Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 9(8)2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38470490

RESUMEN

Excessive lipolysis in white adipose tissue (WAT) leads to insulin resistance (IR) and ectopic fat accumulation in insulin-sensitive tissues. However, the impact of Gi-coupled receptors in restraining adipocyte lipolysis through inhibition of cAMP production remained poorly elucidated. Given that the Gi-coupled P2Y13 receptor (P2Y13-R) is a purinergic receptor expressed in WAT, we investigated its role in adipocyte lipolysis and its effect on IR and metabolic dysfunction-associated steatotic liver disease (MASLD). In humans, mRNA expression of P2Y13-R in WAT was negatively correlated to adipocyte lipolysis. In mice, adipocytes lacking P2Y13-R displayed higher intracellular cAMP levels, indicating impaired Gi signaling. Consistently, the absence of P2Y13-R was linked to increased lipolysis in adipocytes and WAT explants via hormone-sensitive lipase activation. Metabolic studies indicated that mice lacking P2Y13-R showed a greater susceptibility to diet-induced IR, systemic inflammation, and MASLD compared with their wild-type counterparts. Assays conducted on precision-cut liver slices exposed to WAT conditioned medium and on liver-specific P2Y13-R-knockdown mice suggested that P2Y13-R activity in WAT protects from hepatic steatosis, independently of liver P2Y13-R expression. In conclusion, our findings support the idea that targeting adipose P2Y13-R activity may represent a pharmacological strategy to prevent obesity-associated disorders, including type 2 diabetes and MASLD.


Asunto(s)
Adipocitos , Tejido Adiposo Blanco , Hígado Graso , Resistencia a la Insulina , Lipólisis , Receptores Purinérgicos P2 , Animales , Femenino , Humanos , Masculino , Ratones , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Tejido Adiposo Blanco/metabolismo , Hígado Graso/metabolismo , Hígado Graso/genética , Hígado Graso/patología , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/deficiencia
2.
Cell Physiol Biochem ; 56(1): 28-38, 2022 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-35060690

RESUMEN

BACKGROUND/AIMS: Osteoprotegerin (OPG) is a profibrotic mediator produced by myofibro-blasts under influence of transforming growth factor ß (TGFß). Its expression in experimental models of liver fibrosis correlates well with disease severity and treatment responses. The regulation of OPG in liver tissue is largely unknown and we therefore set out to elucidate which growth factors/interleukins associated with fibrosis induce OPG and through which pathways. METHODS: Precision-cut liver slices of wild type and STAT6-deficient mice and 3T3 fibroblasts were used to investigate the effects of TGFß, interleukin (IL) 13 (IL13), IL1ß, and platelet-derived growth factor BB (PDGF-BB) on expression of OPG. OPG protein was measure by ELISA, whereas OPG mRNA and expression of other relevant genes was measured by qPCR. RESULTS: In addition to TGFß, only IL13 and not PDGF-BB or IL1ß could induce OPG expression in 3T3 fibroblasts and liver slices. This IL13-dependent induction was not shown in liver slices of STAT6-deficient mice and when wild type slices were cotreated with TGFß receptor 1 kinase inhibitor galunisertib, STAT6 inhibitor AS1517499, or AP1 inhibitor T5224. This suggests that the OPG-inducing effect of IL13 is mediated through IL13 receptor α1-activation and subsequent STAT6-dependent upregulation of IL13 receptor α2, which in turn activates AP1 and induces production of TGFß and subsequent production of OPG. CONCLUSION: We have shown that IL13 induces OPG release by liver tissue through a TGFß-dependent pathway involving both the α1 and the α2 receptor of IL13 and transcription factors STAT6 and AP1. OPG may therefore be a novel target for the treatment liver fibrosis as it is mechanistically linked to two important regulators of fibrosis in liver, namely IL13 and TGFß1.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-13/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Osteoprotegerina/biosíntesis , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Femenino , Masculino , Ratones
3.
Antioxid Redox Signal ; 37(4-6): 370-393, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34605675

RESUMEN

Significance: The mitochondrial oxidative phosphorylation (OXPHOS) system, comprising the electron transport chain and ATP synthase, generates membrane potential, drives ATP synthesis, governs energy metabolism, and maintains redox balance. OXPHOS dysfunction is associated with a plethora of diseases ranging from rare inherited disorders to common conditions, including diabetes, cancer, neurodegenerative diseases, as well as aging. There has been great interest in studying regulators of OXPHOS. Among these, ATPase inhibitory factor 1 (IF1) is an endogenous inhibitor of ATP synthase that has long been thought to avoid the consumption of cellular ATP when ATP synthase acts as an ATP hydrolysis enzyme. Recent Advances: Recent data indicate that IF1 inhibits ATP synthesis and is involved in a multitude of mitochondrial-related functions, such as mitochondrial quality control, energy metabolism, redox balance, and cell fate. IF1 also inhibits the ATPase activity of cell-surface ATP synthase, and it is used as a cardiovascular disease biomarker. Critical Issues: Although recent data have led to a paradigm shift regarding IF1 functions, these have been poorly studied in entire organisms and in different organs. The understanding of the cellular biology of IF1 is, therefore, still limited. The aim of this review was to provide an overview of the current understanding of the role of IF1 in mitochondrial functions, health, and diseases. Future Directions: Further investigations of IF1 functions at the cell, organ, and whole-organism levels and in different pathophysiological conditions will help decipher the controversies surrounding its involvement in mitochondrial function and could unveil therapeutic strategies in human pathology. Antioxid. Redox Signal. 37, 370-393.


Asunto(s)
ATPasas de Translocación de Protón Mitocondriales , Proteínas , Adenosina Trifosfato/metabolismo , Metabolismo Energético , Humanos , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Proteínas/metabolismo
4.
Am J Physiol Gastrointest Liver Physiol ; 318(2): G336-G351, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31905025

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease, characterized by excess fat accumulation (steatosis). Nonalcoholic steatohepatitis (NASH) develops in 15-20% of NAFLD patients and frequently progresses to liver fibrosis and cirrhosis. We aimed to develop an ex vivo model of inflammation and fibrosis in steatotic murine precision-cut liver slices (PCLS). NASH was induced in C57Bl/6 mice on an amylin and choline-deficient l-amino acid-defined (CDAA) diet. PCLS were prepared from steatohepatitic (sPCLS) and control (cPCLS) livers and cultured for 48 h with LPS, TGFß1, or elafibranor. Additionally, C57Bl/6 mice were placed on CDAA diet for 12 wk to receive elafibranor or vehicle from weeks 7 to 12. Effects were assessed by transcriptome analysis and procollagen Iα1 protein production. The diets induced features of human NASH. Upon culture, all PCLS showed an increased gene expression of fibrosis- and inflammation-related markers but decreased lipid metabolism markers. LPS and TGFß1 affected sPCLS more pronouncedly than cPCLS. TGFß1 increased procollagen Iα1 solely in cPCLS. Elafibranor ameliorated fibrosis and inflammation in vivo but not ex vivo, where it only increased the expression of genes modulated by PPARα. sPCLS culture induced inflammation-, fibrosis-, and lipid metabolism-related transcripts, explained by spontaneous activation. sPCLS remained responsive to proinflammatory and profibrotic stimuli on gene expression. We consider that PCLS represent a useful tool to reproducibly study NASH progression. sPCLS can be used to evaluate potential treatments for NASH, as demonstrated in our elafibranor study, and serves as a model to bridge results from rodent studies to the human system.NEW & NOTEWORTHY This study showed that nonalcoholic steatohepatitis can be studied ex vivo in precision-cut liver slices obtained from murine diet-induced fatty livers. Liver slices develop a spontaneous inflammatory and fibrogenic response during culture that can be augmented with specific modulators. Additionally, the model can be used to test the efficacy of pharmaceutical compounds (as shown in this investigation with elafibranor) and could be a tool for preclinical assessment of potential therapies.


Asunto(s)
Inflamación/patología , Cirrosis Hepática/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Células Cultivadas , Chalconas/farmacología , Colágeno Tipo I/biosíntesis , Dieta , Modelos Animales de Enfermedad , Técnicas In Vitro , Metabolismo de los Lípidos/genética , Lipopolisacáridos/farmacología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Propionatos/farmacología , Transcriptoma/genética , Factor de Crecimiento Transformador beta1/farmacología
5.
Biochim Biophys Acta Mol Basis Dis ; 1866(1): 165582, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676376

RESUMEN

Fibrosis is the hallmark of pathologic tissue remodelling in most chronic diseases. Despite advances in our understanding of the mechanisms of fibrosis, it remains uncured. Fibrogenic processes share conserved core cellular and molecular pathways across organs. In this study, we aimed to elucidate shared and organ-specific features of fibrosis using murine precision-cut tissue slices (PCTS) prepared from small intestine, liver and kidneys. PCTS displayed substantial differences in their baseline gene expression profiles: 70% of the extracellular matrix (ECM)-related genes were differentially expressed across the organs. Culture for 48 h induced significant changes in ECM regulation and triggered the onset of fibrogenesis in all PCTS in organ-specific manner. TGFß signalling was activated during 48 h culture in all PCTS. However, the degree of its involvement varied: both canonical and non-canonical TGFß pathways were activated in liver and kidney slices, while only canonical, Smad-dependent, cascade was involved in intestinal slices. The treatment with galunisertib blocked the TGFßRI/SMAD2 signalling in all PCTS, but attenuated culture-induced dysregulation of ECM homeostasis and mitigated the onset of fibrogenesis with organ-specificity. In conclusion, regardless the many common features in pathophysiology of organ fibrosis, PCTS displayed diversity in culture-induced responses and in response to the treatment with TGFßRI kinase inhibitor galunisertib, even though it targets a core fibrosis pathway. A clear understanding of the common and organ-specific features of fibrosis is the basis for developing novel antifibrotic therapies.


Asunto(s)
Fibrosis/patología , Cirrosis Hepática/patología , Hígado/patología , Animales , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Quinolinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
6.
Arch Toxicol ; 93(12): 3549-3583, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31754732

RESUMEN

Our knowledge of complex pathological mechanisms underlying organ fibrosis is predominantly derived from animal studies. However, relevance of animal models for human disease is limited; therefore, an ex vivo model of human precision-cut tissue slices (PCTS) might become an indispensable tool in fibrosis research and drug development by bridging the animal-human translational gap. This study, presented as two parts, provides comprehensive characterization of the dynamic transcriptional changes in PCTS during culture by RNA sequencing. Part I investigates the differences in culture-induced responses in murine and human PCTS derived from healthy liver, kidney and gut. Part II delineates the molecular processes in cultured human PCTS generated from diseased liver, kidney and ileum. We demonstrated that culture was associated with extensive transcriptional changes and impacted PCTS in a universal way across the organs and two species by triggering an inflammatory response and fibrosis-related extracellular matrix (ECM) remodelling. All PCTS shared mRNA upregulation of IL-11 and ECM-degrading enzymes MMP3 and MMP10. Slice preparation and culturing activated numerous pathways across all PCTS, especially those involved in inflammation (IL-6, IL-8 and HMGB1 signalling) and tissue remodelling (osteoarthritis pathway and integrin signalling). Despite the converging effects of culture, PCTS display species-, organ- and pathology-specific differences in the regulation of genes and canonical pathways. The underlying pathology in human diseased PCTS endures and influences biological processes like cytokine release. Our study reinforces the use of PCTS as an ex vivo fibrosis model and supports future studies towards its validation as a preclinical tool for drug development.


Asunto(s)
Técnicas de Cultivo de Órganos/métodos , Transcriptoma/genética , Animales , Análisis por Conglomerados , Fibrosis/patología , Perfilación de la Expresión Génica/estadística & datos numéricos , Regulación de la Expresión Génica , Humanos , Masculino , Redes y Vías Metabólicas , Ratones Endogámicos C57BL , Análisis de Componente Principal , Análisis de Secuencia de ARN
7.
Biochem Pharmacol ; 169: 113633, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31494146

RESUMEN

BACKGROUND: Liver fibrosis results from continuous inflammation and injury. Despite its high prevalence worldwide, no approved antifibrotic therapies exist. Omipalisib is a selective inhibitor of the PI3K/mTOR pathway that controls nutrient metabolism, growth and proliferation. It has shown antifibrotic properties in vitro. While clinical trials for idiopathic pulmonary fibrosis have been initiated, an in-depth preclinical evaluation is lacking. We evaluated omipalisib's effects on fibrogenesis using the ex vivo model of murine and human precision-cut tissue slices (PCTS). METHODS: Murine and human liver and jejunum PCTS were incubated with omipalisib up to 10 µM for 48 h. PI3K pathway activation was assessed through protein kinase B (Akt) phosphorylation and antifibrotic efficacy was determined via a spectrum of fibrosis markers at the transcriptional and translational level. RESULTS: During incubation of PCTS the PI3K pathway was activated and incubation with omipalisib prevented Akt phosphorylation (IC50 = 20 and 1.5 nM for mouse and human, respectively). Viability of mouse and human liver PCTS was compromised only at the high concentration of 10 and 1-5 µM, respectively. However, viability of jejunum PCTS decreased with 0.1 (mouse) and 0.01 µM (human). Spontaneously increased fibrosis related genes and proteins were significantly and similarly suppressed in mouse and in human liver PCTS. CONCLUSIONS: Omipalisib has antifibrotic properties in ex vivo mouse and human liver PCTS, but higher concentrations showed toxicity in jejunum PCTS. While the PI3K/mTOR pathway appears to be a promising target for the treatment of liver fibrosis, PCTS revealed likely side effects in the intestine at higher doses.


Asunto(s)
Cirrosis Hepática/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/fisiología , Adenosina Trifosfato/análisis , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/fisiología , Piridazinas , Quinolinas/farmacología , Sulfonamidas/farmacología , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
8.
Chem Res Toxicol ; 29(3): 342-51, 2016 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-26881866

RESUMEN

Human toxicity screening is an important stage in the development of safe drug candidates. Hepatotoxicity is one of the major reasons for the withdrawal of drugs from the market because the liver is the major organ involved in drug metabolism, and it can generate toxic metabolites. There is a need to screen molecules for drug-induced hepatotoxicity in humans at an earlier stage. Transcriptomics is a technique widely used to screen molecules for toxicity and to unravel toxicity mechanisms. To date, the majority of such studies were performed using animals or animal cells, with concomitant difficulty in interpretation due to species differences, or in human hepatoma cell lines or cultured hepatocytes, suffering from the lack of physiological expression of enzymes and transporters and lack of nonparenchymal cells. The aim of this study was to classify known hepatotoxicants on their phenotype of toxicity in humans using gene expression profiles ex vivo in human precision-cut liver slices (PCLS). Hepatotoxicants known to induce either necrosis (n = 5) or cholestasis (n = 5) were used at concentrations inducing low (<30%) and medium (30-50%) cytotoxicity, based on ATP content. Random forest and support vector machine algorithms were used to classify hepatotoxicants using a leave-one-compound-out cross-validation method. Optimized biomarker sets were compared to derive a consensus list of markers. Classification correctly predicted the toxicity phenotype with an accuracy of 70-80%. The classification is slightly better for the low than for the medium cytotoxicity. The consensus list of markers includes endoplasmic reticulum stress genes, such as C2ORF30, DNAJB9, DNAJC12, SRP72, TMED7, and UBA5, and a sodium/bile acid cotransporter (SLC10A7). This study shows that human PCLS are a useful model to predict the phenotype of drug-induced hepatotoxicity. Additional compounds should be included to confirm the consensus list of markers, which could then be used to develop a biomarker PCR-array for hepatotoxicity screening.


Asunto(s)
Colestasis/inducido químicamente , Perfilación de la Expresión Génica , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Necrosis/inducido químicamente , Toxicogenética , Acetaminofén/toxicidad , Anciano , Benzofuranos/toxicidad , Ácidos y Sales Biliares/toxicidad , Cloranfenicol/toxicidad , Clorpromazina/toxicidad , Colestasis/genética , Colchicina/toxicidad , Ciclosporina/toxicidad , Dietilnitrosamina/toxicidad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Etinilestradiol/toxicidad , Femenino , Hepatocitos/metabolismo , Humanos , Masculino , Metiltestosterona/toxicidad , Persona de Mediana Edad , Necrosis/genética , Fenotipo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...