Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443222

RESUMEN

Effective therapies for alcohol-associated liver disease (ALD) are limited; therefore, the discovery of new therapeutic agents is greatly warranted. Toll-like receptor 7 (TLR7) is a pattern recognition receptor for single-stranded RNA, and its activation prevents liver fibrosis. We examined liver and intestinal damage in Tlr7-/- mice to determine the role of TLR7 in ALD pathogenesis. In an alcoholic hepatitis (AH) mouse model, hepatic steatosis, injury, and inflammation were induced by chronic binge ethanol feeding in mice, and Tlr7 deficiency exacerbated these effects. Because these results demonstrated that endogenous TLR7 signaling activation is protective in the AH mouse model, we hypothesized that TLR7 activation may be an effective therapeutic strategy for ALD. Therefore, we investigated the therapeutic effect of TLR7 agonistic agent, 1Z1, in the AH mouse model. Oral administration of 1Z1 was well tolerated and prevented intestinal barrier disruption and bacterial translocation, which thus suppressed ethanol-induced hepatic injury, steatosis, and inflammation. Furthermore, 1Z1 treatment up-regulated the expression of antimicrobial peptides, Reg3b and Reg3g, in the intestinal epithelium, which modulated the microbiome by decreasing and increasing the amount of Bacteroides and Lactobacillus, respectively. Additionally, 1Z1 up-regulated intestinal interleukin (IL)-22 expression. IL-22 deficiency abolished the protective effects of 1Z1 in ethanol-induced liver and intestinal damage, suggesting intestinal IL-22 as a crucial mediator for 1Z1-mediated protection in the AH mouse model. Collectively, our results indicate that TLR7 signaling exerts protective effects in the AH mouse model and that a TLR7 ligand, 1Z1, holds therapeutic potential for the treatment of AH.


Asunto(s)
Etanol/toxicidad , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Hepatopatías Alcohólicas/tratamiento farmacológico , Glicoproteínas de Membrana/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 7/metabolismo , Administración Oral , Animales , Bacteroides/efectos de los fármacos , Modelos Animales de Enfermedad , Hígado Graso/complicaciones , Hígado Graso/genética , Hígado Graso/metabolismo , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/complicaciones , Inflamación/genética , Inflamación/metabolismo , Mucosa Intestinal/efectos de los fármacos , Lactobacillus/efectos de los fármacos , Ligandos , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/fisiopatología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/metabolismo , Polietilenglicoles/química , Polietilenglicoles/farmacología , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Transducción de Señal/genética , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/genética , Interleucina-22
2.
Int J Mol Sci ; 25(11)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38891813

RESUMEN

We investigated the pharmacokinetic pathway of berberine and its metabolites in vitro, in Caco-2 cells, and in human participants following the administration of dihydroberberine (DHB) and micellar berberine (LipoMicel®, LMB) formulations. A pilot trial involving nine healthy volunteers was conducted over a 24 h period; blood samples were collected and subjected to Ultra High-Performance Liquid Chromatography-High Resolution Mass Spectrometry (UHPLC-HRMS) analyses to quantify the concentrations of berberine and its metabolites. Pharmacokinetic correlations indicated that berberrubine and thalifendine follow distinct metabolic pathways. Additionally, jatrorrhizine sulfate appeared to undergo metabolism differently compared to the other sulfated metabolites. Moreover, berberrubine glucuronide likely has a unique metabolic pathway distinct from other glucuronides. The human trial revealed significantly higher blood concentrations of berberine metabolites in participants of the DHB treatment group compared to the LMB treatment group-except for berberrubine glucuronide, which was only detected in the LMB treatment group. Similarly, results from in vitro investigations showed significant differences in berberine metabolite profiles between DHB and LMB. Dihydroberberine, dihydroxy-berberrubine/thalifendine and jatrorrhizine sulfate were detected in LMB-treated cells, but not in DHB-treated cells; thalifendine and jatrorrhizine-glucuronide were detected in DHB-treated cells only. While DHB treatment provided higher blood concentrations of berberine and most berberine metabolites, both in vitro (Caco-2 cells) and in vivo human studies showed that treatment with LMB resulted in a higher proportion of unmetabolized berberine compared to DHB. These findings suggest potential clinical implications that merit further investigation in future large-scale trials.


Asunto(s)
Berberina , Micelas , Humanos , Berberina/análogos & derivados , Berberina/farmacocinética , Berberina/sangre , Berberina/metabolismo , Células CACO-2 , Proyectos Piloto , Masculino , Adulto , Femenino , Cromatografía Líquida de Alta Presión
3.
Molecules ; 28(4)2023 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-36838711

RESUMEN

Psoriasis, a chronic inflammation-mediated skin disease, affects 2-3% of the global population. It is characterized by keratinocyte hyperproliferation and immune cell infiltration. The JAK/STAT3 and JAK/STAT1 signaling pathways play an important role in the development of psoriasis when triggered by IL-6 and IFN-γ, which are produced by dendritic cells and T-lymphocytes. Thus, blocking JAK/STAT signaling may be a potential strategy for treating psoriasis. Therefore, we examined the effects of CMX, an extract of Centipeda minima enriched in Brevilin A, Arnicolide D, Arnicolide C, and Microhelenin C, on macrophages and keratinocytes. We established an in vitro model of psoriasis, based on an inflammation-associated keratinocyte proliferation model, and used macrophages and keratinocytes treated with LPS, IL-6, or IFN-γ to evaluate the effect of CMX. We found that CMX reduced pro-inflammatory cytokine production, by inhibiting lipopolysaccharide (LPS)-induced JAK1/2 and STAT1/3 phosphorylation in macrophages. Moreover, CMX-downregulated chemokine expression and cell proliferation compared with components in HaCaT cells, induced by rh-IL-6 and rh-IFN-γ, respectively. Consistently, we demonstrated that the reduction in chemokine expression and hyperproliferation was mediated by the regulation of IFN-γ-activated JAK/STAT1 and IL-6-activated JAK/STAT3 signaling. In conclusion, CMX inhibited JAK/STAT-mediated inflammatory responses and cell proliferation in macrophages and keratinocytes. Consequently, CMX may have potential uses as a therapeutic agent for treating psoriasis.


Asunto(s)
Interleucina-6 , Psoriasis , Humanos , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Queratinocitos , Inflamación/tratamiento farmacológico , Psoriasis/tratamiento farmacológico , Proliferación Celular , Quimiocinas/metabolismo , Macrófagos/metabolismo , Factor de Transcripción STAT1/metabolismo
4.
Molecules ; 28(22)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-38005205

RESUMEN

Zaluzanin C (ZC), a sesquiterpene lactone isolated from Laurus nobilis L., has been reported to have anti-inflammatory and antioxidant effects. However, the mechanistic role of ZC in its protective effects in Kupffer cells and hepatocytes has not been elucidated. The purpose of this study was to elucidate the efficacy and mechanism of action of ZC in Kupffer cells and hepatocytes. ZC inhibited LPS-induced mitochondrial ROS (mtROS) production and subsequent mtROS-mediated NF-κB activity in Kupffer cells (KCs). ZC reduced mRNA levels of pro-inflammatory cytokines (Il1b and Tnfa) and chemokines (Ccl2, Ccl3, Ccl4, Cxcl2 and Cxcl9). Tumor necrosis factor (TNF)-α-induced hepatocyte mtROS production was inhibited by ZC. ZC was effective in alleviating mtROS-mediated mitochondrial dysfunction. ZC enhanced mitophagy and increased mRNA levels of fatty acid oxidation genes (Pparα, Cpt1, Acadm and Hadha) and mitochondrial biosynthetic factors (Pgc1α, Tfam, Nrf1 and Nrf2) in hepatocytes. ZC has proven its anti-lipid effect by improving lipid accumulation in hepatocytes by enhancing mitochondrial function to facilitate lipid metabolism. Therefore, our study suggests that ZC may be an effective compound for hepatoprotection by suppressing inflammation and lipid accumulation through regulating mtROS.


Asunto(s)
Hepatocitos , Macrófagos del Hígado , Humanos , Macrófagos del Hígado/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Mitocondrias/metabolismo , ARN Mensajero/metabolismo , Lípidos/farmacología , Hígado , Metabolismo de los Lípidos
5.
Am J Pathol ; 191(9): 1550-1563, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34126083

RESUMEN

Despite the increasing clinical importance of nonalcoholic fatty liver disease (NAFLD), little is known about its underlying pathogenesis or specific treatment. The senescence marker protein 30 (SMP30), which regulates the biosynthesis of vitamin C (VC) in many mammals, except primates and humans, was recently recognized as a gluconolactonase. However, the precise relation between VC and lipid metabolism in NAFLD is not completely understood. Therefore, this study aimed to clearly reveal the role of VC in NAFLD progression. SMP30 knockout (KO) mice were used as a VC-deficient mouse model. To investigate the precise role of VC on lipid metabolism, 13- to 15-week-old SMP30 KO mice and wild-type mice fed a 60% high-fat diet were exposed to tap water or VC-containing water (1.5 g/L) ad libitum for 11 weeks. Primary mouse hepatocytes isolated from the SMP30 KO and wild-type mice were used to demonstrate the relation between VC and lipid metabolism in hepatocytes. Long-term VC deficiency significantly suppressed the progression of simple steatosis. The high-fat diet-fed VC-deficient SMP30 KO mice exhibited impaired sterol regulatory element-binding protein-1c activation because of excessive cholesterol accumulation in hepatocytes. Long-term VC deficiency inhibits de novo lipogenesis through impaired sterol regulatory element-binding protein-1c activation.


Asunto(s)
Deficiencia de Ácido Ascórbico/metabolismo , Hepatocitos/metabolismo , Lipogénesis/fisiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Progresión de la Enfermedad , Metabolismo de los Lípidos/fisiología , Masculino , Ratones , Ratones Noqueados
6.
Int J Mol Sci ; 23(23)2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36499214

RESUMEN

Mitochondria play a central role in the pathophysiology of inflammatory bowel disease (IBD) and colorectal cancer (CRC). The maintenance of mitochondrial function is necessary for a stable immune system. Mitochondrial dysfunction in the gastrointestinal system leads to the excessive activation of multiple inflammatory signaling pathways, leading to IBD and increased severity of CRC. In this review, we focus on the mitochondria and inflammatory signaling pathways and its related gastrointestinal diseases.


Asunto(s)
Neoplasias Colorrectales , Enfermedades Inflamatorias del Intestino , Humanos , Mitocondrias/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Transducción de Señal , Neoplasias Colorrectales/metabolismo
7.
J Biol Chem ; 294(33): 12359-12369, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31235522

RESUMEN

Excessive alcohol consumption induces intestinal dysbiosis of the gut microbiome and reduces gut epithelial integrity. This often leads to portal circulation-mediated translocation of gut-derived microbial products, such as lipopolysaccharide (LPS), to the liver, where these products engage Toll-like receptor 4 (TLR4) and initiate hepatic inflammation, which promotes alcoholic liver disease (ALD). Although the key self-destructive process of autophagy has been well-studied in hepatocytes, its role in macrophages during ALD pathogenesis remains elusive. Using WT and myeloid cell-specific autophagy-related 7 (Atg7) knockout (Atg7ΔMye) mice, we found that chronic ethanol feeding for 6 weeks plus LPS injection enhances serum alanine aminotransferase and IL-1ß levels and augments hepatic C-C motif chemokine ligand 5 (CCL5) and C-X-C motif chemokine ligand 10 (CXCL10) expression in WT mice, a phenotype that was further exacerbated in Atg7ΔMye mice. Atg7ΔMye macrophages exhibited defective mitochondrial respiration and displayed elevated mitochondrial reactive oxygen species production and inflammasome activation relative to WT cells. Interestingly, compared with WT cells, Atg7ΔMye macrophages also had a drastically increased abundance and nuclear translocation of interferon regulatory factor 1 (IRF1) after LPS stimulation. Mechanistically, LPS induced co-localization of IRF1 with the autophagy adaptor p62 and the autophagosome, resulting in subsequent IRF1 degradation. However, upon p62 silencing or Atg7 deletion, IRF1 started to accumulate in autophagy-deficient macrophages and translocated into the nucleus, where it induced CCL5 and CXCL10 expression. In conclusion, macrophage autophagy protects against ALD by promoting IRF1 degradation and removal of damaged mitochondria, limiting macrophage activation and inflammation.


Asunto(s)
Muerte Celular Autofágica , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Etanol/efectos adversos , Factor 1 Regulador del Interferón/metabolismo , Macrófagos/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteolisis , Animales , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Etanol/farmacología , Factor 1 Regulador del Interferón/genética , Lipopolisacáridos/toxicidad , Hígado/metabolismo , Hígado/patología , Macrófagos/patología , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/patología
8.
Molecules ; 25(6)2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-32183436

RESUMEN

Aucklandia lappa Decne., known as "Mok-hyang" in Korea, has been used for the alleviation of abdominal pain, vomiting, diarrhea, and stress gastric ulcers in traditional oriental medicine. We investigated the anti-inflammatory and antioxidative effects of the ethanol extract of Aucklandia lappa Decne. (ALDE) in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. ALDE significantly inhibited the LPS-induced nitric oxide (NO) production and reduced inducible nitric oxide synthase (iNOS) expression in RAW 264.7 cells. The production of other proinflammatory mediators, including COX-2, interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α, was reduced by ALDE in LPS-stimulated RAW 264.7 cells. The mechanism underlying the anti-inflammatory effects of ALDE was elucidated to be the suppression of LPS-induced nuclear translocation of p65, followed by the degradation of IκB and the inhibition of the phosphorylation of mitogen-activated protein kinases (MAPK). In addition, ALDE showed enhanced radical scavenging activity. The antioxidant effect of ALDE was caused by the enhanced expression of heme oxygenase (HO-1) via stabilization of the expression of the nuclear transcription factor E2-related factor 2 (Nrf2) pathway. Collectively, these results indicated that ALDE not only exerts anti-inflammatory effects via the suppression of the NF-κB and MAPK pathways but also has an antioxidative effect through the activation of the Nrf2/HO-1 pathway.


Asunto(s)
Antiinflamatorios/farmacología , Inflamación/tratamiento farmacológico , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Extractos Vegetales/farmacología , Saussurea/química , Animales , Antioxidantes/metabolismo , Línea Celular , Ciclooxigenasa 2/metabolismo , Hemo-Oxigenasa 1/metabolismo , Proteínas I-kappa B/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
9.
Am J Pathol ; 188(4): 967-980, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29355515

RESUMEN

Excessive alcohol consumption leads to chronic liver diseases. Macrophage-inducible C-type lectin (Mincle) is a C-type lectin receptor that recognizes spliceosome-associated protein 130 (SAP130) known as an endogenous ligand released from dying cells. The aim was to examine the role of Mincle-SAP130 in the pathogenesis of alcoholic liver disease. Alcohol-induced liver injury was induced in wild-type (WT) and Mincle knockout (KO) mice by using a chronic-binge ethanol-feeding model. Mincle KO mice showed significant lower hepatic steatosis, inflammation with neutrophil infiltration, and fibrosis compared with WT mice after alcohol feeding. In contrast, Mincle activation exacerbated alcohol-induced liver injury. Kupffer cells (KCs) are major sources of Mincle. IL-1ß expression was significantly down-regulated in Mincle KO mice compared with that in WT mice after alcohol consumption. Interestingly, expression and production of IL-1ß were significantly decreased in SAP130-treated KCs isolated from leucine-rich-containing family pyrin domain containing-3-deficient mice compared with those in WT KCs. Such results were also observed in cells treated with SAP130 plus Syk inhibitor. Furthermore, infiltration of invariant natural killer T cells was decreased in livers of Mincle KO mice. Finally, inhibition of Syk signaling ameliorated alcohol-induced liver injury. Collectively, these results demonstrated that interaction between Mincle and SAP130 may promote the progression of alcoholic liver disease by IL-1ß production in KCs and consequently increase inflammatory immune cell infiltration.


Asunto(s)
Progresión de la Enfermedad , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Hígado/lesiones , Hígado/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas/metabolismo , Animales , Etanol , Lectinas Tipo C/deficiencia , Lectinas Tipo C/metabolismo , Hígado/patología , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/metabolismo , Infiltración Neutrófila , Transducción de Señal , Quinasa Syk/metabolismo
10.
Am J Pathol ; 188(11): 2574-2588, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30125542

RESUMEN

Toll-like receptor 7 (TLR7) signaling regulates the production of type 1 interferons (IFNs) and proinflammatory cytokines, such as tumor necrosis factor (TNF)-α, implicated in the control of regulatory T (Treg) cell activity. However, the mechanistic interplay between TLR7 signaling and Treg cells in nonalcoholic steatohepatitis (NASH) has not been elucidated. Our aim was to clarify the role of TLR7 signaling in the pathogenesis of NASH. Steatohepatitis was induced in wild-type (WT), TLR7-deficient, IFN-α/ß receptor 1-deficient, and Treg cell-depleted mice. TLR7-deficient and IFN-α/ß receptor 1-deficient mice were more protective to steatohepatitis than WT mice. Of interest, both TNF-α and type 1 IFN promoted apoptosis of Treg cells involved in the prevention of NASH. Indeed, Treg cell-depleted mice had aggravated steatohepatitis compared with WT mice. Finally, treatment with immunoregulatory sequence 661, an antagonist of TLR7, efficiently ameliorated NASH in vivo. These results demonstrate that TLR7 signaling can induce TNF-α production in Kupffer cells and type I IFN production in dendritic cells. These cytokines subsequently induce hepatocyte death and inhibit Treg cells activities, leading to the progression of NASH. Thus, manipulating the TLR7-Treg cell axis might be used as a novel therapeutic strategy to treat NASH.


Asunto(s)
Células Dendríticas/inmunología , Macrófagos del Hígado/inmunología , Glicoproteínas de Membrana/fisiología , Enfermedad del Hígado Graso no Alcohólico/etiología , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 7/fisiología , Animales , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Interferón Tipo I/metabolismo , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Int J Cancer ; 142(1): 81-91, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28875549

RESUMEN

Hepatocyte death is associated with liver inflammation, fibrosis and hepatocellular carcinoma (HCC). Damaged cells trigger inflammation through activation of Toll-like receptors (TLRs). Although the role of TLR4 in HCC development has been reported, the role of TLR9 in the development of HCC remains elusive. To investigate the role of TLR4 and TLR9 signaling in liver inflammation-fibrosis-cancer axis, we took advantage of mice with hepatic deletion of transforming growth factor-ß-activated kinase 1 (Tak1ΔHep) that develop spontaneous liver injury, inflammation, fibrosis, and HCC, recapitulating the pathology of human HCC. We generated double knockout mice lacking genes of our interest with hepatic Tak1. Tak1ΔHep mice and Tlr4-deficient Tak1ΔHep mice had similar serum ALT levels, but Tlr4-deficient Tak1ΔHep mice exhibited significantly reduced macrophage infiltration, myofibroblast activation and tumor formation. Ablation of TLR9 reduced spontaneous liver injury, inflammation, fibrosis, and cancer development in Tak1ΔHep mice. In addition, the common adaptor, myeloid differentiation factor 88 (MyD88)-deficient Tak1ΔHep mice also attenuated liver injury, macrophage recruitment, collagen deposition, and tumor growth compared with control Tak1ΔHep mice. Genetic ablation of TNF receptor type I (TNFR) in Tak1ΔHep mice remarkably reduced liver inflammation-fibrosis-cancer axis. Surprisingly, disruption of interleukin-1 receptor (IL-1R) had no effect on liver injury and tumor formation, although Il1r-deficient Tak1ΔHep showed attenuated macrophage infiltration and collagen deposition. In conclusion, TLR4- and TLR9-MyD88 are driving forces of progression to HCC accompanied by liver inflammation and fibrosis in Tak1ΔHep mice. Importantly, TLR4 and TLR9 downstream TNFR, but not IL-1R signaling is crucial for the development of HCC in Tak1ΔHep mice.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Carcinoma Hepatocelular/patología , Hepatocitos/patología , Cirrosis Hepática/patología , Neoplasias Hepáticas/patología , Quinasas Quinasa Quinasa PAM/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
12.
Adv Exp Med Biol ; 1061: 45-53, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29956205

RESUMEN

Chemokines are chemo-attractants for leukocyte trafficking, growth, and activation in injured and inflammatory tissues. The chemokine system is comprised of 50 chemokine ligands and 20 cognate chemokine receptors. In the context of liver diseases, leukocytes, hepatocytes, hepatic stellate cells, endothelial cells, and vascular smooth muscle cells are capable of producing chemokines. Chemokine receptors are typically expressed in various leukocyte subsets. Given that inflammation is a critical factor for the transition from simple steatosis to non-alcoholic steatohepatitis (NASH), and fibrosis, the chemokine system may play a prominent role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Indeed, accumulating evidence shows elevated expression of chemokines and their receptors in the livers of obese patients with advanced steatosis and NASH. This chapter will discuss the underlying molecular mechanisms and the therapeutic potential of the chemokine systems in the pathogenesis of NAFLD. Among chemokines, we will highlight CCL2, CCL5, CXCL8-10, CX3CL1, and CXCL16 as pivotal mediators in the development of steatosis, NASH, and fibrosis.


Asunto(s)
Quimiocinas/metabolismo , Cirrosis Hepática/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Receptores de Quimiocina/metabolismo , Animales , Humanos , Cirrosis Hepática/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/patología
13.
Molecules ; 22(9)2017 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-28846649

RESUMEN

Type 2 diabetes is characterized by insulin resistance, which leads to increased blood glucose levels. Adipocytes are involved in the development of insulin resistance, resulting from the dysfunction of the insulin signaling pathway. In this study, we investigated whether meso-dihydroguaiaretic acid (MDGA) may modulate glucose uptake in adipocytes, and examined its mechanism of action. MDGA enhanced adipogenesis through up-regulation of peroxisome proliferator-activated receptor γ and CCAAT/enhancer-binding protein α in 3T3-L1 adipocytes partially differentiated with sub-optimal concentrations of insulin. MDGA also increased glucose uptake by stimulating expression and translocation of glucose transporter 4 (GLUT4) in adipocytes. These results suggest that MDGA may increase GLUT4 expression and its translocation by promoting insulin sensitivity, leading to enhanced glucose uptake.


Asunto(s)
Adipocitos/citología , Transportador de Glucosa de Tipo 4/metabolismo , Glucosa/metabolismo , Guayacol/análogos & derivados , Lignanos/farmacología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis , Animales , Guayacol/farmacología , Ratones , PPAR gamma/metabolismo , Regulación hacia Arriba
14.
Am J Physiol Gastrointest Liver Physiol ; 309(1): G30-41, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25930080

RESUMEN

Although previous studies reported the involvement of the TLR4-TRIF pathway in alcohol-induced liver injury, the role of TLR2 and TLR9 signaling in alcohol-mediated neutrophil infiltration and liver injury has not been elucidated. Since alcohol binge drinking is recognized to induce more severe form of alcohol liver disease, we used a chronic-binge ethanol-feeding model as a mouse model for early stage of alcoholic hepatitis. Whereas a chronic-binge ethanol feeding induced alcohol-mediated liver injury in wild-type mice, TLR2- and TLR9-deficient mice showed reduced liver injury. Induction of neutrophil-recruiting chemokines, including Cxcl1, Cxcl2, and Cxcl5, and hepatic neutrophil infiltration were increased in wild-type mice, but not in TLR2- and TLR9-deficient mice. In vivo depletion of Kupffer cells (KCs) by liposomal clodronate reduced liver injury and the expression of Il1b, but not Cxcl1, Cxcl2, and Cxcl5, suggesting that KCs are partly associated with liver injury, but not neutrophil recruitment, in a chronic-binge ethanol-feeding model. Notably, hepatocytes and hepatic stellate cells (HSCs) produce high amounts of CXCL1 in ethanol-treated mice. The treatment with TLR2 and TLR9 ligands synergistically upregulated CXCL1 expression in hepatocytes. Moreover, the inhibitors for CXCR2, a receptor for CXCL1, and MyD88 suppressed neutrophil infiltration and liver injury induced by chronic-binge ethanol treatment. Consistent with the above findings, hepatic CXCL1 expression was highly upregulated in patients with alcoholic hepatitis. In a chronic-binge ethanol-feeding model, the TLR2 and TLR9-dependent MyD88-dependent pathway mediates CXCL1 production in hepatocytes and HSCs; the CXCL1 then promotes neutrophil infiltration into the liver via CXCR2, resulting in the development of alcohol-mediated liver injury.


Asunto(s)
Quimiocina CXCL1/metabolismo , Hepatopatías Alcohólicas/metabolismo , Hígado/inmunología , Infiltración Neutrófila , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Consumo Excesivo de Bebidas Alcohólicas , Células Cultivadas , Quimiocina CXCL1/inmunología , Modelos Animales de Enfermedad , Etanol , Femenino , Células Estrelladas Hepáticas/inmunología , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/inmunología , Hepatocitos/metabolismo , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Hígado/patología , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/inmunología , Hepatopatías Alcohólicas/patología , Hepatopatías Alcohólicas/prevención & control , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Interleucina-8B/inmunología , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Factores de Tiempo , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Regulación hacia Arriba
15.
Hepatology ; 59(2): 483-95, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23996730

RESUMEN

UNLABELLED: Transforming growth factor beta (TGF-ß) signaling activates Smad- and TGF-ß-activated kinase 1 (TAK1)-dependent signaling to regulate cell survival, proliferation, fibrosis, and tumorigenesis. The effects of TGF-ß signaling on metabolic syndrome, including nonalcoholic fatty liver disease, remain elusive. Wild-type (WT) and hepatocyte-specific TGF-ß receptor type II-deficient (Tgfbr2ΔHEP) mice were fed a choline-deficient amino acid (CDAA)-defined diet for 22 weeks to induce NASH. WT mice fed a CDAA diet displayed increased activation of Smad2/3 and had marked lipid accumulation, inflammatory cell infiltration, hepatocyte death, and fibrosis; in comparison, Tgfbr2ΔHEP mice fed a CDAA diet had suppressed liver steatosis, inflammation, and fibrosis. Both palmitate-induced steatotic hepatocytes and hepatocytes isolated from WT mice fed a CDAA diet had increased susceptibility to TGF-ß-mediated death. TGF-ß-mediated death in steatotic hepatocytes was inhibited by silencing Smad2 or blocking reactive oxygen species (ROS) production and was enhanced by inhibiting TAK1 or nuclear factor kappa B. Increased hepatic steatosis in WT mice fed a CDAA diet was associated with the increased expression of lipogenesis genes (Dgat1 and Srebp1c), whereas the decreased steatosis in Tgfbr2ΔHEP mice was accompanied by the increased expression of genes involved in ß-oxidation (Cpt1 and Acox1). In combination with palmitate treatment, TGF-ß signaling promoted lipid accumulation with induction of lipogenesis-related genes and suppression of ß-oxidation-related genes in hepatocytes. Silencing Smad2 decreased TGF-ß-mediated lipid accumulation and corrected altered gene expression related to lipid metabolism in hepatocytes. Finally, we confirmed that livers from patients with nonalcoholic steatohepatitis (NASH) displayed phosphorylation and nuclear translocation of Smad2/3. CONCLUSIONS: TGF-ß signaling in hepatocytes contributes to hepatocyte death and lipid accumulation through Smad signaling and ROS production that promote the development of NASH.


Asunto(s)
Apoptosis/fisiología , Hígado Graso/fisiopatología , Hepatocitos/fisiología , Metabolismo de los Lípidos/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Deficiencia de Colina/metabolismo , Deficiencia de Colina/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hígado Graso/etiología , Hígado Graso/patología , Eliminación de Gen , Hepatocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Proteínas Smad/fisiología
16.
Hepatology ; 60(1): 237-49, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24375615

RESUMEN

UNLABELLED: Toll-like receptor 7 (TLR7) signaling predominantly regulates production of type I interferons (IFNs), which has been suggested in clinical studies to be antifibrotic. However, the mechanistic role of the TLR7-type I IFN axis in liver fibrosis has not been elucidated. In the present study, liver fibrosis was induced in wild-type (WT), TLR7-deficient, and IFN-α/ß receptor-1 (IFNAR1)-deficient mice and TLR7-mediated signaling was assessed in liver cells isolated from these mice. TLR7-deficient and IFNAR1-deficient mice were more susceptible to liver fibrosis than WT mice, indicating that TLR7-type I IFN signaling exerts a protective effect against liver fibrosis. Notably, the hepatic expression of interleukin-1 receptor antagonist (IL-1ra) was suppressed in TLR7- or IFNAR1-deficient mice compared with respective WT mice, and treatment with recombinant IL-1ra reduced liver fibrosis. In vivo activation of TLR7 significantly increased IFNa4 and IL-1ra expression in the liver. Interestingly, each cytokine had a different cellular source, showing that dendritic cells (DCs) are the responsible cell type for production of type I IFN, while Kupffer cells (KCs) mainly produce IL-1ra in response to type I IFN. Furthermore, TLR7 activation by R848 injection suppressed liver fibrosis and production of proinflammatory cytokines, and these effects were dependent on type I IFN signaling. Consistent with in vivo data, IFN-α significantly induced IL-1ra production in primary KCs. CONCLUSION: TLR7 signaling activates DCs to produce type I IFN, which in turn induces antifibrogenic IL-1ra production in KCs. Thus, manipulation of the TLR7-type I IFN-IL-1ra axis may be a new therapeutic strategy for the treatment of liver fibrosis.


Asunto(s)
Colestasis/prevención & control , Interferón Tipo I/metabolismo , Cirrosis Hepática/prevención & control , Glicoproteínas de Membrana/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 7/metabolismo , Animales , Tetracloruro de Carbono/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Colestasis/inmunología , Colestasis/metabolismo , Enfermedad Crónica , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Antagonista del Receptor de Interleucina 1/inmunología , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Cirrosis Hepática/inmunología , Cirrosis Hepática/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología
17.
Dig Dis Sci ; 60(7): 2009-18, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25416630

RESUMEN

BACKGROUND: Accumulating evidence suggests that Foxp3+ regulatory T (Treg) cells act as inhibitory mediators of inflammation; however, the in vivo mechanism underlying this protection remains elusive in liver diseases. AIMS: To clarify the in vivo role of Foxp3+ Treg cells in liver fibrosis, we used the DEREG mouse, which expresses the diphtheria toxin receptor under control of the Foxp3 promoter, allowing for specific deletion of Foxp3+ Treg cells. METHODS: Bile duct ligation-induced liver injury and fibrosis were assessed by histopathology, fibrogenic gene expression, and measurement of cytokine and chemokine levels. RESULTS: Depletion of Foxp3+ Treg cells enhanced Th17 cell response as demonstrated by the increase of IL-17+ cells and related gene expressions including Il17f, Il17ra, and Rorgt in the fibrotic livers of DEREG mice. Of note, infiltration of CD8+ T cells and Cd8 gene expression was significantly increased in the livers of DEREG mice. Consistent with increased IL-17+ and CD8+ T cell responses, DEREG mice generated higher levels of inflammatory cytokines (TNF-α, IL-6, and IL-12p70) and chemokines (MCP-1, MIP-1α, and RANTES). These results were concordant with severity of liver fibrosis and hepatic enzyme levels (ALT and ALP). CONCLUSIONS: The present findings demonstrate that Foxp3+ Treg cells inhibit the profibrogenic inflammatory milieu through suppression of pro-fibrogenic CD8+ and IL-17+ T cells.


Asunto(s)
Colestasis/metabolismo , Factores de Transcripción Forkhead/metabolismo , Cirrosis Hepática/patología , Linfocitos T Reguladores/fisiología , Animales , Conductos Biliares/cirugía , Linfocitos T CD8-positivos , Factores de Transcripción Forkhead/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ligadura , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Mutación , Células Th17
18.
Gastroenterology ; 144(5): 1042-1054.e4, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23391818

RESUMEN

BACKGROUND & AIMS: Transforming growth factor (TGF)-ß-activated kinase 1 (TAK1) is activated in different cytokine signaling pathways. Deletion of Tak1 from hepatocytes results in spontaneous development of hepatocellular carcinoma (HCC), liver inflammation, and fibrosis. TGF-ß activates TAK1 and Smad signaling, which regulate cell death, proliferation, and carcinogenesis. However, it is not clear whether TGF-ß signaling in hepatocytes, via TGF-ß receptor-2 (Tgfbr2), promotes HCC and liver fibrosis. METHODS: We generated mice with hepatocyte-specific deletion of Tak1 (Tak1ΔHep), as well as Tak1/Tgfbr2DHep and Tak1/Smad4ΔHep mice. Tak1flox/flox, Tgfbr2ΔHep, and Smad4ΔHep mice were used as controls, respectively. We assessed development of liver injury, inflammation, fibrosis, and HCC. Primary hepatocytes isolated from these mice were used to assess TGF-ß-mediated signaling. RESULTS: Levels of TGF-ß, TGF-ßR2, and phospho-Smad2/3 were increased in HCCs from Tak1ΔHep mice, which developed liver fibrosis and inflammation by 1 month and HCC by 9 months. However, Tak1/Tgfbr2ΔHep mice did not have this phenotype, and their hepatocytes did not undergo spontaneous cell death or compensatory proliferation. Hepatocytes from Tak1ΔHep mice incubated with TGF-ß did not activate p38, c-Jun N-terminal kinase, or nuclear factor-κB; conversely, TGF-ß-mediated cell death and phosphorylation of Smad2/3 were increased, compared with control hepatocytes. Blocking the Smad pathway inhibited TGF-ß-mediated death of Tak1-/- hepatocytes. Accordingly, disruption of Smad4 reduced the spontaneous liver injury, inflammation, fibrosis, and HCC that develops in Tak1ΔHep mice. Levels of the anti-apoptotic protein Bcl-xL, ß-catenin, connective tissue growth factor, and vascular endothelial growth factor were increased in HCC from Tak1ΔHep mice, but not in HCCs from Tak1/Tgfbr2ΔHep mice. Injection of N-nitrosodiethylamine induced HCC formation in wild-type mice, but less in Tgfbr2ΔHep mice. CONCLUSIONS: TGF-ß promotes development of HCC in Tak1ΔHep mice by inducing hepatocyte apoptosis and compensatory proliferation during early phases of tumorigenesis, and inducing expression of anti-apoptotic, pro-oncogenic, and angiogenic factors during tumor progression.


Asunto(s)
Carcinoma Hepatocelular/genética , ADN de Neoplasias/genética , Eliminación de Gen , Cirrosis Hepática/patología , Neoplasias Hepáticas Experimentales/genética , Quinasas Quinasa Quinasa PAM/genética , Factor de Crecimiento Transformador beta/genética , Animales , Apoptosis , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Análisis Mutacional de ADN , Hepatocitos/metabolismo , Hepatocitos/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Transgénicos , Fenotipo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas
19.
J Zoo Wildl Med ; 45(4): 951-4, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25632690

RESUMEN

An African pygmy hedgehog (Atelerix albiventris) was diagnosed as chylous ascites with biliary cirrhosis. Abdomenocentesis revealed a milky fluid with a 324 mg/dl triglyceride level. On serum biochemical examination, the hedgehog had hypoalbuminemia, hypoglycemia, and high blood urea nitrogen. There was no cytologic or genomic evidence of infection, and a blood culture was negative. Histopathologic examination revealed a liver with proliferative bile ducts that were often surrounded by prominent septa of fibrous connective tissue. In the area of ductular reaction, proliferative cells positive for CD66, an embryogenic antigen of epithelial cells, were revealed. The potential association between chylous ascites and liver cirrhosis is undetermined but could be an aspect of future study. This is the first description of chylous ascites in a hedgehog.


Asunto(s)
Ascitis Quilosa/veterinaria , Erizos , Hepatopatías/veterinaria , Animales , Ascitis Quilosa/diagnóstico , Ascitis Quilosa/patología , Hepatopatías/diagnóstico , Hepatopatías/patología , Masculino
20.
Life Sci ; 342: 122534, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38408637

RESUMEN

AIMS: Sphingolipids are involved in the regulation of insulin signaling, which is linked to the development of insulin resistance, leading to diabetes mellitus. We aimed to study whether modulation of sphingolipid levels by GT-11 may regulate insulin signaling in C2C12 myotubes. MAIN METHODS: We investigated the effects of sphingolipid metabolism on Akt phosphorylation and glucose uptake using C2C12 myotubes. Either GT-11, an inhibitor of dihydroceramide desaturase 1 and S1P lyase, or siRNA targeting Sgpl1, the gene encoding the enzyme, was employed to determine the effect of sphingolipid metabolism modulation on insulin signaling. Western blotting and glucose uptake assays were used to evaluate the effect of treatments on insulin signaling. Sphingolipid metabolites were analyzed by high performance liquid chromatography (HPLC). KEY FINDINGS: Treatment with GT-11 resulted in decreased Akt phosphorylation and reduced glucose uptake. Silencing the Sgpl1 gene, which encodes S1P lyase, mimicked these findings, suggesting the potential for regulating insulin signaling through S1P lyase modulation. GT-11 modulated sphingolipid metabolism, inducing the accumulation of sphingolipids. Using PF-543 and ARN14974 to inhibit sphingosine kinases and acid ceramidase, respectively, we identified a significant interplay between sphingosine, S1P lyase, and insulin signaling. Treatment with either exogenous sphingosine or palmitic acid inhibited Akt phosphorylation, and reduced S1P lyase activity. SIGNIFICANCE: Our findings highlight the importance of close relationship between sphingolipid metabolism and insulin signaling in C2C12 myotubes, pointing to its potential therapeutic relevance for diabetes mellitus.


Asunto(s)
Diabetes Mellitus , Liasas , Humanos , Insulina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esfingosina/metabolismo , Esfingolípidos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Glucosa/metabolismo , Liasas/metabolismo , Liasas/farmacología , Diabetes Mellitus/metabolismo , Lisofosfolípidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA