target genes and more EMT, myofibroblast accumulation, and fibrosis than WT mice. Numbers of Shh-producing cells and Hh-responsive ductular cells that expressed EMT markers increased in parallel with liver fibrosis in patients with NAFLD. CONCLUSIONS: Hh-mediated EMT in ductular cells contributes to the pathogenesis of cirrhosis in NAFLD.


Asunto(s)
Transdiferenciación Celular , Células Epiteliales/metabolismo , Hígado Graso/metabolismo , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Cirrosis Hepática Experimental/etiología , Transducción de Señal , Animales , Estudios de Casos y Controles , Línea Celular , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Deficiencia de Colina/complicaciones , Deficiencia de Colina/metabolismo , Deficiencia de Colina/patología , Progresión de la Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Hígado Graso/etiología , Hígado Graso/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/patología , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Receptores Patched , Receptor Patched-1 , ARN Mensajero/metabolismo , Ratas , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Alcaloides de Veratrum/farmacología
6.
Hepatology ; 50(5): 1421-30, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19676126

RESUMEN

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is a potentially progressive liver disease that culminates in cirrhosis. Cirrhosis occurs more often in individuals with nonalcoholic steatohepatitis (NASH) than in those with steatosis (nonalcoholic fatty liver [NAFL]). The difference between NAFL and NASH is the extent of hepatocyte apoptosis, which is more extensive in NASH. Because phagocytosis of apoptotic cells activates hepatic stellate cells (HSCs), we examined the hypothesis that a pan-caspase inhibitor, VX-166, would reduce progression of fibrosis in a mouse model of NASH. Male db/db mice were fed methionine/choline-deficient (MCD) diets to induce NASH and liver fibrosis. Mice were gavaged once daily with either the pan-caspase inhibitor VX-166 (6 mg/kg/d; Vertex, Abingdon, UK) or vehicle only and sacrificed at 4 or 8 weeks. Treatment with an MCD diet increased alanine aminotransferase (ALT), caspase-3 activity, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells, NASH, and fibrosis. Treatment of MCD-fed mice with VX-166 decreased active caspase-3, TUNEL-positive cells, and triglyceride content (P < 0.05). However, ALT levels were similar in VX-166-treated mice and vehicle-treated controls. Histological findings also confirmed that both groups had comparable liver injury (NAFLD activity score >or=6). Nevertheless, VX-166-treated MCD-fed mice demonstrated decreased alpha-smooth muscle actin expression (4 weeks, P < 0.05; 8 weeks, P < 0.005) and had reduced hepatic levels of collagen 1alpha1 messenger RNA (8 weeks, P < 0.05). Hydroxyproline content and Sirius red staining of VX-166-treated livers confirmed decreases in fibrosis. CONCLUSION: Inhibiting hepatic apoptosis suppresses the development of fibrosis in mice with NASH. Beneficial effects on liver fibrosis were associated with reductions in hepatic steatosis, but occurred without obvious improvement in liver injury. These findings are consistent with evidence that apoptosis triggers HSC activation and liver fibrosis and suggest that caspase inhibitors may be useful as an antifibrotic NASH therapy.


Asunto(s)
Inhibidores de Caspasas , Inhibidores Enzimáticos/uso terapéutico , Hígado Graso/tratamiento farmacológico , Hígado Graso/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Actinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Deficiencia de Colina/complicaciones , Complicaciones de la Diabetes/complicaciones , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Hígado Graso/inducido químicamente , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Masculino , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Obesos , Obesidad/complicaciones
7.
Semin Liver Dis ; 29(2): 200-10, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19387919

RESUMEN

Subpopulations of individuals with alcohol-induced fatty livers and nonalcoholic steatosis develop steatohepatitis. Steatohepatitis is defined histologically: increased numbers of injured and dying hepatocytes distinguish this condition from simple steatosis. The increased hepatocyte death is generally accompanied by hepatic accumulation of inflammatory cells and sometimes increases in myofibroblastic cells, leading to hepatic fibrosis and eventually, cirrhosis. The purpose of this review is to summarize similarities and differences in the pathogenesis of steatohepatitis in alcoholic fatty liver disease and nonalcoholic fatty liver disease.


Asunto(s)
Hígado Graso Alcohólico/patología , Hígado Graso/patología , Hepatocitos/patología , Animales , Muerte Celular , Citocinas/metabolismo , Retículo Endoplásmico/metabolismo , Etanol/toxicidad , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso Alcohólico/etiología , Hígado Graso Alcohólico/metabolismo , Tracto Gastrointestinal/microbiología , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/microbiología , Humanos , Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos , Estrés Oxidativo , Factores de Riesgo
8.
Gynecol Oncol ; 105(2): 312-20, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17276500

RESUMEN

OBJECTIVES: MKK4 is a metastasis suppressor that is downregulated in some ovarian cancers. We sought to investigate whether promoter methylation, loss of heterozygosity, or changes in phosphorylation are involved in MKK4 dysregulation during ovarian carcinogenesis. METHODS: Bisulfite sequencing was used to determine MKK4 promoter methylation. PCR analysis of tumor/normal DNA was performed to determine LOH at the MKK4 locus. Normal human ovarian surface epithelium (HOSE) and SKOV-3 cells were serum starved and treated with EGF, TGFbeta, or wortmannin. Western blotting was performed using antibodies that detect total and phosphorylated MKK4. RESULTS: No MKK4 promoter hypermethylation was detected in 21 ovarian cancers. LOH was detected at the MKK4 intragenic marker D17S969 in 35% of cases and at D17S1303 in 20%. MKK4 protein was detected in 97% of ovarian tumors. The inactivated phosphoserine 80 (ser-80) form comprised 62% of phosphorylated MKK4 protein in ovarian tumors. Treatment of HOSE or SKOV-3 cells with EGF induced a 1.7- to 4.2-fold increase in phosphorylation of ser-80 MKK4 without altering total MKK4 protein. TGFbeta increased MKK4 ser-80 phosphorylation by 5.4-fold above baseline. The PI3K/Akt pathway inhibitor wortmannin decreased the amount of ser-80 MKK4 by 50%, and inhibited EGF stimulation of MKK4 ser-80 phosphorylation by 60%. CONCLUSIONS: LOH of MKK4 occurs in some ovarian cancers, but without loss of MKK4 protein. MKK4 expression does not appear to be downregulated by promoter methylation. Peptide growth factors induce MKK4 ser-80 phosphorylation, which downregulates its activity. PI3K/Akt pathway inhibitors can partially block ser-80 phosphorylation and this may have therapeutic implications.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , MAP Quinasa Quinasa 4/genética , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Secuencia de Bases , Línea Celular Tumoral , Islas de CpG , Metilación de ADN , Femenino , Humanos , Inmunohistoquímica , Pérdida de Heterocigocidad , MAP Quinasa Quinasa 4/biosíntesis , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Datos de Secuencia Molecular , Estadificación de Neoplasias , Neoplasias Ováricas/patología , Fosforilación , Regiones Promotoras Genéticas
9.
Proc Natl Acad Sci U S A ; 102(12): 4512-7, 2005 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-15753288

RESUMEN

Worldwide HIV-1 vaccine efforts are guided by the principle that HIV-specific T cell responses may provide protection from infection or delay overt disease. However, no clear correlates of T cell-mediated immune protection have been identified. Here, we examine in a HLA-B27(+) HIV seronegative vaccinee persistent HIV-specific vaccine-induced anti-Gag CD4(+) and CD8(+) T cell responses. Although these responses exhibited those characteristics (multifunctionality, appropriate memory phenotype, and targeting of epitopes associated with long-term nonprogression) predicted to correlate with protection from infection, the subject became HIV infected. After HIV infection, the vaccine-induced CD8(+) T cells expanded, but both CD4(+) and CD8(+) T cell responses acquired the functional and phenotypic patterns characteristic of chronic HIV infection. The virus quickly escaped the vaccine-induced T cell response, and the subject progressed more rapidly than expected for someone expressing the HLA-B27 allele. These data suggest that control of HIV by vaccine-elicited HIV-specific T cell responses may be difficult, even when the T cell response has those characteristics predicted to provide optimal protection.


Asunto(s)
Vacunas contra el SIDA/farmacología , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , VIH-1/inmunología , Linfocitos T/inmunología , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , ADN Viral/genética , Productos del Gen gag/genética , Infecciones por VIH/genética , Infecciones por VIH/transmisión , Seronegatividad para VIH/inmunología , VIH-1/genética , Antígeno HLA-B27/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA