Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33649241

RESUMEN

α1-Antitrypsin (AAT) deficiency is a common genetic disease presenting with lung and liver diseases. AAT deficiency results from pathogenic variants in the SERPINA1 gene encoding AAT and the common mutant Z allele of SERPINA1 encodes for Z α1-antitrypsin (ATZ), a protein forming hepatotoxic polymers retained in the endoplasmic reticulum of hepatocytes. PiZ mice express the human ATZ and are a valuable model to investigate the human liver disease of AAT deficiency. In this study, we investigated differential expression of microRNAs (miRNAs) between PiZ and control mice and found that miR-34b/c was up-regulated and its levels correlated with intrahepatic ATZ. Furthermore, in PiZ mouse livers, we found that Forkhead Box O3 (FOXO3) driving microRNA-34b/c (miR-34b/c) expression was activated and miR-34b/c expression was dependent upon c-Jun N-terminal kinase (JNK) phosphorylation on Ser574 Deletion of miR-34b/c in PiZ mice resulted in early development of liver fibrosis and increased signaling of platelet-derived growth factor (PDGF), a target of miR-34b/c. Activation of FOXO3 and increased miR-34c were confirmed in livers of humans with AAT deficiency. In addition, JNK-activated FOXO3 and miR-34b/c up-regulation were detected in several mouse models of liver fibrosis. This study reveals a pathway involved in liver fibrosis and potentially implicated in both genetic and acquired causes of hepatic fibrosis.


Asunto(s)
Proteína Forkhead Box O3/metabolismo , Cirrosis Hepática , MAP Quinasa Quinasa 4/metabolismo , Regulación hacia Arriba , Animales , Modelos Animales de Enfermedad , Proteína Forkhead Box O3/genética , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/prevención & control , MAP Quinasa Quinasa 4/genética , Masculino , Ratones , Ratones Noqueados , MicroARNs/biosíntesis , MicroARNs/genética , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
2.
Hepatology ; 76(5): 1360-1375, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35278227

RESUMEN

BACKGROUND AND AIMS: In liver fibrosis, myofibroblasts derive from HSCs and as yet undefined mesenchymal cells. We aimed to identify portal mesenchymal progenitors of myofibroblasts. APPROACH AND RESULTS: Portal mesenchymal cells were isolated from mouse bilio-vascular tree and analyzed by single-cell RNA-sequencing. Thereby, we uncovered the landscape of portal mesenchymal cells in homeostatic mouse liver. Trajectory analysis enabled inferring a small cell population further defined by surface markers used to isolate it. This population consisted of portal fibroblasts with mesenchymal stem cell features (PMSCs), i.e., high clonogenicity and trilineage differentiation potential, that generated proliferative myofibroblasts, contrasting with nonproliferative HSC-derived myofibroblasts (-MF). Using bulk RNA-sequencing, we built oligogene signatures of the two cell populations that remained discriminant across myofibroblastic differentiation. SLIT2, a prototypical gene of PMSC/PMSC-MF signature, mediated profibrotic and angiogenic effects of these cells, which conditioned medium promoted HSC survival and endothelial cell tubulogenesis. Using PMSC/PMSC-MF 7-gene signature and slit guidance ligand 2 fluorescent in situ hybridization, we showed that PMSCs display a perivascular portal distribution in homeostatic liver and largely expand with fibrosis progression, contributing to the myofibroblast populations that form fibrotic septa, preferentially along neovessels, in murine and human liver disorders, irrespective of etiology. We also unraveled a 6-gene expression signature of HSCs/HSC-MFs that did not vary in these disorders, consistent with their low proliferation rate. CONCLUSIONS: PMSCs form a small reservoir of expansive myofibroblasts, which, in interaction with neovessels and HSC-MFs that mainly arise through differentiation from a preexisting pool, underlie the formation of fibrotic septa in all types of liver diseases.


Asunto(s)
Hepatopatías , Células Madre Mesenquimatosas , Ratones , Humanos , Animales , Miofibroblastos/metabolismo , Medios de Cultivo Condicionados/metabolismo , Hibridación Fluorescente in Situ , Ligandos , Cirrosis Hepática/patología , Hígado/patología , Fibroblastos/patología , Hepatopatías/patología , ARN , Células Estrelladas Hepáticas/metabolismo , Células Cultivadas
3.
Int J Mol Sci ; 24(2)2023 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-36674751

RESUMEN

ABCB4 (ATP-binding cassette subfamily B member 4) is a hepatocanalicular floppase involved in biliary phosphatidylcholine (PC) secretion. Variations in the ABCB4 gene give rise to several biliary diseases, including progressive familial intrahepatic cholestasis type 3 (PFIC3), an autosomal recessive disease that can be lethal in the absence of liver transplantation. In this study, we investigated the effect and potential rescue of ten ABCB4 missense variations in NBD1:NBD2 homologous positions (Y403H/Y1043H, K435M/K1075M, E558K/E1200A, D564G/D1206G and H589Y/H1231Y) all localized at the conserved and functionally critical motifs of ABC transporters, six of which are mutated in patients. By combining structure analysis and in vitro studies, we found that all ten mutants were normally processed and localized at the canalicular membrane of HepG2 cells, but showed dramatically impaired PC transport activity that was significantly rescued by treatment with the clinically approved CFTR potentiator ivacaftor. Our results provide evidence that functional ABCB4 mutations are rescued by ivacaftor, paving the way for the repositioning of this potentiator for the treatment of selected patients with PFIC3 caused by mutations in the ATP-binding sites of ABCB4.


Asunto(s)
Colestasis Intrahepática , Mutación Missense , Humanos , Reposicionamiento de Medicamentos , Colestasis Intrahepática/tratamiento farmacológico , Colestasis Intrahepática/genética , Fosfatidilcolinas , Adenosina Trifosfato
4.
Hepatology ; 74(6): 3194-3212, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34297412

RESUMEN

BACKGROUND AND AIMS: Zinc finger E-box binding homeobox 1 (ZEB1) is a transcription factor that promotes metastatic and stem cell features, which has been associated with poor prognosis in cholangiocarcinoma (CCA), a desmoplastic cancer enriched in cancer-associated fibroblasts (CAFs). We aimed to define ZEB1 regulatory functions in malignant and stromal compartments of CCA. APPROACH AND RESULTS: Bioinformatic and immunohistochemical analyses were performed to determine correlations between ZEB1 and markers of progressiveness in human intrahepatic CCA (iCCA). Gain-of-function and loss-of-function models were generated in CCA cells and liver myofibroblasts as a model of CAFs. Conditioned media (CM) was used to unravel tumor-stroma interplay. In vivo experiments were performed using a xenograft CCA model. ZEB1 expression in tumor cells of human iCCA was associated with undifferentiated tumor and vascular invasion. In vitro, ZEB1 promoted epithelial-mesenchymal transition and stemness in tumor cells, leading to cell migration and spheroid formation. In vivo, ZEB1-overexpressing CCA cells formed larger tumors with more abundant stroma. Expression of cellular communication network factor 2 (CCN2, encoding connective tissue growth factor [CTGF]) was increased in tumor cells from ZEB1-overexpressing xenografts and correlated with ZEB1 expression in human tumors. In vitro, CM from ZEB1-overexpressing tumor cells or recombinant CTGF induced myofibroblast proliferation. ZEB1 was also expressed by CAFs in human CCA, and its expression correlated with CCN2 in myofibroblasts and CCA stroma. In mice, cotransplantation of CCA cells with ZEB1-depleted myofibroblasts reduced CCA progressiveness compared to CCA cells/ZEB1-expressing myofibroblasts. Furthermore, ZEB1 controls the expression of paracrine signals (i.e., HGF and IL6) in tumor cells and myofibroblasts. CONCLUSIONS: ZEB1 plays a key role in CCA progression by regulating tumor cell-CAF crosstalk, leading to tumor dedifferentiation and CAF activation.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Desdiferenciación Celular , Colangiocarcinoma/metabolismo , Comunicación Paracrina , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Neoplasias de los Conductos Biliares/patología , Fibroblastos Asociados al Cáncer/patología , Colangiocarcinoma/patología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Células del Estroma
5.
Liver Int ; 41(6): 1344-1357, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33650203

RESUMEN

BACKGROUND & AIM: ABCB4 is expressed at the canalicular membrane of hepatocytes. This ATP-binding cassette (ABC) transporter is responsible for the secretion of phosphatidylcholine into bile canaliculi. Missense genetic variations of ABCB4 are correlated with several rare cholestatic liver diseases, the most severe being progressive familial intrahepatic cholestasis type 3 (PFIC3). In a repurposing strategy to correct intracellularly retained ABCB4 variants, we tested 16 compounds previously validated as cystic fibrosis transmembrane conductance regulator (CFTR) correctors. METHODS: The maturation, intracellular localization and activity of intracellularly retained ABCB4 variants were analyzed in cell models after treatment with CFTR correctors. In addition, in silico molecular docking calculations were performed to test the potential interaction of CFTR correctors with ABCB4. RESULTS: We observed that the correctors C10, C13, and C17, as well as the combinations of C3 + C18 and C4 + C18, allowed the rescue of maturation and canalicular localization of four distinct traffic-defective ABCB4 variants. However, such treatments did not permit a rescue of the phosphatidylcholine secretion activity of these defective variants and were also inhibitory of the activity of wild type ABCB4. In silico molecular docking analyses suggest that these CFTR correctors might directly interact with transmembrane domains and/or ATP-binding sites of the transporter. CONCLUSION: Our results illustrate the uncoupling between the traffic and the activity of ABCB4 because the same molecules can rescue the traffic of defective variants while they inhibit the secretion activity of the transporter. We expect that this study will help to design new pharmacological tools with potential clinical interest.


Asunto(s)
Colestasis Intrahepática , Colestasis , Subfamilia B de Transportador de Casetes de Unión a ATP , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Simulación del Acoplamiento Molecular , Fosfatidilcolinas
6.
Nature ; 523(7558): 92-5, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-25970250

RESUMEN

The tumour microenvironment may contribute to tumorigenesis owing to mechanical forces such as fibrotic stiffness or mechanical pressure caused by the expansion of hyper-proliferative cells. Here we explore the contribution of the mechanical pressure exerted by tumour growth onto non-tumorous adjacent epithelium. In the early stage of mouse colon tumour development in the Notch(+)Apc(+/1638N) mouse model, we observed mechanistic pressure stress in the non-tumorous epithelial cells caused by hyper-proliferative adjacent crypts overexpressing active Notch, which is associated with increased Ret and ß-catenin signalling. We thus developed a method that allows the delivery of a defined mechanical pressure in vivo, by subcutaneously inserting a magnet close to the mouse colon. The implanted magnet generated a magnetic force on ultra-magnetic liposomes, stabilized in the mesenchymal cells of the connective tissue surrounding colonic crypts after intravenous injection. The magnetically induced pressure quantitatively mimicked the endogenous early tumour growth stress in the order of 1,200 Pa, without affecting tissue stiffness, as monitored by ultrasound strain imaging and shear wave elastography. The exertion of pressure mimicking that of tumour growth led to rapid Ret activation and downstream phosphorylation of ß-catenin on Tyr654, imparing its interaction with the E-cadherin in adherens junctions, and which was followed by ß-catenin nuclear translocation after 15 days. As a consequence, increased expression of ß-catenin-target genes was observed at 1 month, together with crypt enlargement accompanying the formation of early tumorous aberrant crypt foci. Mechanical activation of the tumorigenic ß-catenin pathway suggests unexplored modes of tumour propagation based on mechanical signalling pathways in healthy epithelial cells surrounding the tumour, which may contribute to tumour heterogeneity.


Asunto(s)
Carcinogénesis/patología , Neoplasias del Colon/fisiopatología , Presión , Microambiente Tumoral , beta Catenina/genética , Transporte Activo de Núcleo Celular , Animales , Células Epiteliales/citología , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Imanes , Masculino , Nanopartículas del Metal , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , beta Catenina/metabolismo
7.
Int J Mol Sci ; 22(13)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209301

RESUMEN

ABCB4 (ATP-binding cassette subfamily B member 4) is an ABC transporter expressed at the canalicular membrane of hepatocytes where it ensures phosphatidylcholine secretion into bile. Genetic variations of ABCB4 are associated with several rare cholestatic diseases. The available treatments are not efficient for a significant proportion of patients with ABCB4-related diseases and liver transplantation is often required. The development of novel therapies requires a deep understanding of the molecular mechanisms regulating ABCB4 expression, intracellular traffic, and function. Using an immunoprecipitation approach combined with mass spectrometry analyses, we have identified the small GTPase RAB10 as a novel molecular partner of ABCB4. Our results indicate that the overexpression of wild type RAB10 or its dominant-active mutant significantly increases the amount of ABCB4 at the plasma membrane expression and its phosphatidylcholine floppase function. Contrariwise, RAB10 silencing induces the intracellular retention of ABCB4 and then indirectly diminishes its secretory function. Taken together, our findings suggest that RAB10 regulates the plasma membrane targeting of ABCB4 and consequently its capacity to mediate phosphatidylcholine secretion.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Membrana Celular/metabolismo , Hepatocitos/metabolismo , Fosfatidilcolinas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Transporte Biológico Activo , Membrana Celular/genética , Células HEK293 , Células HeLa , Humanos , Fosfatidilcolinas/genética , Proteínas de Unión al GTP rab/genética
8.
Gut ; 69(1): 92-102, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31003979

RESUMEN

OBJECTIVE: Patients with primary sclerosing cholangitis (PSC) were previously shown to display a bacterial gut dysbiosis but fungal microbiota has never been examined in these patients. The aim of this study was to assess the fungal gut microbiota in patients with PSC. DESIGN: We analysed the faecal microbiota of patients with PSC and concomitant IBD (n=27), patients with PSC and no IBD (n=22), patients with IBD and no PSC (n=33) and healthy subjects (n=30). Bacterial and fungal composition of the faecal microbiota was determined using 16S and ITS2 sequencing, respectively. RESULTS: We found that patients with PSC harboured bacterial dysbiosis characterised by a decreased biodiversity, an altered composition and a decreased correlation network density. These alterations of the microbiota were associated with PSC, independently of IBD status. For the first time, we showed that patients with PSC displayed a fungal gut dysbiosis, characterised by a relative increase in biodiversity and an altered composition. Notably, we observed an increased proportion of Exophiala and a decreased proportion of Saccharomyces cerevisiae. Compared with patients with IBD and healthy subjects, the gut microbiota of patients with PSC exhibited a strong disruption in bacteria-fungi correlation network, suggesting an alteration in the interkingdom crosstalk. CONCLUSION: This study demonstrates that bacteria and fungi contribute to gut dysbiosis in PSC.


Asunto(s)
Colangitis Esclerosante/microbiología , Disbiosis/microbiología , Hongos/aislamiento & purificación , Microbioma Gastrointestinal , Adulto , Anciano , Bacterias/clasificación , Bacterias/aislamiento & purificación , Técnicas de Tipificación Bacteriana/métodos , Biodiversidad , Femenino , Hongos/clasificación , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Masculino , Persona de Mediana Edad , Técnicas de Tipificación Micológica/métodos , Adulto Joven
9.
J Hepatol ; 72(4): 627-635, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31760070

RESUMEN

BACKGROUND & AIMS: In non-alcoholic fatty liver disease (NAFLD), hepatocytes can undergo necroptosis: a regulated form of necrotic cell death mediated by the receptor-interacting protein kinase (RIPK) 1. Herein, we assessed the potential for RIPK1 and its downstream effector mixed lineage kinase domain-like protein (MLKL) to act as therapeutic targets and markers of activity in NAFLD. METHODS: C57/BL6J-mice were fed a normal chow diet or a high-fat diet (HFD). The effect of RIPA-56, a highly specific inhibitor of RIPK1, was evaluated in HFD-fed mice and in primary human steatotic hepatocytes. RIPK1 and MLKL concentrations were measured in the serum of patients with NAFLD. RESULTS: When used as either a prophylactic or curative treatment for HFD-fed mice, RIPA-56 caused a downregulation of MLKL and a reduction of liver injury, inflammation and fibrosis, characteristic of non-alcoholic steatohepatitis (NASH), as well as of steatosis. This latter effect was reproduced by treating primary human steatotic hepatocytes with RIPA-56 or necrosulfonamide, a specific inhibitor of human MLKL, and by knockout (KO) of Mlkl in fat-loaded AML-12 mouse hepatocytes. Mlkl-KO led to activation of mitochondrial respiration and an increase in ß-oxidation in steatotic hepatocytes. Along with decreased MLKL activation, Ripk3-KO mice exhibited increased activities of the liver mitochondrial respiratory chain complexes in experimental NASH. In patients with NAFLD, serum concentrations of RIPK1 and MLKL increased in correlation with activity. CONCLUSION: The inhibition of RIPK1 improves NASH features in HFD-fed mice and reverses steatosis via an MLKL-dependent mechanism that, at least partly, involves an increase in mitochondrial respiration. RIPK1 and MLKL are potential serum markers of activity and promising therapeutic targets in NAFLD. LAY SUMMARY: There are currently no pharmacological treatment options for non-alcoholic fatty liver disease (NAFLD), which is now the most frequent liver disease. Necroptosis is a regulated process of cell death that can occur in hepatocytes during NAFLD. Herein, we show that RIPK1, a gatekeeper of the necroptosis pathway that is activated in NAFLD, can be inhibited by RIPA-56 to reduce not only liver injury, inflammation and fibrosis, but also steatosis in experimental models. These results highlight the potential of RIPK1 as a therapeutic target in NAFLD.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/sangre , Acrilamidas/farmacología , Anciano , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Necroptosis/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteínas Quinasas/sangre , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Resultado del Tratamiento
10.
Liver Int ; 40(1): 163-174, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31544333

RESUMEN

BACKGROUND AND AIMS: The ABCC2 gene is implicated in Dubin-Johnson syndrome (DJS), a rare autosomal recessive liver disorder. The primary aim of this study was to determine the diagnostic value of ABCC2 genetic testing in the largest cohort of DJS reported to date. The high number of patients with cholestatic manifestations in this series prompted us to evaluate the genetic contribution of rare, potentially pathogenic ABCC2 variants to other inherited cholestatic disorders. METHODS: The cohort study included 32 patients with clinical DJS diagnosis, and 372 patients referred for the following disorders: low phospholipid-associated cholelithiasis (LPAC) syndrome, intrahepatic cholestasis of pregnancy (ICP) and benign recurrent intrahepatic cholestasis (BRIC). ABCC2 was screened by next-generation sequencing. RESULTS: Most patients with clinical DJS had positive genetic diagnosis (n = 30; 94%), with a great diversity of point mutations and copy number variations in ABCC2. Strikingly, eight (27%) of these patients showed transient cholestatic features at presentation: four neonatal cholestasis, two ICP, one contraceptive-induced cholestasis and one sporadic cholestasis. Conversely, the frequency of rare, heterozygous, potentially pathogenic ABCC2 variants in patients with LPAC, ICP or BRIC did not differ significantly from that of the general population. CONCLUSIONS: This large series reveals that DJS is a highly homogeneous Mendelian disorder involving a large spectrum of ABCC2 variants. Genetic testing is crucial to establish early DJS diagnosis in patients with atypical presentations, such as neonatal cholestasis. This study also provides no evidence for the contribution of rare, potentially pathogenic ABCC2 variants to other inherited cholestatic disorders.


Asunto(s)
Ictericia Idiopática Crónica/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Adolescente , Adulto , Niño , Preescolar , Colestasis/diagnóstico , Colestasis Intrahepática/diagnóstico , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Femenino , Francia , Heterocigoto , Humanos , Lactante , Ictericia Idiopática Crónica/diagnóstico , Masculino , Persona de Mediana Edad , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Mutación , Embarazo , Complicaciones del Embarazo/diagnóstico , Adulto Joven
11.
Clin Gastroenterol Hepatol ; 17(13): 2785-2792.e3, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30880273

RESUMEN

BACKGROUND & AIMS: Primary sclerosing cholangitis (PSC) has a variable, often progressive, course. Magnetic resonance cholangiography (MRC) is used in the diagnosis of PSC. Magnetic resonance risk scoring systems, called Anali without and with gadolinium, are used to predict disease progression, determined by radiologic factors. We aimed to assess the prognostic value of Anali scores in patients with PSC and validate our findings in a separate cohort. METHODS: We performed a retrospective study of patients with large-duct PSC (internal cohort, 119 patients in France; external cohort, 119 patients in Canada, Italy, and the United Kingdom). All the first-available MRC results were reviewed by 2 radiologists and the Anali scores were calculated as follows: Anali without gadolinium = (1× dilatation of intrahepatic bile ducts) + (2× dysmorphy) + (1× portal hypertension); Anali with gadolinium = (1× dysmorphy) + (1× parenchymal enhancement heterogeneity). The primary end point was survival without liver transplantation or cirrhosis decompensation. The prognostic value of Anali scores was assessed by Cox regression modeling. RESULTS: During a total of 549 patient-years for the internal cohort and 497 patient-years for the external cohort, we recorded 2 and 8 liver transplantations, 4 and 3 liver-related deaths, and 26 and 25 cirrhosis decompensations, respectively. In the univariate analysis, factors associated with survival without liver transplantation or cirrhosis decompensation in the internal cohort were as follows: serum levels of bilirubin, aspartate aminotransferase, alanine aminotransferase, γ-glutamyl transferase, alkaline phosphatase, albumin, and Anali scores. Anali scores without and with gadolinium identified patients' survival without liver transplantation or cirrhosis decompensation with a c-statistic of 0.89 (95% CI, 0.84-0.95) and 0.75 (95% CI, 0.64-0.87), respectively. Independent prognostic factors identified by multivariate analysis were Anali scores and bilirubinemia. The prognostic value of Anali scores was confirmed in the external cohort. CONCLUSIONS: In internal and external cohorts, we found that Anali scores, determined from MRC, were associated with outcomes of patients with PSC. These scores might be used as prognostic factors.


Asunto(s)
Conductos Biliares Intrahepáticos/diagnóstico por imagen , Colangiografía , Colangitis Esclerosante/diagnóstico por imagen , Hipertensión Portal/diagnóstico por imagen , Hígado/diagnóstico por imagen , Imagen por Resonancia Magnética , Adulto , Atrofia , Conductos Biliares Intrahepáticos/patología , Colangitis Esclerosante/fisiopatología , Colangitis Esclerosante/cirugía , Dilatación Patológica , Progresión de la Enfermedad , Femenino , Humanos , Hígado/patología , Trasplante de Hígado , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos
12.
Am J Gastroenterol ; 114(12): 1878-1885, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31738286

RESUMEN

OBJECTIVES: Magnetic resonance (MR) risk scores and liver stiffness (LS) have individually been shown to predict clinical outcomes in primary sclerosing cholangitis (PSC). The aim of this study was to assess their complementary prognostic value. METHODS: Patients with PSC from 3 European centers with a 3-dimensional MR cholangiography available for central reviewing and a valid LS measurement assessed by vibration-controlled transient elastography by FibroScan performed within a 6-month interval were included in a longitudinal retrospective study. The MR score (Anali) without gadolinium (Gd) was calculated according to the formula: (1 × dilatation of intrahepatic bile ducts) + (2 × dysmorphy) + (1 × portal hypertension). The primary end point was survival without liver transplantation or cirrhosis decompensation. The prognostic values of LS and Anali score without Gd were assessed using Cox proportional hazard models. RESULTS: One hundred sixty-two patients were included. Over a total follow-up of 753 patient-years, 40 patients experienced an adverse outcome (4 liver transplantations, 6 liver-related deaths, and 30 cirrhosis decompensations). LS and Anali score without Gd were significantly correlated (ρ = 0.51, P < 0.001) and were independently associated with the occurrence of an adverse outcome. Optimal prognostic thresholds were 10.5 kPa for LS and 2 for the Anali score without Gd. Hazard ratios (95% confidence interval) were 2.07 (1.06-4.06) and 3.78 (1.67-8.59), respectively. The use in combination of these 2 thresholds allowed us to separate patients into low-, medium-, and high-risk groups for developing adverse outcomes. The 5-year cumulative rates of adverse outcome in these 3 groups were 8%, 16%, and 38% (P < 0.001), respectively. DISCUSSION: The combined use of MRI and vibration-controlled transient elastography permits easy risk stratification of patients with PSC.


Asunto(s)
Colangiografía , Colangitis Esclerosante/diagnóstico por imagen , Diagnóstico por Imagen de Elasticidad , Cirrosis Hepática Biliar/diagnóstico por imagen , Imagen por Resonancia Magnética , Adulto , Colangiocarcinoma/epidemiología , Colangiocarcinoma/mortalidad , Colangitis/mortalidad , Colangitis Esclerosante/epidemiología , Colangitis Esclerosante/mortalidad , Colangitis Esclerosante/cirugía , Comorbilidad , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/epidemiología , Hígado/diagnóstico por imagen , Cirrosis Hepática Biliar/epidemiología , Cirrosis Hepática Biliar/mortalidad , Cirrosis Hepática Biliar/cirugía , Trasplante de Hígado , Masculino , Persona de Mediana Edad , Pronóstico , Supervivencia sin Progresión , Medición de Riesgo , Choque Séptico/mortalidad , Vibración
13.
Curr Opin Gastroenterol ; 40(2): 61, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-38320196
14.
Curr Opin Gastroenterol ; 35(2): 88-92, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30585791

RESUMEN

PURPOSE OF REVIEW: Cystic fibrosis (CF; OMIM 219700) is caused by variations in the cystic fibrosis transmembrane conductance regulator gene. CF-related liver disease (CFLD) affects approximately one-third of patients with CF, but the severity of CFLD is highly variable. This review provides the latest knowledge in the pathophysiology and CF genetic modifier research in CFLD. RECENT FINDINGS: So far, the only modifier gene validated in CFLD is SERPINA1 (α-1-antitrypsin) Z allele. Recent studies support the view that cholangiopathy arising in CF is the result of an ill-adapted innate immune response to endotoxins coming from the intestine and triggering a pro-inflammatory response. SUMMARY: The pathophysiology of liver disease remains uncertain and so far, no therapy has proven effective to prevent the progression of CFLD. A better understanding of the pathophysiology and the effect of environmental and non-cystic fibrosis transmembrane conductance regulator genetic influences in the context of CFLD development would help improve management and develop new drug therapies.


Asunto(s)
Cirrosis Hepática/genética , Alelos , Animales , Genotipo , Humanos , Fenotipo , alfa 1-Antitripsina/genética
15.
Hepatology ; 65(2): 560-570, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28012258

RESUMEN

ABCB4 (MDR3) is an adenosine triphosphate (ATP)-binding cassette (ABC) transporter expressed at the canalicular membrane of hepatocytes, where it mediates phosphatidylcholine (PC) secretion. Variations in the ABCB4 gene are responsible for several biliary diseases, including progressive familial intrahepatic cholestasis type 3 (PFIC3), a rare disease that can be lethal in the absence of liver transplantation. In this study, we investigated the effect and potential rescue of ABCB4 missense variations that reside in the highly conserved motifs of ABC transporters, involved in ATP binding. Five disease-causing variations in these motifs have been identified in ABCB4 (G535D, G536R, S1076C, S1176L, and G1178S), three of which are homologous to the gating mutations of cystic fibrosis transmembrane conductance regulator (CFTR or ABCC7; i.e., G551D, S1251N, and G1349D), that were previously shown to be function defective and corrected by ivacaftor (VX-770; Kalydeco), a clinically approved CFTR potentiator. Three-dimensional structural modeling predicted that all five ABCB4 variants would disrupt critical interactions in the binding of ATP and thereby impair ATP-induced nucleotide-binding domain dimerization and ABCB4 function. This prediction was confirmed by expression in cell models, which showed that the ABCB4 mutants were normally processed and targeted to the plasma membrane, whereas their PC secretion activity was dramatically decreased. As also hypothesized on the basis of molecular modeling, PC secretion activity of the mutants was rescued by the CFTR potentiator, ivacaftor (VX-770). CONCLUSION: Disease-causing variations in the ATP-binding sites of ABCB4 cause defects in PC secretion, which can be rescued by ivacaftor. These results provide the first experimental evidence that ivacaftor is a potential therapy for selected patients who harbor mutations in the ATP-binding sites of ABCB4. (Hepatology 2017;65:560-570).


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Aminofenoles/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Mutagénesis/efectos de los fármacos , Quinolonas/farmacología , Adenosina Trifosfato/genética , Adolescente , Sitios de Unión , Células Cultivadas , Niño , Fibrosis Quística/patología , Femenino , Células Hep G2 , Humanos , Masculino , Mutación Missense/genética , Fosfatidilcolinas/metabolismo , Muestreo , Transfección , Adulto Joven
16.
Curr Opin Gastroenterol ; 39(2): 57-58, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36821451
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(7): 1699-1708, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28390947

RESUMEN

Nuclear receptors (NR), the largest family of transcription factors, control many physiological and pathological processes. To gain insight into hepatic NR and their potential as therapeutic targets in cholestatis, we determined their expression in individual cell types of the mouse liver in normal and cholestatic conditions. Hepatocytes, cholangiocytes, hepatic stellate cells (HSC), sinusoidal endothelial cells (SEC) and Kupffer cells (KC) were isolated from the liver of mice with acute or chronic cholestasis (i.e. bile duct-ligated or Abcb4-/- mice, respectively) and healthy controls. The expression of 43 out of the 49 NR was evidenced by RT-qPCR in one or several liver cell types. Expression of four NR was restricted to non-parenchymal liver cells. In normal conditions, NR were expressed at higher levels in individual cell types when compared to total liver. Half of the NR expressed in the liver had maximal expression in non-parenchymal cells. After bile duct ligation, NR mRNA changes occurred mostly in non-parenchymal cells and mainly consisted in down-regulations. In Abcb4-/- mice, NR mRNA changes were equally frequent in hepatocytes and non-parenchymal cells. Essentially down-regulations were found in hepatocytes, HSC and cholangiocytes, as opposed to up-regulations in SEC and KC. While undetectable in total liver, Vdr expression was up-regulated in all non-parenchymal cells in Abcb4-/- mice. In conclusion, non-parenchymal liver cells are a major site of NR expression. During cholestasis, NR expression is markedly altered mainly by down-regulations, suggesting major changes in metabolic activity. Thus, non-parenchymal cells are important new targets to consider in NR-directed therapies.


Asunto(s)
Colestasis/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Transcriptoma , Subfamilia B de Transportador de Casetes de Unión a ATP/deficiencia , Animales , Colestasis/genética , Colestasis/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hepatocitos/patología , Hígado/patología , Ratones , Ratones Noqueados , Receptores Citoplasmáticos y Nucleares/genética , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
18.
Hepatology ; 63(5): 1620-31, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26474921

RESUMEN

UNLABELLED: Progressive familial intrahepatic cholestasis type 3 is caused by biallelic variations of ABCB4, most often (≥70%) missense. In this study, we examined the effects of 12 missense variations identified in progressive familial intrahepatic cholestasis type 3 patients. We classified these variations on the basis of the defects thus identified and explored potential rescue of trafficking-defective mutants by pharmacological means. Variations were reproduced in the ABCB4 complementary DNA and the mutants, thus obtained, expressed in HepG2 and HEK293 cells. Three mutants were either fully (I541F and L556R) or largely (Q855L) retained in the endoplasmic reticulum, in an immature form. Rescue of the defect, i.e., increase in the mature form at the bile canaliculi, was obtained by cell treatments with cyclosporin A or C and, to a lesser extent, B, D, or H. Five mutations with little or no effect on ABCB4 expression at the bile canaliculi caused a decrease (F357L, T775M, and G954S) or almost absence (S346I and P726L) of phosphatidylcholine secretion. Two mutants (T424A and N510S) were normally processed and expressed at the bile canaliculi, but their stability was reduced. We found no defect of the T175A mutant or of R652G, previously described as a polymorphism. In patients, the most severe phenotypes appreciated by the duration of transplant-free survival were caused by ABCB4 variants that were markedly retained in the endoplasmic reticulum and expressed in a homozygous status. CONCLUSION: ABCB4 variations can be classified as follows: nonsense variations (I) and, on the basis of current findings, missense variations that primarily affect the maturation (II), activity (III), or stability (IV) of the protein or have no detectable effect (V); this classification provides a strong basis for the development of genotype-based therapies.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/deficiencia , Colestasis Intrahepática/genética , Mutación , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Ciclosporina/farmacología , Células HEK293 , Células Hep G2 , Humanos , Fosfatidilcolinas/metabolismo
20.
J Pediatr Gastroenterol Nutr ; 65(4): 443-448, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28753176

RESUMEN

OBJECTIVES: Hepatobiliary complications are a leading cause of morbidity and mortality in cystic fibrosis (CF) patients. Knowledge of the underlying pathological aspects and optimal clinical management is, however, sorely lacking. METHODS: We provide a summary of the lectures given by international speakers at the European Society for Paediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN) monothematic conference on cystic fibrosis-related liver disease (CFLD) held in Paris in January 2016, to discuss the status of our current knowledge of liver disease in CF patients, to define the critical areas that need to be addressed, and to resolve actions to elucidate relevant mechanisms of disease to optimise future therapeutic options. CONCLUSIONS: The need for a universal consensus on the definition of CFLD to clarify disease stage and to identify relevant biomarkers to assess disease severity was highlighted. A deeper understanding of the pathophysiology and prognostic factors for the long-term evolution of CFLD is fundamental to move forward and has a strong bearing on identifying potential treatments. Novel experimental models and new treatment options under investigation are discussed and offer hope for the near future of CFLD.


Asunto(s)
Fibrosis Quística/complicaciones , Hepatopatías/etiología , Biomarcadores/metabolismo , Fibrosis Quística/diagnóstico , Fibrosis Quística/metabolismo , Fibrosis Quística/terapia , Fármacos Gastrointestinales/uso terapéutico , Humanos , Hepatopatías/diagnóstico , Hepatopatías/metabolismo , Hepatopatías/terapia , Trasplante de Hígado , Trasplante de Páncreas , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA