Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 395
Filtrar
1.
Hum Exp Toxicol ; 41: 9603271221084672, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35303413

RESUMEN

Liver fibrosis is a wound-healing response and the activation of the hepatic stellate cell (HSC) is the core of hepatic fibrosis. MicroRNAs (miRNAs) participate in the development of fibrosis. It is reported that histone deacetylases (HDAC2) tyrosine phosphorylation by cellular-Abelsongene (c-Abl) induces malignant growth of cells. Here, we investigated the effect of miR-122-5p on the proliferation and apoptosis of HSCs. Normal human HSC line LX-2 and LX-2 cells stimulated by transforming growth factor (TGF)-ß1 for 24 h were cultured and assigned into the blank group and the TGF-ß1 group. The miR-122-5p inhibitor and its negative control were transfected into LX-2 cells and miR-122-5p mimic and its negative control were transfected into LX-2 cells stimulated by TGF-ß1. The result showed that miR-122-5p expression was decreased in TGF-ß1-stimulated LX-2 cells. miR-122-5p overexpression reduced the mRNA and protein levels of collagen I and α-smooth muscle actin, inhibited cell proliferation, and accelerated cell apoptosis. miR-122-5p targeted c-Abl. Meanwhile, miR-122-5p overexpression inhibited HSC activation by suppressing the c-Abl/HDAC2 pathway. In summary, miR-122-5p overexpression exerted anti-fibrosis effect and inhibited HSC activation by suppressing the c-Abl/HDAC2 pathway.


Asunto(s)
Apoptosis/fisiología , Proliferación Celular/fisiología , Células Estrelladas Hepáticas/fisiología , Histona Desacetilasa 2/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Histona Desacetilasa 2/genética , Humanos , MicroARNs/genética , Proteínas Proto-Oncogénicas c-abl/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología
2.
Toxicol In Vitro ; 80: 105315, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35051607

RESUMEN

Activation of hepatic stellates (HSCs) is known as the major cause of initiation and progression of liver fibrosis. A wide array of events occurs during HSC activation including induction of hedgehog (Hh) signaling and endoplasmic reticulum (ER) stress. Targeting HSC activation may provide promising insights into liver fibrosis treatment. In this regard, establishing in vitro models which can mimic the molecular pathways of interest is very important. We aimed to activate HSC in which Hh signaling and ER stress are stimulated simultaneously. We used 5 ng/ml TGFß to activate LX-2 cells, HSC cell line. Gene expression analysis using qRT-PCR, immunostaining and immunoblotting were performed to show HSC activation associated markers. Furthermore, the migration capacity of the TGFß treated cells is evaluated. The results demonstrated that major fibrogenic markers including collagen1a, lysyl oxidase, and tissue inhibitor of matrix metalloproteinase 1 genes are up-regulated significantly. In addition, our immunofluorescence and immunoblotting results showed that protein levels of GLI-2 and XBP1, were enhanced. Moreover, we found that TGFß treatment reduced the migration of LX-2 cells. Our results are compatible with high throughput data analysis with respect to differentially expressed genes of activated HSC compared to the quiescent ones. Moreover, our findings suggest that quercetin can reduce fibrogenic markers of activated HSCs as well as osteopontin expression, a target gene of hedgehog signaling.


Asunto(s)
Estrés del Retículo Endoplásmico , Proteínas Hedgehog/metabolismo , Células Estrelladas Hepáticas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular , Movimiento Celular , Colágeno/metabolismo , Expresión Génica , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/fisiología , Humanos , Quercetina/farmacología , Transducción de Señal
3.
J Nutr Biochem ; 100: 108888, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34695558

RESUMEN

This study aimed to investigate the therapeutic effects of nobiletin (NOB) on nonalcoholic steatohepatitis (NASH) and liver fibrosis in mice and to elucidate its underlying molecular mechanisms. BALB/c mice were fed a normal chow diet or a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) for 8 wks and treated with NOB (50 mg/kg) or vehicle by daily intraperitoneally injection for the last 4 wks. In vitro, we used palmitate (PA) stimulated AML12 cells as the model of hepatocyte lipotoxicity to dissect the effect and molecular mechanisms of NOB' action. Our results exhibited that NOB dramatically reduced hepatic steatosis, lipid accumulation and hepatocyte apoptosis, and inhibited the infiltration of F4/80+ macrophages into the NASH livers. Furthermore, NOB limited liver fibrosis and hepatic stellate cells activation in NASH mice. In parallel, NOB alleviated hepatocytes apoptosis and lipid accumulation in PA-treated AML12 cells. Most importantly, these histological ameliorations in NASH and fibrosis in NOB-treated NASH mice were associated with improvement hepatic oxidative stress, lipid peroxidation product, mitochondrial respiratory chain complexes I and restored ATP production. Similarly, NOB attenuated PA-induced reactive oxygen species (ROS) generation and mitochondrial disfunction in cultured AML12 cells. Additionally, NOB diminished the expression of mitochondrial Ca2+ uniporter (MCU) both in NASH livers and in PA-treated AML12. Taken together, our results indicate that NOB mitigated NASH development and fibrosis through modulating hepatic oxidative stress and attenuating mitochondrial dysfunction. Therefore, NOB might be a novel and promising agent for treatment of NASH and liver fibrosis.


Asunto(s)
Apoptosis , Flavonas/farmacología , Hepatocitos/fisiología , Mitocondrias Hepáticas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Línea Celular , Dieta Alta en Grasa , Flavonas/uso terapéutico , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/fisiología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Cirrosis Hepática/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos BALB C , Mitocondrias Hepáticas/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/patología
4.
Mol Med Rep ; 25(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34738631

RESUMEN

It is important to determine the mechanism of liver fibrosis for targeted therapy and the development of targeted therapies for liver fibrosis may offer promise for patients with liver disease. Long non­coding RNAs (lncRNAs) serve a role in hepatic fibrosis. The lncRNA maternally expressed gene 3 (MEG3) has been confirmed to inhibit liver fibrosis. The present study investigated the role of the MEG3 in healthy patients and patients with liver fibrosis. The expression levels of MEG3 and microRNA (miR)­145 in the serum of healthy volunteers and patients with liver fibrosis and in LX­2 cells were detected using reverse transcription­quantitative PCR. A dual­luciferase reporter assay was used to determine the targeting relationship between MEG3 and miR­145, and the targeting relationship between miR­145 and peroxisome proliferator­activated receptor Î³ (PPARγ). The protein expression levels of PPARγ, α­smooth muscle actin (α­SMA) and collagen I (COL1A1) were detected using western blotting. The expression levels of α­SMA and COL1A1 were also determined using immunofluorescence. Finally, a Cell Counting Kit­8 assay was performed to assess the proliferative ability of LX­2 cells. A significantly reduced MEG3 expression level was demonstrated in serum from patients with liver fibrosis compared with serum from healthy controls. TGF­ß1 induced a significantly decreased MEG3 expression level in LX­2 human hepatic stellate cells in vitro. The TGF­ß1­induced increases in cell proliferation and α­SMA and COL1A1 protein expression levels were reversed following MEG3 overexpression. The results also demonstrated that MEG3 sponged miR­145 and competed endogenously with miR­145 to regulate PPARγ. In summary, the present study identified MEG3 as an anti­fibrotic lncRNA and provided new information regarding the role of MEG3 in liver fibrosis. MEG3 may therefore be a potential target in the treatment of liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , MicroARNs/genética , PPAR gamma/metabolismo , ARN Largo no Codificante/metabolismo , Actinas/metabolismo , Adulto , Proliferación Celular/genética , China , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I/análisis , Femenino , Fibrosis/metabolismo , Células Estrelladas Hepáticas/fisiología , Humanos , Cirrosis Hepática/metabolismo , Masculino , MicroARNs/sangre , MicroARNs/metabolismo , Persona de Mediana Edad , PPAR gamma/genética , ARN Largo no Codificante/sangre , ARN Largo no Codificante/genética , Factor de Crecimiento Transformador beta1/metabolismo
5.
J Gastrointest Cancer ; 52(4): 1294-1308, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34927218

RESUMEN

INTRODUCTION: Three-dimensional (3D) cell culture studies are becoming extremely common because of their capability to mimic tumor architecture, such as cell-cell and cell-ECM interactions, more efficiently than 2D monolayer systems. These interactions have important roles in defining the tumor cell behaviors, such as proliferation, differentiation, and most importantly, tumor drug response. OBJECTIVE: This review aims to provide an overview of the methods for 3D tumor spheroid formation to model human tumors, specifically concentrated on studies using hepatocellular carcinoma (HCC) cells. METHOD: We obtained information from previously published articles. In this review, there is discussion of the scaffold and non-scaffold-based approaches, including hanging drop, bioreactors and 3D bioprinting. RESULTS AND CONCLUSION: The mimicking of the tumor microenvironment (TME) as tumor spheroids could provide a valuable platform for studying tumor biology. Multicellular tumor spheroids are self-assembled cultures of mixed cells (tumor and stromal cells) organized in a 3D arrangement. These spheroids closely mimic the main features of human solid tumors, such as structural organization, central hypoxia, and overall oxygen and nutrient gradients. Hepatocellular carcinoma (HCC) is the most common liver malignancy, and most difficult to overcome because of its drug resistance and tumor heterogeneity. In order to mimic this highly heterogeneous environment, 3D cell culture systems are needed.


Asunto(s)
Carcinoma Hepatocelular/fisiopatología , Técnicas de Cultivo Tridimensional de Células/métodos , Células Estrelladas Hepáticas/patología , Células Estrelladas Hepáticas/fisiología , Neoplasias Hepáticas/fisiopatología , Microambiente Tumoral , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Humanos , Neoplasias Hepáticas/patología
6.
Mediators Inflamm ; 2021: 3399357, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34690551

RESUMEN

Liver fibrosis, which results from chronic liver injury due to factors such as chronic alcohol consumption, hepatitis virus infections, and immune attacks, is marked by excessive deposition of extracellular matrix (ECM). Resveratrol (Res), a polyphenol phytoalexin, has been demonstrated to show anti-inflammatory, antioxidative, antiproliferative, and chemopreventive activities. In recent years, Res has been found to inhibit liver fibrosis. Enhanced Hippo pathway activation has also been reported to inhibit tumor progression and liver fibrosis. In the present study, the role of the Hippo pathway in mediating the effects of Res on hepatic stellate cells (HSCs) was examined. We found that Res significantly suppresses HSC proliferation, reducing the cell index. Res induced HSC inactivation, reducing collagen deposition and α-smooth muscle actin (α-SMA) expression. In addition, Res contributed to HSC apoptosis, upregulating Bax and downregulating Bcl-2 expression. Notably, the Hippo pathway was involved in the Res-mediated suppression of HSC activation. Res enhanced the activation of the Hippo pathway and reduced yes-associated protein (YAP) and transcriptional coactivator with the PDZ-binding motif (TAZ) expression. Interestingly, the YAP overexpression inhibited Res-induced HSC inactivation and apoptosis. In conclusion, these results demonstrate that Res inhibits HSC activation, at least in part, via the Hippo pathway. The present study indicates a new antifibrotic mechanism of Res and provides novel insights into Hippo-mediated HSC apoptosis and HSC activation in liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/efectos de los fármacos , Vía de Señalización Hippo/efectos de los fármacos , Resveratrol/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Células Estrelladas Hepáticas/fisiología , Vía de Señalización Hippo/fisiología , Humanos , Cirrosis Hepática/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Resveratrol/uso terapéutico , Proteínas Señalizadoras YAP/fisiología
7.
Mol Biol Rep ; 48(12): 7677-7688, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34648138

RESUMEN

BACKGROUND: Hepatic stellate cells (HSCs) are liver-resident myofibroblast precursors responsible for the production of collagen and maintenance of the hepatic extracellular matrix (ECM). As such, they are generally associated with fibrotic liver diseases. HSCs become "activated" in response to tissue damage or pathogen invasion, a process most commonly driven by transforming growth factor-ß1 (TGF-ß1). Despite this, the full extent of TGF-ß1 signalling in these cells is poorly understood. Clarifying the range and diversity of this signalling will further improve our understanding of the process of HSC activation. METHODS AND RESULTS: RNA sequencing was used to quantitate the transcriptomic changes induced in LX-2 cells, an activated human HSC line, following TGF-b1 treatment. In total, 5,258 genes were found to be significantly differentially expressed with a false discovery rate cut-off of < 0.1. The topmost deregulated of these genes included those with no currently characterised role in either HSC activation or fibrotic processes, including CIITA and SERPINB2. In silico analysis revealed the prominent signalling pathways downstream of TGF-ß1 in LX-2 cells. CONCLUSIONS: In this study, we describe the genes and signalling pathways significantly deregulated in LX-2 cells following TGF-ß1 treatment. We identified several highly deregulated genes with no currently characterised role in HSC activation, which may represent novel mediators of fibrotic responses in HSCs or the liver macroenvironment. This work may be of use in the identification of new markers of liver fibrosis and could provide insight into prospective genes or pathways that might be targeted for the amelioration of fibrotic liver disease in the future.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Actinas/genética , Secuencia de Bases/genética , Línea Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/genética , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Humanos , Hígado/metabolismo , Cirrosis Hepática/patología , Análisis de Secuencia de ARN/métodos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína smad3/metabolismo , Transcriptoma/genética , Factor de Crecimiento Transformador beta1/farmacología
8.
Physiol Res ; 70(6): 821-829, 2021 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-34717063

RESUMEN

Hepatic stellate cells (HSCs) are located in the space of Disse, between liver sinusoidal endothelia cells (LSECs) and hepatocytes. They have surprised and excited hepatologists for their biological characteristics. Under physiological quiescent conditions, HSCs are the major vitamin A-storing cells of the liver, playing crucial roles in the liver development, regeneration, and tissue homeostasis. Upon injury-induced activation, HSCs convert to a pro-fibrotic state, producing the excessive extracellular matrix (ECM) and promoting angiogenesis in the liver fibrogenesis. Activated HSCs significantly contribute to liver fibrosis progression and inactivated HSCs are key to liver fibrosis regression. In this review, we summarize the comprehensive understanding of HSCs features, including their roles in normal liver and liver fibrosis in hopes of advancing the development of emerging diagnosis and treatment for hepatic fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/etiología , Animales , Células Estrelladas Hepáticas/ultraestructura , Humanos , Hígado/embriología
9.
J Gastroenterol Hepatol ; 36(12): 3477-3486, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34547820

RESUMEN

BACKGROUND AND AIM: Protein O-GlcNAcylation is a critical post-translational modification regulating gene expression and fundamental cell functions. O-GlcNAc transferase (OGT) emerged as a key regulator of liver pathophysiology and disease. In this study, we aimed to evaluate the role of OGT in hepatic stellate cells (HSCs) and its consequent role in liver fibrosis. METHODS: Primary HSCs were isolated from C57/B6 mice. Cell morphology and α-SMA immunofluorescence staining were observed under scanning confocal microscope. Transcriptomic profile was evaluated by RNAseq (Illumina). Promoter activity was examined by luciferase and ß--Galactosidase reporter assays. Liver fibrosis mouse models were induced either by intraperitoneal injection of CCl4 at 3 times/week for 4 weeks or by feeding with methionine and choline deficient (MCD) diet for 4 weeks. RESULTS: OGT protein expression and protein O-GlcNAcylation were significantly decreased in CCl4 - or MCD diet-induced liver fibrosis as compared with normal liver in mice. OGT expression and protein O-GlcNAcylation were also decreased in primary HSCs isolated from liver with CCl4 -induced fibrosis compared with those from normal liver. RNA-seq showed that OGT knockdown in HSCs modulated key signaling pathways involved in HSC activation. Promoter sequence analysis of the differentially expressed genes predicted serum response factor (SRF) as a key transcription factor regulated by OGT. Luciferase reporter assay confirmed that OGT repressed activity of SRF to induce α-SMA transcription. Mutations of specific O-GlcNAcylation sites on SRF increased its transcriptional activity, validating negative regulation of SRF by OGT-mediated O-GlcNAcylation. CONCLUSIONS: Our results suggest that OGT functions as a negative regulator of HSC activation by promoting SRF O-GlcNAcylation to protect against liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas , N-Acetilglucosaminiltransferasas , Procesamiento Proteico-Postraduccional , Animales , Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/prevención & control , Ratones , N-Acetilglucosaminiltransferasas/metabolismo
10.
Am J Pathol ; 191(12): 2219-2230, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34428424

RESUMEN

Transdifferentiation (or activation) of hepatic stellate cells (HSCs) to myofibroblasts is a key event in liver fibrosis. Activated HSCs in the tumor microenvironment reportedly promote tumor progression. This study analyzed the effect of an inhibitor of HSC activation, retinol-binding protein-albumin domain III fusion protein (R-III), on protumorigenic functions of HSCs. Although conditioned medium collected from activated HSCs enhanced the migration, invasion, and proliferation of the hepatocellular carcinoma cell line Hepa-1c1c7, this effect was not observed in Hepa-1c1c7 cells treated with conditioned medium from R-III-exposed HSCs. In a subcutaneous tumor model, larger tumors with increased vascular density were formed in mice transplanted with Hepa-1c1c7+HSC than in mice transplanted with Hepa-1c1c7 cells alone. Intriguingly, when Hepa-1c1c7+HSC-transplanted mice were injected intravenously with R-III, a reduction in vascular density and extended tumor necrosis were observed. In an orthotopic tumor model, co-transplantation of HSCs enhanced tumor growth, angiogenesis, and regional metastasis accompanied by increased peritumoral lymphatic vessel density, which was abolished by R-III. In vitro study showed that R-III treatment affected the synthesis of pro-angiogenic and anti-angiogenic factors in activated HSCs, which might be the potential mechanism underlying the R-III effect. These findings suggest that the inhibition of HSC activation abrogates HSC-induced tumor angiogenesis and growth, which represents an attractive therapeutic strategy.


Asunto(s)
Carcinoma Hepatocelular/patología , Células Estrelladas Hepáticas/efectos de los fármacos , Neoplasias Hepáticas/patología , Proteínas Recombinantes de Fusión/farmacología , Albúminas/química , Albúminas/farmacología , Albúminas/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/terapia , Transformación Celular Neoplásica/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Femenino , Células Estrelladas Hepáticas/fisiología , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Dominios y Motivos de Interacción de Proteínas/fisiología , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas de Unión al Retinol/farmacología , Proteínas de Unión al Retinol/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Expert Rev Gastroenterol Hepatol ; 15(8): 879-890, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34225534

RESUMEN

Introduction: Hepatic stellate cells (HSCs) are essential for physiological homeostasis of the liver extracellular matrix (ECM). Excessive transdifferentiation of HSC from a quiescent to an activated phenotype contributes to disrupt this balance and can lead to liver fibrosis. Accumulating evidence has suggested that nuclear receptors (NRs) are involved in the regulation of HSC activation, proliferation, and function. Therefore, these NRs may be therapeutic targets to balance ECM homeostasis and inhibit HSC activation in liver fibrosis.Areas covered: In this review, the authors summarized the recent progress in the understanding of the regulatory role of NRs in HSCs and their potential as drug targets in liver fibrosis.Expert opinion: NRs are still potential therapy targets for inhibiting HSCs activation and liver fibrosis. However, the development of NRs agonists or antagonists to inhibit HSCs requires fully consideration of systemic effects.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/fisiopatología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Matriz Extracelular/fisiología , Células Estrelladas Hepáticas/efectos de los fármacos , Homeostasis , Humanos , Hígado/fisiología , Hígado/fisiopatología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores
12.
Sci Rep ; 11(1): 13380, 2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34183709

RESUMEN

Dietary palmitic acid (PA) promotes liver fibrosis in patients with nonalcoholic steatohepatitis (NASH). Herein, we clarified the intestinal absorption kinetics of dietary PA and effect of trans-portal PA on the activation of hepatic stellate cells (HSCs) involved in liver fibrosis in NASH. Blood PA levels after meals were significantly increased in patients with NASH compared to those in the control. Expression of genes associated with fat absorption and chylomicron formation, such as CD36 and MTP, was significantly increased in the intestine of NASH model rats compared with that in the controls. Plasma levels of glucagon-like peptide-2, involved in the upregulation of CD36 expression, were elevated in NASH rats compared with those in the controls. Furthermore, portal PA levels after meals in NASH rats were significantly higher than those in control and nonalcoholic fatty liver rats. Moreover, PA injection into the portal vein to the liver in control rats increased the mRNA levels associated with the activation of HSCs. Increased intestinal absorption of diet-derived PA was observed in NASH. Thus, the rapid increase in PA levels via the portal vein to the liver may activate HSCs and affect the development of liver fibrosis in NASH.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/fisiología , Absorción Intestinal/fisiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ácido Palmítico/metabolismo , Animales , Quilomicrones/metabolismo , Humanos , Hígado/metabolismo , Hígado/fisiopatología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
13.
Exp Cell Res ; 405(1): 112663, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34051242

RESUMEN

BACKGROUND: Hepatic stellate cell (HSC) differentiation/activation is central to liver fibrosis and is innately linked to the immune response to liver injury. Exosomes (EXOs) are important means of communication between cell populations. This study sought to characterize EXO release from HSCs and the effect of HSC-EXOs on macrophage cytokine release/function. METHODS: Liver from a rat fibrosis model was analyzed for EXO expression and localization. Quiescent and culture-activated rat and mouse HSCs and activated human HSCs were analyzed for microRNA expression. Mouse, rat, and human HSCs were culture-activated and EXOs purified from culture medium prior to addition to macrophages, and interleukin-6 (IL-6) and tumor necrosis factor-α (TNFα) mRNA and protein measured. The effect of activated HSC-EXOs on macrophage migration was assayed. RESULTS: Activation of rat HSCs led to increased EXO production in vivo, an effect mirrored by in vitro rat HSC culture-activation. Culture activation of mouse and rat HSCs led to altered EXO microRNA profiles, with a similar microRNA profile detected in activated human HSCs. Addition of activated HSC-EXOs to macrophages stimulated IL-6 and TNFα mRNA expression and protein secretion in mouse and human macrophages, but not for rat HSC-EXO-macrophages. Addition of human EXOs to macrophages stimulated migration, effects mirrored by the direct addition of rhIL-6 and rhTNFα. CONCLUSIONS: HSC-EXOs associate with macrophages and stimulate cytokine synthesis-release and macrophage migration. Constructing a comprehensive understanding of EXO interactions between liver cell populations in the setting of inflammation/fibrosis increases the potential for developing new diagnostic/therapeutic approaches.


Asunto(s)
Exosomas/fisiología , Células Estrelladas Hepáticas/fisiología , Inflamación/inmunología , Macrófagos/inmunología , Animales , Células Cultivadas , Citocinas/metabolismo , Células Estrelladas Hepáticas/citología , Humanos , Inflamación/metabolismo , Inflamación/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
14.
Hepatology ; 74(4): 2170-2185, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33932306

RESUMEN

BACKGROUND AND AIMS: The important roles of glutamate and metabotropic glutamate receptor 5 (mGluR5) in HSCs have recently been reported in various liver diseases; however, the mechanism linking the glutamine/glutamate metabolism and mGluR5 in liver fibrosis remains unclear. Here, we report that mGluR5 activation in natural killer (NK) cells attenuates liver fibrosis through increased cytotoxicity and interferon-γ (IFN-γ) production in both mice and humans. APPROACH AND RESULTS: Following 2-week injection of carbon tetrachloride (CCl4 ) or 5-week methionine-deficient and choline-deficient diet, liver fibrosis was more aggravated in mGluR5 knockout mice with significantly decreased frequency of NK cells compared with wild-type mice. Consistently, NK cell-specific mGluR5 knockout mice had aggravated CCl4 -induced liver fibrosis with decreased production of IFN-γ. Conversely, in vitro activation of mGluR5 in NK cells significantly increased the expression of anti-fibrosis-related genes including Ifng, Prf1 (perforin), and Klrk1 (killer cell lectin like receptor K1) and the production of IFN-γ through the mitogen-activated extracellular signal-regulated kinase/extracellular signal-related kinase pathway, contributing to the increased cytotoxicity against activated HSCs. However, we found that the uptake of glutamate was increased in activated HSCs, resulting in shortage of extracellular glutamate and reduced stimulation of mGluR5 in NK cells. Consequently, this could enable HSCs to evade NK cell cytotoxicity in advanced liver fibrosis. In vivo, pharmacologic activation of mGluR5 accelerated CCl4 -induced liver fibrosis regression by restoring NK cell cytotoxicity. In humans, mGluR5 activation enhanced the cytotoxicity of NK cells isolated from healthy donors, but not from patients with cirrhosis with significantly reduced mGluR5 expression in NK cells. CONCLUSIONS: mGluR5 plays important roles in attenuating liver fibrosis by augmenting NK cell cytotoxicity, which could be used as a potential therapeutic target for liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Interferón gamma/inmunología , Cirrosis Hepática , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Animales , Células Cultivadas , Citotoxicidad Inmunológica/inmunología , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Humanos , Células Asesinas Naturales/fisiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones
15.
Med Sci Monit ; 27: e929438, 2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33850093

RESUMEN

BACKGROUND Hepatic stellate cells (HSCs) play a vital role in hepatic fibrogenesis. Our recent clinical study indicated that the Zi Qi decoction, a Traditional Chinese Medicine formula, exhibited good efficacy in alleviating liver fibrosis, but the underlying mechanism remains elusive. MATERIAL AND METHODS Rats repeatedly injected with CCl4 and cells stimulated with lipopolysaccharide were used as in vivo and in vitro models for liver fibrosis, respectively. The viability of LX-2 cells was evaluated with MTT assay. Relative messenger RNA (mRNA) expression of representative extracellular matrix (ECM) components was detected with real-time quantitative polymerase chain reaction (RT-qPCR). Moreover, total and phosphorylation levels of ECM proteins and pathway-related proteins were detected with western blotting. Immunofluorescent staining was used to show the nuclear translocation of nuclear factor kappa b (NF-kappaB) p65. Hematoxylin & eosin (H&E) and Masson trichrome staining and immunohistochemistry were performed to evaluate the extent of liver fibrosis. The levels of alanine transaminase (ALT), aspartate transaminase (AST), gamma-glutamyl transpeptidase (GGT), Hyp, tumor necrosis factor-alpha (TNF-alpha), and interleukin-6 (IL-6) were tested with an enzyme-linked immunosorbent assay. In addition, 7.0T micro-magnetic resonance imaging (micro-MRI) was used to evaluate the severity of hepatic damage. RESULTS The Zi Qi decoction inhibited lipopolysaccharide-mediated upregulation of mRNA and protein levels of representative ECM proteins both in vivo and in vitro. The Zi Qi decoction also suppressed activation of the Toll-like receptor 4 (TLR4)-related NF-kappaB signaling pathway and subsequently inhibited the nuclear translocation of activated NF-kappaB. Moreover, another TLR4 downstream pathway, mitogen-activated protein kinase (MAPK), was simultaneously restrained. The results of liver pathology and MRI in rat models also suggested the efficacy of the Zi Qi decoction in attenuating liver damage. CONCLUSIONS The Zi Qi decoction inhibited liver fibrosis by inhibiting the TLR4-related NF-kB and MAPK signaling pathways and preventing activation of HSCs.


Asunto(s)
Mezclas Complejas/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/terapia , Hígado/metabolismo , Alanina Transaminasa/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Hígado/patología , Sistema de Señalización de MAP Quinasas , Masculino , Medicina Tradicional China , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Toll-Like 4/metabolismo
16.
Hepatol Commun ; 5(3): 358-370, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33681672

RESUMEN

The hepatic mesenchyme has been studied extensively in the context of liver fibrosis; however, much less is known regarding the role of mesenchymal cells during liver regeneration. As our knowledge of the cellular and molecular mechanisms driving hepatic regeneration deepens, the key role of the mesenchymal compartment during the regenerative response has been increasingly appreciated. Single-cell genomics approaches have recently uncovered both spatial and functional zonation of the hepatic mesenchyme in homeostasis and following liver injury. Here we discuss how the use of preclinical models, from in vivo mouse models to organoid-based systems, are helping to shape our understanding of the role of the mesenchyme during liver regeneration, and how these approaches should facilitate the precise identification of highly targeted, pro-regenerative therapies for patients with liver disease.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Hepatopatías/fisiopatología , Regeneración Hepática/fisiología , Hígado/citología , Mesodermo/citología , Animales , Células Cultivadas , Humanos , Hígado/fisiopatología , Mesodermo/fisiopatología , Ratones
17.
Biol Pharm Bull ; 44(1): 112-117, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390537

RESUMEN

Advanced glycation end-products (AGEs) are produced by the non-enzymatic reaction of sugars with proteins. It has been revealed that glyceraldehyde-derived toxic AGEs (TAGE) are elevated in the serum of non-alcoholic steatohepatitis (NASH) patients. NASH causes liver fibrosis and progresses to cirrhosis and hepatocellular carcinoma. However, the impact of TAGE in liver fibrosis caused by extracellular matrix accumulation remains poorly understood. In this study, we examined the effect of TAGE on the activation of hepatic stellate cells that are involved in liver fibrosis. LX-2 cells treated with transforming growth factor-ß1 (TGF-ß1) significantly reduced cell viability by apoptosis. However, the decrease in cell viability with TGF-ß1 treatment was significantly suppressed by TAGE co-treatment. The levels of α-smooth muscle actin (α-SMA) and platelet-derived growth factor (PDGF)-Rß and its ligand PDGF-B were increased in LX-2 cells following TGF-ß1 treatment, suggesting that these cells were activated; however, these increases were unaffected by TAGE co-treatment. Moreover, collagen I level was increased with TGF-ß1 treatment, and this increase was further increased by TAGE co-treatment. These results suggested that the suppression of apoptosis in activated LX-2 cells by TGF-ß1 and TAGE co-treatment is related to an increase in the production of the extracellular matrix such as collagen I. Therefore, it was suggested that TAGE might aggravate the liver fibrosis of chronic hepatitis, such as NASH.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Productos Finales de Glicación Avanzada/toxicidad , Células Estrelladas Hepáticas/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Células Estrelladas Hepáticas/patología , Células Estrelladas Hepáticas/fisiología , Humanos
18.
Nat Rev Gastroenterol Hepatol ; 18(3): 151-166, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33128017

RESUMEN

Chronic liver injury leads to liver inflammation and fibrosis, through which activated myofibroblasts in the liver secrete extracellular matrix proteins that generate the fibrous scar. The primary source of these myofibroblasts are the resident hepatic stellate cells. Clinical and experimental liver fibrosis regresses when the causative agent is removed, which is associated with the elimination of these activated myofibroblasts and resorption of the fibrous scar. Understanding the mechanisms of liver fibrosis regression could identify new therapeutic targets to treat liver fibrosis. This Review summarizes studies of the molecular mechanisms underlying the reversibility of liver fibrosis, including apoptosis and the inactivation of hepatic stellate cells, the crosstalk between the liver and the systems that orchestrate the recruitment of bone marrow-derived macrophages (and other inflammatory cells) driving fibrosis resolution, and the interactions between various cell types that lead to the intracellular signalling that induces fibrosis or its regression. We also discuss strategies to target hepatic myofibroblasts (for example, via apoptosis or inactivation) and the myeloid cells that degrade the matrix (for example, via their recruitment to fibrotic liver) to facilitate fibrosis resolution and liver regeneration.


Asunto(s)
Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Células Estrelladas Hepáticas/fisiología , Hepatocitos/fisiología , Humanos , Miofibroblastos/fisiología
19.
Drug Deliv ; 28(1): 10-18, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33336604

RESUMEN

Hepatic stellate cells (HSCs) play a crucial role in the progression of liver fibrosis, which can be considered as the specific therapeutic target of anti-fibrotic treatment. Targeted induction of HSCs to hepatocytes via delivery of clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (dCas9) system holds promise for hepatic fibrosis treatment. Our study here revealed that CRISPR/dCas9-VP64 system encapsulated in AML12 cell-derived exosomes could efficiently and successfully be delivered into the HSCs. In turn, the CRISPR/dCas9-VP64 system loaded in the exosomes can be efficiently released into the HSCs. As a proof-of-concept study, gRNA against hepatocyte nuclear factor 4α (HNF4α) together with the delivery of CRISPR/dCas9-VP64 system induced the HSCs to hepatocyte-like phenotype. In conclusion, our study here revealed that CRISPR/dCas9-VP64 system encapsulated in AML12 cell-derived exosomes could be functional in HSCs, emerging as a gene therapy strategy for hepatic fibrosis.


Asunto(s)
Sistemas CRISPR-Cas/genética , Exosomas/fisiología , Edición Génica/métodos , Células Estrelladas Hepáticas/fisiología , Animales , Línea Celular , Portadores de Fármacos , Humanos , Ratones , Ratones Endogámicos C57BL
20.
Am J Pathol ; 191(3): 438-453, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33345995

RESUMEN

Hepatic stellate cells (HSCs) are resident mesenchymal cells in the space of Disse interposed between liver sinusoidal endothelial cells and hepatocytes. Thorn-like microprojections, or spines, project out from the cell surface of HSCs, crossing the space of Disse, to establish adherens junctions with neighboring hepatocytes. Although HSC activation is initiated largely from stimulation by adjacent cells, isolated HSCs also activate spontaneously in primary culture on plastic. Therefore, other unknown HSC-initiating factors apart from paracrine stimuli may promote activation. The dissociation of adherens junctions between HSCs and hepatocytes as an activating signal for HSCs was explored, establishing epithelial cadherin (E-cadherin) as an adhesion molecule linking hepatocytes and HSCs. In vivo, following carbon tetrachloride-induced liver injury, HSCs lost their spines and dissociated from adherens junctions in the early stages of injury, and were subsequently activated along with an increase in YAP/TAZ expression. After abrogation of liver injury, HSCs reconstructed their spines and adherens junctions. In vitro, reconstitution of E-cadherin-containing adherens junctions by forced E-cadherin expression quiesced HSCs and suppressed TAZ expression. Additionally, increase of TAZ expression leading to the activation of HSCs by autocrine stimulation of transforming growth factor-ß, was revealed as a mechanism of spontaneous activation. Thus, we have uncovered a critical event required for HSC activation through enhanced TAZ-mediated mechanotransduction after the loss of adherens junctions between HSCs and hepatocytes.


Asunto(s)
Uniones Adherentes/fisiología , Cadherinas/metabolismo , Células Endoteliales/fisiología , Células Estrelladas Hepáticas/fisiología , Hepatocitos/fisiología , Mecanotransducción Celular , Animales , Proliferación Celular , Células Cultivadas , Células Endoteliales/citología , Células Estrelladas Hepáticas/citología , Hepatocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...