Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.127
Filtrer
1.
Front Immunol ; 15: 1425384, 2024.
Article de Anglais | MEDLINE | ID: mdl-39139565

RÉSUMÉ

Background: Schistosomiasis is a zoonotic parasitic disorder induced by the infestation of schistosomes, a genus of trematodes. MicroRNAs (miRNAs) in egg-derived exosomes are crucial for modulating the host's immune responses and orchestrating the pathophysiological mechanisms. Although the exosomes secreted by S. japonicum contain abundant miRNAs, the specific roles of these miRNAs in the pathogenesis of schistosomiasis-induced hepatic fibrosis are yet to be comprehensively elucidated. The egg exosomes of S. japonicum secrete miRNA-30, a novel miRNA. Methods: In vitro, the effect of miRNA-30 was evaluated by transfecting HSCs with miRNA mimics. The target gene biosignature for miRNA-30 was predicted using the miRDB software. The effect of miRNA-30 in hepatic fibrosis was evaluated by either elevating its expression in healthy mice or by inhibiting its activity in infected mice by administration of recombinant adeno-associated virus serotype eight vectors expressing miRNA-30 or miRNA sponges. Results: This novel miRNA can activate hepatic stellate cells (HSCs), the prinary effector cells of hepatic fibrosis, in vitro, i.e., it significantly increases the fibrogenic factors Col1(α1), Col3(α1), and α-SMA at both mRNA and protein levels. In addition, miRNA-30 may activate HSCs by targeting the host RORA gene. In addition, in vivo experiments were conducted by administering a recombinant adeno-associated viral vector to modulate the expression levels of miRNA-30. The overexpression of miRNA-30 in healthy mice significantly elevated the expression of Col1(α1), Col3(α1), and α-SMA at both the transcriptomic and proteomic scales. This overexpression was coupled with a pronounced augmentation in the hepatic hydroxyproline content. Conversely, the in vivo silencing of miRNA-30 in infected mice induced a considerable reduction in the size of hepatic granulomas and areas of collagen deposition. Hence, in vivo, modulation of miRNA-30 expression may play a pivotal role in ameliorating the severity of hepatic fibrosis in mice afflicted with S. japonica. Conclusions: The study results suggest that miRNA-30 may augment schistosomiasis-induced hepatic fibrosis through a probable interaction with the host RORA. Our study may improve the current theoretical framework regarding cross-species regulation by miRNAs of hepatic fibrosis in schistosomiasis.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , microARN , Schistosoma japonicum , Schistosomiase artérioveineuse , Animaux , microARN/génétique , Cirrhose du foie/parasitologie , Cirrhose du foie/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Souris , Schistosomiase artérioveineuse/immunologie , Schistosomiase artérioveineuse/génétique , Schistosomiase artérioveineuse/parasitologie , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/parasitologie , Exosomes/métabolisme , Exosomes/génétique , Femelle , Modèles animaux de maladie humaine , Ovule/métabolisme
2.
FASEB J ; 38(15): e23495, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39126242

RÉSUMÉ

Hepatic stellate cell (HSC) activation is the essential pathological process of liver fibrosis (LF). The molecular mechanisms regulating HSC activation and LF are incompletely understood. Here, we explored the effect of transcription factor SRY-related high mobility group box 7 (SOX7) on HSC activation and LF, and the underlying molecular mechanism. We found the expression levels of SOX7 were decreased in human and mouse fibrotic livers, particularly at the fibrotic foci. SOX7 was also downregulated in primary activated HSCs and TGF-ß1 stimulated LX-2 cells. SOX7 knockdown promoted activation and proliferation of LX-2 cells while inhibiting their apoptosis. On the other hand, overexpression of SOX7 suppressed the activation and proliferation of HSCs. Mechanistically, SOX7 attenuates HSC activation and LF by decreasing the expression of ß-catenin and phosphorylation of Smad2 and Smad3 induced by TGF-ß1. Furthermore, overexpression of SOX7 using AAV8-SOX7 mouse models ameliorated the extent of LF in response to CCl4 treatment in vivo. Collectively, SOX7 suppressed HSC activation and LF. Targeting SOX7, therefore, could be a potential novel strategy to protect against LF.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , Facteurs de transcription SOX-F , Cellules étoilées du foie/métabolisme , Animaux , Cirrhose du foie/métabolisme , Cirrhose du foie/génétique , Cirrhose du foie/anatomopathologie , Souris , Humains , Mâle , Facteurs de transcription SOX-F/métabolisme , Facteurs de transcription SOX-F/génétique , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Prolifération cellulaire , Souris de lignée C57BL , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Apoptose , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Lignée cellulaire , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique
3.
Sci Rep ; 14(1): 18840, 2024 08 13.
Article de Anglais | MEDLINE | ID: mdl-39138336

RÉSUMÉ

The combination of lineage tracing and immunohistochemistry has helped to identify subpopulations and fate of hepatic stellate cells (HSC) in murine liver. HSC are sinusoidal pericytes that act as myofibroblast precursors after liver injury. Single cell RNA sequencing approaches have recently helped to differentiate central and portal HSC. A specific Cre line to lineage trace portal HSC has not yet been described. We used three Cre lines (Lrat-Cre, PDGFRß-CreERT2 and SMMHC-CreERT2) known to label mesenchymal cells including HSC in combination with a tdTomato-expressing reporter. All three Cre lines labeled populations of HSC as well as smooth muscle cells (SMC). Using the SMMHC-CreERT2, we identified a subtype of HSC in the periportal area of the hepatic lobule (termed zone 1-HSC). We lineage traced tdTomato-expressing zone 1-HSC over 1 year, described fibrotic behavior in two fibrosis models and investigated their possible role during fibrosis. This HSC subtype resides in zone 1 under healthy conditions; however, zonation is disrupted in preclinical models of liver fibrosis (CCl4 and MASH). Zone 1-HSC do not transform into αSMA-expressing myofibroblasts. Rather, they participate in sinusoidal capillarization. We describe a novel subtype of HSC restricted to zone 1 under physiological conditions and its possible function after liver injury. In contrast to the accepted notion, this HSC subtype does not transform into αSMA-positive myofibroblasts; rather, zone 1-HSC adopt properties of capillary pericytes, thereby participating in sinusoidal capillarization.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , Myofibroblastes , Animaux , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Souris , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Foie/anatomopathologie , Foie/métabolisme , Péricytes/métabolisme , Péricytes/anatomopathologie , Lignage cellulaire , Mâle , Différenciation cellulaire , Modèles animaux de maladie humaine , Souris de lignée C57BL
4.
Clin Transl Med ; 14(8): e1793, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39113232

RÉSUMÉ

INTRODUCTION: Liver fibrosis is primarily driven by the activation of hepatic stellate cells (HSCs), which involves various epigenetic modifications. OBJECTIVES: N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotic cells, influences numerous physiological and pathological processes. Nevertheless, the role of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), a reader gene mediating m6A modifications, in liver fibrosis remains unclear. METHODS AND RESULTS: This study demonstrated that IGF2BP3 knockout reduces liver fibrosis by promoting HSC ferroptosis (FPT) and inactivating HSCs. Multi-omics analysis revealed that HSC-specific IGF2BP3 knockout decreased m6A content in Jagged1 (Jag1), a key component of the Notch signalling pathway. Furthermore, IGF2BP3 deficiency significantly reduced the expression of hairy and enhancer of split-1 (Hes1), a transcription factor in the Notch/Jag1 signalling pathway, with mRNA levels declining to 35%-62% and protein levels to 28%-35%. Additionally, it suppressed glutathione peroxidase 4 (GPX4) (decreased to approximately 31%-38%), a negative regulator of FPT, thereby facilitating HSC FPT progression and reducing profibrotic gene expression. CONCLUSION: These findings uncover a novel IGF2BP3/Notch/Jag1 signalling pathway involving HSC FPT, suggesting promising targets for ameliorating liver fibrosis. KEY POINTS/HIGHLIGHTS: IGF2BP3 deficiency inactivates Jag1 signalling. IGF2BP3 deficiency-mediated m6A modifications promote HSC ferroptosis. IGF2BP3 inhibition facilitates ferroptosis in HSCs via the Hes1/GPX4 axis. IGF2BP3 deficiency inactivates Jag1/Notch1/3/Hes1 signalling pathway inactivation, leading to the decrease in GPX4, which contributes to HSC ferroptosis.


Sujet(s)
Ferroptose , Cellules étoilées du foie , Protéine jagged-1 , Cirrhose du foie , Protéines de liaison à l'ARN , Récepteurs Notch , Transduction du signal , Ferroptose/génétique , Cellules étoilées du foie/métabolisme , Animaux , Cirrhose du foie/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Souris , Protéine jagged-1/génétique , Protéine jagged-1/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Transduction du signal/génétique , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique , Souris knockout , Mâle , Humains
5.
Stem Cell Res Ther ; 15(1): 223, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39044210

RÉSUMÉ

BACKGROUND: Hepatic stellate cells (HSC) have numerous critical roles in liver function and homeostasis, while they are also known for their importance during liver injury and fibrosis. There is therefore a need for relevant in vitro human HSC models to fill current knowledge gaps. In particular, the roles of vitamin A (VA), lipid droplets (LDs), and energy metabolism in human HSC activation are poorly understood. METHODS: In this study, human pluripotent stem cell-derived HSCs (scHSCs), benchmarked to human primary HSC, were exposed to 48-hour starvation of retinol (ROL) and palmitic acid (PA) in the presence or absence of the potent HSC activator TGF-ß. The interventions were studied by an extensive set of phenotypic and functional analyses, including transcriptomic analysis, measurement of activation-related proteins and cytokines, VA- and LD storage, and cell energy metabolism. RESULTS: The results show that though the starvation of ROL and PA alone did not induce scHSC activation, the starvation amplified the TGF-ß-induced activation-related transcriptome. However, TGF-ß-induced activation alone did not lead to a reduction in VA or LD stores. Additionally, reduced glycolysis and increased mitochondrial fission were observed in response to TGF-ß. CONCLUSIONS: scHSCs are robust models for activation studies. The loss of VA and LDs is not sufficient for scHSC activation in vitro, but may amplify the TGF-ß-induced activation response. Collectively, our work provides an extensive framework for studying human HSCs in healthy and diseased conditions.


Sujet(s)
Cellules étoilées du foie , Acide palmitique , Facteur de croissance transformant bêta , Rétinol , Humains , Rétinol/pharmacologie , Rétinol/métabolisme , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Acide palmitique/pharmacologie , Facteur de croissance transformant bêta/métabolisme , Gouttelettes lipidiques/métabolisme , Gouttelettes lipidiques/effets des médicaments et des substances chimiques , Cellules souches pluripotentes/métabolisme , Cellules souches pluripotentes/effets des médicaments et des substances chimiques , Cellules souches pluripotentes/cytologie , Métabolisme énergétique/effets des médicaments et des substances chimiques
6.
Signal Transduct Target Ther ; 9(1): 169, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38956074

RÉSUMÉ

More than 90% of hepatocellular carcinoma (HCC) cases develop in the presence of fibrosis or cirrhosis, making the tumor microenvironment (TME) of HCC distinctive due to the intricate interplay between cancer-associated fibroblasts (CAFs) and cancer stem cells (CSCs), which collectively regulate HCC progression. However, the mechanisms through which CSCs orchestrate the dynamics of the tumor stroma during HCC development remain elusive. Our study unveils a significant upregulation of Sema3C in fibrotic liver, HCC tissues, peripheral blood of HCC patients, as well as sorafenib-resistant tissues and cells, with its overexpression correlating with the acquisition of stemness properties in HCC. We further identify NRP1 and ITGB1 as pivotal functional receptors of Sema3C, activating downstream AKT/Gli1/c-Myc signaling pathways to bolster HCC self-renewal and tumor initiation. Additionally, HCC cells-derived Sema3C facilitated extracellular matrix (ECM) contraction and collagen deposition in vivo, while also promoting the proliferation and activation of hepatic stellate cells (HSCs). Mechanistically, Sema3C interacted with NRP1 and ITGB1 in HSCs, activating downstream NF-kB signaling, thereby stimulating the release of IL-6 and upregulating HMGCR expression, consequently enhancing cholesterol synthesis in HSCs. Furthermore, CAF-secreted TGF-ß1 activates AP1 signaling to augment Sema3C expression in HCC cells, establishing a positive feedback loop that accelerates HCC progression. Notably, blockade of Sema3C effectively inhibits tumor growth and sensitizes HCC cells to sorafenib in vivo. In sum, our findings spotlight Sema3C as a novel biomarker facilitating the crosstalk between CSCs and stroma during hepatocarcinogenesis, thereby offering a promising avenue for enhancing treatment efficacy and overcoming drug resistance in HCC.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Sémaphorines , Microenvironnement tumoral , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Humains , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Microenvironnement tumoral/génétique , Sémaphorines/génétique , Sémaphorines/métabolisme , Antigènes CD29/génétique , Antigènes CD29/métabolisme , Souris , Transduction du signal/génétique , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Neuropiline 1/génétique , Neuropiline 1/métabolisme , Lignée cellulaire tumorale , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Animaux , Régulation de l'expression des gènes tumoraux/génétique , Sorafénib/pharmacologie , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Évolution de la maladie
7.
J Cell Mol Med ; 28(13): e18529, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38984945

RÉSUMÉ

In this in vitro study, for the first time, we evaluate the effects of simvastatin-loaded liposome nanoparticles (SIM-LipoNPs) treatment on fibrosis-induced liver microtissues, as simvastatin (SIM) has shown potential benefits in the non-alcoholic fatty liver disease process. We developed multicellular liver microtissues composed of hepatic stellate cells, hepatoblastoma cells and human umbilical vein endothelial cells. The microtissues were supplemented with a combination of palmitic acid and oleic acid to develop fibrosis models. Subsequently, various groups of microtissues were exposed to SIM and SIM-LipoNPs at doses of 5 and 10 mg/mL. The effectiveness of the treatments was evaluated by analysing cell viability, production of reactive oxygen species (ROS) and nitric oxide (NO), the expression of Kruppel-like factor (KLF) 2, and pro-inflammatory cytokines (interleukin(IL)-1 α, IL-1 ß, IL-6 and tumour necrosis factor-α), and the expression of collagen I. Our results indicated that SIM-LipoNPs application showed promising results. SIM-LipoNPs effectively amplified the SIM-klf2-NO pathway at a lower dosage compatible with a high dosage of free SIM, which also led to reduced oxidative stress by decreasing ROS levels. SIM-LipoNPs administration also resulted in a significant reduction in pro-inflammatory cytokines and Collagen I mRNA levels, as a marker of fibrosis. In conclusion, our study highlights the considerable therapeutic potential of using SIM-LipoNPs to prevent liver fibrosis progress, underscoring the remarkable properties of SIM-LipoNPs in activating the KLF2-NO pathway and anti-oxidative and anti-inflammatory response.


Sujet(s)
Cellules étoilées du foie , Facteurs de transcription Krüppel-like , Liposomes , Cirrhose du foie , Nanoparticules , Espèces réactives de l'oxygène , Simvastatine , Humains , Simvastatine/pharmacologie , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Espèces réactives de l'oxygène/métabolisme , Nanoparticules/composition chimique , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Monoxyde d'azote/métabolisme
8.
Bull Exp Biol Med ; 177(1): 74-78, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38955854

RÉSUMÉ

Activated hepatic stellate cells differentiate into myofibroblasts, which synthesize and secrete extracellular matrix (ECM) leading to liver fibrosis. It was previously demonstrated that bulleyaconitine A (BLA), an alkaloid from Aconitum bulleyanum, inhibits proliferation and promotes apoptosis of human hepatic Lieming Xu-2 (LX-2) cells. In this study, we analyzed the effect of BLA on the production of ECM and related proteins by LX-2 cells activated with acetaldehyde (AA). The cells were randomized into the control group, AA group (cells activated with 400 µM AA), and BLA+AA group (cells cultured in the presence of 400 µM AA and 18.75 µg/ml BLA). In the BLA+AA group, the contents of collagens I and III and the expression of α-smooth muscle actin and transforming growth factor-ß1 (TGF-ß1) were statistically significantly higher than in the control, but lower than in the AA group. Expression of MMP-1 in the BLA+AA group was also significantly higher than in the AA group, but lower than in the control. Expression of TIMP-1 in the BLA+AA group was significantly higher than in the control, but lower than in the AA group. Thus, BLA suppressed activation and proliferation of LX-2 cells by inhibiting TGF-ß1 signaling pathway and decreasing the content of collagens I and III by reducing the MMP-1/TIMP-1 ratio.


Sujet(s)
Acétaldéhyde , Aconitine , Actines , Collagène de type I , Matrice extracellulaire , Cellules étoilées du foie , Inhibiteur tissulaire de métalloprotéinase-1 , Facteur de croissance transformant bêta-1 , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Humains , Acétaldéhyde/pharmacologie , Acétaldéhyde/analogues et dérivés , Aconitine/pharmacologie , Aconitine/analogues et dérivés , Collagène de type I/métabolisme , Collagène de type I/génétique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Inhibiteur tissulaire de métalloprotéinase-1/métabolisme , Inhibiteur tissulaire de métalloprotéinase-1/génétique , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Actines/métabolisme , Actines/génétique , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 1/génétique , Lignée cellulaire , Collagène de type III/métabolisme , Collagène de type III/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Aconitum/composition chimique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie
9.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39000518

RÉSUMÉ

While obesity-related nonalcoholic fatty liver disease (NAFLD) is linked with metabolic dysfunctions such as insulin resistance and adipose tissue inflammation, lean NAFLD more often progresses to liver fibrosis even in the absence of metabolic syndrome. This review aims to summarize the current knowledge regarding the mechanisms of liver fibrosis in lean NAFLD. The most commonly used lean NAFLD models include a methionine/choline-deficient (MCD) diet, a high-fat diet with carbon tetrachloride (CCl4), and a high-fructose and high-cholesterol diet. The major pro-fibrogenic mechanisms in lean NAFLD models include increased activation of the extracellular signal-regulated kinase (ERK) pathway, elevated expression of α-smooth muscle actin (α-SMA), collagen type I, and TGF-ß, and modulation of fibrogenic markers such as tenascin-X and metalloproteinase inhibitors. Additionally, activation of macrophage signaling pathways promoting hepatic stellate cell (HSC) activation further contributes to fibrosis development. Animal models cannot cover all clinical features that are evident in patients with lean or obese NAFLD, implicating the need for novel models, as well as for deeper comparisons of clinical and experimental studies. Having in mind the prevalence of fibrosis in lean NAFLD patients, by addressing specific pathways, clinical studies can reveal new targeted therapies along with novel biomarkers for early detection and enhancement of clinical management for lean NAFLD patients.


Sujet(s)
Cirrhose du foie , Stéatose hépatique non alcoolique , Obésité , Humains , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie , Obésité/métabolisme , Obésité/complications , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/étiologie , Animaux , Cellules étoilées du foie/métabolisme , Modèles animaux de maladie humaine
10.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-39000553

RÉSUMÉ

Nonalcoholic steatohepatitis (NASH) and alcoholic hepatitis (AH) affect a large part of the general population worldwide. Dysregulation of lipid metabolism and alcohol toxicity drive disease progression by the activation of hepatic stellate cells and the capillarization of liver sinusoidal endothelial cells. Collagen deposition, along with sinusoidal remodeling, alters sinusoid structure, resulting in hepatic inflammation, portal hypertension, liver failure, and other complications. Efforts were made to develop treatments for NASH and AH. However, the success of such treatments is limited and unpredictable. We report a strategy for NASH and AH treatment involving the induction of integrin αvß3-mediated cell apoptosis using a rationally designed protein (ProAgio). Integrin αvß3 is highly expressed in activated hepatic stellate cells (αHSCs), the angiogenic endothelium, and capillarized liver sinusoidal endothelial cells (caLSECs). ProAgio induces the apoptosis of these disease-driving cells, therefore decreasing collagen fibril, reversing sinusoid remodeling, and reducing immune cell infiltration. The reversal of sinusoid remodeling reduces the expression of leukocyte adhesion molecules on LSECs, thus decreasing leukocyte infiltration/activation in the diseased liver. Our studies present a novel and effective approach for NASH and AH treatment.


Sujet(s)
Cellules endothéliales , Cellules étoilées du foie , Hépatite alcoolique , Foie , Stéatose hépatique non alcoolique , Cellules étoilées du foie/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/traitement médicamenteux , Animaux , Cellules endothéliales/métabolisme , Hépatite alcoolique/métabolisme , Hépatite alcoolique/anatomopathologie , Foie/métabolisme , Foie/anatomopathologie , Apoptose , Humains , Intégrine alphaVbêta3/métabolisme , Mâle , Souris
11.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39063116

RÉSUMÉ

The burden of chronic liver disease is globally increasing at an alarming rate. Chronic liver injury leads to liver inflammation and fibrosis (LF) as critical determinants of long-term outcomes such as cirrhosis, liver cancer, and mortality. LF is a wound-healing process characterized by excessive deposition of extracellular matrix (ECM) proteins due to the activation of hepatic stellate cells (HSCs). In the healthy liver, quiescent HSCs metabolize and store retinoids. Upon fibrogenic activation, quiescent HSCs transdifferentiate into myofibroblasts; lose their vitamin A; upregulate α-smooth muscle actin; and produce proinflammatory soluble mediators, collagens, and inhibitors of ECM degradation. Activated HSCs are the main effector cells during hepatic fibrogenesis. In addition, the accumulation and activation of profibrogenic macrophages in response to hepatocyte death play a critical role in the initiation of HSC activation and survival. The main source of myofibroblasts is resident HSCs. Activated HSCs migrate to the site of active fibrogenesis to initiate the formation of a fibrous scar. Single-cell technologies revealed that quiescent HSCs are highly homogenous, while activated HSCs/myofibroblasts are much more heterogeneous. The complex process of inflammation results from the response of various hepatic cells to hepatocellular death and inflammatory signals related to intrahepatic injury pathways or extrahepatic mediators. Inflammatory processes modulate fibrogenesis by activating HSCs and, in turn, drive immune mechanisms via cytokines and chemokines. Increasing evidence also suggests that cellular stress responses contribute to fibrogenesis. Recent data demonstrated that LF can revert even at advanced stages of cirrhosis if the underlying cause is eliminated, which inhibits the inflammatory and profibrogenic cells. However, despite numerous clinical studies on plausible drug candidates, an approved antifibrotic therapy still remains elusive. This state-of-the-art review presents cellular and molecular mechanisms involved in hepatic fibrogenesis and its resolution, as well as comprehensively discusses the drivers linking liver injury to chronic liver inflammation and LF.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , Humains , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Animaux , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie
12.
Drug Des Devel Ther ; 18: 2715-2727, 2024.
Article de Anglais | MEDLINE | ID: mdl-38974122

RÉSUMÉ

Hepatic fibrosis (HF) is a pathological process of structural and functional impairment of the liver and is a key component in the progression of chronic liver disease. There are no specific anti-hepatic fibrosis (anti-HF) drugs, and HF can only be improved or prevented by alleviating the cause. Autophagy of hepatic stellate cells (HSCs) is closely related to the development of HF. In recent years, traditional Chinese medicine (TCM) has achieved good therapeutic effects in the prevention and treatment of HF. Several active ingredients from TCM (AITCM) can regulate autophagy in HSCs to exert anti-HF effects through different pathways, but relevant reviews are lacking. This paper reviewed the research progress of AITCM regulating HSCs autophagy against HF, and also discussed the relationship between HSCs autophagy and HF, pointing out the problems and limitations of the current study, in order to provide references for the development of anti-HF drugs targeting HSCs autophagy in TCM. By reviewing the literature in PubMed, Web of Science, Embase, CNKI and other databases, we found that the relationship between autophagy of HSCs and HF is currently controversial. HSCs autophagy may promote HF by consuming lipid droplets (LDs) to provide energy for their activation. However, in contrast, inducing autophagy in HSCs can exert the anti-HF effect by stimulating their apoptosis or senescence, reducing type I collagen accumulation, inhibiting the extracellular vesicles release, degrading pro-fibrotic factors and other mechanisms. Some AITCM inhibit HSCs autophagy to resist HF, with the most promising direction being to target LDs. While, others induce HSCs autophagy to resist HF, with the most promising direction being to target HSCs apoptosis. Future research needs to focus on cell targeting research, autophagy targeting research and in vivo verification research, and to explore the reasons for the contradictory effects of HSCs autophagy on HF.


Sujet(s)
Autophagie , Médicaments issus de plantes chinoises , Cellules étoilées du foie , Cirrhose du foie , Médecine traditionnelle chinoise , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Autophagie/effets des médicaments et des substances chimiques , Humains , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Animaux
13.
Anal Cell Pathol (Amst) ; 2024: 2751280, 2024.
Article de Anglais | MEDLINE | ID: mdl-38946862

RÉSUMÉ

Background: Biliary atresia (BA) is a devastating congenital disease characterized by inflammation and progressive liver fibrosis. Activation of hepatic stellate cells (HSCs) plays a central role in the pathogenesis of hepatic fibrosis. Our study aimed to investigate the pharmacological effect and potential mechanism of pirfenidone (PFD) and andrographolide (AGP) separately and together on liver fibrosis of BA. Materials and Methods: The bile ducts of male C57BL/6J mice were ligated or had the sham operation. The in vivo effects of PFD and/or AGP on liver fibrosis of BA were evaluated. Human hepatic stellate cells (LX-2) were also treated with PFD and/or AGP in vitro. Results: PFD and/or AGP ameliorates liver fibrosis and inflammation in the mice model of BA, as evidenced by significant downregulated in the accumulation of collagen fibers, hepatic fibrosis markers (α-SMA, collagen I, and collagen IV), and inflammatory markers (IL-1ß, IL-6, and TNF-α). Moreover, compared with monotherapy, these changes are more obvious in the combined treatment of PFD and AGP. Consistent with animal experiments, hepatic fibrosis markers (α-SMA, collagen I, and CTGF) and inflammatory markers (IL-1ß, IL-6, and TNF-α) were significantly decreased in activated LX-2 cells after PFD and/or AGP treatment. In addition, PFD and/or AGP inhibited the activation of HSCs by blocking the TGF-ß/Smad signaling pathway, and the combined treatment of PFD and AGP synergistically inhibited the phosphorylation of Smad2 and Smad3. Conclusion: The combined application of PFD and AGP exerted superior inhibitive effects on HSC activation and liver fibrosis by mediating the TGF-ß/Smad signaling pathway as compared to monotherapy. Therefore, the combination of PFD and AGP may be a promising treatment strategy for liver fibrosis in BA.


Sujet(s)
Diterpènes , Cellules étoilées du foie , Cirrhose du foie , Souris de lignée C57BL , Pyridones , Transduction du signal , Protéines Smad , Facteur de croissance transformant bêta , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Animaux , Cirrhose du foie/anatomopathologie , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Diterpènes/pharmacologie , Diterpènes/usage thérapeutique , Mâle , Facteur de croissance transformant bêta/métabolisme , Protéines Smad/métabolisme , Humains , Pyridones/pharmacologie , Lignée cellulaire , Souris , Atrésie des voies biliaires/anatomopathologie , Atrésie des voies biliaires/traitement médicamenteux , Atrésie des voies biliaires/métabolisme , Modèles animaux de maladie humaine , Association de médicaments
14.
Mol Med ; 30(1): 99, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982366

RÉSUMÉ

BACKGROUND: Enhanced glycolysis is a crucial metabolic event that drives the development of liver fibrosis, but the molecular mechanisms have not been fully understood. Lactate is the endproduct of glycolysis, which has recently been identified as a bioactive metabolite binding to G-protein-coupled receptor 81 (GPR81). We then questioned whether GPR81 is implicated in the development of liver fibrosis. METHODS: The level of GPR81 was determined in mice with carbon tetrachloride (CCl4)-induced liver fibrosis and in transforming growth factor beta 1 (TGF-ß1)-activated hepatic stellate cells (HSCs) LX-2. To investigate the significance of GPR81 in liver fibrosis, wild-type (WT) and GPR81 knockout (KO) mice were exposed to CCl4, and then the degree of liver fibrosis was determined. In addition, the GPR81 agonist 3,5-dihydroxybenzoic acid (DHBA) was supplemented in CCl4-challenged mice and TGF-ß1-activated LX-2 cells to further investigate the pathological roles of GPR81 on HSCs activation. RESULTS: CCl4 exposure or TGF-ß1 stimulation significantly upregulated the expression of GPR81, while deletion of GPR81 alleviated CCl4-induced elevation of aminotransferase, production of pro-inflammatory cytokines, and deposition of collagen. Consistently, the production of TGF-ß1, the expression of alpha-smooth muscle actin (α-SMA) and collagen I (COL1A1), as well as the elevation of hydroxyproline were suppressed in GPR81 deficient mice. Supplementation with DHBA enhanced CCl4-induced liver fibrogenesis in WT mice but not in GPR81 KO mice. DHBA also promoted TGF-ß1-induced LX-2 activation. Mechanistically, GPR81 suppressed cAMP/CREB and then inhibited the expression of Smad7, a negative regulator of Smad3, which resulted in increased phosphorylation of Smad3 and enhanced activation of HSCs. CONCLUSION: GPR81 might be a detrimental factor that promotes the development of liver fibrosis by regulating CREB/Smad7 pathway.


Sujet(s)
Tétrachloro-méthane , Protéine de liaison à l'élément de réponse à l'AMP cyclique , Cellules étoilées du foie , Cirrhose du foie , Souris knockout , Récepteurs couplés aux protéines G , Transduction du signal , Protéine Smad7 , Animaux , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/génétique , Cirrhose du foie/anatomopathologie , Cirrhose du foie/étiologie , Cirrhose du foie/induit chimiquement , Souris , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Cellules étoilées du foie/métabolisme , Protéine Smad7/métabolisme , Protéine Smad7/génétique , Facteur de croissance transformant bêta-1/métabolisme , Mâle , Humains , Lignée cellulaire , Modèles animaux de maladie humaine , Souris de lignée C57BL , Délétion de gène
15.
Sci Rep ; 14(1): 16897, 2024 07 23.
Article de Anglais | MEDLINE | ID: mdl-39043893

RÉSUMÉ

The chemokine (CCL)-chemokine receptor (CCR2) interaction, importantly CCL2-CCR2, involved in the intrahepatic recruitment of monocytes upon liver injury promotes liver fibrosis. CCL2-CCR2 antagonism using Cenicriviroc (CVC) showed promising results in several preclinical studies. Unfortunately, CVC failed in phase III clinical trials due to lack of efficacy to treat liver fibrosis. Lack of efficacy could be attributed to the fact that macrophages are also involved in disease resolution by secreting matrix metalloproteinases (MMPs) to degrade extracellular matrix (ECM), thereby inhibiting hepatic stellate cells (HSCs) activation. HSCs are the key pathogenic cell types in liver fibrosis that secrete excessive amounts of ECM causing liver stiffening and liver dysfunction. Knowing the detrimental role of intrahepatic monocyte recruitment, ECM, and HSCs activation during liver injury, we hypothesize that combining CVC and MMP (MMP1) could reverse liver fibrosis. We evaluated the effects of CVC, MMP1 and CVC + MMP1 in vitro and in vivo in CCl4-induced liver injury mouse model. We observed that CVC + MMP1 inhibited macrophage migration, and TGF-ß induced collagen-I expression in fibroblasts in vitro. In vivo, MMP1 + CVC significantly inhibited normalized liver weights, and improved liver function without any adverse effects. Moreover, MMP1 + CVC inhibited monocyte infiltration and liver inflammation as confirmed by F4/80 and CD11b staining, and TNFα gene expression. MMP1 + CVC also ameliorated liver fibrogenesis via inhibiting HSCs activation as assessed by collagen-I staining and collagen-I and α-SMA mRNA expression. In conclusion, we demonstrated that a combination therapeutic approach by combining CVC and MMP1 to inhibit intrahepatic monocyte recruitment and increasing collagen degradation respectively ameliorate liver inflammation and fibrosis.


Sujet(s)
Matrice extracellulaire , Cellules étoilées du foie , Cirrhose du foie , Matrix metalloproteinase 1 , Monocytes , Animaux , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 1/génétique , Souris , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Monocytes/métabolisme , Monocytes/effets des médicaments et des substances chimiques , Matrice extracellulaire/métabolisme , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL , Tétrachloro-méthane , Modèles animaux de maladie humaine , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Humains , Mouvement cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Imidazoles , Sulfoxydes
16.
J Agric Food Chem ; 72(28): 15740-15754, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38970822

RÉSUMÉ

Hepatic fibrosis is a compensatory response to chronic liver injury and inflammation, and dietary intervention is recommended as one of the fundamental prevention strategies. Raspberry ketone (RK) is an aromatic compound first isolated from raspberry and widely used to prepare food flavors. The current study investigated the hepatoprotection and potential mechanism of RK against hepatic fibrosis. In vitro, hepatic stellate cell (HSC) activation was stimulated with TGF-ß and cultured with RK, farnesoid X receptor (FXR), or peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) agonist or inhibitor, respectively. In vivo, C57BL/6 mice were injected intraperitoneally with thioacetamide (TAA) at 100/200 mg/kg from the first to the fifth week. Mice were intragastrically administrated with RK or Cur once a day from the second to the fifth week. In activated HSCs, RK inhibited extracellular matrix (ECM) accumulation, inflammation, and epithelial-mesenchymal transition (EMT) process. RK both activated FXR/PGC-1α and regulated their crosstalk, which were verified by their inhibitors and agonists. Deficiency of FXR or PGC-1α also attenuated the effect of RK on the reverse of activated HSCs. RK also decreased serum ALT/AST levels, liver histopathological change, ECM accumulation, inflammation, and EMT in mice caused by TAA. Double activation of FXR/PGC-1α might be the key targets for RK against hepatic fibrosis. Above all, these discoveries supported the potential of RK as a novel candidate for the dietary intervention of hepatic fibrosis.


Sujet(s)
Butanones , Cellules étoilées du foie , Cirrhose du foie , Souris de lignée C57BL , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Récepteurs cytoplasmiques et nucléaires , Transduction du signal , Animaux , Humains , Mâle , Souris , Butanones/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Cirrhose du foie/métabolisme , Cirrhose du foie/génétique , Cirrhose du foie/traitement médicamenteux , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Récepteurs cytoplasmiques et nucléaires/métabolisme , Récepteurs cytoplasmiques et nucléaires/génétique , Rubus/composition chimique , Transduction du signal/effets des médicaments et des substances chimiques , Rats
17.
J Cell Mol Med ; 28(12): e18458, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39031798

RÉSUMÉ

Schistosomiasis is a parasitic disease characterized by liver fibrosis, a process driven by the activation of hepatic stellate cells (HSCs) and subsequent collagen production. Previous studies from our laboratory have demonstrated the ability of Schistosoma japonicum protein P40 (SjP40) to inhibit HSCs activation and exert an antifibrotic effect. In this study, we aimed to elucidate the molecular mechanism underlying the inhibitory effect of recombinant SjP40 (rSjP40) on HSCs activation. Using a cell model in which rSjP40 inhibited LX-2 cell activation, we performed RNA-seq analyses and identified ATF3 as the most significantly altered gene. Further investigation revealed that rSjP40 inhibited HSCs activation partly by suppressing ATF3 activation. Knockdown of ATF3 in mouse liver significantly alleviated S. japonicum-induced liver fibrosis. Moreover, our results indicate that ATF3 is a direct target of microRNA-494-3p, a microRNA associated with anti-liver fibrosis effects. rSjP40 was found to downregulate ATF3 expression by upregulating microRNA-494-3p in LX-2 cells. This downregulation led to the inhibition of the expression of liver fibrosis proteins α-SMA and COL1A1, ultimately alleviating liver fibrosis caused by S. japonicum.


Sujet(s)
Facteur de transcription ATF-3 , Protéines d'helminthes , Cellules étoilées du foie , Cirrhose du foie , microARN , Schistosoma japonicum , Schistosomiase artérioveineuse , Animaux , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/parasitologie , Schistosomiase artérioveineuse/parasitologie , Schistosomiase artérioveineuse/métabolisme , Schistosomiase artérioveineuse/génétique , Cirrhose du foie/parasitologie , Cirrhose du foie/génétique , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Souris , Humains , microARN/génétique , microARN/métabolisme , Protéines d'helminthes/génétique , Protéines d'helminthes/métabolisme , Actines/métabolisme , Actines/génétique , Lignée cellulaire , Régulation de l'expression des gènes , Foie/métabolisme , Foie/parasitologie , Foie/anatomopathologie , Modèles animaux de maladie humaine , Antigènes d'helminthe
18.
Chem Biol Interact ; 399: 111139, 2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-38992766

RÉSUMÉ

Carbon tetrachloride (CCl4) has a wide range of toxic effects, especially causing acute liver injury (ALI), in which rapid compensation for hepatocyte loss ensures liver survival, but proliferation of surviving hepatocytes (known as endoreplication) may imply impaired residual function. Yes-associated protein (YAP) drives hepatocytes to undergo endoreplication and ploidy, the underlying mechanisms of which remain a mystery. In the present study, we uncover during CCl4-mediated ALI accompanied by increased hepatocytes proliferation and YAP activation. Notably, bioinformatics analyses elucidate that hepatic-specific deletion of YAP substantially ameliorated CCl4-induced hepatic proliferation, effectively decreased the vitamin D receptor (VDR) expression. Additionally, a mouse model of acute liver injury substantiated that inhibition of YAP could suppress hepatocytes proliferation via VDR. Furthermore, we also disclosed that the VDR agonist nullifies CCl4-induced ALI alleviated by the YAP inhibitor in vivo. Importantly, hepatocytes were isolated from mice, and it was spotlighted that the anti-proliferative impact of the YAP inhibitor was abolished by the activation of VDR within these hepatocytes. Similarly, primary hepatic stellate cells (HSCs) were isolated and it was manifested that YAP inhibitor suppressed HSC activation via VDR during acute liver injury. Our findings further elucidate the YAP's role in ALI and may provide new avenues for protection against CCl4-drived acute liver injury.


Sujet(s)
Tétrachloro-méthane , Prolifération cellulaire , Lésions hépatiques dues aux substances , Cellules étoilées du foie , Hépatocytes , Récepteur calcitriol , Protéines de signalisation YAP , Animaux , Récepteur calcitriol/métabolisme , Tétrachloro-méthane/toxicité , Souris , Protéines de signalisation YAP/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/traitement médicamenteux , Lésions hépatiques dues aux substances/anatomopathologie , Mâle , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Protéines adaptatrices de la transduction du signal/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie
19.
Int Immunopharmacol ; 138: 112554, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38968861

RÉSUMÉ

BACKGROUND: Human placental mesenchymal stromal cells (hPMSCs) are known to limit graft-versus-host disease (GVHD). CD8+CD122+PD-1+Tregs have been shown to improve the survival of GVHD mice. However, the regulatory roles of hPMSCs in this subgroup remain unclear. Here, the regulatory mechanism of hPMSCs in reducing liver fibrosis in GVHD mice by promoting CD8+CD122+PD-1+Tregs formation and controlling the balance of IL-6 and IL-10 were explored. METHODS: A GVHD mouse model was constructed using C57BL/6J and BALB/c mice and treated with hPMSCs. LX-2 cells were explored to study the effects of IL-6 and IL-10 on the activation of hepatic stellate cells (HSCs). The percentage of CD8+CD122+PD-1+Tregs and IL-10 secretion were determined using FCM. Changes in hepatic tissue were analysed by HE, Masson, multiple immunohistochemical staining and ELISA, and the effects of IL-6 and IL-10 on LX-2 cells were detected using western blotting. RESULTS: hPMSCs enhanced CD8+CD122+PD-1+Treg formation via the CD73/Foxo1 and promoted IL-10, p53, and MMP-8 levels, but inhibited IL-6, HLF, α-SMA, Col1α1, and Fn levels in the liver of GVHD mice through CD73. Positive and negative correlations of IL-6 and IL-10 between HLF were found in liver tissue, respectively. IL-6 upregulated HLF, α-SMA, and Col1α1 expression via JAK2/STAT3 pathway, whereas IL-10 upregulated p53 and inhibited α-SMA and Col1α1 expression in LX-2 cells by activating STAT3. CONCLUSIONS: hPMSCs promoted CD8+CD122+PD-1+Treg formation and IL-10 secretion but inhibited HSCs activation and α-SMA and Col1α1 expression by CD73, thus controlling the balance of IL-6 and IL-10, and alleviating liver injury in GVHD mice.


Sujet(s)
Protéine O1 à motif en tête de fourche , Maladie du greffon contre l'hôte , Cellules souches mésenchymateuses , Lymphocytes T régulateurs , Animaux , Femelle , Humains , Souris , Grossesse , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Modèles animaux de maladie humaine , Protéine O1 à motif en tête de fourche/métabolisme , Maladie du greffon contre l'hôte/immunologie , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/immunologie , Interleukine-10/métabolisme , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Interleukine-6/métabolisme , Foie/anatomopathologie , Foie/immunologie , Foie/métabolisme , Cirrhose du foie/immunologie , Cirrhose du foie/thérapie , Cirrhose du foie/métabolisme , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/immunologie , Souris de lignée BALB C , Souris de lignée C57BL , Placenta/cytologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme
20.
J Med Chem ; 67(14): 12085-12098, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38991128

RÉSUMÉ

Liver fibrosis is a condition characterized by aberrant proliferation of connective tissue in the liver resulting from diverse etiological factors. G protein-coupled receptor GPR55 has recently been identified as a regulator of liver diseases. Herein, we report the discovery of a cyclic peptide P1-1 that antagonizes GPR55 and suppresses collagen secretion in hepatic stellate cells. The alanine scanning and docking study was carried out to predict the binding mode and allowed for further structural optimization of peptide antagonists for GPR55. The subsequent in vivo study demonstrated that P1-1 ameliorates CCl4-induce and MCD-diet-induce acute liver inflammation and fibrosis. Further study indicates that P1-1 reduces reactive oxygen species (ROS) production, attenuates ER stress, and inhibits mitochondria-associated hepatocyte apoptosis. In this work, we provided the first successful example of antagonizing GPR55 for liver inflammation and fibrosis, which validates GPR55 as a promising target for the treatment of liver fibrosis and affords a high-potent GPR55 antagonist P1-1 as a potential therapeutic candidate.


Sujet(s)
Cirrhose du foie , Récepteurs de cannabinoïdes , Récepteurs couplés aux protéines G , Animaux , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Récepteurs couplés aux protéines G/antagonistes et inhibiteurs , Récepteurs couplés aux protéines G/métabolisme , Humains , Récepteurs de cannabinoïdes/métabolisme , Souris , Mâle , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Simulation de docking moléculaire , Souris de lignée C57BL , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Peptides cycliques/pharmacologie , Peptides cycliques/composition chimique , Peptides cycliques/synthèse chimique , Peptides cycliques/usage thérapeutique , Découverte de médicament , Relation structure-activité , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE