Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
JHEP Rep ; 4(5): 100463, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35462858

RESUMEN

Background & Aims: Organic solute transporter (OST) subunits OSTα and OSTß facilitate bile acid efflux from the enterocyte into the portal circulation. Patients with deficiency of OSTα or OSTß display considerable variation in the level of bile acid malabsorption, chronic diarrhea, and signs of cholestasis. Herein, we generated and characterized a mouse model of OSTß deficiency. Methods: Ostß -/- mice were generated using CRISR/Cas9 and compared to wild-type and Ostα -/- mice. OSTß was re-expressed in livers of Ostß -/- mice using adeno-associated virus serotype 8 vectors. Cholestasis was induced in both models by bile duct ligation (BDL) or 3.5-diethoxycarbonyl-1.4-dihydrocollidine (DDC) feeding. Results: Similar to Ostα -/- mice, Ostß -/- mice exhibited elongated small intestines with blunted villi and increased crypt depth. Increased expression levels of ileal Fgf15, and decreased Asbt expression in Ostß -/- mice indicate the accumulation of bile acids in the enterocyte. In contrast to Ostα -/- mice, induction of cholestasis in Ostß -/- mice by BDL or DDC diet led to lower survival rates and severe body weight loss, but an improved liver phenotype. Restoration of hepatic Ostß expression via adeno-associated virus-mediated overexpression did not rescue the phenotype of Ostß -/- mice. Conclusions: OSTß is pivotal for bile acid transport in the ileum and its deficiency leads to an intestinal phenotype similar to Ostα -/- mice, but it exerts distinct effects on survival and the liver phenotype, independent of its expression in the liver. Our findings provide insights into the variable clinical presentation of patients with OSTα and OSTß deficiencies. Lay summary: Organic solute transporter (OST) subunits OSTα and OSTß together facilitate the efflux of conjugated bile acids into the portal circulation. Ostα knockout mice have longer and thicker small intestines and are largely protected against experimental cholestatic liver injury. Herein, we generated and characterized Ostß knockout mice for the first time. Ostα and Ostß knockout mice shared a similar phenotype under normal conditions. However, in cholestasis, Ostß knockout mice had a worsened overall phenotype which indicates a separate and specific role of OSTß, possibly as an interacting partner of other intestinal proteins.

2.
Biochim Biophys Acta Mol Basis Dis ; 1867(8): 166153, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33895309

RESUMEN

IsoBAs, stereoisomers of primary and secondary BAs, are found in feces and plasma of human individuals. BA signaling via the nuclear receptor FXR is crucial for regulation of hepatic and intestinal physiology/pathophysiology. AIM: Investigate the ability of BA-stereoisomers to bind and modulate FXR under physiological/pathological conditions. METHODS: Expression-profiling, luciferase-assays, fluorescence-based coactivator-association assays, administration of (iso)-BAs to WT and cholestatic mice. RESULTS: Compared to CDCA/isoCDCA, administration of DCA/isoDCA, UDCA/isoUDCA only slightly increased mRNA expression of FXR target genes; the induction was more evident looking at pre-mRNAs. Notably, almost 50% of isoBAs were metabolized to 3-oxo-BAs within 4 h in cell-based assays, making it difficult to study their actions. FRET-based real-time monitoring of FXR activity revealed that isoCDCA>CDCA stimulated FXR, and isoDCA and isoUDCA allowed fully activated FXR to be re-stimulated by a second dose of GW4064. In vivo co-administration of a single dose of isoBAs followed by GW4064 cooperatively activated FXR, as did feeding of UDCA in a background of endogenous FXR ligands. However, in animals with biliary obstruction and concomitant loss of intestinal BAs, UDCA was unable to increase intestinal Fgf15. In contrast, mice with an impaired enterohepatic circulation of BAs (Asbt-/-, Ostα-/-), administration of UDCA was still able to induce ileal Fgf15 and repress hepatic BA-synthesis, arguing that UDCA is only effective in the presence of endogenous FXR ligands. CONCLUSION: Secondary (iso)BAs cooperatively activate FXR in the presence of endogenous BAs, which is important to consider in diseases linked to disturbances in BA enterohepatic cycling.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Células CACO-2 , Línea Celular , Línea Celular Tumoral , Colestasis/tratamiento farmacológico , Colestasis/metabolismo , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Íleon/efectos de los fármacos , Íleon/metabolismo , Isoxazoles/farmacología , Ligandos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
3.
Sci Rep ; 9(1): 2193, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30778102

RESUMEN

The farnesoid X receptor (FXR) belongs to the nuclear receptor family and is activated by bile acids. Multiple, chemically rather diverse, FXR agonists have been developed and several of these compounds are currently tested in clinical trials for NAFLD and cholestasis. Here, we investigated possible FXR-agonism or antagonism of existing FDA/EMA-approved drugs. By using our recently developed FRET-sensor, containing the ligand binding domain of FXR (FXR-LBD), 1280 FDA-approved drugs were screened for their ability to activate FXR in living cells using flow cytometry. Fifteen compounds induced the sensor for more than twenty percent above background. Real-time confocal microscopy confirmed that avermectin B1a, gliquidone, nicardipine, bepridil and triclosan activated the FRET sensor within two minutes. These compounds, including fluticasone, increased mRNA expression of FXR target genes OSTα and OSTß in Huh7 cells, and in most cases also of MRP2, SHP and FGF19. Finally, avermectin B1a, gliquidone, nicardipine and bepridil significantly increased IBABP promoter activity in a luciferase reporter assay in a dose-dependent manner. In conclusion, six FDA/EMA-approved drugs currently used in the clinical practice exhibit moderate agonistic FXR activity. This may on the one hand explain (undesired) side-effects, but on the other hand may form an opportunity for polypharmacology.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/agonistas , Ácidos y Sales Biliares/metabolismo , Técnicas Biosensibles , Línea Celular , Aprobación de Drogas , Reposicionamiento de Medicamentos , Transferencia Resonante de Energía de Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Humanos , Isoxazoles/química , Isoxazoles/farmacología , Ivermectina/análogos & derivados , Ivermectina/química , Ivermectina/farmacología , Ligandos , Estructura Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Estados Unidos , United States Food and Drug Administration
4.
Cell Mol Gastroenterol Hepatol ; 5(3): 223-237, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29675448

RESUMEN

BACKGROUND & AIMS: The organic solute transporter α-ß (OSTα-OSTß) mainly facilitates transport of bile acids across the basolateral membrane of ileal enterocytes. Therefore, inhibition of OSTα-OSTß might have similar beneficial metabolic effects as intestine-specific agonists of the major nuclear receptor for bile acids, the farnesoid X receptor (FXR). However, no OSTα-OSTß inhibitors have yet been identified. METHODS: Here, we developed a screen to identify specific inhibitors of OSTα-OSTß using a genetically encoded Förster Resonance Energy Transfer (FRET)-bile acid sensor that enables rapid visualization of bile acid efflux in living cells. RESULTS: As proof of concept, we screened 1280 Food and Drug Administration-approved drugs of the Prestwick chemical library. Clofazimine was the most specific hit for OSTα-OSTß and reduced transcellular transport of taurocholate across Madin-Darby canine kidney epithelial cell monolayers expressing apical sodium bile acid transporter and OSTα-OSTß in a dose-dependent manner. Moreover, pharmacologic inhibition of OSTα-OSTß also moderately increased intracellular taurocholate levels and increased activation of intestinal FXR target genes. Oral administration of clofazimine in mice (transiently) increased intestinal FXR target gene expression, confirming OSTα-OSTß inhibition in vivo. CONCLUSIONS: This study identifies clofazimine as an inhibitor of OSTα-OSTß in vitro and in vivo, validates OSTα-OSTß as a drug target to enhance intestinal bile acid signaling, and confirmed the applicability of the Förster Resonance Energy Transfer-bile acid sensor to screen for inhibitors of bile acid efflux pathways.

5.
Expert Opin Drug Discov ; 9(11): 1293-306, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25217125

RESUMEN

INTRODUCTION: There are currently no treatments approved by the FDA to effectively treat cocaine dependence. Research of recent years has gradually revealed the importance of 5-hydroxytryptamine (5-HT) in the reinforcing and rewarding effects of cocaine and the potential for relapse. Brain-derived neurotropic factor (BDNF) is an important modulator of the serotonergic system and 5-HT modulates BDNF expression. Their reciprocal interaction is of crucial importance for synaptic plasticity during long-term cocaine intake. Thus, agents modifying BDNF-5-HT interactions might have therapeutic potential for cocaine dependence by reversing the altered brain structure that underlies relapse after cocaine withdrawal. AREAS COVERED: On the basis of the available literature, the authors propose an interaction between BDNF and the serotonergic system in the response to cocaine and during cocaine intake. Furthermore, they discuss putative therapies that are based on 5-HT and BDNF. EXPERT OPINION: Recent studies are beginning to elucidate the role of 5-HT and BDNF in cocaine addiction. Additionally, animal studies modeling addiction-like drug intake will only further help to gain a better understanding of how to treat cocaine addiction. Based on the current evidence, the authors believe that BDNF, as a modulator of the serotonergic pathway, or 5-HT, as a modulator of the BDNF system, represent a valuable target to treat drug addiction, which may yield novel therapeutics in the future.


Asunto(s)
Trastornos Relacionados con Cocaína/tratamiento farmacológico , Serotoninérgicos/uso terapéutico , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos Relacionados con Cocaína/metabolismo , Diseño de Fármacos , Humanos , Serotonina/metabolismo , Transducción de Señal
6.
Endocrinology ; 155(6): 2320-30, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24693966

RESUMEN

Allan-Herndon-Dudley syndrome (AHDS) is an inherited disorder of brain development characterized by severe psychomotor retardation. This X-linked disease is caused by mutations in the monocarboxylate transporter 8 (MCT8), an important thyroid hormone transporter in brain neurons. MCT8-knockout mice lack the 2 major neurological symptoms of AHDS, namely locomotor problems and cognitive impairment. The pathological mechanism explaining the symptoms is still obscure, and no cure for this condition is known. The development of an animal model that carries MCT8-related neurological symptoms is warranted. We have employed morpholino-based gene knockdown to create zebrafish deficient for mct8. Knockdown of mct8 results in specific symptoms in the thyroid axis and brain. The mct8-morphants showed impaired locomotor behavior and brain development. More specifically, we observed maldevelopment of the cerebellum and mid-hindbrain boundary and apoptotic clusters in the zebrafish brain. The mRNA expression of zebrafish orthologs of mammalian TSH, thyroid hormone transporters, and deiodinases was altered in mct8 morphants. In particular, deiodinase type 3 gene expression was consistently up-regulated in zebrafish mct8 morphants. The thyroid hormone metabolite tetrac, but not T3, partly ameliorated the affected phenotype and locomotion disability of morphant larvae. Our results show that mct8 knockdown in zebrafish larvae results in disturbances in the thyroid axis, brain, and locomotion behavior, which is congruent with the clinical aspect of impaired locomotion and cognition in patients with AHDS. Taken together, the zebrafish is a suitable animal model for the study of the pathophysiology of AHDS.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Inmunohistoquímica , Locomoción/genética , Locomoción/fisiología , Ratones , Transportadores de Ácidos Monocarboxílicos/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA