Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Ann Hepatol ; 29(5): 101517, 2024.
Article in English | MEDLINE | ID: mdl-38852781

ABSTRACT

INTRODUCTION AND OBJECTIVES: Liver fibrosis remains a complication derived from a chronic Hepatitis C Virus (HCV) infection even when it is resolved, and no liver antifibrotic drug has been approved. Molecular mechanisms on hepatocytes and activation of hepatic stellate cells (HSCs) play a central role in liver fibrogenesis. To elucidate molecular mechanisms, it is important to analyze pathway regulation during HSC activation and HCV infection. MATERIALS AND METHODS: We evaluate the fibrosis-associated molecular mechanisms during a co-culture of human HSCs (LX2), with human hepatocytes (Huh7) that express HCV NS5A or Core protein. We evaluated LX2 activation induced by HCV NS5A or Core expression in Huh7 cells during co-culture. We determined a fibrosis-associated gene expression profile in Huh7 that expresses NS5A or Core proteins during the co-culture with LX2. RESULTS: We observed that NS5A induced 8.3-, 6.7- and 4-fold changes and that Core induced 6.5-, 1.8-, and 6.2-fold changes in the collagen1, TGFß1, and timp1 gene expression, respectively, in LX2 co-cultured with transfected Huh7. In addition, NS5A induced the expression of 30 genes while Core induced 41 genes and reduced the expression of 30 genes related to fibrosis in Huh7 cells during the co-culture with LX2, compared to control. The molecular pathways enriched from the gene expression profile were involved in TGFB signaling and the organization of extracellular matrix. CONCLUSIONS: We demonstrated that HCV NS5A and Core protein expression regulate LX2 activation. NS5A and Core-induced LX2 activation, in turn, regulates diverse fibrosis-related gene expression at different levels in Huh7, which can be further analyzed as potential antifibrotic targets during HCV infection.


Subject(s)
Coculture Techniques , Collagen Type I , Hepacivirus , Hepatic Stellate Cells , Hepatocytes , Liver Cirrhosis , Tissue Inhibitor of Metalloproteinase-1 , Transforming Growth Factor beta1 , Viral Core Proteins , Viral Nonstructural Proteins , Humans , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Hepacivirus/genetics , Hepatocytes/metabolism , Hepatocytes/virology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Collagen Type I/metabolism , Collagen Type I/genetics , Viral Core Proteins/genetics , Viral Core Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Gene Expression Regulation , Signal Transduction , Collagen Type I, alpha 1 Chain/genetics , Collagen Type I, alpha 1 Chain/metabolism , Gene Expression Profiling/methods , Cell Line, Tumor , RNA-Dependent RNA Polymerase
2.
Arch Med Res ; 55(2): 102937, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38301446

ABSTRACT

BACKGROUND: The nasal vaccine HB-ATV-8 has emerged as a promising approach for NAFLD (non-alcoholic fatty liver disease) and atherosclerosis prevention. HB-ATV-8 contains peptide seq-1 derived from the carboxy-end of the Cholesteryl Ester Transfer Protein (CETP), shown to reduce liver fibrosis, inflammation, and atherosclerotic plaque formation in animal models. Beyond the fact that this vaccine induces B-cell lymphocytes to code for antibodies against the seq-1 sequence, inhibiting CETP's cholesterol transfer activity, we have hypothesized that beyond the modulation of CETP activity carried out by neutralizing antibodies, the observed molecular effects may also correspond to the direct action of peptide seq-1 on diverse cellular systems and molecular features involved in the development of liver fibrosis. METHODS: The HepG2 hepatoma-derived cell line was employed to establish an in vitro steatosis model. To obtain a conditioned cell medium to be used with hepatic stellate cell (HSC) cultures, HepG2 cells were exposed to fatty acids or fatty acids plus peptide seq-1, and the culture medium was collected. Gene regulation of COL1A1, ACTA2, TGF-ß, and the expression of proteins COL1A1, MMP-2, and TIMP-2 were studied. AIM: To establish an in vitro steatosis model employing HepG2 cells that mimics molecular processes observed in vivo during the onset of liver fibrosis. To evaluate the effect of peptide Seq-1 on lipid accumulation and pro-fibrotic responses. To study the effect of Seq-1-treated steatotic HepG2 cell supernatants on lipid accumulation, oxidative stress, and pro-fibrotic responses in HSC. RESULTS AND CONCLUSION: Peptide seq-1-treated HepG2 cells show a downregulation of COLIA1, ACTA2, and TGF-ß genes, and a decreased expression of proteins such as COL1A1, MMP-2, and TIMP-2, associated with the remodeling of extracellular matrix components. The same results are observed when HSCs are incubated with peptide Seq-1-treated steatotic HepG2 cell supernatants. The present study consolidates the nasal vaccine HB-ATV-8 as a new prospect in the treatment of NASH directly associated with the development of cardiovascular disease.


Subject(s)
Non-alcoholic Fatty Liver Disease , Vaccines , Animals , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Matrix Metalloproteinase 2 , Cholesterol Ester Transfer Proteins/metabolism , Down-Regulation , Hepatocytes/metabolism , Fibrosis , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/prevention & control , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Fatty Acids/metabolism , Lipids/pharmacology , Liver/metabolism
3.
Cells ; 13(3)2024 Feb 04.
Article in English | MEDLINE | ID: mdl-38334674

ABSTRACT

Different cellular mechanisms influence steatotic liver disease (SLD) progression. The influence of different levels of steatogenic inputs has not been studied in hepatocytes and hepatic stellate cells (HSCs). METHODS: HepG2 hepatocytes and LX-2 HSCs were cultured in mild (MS) and severe (SS) steatogenic conditions. TGF-ß stimulation was also tested for HSCs in control (T) and steatogenic conditions (MS-T and SS-T). Steatosis was stained with Oil Red, and the proliferation was assayed via WST-8 reduction, apoptosis via flow cytometry, and senescence via SA-ß-galactosidase activity. RESULTS: Regarding hepatocytes, steatosis progressively increased; proliferation was lower in MS and SS; and the viability of both conditions significantly decreased at 72 h. Apoptosis increased in MS at 72 h, while it decreased in SS. Senescence increased in MS and diminished in SS. Regarding HSCs, the SS and SS-T groups showed no proliferation, and the viability was reduced in MS at 72 h and in SS and SS-T. The LX-2 cells showed increased apoptosis in SS and SS-T at 24 h, and in MS and MS-T at 72 h. Senescence decreased in MS, SS, and SS-T. CONCLUSIONS: Lipid overload induces differential effects depending on the cell type, the steatogenic input level, and the exposure time. Hepatocytes are resilient to mild steatosis but susceptible to high lipotoxicity. HSCs are sensitive to lipid overload, undergoing apoptosis and lowering senescence and proliferation. Collectively, these data may help explain the development of steatosis and fibrosis in SLD.


Subject(s)
Fatty Liver , Hepatic Stellate Cells , Humans , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Fatty Liver/metabolism , Cell Proliferation , Hyperplasia/metabolism , Apoptosis , Lipids
4.
Technol Cancer Res Treat ; 22: 15330338231163677, 2023.
Article in English | MEDLINE | ID: mdl-36938618

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common liver cancer. It is highly lethal and has high recurrence. Death among HCC patients occur mainly due to tumor progression, recurrence, metastasis, and chemoresistance. Cancer stem cells (CSCs) are cell subpopulations within the tumor that promote invasion, recurrence, metastasis, and drug resistance. Hepatic stellate cells (HSCs) are important components of the tumor microenvironment (TME) responsible for primary secretory ECM proteins during liver injury and inflammation. These cells promote fibrogenesis, infiltrate the tumor stroma, and contribute to HCC development. Interactions between HSC and CSC and their microenvironment help promote carcinogenesis through different mechanisms. This review summarizes the roles of CSCs and HSCs in establishing the TME in primary liver tumors and describes their involvement in HCC chemoresistance.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Cell Line, Tumor , Cell Movement , Neoplastic Stem Cells/metabolism , Tumor Microenvironment
5.
Ann Hepatol ; 28(1): 100775, 2023.
Article in English | MEDLINE | ID: mdl-36280014

ABSTRACT

INTRODUCTION AND OBJECTIVES: Liver fibrosis is a common pathological change in many chronic liver diseases. Activation of hepatic stellate cells (HSCs) is the core event in liver fibrosis. This study aimed to investigate the role of testicular orphan receptor 4 (TR4) in the activation of HSCs. MATERIALS AND METHODS: In vivo, bile duct ligation (BDL)-induced rat liver fibrosis model was established, and the expressions of TR4 and α-smooth muscle actin (α-SMA) in liver tissues were detected. In vitro, TR4 knockdown and overexpression in JS-1 cells using lentiviral vectors were constructed, and the expressions of TR4, α-SMA, Col-I, and TGF-ß1/smads and retinoid X receptor (RXR) pathway-related genes were detected. RESULTS: TR4 was highly expressed in BDL-induced fibrotic liver, accompanied by increased expression of α-SMA. Knockdown of TR4 significantly inhibited the expressions of α-SMA, Col-I, p-TßRI, and p-Smad2/3, and up-regulated the expression of RXRα in HSCs in vitro. In contrast, TR4 overexpression significantly increased the expressions of α-SMA, Col-I, p-TßRI, and p-Smad2/3, and inhibited the expression of RXRα. CONCLUSIONS: TR4 may promote the activation of HSCs by up-regulating TßR I/Smad2/3 signaling pathway and down-regulating RXRα signaling, thereby promoting the progression of liver fibrosis. Our findings may provide a new therapeutic target against hepatic fibrosis.


Subject(s)
Hepatic Stellate Cells , Transforming Growth Factor beta1 , Rats , Animals , Hepatic Stellate Cells/metabolism , Transforming Growth Factor beta1/metabolism , Liver Cirrhosis/metabolism , Signal Transduction , Liver/pathology , Receptors, Transforming Growth Factor beta/metabolism
6.
Phytomedicine ; 107: 154466, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36182796

ABSTRACT

BACKGROUND: Liver fibrosis is a common scarring response and may ultimately lead to liver cancer, unfortunately, there is currently no effective antifibrotic drug approved for human use. Limonoids exhibit a broad spectrum of biological activities; however, the potential role of limonoids against fibrosis is largely unknown. PURPOSE: This study investigates the antifibrotic activities and potential mechanisms of TKF (3-tigloyl-khasenegasin F), a natural mexicanolide-type limonoid derivative. STUDY DESIGN/METHODS: Two well-established mouse models (CCl4 challenge and bile duct ligation) were used to assess anti-fibrotic effects of TKF in vivo. Human hepatic stellate cell (HSC) line LX-2 and mouse primary hepatic stellate cells (pHSCs) also served as in vitro liver fibrosis models. RESULT: TKF administration significantly attenuated hepatic histopathological injury and collagen accumulation and suppressed fibrogenesis-associated gene expression including Col1a1, Acta2, and Timp1. In LX-2 cells and mouse pHSCs, TKF dose-dependently suppressed HSC activation and the expression levels of fibrogenic markers. Mechanistic studies showed that TKF inhibited Notch3-Hes1 and YAP signalings in vivo and in vitro. Furthermore, YAP inhibition or knockdown downregulated the Notch3 expression; however, Notch3 inhibition or knockdown did not affect the level of YAP in activated HSC. We revealed that TKF inhibited Notch3-Hes1 activation and downregulated hepatic fibrogenic gene expression via inhibiting YAP. CONCLUSION: The therapeutic benefit of TKF against liver fibrosis results from inhibition of YAP and Notch3-Hes1 pathways, indicating that TKF may be a novel therapeutic candidate for liver fibrosis.


Subject(s)
Hepatic Stellate Cells , Limonins , Animals , Fibrosis , Humans , Limonins/pharmacology , Liver/metabolism , Liver Cirrhosis/metabolism , Mice , Receptor, Notch3/metabolism
7.
Environ Toxicol ; 36(2): 168-176, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32918399

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) affects around 25% of the worldwide population. Non-alcoholic steatohepatitis (NASH), the more progressive variant of NAFLD, is characterized by steatosis, cellular ballooning, lobular inflammation, and may culminate on hepatic stellate cell (HSC) activation, thus increasing the risk for fibrosis, cirrhosis, and HCC development. Conversely, the antifibrotic effects of sorafenib, an FDA-approved drug for HCC treatment, have been demonstrated in 2D cell cultures and animal models, but its mechanisms in a NAFLD-related microenvironment in vitro requires further investigation. Thus, a human 3D co-culture model of fatty hepatocytes and HSC was established by culturing hepatoma C3A cells, pre-treated with 1.32 mM oleic acid, with HSC LX-2 cells. The fatty C3A/LX-2 spheroids showed morphological and molecular hallmarks of altered lipid metabolism and steatosis-induced fibrogenesis, similarly to the human disease. Sorafenib (15 µM) for 72 hours reduced fatty spheroid viability, and upregulated the expression of lipid oxidation- and hydrolysis-related genes, CPT1 and LIPC, respectively. Sorafenib also inhibited steatosis-induced fibrogenesis by downregulating COL1A1, TGFB1, PDGF, and TIMP1 and by decreasing protein levels of IL-6, TGF-ß1, and TNF-α in fatty spheroids. The demonstration of the antifibrotic properties of sorafenib on steatosis-induced fibrogenesis in a 3D in vitro model of NAFLD supports its clinical use as a therapeutic agent for the treatment of NAFLD/NASH patients.


Subject(s)
Hepatic Stellate Cells/drug effects , Hepatocytes/drug effects , Liver Cirrhosis/prevention & control , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Sorafenib/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Fibrosis , Hepatic Stellate Cells/pathology , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/metabolism
8.
Mol Cell Biochem ; 476(2): 649-661, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33073314

ABSTRACT

The phytoalexin Resveratrol (3,5,4'-trihydroxystilbene; RSV) has been related to numerous beneficial effects on health by its cytoprotection and chemoprevention activities. Liver fibrosis is characterized by the extracellular matrix accumulation after hepatic injury and can lead to cirrhosis. Hepatic stellate cells (HSC) play a crucial role during fibrogenesis and liver wound healing by changing their quiescent phenotype to an activated phenotype for protecting healthy areas from damaged areas. Strategies on promoting the activated HSC death, the quiescence return or the cellular activation stimuli decrease play an important role on reducing liver fibrosis. Here, we evaluated the RSV effects on some markers of activation in GRX, an HSC model. We further evaluated the RSV influence in the ability of GRX on releasing inflammatory mediators. RSV at 1 and 10 µM did not alter the protein content of α-SMA, collagen I and GFAP; but 50 µM increased the content of these activation-related proteins. Also, RSV did not change the myofibroblast-like morphology of GRX. Interestingly, RSV at 10 and 50 µM decreased the GRX migration and collagen-I gel contraction. Finally, we showed that RSV triggered the increase in the TNF-α and IL-10 content in culture media of GRX while the opposite occurred for the IL-6 content. Altogether, these results suggested that RSV did not decrease the activation state of GRX and oppositely, triggered a pro-activation effect at the 50 µM concentration. However, despite the increase of TNF- α in culture media, these results on IL-6 and IL-10 secretion were in accordance with the anti-inflammatory role of RSV in our model.


Subject(s)
Antioxidants/pharmacology , Cytokines/metabolism , Hepatic Stellate Cells/drug effects , Inflammation/drug therapy , Liver Cirrhosis/drug therapy , Resveratrol/pharmacology , Animals , Cell Line , Cell Proliferation , Hepatic Stellate Cells/immunology , Hepatic Stellate Cells/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Myofibroblasts/drug effects , Myofibroblasts/metabolism
9.
Ann Hepatol ; 22: 100281, 2021.
Article in English | MEDLINE | ID: mdl-33220464

ABSTRACT

INTRODUCTION AND OBJECTIVES: Hepatic fibrosis is characterized by the accumulation of extracellular matrix which includes the accumulation of α-smooth muscle actin (α-SMA), collagen type I (COL1α1), as well as remodeling induced by metalloproteinases and tissue inhibitor of metalloproteinase (TIMPs), where hepatic stellate cells (HSCs) play a central role. In addition, the transcription factor SNAI1 (which participates in epithelial-mesenchymal transition, EMT) and mitofusin 2 (MFN2, a mitochondrial marker) plays an important role in chronic liver disease. Turnera diffusa (TD), a Mexican endemic plant, has been shown to possess antioxidant and hepatoprotective activity in vitro. We treated human HSC (LX2 cells) with a methanolic extract of Turnera diffusa (METD) to evaluate the mechanism involved in its hepatoprotective effect measured as fibrosis modulation, EMT, and mitochondrial markers. MATERIALS AND METHODS: HSC LX-2 cells were treated with METD (100 and 200ng/mL) alone or combined with TGF-ß (10ng/mL) at different time points (24, 48, and 72h). α-SMA, COL1α1, MMP2, TIMP1, SNAI1, and MFN2 mRNAs and protein levels were determined by real-time quantitative PCR and Western Blot analysis. RESULTS: We found that METD decreases COL1α1-mRNA, α-SMA, and TIMP1 protein expression in LX2 cells treated with and TGF-ß. This treatment also decreases MFN2 and TIMP1 protein expression and induces overexpression of MMP2-mRNA. CONCLUSIONS: Our results suggest that a methanolic extract of Turnera diffusa is associated with an antifibrotic effect by decreasing profibrotic and mitochondrial markers together with the possible induction of apoptosis through SNAI1 expression in activated HSC cells.


Subject(s)
Hepatic Stellate Cells/drug effects , Liver Cirrhosis/prevention & control , Plant Extracts/pharmacology , Turnera , Actins/metabolism , Cell Culture Techniques , Collagen Type I, alpha 1 Chain/metabolism , GTP Phosphohydrolases/metabolism , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Matrix Metalloproteinase 2/metabolism , Mitochondrial Proteins/metabolism , Snail Family Transcription Factors/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism
10.
Antioxidants (Basel) ; 9(12)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333846

ABSTRACT

Activated hepatic stellate cells (HSCs) and myofibroblasts are the main producers of extracellular matrix (ECM) proteins that form the fibrotic tissue that leads to hepatic fibrosis. Reactive oxygen species (ROS) can directly activate HSCs or induce inflammation or programmed cell death, especially pyroptosis, in hepatocytes, which in turn activates HSCs and fibroblasts to produce ECM proteins. Therefore, antioxidants and the nuclear factor E2-related factor-2 signaling pathway play critical roles in modulating the profibrogenic response. The master proinflammatory factors nuclear factor-κB (NF-κB) and the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome may coordinate to produce and activate profibrogenic molecules such as interleukins 1ß and 18, which effectively activate HSCs, to produce large amounts of fibrotic proteins. Furthermore, the NLRP3 inflammasome activates pro-caspase 1, which is upregulated by NF-κB, to produce caspase 1, which induces pyroptosis via gasdermin and the activation of HSCs. ROS play central roles in the activation of the NF-κB and NLRP3 signaling pathways via IκB (an inhibitor of NF-κB) and thioredoxin-interacting protein, respectively, thereby linking the molecular mechanisms of oxidative stress, inflammation and fibrosis. Elucidating these molecular pathways may pave the way for the development of therapeutic tools to interfere with specific targets.

11.
Cells ; 9(1)2020 01 11.
Article in English | MEDLINE | ID: mdl-31940814

ABSTRACT

Cirrhosis, a late form of liver disease, is characterized by extensive scarring due to exacerbated secretion of extracellular matrix proteins by myofibroblasts that develop during this process. These myofibroblasts arise mainly from hepatic stellate cells (HSCs), liver-specific pericytes that become activated at the onset of liver injury. Consequently, HSCs tend to be viewed mainly as myofibroblast precursors in a fibrotic process driven by inflammation. Here, the molecular interactions between liver pericytes and inflammatory cells such as macrophages and neutrophils at the first moments after injury and during the healing process are brought into focus. Data on HSCs and pericytes from other tissues indicate that these cells are able to sense pathogen- and damage-associated molecular patterns and have an important proinflammatory role in the initial stages of liver injury. On the other hand, further data suggest that as the healing process evolves, activated HSCs play a role in skewing the initial proinflammatory (M1) macrophage polarization by contributing to the emergence of alternatively activated, pro-regenerative (M2-like) macrophages. Finally, data suggesting that some HSCs activated during liver injury could behave as hepatic progenitor or stem cells will be discussed.


Subject(s)
Inflammation/metabolism , Liver Diseases/metabolism , Liver/metabolism , Myofibroblasts/metabolism , Pericytes/metabolism , Animals , Humans , Inflammation/pathology , Liver/pathology , Liver Diseases/pathology , Myofibroblasts/pathology , Pericytes/pathology
12.
J Cell Biochem ; 120(11): 19031-19043, 2019 11.
Article in English | MEDLINE | ID: mdl-31257611

ABSTRACT

Caveolin-1 (Cav-1) expression is increased in hepatic stellate cells (HSC) upon liver cirrhosis and it functions as an integral membrane protein of lipid rafts and caveolae that regulates and integrates multiple signals as a platform. This study aimed to evaluate the role of Cav-1 in HSC. Thus, the effects of exogenous expression of Cav-1 in GRX cells, a model of activated HSC, were determined. Here, we demonstrated through evaluating well-known HSC activation markers - such as α-smooth muscle actin, collagen I, and glial fibrillary acidic protein - that up regulation of Cav-1 induced GRX to a more activated phenotype. GRXEGFP-Cav1 presented an increased migration, an altered adhesion pattern, a reorganization f-actin cytoskeleton, an arrested cell cycle, a modified cellular ultrastructure, and a raised endocytic flux. Based on this, GRX EGFP-Cav1 represents a new cellular model that can be an important tool for understanding of events related to HSC activation. Furthermore, our results reinforce the role of Cav-1 as a molecular marker of HSC activation.


Subject(s)
Caveolin 1/biosynthesis , Cell Cycle Checkpoints , Gene Expression , Hepatic Stellate Cells/metabolism , Caveolin 1/genetics , Cell Line , Hepatic Stellate Cells/cytology , Humans
13.
Nutr Res Rev ; 32(1): 128-145, 2019 06.
Article in English | MEDLINE | ID: mdl-30707092

ABSTRACT

Early-life nutrition plays a critical role in fetal growth and development. Food intake absence and excess are the two main types of energy malnutrition that predispose to the appearance of diseases in adulthood, according to the hypothesis of 'developmental origins of health and disease'. Epidemiological data have shown an association between early-life malnutrition and the metabolic syndrome in later life. Evidence has also demonstrated that nutrition during this period of life can affect the development of the immune system through epigenetic mechanisms. Thus, epigenetics has an essential role in the complex interplay between environmental factors and genetics. Altogether, this leads to the inflammatory response that is commonly seen in non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of the metabolic syndrome. In conjunction, DNA methylation, covalent modification of histones and the expression of non-coding RNA are the epigenetic phenomena that affect inflammatory processes in the context of NAFLD. Here, we highlight current understanding of the mechanisms underlying developmental programming of NAFLD linked to epigenetic modulation of the immune system and environmental factors, such as malnutrition.


Subject(s)
Epigenesis, Genetic , Immune System/physiology , Liver/pathology , Malnutrition/complications , Maternal Nutritional Physiological Phenomena , Non-alcoholic Fatty Liver Disease/etiology , Nutritional Status , Carcinoma, Hepatocellular/etiology , DNA Methylation , Female , Histones , Humans , Inflammation/etiology , Metabolic Syndrome/etiology , MicroRNAs , Pregnancy , Prenatal Exposure Delayed Effects
14.
Front Immunol ; 10: 3036, 2019.
Article in English | MEDLINE | ID: mdl-32038610

ABSTRACT

In human brucellosis, the liver is frequently affected. Brucella abortus triggers a profibrotic response on hepatic stellate cells (HSCs) characterized by inhibition of MMP-9 with concomitant collagen deposition and TGF-ß1 secretion through type 4 secretion system (T4SS). Taking into account that it has been reported that the inflammasome is necessary to induce a fibrotic phenotype in HSC, we hypothesized that Brucella infection might create a microenvironment that would promote inflammasome activation with concomitant profibrogenic phenotype in HSCs. B. abortus infection induces IL-1ß secretion in HSCs in a T4SS-dependent manner. The expression of caspase-1 (Casp-1), absent in melanoma 2 (AIM2), Nod-like receptor (NLR) containing a pyrin domain 3 (NLRP3), and apoptosis-associated speck-like protein containing a CARD (ASC) was increased in B. abortus-infected HSC. When infection experiments were performed in the presence of glyburide, a compound that inhibits NLRP3 inflammasome, or A151, a specific AIM2 inhibitor, the secretion of IL-1ß was significantly inhibited with respect to uninfected controls. The role of inflammasome activation in the induction of a fibrogenic phenotype in HSCs was determined by performing B. abortus infection experiments in the presence of the inhibitors Ac-YVAD-cmk and glyburide. Both inhibitors were able to reverse the effect of B. abortus infection on the fibrotic phenotype in HSCs. Finally, the role of inflammasome in fibrosis was corroborated in vivo by the reduction of fibrotic patches in liver from B. abortus-infected ASC, NLRP, AIM2, and cCasp-1/11 knock-out (KO) mice with respect to infected wild-type mice.


Subject(s)
Brucella abortus/physiology , Brucellosis/immunology , Hepatic Stellate Cells/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Animals , Brucella abortus/genetics , Brucellosis/genetics , Brucellosis/microbiology , Caspase 1/genetics , Caspase 1/immunology , Fibrosis/genetics , Fibrosis/immunology , Fibrosis/microbiology , Hepatic Stellate Cells/microbiology , Humans , Inflammasomes/genetics , Interleukin-1beta/genetics , Liver/immunology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology
15.
Cell Biol Int ; 43(3): 333-343, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30468282

ABSTRACT

Hepatic fibrosis and its end-stage cirrhosis have increased worldwide, and, despite all the efforts, no successful therapy is available. More recently, the heptapeptide angiotensin-(1-7) [ang-(1-7)] was reported to be able to modulate liver fibrosis and even steatosis; however, the molecular bases of these effects are not clear. In this study, we investigated the overexpression of the microRNA-1254 in the human hepatic stellate cell line LX-2, based on the effect of the heptapeptide in such cells, previously, demonstrated by our research group. In addition, this miRNA was chosen based on the identification of putative binding site of this small molecule in the mRNA sequences of different molecular connectors of the AKT/ PI3K pathway, which is modulated by the heptapeptide and connects to the control of several cellular mechanisms, including proliferation, survival, migration, and even liver fibrogenesis. The results revealed an innovative function of the miR-1254 in controlling SMAD3 and pro-fibrosing elements as well as the wound healing response in LX-2, attenuating the scaring repair of the injured tissue. The combined findings provide useful information for future studies on the controlling of hepatic fibrogenesis.


Subject(s)
Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , MicroRNAs/metabolism , Smad3 Protein/metabolism , Wound Healing , Base Sequence , Cell Line , Humans , MicroRNAs/genetics , Transcription, Genetic
16.
World J Gastroenterol ; 24(16): 1679-1707, 2018 Apr 28.
Article in English | MEDLINE | ID: mdl-29713125

ABSTRACT

Liver diseases are caused by different etiological agents, mainly alcohol consumption, viruses, drug intoxication or malnutrition. Frequently, liver diseases are initiated by oxidative stress and inflammation that lead to the excessive production of extracellular matrix (ECM), followed by a progression to fibrosis, cirrhosis and hepatocellular carcinoma (HCC). It has been reported that some natural products display hepatoprotective properties. Naringenin is a flavonoid with antioxidant, antifibrogenic, anti-inflammatory and anticancer properties that is capable of preventing liver damage caused by different agents. The main protective effects of naringenin in liver diseases are the inhibition of oxidative stress, transforming growth factor (TGF-ß) pathway and the prevention of the transdifferentiation of hepatic stellate cells (HSC), leading to decreased collagen synthesis. Other effects include the inhibition of the mitogen activated protein kinase (MAPK), toll-like receptor (TLR) and TGF-ß non-canonical pathways, the inhibition of which further results in a strong reduction in ECM synthesis and deposition. In addition, naringenin has shown beneficial effects on nonalcoholic fatty liver disease (NAFLD) through the regulation of lipid metabolism, modulating the synthesis and oxidation of lipids and cholesterol. Moreover, naringenin protects from HCC, since it inhibits growth factors such as TGF-ß and vascular endothelial growth factor (VEGF), inducing apoptosis and regulating MAPK pathways. Naringenin is safe and acts by targeting multiple proteins. However, it possesses low bioavailability and high intestinal metabolism. In this regard, formulations, such as nanoparticles or liposomes, have been developed to improve naringenin bioavailability. We conclude that naringenin should be considered in the future as an important candidate in the treatment of different liver diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Anticarcinogenic Agents/therapeutic use , Antioxidants/therapeutic use , Flavanones/therapeutic use , Liver Diseases/drug therapy , Liver/drug effects , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , Anticarcinogenic Agents/adverse effects , Anticarcinogenic Agents/pharmacokinetics , Antioxidants/adverse effects , Antioxidants/pharmacokinetics , Collagen/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Flavanones/adverse effects , Flavanones/pharmacokinetics , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Inflammation Mediators/metabolism , Lipid Metabolism/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases/diagnosis , Liver Diseases/etiology , Liver Diseases/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects
17.
Ann Hepatol ; 16(6): 959-965, 2017.
Article in English | MEDLINE | ID: mdl-29055933

ABSTRACT

BACKGROUND: Acute exposure to high concentrations of microcystin-LR (MC-LR) can cause significant hepatocyte injury. AIM: To document the effects of long-term, low-dose MC-LR exposure on hepatic inflammation and fibrosis in mice with healthy and diseased livers. MATERIAL AND METHODS: Male CD1 mice (N = 20/group) were exposed to 1.0 µg/L of MC-LR in drinking water; 1.0 µg/L MC-LR plus 300 mg/L of the hepatotoxin thioacetamide (MC-LR/TAA); or 300 mg/L TAA alone for 28 weeks. Liver biochemistry and histology were documented at the end of the study period. In addition, hepatic stellate cells (HSCs), were exposed in vitro to MC-LR (0.1-10,000 µg/L) and monitored for changes in cell metabolism, proliferation and activation. RESULTS: Liver biochemistry and histology were essentially normal in MC-LR alone exposed mice. MC-LR/TAA and TAA alone exposed mice had significant hepatic inflammation and fibrosis but the extent of the changes were similar in the two groups. In vitro, MC-LR had no effect on HSC metabolism, proliferation or activation. CONCLUSION: Long-term, low-dose exposure to MC-LR is unlikely to lead to chronic liver disease in the setting of a normal liver or exacerbate existing liver disease in the setting of ongoing hepatitis.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Experimental/chemically induced , Liver/drug effects , Microcystins/toxicity , Animals , Cell Line , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Energy Metabolism/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Marine Toxins , Mice , Risk Assessment , Severity of Illness Index , Thioacetamide , Time Factors
18.
Fundam Clin Pharmacol ; 31(6): 610-624, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28802065

ABSTRACT

The ability of quercetin to reverse an established cirrhosis has not yet been investigated. Therefore, the aim of this study was to examine the efficacy of this flavonoid in reversing experimental cirrhosis. Cirrhosis was induced by intraperitoneal administration of TAA (200 mg/kg of body weight) three times per week for 8 weeks or by intraperitoneal petrolatum-CCl4 (400 mg/kg of body weight) administration three times per week for 8 weeks. To determine the capacity of quercetin to prevent liver fibrosis, the flavonoid (50 mg/kg of body weight, p.o.) was administered daily to rats during the CCl4 or TAA treatment. To evaluate the ability of quercetin to reverse the previously induced cirrhosis, we first treated rats with CCl4 for 8 weeks, as previously described and then the flavonoid was administered for four more weeks. We found that the liver anti-inflammatory and antinecrotic effects of quercetin are associated with its antioxidant properties, to the ability of the flavonoid to block NF-κB activation and in consequence to reduce cytokine IL-1. The ability of quercetin to reverse fibrosis may be associated with the capacity of the flavonoid to decrease TGF-ß levels, hepatic stellate cell activation, and to promote degradation of the ECM by increasing metalloproteinases. The main conclusion is that quercetin, in addition to its liver protective activity against TAA chronic intoxication, is also capable of reversing a well-stablished cirrhosis by blocking the prooxidant processes and by downregulating the inflammatory and profibrotic responses.


Subject(s)
Antioxidants/pharmacology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis/immunology , Quercetin/pharmacology , Animals , Disease Models, Animal , Immunomodulation , Liver Cirrhosis, Experimental/pathology , Male , NF-kappa B/drug effects , Rats , Rats, Wistar
19.
Cell Biol Int ; 41(9): 946-959, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28498509

ABSTRACT

Liver fibrosis is a pathophysiological process correlated with intense repair and cicatrization mechanisms in injured liver, and over the past few years, the characterization of the fine-tuning of molecular interconnections that support the development of liver fibrosis has been investigated. In this cellular process, the hepatic stellate cells (HSCs) support the organ fibrogenesis. The HSCs are found in two distinct morpho-physiological states: quiescent and activated. In normal liver, most HSCs are found in quiescent state, presenting a considerable amount of lipid droplets in the cytoplasm, while in injured liver, the activated phenotype of HSCs is a myofibroblast, that secrete extracellular matrix elements and contribute to the establishment of the fibrotic process. Studies on the molecular mechanisms by which HSCs try to restore their quiescent state have been performed; however, no effective treatment to reverse fibrosis has been so far prescribed. Therefore, the elucidation of the cellular and molecular mechanisms of apoptosis, senescence, and the cell reversion phenotype process from activate to quiescent state will certainly contribute to the development of effective therapies to treat hepatic fibrosis. In this context, this review aimed to address central elements of apoptosis, senescence, and reversal of HSC phenotype in the control of hepatic fibrogenesis, as a guide to future development of therapeutic strategies.


Subject(s)
Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Aging/physiology , Animals , Apoptosis/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Liver Cirrhosis/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Signal Transduction
20.
Hepatol Res ; 47(9): 928-940, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27653239

ABSTRACT

AIM: The aim of this study was to investigate the effects of rosuvastatin in a model of diet-induced obesity and non-alcoholic fatty liver disease, with attention to the activation of hepatic stellate cells (HSCs). METHOD: Male C57BL/6 mice received a control diet (C; 10% energy as lipids) or a high-fat diet (HF; 50% energy as lipids) for 12 weeks, followed by 7 weeks of treatment. Group CR received control diet + rosuvastatin; group HFR received high-fat diet + rosuvastatin. RESULTS: The HF group showed higher insulin, total cholesterol, triacylglycerol, and leptin levels than the C group, all of which were significantly diminished by rosuvastatin in the HFR group. The HF group had greater steatosis and activated HSCs than the C group, whereas rosuvastatin diminished the steatosis (less 21%, P < 0.001) and significantly inhibited the activation of the HSCs in the HFR group compared to the HF group. The sterol regulatory element-binding protein-1 and the peroxisome proliferator-activated receptor (PPAR)-γ protein expressions were increased in HF animals and reduced after treatment in the HFR group. By contrast, low PPAR-α and carnitine palmitoyltransferase-1 expressions were found in the HF group, and were restored by rosuvastatin treatment in the HFR group. CONCLUSION: Rosuvastatin mitigated hepatic steatosis by modulating PPAR balance, favoring PPAR-α over PPAR-γ downstream effects. The effects were accompanied by a diminishing of insulin resistance, the anti-inflammatory adipokine profile, and HSC activation, avoiding non-alcoholic fatty liver disease progression and non-alcoholic steatohepatitis onset in this model.

SELECTION OF CITATIONS
SEARCH DETAIL