Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Am J Pathol ; 193(1): 11-26, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36243043

RESUMEN

Patients with cholestatic liver disease, including those with primary biliary cholangitis, can experience symptoms of impaired cognition or brain fog. This phenomenon remains unexplained and is currently untreatable. Bile duct ligation (BDL) is an established rodent model of cholestasis. In addition to liver changes, BDL animals develop cognitive symptoms early in the disease process (before development of cirrhosis and/or liver failure). The cellular mechanisms underpinning these cognitive symptoms are poorly understood. Herein, the study explored the neurocognitive symptom manifestations, and tested potential therapies, in BDL mice, and used human neuronal cell cultures to explore translatability to humans. BDL animals exhibited short-term memory loss and showed reduced astrocyte coverage of the blood-brain barrier, destabilized hippocampal network activity, and neuronal senescence. Ursodeoxycholic acid (first-line therapy for most human cholestatic diseases) did not reverse symptomatic or mechanistic aspects. In contrast, obeticholic acid (OCA), a farnesoid X receptor agonist and second-line anti-cholestatic agent, normalized memory function, suppressed blood-brain barrier changes, prevented hippocampal network deficits, and reversed neuronal senescence. Co-culture of human neuronal cells with either BDL or human cholestatic patient serum induced cellular senescence and increased mitochondrial respiration, changes that were limited again by OCA. These findings provide new insights into the mechanism of cognitive symptoms in BDL animals, suggesting that OCA therapy or farnesoid X receptor agonism could be used to limit cholestasis-induced neuronal senescence.


Asunto(s)
Colestasis , Memoria a Corto Plazo , Humanos , Ratones , Animales , Colestasis/tratamiento farmacológico , Ácido Quenodesoxicólico/farmacología , Conductos Biliares/cirugía , Hígado , Ligadura
2.
Hepatology ; 74(6): 3441-3459, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34233024

RESUMEN

BACKGROUND AND AIMS: NAFLD is the most common hepatic pathology in western countries and no treatment is currently available. NAFLD is characterized by the aberrant hepatocellular accumulation of fatty acids in the form of lipid droplets (LDs). Recently, it was shown that liver LD degradation occurs through a process termed lipophagy, a form of autophagy. However, the molecular mechanisms governing liver lipophagy are elusive. Here, we aimed to ascertain the key molecular players that regulate hepatic lipophagy and their importance in NAFLD. APPROACH AND RESULTS: We analyzed the formation and degradation of LD in vitro (fibroblasts and primary mouse hepatocytes), in vivo and ex vivo (mouse and human liver slices) and focused on the role of the autophagy master regulator mammalian target of rapamycin complex (mTORC) 1 and the LD coating protein perilipin (Plin) 3 in these processes. We show that the autophagy machinery is recruited to the LD on hepatic overload of oleic acid in all experimental settings. This led to activation of lipophagy, a process that was abolished by Plin3 knockdown using RNA interference. Furthermore, Plin3 directly interacted with the autophagy proteins focal adhesion interaction protein 200 KDa and autophagy-related 16L, suggesting that Plin3 functions as a docking protein or is involved in autophagosome formation to activate lipophagy. Finally, we show that mTORC1 phosphorylated Plin3 to promote LD degradation. CONCLUSIONS: These results reveal that mTORC1 regulates liver lipophagy through a mechanism dependent on Plin3 phosphorylation. We propose that stimulating this pathway can enhance lipophagy in hepatocytes to help protect the liver from lipid-mediated toxicity, thus offering a therapeutic strategy in NAFLD.


Asunto(s)
Autofagia , Hígado Graso/metabolismo , Hepatocitos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Perilipina-3/metabolismo , Transducción de Señal , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
3.
Eur J Pharmacol ; 913: 174618, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34762934

RESUMEN

Fibrosis is the formation of scar tissue due to injury or long-term inflammation and is a leading cause of morbidity and mortality. Activation of the pro-fibrotic cytokine transforming growth factor-ß (TGFß) via the alpha-V beta-6 (αvß6) integrin has been identified as playing a key role in the development of fibrosis. Therefore, a drug discovery programme to identify an orally bioavailable small molecule αvß6 arginyl-glycinyl-aspartic acid (RGD)-mimetic was initiated. As part of a medicinal chemistry programme GSK3335103 was identified and profiled in a range of pre-clinical in vitro and in vivo systems. GSK3335103 was shown to bind to the αvß6 with high affinity and demonstrated fast binding kinetics. In primary human lung epithelial cells, GSK3335103-induced concentration- and time-dependent internalisation of αvß6 with a rapid return of integrin to the cell surface observed after washout. Following sustained engagement of the αvß6 integrin in vitro, lysosomal degradation was induced by GSK3335103. GSK3335103 was shown to engage with the αvß6 integrin and inhibit the activation of TGFß in both ex vivo IPF tissue and in a murine model of bleomycin-induced lung fibrosis, as measured by αvß6 engagement, TGFß signalling and collagen deposition, with a prolonged duration of action observed in vivo. In summary, GSK3335103 is a potent αvß6 inhibitor that attenuates TGFß signalling in vitro and in vivo with a well-defined pharmacokinetic/pharmacodynamic relationship. This translates to a significant reduction of collagen deposition in vivo and therefore GSK3335103 represents a potential novel oral therapy for fibrotic disorders.


Asunto(s)
Antifibróticos/farmacología , Integrinas/antagonistas & inhibidores , Fibrosis Pulmonar/tratamiento farmacológico , Administración Oral , Animales , Antifibróticos/química , Antifibróticos/uso terapéutico , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Disponibilidad Biológica , Bleomicina/administración & dosificación , Bleomicina/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Integrinas/química , Integrinas/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Lisosomas/metabolismo , Masculino , Ratones , Oligopéptidos/química , Cultivo Primario de Células , Proteolisis/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Factor de Crecimiento Transformador beta/metabolismo
4.
JCI Insight ; 5(4)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32102985

RESUMEN

Neutrophils are the most abundant inflammatory cells at the earliest stages of wound healing and play important roles in wound repair and fibrosis. Formyl peptide receptor 1 (FPR-1) is abundantly expressed on neutrophils and has been shown to regulate their function, yet the importance of FPR-1 in fibrosis remains ill defined. FPR-1-deficient (fpr1-/-) mice were protected from bleomycin-induced pulmonary fibrosis but developed renal and hepatic fibrosis normally. Mechanistically, we observed a failure to effectively recruit neutrophils to the lungs of fpr1-/- mice, whereas neutrophil recruitment was unaffected in the liver and kidney. Using an adoptive transfer model we demonstrated that the defect in neutrophil recruitment to the lung was intrinsic to the fpr1-/- neutrophils, as C57BL/6 neutrophils were recruited normally to the damaged lung in fpr1-/- mice. Finally, C57BL/6 mice in which neutrophils had been depleted were protected from pulmonary fibrosis. In conclusion, FPR-1 and FPR-1 ligands are required for effective neutrophil recruitment to the damaged lung. Failure to recruit neutrophils or depletion of neutrophils protects from pulmonary fibrosis.


Asunto(s)
Infiltración Neutrófila/fisiología , Fibrosis Pulmonar/fisiopatología , Receptores de Formil Péptido/fisiología , Animales , Bleomicina/toxicidad , Humanos , Ligandos , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo
5.
Nat Commun ; 11(1): 4659, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32938936

RESUMEN

The αvß6 integrin plays a key role in the activation of transforming growth factor-ß (TGFß), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvß6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvß6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFß signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvß6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvß6, induces prolonged inhibition of TGFß signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy.


Asunto(s)
Butiratos/farmacología , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Integrinas/antagonistas & inhibidores , Naftiridinas/farmacología , Pirazoles/farmacología , Pirrolidinas/farmacología , Administración por Inhalación , Animales , Antígenos de Neoplasias/metabolismo , Bleomicina/toxicidad , Butiratos/administración & dosificación , Butiratos/metabolismo , Butiratos/farmacocinética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/patología , Integrinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Naftiridinas/administración & dosificación , Naftiridinas/metabolismo , Naftiridinas/farmacocinética , Pirazoles/administración & dosificación , Pirazoles/metabolismo , Pirazoles/farmacocinética , Pirrolidinas/administración & dosificación , Pirrolidinas/metabolismo , Pirrolidinas/farmacocinética , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Tomografía Computarizada de Emisión de Fotón Único , Factor de Crecimiento Transformador beta/metabolismo , Investigación Biomédica Traslacional
6.
Nat Metab ; 2(11): 1350-1367, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33168981

RESUMEN

Fibrosis is a common pathological feature of chronic disease. Deletion of the NF-κB subunit c-Rel limits fibrosis in multiple organs, although the mechanistic nature of this protection is unresolved. Using cell-specific gene-targeting manipulations in mice undergoing liver damage, we elucidate a critical role for c-Rel in controlling metabolic changes required for inflammatory and fibrogenic activities of hepatocytes and macrophages and identify Pfkfb3 as the key downstream metabolic mediator of this response. Independent deletions of Rel in hepatocytes or macrophages suppressed liver fibrosis induced by carbon tetrachloride, while combined deletion had an additive anti-fibrogenic effect. In transforming growth factor-ß1-induced hepatocytes, c-Rel regulates expression of a pro-fibrogenic secretome comprising inflammatory molecules and connective tissue growth factor, the latter promoting collagen secretion from HMs. Macrophages lacking c-Rel fail to polarize to M1 or M2 states, explaining reduced fibrosis in RelΔLysM mice. Pharmacological inhibition of c-Rel attenuated multi-organ fibrosis in both murine and human fibrosis. In conclusion, activation of c-Rel/Pfkfb3 in damaged tissue instigates a paracrine signalling network among epithelial, myeloid and mesenchymal cells to stimulate fibrogenesis. Targeting the c-Rel-Pfkfb3 axis has potential for therapeutic applications in fibrotic disease.


Asunto(s)
Epitelio/patología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Macrófagos/patología , Proteínas Proto-Oncogénicas c-rel/genética , Animales , Polaridad Celular/genética , Marcación de Gen , Hepatocitos/patología , Hidroxiprolina/metabolismo , Cirrosis Hepática/prevención & control , Regeneración Hepática/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis/genética , Comunicación Paracrina/genética , Fosfofructoquinasa-2/genética , Proteínas Proto-Oncogénicas c-rel/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-rel/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA