Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Pathol ; 186(3): 587-99, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26772417

RESUMEN

All serious liver injuries alter metabolism and initiate hepatic regeneration. Recent studies using partial hepatectomy (PH) and other experimental models of liver regeneration implicate the metabolic response to hepatic insufficiency as an important source of signals that promote regeneration. Based on these considerations, the analyses reported here were undertaken to assess the impact of interrupting the hypoglycemic response to PH on liver regeneration in mice. A regimen of parenteral dextrose infusion that delays PH-induced hypoglycemia for 14 hours after surgery was identified, and the hepatic regenerative response to PH was compared between dextrose-treated and control mice. The results showed that regenerative recovery of the liver was postponed in dextrose-infused mice (versus vehicle control) by an interval of time comparable to the delay in onset of PH-induced hypoglycemia. The regulation of specific liver regeneration-promoting signals, including hepatic induction of cyclin D1 and S-phase kinase-associated protein 2 expression and suppression of peroxisome proliferator-activated receptor γ and p27 expression, was also disrupted by dextrose infusion. These data support the hypothesis that alterations in metabolism that occur in response to hepatic insufficiency promote liver regeneration, and they define specific pro- and antiregenerative molecular targets whose regenerative regulation is postponed when PH-induced hypoglycemia is delayed.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/administración & dosificación , Insuficiencia Hepática/prevención & control , Hipoglucemia/tratamiento farmacológico , Regeneración Hepática/efectos de los fármacos , Animales , Ciclina D1/genética , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Hepatectomía/efectos adversos , Concentración de Iones de Hidrógeno , Hipoglucemia/etiología , Hígado/citología , Hígado/metabolismo , Hígado/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , PPAR gamma/genética , PPAR gamma/metabolismo , Fosforilación , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo
2.
Hepatology ; 64(5): 1587-1605, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27097116

RESUMEN

Hepatocellular cancer (HCC) remains a significant therapeutic challenge due to its poorly understood molecular basis. In the current study, we investigated two independent cohorts of 249 and 194 HCC cases for any combinatorial molecular aberrations. Specifically we assessed for simultaneous HMET expression or hMet activation and catenin ß1 gene (CTNNB1) mutations to address any concomitant Met and Wnt signaling. To investigate cooperation in tumorigenesis, we coexpressed hMet and ß-catenin point mutants (S33Y or S45Y) in hepatocytes using sleeping beauty transposon/transposase and hydrodynamic tail vein injection and characterized tumors for growth, signaling, gene signatures, and similarity to human HCC. Missense mutations in exon 3 of CTNNB1 were identified in subsets of HCC patients. Irrespective of amino acid affected, all exon 3 mutations induced similar changes in gene expression. Concomitant HMET overexpression or hMet activation and CTNNB1 mutations were evident in 9%-12.5% of HCCs. Coexpression of hMet and mutant-ß-catenin led to notable HCC in mice. Tumors showed active Wnt and hMet signaling with evidence of glutamine synthetase and cyclin D1 positivity and mitogen-activated protein kinase/extracellular signal-regulated kinase, AKT/Ras/mammalian target of rapamycin activation. Introduction of dominant-negative T-cell factor 4 prevented tumorigenesis. The gene expression of mouse tumors in hMet-mutant ß-catenin showed high correlation, with subsets of human HCC displaying concomitant hMet activation signature and CTNNB1 mutations. CONCLUSION: We have identified cooperation of hMet and ß-catenin activation in a subset of HCC patients and modeled this human disease in mice with a significant transcriptomic intersection; this model will provide novel insight into the biology of this tumor and allow us to evaluate novel therapies as a step toward precision medicine. (Hepatology 2016;64:1587-1605).


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Mutación Puntual , Proteínas Proto-Oncogénicas c-met/genética , beta Catenina/genética , Animales , Regulación Neoplásica de la Expresión Génica , Masculino , Ratones , Modelos Biológicos
3.
J Hepatol ; 64(2): 316-325, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26505118

RESUMEN

BACKGROUND & AIMS: During liver regeneration, hepatocytes are derived from pre-existing hepatocytes. However, if hepatocyte proliferation is compromised, biliary epithelial cells (BECs) become the source of new hepatocytes. We recently reported on a zebrafish liver regeneration model in which BECs extensively contribute to hepatocytes. Using this model, we performed a targeted chemical screen to identify important factors that regulate BEC-driven liver regeneration, the mechanisms of which remain largely unknown. METHODS: Using Tg(fabp10a:CFP-NTR) zebrafish, we examined the effects of 44 selected compounds on BEC-driven liver regeneration. Liver size was assessed by fabp10a:DsRed expression; liver marker expression was analyzed by immunostaining, in situ hybridization and quantitative PCR. Proliferation and apoptosis were also examined. Moreover, we used a mouse liver injury model, choline-deficient, ethionine-supplemented (CDE) diet. RESULTS: We identified 10 compounds that affected regenerating liver size. Among them, only bromodomain and extraterminal domain (BET) inhibitors, JQ1 and iBET151, blocked both Prox1 and Hnf4a induction in BECs. BET inhibition during hepatocyte ablation blocked BEC dedifferentiation into hepatoblast-like cells (HB-LCs). Intriguingly, after JQ1 washout, liver regeneration resumed, indicating temporal, but not permanent, perturbation of liver regeneration by BET inhibition. BET inhibition after hepatocyte ablation suppressed the proliferation of newly generated hepatocytes and delayed hepatocyte maturation. Importantly, Myca overexpression, in part, rescued the proliferation defect. Furthermore, oval cell numbers in mice fed CDE diet were greatly reduced upon JQ1 administration, supporting the zebrafish findings. CONCLUSIONS: BET proteins regulate BEC-driven liver regeneration at multiple steps: BEC dedifferentiation, HB-LC proliferation, the proliferation of newly generated hepatocytes, and hepatocyte maturation.


Asunto(s)
Azepinas/metabolismo , Células Epiteliales/fisiología , Hepatocitos/fisiología , Compuestos Heterocíclicos de 4 o más Anillos/metabolismo , Regeneración Hepática/fisiología , Triazoles/metabolismo , Animales , Sistema Biliar/patología , Línea Celular , Proliferación Celular/fisiología , Transdiferenciación Celular/fisiología , Hígado/metabolismo , Hígado/patología , Ratones , Tamaño de los Órganos , Factores de Transcripción/antagonistas & inhibidores , Activación Transcripcional/fisiología , Pez Cebra
4.
Gene Expr ; 17(1): 19-34, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27226410

RESUMEN

Triiodothyronine (T3) induces hepatocyte proliferation in rodents. Recent work has shown molecular mechanism for T3's mitogenic effect to be through activation of ß-catenin signaling. Since systemic side effects of T3 may preclude its clinical use, and hepatocytes mostly express T3 hormone receptor ß (TRß), we investigated if selective TRß agonists like GC-1 may also have ß-catenin-dependent hepatocyte mitogenic effects. Here we studied the effect of GC-1 and T3 in conditional knockouts of various Wnt pathway components. We also assessed any regenerative advantage of T3 or GC-1 when given prior to partial hepatectomy in mice. Mice administered GC-1 showed increased pSer675-ß-catenin, cyclin D1, BrdU incorporation, and PCNA. No abnormalities in liver function tests were noted. GC-1-injected liver-specific ß-catenin knockouts (ß-catenin LKO) showed decreased proliferation when compared to wild-type littermates. To address if Wnt signaling was required for T3- or GC-1-mediated hepatocyte proliferation, we used LRP5-6-LKO, which lacks the two redundant Wnt coreceptors. Surprisingly, decreased hepatocyte proliferation was also evident in LRP5-6-LKO in response to T3 and GC-1, despite increased pSer675-ß-catenin. Further, increased levels of active ß-catenin (hypophosphorylated at Ser33, Ser37, and Thr41) were evident after T3 and GC-1 treatment. Finally, mice pretreated with T3 or GC-1 for 7 days followed by partial hepatectomy showed a significant increase in hepatocyte proliferation both at the time (T0) and 24 h after surgery. In conclusion, like T3, TRß-selective agonists induce hepatocyte proliferation through ß-catenin activation via both PKA- and Wnt-dependent mechanisms and confer a regenerative advantage following surgical resection. Hence, these agents may be useful regenerative therapies in liver transplantation or other surgical settings.


Asunto(s)
Acetatos/farmacología , Proliferación Celular/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Regeneración Hepática/efectos de los fármacos , Fenoles/farmacología , Receptores beta de Hormona Tiroidea/agonistas , Triyodotironina/farmacología , beta Catenina/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hepatectomía/métodos , Hepatocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores beta de Hormona Tiroidea/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
5.
J Hepatol ; 62(2): 380-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25457204

RESUMEN

BACKGROUND & AIMS: Hepatocellular cancer (HCC) remains a disease of poor prognosis, highlighting the relevance of elucidating key molecular aberrations that may be targeted for novel therapies. Wnt signalling activation, chiefly due to mutations in CTNNB1, have been identified in a major subset of HCC patients. While several in vitro proof of concept studies show the relevance of suppressing Wnt/ß-catenin signalling in HCC cells or tumour xenograft models, no study has addressed the impact of ß-catenin inhibition in a relevant murine HCC model driven by Ctnnb1 mutations. METHODS: We studied the in vivo impact of ß-catenin suppression by locked nucleic acid (LNA) antisense treatment, after establishing Ctnnb1 mutation-driven HCC by diethylnitrosamine and phenobarbital (DEN/PB) administration. RESULTS: The efficacy of LNA directed against ß-catenin vs. scrambled on Wnt signalling was demonstrated in vitro in HCC cells and in vivo in normal mice. The DEN/PB model leads to HCC with Ctnnb1 mutations. A complete therapeutic response in the form of abrogation of HCC was observed after ten treatments of tumour-bearing mice with ß-catenin LNA every 48h as compared to the scrambled control. A decrease in ß-catenin activity, cell proliferation and increased cell death was evident after ß-catenin suppression. No effect of ß-catenin suppression was evident in non-Ctnnb1 mutated HCC, observed after DEN-only administration. CONCLUSIONS: Thus, we provide the in vivo proof of concept that ß-catenin suppression in HCC will be of significant therapeutic benefit, provided the tumours display Wnt activation via mechanisms like CTNNB1 mutations.


Asunto(s)
Carcinogénesis/genética , Carcinoma Hepatocelular/genética , ADN de Neoplasias/genética , Neoplasias Hepáticas Experimentales/genética , Mutación , Oligonucleótidos/metabolismo , beta Catenina/genética , Alquilantes/uso terapéutico , Animales , Western Blotting , Carcinogénesis/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Análisis Mutacional de ADN , Dietilnitrosamina/farmacología , Ensayo de Inmunoadsorción Enzimática , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Inmunohistoquímica , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Fenobarbital/farmacología , Células Tumorales Cultivadas , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo
6.
Gastroenterology ; 147(3): 690-701, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24837480

RESUMEN

BACKGROUND & AIMS: Aberrant activation of ß-catenin and Yes-associated protein 1 (Yap1) signaling pathways have been associated with the development of multiple tumor types. Yap functions as a transcriptional coactivator by interacting with TEA domain DNA binding proteins. We investigated the interactions among these pathways during hepatic tumorigenesis. METHODS: We used immunohistochemical analysis to determine expression of ß-catenin and Yap1 in liver cancer specimens collected from patients in Europe and the United States, consisting of 104 hepatocellular carcinoma, 62 intrahepatic cholangiocarcinoma, and 94 hepatoblastoma samples. We assessed ß-catenin and Yap1 signaling and interactions in hepatoblastoma cell lines ((HuH6, HepG2, HepT1, HC-AFW1, HepG2, and HC-AFW1); proteins were knocked down with small interfering RNAs, and effects on proliferation and cell death were measured. Sleeping beauty-mediated hydrodynamic transfection was used to overexpress constitutively active forms of ß-catenin (ΔN90/ß-catenin) and Yap1 (YapS127A) in livers of mice; tissues were collected, and histological and immunohistochemical analyses were performed. RESULTS: We observed nuclear localization of ß-catenin and Yap1 in 79% of hepatoblastoma samples but not in most hepatocellular carcinoma or intrahepatic cholangiocarcinoma samples. Yap1 and ß-catenin coprecipitated in hepatoblastoma but not hepatocellular carcinoma cells. Small interfering RNA-mediated knockdown of Yap1 or ß-catenin in hepatoblastoma cells reduced proliferation in an additive manner. Knockdown of Yap1 reduced its ability to coactivate transcription with ß-catenin; ß-catenin inhibitors inactivated Yap1. Overexpression of constitutively active forms of Yap1 and ß-catenin in mouse liver led to rapid tumorigenesis, with 100% mortality by 11 weeks. Tumor cells expressed both proteins, and human hepatoblastoma cells expressed common targets of their 2 signaling pathways. Yap1 binding of TEA domain factors was required for tumorigenesis in mice. CONCLUSIONS: ß-catenin and the transcriptional regulator Yap1 interact physically and are activated in most human hepatoblastoma tissues; overexpression of activated forms of these proteins in livers of mice leads to rapid tumor development. Further analysis of these mice will allow further studies of these pathways in hepatoblastoma pathogenesis and could lead to the identification of new therapeutic targets.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transformación Celular Neoplásica/metabolismo , Hepatoblastoma/metabolismo , Neoplasias Hepáticas/metabolismo , Fosfoproteínas/metabolismo , beta Catenina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Muerte Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Europa (Continente) , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Hepatoblastoma/genética , Hepatoblastoma/patología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Fosfoproteínas/genética , Unión Proteica , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Factores de Transcripción , Transcripción Genética , Transfección , Estados Unidos , Proteínas Señalizadoras YAP , beta Catenina/genética
7.
Am J Pathol ; 184(2): 306-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24418096

RESUMEN

This Guest Editorial introduces this month's special Liver Pathobiology Theme Issue, a series of reviews that encompass the discipline of hepatic regenerative medicine.


Asunto(s)
Regeneración Hepática , Medicina Regenerativa , Matriz Extracelular/metabolismo , Hepatocitos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Hígado/citología , Hígado/metabolismo , Poliploidía , Ingeniería de Tejidos
8.
Am J Pathol ; 184(11): 3013-25, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25193591

RESUMEN

Acetaminophen (APAP) overdose results in acute liver failure and has limited treatment options. Previous studies show that stimulating liver regeneration is critical for survival after APAP overdose, but the mechanisms remain unclear. In this study, we identified major signaling pathways involved in liver regeneration after APAP-induced acute liver injury using a novel incremental dose model. Liver injury and regeneration were studied in C57BL/6 mice treated with either 300 mg/kg (APAP300) or 600 mg/kg (APAP600) APAP. Mice treated with APAP300 developed extensive liver injury and robust liver regeneration. In contrast, APAP600-treated mice exhibited significant liver injury but substantial inhibition of liver regeneration, resulting in sustained injury and decreased survival. The inhibition of liver regeneration in the APAP600 group was associated with cell cycle arrest and decreased cyclin D1 expression. Several known regenerative pathways, including the IL-6/STAT-3 and epidermal growth factor receptor/c-Met/mitogen-activated protein kinase pathways, were activated, even at APAP600, where regeneration was inhibited. However, canonical Wnt/ß-catenin and NF-κB pathways were activated only in APAP300-treated mice, where liver regeneration was stimulated. Furthermore, overexpression of a stable form of ß-catenin, where serine 45 is mutated to aspartic acid, in mice resulted in improved liver regeneration after APAP overdose. Taken together, our incremental dose model has identified a differential role of several signaling pathways in liver regeneration after APAP overdose and highlighted canonical Wnt signaling as a potential target for regenerative therapies for APAP-induced acute liver failure.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Regeneración Hepática/fisiología , Transducción de Señal/fisiología , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
9.
J Biol Chem ; 288(29): 21389-21398, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23740243

RESUMEN

Tissue regeneration requires the activation of a set of specific growth signaling pathways. The identity of these cascades and their biological roles are known; however, the molecular mechanisms regulating the interplay between these pathways remain poorly understood. Here, we define a new role for SULFATASE 2 (SULF2) in regulating tissue regeneration and define the WNT-GLI1 axis as a novel downstream effector for this sulfatase in a liver model of tissue regeneration. SULF2 is a heparan sulfate 6-O-endosulfatase, which releases growth factors from extracellular storage sites turning active multiple signaling pathways. We demonstrate that SULF2-KO mice display delayed regeneration after partial hepatectomy (PH). Mechanistic analysis of the SULF2-KO phenotype showed a decrease in WNT signaling pathway activity in vivo. In isolated hepatocytes, SULF2 deficiency blocked WNT-induced ß-CATENIN nuclear translocation, TCF activation, and proliferation. Furthermore, we identified the transcription factor GLI1 as a novel target of the SULF2-WNT cascade. WNT induces GLI1 expression in a SULF2- and ß-CATENIN-dependent manner. GLI1-KO mice phenocopied the SULF2-KO, showing delayed regeneration and decreased hepatocyte proliferation. Moreover, we identified CYCLIN D1, a key mediator of cell growth during tissue regeneration, as a GLI1 transcriptional target. GLI1 binds to the cyclin d1 promoter and regulates its activity and expression. Finally, restoring GLI1 expression in the liver of SULF2-KO mice after PH rescues CYCLIN D1 expression and hepatocyte proliferation to wild-type levels. Thus, together these findings define a novel pathway in which SULF2 regulates tissue regeneration in part via the activation of a novel WNT-GLI1-CYCLIN D1 pathway.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Regeneración Hepática , Sulfatasas/metabolismo , Vía de Señalización Wnt , Animales , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Hepatectomía , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Regeneración Hepática/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Sulfatasas/deficiencia , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt3A/farmacología , Proteína con Dedos de Zinc GLI1 , beta Catenina/metabolismo
10.
Am J Pathol ; 182(5): 1648-58, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23529017

RESUMEN

Aberrant platelet-derived growth factor receptor-α (PDGFRα) signaling is evident in a subset of hepatocellular cancers (HCCs). However, its role and regulation in hepatic physiology remains elusive. In the current study, we examined PDGFRα signaling in liver development and regeneration. We identified notable PDGFRα activation in hepatic morphogenesis that, when interrupted by PDGFRα-blocking antibody, led to decreased hepatoblast proliferation and survival in embryonic liver cultures. We also identified temporal PDGFRα overexpression, which is regulated by epidermal growth factor (EGF) and tumor necrosis factor α, and its activation at 3 to 24 hours after partial hepatectomy. Through generation of hepatocyte-specific PDGFRA knockout (KO) mice that lack an overt phenotype, we show that absent PDGFRα compromises extracelluar signal-regulated kinases and AKT activation 3 hours after partial hepatectomy, which, however, is alleviated by temporal compensatory increases in the EGF receptor (EGFR) and the hepatocyte growth factor receptor (Met) expression and activation along with rebound activation of extracellular signal-regulated kinases and AKT at 24 hours. These untimely increases in EGFR and Met allow for normal hepatocyte proliferation at 48 hours in KO, which, however, are aberrantly prolonged up to 72 hours. Intriguingly, such compensation also was visible in primary KO hepatocyte cultures but not in HCC cells after siRNA-mediated PDGFRα knockdown. Thus, temporal activation of PDGFRα in liver development is important in hepatic morphogenesis. In liver regeneration, despite increased signaling, PDGFRα is dispensable owing to EGFR and Met compensation, which is unique to normal hepatocytes but not HCC cells.


Asunto(s)
Regeneración Hepática , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Receptores ErbB/metabolismo , Femenino , Hepatectomía , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hígado/embriología , Hígado/cirugía , Masculino , Ratones , Ratones Noqueados , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Factores de Tiempo , Técnicas de Cultivo de Tejidos
11.
Hepatology ; 57(2): 763-74, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22941935

RESUMEN

UNLABELLED: Wnt/ß-catenin signaling plays an important role in hepatic homeostasis, especially in liver development, regeneration, and cancer, and loss of ß-catenin signaling is often associated with increased apoptosis. To elucidate how ß-catenin may be regulating hepatocyte survival, we investigated the susceptibility of ß-catenin conditional knockout (KO) mice and their wild-type (WT) littermates to Fas and tumor necrosis factor-α (TNF-α), two common pathways of hepatocyte apoptosis. While comparable detrimental effects from Fas activation were observed in WT and KO, a paradoxical survival benefit was observed in KO mice challenged with D-galactosamine/lipopolysaccharide. KO mice showed significantly lower morbidity and liver injury due to early, robust, and protracted activation of NF-κB in the absence of ß-catenin. Enhanced NF-κB activation in KO mice was associated with increased basal inflammation and Toll-like receptor 4 expression and lack of the p65/ß-catenin complex in hepatocytes. The p65/ß-catenin complex in WT livers underwent temporal dissociation allowing for NF-κB activation to regulate hepatocyte survival following TNF-α-induced hepatic injury. Decrease of total ß-catenin protein but not its inactivation induced p65 activity, whereas ß-catenin stabilization either chemically or due to mutations repressed it in hepatomas in a dose-dependent manner, whereas ß-catenin stabilization repressed it either chemically or due to mutations. CONCLUSION: The p65/ß-catenin complex in hepatocytes undergoes dynamic changes during TNF-α-induced hepatic injury and plays a critical role in NF-κB activation and cell survival. Modulation of ß-catenin levels is a unique mode of regulating NF-κB activity and thus may present novel opportunities in devising therapeutics in specific hepatic injuries.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , FN-kappa B/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Hepatocitos/metabolismo , Hepatocitos/patología , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Transducción de Señal/fisiología , Receptor Toll-Like 4/fisiología , Factor de Transcripción ReIA/fisiología , beta Catenina/genética , beta Catenina/fisiología , Receptor fas/fisiología
12.
Nat Commun ; 15(1): 5442, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937436

RESUMEN

Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.


Asunto(s)
Antígenos de Neoplasias , Antígeno B7-H1 , Linfocitos T CD8-positivos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Viroterapia Oncolítica , Virus Oncolíticos , Microambiente Tumoral , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Animales , Viroterapia Oncolítica/métodos , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/inmunología , Microambiente Tumoral/inmunología , Ratones , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Humanos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Interferón beta/metabolismo , Interferón beta/inmunología , Ratones Endogámicos C57BL , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos T/inmunología , Femenino , Vesiculovirus/inmunología , Vesiculovirus/genética
13.
J Biol Chem ; 287(27): 22789-98, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22613727

RESUMEN

Hepatic competence, specification, and liver bud expansion during development depend on precise temporal modulation of the Wnt/ß-catenin signaling. Also, loss- and gain-of-function studies have revealed pleiotropic roles of ß-catenin in proliferation and hepatocyte and biliary epithelial cell differentiation, but precise mechanisms remain unknown. Here we utilize livers from different stages of murine development to determine ß-catenin signaling and downstream targets. Although during early liver development full-length ß-catenin is the predominant form, at late stages, where full-length ß-catenin localizes to developing biliary epithelial cells only, a 75-kDa truncated ß-catenin species is the principal form localizing at the membrane and in the nucleus of differentiating hepatocytes. The truncated species lacks 95 N-terminal amino acids and is transcriptionally active. Our evidence points to proteolytic cleavage of ß-catenin by calpain as the mechanism of truncation in cell-free and cell-based assays. Intraperitoneal injection of a short term calpain inhibitor to timed pregnant female mice abrogated ß-catenin truncation in the embryonic livers. RNA-seq revealed a unique set of targets transcribed in cells expressing truncated versus full-length ß-catenin, consistent with different functionalities. A further investigation using N- and C-terminal-specific ß-catenin antibodies on human hepatoblastomas revealed a correlation between full-length versus truncated ß-catenin and differentiation status, with embryonal hepatoblastomas expressing full-length ß-catenin and fetal hepatoblastomas expressing ß-catenin lacking its N terminus. Thus we conclude that calpain-mediated cleavage of ß-catenin plays a role in regulating hepatoblast differentiation in mouse and human liver, and the presence of the ß-catenin N terminus correlates with differentiation status in hepatoblastomas.


Asunto(s)
Calpaína/metabolismo , Hepatoblastoma/diagnóstico , Neoplasias Hepáticas Experimentales/diagnóstico , beta Catenina/metabolismo , Empalme Alternativo/fisiología , Animales , Calpaína/antagonistas & inhibidores , Diferenciación Celular/fisiología , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Femenino , Células HEK293 , Hepatoblastoma/metabolismo , Hepatocitos/citología , Hepatocitos/enzimología , Humanos , Hígado/embriología , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo , Estructura Terciaria de Proteína , beta Catenina/química , beta Catenina/genética
14.
Hepatology ; 55(3): 931-40, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22031168

RESUMEN

UNLABELLED: The liver plays a central role in ethanol metabolism, and oxidative stress is implicated in alcohol-mediated liver injury. ß-Catenin regulates hepatic metabolic zonation and adaptive response to oxidative stress. We hypothesized that ß-catenin regulates the hepatic response to ethanol ingestion. Female liver-specific ß-catenin knockout (KO) mice and wild-type (WT) littermates were fed the Lieber-Decarli liquid diet (5% ethanol) in a pairwise fashion. Liver histology, biochemistry, and gene-expression studies were performed. Plasma alcohol and ammonia levels were measured using standard assays. Ethanol-fed (EtOH) KO mice exhibited systemic toxicity and early mortality. KO mice exhibited severe macrovesicular steatosis and 5 to 6-fold higher serum alanine aminotransferase and aspartate aminotransferase levels. KO mice had a modest increase in hepatic oxidative stress, lower expression of mitochondrial superoxide dismutase (SOD2), and lower citrate synthase activity, the first step in the tricarboxylic acid cycle. N-Acetylcysteine did not prevent ethanol-induced mortality in KO mice. In WT livers, ß-catenin was found to coprecipitate with forkhead box O3, the upstream regulator of SOD2. Hepatic alcohol dehydrogenase and aldehyde dehydrogenase activities and expression were lower in KO mice. Hepatic cytochrome P450 2E1 protein levels were up-regulated in EtOH WT mice, but were nearly undetectable in KO mice. These changes in ethanol-metabolizing enzymes were associated with 30-fold higher blood alcohol levels in KO mice. CONCLUSION: ß-Catenin is essential for hepatic ethanol metabolism and plays a protective role in alcohol-mediated liver steatosis. Our results strongly suggest that integration of these functions by ß-catenin is critical for adaptation to ethanol ingestion in vivo.


Asunto(s)
Etanol/efectos adversos , Etanol/metabolismo , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Hígado/metabolismo , beta Catenina/metabolismo , Alanina Transaminasa/sangre , Amoníaco/sangre , Animales , Aspartato Aminotransferasas/sangre , Modelos Animales de Enfermedad , Etanol/farmacología , Hígado Graso/mortalidad , Femenino , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Superóxido Dismutasa/metabolismo , beta Catenina/deficiencia , beta Catenina/genética
15.
Hepatology ; 55(6): 1863-75, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22234969

RESUMEN

UNLABELLED: Hypoxia is often found in solid tumors and is associated with tumor progression and poor clinical outcomes. The exact mechanisms related to hypoxia-induced invasion and metastasis remain unclear. We elucidated the mechanism by which the nuclear-damage-associated molecular pattern molecule, high-mobility group box 1 (HMGB1), released under hypoxic stress, can induce an inflammatory response to promote invasion and metastasis in hepatocellular carcinoma (HCC) cells. Caspase-1 activation was found to occur in hypoxic HCC cells in a process that was dependent on the extracellular release of HMGB1 and subsequent activation of both Toll-like receptor 4 (TLR4)- and receptor for advanced glycation endproducts (RAGE)-signaling pathways. Downstream from hypoxia-induced caspase-1 activation, cleavage and release of proinflammatory cytokines interleukin (IL)-1ß and -18 occurred. We further demonstrate that overexpression of HMGB1 or treatment with recombinant HMGB1 enhanced the invasiveness of HCC cells, whereas stable knockdown of HMGB1 remarkably reduced HCC invasion. Moreover, in a murine model of HCC pulmonary metastasis, stable knockdown of HMGB1 suppressed HCC invasion and metastasis. CONCLUSION: These results suggest that in hypoxic HCC cells, HMGB1 activates TLR4- and RAGE-signaling pathways to induce caspase-1 activation with the subsequent production of multiple inflammatory mediators, which, in turn, promote cancer invasion and metastasis.


Asunto(s)
Carcinoma Hepatocelular/patología , Caspasa 1/metabolismo , Proteína HMGB1/fisiología , Neoplasias Hepáticas/patología , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Activación Enzimática , Humanos , Interleucina-1beta/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/fisiología , Transducción de Señal , Receptor Toll-Like 4/fisiología
16.
Protein Expr Purif ; 87(2): 111-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23137940

RESUMEN

Expression of recombinant proteins in bacterial or eukaryotic systems often results in aggregation rendering them unavailable for biochemical or structural studies. Protein aggregation is a costly problem for biomedical research. It forces research laboratories and the biomedical industry to search for alternative, more soluble, non-human proteins and limits the number of potential "druggable" targets. In this study we present a highly reproducible protocol that introduces the systematic use of an extensive number of detergents to solubilize aggregated proteins expressed in bacterial and eukaryotic systems. We validate the usefulness of this protocol by solubilizing traditionally difficult human protein targets to milligram quantities and confirm their biological activity. We use this method to solubilize monomeric or multimeric components of multi-protein complexes and demonstrate its efficacy to reconstitute large cellular machines. This protocol works equally well on cytosolic, nuclear and membrane proteins and can be easily adapted to a high throughput format.


Asunto(s)
Biotecnología/métodos , Detergentes/química , Proteínas de la Membrana/aislamiento & purificación , Complejos Multiproteicos/aislamiento & purificación , Proteínas Recombinantes/aislamiento & purificación , Electroforesis en Gel de Poliacrilamida , Escherichia coli , Proteínas de la Membrana/química , Complejos Multiproteicos/química , Proteínas Recombinantes/química , Saccharomyces cerevisiae , Células Sf9 , Solubilidad
17.
Semin Cancer Biol ; 21(1): 44-58, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21182948

RESUMEN

Among the adult organs, liver is unique for its ability to regenerate. A concerted signaling cascade enables optimum initiation of the regeneration process following insults brought about by surgery or a toxicant. Additionally, there exists a cellular redundancy, whereby a transiently amplifying progenitor population appears and expands to ensure regeneration, when differentiated cells of the liver are unable to proliferate in both experimental and clinical scenarios. One such pathway of relevance in these phenomena is Wnt/ß-catenin signaling, which is activated relatively early during regeneration mostly through post-translational modifications. Once activated, ß-catenin signaling drives the expression of target genes that are critical for cell cycle progression and contribute to initiation of the regeneration process. The role and regulation of Wnt/ß-catenin signaling is now documented in rats, mice, zebrafish and patients. More recently, a regenerative advantage of the livers in ß-catenin overexpressing mice was reported, as was also the case after exogenous Wnt-1 delivery to the liver paving the way for assessing means to stimulate the pathway for therapeutics in liver failure. ß-Catenin is also pertinent in hepatic oval cell activation and differentiation. However, aberrant activation of the Wnt/ß-catenin signaling is reported in a significant subset of hepatocellular cancers (HCC). While many mechanisms of such activation have been reported, the most functional means of aberrant and sustained activation is through mutations in the ß-catenin gene or in AXIN1/2, which encodes for a scaffolding protein critical for ß-catenin degradation. Intriguingly, in experimental models hepatic overexpression of normal or mutant ß-catenin is insufficient for tumorigenesis. In fact ß-catenin loss promoted chemical carcinogenesis in the liver due to alternate mechanisms. Since most HCC occur in the backdrop of chronic hepatic injury, where hepatic regeneration is necessary for maintenance of liver function, but at the same time serves as the basis of dysplastic changes, this Promethean attribute exhibits a Jekyll and Hyde behavior that makes distinguishing good regeneration from bad regeneration essential for targeting selective molecular pathways as personalized medicine becomes a norm in clinical practice. Could ß-catenin signaling be one such pathway that may be redundant in regeneration and indispensible in HCC in a subset of cases?


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Regeneración Hepática , Transducción de Señal , beta Catenina/metabolismo , Animales , Carcinoma Hepatocelular/patología , Humanos , Neoplasias Hepáticas/patología , Células Madre/citología , Células Madre/metabolismo , Proteínas Wnt/metabolismo
18.
Hepatol Commun ; 7(9)2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37556373

RESUMEN

BACKGROUND: We previously showed that loss of yes-associated protein 1 (YAP) in early liver development (YAPKO) leads to an Alagille syndrome-like phenotype, with failure of intrahepatic bile duct development, severe cholestasis, and chronic hepatocyte adaptations to reduce liver injury. TAZ, a paralog of YAP, was significantly upregulated in YAPKO hepatocytes and interacted with TEA domain family member (TEAD) transcription factors, suggesting possible compensatory activity. METHODS: We deleted both Yap1 and Wwtr1 (which encodes TAZ) during early liver development using the Foxa3 promoter to drive Cre expression, similar to YAPKO mice, resulting in YAP/TAZ double knockout (DKO) and YAPKO with TAZ heterozygosity (YAPKO TAZHET). We evaluated these mice using immunohistochemistry, serum biochemistry, bile acid profiling, and RNA sequencing. RESULTS: DKO mice were embryonic lethal, but their livers were similar to YAPKO, suggesting an extrahepatic cause of death. Male YAPKO TAZHET mice were also embryonic lethal, with insufficient samples to determine the cause. However, YAPKO TAZHET females survived and were phenotypically similar to YAPKO mice, with increased bile acid hydrophilicity and similar global gene expression adaptations but worsened the hepatocellular injury. TAZ heterozygosity in YAPKO impacted the expression of canonical YAP targets Ctgf and Cyr61, and we found changes in pathways regulating cell division and inflammatory signaling correlating with an increase in hepatocyte cell death, cell cycling, and macrophage recruitment. CONCLUSIONS: YAP loss (with or without TAZ loss) aborts biliary development. YAP and TAZ play a codependent critical role in foregut endoderm development outside the liver, but they are not essential for hepatocyte development. TAZ heterozygosity in YAPKO livers increased cell cycling and inflammatory signaling in the setting of chronic injury, highlighting genes that are especially sensitive to TAZ regulation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Carcinoma Hepatocelular , Colestasis , Neoplasias Hepáticas , Proteínas Señalizadoras YAP , Animales , Masculino , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Endodermo/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Transactivadores/metabolismo , Factores de Transcripción/genética , Proteínas Señalizadoras YAP/genética , Femenino
19.
Hepatology ; 54(4): 1333-43, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21721031

RESUMEN

UNLABELLED: Prolonged exposure of mice to diet containing 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) results in hepatobiliary injury, atypical ductular proliferation, oval cell appearance, and limited fibrosis. Previously, we reported that short-term ingestion of DDC diet by hepatocyte-specific ß-catenin conditional knockout (KO) mice led to fewer A6-positive oval cells than wildtype (WT) littermates. To examine the role of ß-catenin in chronic hepatic injury and repair, we exposed WT and KO mice to DDC for 80 and 150 days. Paradoxically, long-term DDC exposure led to significantly more A6-positive cells, indicating greater atypical ductular proliferation in KO, which coincided with increased fibrosis and cholestasis. Surprisingly, at 80 and 150 days in KO we observed a significant amelioration of hepatocyte injury. This coincided with extensive repopulation of ß-catenin null livers with ß-catenin-positive hepatocytes at 150 days, which was preceded by appearance of ß-catenin-positive hepatocyte clusters at 80 days and a few ß-catenin-positive hepatocytes at earlier times. Intriguingly, occasional ß-catenin-positive hepatocytes that were negative for progenitor markers were also observed at baseline in the KO livers, suggesting spontaneous escape from cre-mediated recombination. These cells with hepatocyte morphology expressed mature hepatocyte markers but lacked markers of hepatic progenitors. The gradual repopulation of KO livers with ß-catenin-positive hepatocytes occurred only following DDC injury and coincided with a progressive loss of hepatic cre-recombinase expression. A few ß-catenin-positive cholangiocytes were observed albeit only after long-term DDC exposure and trailed the appearance of ß-catenin-positive hepatocytes. CONCLUSION: In a chronic liver injury model, ß-catenin-positive hepatocytes exhibit growth and survival advantages and repopulate KO livers, eventually limiting hepatic injury and dysfunction despite increased fibrosis and intrahepatic cholestasis.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/patología , Hepatocitos/efectos de los fármacos , Piridinas/farmacología , beta Catenina/metabolismo , Animales , Western Blotting , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Hepatocitos/patología , Inmunohistoquímica , Pruebas de Función Hepática , Regeneración Hepática/fisiología , Ratones , Ratones Noqueados , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
Dev Dyn ; 240(3): 486-500, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21337461

RESUMEN

The Wnt/ß-catenin pathway is an evolutionarily conserved signaling cascade that plays key roles in development and adult tissue homeostasis and is aberrantly activated in many tumors. Over a decade of work in mouse, chick, xenopus, and zebrafish models has uncovered multiple functions of this pathway in hepatic pathophysiology. Specifically, beta-catenin, the central component of the canonical Wnt pathway, is implicated in the regulation of liver regeneration, development, and carcinogenesis. Wnt-independent activation of beta-catenin by receptor tyrosine kinases has also been observed in the liver. In liver development across various species, through regulation of cell proliferation, differentiation, and maturation, beta-catenin directs foregut endoderm specification, hepatic specification of the foregut, and hepatic morphogenesis. Its role has also been defined in adult hepatic progenitors or oval cells especially in their expansion and differentiation. Thus, beta-catenin undergoes tight temporal regulation to exhibit pleiotropic effects during hepatic development and in hepatic progenitor biology.


Asunto(s)
Hígado/citología , Hígado/metabolismo , Células Madre/citología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Humanos , Hígado/embriología , Células Madre/metabolismo , Proteínas Wnt/genética , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA