Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Sci (Lond) ; 129(10): 875-83, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26201095

RESUMEN

Schistosomiasis is a major cause of portal hypertension worldwide. It associates with portal fibrosis that develops during chronic infection. The mechanisms by which the pathogen evokes these host responses remain unclear. We evaluated the hypothesis that schistosome eggs release factors that directly stimulate liver cells to produce osteopontin (OPN), a pro-fibrogenic protein that stimulates hepatic stellate cells to become myofibroblasts. We also investigated the utility of OPN as a biomarker of fibrosis and/or severity of portal hypertension. Cultured cholangiocytes, Kupffer cells and hepatic stellate cells were treated with soluble egg antigen (SEA); OPN production was quantified by quantitative reverse transcriptase polymerase chain reaction (qRTPCR) and ELISA; cell proliferation was assessed by BrdU (5-bromo-2'-deoxyuridine). Mice were infected with Schistosoma mansoni for 6 or 16 weeks to cause early or advanced fibrosis. Liver OPN was evaluated by qRTPCR and immunohistochemistry (IHC) and correlated with liver fibrosis and serum OPN. Livers from patients with schistosomiasis mansoni (early fibrosis n=15; advanced fibrosis n=72) or healthy adults (n=22) were immunostained for OPN and fibrosis markers. Results were correlated with plasma OPN levels and splenic vein pressures. SEA-induced cholangiocyte proliferation and OPN secretion (P<0.001 compared with controls). Cholangiocytes were OPN (+) in Schistosoma-infected mice and humans. Liver and serum OPN levels correlated with fibrosis stage (mice: r=0.861; human r=0.672, P=0.0001) and myofibroblast accumulation (mice: r=0.800; human: r=0.761, P=0.0001). Numbers of OPN (+) bile ductules strongly correlated with splenic vein pressure (r=0.778; P=0.001). S. mansoni egg antigens stimulate cholangiocyte proliferation and OPN secretion. OPN levels in liver and blood correlate with fibrosis stage and portal hypertension severity.


Asunto(s)
Proliferación Celular , Hipertensión Portal/metabolismo , Cirrosis Hepática/metabolismo , Osteopontina/metabolismo , Esquistosomiasis mansoni/metabolismo , Adolescente , Adulto , Animales , Antígenos Helmínticos/farmacología , Conductos Biliares/citología , Conductos Biliares/efectos de los fármacos , Conductos Biliares/metabolismo , Línea Celular , Células Cultivadas , Femenino , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Interacciones Huésped-Parásitos , Humanos , Hipertensión Portal/genética , Hipertensión Portal/parasitología , Inmunohistoquímica , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/parasitología , Masculino , Ratones , Persona de Mediana Edad , Osteopontina/sangre , Osteopontina/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Schistosoma/fisiología , Esquistosomiasis mansoni/genética , Esquistosomiasis mansoni/parasitología , Adulto Joven
2.
Liver Int ; 33(1): 149-61, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23121638

RESUMEN

BACKGROUND: Schistosomiasis mansoni is a major cause of portal fibrosis and portal hypertension. The Hedgehog pathway regulates fibrogenic repair in some types of liver injury. AIMS: Determine if Hedgehog pathway activation occurs during fibrosis progression in schistosomiasis and to determine if macrophage-related mechanisms are involved. METHODS: Immunohistochemistry was used to characterize the cells that generate and respond to Hedgehog ligands in 28 liver biopsies from patients with different grades of schistosomiasis fibrosis staged by ultrasound. Cultured macrophages (RAW264.7 and primary rat Kupffer cells) and primary rat liver sinusoidal endothelial cells (LSEC) were treated with schistosome egg antigen (SEA) and evaluated using qRT-PCR. Inhibition of the Hedgehog pathway was used to investigate its role in alternative activation of macrophages (M2) and vascular tube formation. RESULTS: Patients with schistosomiasis expressed more ligands (Shh and Ihh) and target genes (Patched and Gli2) than healthy individuals. Activated LSEC and myofibroblasts were Hedgehog responsive [Gli2(+)] and accumulated in parallel with fibrosis stage (P < 0.05). Double IHC for Ihh/CD68 showed that Ihh(+) cells were macrophages. In vitro studies demonstrated that SEA-stimulated macrophages to express Ihh and Shh mRNA (P < 0.05). Conditioned media from such macrophages induced luciferase production by Shh-LightII cells (P < 0.001) and Hedgehog inhibitors blocked this effect (P < 0.001). SEA-treated macrophages also up-regulated their own expression of M2 markers, and Hh pathway inhibitors abrogated this response (P < 0.01). Inhibition of the Hedgehog pathway in LSEC blocked SEA-induced migration and tube formation. CONCLUSION: SEA stimulates liver macrophages to produce Hh ligands, which promote alternative activation of macrophages, fibrogenesis and vascular remodelling in schistosomiasis.


Asunto(s)
Proteínas Hedgehog/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Neovascularización Patológica , Esquistosomiasis mansoni/complicaciones , Transducción de Señal , Adulto , Animales , Biopsia , Línea Celular , Células Endoteliales/metabolismo , Células Endoteliales/parasitología , Femenino , Genes Reporteros , Humanos , Inmunohistoquímica , Macrófagos del Hígado/metabolismo , Ligandos , Hígado/diagnóstico por imagen , Hígado/parasitología , Hígado/patología , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/parasitología , Cirrosis Hepática/fisiopatología , Activación de Macrófagos , Macrófagos/parasitología , Macrófagos/patología , Masculino , Ratones , Persona de Mediana Edad , Miofibroblastos/metabolismo , Miofibroblastos/parasitología , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/diagnóstico , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/fisiopatología , Índice de Severidad de la Enfermedad , Transfección , Ultrasonografía , Adulto Joven
3.
Crit Rev Toxicol ; 42(6): 501-48, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22582993

RESUMEN

Prediction of chemical-induced hepatotoxicity in humans from in vitro data continues to be a significant challenge for the pharmaceutical and chemical industries. Generally, conventional in vitro hepatic model systems (i.e. 2-D static monocultures of primary or immortalized hepatocytes) are limited by their inability to maintain histotypic and phenotypic characteristics over time in culture, including stable expression of clearance and bioactivation pathways, as well as complex adaptive responses to chemical exposure. These systems are less than ideal for longer-term toxicity evaluations and elucidation of key cellular and molecular events involved in primary and secondary adaptation to chemical exposure, or for identification of important mediators of inflammation, proliferation and apoptosis. Progress in implementing a more effective strategy for in vitro-in vivo extrapolation and human risk assessment depends on significant advances in tissue culture technology and increasing their level of biological complexity. This article describes the current and ongoing need for more relevant, organotypic in vitro surrogate systems of human liver and recent efforts to recreate the multicellular architecture and hemodynamic properties of the liver using novel culture platforms. As these systems become more widely used for chemical and drug toxicity testing, there will be a corresponding need to establish standardized testing conditions, endpoint analyses and acceptance criteria. In the future, a balanced approach between sample throughput and biological relevance should provide better in vitro tools that are complementary with animal testing and assist in conducting more predictive human risk assessment.


Asunto(s)
Hígado/metabolismo , Técnicas de Cultivo de Tejidos , Pruebas de Toxicidad/métodos , Animales , Técnicas de Cultivo de Célula , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Industria Farmacéutica , Hemodinámica , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Medición de Riesgo/métodos
4.
Liver Int ; 32(2): 312-20, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22098068

RESUMEN

BACKGROUND: Somatostatin is a pleiotropic peptide, exerting a variety of effects through its receptor subtypes. Recently, somatostatin has been shown to act as a chemoattractant for haematopoietic progenitor cells and hepatic oval cells (HOC) via receptor subtype 2 and subtype 4 (SSTR4) respectively. AIMS: We investigated the in vivo effect of somatostatin/SSTR4 on HOC migration in the injured liver model of rats and the type of signalling molecules associated with the chemotactic function. METHODS: Migration assay, HOC transplantation and phosphatidylinositol-3-kinase (PI3K) signalling were assessed with or without somatostatin and an analogue of somatostatin (TT232) that specifically binds to SSTR4. RESULTS: TT232 was shown to have an antimigratory action on HOC induced by somatostatin in vitro. In HOC transplantation experiments, a lower number of donor-derived cells were detected in TT232-treated animals, as compared with control animals. Activation of PI3K was observed in HOC exposed to somatostatin, and this activation was suppressed by either SSTR4 antibody or TT232-pretreatment. In addition, a PI3K inhibitor abrogated the motility of HOC. CONCLUSION: Together, these data suggest that somatostatin stimulates the migration of HOC within injured liver through SSTR4, and this action appears to be mediated by the PI3K pathway.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Péptidos Cíclicos/farmacología , Somatostatina/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Trasplante de Células , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Hepatocitos/patología , Hígado/patología , Masculino , Péptidos Cíclicos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores de Somatostatina/metabolismo , Transducción de Señal/efectos de los fármacos , Somatostatina/metabolismo
5.
J Biol Chem ; 285(47): 36551-60, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20843817

RESUMEN

Trans-differentiation of quiescent hepatic stellate cells (Q-HSCs), which exhibit epithelial and adipocytic features, into myofibroblastic-HSC (MF-HSCs) is a key event in liver fibrosis. Culture models demonstrated that Hedgehog (Hh) pathway activation is required for transition of epithelioid/adipocytic Q-HSCs into MF-HSCs. Hh signaling inhibits adiposity and promotes epithelial-to-mesenchymal transitions (EMTs). Leptin (anti-adipogenic, pro-EMT factor) promotes HSC trans-differentiation and liver fibrosis, suggesting that the pathways may interact to modulate cell fate. This study aimed to determine whether leptin activates Hh signaling and whether this is required for the fibrogenic effects of leptin. Cultures of primary HSCs from lean and fa/fa rats with an inherited ObRb defect were examined. Inhibitors of PI3K/Akt, JAK/STAT, and Hh signaling were used to delineate how ObRb activation influenced Hh signaling and HSC trans-differentiation. Fibrogenesis was compared in wild type and db/db mice (impaired ObRb function) to assess the profibrotic role of leptin. The results demonstrate that leptin-ObR interactions activate Hh signaling with the latter necessary to promote trans-differentiation. Leptin-related increases in Hh signaling required ObR induction of PI3K/Akt, which was sufficient for leptin to repress the epithelioid/adipocytic program. Leptin-mediated induction of JAK/STAT was required for mesenchymal gene expression. Leptin-ObRb interactions were not necessary for HSC trans-differentiation to occur in vitro or in vivo but are important because liver fibrogenesis was attenuated in db/db mice. These findings reveal that leptin activates Hh signaling to alter gene expression programs that control cell fate and have important implications for liver fibrosis and other leptin-regulated processes involving EMTs, including development, obesity, and cancer metastasis.


Asunto(s)
Proteínas Hedgehog/metabolismo , Células Estrelladas Hepáticas/metabolismo , Leptina/metabolismo , Miofibroblastos/metabolismo , Obesidad/metabolismo , Transducción de Señal , Animales , Western Blotting , Diferenciación Celular , Células Cultivadas , Células Epiteliales/metabolismo , Proteínas Hedgehog/genética , Leptina/genética , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Obesos , Miofibroblastos/citología , Fenotipo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Ratas Zucker , Receptores de Leptina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
J Clin Invest ; 118(10): 3331-42, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18802480

RESUMEN

Epithelial-mesenchymal transitions (EMTs) play an important role in tissue construction during embryogenesis, and evidence suggests that this process may also help to remodel some adult tissues after injury. Activation of the hedgehog (Hh) signaling pathway regulates EMT during development. This pathway is also induced by chronic biliary injury, a condition in which EMT has been suggested to have a role. We evaluated the hypothesis that Hh signaling promotes EMT in adult bile ductular cells (cholangiocytes). In liver sections from patients with chronic biliary injury and in primary cholangiocytes isolated from rats that had undergone bile duct ligation (BDL), an experimental model of biliary fibrosis, EMT was localized to cholangiocytes with Hh pathway activity. Relief of ductal obstruction in BDL rats reduced Hh pathway activity, EMT, and biliary fibrosis. In mouse cholangiocytes, coculture with myofibroblastic hepatic stellate cells, a source of soluble Hh ligands, promoted EMT and cell migration. Addition of Hh-neutralizing antibodies to cocultures blocked these effects. Finally, we found that EMT responses to BDL were enhanced in patched-deficient mice, which display excessive activation of the Hh pathway. Together, these data suggest that activation of Hh signaling promotes EMT and contributes to the evolution of biliary fibrosis during chronic cholestasis.


Asunto(s)
Epitelio/metabolismo , Proteínas Hedgehog/metabolismo , Cirrosis Hepática Biliar/fisiopatología , Mesodermo/metabolismo , Transducción de Señal , Animales , Conductos Biliares Intrahepáticos/citología , Conductos Biliares Intrahepáticos/lesiones , Conductos Biliares Intrahepáticos/metabolismo , Línea Celular , Colestasis Intrahepática/metabolismo , Colestasis Intrahepática/fisiopatología , Fibroblastos/metabolismo , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ligandos , Cirrosis Hepática Biliar/metabolismo , Masculino , Ratones , Ratas , Ratas Sprague-Dawley
7.
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
8.
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
9.
Mol Ther ; 18(8): 1553-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20551917

RESUMEN

Adult stem cell-based gene therapy holds several unique advantages including avoidance of germline or other undesirable cell transductions. We have previously shown that liver progenitor (oval) cells can be used as a platform for liver gene delivery of human alpha1-antitrypsin (hAAT). However, this cell source cannot be used in humans for autologous transplantation. In the present study, we tested the feasibility of bone marrow (BM) cell-based liver gene delivery of hAAT. In vitro studies showed that BM cells can be transduced by lentiviral vector (Lenti-CB-hAAT) and recombinant adeno-associated viral vectors (rAAV1-CB-hAAT, and rAAV8-CB-hAAT). Transplantation studies showed that transplanted BM cells homed into liver, differentiated into hepatocytes and expressed hAAT in the liver. Importantly, we showed that transplantation of rAAV8-CB-hAAT vector-transduced BM cells resulted in sustained levels of hAAT in the systemic circulation of recipient mice. These results demonstrated that rAAV vector-mediated BM cell-based liver gene therapy is feasible for the treatment of AAT deficiency and implies a novel therapy for the treatment of liver diseases.


Asunto(s)
Hígado/metabolismo , alfa 1-Antitripsina/metabolismo , Adenoviridae/genética , Albúminas/genética , Albúminas/metabolismo , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción Genética/métodos , Cromosoma Y/metabolismo , alfa 1-Antitripsina/sangre , alfa 1-Antitripsina/genética
10.
Gut ; 59(5): 655-65, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20427400

RESUMEN

OBJECTIVE: The death rate of mature hepatocytes is chronically increased in various liver diseases, triggering responses that prevent liver atrophy, but often cause fibrosis. Mice with targeted disruption of inhibitor kappa B kinase (Ikk) in hepatocytes (HEP mice) provide a model to investigate this process because inhibiting Ikk-nuclear factor-kappaB (NF-kappaB) signalling in hepatocytes increases their apoptosis. METHODS: Cell proliferation, apoptosis, progenitors, fibrosis and production of Hedgehog (Hh) ligands (progenitor and myofibroblast growth factors) were compared in HEP and control mice before and after feeding methionine choline-deficient ethionine-supplemented (MCDE) diets. Ikkbeta was deleted from primary hepatocytes to determine the effects on Hh ligand production; Hh signalling was inhibited directly in progenitors to determine the effects on viability. Liver sections from patients were examined to assess relationships between hepatocyte production of Hh ligands, accumulation of myofibroblastic cells and liver fibrosis. RESULTS: Disrupting the Ikk-NF-kappaB pathway in hepatocytes inhibited their proliferation but induced their production of Hh ligands. The latter provided viability signals for progenitors and myofibroblasts, enhancing accumulation of these cell types and causing fibrogenesis. Findings in the mouse models were recapitulated in diseased human livers. CONCLUSION: Dying mature hepatocytes produce Hh ligands which promote the compensatory outgrowth of progenitors and myofibroblasts. These results help to explain why diseases that chronically increase hepatocyte death promote cirrhosis.


Asunto(s)
Hepatocitos/patología , Hepatopatías/patología , Células Madre/patología , Animales , Apoptosis , Núcleo Celular/metabolismo , Hígado Graso/metabolismo , Hígado Graso/patología , Proteínas Hedgehog/metabolismo , Hepatocitos/metabolismo , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Antígeno Ki-67/metabolismo , Ligandos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/patología , Hepatopatías/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal/fisiología , Células Madre/metabolismo , Factor de Transcripción ReIA/metabolismo
11.
Cell Transplant ; 30: 963689721999617, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33757335

RESUMEN

The origins of low-temperature tissue storage research date back to the late 1800s. Over half a century later, osmotic stress was revealed to be a main contributor to cell death during cryopreservation. Consequently, the addition of cryoprotective agents (CPAs) such as dimethyl sulfoxide (DMSO), glycerol (GLY), ethylene glycol (EG), or propylene glycol (PG), although toxic to cells at high concentrations, was identified as a necessary step to protect against rampant cell death during cryopreservation. In addition to osmotic stress, cooling and thawing rates were also shown to have significant influence on cell survival during low temperature storage. In general, successful low-temperature cell preservation consists of the addition of a CPA (commonly 10% DMSO), alone or in combination with additional permeating or non-permeating agents, cooling rates of approximately 1ºC/min, and storage in either liquid or vapor phase nitrogen. In addition to general considerations, cell-specific recommendations for hepatocytes, pancreatic islets, sperm, oocytes, and stem cells should be observed to maximize yields. For example, rapid cooling is associated with better cryopreservation outcomes for oocytes, pancreatic islets, and embryonic stem cells while slow cooling is recommended for cryopreservation of hepatocytes, hematopoietic stem cells, and mesenchymal stem cells. Yields can be further maximized by implementing additional pre-cryo steps such as: pre-incubation with glucose and anti-oxidants, alginate encapsulation, and selecting cells within an optimal age range and functional ability. Finally, viability and functional assays are critical steps in determining the quality of the cells post-thaw and improving the efficiency of the current cryopreservation methods.


Asunto(s)
Supervivencia Celular/fisiología , Criopreservación/métodos , Crioprotectores/uso terapéutico , Humanos
12.
NPJ Syst Biol Appl ; 7(1): 7, 2021 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-33504769

RESUMEN

The ToxCast in vitro screening program has provided concentration-response bioactivity data across more than a thousand assay endpoints for thousands of chemicals found in our environment and commerce. However, most ToxCast screening assays have evaluated individual biological targets in cancer cell lines lacking integrated physiological functionality (such as receptor signaling, metabolism). We evaluated differentiated HepaRGTM cells, a human liver-derived cell model understood to effectively model physiologically relevant hepatic signaling. Expression of 93 gene transcripts was measured by quantitative polymerase chain reaction using Fluidigm 96.96 dynamic arrays in response to 1060 chemicals tested in eight-point concentration-response. A Bayesian framework quantitatively modeled chemical-induced changes in gene expression via six transcription factors including: aryl hydrocarbon receptor, constitutive androstane receptor, pregnane X receptor, farnesoid X receptor, androgen receptor, and peroxisome proliferator-activated receptor alpha. For these chemicals the network model translates transcriptomic data into Bayesian inferences about molecular targets known to activate toxicological adverse outcome pathways. These data also provide new insights into the molecular signaling network of HepaRGTM cell cultures.


Asunto(s)
Hepatocitos/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Toxicogenética/métodos , Teorema de Bayes , Técnicas de Cultivo de Célula , Línea Celular , Humanos , Hígado/citología , Bibliotecas de Moléculas Pequeñas , Factores de Transcripción/efectos de los fármacos , Transcriptoma/genética
13.
Lab Invest ; 90(12): 1690-703, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20697376

RESUMEN

Hedgehog (Hh) pathway activation promotes many processes that occur during fibrogenic liver repair. Whether the Hh pathway modulates the outcomes of virally mediated liver injury has never been examined. Gene-profiling studies of human hepatocellular carcinomas (HCCs) demonstrate Hh pathway activation in HCCs related to chronic infection with hepatitis B virus (HBV) or hepatitis C virus (HCV). Because most HCCs develop in cirrhotic livers, we hypothesized that Hh pathway activation occurs during fibrogenic repair of liver damage due to chronic viral hepatitis, and that Hh-responsive cells mediate disease progression and hepatocarciongenesis in chronic viral hepatitis. Immunohistochemistry and qRT-PCR analysis were used to analyze Hh pathway activation and identify Hh-responsive cell types in liver biopsies from 45 patients with chronic HBV or HCV. Hh signaling was then manipulated in cultured liver cells to directly assess the impact of Hh activity in relevant cell types. We found increased hepatic expression of Hh ligands in all patients with chronic viral hepatitis, and demonstrated that infection with HCV stimulated cultured hepatocytes to produce Hh ligands. The major cell populations that expanded during cirrhosis and HCC (ie, liver myofibroblasts, activated endothelial cells, and progenitors expressing markers of tumor stem/initiating cells) were Hh responsive, and higher levels of Hh pathway activity associated with cirrhosis and HCC. Inhibiting pathway activity in Hh-responsive target cells reduced fibrogenesis, angiogenesis, and growth. In conclusion, HBV/HCV infection increases hepatocyte production of Hh ligands and expands the types of Hh-responsive cells that promote liver fibrosis and cancer.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Proteínas Hedgehog/metabolismo , Hepatitis C Crónica/complicaciones , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Proteínas Virales/farmacología , Adulto , Anciano , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Células Cultivadas , Progresión de la Enfermedad , Femenino , Hepatitis C Crónica/patología , Virus de Hepatitis/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Transducción de Señal
14.
Gastroenterology ; 136(1): 320-330.e2, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19013163

RESUMEN

BACKGROUND & AIMS: Angiogenesis contributes to vascular remodeling during cirrhosis. In cirrhotic livers, cholangiocytes, and myofibroblastic hepatic stellate cells (MF-HSC) produce Hedgehog (Hh) ligands. During embryogenesis Hh ligands are released from ligand-producing cells in microparticles and activate Hh signaling in endothelial cells. We studied whether adult liver cell-derived microparticles contain Hh ligands that alter hepatic sinusoidal endothelial cells (SEC). METHODS: MF-HSC and cholangiocytes were exposed to platelet-derived growth factor to induce Hh ligands; microparticles were isolated from medium, analyzed by transmission electron microscopy and immunoblots, and applied to Hh-reporter-containing cells. Microparticles were obtained from serum and bile of rats after bile duct ligation (BDL) or sham surgery and applied to normal primary liver SEC with or without cyclopamine, an Hh signaling inhibitor. Effects on SEC gene expression were evaluated by quantitative reverse-transcription polymerase chain reaction and immunoblotting. Hh target gene expression and SEC activation markers were compared in primary SEC and in liver sections from healthy and BDL rats. RESULTS: Platelet-derived growth factor-treated MF-HSC and cholangiocytes released exosome-enriched microparticles containing biologically-active Hh ligands. BDL increased release of Hh-containing exosome-enriched microparticles into plasma and bile. Transmission electron microscopy and immunoblots revealed similarities among microparticles from all sources; all microparticles induced similar Hh-dependent changes in SEC gene expression. SEC from healthy livers did not express Hh target genes or activation markers, but both were up-regulated in SEC after BDL. CONCLUSIONS: Hh-containing exosome-enriched microparticles released from liver cells alter hepatic SEC gene expression, suggesting a novel mechanism for cirrhotic vasculopathy.


Asunto(s)
Micropartículas Derivadas de Células/fisiología , Células Endoteliales/fisiología , Proteínas Hedgehog/fisiología , Hepatocitos/fisiología , Cirrosis Hepática/patología , Transducción de Señal/fisiología , Animales , Becaplermina , Conductos Biliares/fisiología , Células Cultivadas , Expresión Génica , Células Estrelladas Hepáticas/fisiología , Hipertensión Portal/etiología , Ligadura , Masculino , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-sis , Ratas , Ratas Sprague-Dawley
15.
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
16.
Eur J Immunol ; 39(7): 1879-92, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19544307

RESUMEN

Lymphocyte accumulation is characteristic of chronic hepatitis, but the mechanisms regulating lymphocyte numbers and their roles in liver disease progression are poorly understood. The Hedgehog (Hh) pathway regulates thymic development and lymphopoeisis during embryogenesis, and is activated in fibrosing liver disease in adults. Our objective was to determine if Hh ligands regulate the viability and phenotype of NKT cells, which comprise a substantial sub-population of resident lymphocytes in healthy adult livers and often accumulate during liver fibrosis. The results demonstrate that a mouse invariant NKT cell line (DN32 iNKT cells), mouse primary liver iNKT cells, and human peripheral blood iNKT cells are all responsive to sonic hedgehog (Shh). In cultured iNKT cells, Shh enhances proliferation, inhibits apoptosis, induces activation, and stimulates expression of the pro-fibrogenic cytokine, IL-13. Livers of transgenic mice with an overly active Hh pathway harbor increased numbers of iNKT cells. iNKT cells also express Shh. These results demonstrate that iNKT cells produce and respond to Hh ligands, and that Hh pathway activation regulates the size and cytokine production of liver iNKT cell populations. Therefore, Hh pathway activation may contribute to the local expansion of pro-fibrogenic iNKT cell populations during certain types of fibrosing liver damage.


Asunto(s)
Proliferación Celular , Proteínas Hedgehog/metabolismo , Células T Asesinas Naturales/metabolismo , Transducción de Señal/fisiología , Animales , Conductos Biliares/citología , Conductos Biliares/inmunología , Western Blotting , Línea Celular , Células Cultivadas , Citotoxicidad Inmunológica , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Proteínas Hedgehog/genética , Proteínas Hedgehog/farmacología , Humanos , Hibridomas , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/inmunología , Receptores Patched , Receptores de Superficie Celular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
17.
Hepatology ; 50(2): 518-27, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19575365

RESUMEN

UNLABELLED: The mechanisms mediating hepatic accumulation of inflammatory cells in cholestatic liver disease remain enigmatic. Our thesis is that Hedgehog (Hh) pathway activation promotes hepatic accumulation of immune cells that interact with cholangiocytes. We believe that myofibroblastic hepatic stellate cells (MF-HSCs) release soluble Hh ligands that stimulate cholangiocytes to express chemokines that recruit mononuclear cell types with cognate receptors for these chemokines, thereby orchestrating a repair-related mechanism for liver inflammation. To address this thesis, we used three experimental systems that allow the definition of Hh-dependent mechanisms that induce phenotypic changes in cholangiocytes. First, cholangiocytes were cultured alone or in the presence of Hh-producing MF-HSCs in a transwell coculture system and/or treated with MF-HSC-conditioned medium with or without Hh-neutralizing antibodies. Changes in the cholangiocyte phenotype were then evaluated by microarray analysis, quantitative reverse-transcriptase polymerase chain reaction (QRT-PCR), and/or enzyme-linked immunosorbent assay for chemokine (C-X-C) motif ligand 16 (Cxcl16). Bile duct ligation was chosen to model biliary fibrosis in mice with an overly active Hh pathway, control littermates, and healthy rats, and the gene profile was evaluated by QRT-PCR in whole liver tissue. Second, a transwell chemotaxis assay was used to examine natural killer T (NKT) cell migration in response to cholangiocytes and particularly cholangiocyte-derived Cxcl16. Finally, we studied liver samples from primary biliary cirrhosis patients and controls by QRT-PCR to compare differences in the Hh pathway and Cxcl16. Co-immunostaining of cytokeratin-7 and Cxcl16 was then performed to localize the phenotypic source of Cxcl16. We found that MF-HSCs release soluble Hh ligands that stimulate cholangiocytes to produce Cxcl16 and recruit NKT cells. Hh pathway activation during cholestatic liver injury also induces cholangiocyte expression of Cxcl16. CONCLUSION: During biliary injury, Hh pathway activation induces cholangiocyte production of chemokines that recruit NKT cells to portal tracts.


Asunto(s)
Conductos Biliares/inmunología , Quimiocinas CXC/biosíntesis , Proteínas Hedgehog/metabolismo , Células T Asesinas Naturales/fisiología , Comunicación Paracrina , Receptores Depuradores/biosíntesis , Animales , Conductos Biliares/metabolismo , Conductos Biliares/cirugía , Estudios de Casos y Controles , Línea Celular , Quimiocina CXCL16 , Quimiocina CXCL6/metabolismo , Quimiocinas CXC/metabolismo , Quimiotaxis , Expresión Génica , Humanos , Ligadura , Cirrosis Hepática Biliar/metabolismo , Ratones , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344
18.
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
19.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1093-106, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19815628

RESUMEN

Myofibroblastic hepatic stellate cells (MF-HSC) are derived from quiescent hepatic stellate cells (Q-HSC). Q-HSC express certain epithelial cell markers and have been reported to form junctional complexes similar to epithelial cells. We have shown that Hedgehog (Hh) signaling plays a key role in HSC growth. Because Hh ligands regulate epithelial-to-mesenchymal transition (EMT), we determined whether Q-HSC express EMT markers and then assessed whether these markers change as Q-HSC transition into MF-HSC and whether the process is modulated by Hh signaling. Q-HSC were isolated from healthy livers and cultured to promote myofibroblastic transition. Changes in mRNA and protein expression of epithelial and mesenchymal markers, Hh ligands, and target genes were monitored in HSC treated with and without cyclopamine (an Hh inhibitor). Studies were repeated in primary human HSC and clonally derived HSC from a cirrhotic rat. Q-HSC activation in vitro (culture) and in vivo (CCl(4)-induced cirrhosis) resulted in decreased expression of Hh-interacting protein (Hhip, an Hh antagonist), the EMT inhibitors bone morphogenic protein (BMP-7) and inhibitor of differentiation (Id2), the adherens junction component E-cadherin, and epithelial keratins 7 and 19 and increased expression of Gli2 (an Hh target gene) and mesenchymal markers, including the mesenchyme-associated transcription factors Lhx2 and Msx2, the myofibroblast marker alpha-smooth muscle actin, and matrix molecules such as collagen. Cyclopamine reverted myofibroblastic transition, reducing mesenchymal gene expression while increasing epithelial markers in rodent and human HSC. We conclude that Hh signaling plays a key role in transition of Q-HSC into MF-HSC. Our findings suggest that Q-HSC are capable of transitioning between epithelial and mesenchymal fates.


Asunto(s)
Transdiferenciación Celular , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática Experimental/metabolismo , Hígado/metabolismo , Transducción de Señal , Animales , Tetracloruro de Carbono , Proliferación Celular , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/genética , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/genética , Cirrosis Hepática Experimental/patología , Regeneración Hepática , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Alcaloides de Veratrum/farmacología
20.
Gastroenterology ; 134(5): 1532-43, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18471524

RESUMEN

BACKGROUND & AIMS: Improving outcomes in alcoholic liver disease (ALD) necessitates better understanding of how habitual ethanol (EtOH) consumption alters normal regenerative mechanisms within the liver. Hedgehog (Hh) pathway activation promotes expansion of progenitor populations in other tissues. We evaluated the hypothesis that chronic EtOH exposure activates Hh signaling in liver. METHODS: Hh signaling, liver progenitors, transforming growth factor (TGF)-beta induction, and liver damage were compared in mice fed chow, high-fat diets (HF), or HF + EtOH for 4 weeks. Susceptibility to TGF-beta-mediated apoptosis was compared in Hh-responsive liver cells (eg, immature cholangiocytes and oval cells) and mature hepatocytes (which are unresponsive to Hh). Hepatic accumulation of Hh-responsive cells were compared in controls and ALD patients and correlated with a discriminant function (DF) that predicts subacute mortality. RESULTS: Hh signaling and numbers of Hh-responsive cells were increased in HF mice and greatest in HF+EtOH mice. In both, progenitor and stromal cell populations harbored Hh-responsive cells. More ductular-type progenitors and fibrosis markers were noted in HF+EtOH mice than in HF mice. The former also expressed more TGF-beta-1. TGF-beta-1 treatment selectively promoted the viability of Hh-responsive immature liver cells and caused mature hepatocytes that survived to produce Hh ligands. Hh-responsive cells were increased in ALD patients. Lobular accumulation of Hh-responsive immature ductular cells was greater in those with a DF >32 than those with a DF <32. CONCLUSIONS: Hh signaling is increased in ALD and may influence ALD outcomes by promoting hepatic accumulation of immature ductular cells.


Asunto(s)
Etanol/efectos adversos , Expresión Génica , Proteínas Hedgehog/metabolismo , Hepatopatías Alcohólicas/metabolismo , Regeneración Hepática/fisiología , ARN/genética , Factor de Crecimiento Transformador beta1/genética , Adulto , Animales , Biopsia , Recuento de Células , Células Cultivadas , Depresores del Sistema Nervioso Central/efectos adversos , Depresores del Sistema Nervioso Central/toxicidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Etanol/toxicidad , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inmunohistoquímica , Ligandos , Hepatopatías Alcohólicas/tratamiento farmacológico , Hepatopatías Alcohólicas/patología , Masculino , Ratones , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Índice de Severidad de la Enfermedad , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA