Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 325(3): G213-G229, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37366545

RESUMEN

The endocannabinoid system (ECS) is dysregulated in various liver diseases. Previously, we had shown that the major endocannabinoid 2-arachidonoyl glycerol (2-AG) promoted tumorigenesis of intrahepatic cholangiocarcinoma (ICC). However, biosynthesis regulation and clinical significance of 2-AG remain elusive. In the present study, we quantified 2-AG by gas chromatography/mass spectrometry (GC/MS) and showed that 2-AG was enriched in patients with ICC samples as well as in thioacetamide-induced orthotopic rat ICC model. Moreover, we found that diacylglycerol lipase ß (DAGLß) was the principal synthesizing enzyme of 2-AG that significantly upregulated in ICC. DAGLß promoted tumorigenesis and metastasis of ICC in vitro and in vivo and positively correlated with clinical stage and poor survival in patients with ICC. Functional studies showed that activator protein-1 (AP-1; heterodimers of c-Jun and FRA1) directly bound to the promoter and regulated transcription of DAGLß, which can be enhanced by lipopolysaccharide (LPS). miR-4516 was identified as the tumor-suppressing miRNA of ICC that can be significantly suppressed by LPS, 2-AG, or ectopic DAGLß overexpression. FRA1 and STAT3 were targets of miR-4516 and overexpression of miRNA-4516 significantly suppressed expression of FRA1, SATA3, and DAGLß. Expression of miRNA-4516 was negatively correlated with FRA1, SATA3, and DAGLß in patients with ICC samples. Our findings identify DAGLß as the principal synthesizing enzyme of 2-AG in ICC. DAGLß promotes oncogenesis and metastasis of ICC and is transcriptionally regulated by a novel AP-1/DAGLß/miR4516 feedforward circuitry.NEW & NOTEWORTHY Dysregulated endocannabinoid system (ECS) had been confirmed in various liver diseases. However, regulation and function of 2-arachidonoyl glycerol (2-AG) and diacylglycerol lipase ß (DAGLß) in intrahepatic cholangiocarcinoma (ICC) remain to be elucidated. Here, we demonstrated that 2-AG was enriched in ICC, and DAGLß was the principal synthesizing enzyme of 2-AG in ICC. DAGLß promotes tumorigenesis and metastasis in ICC via a novel activator protein-1 (AP-1)/DAGLß/miR4516 feedforward circuitry.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , MicroARNs , Ratas , Animales , Factor de Transcripción AP-1/genética , Endocannabinoides , Lipoproteína Lipasa , Glicerol , Lipopolisacáridos , Colangiocarcinoma/patología , MicroARNs/genética , MicroARNs/metabolismo , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Neoplasias de los Conductos Biliares/metabolismo , Carcinogénesis , Línea Celular Tumoral
2.
Am J Pathol ; 192(3): 484-502, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34896073

RESUMEN

Leptin is an adipokine with roles in food intake and energy metabolism through its actions on neurons in the hypothalamus. The role of leptin in obesity and cardiovascular disorders is well documented. However, its influence on liver conditions such as cholestasis is poorly understood. The effects of exogenous leptin and leptin-neutralizing antibody on biliary hyperplasia, hepatic fibrosis, and inflammation in the multidrug resistance protein 2 knockout (Mdr2KO) mouse model of cholestasis were assessed by quantifying markers specific for cholangiocytes, activated hepatic stellate cells (HSCs), and cytokines. Serum and hepatic leptin were increased in Mdr2KO mice compared with FVB/NJ (FVBN) controls, and exogenous leptin enhanced biliary hyperplasia and liver fibrosis in Mdr2KO and FVBN mice. Leptin administration increased hepatic expression of C-C motif chemokine ligand 2 and IL-6 in Mdr2KO mice. In contrast, leptin-neutralizing antibody reduced intrahepatic bile duct mass and decreased HSC activation in Mdr2KO mice compared with FVBN controls. Sex-related differences were noted, with female Mdr2KO mice having more leptin than males. In cholangiocytes and LX2 cells in vitro, leptin increased phosphorylated Akt and stimulated cell proliferation. Leptin receptor siRNA and inhibitors of Akt phosphorylation impaired leptin-induced cell proliferation and proinflammatory cytokines. The current data suggest that leptin is abnormally increased in cholestatic mice, and excess leptin increases ductular reaction, hepatic fibrosis, and inflammation via leptin receptor-mediated phosphorylation of Akt in cholangiocytes and HSCs.


Asunto(s)
Colestasis , Receptores de Leptina , Animales , Anticuerpos Neutralizantes , Colestasis/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Células Estrelladas Hepáticas/metabolismo , Hiperplasia/patología , Inflamación/patología , Leptina/metabolismo , Leptina/farmacología , Hígado/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Leptina/metabolismo
3.
Front Oncol ; 11: 662135, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34262860

RESUMEN

Variations in tumor biology from patient to patient combined with the low overall survival rate of hepatocellular carcinoma (HCC) present significant clinical challenges. During the progression of chronic liver diseases from inflammation to the development of HCC, microenvironmental properties, including tissue stiffness and oxygen concentration, change over time. This can potentially impact drug metabolism and subsequent therapy response to commonly utilized therapeutics, such as doxorubicin, multi-kinase inhibitors (e.g., sorafenib), and other drugs, including immunotherapies. In this study, we utilized four common HCC cell lines embedded in 3D collagen type-I gels of varying stiffnesses to mimic normal and cirrhotic livers with environmental oxygen regulation to quantify the impact of these microenvironmental factors on HCC chemoresistance. In general, we found that HCC cells with higher baseline levels of cytochrome p450-3A4 (CYP3A4) enzyme expression, HepG2 and C3Asub28, exhibited a cirrhosis-dependent increase in doxorubicin chemoresistance. Under the same conditions, HCC cell lines with lower CYP3A4 expression, HuH-7 and Hep3B2, showed a decrease in doxorubicin chemoresistance in response to an increase in microenvironmental stiffness. This differential therapeutic response was correlated with the regulation of CYP3A4 expression levels under the influence of stiffness and oxygen variation. In all tested HCC cell lines, the addition of sorafenib lowered the required doxorubicin dose to induce significant levels of cell death, demonstrating its potential to help reduce systemic doxorubicin toxicity when used in combination. These results suggest that patient-specific tumor microenvironmental factors, including tissue stiffness, hypoxia, and CYP3A4 activity levels, may need to be considered for more effective use of chemotherapeutics in HCC patients.

4.
Sci Rep ; 10(1): 16024, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32994489

RESUMEN

The orexigenic peptide ghrelin (Ghr) stimulates hunger signals in the hypothalamus via growth hormone secretagogue receptor (GHS-R1a). Gastric Ghr is synthetized as a preprohormone which is proteolytically cleaved, and acylated by a membrane-bound acyl transferase (MBOAT). Circulating Ghr is reduced in cholestatic injuries, however Ghr's role in cholestasis is poorly understood. We investigated Ghr's effects on biliary hyperplasia and hepatic fibrosis in Mdr2-knockout (Mdr2KO) mice, a recognized model of cholestasis. Serum, stomach and liver were collected from Mdr2KO and FVBN control mice treated with Ghr, des-octanoyl-ghrelin (DG) or vehicle. Mdr2KO mice had lower expression of Ghr and MBOAT in the stomach, and lower levels of circulating Ghr compared to WT-controls. Treatment of Mdr2KO mice with Ghr improved plasma transaminases, reduced biliary and fibrosis markers. In the liver, GHS-R1a mRNA was expressed predominantly in cholangiocytes. Ghr but not DG, decreased cell proliferation via AMPK activation in cholangiocytes in vitro. AMPK inhibitors prevented Ghr-induced FOXO1 nuclear translocation and negative regulation of cell proliferation. Ghr treatment reduced ductular reaction and hepatic fibrosis in Mdr2KO mice, regulating cholangiocyte proliferation via GHS-R1a, a G-protein coupled receptor which causes increased intracellular Ca2+ and activation of AMPK and FOXO1, maintaining a low rate of cholangiocyte proliferation.


Asunto(s)
Colestasis/tratamiento farmacológico , Ghrelina/administración & dosificación , Cirrosis Hepática/prevención & control , Receptores de Ghrelina/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Acetiltransferasas/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colestasis/genética , Colestasis/metabolismo , Modelos Animales de Enfermedad , Proteína Forkhead Box O1/metabolismo , Ghrelina/metabolismo , Ghrelina/farmacología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Ratones , Ratones Noqueados , Transaminasas/sangre , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
5.
Micromachines (Basel) ; 11(5)2020 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-32397454

RESUMEN

In chronic liver diseases and hepatocellular carcinoma, the cells and extracellular matrix of the liver undergo significant alteration in response to chronic injury. Recent literature has highlighted the critical, but less studied, role of the liver vasculature in the progression of chronic liver diseases. Recent advancements in liver-on-a-chip systems has allowed in depth investigation of the role that the hepatic vasculature plays both in response to, and progression of, chronic liver disease. In this review, we first introduce the structure, gradients, mechanical properties, and cellular composition of the liver and describe how these factors influence the vasculature. We summarize state-of-the-art vascularized liver-on-a-chip platforms for investigating biological models of chronic liver disease and their influence on the liver sinusoidal endothelial cells of the hepatic vasculature. We conclude with a discussion of how future developments in the field may affect the study of chronic liver diseases, and drug development and testing.

6.
Am J Pathol ; 190(3): 586-601, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31953035

RESUMEN

Galanin (Gal) is a peptide with a role in neuroendocrine regulation of the liver. In this study, we assessed the role of Gal and its receptors, Gal receptor 1 (GalR1) and Gal receptor 2 (GalR2), in cholangiocyte proliferation and liver fibrosis in multidrug resistance protein 2 knockout (Mdr2KO) mice as a model of chronic hepatic cholestasis. The distribution of Gal, GalR1, and GalR2 in specific liver cell types was assessed by laser-capture microdissection and confocal microscopy. Galanin immunoreactivity was detected in cholangiocytes, hepatic stellate cells (HSCs), and hepatocytes. Cholangiocytes expressed GalR1, whereas HSCs and hepatocytes expressed GalR2. Strategies were used to either stimulate or block GalR1 and GalR2 in FVB/N (wild-type) and Mdr2KO mice and measure biliary hyperplasia and hepatic fibrosis by quantitative PCR and immunostaining of specific markers. Galanin treatment increased cholangiocyte proliferation and fibrogenesis in both FVB/N and Mdr2KO mice. Suppression of GalR1, GalR2, or both receptors in Mdr2KO mice resulted in reduced bile duct mass and hepatic fibrosis. In vitro knockdown of GalR1 in cholangiocytes reduced α-smooth muscle actin expression in LX-2 cells treated with cholangiocyte-conditioned media. A GalR2 antagonist inhibited HSC activation when Gal was administered directly to LX-2 cells, but not via cholangiocyte-conditioned media. These data demonstrate that Gal contributes not only to cholangiocyte proliferation but also to liver fibrogenesis via the coordinate activation of GalR1 in cholangiocytes and GalR2 in HSCs.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Colestasis/metabolismo , Galanina/metabolismo , Cirrosis Hepática/metabolismo , Receptor de Galanina Tipo 1/metabolismo , Receptor de Galanina Tipo 2/metabolismo , Animales , Conductos Biliares/metabolismo , Proliferación Celular , Colestasis/patología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Galanina/genética , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Ratones , Ratones Noqueados , Receptor de Galanina Tipo 1/genética , Receptor de Galanina Tipo 2/genética , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
7.
Am J Pathol ; 190(2): 347-357, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31734229

RESUMEN

Severe hepatic insults can lead to acute liver failure and hepatic encephalopathy (HE). Transforming growth factor ß1 (TGFß1) has been shown to contribute to HE during acute liver failure; however, TGFß1 must be activated to bind its receptor and generate downstream effects. One protein that can activate TGFß1 is thrombospondin-1 (TSP-1). Therefore, the aim of this study was to assess TSP-1 during acute liver failure and HE pathogenesis. C57Bl/6 or TSP-1 knockout (TSP-1-/-) mice were injected with azoxymethane (AOM) to induce acute liver failure and HE. Liver damage, neurologic decline, and molecular analyses of TSP-1 and TGFß1 signaling were performed. AOM-treated mice had increased TSP-1 and TGFß1 mRNA and protein expression in the liver. TSP-1-/- mice administered AOM had reduced liver injury as assessed by histology and serum transaminase levels compared with C57Bl/6 AOM-treated mice. TSP-1-/- mice treated with AOM had reduced TGFß1 signaling that was associated with less hepatic cell death as assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining and cleaved caspase 3 expression. TSP-1-/- AOM-treated mice had a reduced rate of neurologic decline, less cerebral edema, and a decrease in microglia activation in comparison with C57Bl/6 mice treated with AOM. Taken together, TSP-1 is an activator of TGFß1 signaling during AOM-induced acute liver failure and contributes to both liver pathology and HE progression.


Asunto(s)
Modelos Animales de Enfermedad , Encefalopatía Hepática/patología , Fallo Hepático Agudo/patología , Trombospondina 1/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Azoximetano/toxicidad , Carcinógenos/toxicidad , Muerte Celular , Encefalopatía Hepática/etiología , Encefalopatía Hepática/metabolismo , Fallo Hepático Agudo/etiología , Fallo Hepático Agudo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
8.
Cells ; 8(9)2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31510045

RESUMEN

Wnt5a signaling has been implicated in the progression of cancer by regulating multiple cellular processes, largely migration and invasion, epithelial-mesenchymal transition (EMT), and metastasis. Since Wnt5a signaling has also been involved in inflammatory processes in infectious and inflammatory diseases, we addressed the role of Wnt5a in regulating NF-κB, a pivotal mediator of inflammatory responses, in the context of cancer. The treatment of melanoma cells with Wnt5a induced phosphorylation of the NF-κB subunit p65 as well as IKK phosphorylation and IκB degradation. By using cDNA overexpression, RNA interference, and dominant negative mutants we determined that ROR1, Dvl2, and Akt (from the Wnt5a pathway) and TRAF2 and RIP (from the NF-κB pathway) are required for the Wnt5a/NF-κB crosstalk. Wnt5a also induced p65 nuclear translocation and increased NF-κB activity as evidenced by reporter assays and a NF-κB-specific upregulation of RelB, Bcl-2, and Cyclin D1. Further, stimulation of melanoma cells with Wnt5a increased the secretion of cytokines and chemokines, including IL-6, IL-8, IL-11, and IL-6 soluble receptor, MCP-1, and TNF soluble receptor I. The inhibition of endogenous Wnt5a demonstrated that an autocrine Wnt5a loop is a major regulator of the NF-κB pathway in melanoma. Taken together, these results indicate that Wnt5a activates the NF-κB pathway and has an immunomodulatory effect on melanoma through the secretion of cytokines and chemokines.


Asunto(s)
Melanoma/metabolismo , FN-kappa B/metabolismo , Proteína Wnt-5a/metabolismo , Comunicación Autocrina , Línea Celular Tumoral , Quimiocinas/metabolismo , Citocinas/metabolismo , Proteínas Dishevelled/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , FN-kappa B/antagonistas & inhibidores , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Transducción de Señal , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción ReIA/metabolismo
9.
Toxicol Sci ; 170(2): 549-561, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31132129

RESUMEN

Acute liver failure is a serious consequence of acetaminophen (APAP)-induced hepatotoxic liver injury with high rates of morbidity and mortality. Transforming growth factor beta 1 (TGFß1) is elevated during liver injury and influences hepatocyte senescence during APAP-induced hepatotoxicity. This study investigated TGFß1 signaling in the context of inflammation, necrotic cell death, and oxidative stress during APAP-induced liver injury. Male C57Bl/6 mice were injected with 600 mg/kg APAP to generate liver injury in the presence or absence of the TGFß receptor 1 inhibitor, GW788388, 1 h prior to APAP administration. Acetaminophen-induced liver injury was characterized using histological and biochemical measures. Transforming growth factor beta 1 expression and signal transduction were assessed using immunohistochemistry, Western blotting and ELISA assays. Hepatic necrosis, liver injury, cell proliferation, hepatic inflammation, and oxidative stress were assessed in all mice. Acetaminophen administration significantly induced necrosis and elevated serum transaminases compared with control mice. Transforming growth factor beta 1 staining was observed in and around areas of necrosis with phosphorylation of SMAD3 observed in hepatocytes neighboring necrotic areas in APAP-treated mice. Pretreatment with GW788388 prior to APAP administration in mice reduced hepatocyte cell death and stimulated regeneration. Phosphorylation of SMAD3 was reduced in APAP mice pretreated with GW788388 and this correlated with reduced hepatic cytokine production and oxidative stress. These results support that TGFß1 signaling plays a significant role in APAP-induced liver injury by influencing necrotic cell death, inflammation, oxidative stress, and hepatocyte regeneration. In conclusion, targeting TGFß1 or downstream signaling may be a possible therapeutic target for the management of APAP-induced liver injury.


Asunto(s)
Acetaminofén/toxicidad , Benzamidas/farmacología , Hepatocitos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pirazoles/farmacología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antioxidantes , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Glutatión/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Inflamación , Hígado/efectos de los fármacos , Fallo Hepático Agudo/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Sustancias Protectoras/farmacología , Regeneración , Transducción de Señal/efectos de los fármacos
10.
Proc (Bayl Univ Med Cent) ; 32(1): 26-29, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30956575

RESUMEN

The nonalcoholic fatty liver disease fibrosis score (NFS) has been shown to be a cost-effective screening strategy in the primary care setting to determine when gastroenterology referral is needed, but NFS as a predictor of hospitalization within 1 year is uncertain. This retrospective observational cohort study involved 1803 patients with a diagnosis of nonalcoholic fatty liver disease or nonalcoholic steatohepatitis. The NFS was categorized into the following: low (less than -1.455), moderate (between -1.455 and 0.676), and high (>0.676). The average NFS score by hospital admission was -0.760, the average number of admissions was 1.81, and the median number of days to hospital admission was 135.8 days (45.5-363, 25th to 75th percentile). A univariate logistic regression model showed that NFS significantly predicted hospital admission (P = 0.007); however, a multivariate logistic regression model, after adjusting for hypertension and tobacco use, indicated that NFS was not significantly associated with hospital admission. Using the logistic regression model, hypertension predicted admission at low (P < 0.0001) and moderate (P = 0.0005) NFS. Using this same model, tobacco use also predicted admission at low (P < 0.0001) and moderate (P = 0.0002) NFS. The NFS should not be used to determine which patients are at increased risk of hospitalization.

11.
J Neuroinflammation ; 16(1): 69, 2019 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-30940161

RESUMEN

BACKGROUND: Acute liver failure resulting from drug-induced liver injury can lead to the development of neurological complications called hepatic encephalopathy (HE). Hepatic transforming growth factor beta 1 (TGFß1) is upregulated due to liver failure in mice and inhibiting circulating TGFß reduced HE progression. However, the specific contributions of TGFß1 on brain cell populations and neuroinflammation during HE are not known. Therefore, the aim of this study was to characterize hepatic and brain TGFß1 signaling during acute liver failure and its contribution to HE progression using a combination of pharmacological and genetic approaches. METHODS: C57Bl/6 or neuron-specific transforming growth factor beta receptor 2 (TGFßR2) null mice (TGFßR2ΔNeu) were treated with azoxymethane (AOM) to induce acute liver failure and HE. The activity of circulating TGFß1 was inhibited in C57Bl/6 mice via injection of a neutralizing antibody against TGFß1 (anti-TGFß1) prior to AOM injection. In all mouse treatment groups, liver damage, neuroinflammation, and neurological deficits were assessed. Inflammatory signaling between neurons and microglia were investigated in in vitro studies through the use of pharmacological inhibitors of TGFß1 signaling in HT-22 and EOC-20 cells. RESULTS: TGFß1 was expressed and upregulated in the liver following AOM injection. Pharmacological inhibition of TGFß1 after AOM injection attenuated neurological decline, microglia activation, and neuroinflammation with no significant changes in liver damage. TGFßR2ΔNeu mice administered AOM showed no effect on liver pathology but significantly reduced neurological decline compared to control mice. Microglia activation and neuroinflammation were attenuated in mice with pharmacological inhibition of TGFß1 or in TGFßR2ΔNeu mice. TGFß1 increased chemokine ligand 2 (CCL2) and decreased C-X3-C motif ligand 1 (CX3CL1) expression in HT-22 cells and reduced interleukin-1 beta (IL-1ß) expression, tumor necrosis factor alpha (TNFα) expression, and phagocytosis activity in EOC-20 cells. CONCLUSION: Increased circulating TGFß1 following acute liver failure results in activation of neuronal TGFßR2 signaling, driving neuroinflammation and neurological decline during AOM-induced HE.


Asunto(s)
Corteza Cerebral/patología , Encefalopatía Hepática/etiología , Fallo Hepático Agudo/complicaciones , Fallo Hepático Agudo/patología , Neuronas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta1/sangre , Animales , Anticuerpos/uso terapéutico , Azoximetano/toxicidad , Benzamidas/farmacología , Carcinógenos/toxicidad , Línea Celular Transformada , Modelos Animales de Enfermedad , Encefalopatía Hepática/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inflamación/etiología , Isoquinolinas/farmacología , Hígado/metabolismo , Hígado/patología , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Fagocitosis/genética , Pirazoles/farmacología , Piridinas/farmacología , Pirroles/farmacología , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
12.
J Clin Exp Hepatol ; 9(1): 117-124, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30774268

RESUMEN

Hepatic encephalopathy describes the array of neurological complications that arise due to liver insufficiency and/or portal-systemic shunt. The pathogenesis of hepatic encephalopathy shares a longstanding association with hyperammonemia and inflammation. Recently, aberrant bile acid signaling has been implicated in the development of key features of hepatic encephalopathy due to acute liver failure including neuronal dysfunction, neuroinflammation and blood-brain barrier permeability. This review summarizes the findings of recent studies demonstrating a role for bile acids in hepatic encephalopathy and speculates on the possible downstream consequences of bile acid signaling.

13.
Sci Rep ; 8(1): 13147, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30177688

RESUMEN

Gulf War Illness (GWI) is a chronic multisymptom disorder affecting veterans of the 1990-91 Gulf war. GWI was linked with exposure to chemicals including the nerve gas prophylactic drug pyridostigmine-bromide (PB) and pesticides (DEET, permethrin). Veterans with GWI exhibit prolonged, low-level systemic inflammation, though whether this impacts the liver is unknown. While no evidence exists that GWI-related chemicals are hepatotoxic, the prolonged inflammation may alter the liver's response to insults such as cholestatic injury. We assessed the effects of GWI-related chemicals on macrophage infiltration and its subsequent influence on hepatic cholestasis. Sprague Dawley rats were treated daily with PB, DEET and permethrin followed by 15 minutes of restraint stress for 28 days. Ten weeks afterward, GWI rats or naïve age-matched controls underwent bile duct ligation (BDL) or sham surgeries. Exposure to GWI-related chemicals alone increased IL-6, and CD11b+F4/80- macrophages in the liver, with no effect on biliary mass or hepatic fibrosis. However, pre-exposure to GWI-related chemicals enhanced biliary hyperplasia and fibrogenesis caused by BDL, compared to naïve rats undergoing the same surgery. These data suggest that GWI patients could be predisposed to developing worse liver pathology due to sustained low-level inflammation of the liver when compared to patients without GWI.


Asunto(s)
Colestasis/inmunología , DEET/toxicidad , Permetrina/toxicidad , Síndrome del Golfo Pérsico/inmunología , Bromuro de Piridostigmina/toxicidad , Estrés Psicológico/inmunología , Animales , Conductos Biliares/efectos de los fármacos , Conductos Biliares/inmunología , Conductos Biliares/patología , Conductos Biliares/cirugía , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Movimiento Celular/efectos de los fármacos , Colestasis/genética , Colestasis/psicología , Colestasis/cirugía , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inmovilización , Inflamación , Interleucina-6/genética , Interleucina-6/inmunología , Ligadura , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Masculino , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/patología , Síndrome del Golfo Pérsico/inducido químicamente , Síndrome del Golfo Pérsico/genética , Síndrome del Golfo Pérsico/psicología , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/genética , Estrés Psicológico/patología
14.
Hepatology ; 68(3): 1042-1056, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29601088

RESUMEN

Primary sclerosing cholangitis (PSC) patients are at risk of developing cholangiocarcinoma (CCA). We have shown that (1) histamine increases biliary hyperplasia through H1/H2 histamine receptors (HRs) and (2) histamine levels increase and mast cells (MCs) infiltrate during PSC and CCA. We examined the effects of chronic treatment with H1/H2HR antagonists on PSC and CCA. Wild-type and multidrug-resistant knockout (Mdr2-/- ) mice were treated by osmotic minipumps with saline, mepyramine, or ranitidine (10 mg/kg body weight/day) or a combination of mepyramine/ranitidine for 4 weeks. Liver damage was assessed by hematoxylin and eosin. We evaluated (1) H1/H2HR expression, (2) MC presence, (3) L-histidine decarboxylase/histamine axis, (4) cholangiocyte proliferation/bile duct mass, and (5) fibrosis/hepatic stellate cell activation. Nu/nu mice were implanted with Mz-ChA-1 cells into the hind flanks and treated with saline, mepyramine, or ranitidine. Tumor growth was measured, and (1) H1/H2HR expression, (2) proliferation, (3) MC activation, (4) angiogenesis, and (5) epithelial-mesenchymal transition (EMT) were evaluated. In vitro, human hepatic stellate cells were evaluated for H1HR and H2HR expression. Cultured cholangiocytes and CCA lines were treated with saline, mepyramine, or ranitidine (25 µM) before evaluating proliferation, angiogenesis, EMT, and potential signaling mechanisms. H1/H2HR and MC presence increased in human PSC and CCA. In H1/H2HR antagonist (alone or in combination)-treated Mdr2-/- mice, liver and biliary damage and fibrosis decreased compared to saline treatment. H1/H2HR antagonists decreased tumor growth, serum histamine, angiogenesis, and EMT. In vitro, H1/H2HR blockers reduced biliary proliferation, and CCA cells had decreased proliferation, angiogenesis, EMT, and migration. Conclusion: Inhibition of H1/H2HR reverses PSC-associated damage and decreases CCA growth, angiogenesis, and EMT; because PSC patients are at risk of developing CCA, using HR blockers may be therapeutic for these diseases. (Hepatology 2018).


Asunto(s)
Colangiocarcinoma/prevención & control , Colangitis Esclerosante/tratamiento farmacológico , Antagonistas de los Receptores Histamínicos H1/uso terapéutico , Antagonistas de los Receptores H2 de la Histamina/uso terapéutico , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Colangiocarcinoma/etiología , Colangitis Esclerosante/complicaciones , Evaluación Preclínica de Medicamentos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Antagonistas de los Receptores Histamínicos H1/farmacología , Antagonistas de los Receptores H2 de la Histamina/farmacología , Humanos , Hígado/efectos de los fármacos , Masculino , Mastocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Neovascularización Patológica/prevención & control , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
15.
Am J Physiol Gastrointest Liver Physiol ; 313(5): G410-G418, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28751425

RESUMEN

Melatonin is a hormone produced by the pineal gland with increased circulating levels shown to inhibit biliary hyperplasia and fibrosis during cholestatic liver injury. Melatonin also has the capability to suppress the release of hypothalamic gonadotropin-releasing hormone (GnRH), a hormone that promotes cholangiocyte proliferation when serum levels are elevated. However, the interplay and contribution of neural melatonin and GnRH to cholangiocyte proliferation and fibrosis in bile duct-ligated (BDL) rats have not been investigated. To test this, cranial levels of melatonin were increased by implanting osmotic minipumps that performed an intracerebroventricular (ICV) infusion of melatonin or saline for 7 days starting at the time of BDL. Hypothalamic GnRH mRNA and cholangiocyte secretion of GnRH and melatonin were assessed. Cholangiocyte proliferation and fibrosis were measured. Primary human hepatic stellate cells (HSCs) were treated with cholangiocyte supernatants, GnRH, or the GnRH receptor antagonist cetrorelix acetate, and cell proliferation and fibrosis gene expression were assessed. Melatonin infusion reduced hypothalamic GnRH mRNA expression and led to decreased GnRH and increased melatonin secretion from cholangiocytes. Infusion of melatonin was found to reduce hepatic injury, cholangiocyte proliferation, and fibrosis during BDL-induced liver injury. HSCs supplemented with BDL cholangiocyte supernatant had increased proliferation, and this increase was reversed when HSCs were supplemented with supernatants from melatonin-infused rats. GnRH stimulated fibrosis gene expression in HSCs, and this was reversed by cetrorelix acetate cotreatment. Increasing bioavailability of melatonin in the brain may improve outcomes during cholestatic liver disease.NEW & NOTEWORTHY We have previously demonstrated that GnRH is expressed in cholangiocytes and promotes their proliferation during cholestasis. In addition, dark therapy, which increases melatonin, reduced cholangiocyte proliferation and fibrosis during cholestasis. This study expands these findings by investigating neural GnRH regulation by melatonin during BDL-induced cholestasis by infusing melatonin into the brain. Melatonin infusion reduced cholangiocyte proliferation and fibrosis, and these effects are due to GNRH receptor 1-dependent paracrine signaling between cholangiocytes and hepatic stellate cells.


Asunto(s)
Conductos Biliares , Colestasis , Hormona Liberadora de Gonadotropina , Cirrosis Hepática , Melatonina , Glándula Pineal/fisiología , Animales , Conductos Biliares/efectos de los fármacos , Conductos Biliares/metabolismo , Conductos Biliares/patología , Proliferación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Depresores del Sistema Nervioso Central/sangre , Depresores del Sistema Nervioso Central/metabolismo , Colestasis/complicaciones , Colestasis/metabolismo , Modelos Animales de Enfermedad , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Antagonistas de Hormonas/farmacología , Humanos , Hiperplasia , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Melatonina/administración & dosificación , Melatonina/sangre , Melatonina/metabolismo , Ratas , Receptores LHRH/antagonistas & inhibidores
16.
Lab Invest ; 97(7): 843-853, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28581486

RESUMEN

The Hippo signaling pathway and the Notch signaling pathway are evolutionary conserved signaling cascades that have important roles in embryonic development of many organs. In murine liver, disruption of either pathway impairs intrahepatic bile duct development. Recent studies suggested that the Notch signaling receptor Notch2 is a direct transcriptional target of the Hippo signaling pathway effector YAP, and the Notch signaling is a major mediator of the Hippo signaling in maintaining biliary cell characteristics in adult mice. However, it remains to be determined whether the Hippo signaling pathway functions through the Notch signaling in intrahepatic bile duct development. We found that loss of the Hippo signaling pathway tumor suppressor Nf2 resulted in increased expression levels of the Notch signaling pathway receptor Notch2 in cholangiocytes but not in hepatocytes. When knocking down Notch2 on the background of Nf2 deficiency in mouse livers, the excessive bile duct development induced by Nf2 deficiency was suppressed by heterozygous and homozygous deletion of Notch2 in a dose-dependent manner. These results implicated that Notch signaling is one of the downstream effectors of the Hippo signaling pathway in regulating intrahepatic bile duct development.


Asunto(s)
Conductos Biliares Intrahepáticos/crecimiento & desarrollo , Neurofibromatosis 2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Notch2/metabolismo , Animales , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Femenino , Vía de Señalización Hippo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurofibromatosis 2/genética , Proteínas Serina-Treonina Quinasas/genética , Receptor Notch2/genética
17.
FASEB J ; 31(10): 4305-4324, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28634212

RESUMEN

Melatonin therapy or prolonged exposure to complete darkness reduces biliary hyperplasia and liver fibrosis in bile-duct-ligated (BDL) rats; however, no information exists in primary sclerosing cholangitis (PSC). Thus, we aimed to determine the therapeutic effects of prolonged dark therapy or melatonin administration on hepatic fibrosis in the multidrug resistance gene 2-knockout (Mdr2-/-) mouse model of PSC. Melatonin levels, biliary mass, liver fibrosis, angiogenesis and miR-200b expression were evaluated in wild-type and Mdr2-/- mice exposed to darkness or melatonin treatment or in male patients with PSC and healthy controls. Mdr2-/- mice were also treated with miR-200b inhibitor or control before evaluating biliary mass, liver fibrosis, and angiogenesis. After overexpression of arylalkylamine N-acetyltransferase (AANAT; the enzyme regulating melatonin synthesis) or inhibition of miR-200b in cholangiocytes and hepatic stellate cells in vitro, we evaluated angiogenesis and fibrosis gene expression. After exposure to darkness or administration of melatonin, Mdr2-/- mice show elevated serum melatonin levels and inhibition of biliary mass, along with reduction of liver fibrosis and angiogenesis. MicroRNA PCR analysis demonstrated that miR-200b expression increased in Mdr2-/- mice and patients with PSC compared with controls and decreased in Mdr2-/- mice subjected to dark exposure or melatonin treatment. Inhibition of miR-200b in Mdr2-/- ablates biliary proliferation, liver fibrosis, and angiogenesis. In vitro, overexpression of AANAT or inhibition of miR-200b in cholangiocytes and hepatic stellate cells decreased the expression of miR-200b, angiogenesis, and fibrosis genes. Dark therapy or targeting melatonin/miR-200b axis may be important in the management of biliary damage and liver fibrosis in cholangiopathies including PSC.-Wu, N., Meng, F., Zhou, T., Han, Y., Kennedy, L., Venter, J., Francis, H., DeMorrow, S., Onori, P., Invernizzi, P., Bernuzzi, F., Mancinelli, R., Gaudio, E., Franchitto, A., Glaser, S., Alpini G. Prolonged darkness reduces liver fibrosis in a mouse model of primary sclerosing cholangitis by miR-200b down-regulation.


Asunto(s)
Colangitis Esclerosante/metabolismo , Oscuridad , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/metabolismo , Melatonina/metabolismo , MicroARNs/genética , Inductores de la Angiogénesis/metabolismo , Animales , Proliferación Celular/fisiología , Colangitis Esclerosante/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Fibrosis/metabolismo , Masculino , Ratones Transgénicos
18.
Am J Pathol ; 187(4): 819-830, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28196718

RESUMEN

During the course of cholestatic liver diseases, mitotically dormant cholangiocytes proliferate and subsequently acquire a neuroendocrine phenotype. Galanin is a neuroendocrine factor responsible for regulation of physiological responses, such as feeding behavior and mood, and has been implicated in the development of fatty liver disease, although its role in biliary hyperplasia is unknown. Biliary hyperplasia was induced in rats via bile duct ligation (BDL) surgery, and galanin was increased in serum and liver homogenates from BDL rats. Treatment of sham and BDL rats with recombinant galanin increased cholangiocyte proliferation and intrahepatic biliary mass, liver damage, and inflammation, whereas blocking galanin expression with specific vivo-morpholino sequences inhibited hyperplastic cholangiocyte proliferation, liver damage, inflammation, and subsequent fibrosis. The proliferative effects of galanin were via activation of galanin receptor 1 expressed specifically on cholangiocytes and were associated with an activation of extracellular signal-regulated kinase 1/2, and ribosomal S6 kinase 1 signal transduction pathways and subsequent increase in cAMP responsive element binding protein DNA-binding activity and induction of Yes-associated protein expression. Strategies to inhibit extracellular signal-regulated kinase 1/2, ribosomal S6 kinase 1, or cAMP responsive element binding protein DNA-binding activity prevented the proliferative effects of galanin. Taken together, these data suggest that targeting galanin signaling may be effective for the maintenance of biliary mass during cholestatic liver diseases.


Asunto(s)
Conductos Biliares/patología , Colestasis/metabolismo , Colestasis/patología , Galanina/metabolismo , Regulación hacia Arriba , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , ADN/metabolismo , Inflamación/patología , Ligadura , Masculino , Ratones , Morfolinos/farmacología , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de Galanina/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteínas Señalizadoras YAP
19.
Hepatology ; 65(6): 1991-2004, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28120369

RESUMEN

Activated mast cells (MCs) release histamine (HA) and MCs infiltrate the liver following bile duct ligation (BDL), increasing intrahepatic bile duct mass (IBDM) and fibrosis. We evaluated the effects of BDL in MC-deficient (KitW-sh ) mice. Wild-type (WT) and KitW-sh mice were subjected to sham or BDL for up to 7 days and KitW-sh mice were injected with cultured mast cells or 1× phosphate-buffered saline (PBS) before collecting serum, liver, and cholangiocytes. Liver damage was assessed by hematoxylin and eosin and alanine aminotransferase levels. IBDM was detected by cytokeratin-19 expression and proliferation by Ki-67 immunohistochemistry (IHC). Fibrosis was detected by IHC, hydroxyproline content, and by qPCR for fibrotic markers. Hepatic stellate cell (HSC) activation and transforming growth factor-beta 1 (TGF-ß1) expression/secretion were evaluated. Histidine decarboxylase (HDC) and histamine receptor (HR) expression were detected by qPCR and HA secretion by enzymatic immunoassay. To evaluate vascular cells, von Willebrand factor (vWF) and vascular endothelial growth factor (VEGF)-C expression were measured. In vitro, cultured HSCs were stimulated with cholangiocyte supernatants and alpha-smooth muscle actin levels were measured. BDL-induced liver damage was reduced in BDL KitW-sh mice, whereas injection of MCs did not mimic BDL-induced damage. In BDL KitW-sh mice, IBDM, proliferation, HSC activation/fibrosis, and TGF-ß1 expression/secretion were decreased. The HDC/HA/HR axis was ablated in sham and BDL KitW-sh mice. vWF and VEGF-C expression decreased in BDL KitW-sh mice. In KitW-sh mice injected with MCs, IBDM, proliferation, fibrosis, and vascular cell activation increased. Stimulation with cholangiocyte supernatants from BDL WT or KitW-sh mice injected with MCs increased HSC activation, which decreased with supernatants from BDL KitW-sh mice. CONCLUSION: MCs promote hyperplasia, fibrosis, and vascular cell activation. Knockout of MCs decreases BDL-induced damage. Modulation of MCs may be important in developing therapeutics for cholangiopathies. (Hepatology 2017;65:1991-2004).


Asunto(s)
Enfermedades de las Vías Biliares/patología , Cirrosis Hepática/patología , Hígado/lesiones , Mastocitos/trasplante , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Conductos Biliares Intrahepáticos/cirugía , Enfermedades de las Vías Biliares/fisiopatología , Biopsia con Aguja , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hiperplasia/patología , Inmunohistoquímica , Ligadura/métodos , Hígado/patología , Masculino , Mastocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Valores de Referencia
20.
J Neuroinflammation ; 13(1): 198, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27561705

RESUMEN

BACKGROUND: Acute liver failure is associated with numerous systemic consequences including neurological dysfunction, termed hepatic encephalopathy, which contributes to mortality and is a challenge to manage in the clinic. During hepatic encephalopathy, microglia activation and neuroinflammation occur due to dysregulated cell signaling and an increase of toxic metabolites in the brain. Fractalkine is a chemokine that is expressed primarily in neurons and through signaling with its receptor CX3CR1 on microglia, leads to microglia remaining in a quiescent state. Fractalkine is often suppressed during neuropathies that are characterized by neuroinflammation. However, the expression and subsequent role of fractalkine on microglia activation and the pathogenesis of hepatic encephalopathy due to acute liver failure is unknown. METHODS: Hepatic encephalopathy was induced in mice via injection of azoxymethane (AOM) or saline for controls. Subsets of these mice were implanted with osmotic minipumps that infused soluble fractalkine or saline into the lateral ventricle of the brain. Neurological decline and the latency to coma were recorded in these mice, and brain, serum, and liver samples were collected. Neurons or microglia were isolated from whole brain samples using immunoprecipitation. Liver damage was assessed using hematoxylin and eosin staining and by measuring serum liver enzyme concentrations. Fractalkine and CX3CR1 expression were assessed by real-time PCR, and proinflammatory cytokine expression was assessed using ELISA assays. RESULTS: Following AOM administration, fractalkine expression is suppressed in the cortex and in isolated neurons compared to vehicle-treated mice. CX3CR1 is suppressed in isolated microglia from AOM-treated mice. Soluble fractalkine infusion into the brain significantly reduced neurological decline in AOM-treated mice compared to saline-infused AOM-treated mice. Infusion of soluble fractalkine into AOM-treated mice reduced liver damage, lessened microglia activation, and suppressed expression of chemokine ligand 2, interleukin-6, and tumor necrosis factor alpha compared to saline-infused mice. CONCLUSIONS: These findings suggest that fractalkine-mediated signaling is suppressed in the brain following the development of hepatic encephalopathy. Supplementation of AOM-treated mice with soluble fractalkine led to improved outcomes, which identifies this pathway as a possible therapeutic target for the management of hepatic encephalopathy following acute liver injury.


Asunto(s)
Quimiocina CX3CL1/uso terapéutico , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Encefalopatía Hepática/complicaciones , Alanina Transaminasa/sangre , Animales , Azoximetano/toxicidad , Bilirrubina/sangre , Proteínas de Unión al Calcio/metabolismo , Carcinógenos/toxicidad , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Encefalitis/patología , Citometría de Flujo , Encefalopatía Hepática/inducido químicamente , Infusiones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/patología , Fosfopiruvato Hidratasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA