Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.583
Filtrar
1.
Sci Rep ; 14(1): 9425, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658618

RESUMO

Liver fibrosis, as a consequence of chronic liver disease, involves the activation of hepatic stellate cell (HSC) caused by various chronic liver injuries. Emerging evidence suggests that activation of HSC during an inflammatory state can lead to abnormal accumulation of extracellular matrix (ECM). Investigating novel strategies to inhibit HSC activation and proliferation holds significant importance for the treatment of liver fibrosis. As a member of the doublecortin domain-containing family, doublecortin domain containing 2 (DCDC2) mutations can lead to neonatal sclerosing cholangitis, but its involvement in liver fibrosis remains unclear. Therefore, this study aims to elucidate the role of DCDC2 in liver fibrosis. Our findings revealed a reduction in DCDC2 expression in both human fibrotic liver tissues and carbon tetrachloride (CCl4)-induced mouse liver fibrotic tissues. Furthermore, exposure to transforming growth factor beta-1(TGF-ß1) stimulation resulted in a dose- and time-dependent decrease in DCDC2 expression. The overexpression of DCDC2 inhibited the expression of α-smooth muscle actin (α-SMA) and type I collagen alpha 1 (Col1α1), and reduced the activation of HSC stimulated with TGF-ß1. Additionally, we provided evidence that the Wnt/ß-catenin signaling pathway was involved in this process, wherein DCDC2 was observed to inhibit ß-catenin activation, thereby preventing its nuclear translocation. Furthermore, our findings demonstrated that DCDC2 could attenuate the proliferation and epithelial-mesenchymal transition (EMT)-like processes of HSC. In vivo, exogenous DCDC2 could ameliorate CCl4-induced liver fibrosis. In summary, DCDC2 was remarkably downregulated in liver fibrotic tissues of both humans and mice, as well as in TGF-ß1-activated HSC. DCDC2 inhibited the activation of HSC induced by TGF-ß1 in vitro and fibrogenic changes in vivo, suggesting that it is a promising therapeutic target for liver fibrosis and warrants further investigation in clinical practice.


Assuntos
Tetracloreto de Carbono , Células Estreladas do Fígado , Cirrose Hepática , Via de Sinalização Wnt , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Cirrose Hepática/metabolismo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Cirrose Hepática/tratamento farmacológico , Humanos , Camundongos , Masculino , beta Catenina/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proliferação de Células , Camundongos Endogâmicos C57BL
2.
Cell Mol Biol (Noisy-le-grand) ; 70(2): 183-188, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38430023

RESUMO

This study aimed to elucidate the effect of mitochondria-targeted reactive oxygen species (ROS) blockor SS-31 on hepatic stellate cells (HSC) activation during liver fibrosis. TGF-ß1 was employed to induce HSC activation, while MitoSOX Red was utilized to assess the presence of mitochondrial ROS. The mitochondrial membrane potential (MMP) was measured using the JC-1 probe, and the ATP level was determined using a specific kit. The proliferation of HSCs was assessed using CCK-8 and colony formation assays, whereas flow cytometry was employed to detect HSC apoptosis. Fibrotic markers (COL1A1 and α-SMA) and NLRP3 inflammasome components (NLRP3, caspase-1, and ASC) were analyzed via Western blotting. Liver fibrosis was induced in mice using CCl4, and subsequently, histopathological changes were observed through HE staining and Masson staining. In TGF-ß1-activated HSCs, mitochondrial ROS expression increased, MMP and ATP content decreased, indicating mitochondrial damage. After TGF-ß1 induction, HSC proliferation increased, apoptosis decreased, and COL1A1, α-SMA, and NLRP3 inflammasome protein expression increased. After SS-31 treatment, mitochondrial ROS expression decreased, MMP recovered, ATP level increased, HSC proliferation decreased, apoptosis increased, and the expressions of COL1A1, α-SMA, and NLRP3 inflammasome decreased. NLRP3 blockor MCC950 treatment blocked HSC activation. CCL4-induced liver fibrosis mice had inflammatory cell infiltration and significant collagen fiber deposition in the liver. After SS-31 treatment, liver inflammation and collagen deposition were significantly reduced. SS-31, as a mitochondria-targeted ROS blockor, can block HSC activation by regulating the NLRP3 inflammasome, thereby alleviating liver fibrosis.


Assuntos
Células Estreladas do Fígado , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Animais , Camundongos , Trifosfato de Adenosina/metabolismo , Colágeno/metabolismo , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
3.
Liver Int ; 43(11): 2523-2537, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37641479

RESUMO

BACKGROUND AND PURPOSE: Liver fibrosis is a wound-healing reaction that eventually leads to cirrhosis. Hydronidone is a new pyridine derivative with the potential to treat liver fibrosis. In this study, we explored the antifibrotic effects of hydronidone and its potential mode of action. METHODS: The anti-hepatic fibrosis effects of hydronidone were studied in carbon tetrachloride (CCl4 )- and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)- induced animal liver fibrosis. The antifibrotic mechanisms of hydronidone were investigated in hepatic stellate cells (HSCs). The antifibrotic effect of hydronidone was further tested after Smad7 knockdown in HSCs in mouse models of fibrosis. RESULTS: In animal models, hydronidone attenuated liver damage and collagen accumulation, and reduced the expression of fibrosis-related genes. Hydronidone decreased the expression of fibrotic genes in HSCs. Impressively, hydronidone significantly upregulated Smad7 expression and promoted the degradation of transforming growth factor ß receptor I (TGFßRI) in HSCs and thus inhibited the TGFß-Smad signalling pathway. Specific knockdown of Smad7 in HSCs in vivo blocked the antifibrotic effect of hydronidone. CONCLUSION: Hydronidone ameliorates liver fibrosis by inhibiting HSCs activation via Smad7-mediated TGFßRI degradation. Hydronidone is a potential drug candidate for the treatment of liver fibrosis.


Assuntos
Cirrose Hepática , Transdução de Sinais , Fator de Crescimento Transformador beta , Animais , Camundongos , Tetracloreto de Carbono/toxicidade , Tetracloreto de Carbono/metabolismo , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Receptor do Fator de Crescimento Transformador beta Tipo I , Fator de Crescimento Transformador beta/metabolismo , Proteína Smad7/efeitos dos fármacos , Proteína Smad7/metabolismo
4.
Cell Death Dis ; 13(11): 1011, 2022 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-36446766

RESUMO

11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1) is a key enzyme that catalyzes the intracellular conversion of cortisone to physiologically active cortisol. Although 11ßHSD1 has been implicated in numerous metabolic syndromes, such as obesity and diabetes, the functional roles of 11ßHSD1 during progression of nonalcoholic steatohepatitis (NASH) and consequent fibrosis have not been fully elucidated. We found that pharmacological and genetic inhibition of 11ßHSD1 resulted in reprogramming of hepatic stellate cell (HSC) activation via inhibition of p-SMAD3, α-SMA, Snail, and Col1A1 in a fibrotic environment and in multicellular hepatic spheroids (MCHSs). We also determined that 11ßHSD1 contributes to the maintenance of NF-κB signaling through modulation of TNF, TLR7, ITGB3, and TWIST, as well as regulating PPARα signaling and extracellular matrix accumulation in activated HSCs during advanced fibrogenesis in MCHSs. Of great interest, the 11ßHSD1 inhibitor J2H-1702 significantly attenuated hepatic lipid accumulation and ameliorated liver fibrosis in diet- and toxicity-induced NASH mouse models. Together, our data indicate that J2H-1702 is a promising new clinical candidate for the treatment of NASH.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases , Células Estreladas do Fígado , Cirrose Hepática , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Células de Kupffer , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/genética , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/genética
5.
J Biochem Mol Toxicol ; 36(11): e23177, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35983967

RESUMO

We focus on exploring the antihepatic fibrosis effect of Myrrhone (Myr), a compound extracted from myrrh, and its effective target. Mouse hepatic stellate cells (HSCs) were cultured in vitro and activated by transforming growth factor-ß induction. After Myr intervention, cell viability was assessed by the Cell Counting Kit-8 assay. The α-smooth muscle actin(α-SMA) and Collagen I levels were measured by immunofluorescence, and the expressions of tumor necrosis factor-α, interleukin-6, and matrix metalloproteinase-9 were examined by enzyme-linked immunosorbent assay, and the p-Smad3 protein level in HSCs was determined by Western Blot. Small molecule-protein docking and pull-down experiments were conducted to validate the binding capacity between Nard and Smad3. In animal experiments, a mouse model of hepatic fibrosis was established with carbon tetrachloride. Myr was administered by gavage daily to determine the serum alanine aminotransferase and aspartate transaminase levels. The severity of hepatic fibrosis was evaluated by Masson staining, the α-SMA and Collagen I expressions were measured by immunohistochemistry, and the histopathological changes were examined by Sirius red and hematoxylin and eosin staining. Myr suppressed the abnormal activation of HSCs, inhibited the cell viability, downregulated the α-SMA and Collagen I, and inhibited the p-Smad3 expression. After silencing Smad3, the effect of Myr was inhibited. Molecular docking and pull-down experiments revealed the presence of a targeted binding relationship between Myr and Smad3. In mouse experiments, Myr could inhibit hepatic fibrosis. This study discovers that Myr can affect the phosphorylation of Smad3, and inhibit the activation of HSCs and the progression of hepatic fibrosis.


Assuntos
Commiphora , Células Estreladas do Fígado , Cirrose Hepática , Fígado , Fitoterapia , Extratos Vegetais , Animais , Camundongos , Tetracloreto de Carbono/toxicidade , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo I/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Fígado/metabolismo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Simulação de Acoplamento Molecular , Fator de Crescimento Transformador beta1/metabolismo , Extratos Vegetais/metabolismo , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico
6.
Chem Biol Interact ; 358: 109899, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35305974

RESUMO

Increasing evidences indicate the vital role of exosomes-mediated intercellular communication in the pathogenesis of liver fibrosis. However, the underlying mechanisms are still not clearly defined. In this study, we found that citreoviridin (CIT), a mycotoxin and ectopic ATP synthase (e-ATPS) inhibitor, induced liver fibrosis in mice. The exosomes derived from CIT-treated L-02 hepatocytes activated hepatic stellate cells (HSC) LX-2. With exosomal small RNA sequencing, we found 156 differentially expressed miRNAs in the exosomes from CIT-treated L-02 cells, and the predicted target genes of exosomal miRNAs were enriched in calcium signaling pathway. The exosomes from CIT-treated L-02 cells induced mitochondrial calcium accumulation in LX-2 cells. And pharmacological inhibition of mitochondrial calcium uptake relieved exosomes-activated fibrogenic response in LX-2 cells. The miR-181a-2-3p that was predicted to target-regulate mitochondrial calcium uptake 1 (MICU1) was significantly increased in the exosomes from CIT-treated L-02 cells. Exosomes-induced reduction of MICU1, mitochondrial calcium overload and activation of LX-2 cells were reversed by AntagomiR-181a-2-3p. In this study, we pointed out that exosomal miR-181a-2-3p from CIT-treated hepatocytes induced mitochondrial calcium accumulation and activated HSC subsequently through inhibiting the expression of MICU1, shedding new light on the mechanism underlying liver fibrosis and CIT hepatotoxicity.


Assuntos
Cálcio , Células Estreladas do Fígado , Cirrose Hepática , MicroRNAs , Mitocôndrias Hepáticas , Animais , Aurovertinas/farmacologia , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Exossomos/metabolismo , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Transdução de Sinais/genética
7.
Hum Exp Toxicol ; 41: 9603271221084672, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35303413

RESUMO

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


Assuntos
Apoptose/fisiologia , Proliferação de Células/fisiologia , Células Estreladas do Fígado/fisiologia , Histona Desacetilase 2/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Células Estreladas do Fígado/efeitos dos fármacos , Histona Desacetilase 2/genética , Humanos , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-abl/genética , Transdução de Sinais , Fator de Crescimento Transformador beta1/farmacologia
8.
Eur J Pharmacol ; 919: 174770, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35120860

RESUMO

This study aimed to investigate the protective mechanisms of helenalin on hepatic fibrosis. In brief, rats were intragastrically administrated with 50% CCl4 for 9 weeks to induce liver fibrosis, followed by treatment with various agents for 6 weeks. The effects of helenalin on hepatic injury were assessed by pathological examinations. The potential targets were predicted by the "Drug-Disease" bioinformatic analysis and then verified by multiple experiments. Moreover, the underlying mechanism was investigated by transcriptomics and metabolomics as a whole. The results showed that helenalin significantly alleviated hepatocyte necrosis and hepatic injury, as proved by the pathological examinations. Also, helenalin markedly attenuated hepatocyte apoptosis by regulating the expression of caspase-3 and Bcl-2 families. Besides, helenalin could significantly reduce collagen accumulation, as evidenced by the decreased contents of collagen, hyaluronic acid and laminin. Moreover, helenalin significantly down-regulated the phosphorylation of PI3K, Akt, FAK, mTOR and P70S6K, and PTEN protein expression, suggesting that helenalin inhibited the PI3K/Akt signaling cascade. Meanwhile, helenalin inhibited the NF-κB signaling pathway by reducing the phosphorylation of IκBα, NF-κB p65 and IKKα/ß, alleviating inflammation response. Interestingly, the analysis of transcriptomics and metabolomics indicated that helenalin inhibited the glycerophospholipid metabolism pathway by down-regulating the target genes (CHKA, ETNPPL, LYPLA1, PCYT2, PLD4 and PNPLA6), ultimately ameliorating hepatocyte damage. In conclusion, helenalin ameliorates hepatic fibrosis by regulating the PI3K/Akt and NF-κB signaling pathways and the glycerophospholipid metabolism pathway.


Assuntos
Antioxidantes/farmacologia , Asteraceae , Medicamentos de Ervas Chinesas/farmacologia , Cirrose Hepática/tratamento farmacológico , Sesquiterpenos de Guaiano/farmacologia , Animais , Antioxidantes/química , Antioxidantes/uso terapêutico , Tetracloreto de Carbono , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/uso terapêutico , Células Estreladas do Fígado/efeitos dos fármacos , Humanos , Cirrose Hepática/patologia , Masculino , Metabolômica , Ratos , Ratos Sprague-Dawley , Sesquiterpenos de Guaiano/química , Sesquiterpenos de Guaiano/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transcriptoma
9.
Sci Rep ; 12(1): 2453, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35165344

RESUMO

Inflammation plays a critical role in the promotion of hepatocyte damage and liver fibrosis. In recent years the protective role of Akkermansia muciniphila, a next-generation beneficial microbe, has been suggested for metabolic and inflammatory disorders. In this study, we aimed to evaluate the effects of live and pasteurized A. muciniphila and its extra cellular vesicles (EVs) on inflammatory markers involved in liver fibrosis in a mouse model of a high-fat diet (HFD)/carbon tetrachloride (CCl4)-induced liver injury. Firstly, the responses of hepatic stellate cells (HSCs) to live and pasteurized A. muciniphila and its EVs were examined in the quiescent and LPS-activated LX-2 cells. Next, the anti-inflammatory effects of different forms of A. muciniphila were examined in the mouse model of HFD/CCl4-induced liver injury. The gene expression of various inflammatory markers was evaluated in liver, colon, and white adipose tissues. The cytokine secretion in the liver and white adipose tissues was also measured by ELISA. The results showed that administration of live and pasteurized A. muciniphila and its EVs leads to amelioration in HSCs activation. Based on data obtained from the histopathological analysis, an improvement in gut health was observed through enhancing the epithelium and mucosal layer thickness and strengthening the intestinal integrity in all treatments. Moreover, live A. muciniphila and its EVs had inhibitory effects on liver inflammation and hepatocytes damage. In addition, the tissue cytokine production and inflammatory gene expression levels revealed that live A. muciniphila and its EVs had more pronounced anti-inflammatory effects on liver and adipose tissues. Furthermore, EVs had better effects on the modulation of gene expression related to TLRs, PPARs, and immune response in the liver. In conclusion, the present results showed that oral administration of A. muciniphila and its derivatives for four weeks could enhance the intestinal integrity and anti-inflammatory responses of the colon, adipose, and liver tissues and subsequently prevent liver injury in HFD/CCL4 mice.


Assuntos
Anti-Inflamatórios/administração & dosagem , Tetracloreto de Carbono/efeitos adversos , Dieta Hiperlipídica/efeitos adversos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/prevenção & controle , Probióticos/administração & dosagem , Tecido Adiposo/metabolismo , Administração Oral , Akkermansia/citologia , Animais , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Vesículas Extracelulares , Microbioma Gastrointestinal , Expressão Gênica , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/microbiologia , Humanos , Fígado/metabolismo , Cirrose Hepática/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Biochem Biophys Res Commun ; 600: 136-141, 2022 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-35219102

RESUMO

Liver fibrosis is a major consequence of chronic liver disease, where excess extracellular matrix is deposited, due caused by the activation of hepatic stellate cells (HSCs). The suppression of collagen production in HSCs is therefore regarded as a therapeutic target of liver fibrosis. The present study investigated effects of harmine, which is a ß-carboline alkaloid and known as an inhibitor of dual-specificity tyrosine-regulated kinases (DYRKs), on the production of collagen in HSCs. LX-2 cells, a human HSC cell line, were treated with harmine (0-10 µM) for 48 h in the presence or absence of TGF-ß1 (5 ng/ml). The expression of collagen type I α1 (COL1A1) and DYRK isoforms was investigated by Western blotting, quantitative RT-PCR, or immunofluorescence. The influence of knockdown of each DYRK isoform on the COL1A1 expression was further investigated. The expression of COL1A1 was markedly increased by treating with TGF-ß1 for 48 h in LX-2 cells. Harmine (10 µM) significantly inhibited the increased expression of COL1A1. LX-2 cells expressed mRNAs of DYRK1A, DYRK1B, DYRK2, and DYRK4, although the expression of DYRK4 was much lower than the others. Knockdown of DYRK1B, but not DYRK1A or DYRK2, with siRNA significantly suppressed TGF-ß1-induced increase in COL1A1 expression. These results suggest that harmine suppresses COL1A1 expression via inhibiting DYRK1B in HSCs and therefore might be effective for the treatment of liver fibrosis.


Assuntos
Cadeia alfa 1 do Colágeno Tipo I , Harmina , Células Estreladas do Fígado , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases , Fator de Crescimento Transformador beta1 , Cadeia alfa 1 do Colágeno Tipo I/antagonistas & inibidores , Cadeia alfa 1 do Colágeno Tipo I/biossíntese , Harmina/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Humanos , Cirrose Hepática/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
12.
J Nanobiotechnology ; 20(1): 9, 2022 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-34983554

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) developed in fibrotic liver does not respond well to immunotherapy, mainly due to the stromal microenvironment and the fibrosis-related immunosuppressive factors. The characteristic of liver sinusoidal endothelial cells (LSECs) in contributing to fibrosis and orchestrating immune response is responsible for the refractory to targeted therapy or immunotherapy of HCC. We aim to seek a new strategy for HCC treatment based on an old drug simvastatin which shows protecting effect on LSEC. METHOD: The features of LSECs in mouse fibrotic HCC model and human HCC patients were identified by immunofluorescence and scanning electron microscopy. The effect of simvastatin on LSECs and hepatic stellate cells (HSCs) was examined by immunoblotting, quantitative RT-PCR and RNA-seq. LSEC-targeted delivery of simvastatin was designed using nanotechnology. The anti-HCC effect and toxicity of the nano-drug was evaluated in both intra-hepatic and hemi-splenic inoculated mouse fibrotic HCC model. RESULTS: LSEC capillarization is associated with fibrotic HCC progression and poor survival in both murine HCC model and HCC patients. We further found simvastatin restores the quiescence of activated hepatic stellate cells (aHSCs) via stimulation of KLF2-NO signaling in LSECs, and up-regulates the expression of CXCL16 in LSECs. In intrahepatic inoculated fibrotic HCC mouse model, LSEC-targeted nano-delivery of simvastatin not only alleviates LSEC capillarization to regress the stromal microenvironment, but also recruits natural killer T (NKT) cells through CXCL16 to suppress tumor progression. Together with anti-programmed death-1-ligand-1 (anti-PD-L1) antibody, targeted-delivery of simvastatin achieves an improved therapeutic effect in hemi-splenic inoculated advanced-stage HCC model. CONCLUSIONS: These findings reveal an immune-based therapeutic mechanism of simvastatin for remodeling immunosuppressive tumor microenvironment, therefore providing a novel strategy in treating HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Sistemas de Liberação de Fármacos por Nanopartículas , Sinvastatina/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Estreladas do Fígado/efeitos dos fármacos , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química , Transdução de Sinais/efeitos dos fármacos
13.
Exp Cell Res ; 411(2): 113001, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34973945

RESUMO

Autophagy is involved in the activation of hepatic stellate cells (HSCs) and liver fibrosis. Previous studies have shown that interleukin 10 (IL-10) has a marked therapeutic effect against liver fibrosis. However, few studies have evaluated the effect of IL-10 on autophagy in HSCs and fibrotic livers. The aim of this study was to assess the effect of IL-10 on the autophagy of HSCs in vitro and in vivo and then to explore the underlying pathway. In vitro, The results revealed that IL-10 had inhibitory effects on hydrogen peroxide (H2O2)-induced autophagy, as evidenced by the decreased LC3II/I ratio and Beclin1 expression, increased p62 expression, reduced numbers of autophagosomes, and blocked autophagy initiation in HSCs. Mechanistically, IL-10 significantly promoted the phosphorylation of the signal transducer and activator of transcription 3(STAT3) and mammalian target of rapamycin (mTOR), leading to the activation of STAT3 and mTOR, which in turn inhibited autophagy. In vivo, the increased expression of IL-10 in fibrotic livers inhibited significantly liver fibrosis and decreased the autophagic activity in fibrotic livers and HSCs. Overall, our results indicate that IL-10 suppressed H2O2-induced autophagy in HSCs by activating the STAT3-mTOR signaling pathway. Present study provides a new theoretical basis for the anti-fibrotic effects of IL-10.


Assuntos
Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Interleucina-10/metabolismo , Interleucina-10/farmacologia , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/patologia , Autofagia/efeitos dos fármacos , Linhagem Celular , Células Estreladas do Fígado/patologia , Humanos , Peróxido de Hidrogênio/farmacologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/prevenção & controle , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
14.
Drug Deliv ; 29(1): 440-453, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35099348

RESUMO

Liver fibrosis is a common pathologic stage of the development of liver failure. It has showed that exosomes loaded with therapeutic circRNAs can be manufactured in bulk by exosome secreted cells in vitro, thus enabling personalized treatment. This study aimed to investigate the role of exosome-based delivery of circDIDO1 in liver fibrosis. Levels of genes and proteins were examined by qRT-PCR and Western blot. Cell proliferation, apoptosis, and cell cycle were analyzed by using cell counting kit-8 (CCK-8) assay, EdU assay, and flow cytometry, respectively. The binding between circDIDO1 and miR-141-3p was confirmed by dual-luciferase reporter, RNA pull-down and RIP assays. Exosomes were isolated by ultracentrifugation, and qualified by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and Western blot. CircDIDO1 overexpression or miR-141-3p inhibition suppressed the proliferation, reduced pro-fibrotic markers, and induced apoptosis as well as cell cycle arrest in hepatic stellate cells (HSCs) by blocking PTEN/AKT pathway. Mechanistically, circDIDO1 acted as an endogenous sponge for miR-141-3p, further rescue experiments showed that circDIDO1 suppressed HSC activation by targeting miR-141-3p. Extracellular circDIDO1 could be incorporated into exosomes isolated from mesenchymal stem cells (MSCs), and transmitted to HSCs to restrain HSC activation. Clinically, low levels of serum circDIDO1 in exosome were correlated with liver failure, and serum exosomal circDIDO1 had a well diagnostic value for liver fibrosis in liver failure patients. Transfer of circDIDO1 mediated by MSC-isolated exosomes suppressed HSC activation through the miR-141-3p/PTEN/AKT pathway, gaining a new insight into the prevention of liver fibrosis in liver failure patients.


Assuntos
Proteínas de Ligação a DNA/farmacologia , Cirrose Hepática/patologia , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/efeitos dos fármacos , PTEN Fosfo-Hidrolase/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Exossomos/metabolismo , Células Estreladas do Fígado/efeitos dos fármacos , Humanos , RNA Circular , Transdução de Sinais
15.
PLoS One ; 17(1): e0262767, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35051234

RESUMO

Hepatic fibrosis is caused by the increased accumulation and improper degradation of extracellular matrix (ECM) proteins in the liver. Hepatic stellate cells (HSCs) activation is a key process in initiating hepatic fibrosis and can be ameliorated by the administration of probiotic strains. This study hypothesized that LAB strains (Lactiplantibacillus plantarum, Lactobacillus brevis, and Weissella cibaria) might attenuate pro-fibrogenic cytokine TGF-ß mediated HSCs activation and induce collagen deposition, expression of other fibrogenic/inflammatory markers, autophagy, and apoptotic processes in vitro. Few studies have evaluated the probiotic effects against fibrogenesis in vitro. In this study, TGF-ß exposure increased collagen deposition in LX-2 cells, but this increase was diminished when the cells were pretreated with LAB strains before TGF-ß stimulation. TGF-ß not only increased collagen deposition, but it also significantly upregulated the mRNA levels of Col1A1, alpha-smooth muscle actin (α-SMA), matrix metalloproteinases-2 (MMP-2), IL-6, CXCL-8, CCL2, and IL-1ß in LX-2 cells. Pretreatment of the cells with LAB strains counteracted the TGF-ß-induced pro-fibrogenic and inflammatory markers by modulating SMAD-dependent and SMAD-independent TGF-ß signaling. In addition, LX-2 cells exposed to TGF-ß induced the autophagic and apoptotic associated proteins that were also positively regulated by the LAB strains. These findings suggest that LAB can attenuate TGF-ß signaling that is associated with liver fibrogenesis.


Assuntos
Autofagia/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Células Estreladas do Fígado/efeitos dos fármacos , Cirrose Hepática/genética , Fígado/efeitos dos fármacos , Probióticos/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular , Colágeno/metabolismo , Células Estreladas do Fígado/metabolismo , Humanos , Fígado/metabolismo , Cirrose Hepática/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo
16.
Toxicol In Vitro ; 80: 105315, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35051607

RESUMO

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


Assuntos
Estresse do Retículo Endoplasmático , Proteínas Hedgehog/metabolismo , Células Estreladas do Fígado/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular , Movimento Celular , Colágeno/metabolismo , Expressão Gênica , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/fisiologia , Humanos , Quercetina/farmacologia , Transdução de Sinais
17.
Toxicol Appl Pharmacol ; 437: 115891, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35077758

RESUMO

Liver fibrosis has the potential to progress into liver cirrhosis, liver failure, and even death. Hepatic stellate cells (HSCs) activation play a central role in liver fibrosis, and persistently damaged hepatocytes secrete soluble factors that activate transdifferentiation of HSCs into myofibroblasts. Our previous studies indicated that fine particulate matter (PM2.5) can activate HSCs by stimulating hepatocytes to secrete TGF-ß1. However, whether PM2.5 activates HSCs by regulating oxidative stress in hepatocytes remains uncertain. Oleanolic acid (OA) has been widely used in the clinic for hepatoprotection in Chinese medicine. In the present study, OA-loaded nanoparticles (OA-NP) with high solubility were used to attenuate the activation of HSCs induced by PM2.5-treated hepatocytes, and further studies were performed to explore the mechanism in which OA-NP plays a vital part. Our results showed that consistently PM2.5 treatment induced oxidative stress in hepatocytes. Moreover, the activation of HSCs induced by PM2.5-treated hepatocytes was reversed by antioxidant N-acetylcysteine treatment. Hence, PM2.5 may participate in the activation of HSCs by regulating oxidative stress in hepatocytes. Using a co-cultivation system, our results proved pretreatment with OA-NP significantly attenuates the activation of HSCs induced by PM2.5-exposed hepatocytes. In addition, the TGF-ß1 expression and oxidative stress in hepatocytes with PM2.5 treated were reduced by the incubation with OA-NP. These observations demonstrated that OA-NP protects against the activation of HSCs by decreasing the TGF-ß1 level and oxidative stress in PM2.5-exposed hepatocytes.


Assuntos
Células Estreladas do Fígado/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Nanopartículas/química , Ácido Oleanólico/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Material Particulado/toxicidade , Fator de Crescimento Transformador beta1/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/metabolismo , Ácido Oleanólico/química , Tamanho da Partícula , Fator de Crescimento Transformador beta1/genética
18.
Comput Math Methods Med ; 2022: 8920861, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35047060

RESUMO

BACKGROUND: Hepatic stellate cells (HSCs) are reported to play significant roles in the development of liver fibrosis. Heme oxygenase-1 (HO-1) is a key rate-limiting enzyme, which could decrease collagen synthesis and liver damage. Nevertheless, it was yet elusive towards the function and mechanism of HO-1. METHODS: An HO-1 inducer Hemin or an HO-1 inhibitor ZnPP-IX was used to treat the activated HSC-T6, respectively. MTT assay was adopted to detect cell proliferation. Immunocytochemical staining was employed to test the levels of alpha-smooth muscle actin (α-SMA), peroxisome proliferator-activated receptor-γ (PPARγ), and nuclear factor-kappa B (NF-kappa B) levels in HSC-T6. HO-1, PPARγ, and NF-κB expression levels were measured by qRT-PCR and Western blotting. ELISA was then used to detect the levels of transforming growth factor- (TGF-) beta 1 (TGF-ß1), interleukin-6 (IL-6), serum hyaluronic acid (HA), and serum type III procollagen aminopeptide (PIIIP). RESULTS: HSC-T6 proliferation was inhibited in Hemin-treated HSCs. The levels of α-SMA, HA, and PIIIP and the production of ECM were lower in Hemin-treated HSCs, whereas those could be rescued by ZnPP-IX. NF-κB activation was decreased, but PPARγ expression was increased after HO-1 upregulation. Furthermore, the levels of TGF-ß1 and IL-6, which were downstream of activated NF-κB in HSC-T6, were reduced. The PPAR-specific inhibitor GW9662 could block those mentioned effects. CONCLUSIONS: Our data demonstrated that HO-1 induction could inhibit HSC proliferation and activation by regulating PPARγ expression and NF-κB activation directly or indirectly, which makes it a promising therapeutic target for liver fibrosis.


Assuntos
Heme Oxigenase (Desciclizante)/metabolismo , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/metabolismo , NF-kappa B/metabolismo , PPAR gama/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Biologia Computacional , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Heme Oxigenase (Desciclizante)/biossíntese , Hemina/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Humanos , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Modelos Biológicos , NF-kappa B/antagonistas & inibidores , PPAR gama/agonistas , PPAR gama/genética , Protoporfirinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos
19.
Mech Ageing Dev ; 201: 111617, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34958827

RESUMO

BACKGROUND: Activated hepatic stellate cells (aHSCs) are the main effector cells during liver fibrogenesis. α-1 adrenergic antagonist doxazosin (DX) was shown to be anti-fibrotic in an in vivo model of liver fibrosis (LF), but the mechanism remains to be elucidated. Recent studies suggest that reversion of LF can be achieved by inducing cellular senescence characterized by irreversible cell-cycle arrest and acquisition of the senescence-associated secretory phenotype (SASP). AIM: To elucidate the mechanism of the anti-fibrotic effect of DX and determine whether it induces senescence. METHODS: Primary culture-activated rat HSCs were used. mRNA and protein expression were measured by qPCR and Western blot, respectively. Cell proliferation was assessed by BrdU incorporation and xCelligence analysis. TGF-ß was used for maximal HSC activation. Norepinephrine (NE), PMA and m-3M3FBS were used to activate alpha-1 adrenergic signaling. RESULTS: Expression of Col1α1 was significantly decreased by DX (10 µmol/L) at mRNA (-30 %) and protein level (-50 %) in TGF-ß treated aHSCs. DX significantly reduced aHSCs proliferation and increased expression of senescence and SASP markers. PMA and m-3M3FBS reversed the effect of DX on senescence markers. CONCLUSION: Doxazosin reverses the fibrogenic phenotype of aHSCs and induces the senescence phenotype.


Assuntos
Senescência Celular , Doxazossina/farmacologia , Células Estreladas do Fígado , Cirrose Hepática , Fenótipo Secretor Associado à Senescência/fisiologia , Transdução de Sinais/efeitos dos fármacos , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/prevenção & controle , Norepinefrina/farmacologia , Ratos , Receptores Adrenérgicos alfa 1/metabolismo , Sulfonamidas/farmacologia
20.
Cell Prolif ; 55(1): e13158, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34811833

RESUMO

OBJECTIVES: Evidences demonstrate that sorafenib alleviates liver fibrosis via inhibiting HSC activation and ECM accumulation. The underlying mechanism remains unclear. Ferroptosis, a novel programmed cell death, regulates diverse physiological/pathological processes. In this study, we aim to investigate the functional role of HSC ferroptosis in the anti-fibrotic effect of sorafenib. MATERIALS AND METHODS: The effects of sorafenib on HSC ferroptosis and ECM expression were assessed in mouse model of liver fibrosis induced by CCl4 . In vitro, Fer-1 and DFO were used to block ferroptosis and then explored the anti-fibrotic effect of sorafenib by detecting α-SMA, COL1α1 and fibronectin proteins. Finally, HIF-1α siRNA, plasmid and stabilizers were applied to assess related signalling pathway. RESULTS: Sorafenib attenuated liver injury and ECM accumulation in CCl4 -induced fibrotic livers, accompanied by reduction of SLC7A11 and GPX4 proteins. In sorafenib-treated HSC-T6 cells, ferroptotic events (depletion of SLC7A11, GPX4 and GSH; accumulation iron, ROS and MDA) were discovered. Intriguingly, these ferroptotic events were not appeared in hepatocytes or macrophages. Sorafenib-elicited HSC ferroptosis and ECM reduction were abrogated by Fer-1 and DFO. Additionally, both HIF-1α and SLC7A11 proteins were reduced in sorafenib-treated HSC-T6 cells. SLC7A11 was positively regulated by HIF-1α, inactivation of HIF-1α/SLC7A11 pathway was required for sorafenib-induced HSC ferroptosis, and elevation of HIF-1α could inhibit ferroptosis, ultimately limited the anti-fibrotic effect. CONCLUSIONS: Sorafenib triggers HSC ferroptosis via HIF-1α/SLC7A11 signalling, which in turn attenuates liver injury and fibrosis.


Assuntos
Sistema y+ de Transporte de Aminoácidos/metabolismo , Ferroptose , Células Estreladas do Fígado/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Cirrose Hepática/tratamento farmacológico , Transdução de Sinais , Sorafenibe/uso terapêutico , Actinas/metabolismo , Animais , Linhagem Celular , Colágeno Tipo I/metabolismo , Ferroptose/efeitos dos fármacos , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Cirrose Hepática/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Estabilidade Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sorafenibe/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...