Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Pharmacol Res ; 208: 107409, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39284429

RESUMO

The pathogenesis of liver diseases is multifaceted and intricate, posing a persistent global public health challenge with limited therapeutic options. Therefore, further research into liver diseases is imperative for better comprehension and advancement in treatment strategies. Numerous studies have confirmed the endoplasmic reticulum (ER) and mitochondria as key organelles driving liver diseases. Notably, the mitochondrial-associated ER membranes (MAMs) establish a physical and functional connection between the ER and mitochondria, highlighting the importance of inter-organelle communication in maintaining their functional homeostasis. This review delves into the intricate architecture and regulative mechanism of the integrated MAM that facilitate the physiological transfer of signals and substances between organelles. Additionally, we also provide a detailed overview regarding the varied pathogenic roles of malfunctioning MAM in liver diseases, focusing on its involvement in the progression of ER stress and mitochondrial dysfunction, the regulation of mitochondrial dynamics and Ca2+ transfer, as well as the disruption of lipid and glucose homeostasis. Furthermore, the current challenges and prospects associated with MAM in liver disease research are thoroughly discussed. In conclusion, elucidating the specific structure and function of MAM in different liver diseases may pave the way for novel therapeutic strategies.


Assuntos
Retículo Endoplasmático , Hepatopatias , Humanos , Retículo Endoplasmático/metabolismo , Hepatopatias/metabolismo , Hepatopatias/patologia , Animais , Mitocôndrias/metabolismo , Estresse do Retículo Endoplasmático , Membranas Intracelulares/metabolismo
2.
Acta Pharmacol Sin ; 45(6): 1115-1129, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38233527

RESUMO

Numerous liver diseases, such as nonalcoholic fatty liver disease, hepatitis, hepatocellular carcinoma, and hepatic ischemia-reperfusion injury, have been increasingly prevalent, posing significant threats to global health. In recent decades, there has been increasing evidence linking the dysregulation of cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING)-related immune signaling to liver disorders. Both hyperactivation and deletion of STING can disrupt the immune microenvironment dysfunction, exacerbating liver disorders. Consequently, there has been a surge in research investigating medical agents or mediators targeting cGAS-STING signaling. Interestingly, therapeutic manipulation of the cGAS-STING pathway has yielded inconsistent and even contradictory effects on different liver diseases due to the distinct physiological characteristics of intrahepatic cells that express and respond to STING. In this review, we comprehensively summarize recent advancements in understanding the dual roles of the STING pathway, highlighting that the benefits of targeting STING signaling depend on the specific types of target cells and stages of liver injury. Additionally, we offer a novel perspective on the suitability of STING agonists and antagonists for clinical assessment. In conclusion, STING signaling remains a highly promising therapeutic target, and the development of STING pathway modulators holds great potential for the treatment of liver diseases.


Assuntos
Hepatopatias , Proteínas de Membrana , Nucleotidiltransferases , Transdução de Sinais , Humanos , Nucleotidiltransferases/metabolismo , Proteínas de Membrana/metabolismo , Hepatopatias/metabolismo , Hepatopatias/imunologia , Animais
3.
Phytother Res ; 38(2): 620-635, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37953063

RESUMO

Idiopathic pulmonary fibrosis (IPF), as the most common idiopathic interstitial pneumonia, is caused by a complex interaction of pathological mechanisms. Interestingly, IPF frequently occurs in the middle-aged and elderly populations but rarely affects young people. Salvianolic acid B (SAB) exerts antioxidant, antiinflammatory, and antifibrotic bioactivities and is considered a promising drug for pulmonary disease treatment. However, the pharmacological effects and mechanisms of SAB on cellular senescence of lung cells and IPF development remain unclear. We used bleomycin (BLM)-induced pulmonary fibrosis mice and different lung cells to investigate the antisenescence impact of SAB and explain its underlying mechanism by network pharmacology and the Human Protein Atlas database. Here, we found that SAB significantly prevented pulmonary fibrosis and cellular senescence in mice, and reversed the senescence trend and typical senescence-associated secretory phenotype (SASP) factors released from lung macrophages and alveolar type II (AT2) epithelial cells, which further reduced lung fibroblasts activation. Additionally, SAB alleviated the epithelial-mesenchymal transition process of AT2 cells induced by transforming growth factor beta. By predicting potential targets of SAB that were then confirmed by chromatin immunoprecipitation-qPCR technology, we determined that SAB directly hampered the binding of transcription factor stimulating protein 1 to the promoters of SASPs (P21 and P16), thus halting lung cell senescence. We demonstrated that SAB reduced BLM-induced AT2 and macrophage senescence, and the subsequent release of SASP factors that activated lung fibroblasts, thereby dual-relieving IPF. This study provides a new scientific foundation and perspective for pulmonary fibrosis therapy.


Assuntos
Benzofuranos , Depsídeos , Fibrose Pulmonar Idiopática , Pulmão , Pessoa de Meia-Idade , Idoso , Humanos , Camundongos , Animais , Adolescente , Pulmão/patologia , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/genética , Senescência Celular/fisiologia , Macrófagos Alveolares , Bleomicina/efeitos adversos
4.
Acta Pharmacol Sin ; 44(12): 2479-2491, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37580495

RESUMO

Liver fibrosis is a wound-healing process characterized by excess formation of extracellular matrix (ECM) from activated hepatic stellate cells (HSCs). Previous studies show that both EZH2, an epigenetic regulator that catalyzes lysine 27 trimethylation on histone 3 (H3K27me3), and long non-coding RNA H19 are highly correlated with fibrogenesis. In the current study, we investigated the underlying mechanisms. Various models of liver fibrosis including Mdr2-/-, bile duct ligation (BDL) and CCl4 mice were adapted. We found that EZH2 was markedly upregulated and correlated with H19 and fibrotic markers expression in these models. Administration of EZH2 inhibitor 3-DZNeP caused significant protective effects in these models. Furthermore, treatment with 3-DZNeP or GSK126 significantly inhibited primary HSC activation and proliferation in TGF-ß-treated HSCs and H19-overexpreesing LX2 cells in vivo. Using RNA-pull down assay combined with RNA immunoprecipitation, we demonstrated that H19 could directly bind to EZH2. Integrated analysis of RNA-sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) further revealed that H19 regulated the reprogramming of EZH2-mediated H3K27me3 profiles, which epigenetically promoted several pathways favoring HSCs activation and proliferation, including epithelial-mesenchymal transition and Wnt/ß-catenin signaling. In conclusion, highly expressed H19 in chronic liver diseases promotes fibrogenesis by reprogramming EZH2-mediated epigenetic regulation of HSCs activation. Targeting the H19-EZH2 interaction may serve as a novel therapeutic approach for liver fibrosis.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste , Histonas , Cirrose Hepática , RNA Longo não Codificante , Animais , Camundongos , Epigênese Genética , Células Estreladas do Fígado/metabolismo , Histonas/metabolismo , Fígado/metabolismo , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Lisina/química , Lisina/metabolismo , Metilação , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo
5.
Acta Pharmacol Sin ; 44(9): 1826-1840, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37095199

RESUMO

Obesity contributes to the progression of various chronic diseases, and shortens life expectancy. With abundant mitochondria, brown adipose tissue (BAT) dissipates energy through heat to limit weight gain and metabolic dysfunction in obesity. Our previous studies have shown that aurantio-obtusin (AO), a bioactive ingredient in Chinese traditional medicine Cassiae semen significantly improves hepatic lipid metabolism in a steatotic mouse model. In the current study we investigated the effects of AO on lipid metabolism in the BAT of diet-induced obesity mice and in oleic acid and palmitic acid (OAPA)-stimulated primary mature BAT adipocytes. Obese mice were established by feeding a HFHS diet for 4 weeks, and then administered AO (10 mg/kg, i.g.) for another 4 weeks. We showed that AO administration significantly increased the weight of BAT and accelerated energy expenditure to protect the weight increase in the obese mice. Using RNA sequencing and molecular biology analysis we found that AO significantly enhanced mitochondrial metabolism and UCP1 expression by activating PPARα both in vivo and in vitro in the primary BAT adipocytes. Interestingly, AO administration did not improve metabolic dysfunction in the liver and white adipose tissue of obese mice after interscapular BAT excision. We demonstrated that low temperature, a trigger of BAT thermogenesis, was not a decisive factor for AO to stimulate the growth and activation of BATs. This study uncovers a regulatory network of AO in activating BAT-dependent lipid consumption and brings up a new avenue for the pharmaceutical intervention in obesity and related comorbidities.


Assuntos
Tecido Adiposo Marrom , PPAR alfa , Camundongos , Animais , Tecido Adiposo Marrom/metabolismo , PPAR alfa/metabolismo , Camundongos Obesos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Mitocôndrias/metabolismo , Metabolismo Energético , Tecido Adiposo Branco/metabolismo , Termogênese , Camundongos Endogâmicos C57BL
6.
IUBMB Life ; 74(9): 880-895, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35514074

RESUMO

Acetaminophen (APAP), one of the most widely used antipyretics and analgesics, principally results in acute liver injury (ALI) in developed countries when taken overdose. Ferulic acid (FA) is a natural polyphenol compound existing in many plants that has free radical scavenging, anti-inflammatory, and liver-protective properties. However, the effect and underlying mechanism of FA in treating APAP-induced ALI have not been fully elucidated. Herein, we established a mouse model of APAP-induced ALI and used APAP-stimulated MPHs for biochemical assessment of molecular parameters. After constructing networks and obtaining predicted targets from public databases, we further verified the putative pathways using immune-blotting assays both in vivo and in vitro. The reign of liver necrosis, serum levels of ALT and AST, and oxidative stress in livers significantly elevated after APAP treatment, which were almost recovered back to normal levels by FA administration. In addition, FA significantly upregulated the APAP-induced downregulation of hepatic specific markers, including HNF4a, Foxa2, and ALB. Then, the results of functional enrichment indicated the possible signaling pathways of FA against APAP challenge, mainly including AMPK, autophagy, apoptosis, and other metabolic process. Furthermore, FA markedly reversed the APAP-induced decline of mitochondria membrane potential, increased ratio of BAX/BCL2 and CASPASE 3 expression, and promoted autophagy flux of hepatocytes by upregulating AMPK phosphorylation, which were abrogated by a specific AMPK inhibitor, compound C. Overall, the hepatoprotective effect of FA on APAP-induced ALI might be associated with anti-oxidant and anti-apoptosis, which were at least partly attributed to AMPK-mediated protective autophagy.


Assuntos
Acetaminofen , Doença Hepática Induzida por Substâncias e Drogas , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Acetaminofen/metabolismo , Acetaminofen/toxicidade , Animais , Autofagia/fisiologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Ácidos Cumáricos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo
7.
Acta Pharmacol Sin ; 43(8): 2026-2041, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35027662

RESUMO

Liver fibrosis is the common consequence of almost all liver diseases and has become an urgent clinical problem without efficient therapies. Recent evidence has shown that hepatocytes-derived extracellular vesicles (EVs) play important roles in liver pathophysiology, but little is known about the role of damaged hepatocytes-derived EVs in hepatic stellate cell (HSC) activation and following fibrosis. Tetramethylpyrazine (TMP) from Ligusticum wallichii Franchat exhibits a broad spectrum of biological activities including liver protection. In this study, we investigated whether TMP exerted liver-protective action through regulating EV-dependent intercellular communication between hepatocytes and HSCs. Chronic liver injury was induced in mice by CCl4 (1.6 mg/kg, i.g.) twice a week for 8 weeks. In the last 4 weeks of CCl4 administration, mice were given TMP (40, 80, 160 mg·kg-1·d-1, i.g.). Acute liver injury was induced in mice by injection of a single dose of CCl4 (0.8 mg/kg, i.p.). After injection, mice were treated with TMP (80 mg/kg) every 24 h. We showed that TMP treatment dramatically ameliorated CCl4-induced oxidative stress and hepatic inflammation as well as acute or chronic liver fibrosis. In cultured mouse primary hepatocytes (MPHs), treatment with CCl4 or acetaminophen resulted in mitochondrial dysfunction, release of mitochondrial DNA (mtDNA) from injured hepatocytes to adjacent hepatocytes and HSCs through EVs, mediating hepatocyte damage and fibrogenic responses in activated HSCs; pretreatment of MPHs with TMP (25 µM) prevented all these pathological effects. Transplanted serum EVs from TMP-treated mice prevented both initiation and progression of liver fibrosis caused by CCl4. Taken together, this study unravels the complex mechanisms underlying the protective effects of TMP against mtDNA-containing EV-mediated hepatocyte injury and HSC activation during liver injury, and provides critical evidence inspiring the development of TMP-based innovative therapeutic agents for the treatment of liver fibrosis.


Assuntos
Vesículas Extracelulares , Hepatopatias , Animais , Tetracloreto de Carbono/efeitos adversos , Tetracloreto de Carbono/metabolismo , DNA Mitocondrial/metabolismo , DNA Mitocondrial/farmacologia , DNA Mitocondrial/uso terapêutico , Fibrose , Células Estreladas do Fígado , Hepatócitos , Fígado/metabolismo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/prevenção & controle , Hepatopatias/metabolismo , Camundongos , Mitocôndrias/patologia , Pirazinas
8.
Hepatology ; 71(2): 611-626, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31220352

RESUMO

Cirrhosis and hepatic encephalopathy (HE) is associated with an altered gut-liver-brain axis. Fecal microbial transplant (FMT) after antibiotics improves outcomes in HE, but the impact on brain function is unclear. The aim of this study is to determine the effect of colonization using human donors in germ-free (GF) mice on the gut-liver-brain axis. GF and conventional mice were made cirrhotic using carbon tetrachloride and compared with controls in GF and conventional state. Additional GF mice were colonized with stool from controls (Ctrl-Hum) and patients with cirrhosis (Cirr-Hum). Stools from patients with HE cirrhosis after antibiotics were pooled (pre-FMT). Stools from the same patients 15 days after FMT from a healthy donor were also pooled (post-FMT). Sterile supernatants were created from pre-FMT and post-FMT samples. GF mice were colonized using stools/sterile supernatants. For all mice, frontal cortex, liver, and small/large intestines were collected. Cortical inflammation, synaptic plasticity and gamma-aminobutyric acid (GABA) signaling, and liver inflammation and intestinal 16s ribosomal RNA microbiota sequencing were performed. Conventional cirrhotic mice had higher degrees of neuroinflammation, microglial/glial activation, GABA signaling, and intestinal dysbiosis compared with other groups. Cirr-Hum mice had greater neuroinflammation, microglial/glial activation, and GABA signaling and lower synaptic plasticity compared with Ctrl-Hum mice. This was associated with greater dysbiosis but no change in liver histology. Pre-FMT material colonization was associated with neuroinflammation and microglial activation and dysbiosis, which was reduced significantly with post-FMT samples. Sterile pre-FMT and post-FMT supernatants did not affect brain parameters. Liver inflammation was unaffected. Conclusion: Fecal microbial colonization from patients with cirrhosis results in higher degrees of neuroinflammation and activation of GABAergic and neuronal activation in mice regardless of cirrhosis compared with those from healthy humans. Reduction in neuroinflammation by using samples from post-FMT patients to colonize GF mice shows a direct effect of fecal microbiota independent of active liver inflammation or injury.


Assuntos
Córtex Cerebral , Disbiose/complicações , Encefalite/microbiologia , Encefalite/terapia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiologia , Cirrose Hepática/microbiologia , Cirrose Hepática/terapia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
FEMS Yeast Res ; 21(2)2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33606011

RESUMO

SLX4 is a scaffold to coordinate the action of structure-specific endonucleases that are required for homologous recombination and DNA repair. In view of ScSLX4 functions in the maintenance and stability of the genome in Saccharomyces cerevisiae, we have explored the roles of CaSLX4 in Candida albicans. Here, we constructed slx4Δ/Δ mutant and found that it exhibited increased sensitivity to the DNA damaging agent, methyl methanesulfonate (MMS) but not the DNA replication inhibitor, hydroxyurea (HU). Accordingly, RT-qPCR and western blotting analysis revealed the activation of SLX4 expression in response to MMS. The deletion of SLX4 resulted in a defect in the recovery from MMS-induced filamentation to yeast form and re-entry into the cell cycle. Like many other DNA repair genes, SLX4 expression was activated by the checkpoint kinase Rad53 under MMS-induced DNA damage. In addition, SLX4 was not required for the inactivation of the DNA damage checkpoint, as indicated by normal phosphorylation of Rad53 in slx4Δ/Δ cells. Therefore, our results demonstrate SLX4 plays an important role in cell recovery from MMS-induced DNA damage in C. albicans.


Assuntos
Candida albicans/efeitos dos fármacos , Candida albicans/genética , Dano ao DNA/efeitos dos fármacos , Endodesoxirribonucleases/genética , Proteínas Fúngicas/genética , Metanossulfonato de Metila/farmacologia , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Endodesoxirribonucleases/metabolismo , Proteínas Fúngicas/metabolismo , Hidroxiureia/farmacologia , Fosforilação , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
10.
Hepatology ; 70(5): 1658-1673, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31063660

RESUMO

Biliary atresia (BA) is a neonatal liver disease featuring cholestasis and severe liver fibrosis (LF). Despite advances in the development of surgical treatment, lacking an early diagnostic marker and intervention of LF invariably leads to death from end-stage liver disease in the early years of life. We previously reported that knockout of sphingosine 1-phosphate receptor 2 (S1PR2) protected mice from bile duct ligation (BDL)-induced cholangiocyte proliferation and LF. Our recent studies further showed that both hepatic and serum exosomal long noncoding RNA H19 (lncRNAH19) levels are correlated with cholestatic injury in multidrug resistance 2 knockout (Mdr2-/- ) mice. However, the role of lncRNAH19 in BA progression remains unclear. Here, we show that both hepatic and serum exosomal H19 levels are positively correlated with severity of fibrotic liver injuries in BA patients. H19 deficiency protects mice from BDL-induced cholangiocyte proliferation and LF by inhibiting bile-acid-induced expression and activation of S1PR2 and sphingosine kinase 2 (SphK2). Furthermore, H19 acts as a molecular sponge for members of the microRNA let-7 family, which results in up-regulation of high-mobility group AT-hook 2 (HMGA2), a known target of let-7 and enhancement of biliary proliferation. Conclusion: These results indicate that H19 plays a critical role in cholangiocyte proliferation and cholestatic liver injury in BA by regulating the S1PR2/SphK2 and let-7/HMGA2 axis. Serum exosomal H19 may represent a noninvasive diagnostic biomarker and potential therapeutic target for BA.


Assuntos
Ductos Biliares/citologia , Atresia Biliar/complicações , Proliferação de Células , Colestase/complicações , Células Epiteliais/fisiologia , Cirrose Hepática/etiologia , RNA Longo não Codificante/fisiologia , Animais , Atresia Biliar/metabolismo , Células Cultivadas , Colestase/metabolismo , Exossomos/metabolismo , Feminino , Humanos , Lactente , Cirrose Hepática/metabolismo , Masculino , Camundongos , RNA Longo não Codificante/análise , RNA Longo não Codificante/biossíntese
11.
Hepatology ; 70(4): 1317-1335, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30985008

RESUMO

Activation of hepatic stellate cells (HSCs) represents the primary driving force to promote the progression of chronic cholestatic liver diseases. We previously reported that cholangiocyte-derived exosomal long noncoding RNA-H19 (lncRNA-H19) plays a critical role in promoting cholestatic liver injury. However, it remains unclear whether cholangiocyte-derived lncRNA-H19 regulates HSC activation, which is the major focus of this study. Both bile duct ligation (BDL) and Mdr2 knockout (Mdr2-/- ) mouse models were used. Wild-type and H19maternalΔExon1/+ (H19KO) mice were subjected to BDL. Mdr2-/- H19maternalΔExon1/+ (DKO) mice were generated. Exosomes isolated from cultured mouse and human cholangiocytes or mouse serum were used for in vivo transplantation and in vitro studies. Fluorescence-labeled exosomes and flow cytometry were used to monitor exosome uptake by hepatic cells. Collagen gel contraction and bromodeoxyuridine assays were used to determine the effect of exosomal-H19 on HSC activation and proliferation. Mouse and human primary sclerosing cholangitis (PSC)/primary biliary cholangitis (PBC) liver samples were analyzed by real-time PCR, western blot analysis, histology, and immunohistochemistry. The results demonstrated that hepatic H19 level was closely correlated with the severity of liver fibrosis in both mouse models and human patients with PSC and PBC. H19 deficiency significantly protected mice from liver fibrosis in BDL and Mdr2-/- mice. Transplanted cholangiocyte-derived H19-enriched exosomes were rapidly and preferentially taken up by HSCs and HSC-derived fibroblasts, and promoted liver fibrosis in BDL-H19KO mice and DKO mice. H19-enriched exosomes enhanced transdifferentiation of cultured mouse primary HSCs and promoted proliferation and matrix formation in HSC-derived fibroblasts. Conclusion: Cholangiocyte-derived exosomal H19 plays a critical role in the progression of cholestatic liver fibrosis by promoting HSC differentiation and activation and represents a potential diagnostic biomarker and therapeutic target for cholangiopathies.


Assuntos
Colangite Esclerosante/genética , Colestase/patologia , Exossomos/genética , Regulação da Expressão Gênica , Cirrose Hepática/genética , RNA Longo não Codificante/genética , Animais , Proliferação de Células/genética , Células Cultivadas , Colangite Esclerosante/patologia , Colestase/genética , Modelos Animais de Doenças , Citometria de Fluxo , Células Estreladas do Fígado/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Cirrose Hepática/patologia , Masculino , Camundongos , Camundongos Knockout , Distribuição Aleatória , Especificidade da Espécie
12.
Pharmacol Res ; 155: 104684, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32045667

RESUMO

Human antigen R (HuR), also known as HuA and embryonic lethal abnormal vision-like 1 (ELAVL1), is a ubiquitously expressed RNA binding protein and functions as an RNA regulator and mediates the expression of various proteins by diverse post-transcriptional mechanisms. HuR has been well characterized in the inflammatory responses and in the development of various cancers. The importance of HuR-mediated roles in cell signaling, inflammation, fibrogenesis and cancer development in the liver has attracted a great deal of attention. However, there is still a substantial gap between the current understanding of the potential roles of HuR in the progression of liver disease and whether HuR can be targeted for the treatment of liver diseases. In this review, we introduce the function and mechanistic characterization of HuR, and then focus on the physiopathological roles of HuR in the development of different liver diseases, including hepatic inflammation, alcoholic liver diseases, non-alcoholic fatty liver diseases, viral hepatitis, liver fibrosis and liver cancers. We also summarize existing approaches targeting HuR function. In conclusion, although characterizing the liver-specific HuR function and demonstrating the multi-level regulative networks of HuR in the liver are still required, emerging evidence supports the notion that HuR represents a potential therapeutic target for the treatment of chronic liver diseases.


Assuntos
Proteína Semelhante a ELAV 1/imunologia , Hepatopatias/terapia , Animais , Humanos , Hepatopatias/imunologia
13.
Exp Mol Pathol ; 115: 104472, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32454104

RESUMO

BACKGROUND AND AIMS: Liver diseases and related complications represent an increasing source of morbidity and mortality and have become a significant economic burden worldwide. Dysregulated energy metabolism, alteration of redox homeostasis, immune responses, inflammation, fibrosis, and malfunctioning of the gut-liver axis are key driving forces of liver diseases. Emerging evidence suggested that long coding RNAs (lncRNAs) played crucial roles in the pathogenesis and development of various diseases. Among them, lncRNA H19 is a maternally expressed lncRNA and has attracted great attention in the research of liver diseases due to its extensive involvement in the epigenetic regulation, inflammation, tissue regeneration, and cancer initiation and development. RESULTS: In the current review, we will first introduce the definition of lncRNA H19, the regulation of lncRNA H19 expression and molecular targets of lncRNA H19. We then summarize the recent advances of studies focusing on the role of H19 in the pathogenesis of liver diseases, including fatty liver diseases, cholestasis, fibrosis and cancer, and in the modulation of gut-liver axis. CONCLUSION: Based on the complex roles of lncRNA H19 played in liver injury and gastrointestinal disorders, targeting H19 has great potency to become a new strategy to diagnose and treat gut- and liver-related disorders.


Assuntos
Biomarcadores/metabolismo , Trato Gastrointestinal/metabolismo , Hepatopatias/diagnóstico , Hepatopatias/genética , Fígado/metabolismo , Terapia de Alvo Molecular , RNA Longo não Codificante/metabolismo , Humanos , Hepatopatias/terapia , RNA Longo não Codificante/genética
14.
Am J Pathol ; 188(9): 2042-2058, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29963993

RESUMO

Esophageal adenocarcinoma (EAC) is the sixth leading cause of cancer deaths worldwide and has been dramatically increasing in incidence over the past decade. Gastroesophageal reflux and Barrett esophagus are well-established risk factors for disease progression. Conjugated bile acids (CBAs), including taurocholate (TCA), represent the major bile acids in the gastroesophageal refluxate of advanced Barrett esophagus and EAC patients. Our previous studies suggested that CBA-induced activation of sphingosine 1-phosphate receptor 2 (S1PR2) plays a critical role in promoting cholangiocarcinoma cell invasive growth. However, the role of CBAs in EAC development and underlying mechanisms remains elusive. In the current study, we identified that the expression level of S1PR2 is correlated to invasiveness of EAC cells. TCA significantly promoted cell proliferation, migration, invasion, transformation, and cancer stem cell expansion in highly invasive EAC cells (OE-33 cells), but had less effect on the lower invasive EAC cells (OE-19 cells). Pharmacologic inhibition of S1PR2 with specific antagonist JTE-013 or knockdown of S1PR2 expression significantly reduced TCA-induced invasive growth of OE-33 cells, whereas overexpression of S1PR2 sensitized OE-19 cells to TCA-induced invasive growth. Furthermore, TCA-induced activation of S1PR2 was closely associated with YAP and ß-catenin signaling pathways. In conclusion, CBA-induced activation of the S1PR2 signaling pathway is critically involved in invasive growth of EAC cells and represents a novel therapeutic target for EAC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenocarcinoma/patologia , Neoplasias Esofágicas/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Fosfoproteínas/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Ácido Taurocólico/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Colagogos e Coleréticos/farmacologia , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Humanos , Invasividade Neoplásica , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Fosfoproteínas/genética , Receptores de Lisoesfingolipídeo/genética , Receptores de Esfingosina-1-Fosfato , Fatores de Transcrição , Células Tumorais Cultivadas , Proteínas de Sinalização YAP
15.
Hepatology ; 67(4): 1441-1457, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28926118

RESUMO

Impaired intestinal barrier function promotes the progression of various liver diseases, including cholestatic liver diseases. The close association of primary sclerosing cholangitis (PSC) with inflammatory bowel disease highlights the importance of the gut-liver axis. It has been reported that bile duct ligation (BDL)-induced liver fibrosis is significantly reduced in C/EBP homologous protein knockout (CHOP-/- ) mice. However, the underlying mechanisms remain unclear. In the current study, we demonstrate that BDL induces striking and acute hepatic endoplasmic reticulum (ER) stress responses after 1 day, which return to normal after 3 days. No significant hepatocyte apoptosis is detected 7-14 days following BDL. However, the inflammatory response is significantly increased after 7 days, which is similar to what we found in human PSC liver samples. BDL-induced loss of stemness in intestinal stem cells (ISCs), disruption of intestinal barrier function, bacterial translocation, activation of hepatic inflammation, M2 macrophage polarization and liver fibrosis are significantly reduced in CHOP-/- mice. In addition, intestinal organoids derived from CHOP-/- mice contain more and longer crypt structures than those from wild-type (WT) mice, which is consistent with the upregulation of stem cell markers (leucine-rich repeat-containing G-protein-coupled receptor 5, olfactomedin 4, and SRY [sex determining region Y]-box 9) and in vivo findings that CHOP-/- mice have longer villi and crypts as compared to WT mice. Similarly, mRNA levels of CD14, interleukin-1ß, tumor necrosis factor-alpha, and monocyte chemotactic protein-1 are increased and stem cell proliferation is suppressed in the duodenum of patients with cirrhosis. CONCLUSION: Activation of ER stress and subsequent loss of stemness of ISCs plays a critical role in BDL-induced systemic inflammation and cholestatic liver injury. Modulation of the ER stress response represents a potential therapeutic strategy for cholestatic liver diseases as well as other inflammatory diseases. (Hepatology 2018;67:1441-1457).


Assuntos
Ductos Biliares/patologia , Colestase/patologia , Mucosa Intestinal/patologia , Células-Tronco/metabolismo , Fator de Transcrição CHOP/metabolismo , Animais , Apoptose/genética , Técnicas de Cultura de Células , Estresse do Retículo Endoplasmático/genética , Feminino , Hepatócitos/patologia , Humanos , Ligadura/efeitos adversos , Fígado/patologia , Hepatopatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/fisiologia , Fator de Transcrição CHOP/genética
16.
Hepatology ; 68(2): 599-615, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29425397

RESUMO

Cholestatic liver injury is an important clinical problem with limited understanding of disease pathologies. Exosomes are small extracellular vesicles released by a variety of cells, including cholangiocytes. Exosome-mediated cell-cell communication can modulate various cellular functions by transferring a variety of intracellular components to target cells. Our recent studies indicate that the long noncoding RNA (lncRNA), H19, is mainly expressed in cholangiocytes, and its aberrant expression is associated with significant down-regulation of small heterodimer partner (SHP) in hepatocytes and cholestatic liver injury in multidrug resistance 2 knockout (Mdr2-/- ) mice. However, how cholangiocyte-derived H19 suppresses SHP in hepatocytes remains unknown. Here, we report that cholangiocyte-derived exosomes mediate transfer of H19 into hepatocytes and promote cholestatic injury. Hepatic H19 level is correlated with severity of cholestatic injury in both fibrotic mouse models, including Mdr2-/- mice, a well-characterized model of primary sclerosing cholangitis (PSC), or CCl4 -induced cholestatic liver injury mouse models, and human PSC patients. Moreover, serum exosomal-H19 level is gradually up-regulated during disease progression in Mdr2-/- mice and patients with cirrhosis. H19-carrying exosomes from the primary cholangiocytes of wild-type (WT) mice suppress SHP expression in hepatocytes, but not the exosomes from the cholangiocytes of H19-/- mice. Furthermore, overexpression of H19 significantly suppressed SHP expression at both transcriptional and posttranscriptional levels. Importantly, transplant of H19-carrying serum exosomes of old fibrotic Mdr2-/- mice significantly promoted liver fibrosis (LF) in young Mdr2-/- mice. CONCLUSION: Cholangiocyte-derived exosomal-H19 plays a critical role in cholestatic liver injury. Serum exosomal H19 represents a noninvasive biomarker and potential therapeutic target for cholestatic diseases. (Hepatology 2018).


Assuntos
Ductos Biliares/patologia , Colestase/genética , Fígado/patologia , RNA Longo não Codificante/metabolismo , Animais , Ductos Biliares/metabolismo , Colangite Esclerosante/patologia , Colestase/patologia , Exossomos/genética , Feminino , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Receptores Citoplasmáticos e Nucleares/metabolismo
17.
Pharmacol Res ; 144: 210-226, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31022523

RESUMO

Liver diseases related complications represent a significant source of morbidity and mortality worldwide, creating a substantial economic burden. Oxidative stress, excessive inflammation, and dysregulated energy metabolism significantly contributed to liver diseases. Therefore, discovery of novel therapeutic drugs for the treatment of liver diseases are urgently required. Licorice is one of the most commonly used herbal drugs in Traditional Chinese Medicine for the treatment of liver diseases and drug-induced liver injury (DILI). Various bioactive components have been isolated and identified from the licorice, including glycyrrhizin, glycyrrhetinic acid, liquiritigenin, Isoliquiritigenin, licochalcone A, and glycycoumarin. Emerging evidence suggested that these natural products relieved liver diseases and prevented DILI through multi-targeting therapeutic mechanisms, including anti-steatosis, anti-oxidative stress, anti-inflammation, immunoregulation, anti-fibrosis, anti-cancer, and drug-drug interactions. In the current review, we summarized the recent progress in the research of hepatoprotective and toxic effects of different licorice-derived bioactive ingredients and also highlighted the potency of these compounds as promising therapeutic options for the treatment of liver diseases and DILI. We also outlined the networks of underlying molecular signaling pathways. Further pharmacology and toxicology research will contribute to the development of natural products in licorice and their derivatives as medicines with alluring prospect in the clinical application.


Assuntos
Anti-Inflamatórios/uso terapêutico , Produtos Biológicos/uso terapêutico , Glycyrrhiza/química , Ácido Glicirrízico/uso terapêutico , Hepatopatias/tratamento farmacológico , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Ácido Glicirrízico/química , Ácido Glicirrízico/farmacologia , Humanos , Hepatopatias/metabolismo , Estresse Oxidativo/efeitos dos fármacos
18.
Hepatology ; 66(3): 869-884, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28271527

RESUMO

The multidrug resistance 2 knockout (Mdr2-/- ) mouse is a well-established model of cholestatic cholangiopathies. Female Mdr2-/- mice develop more severe hepatobiliary damage than male Mdr2-/- mice, which is correlated with a higher proportion of taurocholate in the bile. Although estrogen has been identified as an important player in intrahepatic cholestasis, the underlying molecular mechanisms of gender-based disparity of cholestatic injury remain unclear. The long noncoding RNA H19 is an imprinted, maternally expressed, and estrogen-targeted gene, which is significantly induced in human fibrotic/cirrhotic liver and bile duct-ligated mouse liver. However, whether aberrant expression of H19 accounts for gender-based disparity of cholestatic injury in Mdr2-/- mice remains unknown. The current study demonstrated that H19 was markedly induced (∼200-fold) in the livers of female Mdr2-/- mice at advanced stages of cholestasis (100 days old) but not in age-matched male Mdr2-/- mice. During the early stages of cholestasis, H19 expression was minimal. We further determined that hepatic H19 was mainly expressed in cholangiocytes, not hepatocytes. Both taurocholate and estrogen significantly activated the extracellular signal-regulated kinase 1/2 signaling pathway and induced H19 expression in cholangiocytes. Knocking down H19 not only significantly reduced taurocholate/estrogen-induced expression of fibrotic genes and sphingosine 1-phosphate receptor 2 in cholangiocytes but also markedly reduced cholestatic injury in female Mdr2-/- mice. Furthermore, expression of small heterodimer partner was substantially inhibited at advanced stages of liver fibrosis, which was reversed by H19 short hairpin RNA in female Mdr2-/- mice. Similar findings were obtained in human primary sclerosing cholangitis liver samples. CONCLUSION: H19 plays a critical role in the disease progression of cholestasis and represents a key factor that causes the gender disparity of cholestatic liver injury in Mdr2-/- mice. (Hepatology 2017;66:869-884).


Assuntos
Colangite Esclerosante/patologia , Colestase/patologia , Regulação da Expressão Gênica , RNA Longo não Codificante/genética , Receptores de Lisoesfingolipídeo/genética , Animais , Colangite Esclerosante/genética , Colestase/genética , Modelos Animais de Doenças , Regulação para Baixo , Estrogênios/farmacologia , Feminino , Técnicas de Inativação de Genes , Genes MDR , Humanos , Cirrose Hepática/genética , Cirrose Hepática/patologia , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Papel (figurativo) , Fatores Sexuais , Receptores de Esfingosina-1-Fosfato , Ácido Taurocólico/farmacologia
19.
Hepatology ; 65(6): 2005-2018, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28120434

RESUMO

Bile duct obstruction is a potent stimulus for cholangiocyte proliferation, especially for large cholangiocytes. Our previous studies reported that conjugated bile acids (CBAs) activate the protein kinase B (AKT) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling pathways through sphingosine 1-phosphate receptor (S1PR) 2 in hepatocytes and cholangiocarcinoma cells. It also has been reported that taurocholate (TCA) promotes large cholangiocyte proliferation and protects cholangiocytes from bile duct ligation (BDL)-induced apoptosis. However, the role of S1PR2 in bile-acid-mediated cholangiocyte proliferation and cholestatic liver injury has not been elucidated. Here, we report that S1PR2 is the predominant S1PR expressed in cholangiocytes. Both TCA- and sphingosine-1-phosphate (S1P)-induced activation of ERK1/2 and AKT were inhibited by JTE-013, a specific antagonist of S1PR2, in cholangiocytes. In addition, TCA- and S1P-induced cell proliferation and migration were inhibited by JTE-013 and a specific short hairpin RNA of S1PR2, as well as chemical inhibitors of ERK1/2 and AKT in mouse cholangiocytes. In BDL mice, expression of S1PR2 was up-regulated in whole liver and cholangiocytes. S1PR2 deficiency significantly reduced BDL-induced cholangiocyte proliferation and cholestatic injury, as indicated by significant reductions in inflammation and liver fibrosis in S1PR2 knockout mice. Treatment of BDL mice with JTE-013 significantly reduced total bile acid levels in serum and cholestatic liver injury. CONCLUSION: This study suggests that CBA-induced activation of S1PR2-mediated signaling pathways plays a critical role in obstructive cholestasis and may represent a novel therapeutic target for cholestatic liver diseases. (Hepatology 2017;65:2005-2018).


Assuntos
Colangiocarcinoma/patologia , Colangite Esclerosante/patologia , Colestase/patologia , Cirrose Hepática/patologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Análise de Variância , Animais , Ácidos e Sais Biliares/farmacologia , Neoplasias dos Ductos Biliares/metabolismo , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares/cirurgia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Colangiocarcinoma/metabolismo , Colangite Esclerosante/metabolismo , Colestase/complicações , Modelos Animais de Doenças , Ligadura , Fígado/lesões , Fígado/patologia , Cirrose Hepática/metabolismo , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Knockout , Distribuição Aleatória , Papel (figurativo) , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Regulação para Cima
20.
Pharmacol Res ; 125(Pt B): 105-113, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28889972

RESUMO

AMP-activated protein kinase (AMPK), recognized as an energy sensor with three heterotrimeric subunits (α, ß and γ), not only maintains basal intracellular adenosine triphosphate levels but also regulates energy-intensive pathological responses, such as neurodegenerative and metabolic diseases, through multiple signaling pathways. Recent studies open a new direction for AMPK research and demonstrate that AMPK is a critical player in the pathogenesis of cholestatic liver injury and plays paradoxical roles in the regulation of different pathological processes, including the disruption of bile acid homeostasis and the regulation of hepatic polarity, inflammation and fibrosis. In the present review, we summarize recent findings that implicate AMPK-mediated signaling pathways in the pathogenesis of cholestatic liver injury. These findings provide novel insight regarding the potential use of AMPK as a therapeutic target for the treatment of cholestatic liver injury.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Colestase/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Canalículos Biliares/patologia , Microbioma Gastrointestinal , Humanos , Cirrose Hepática/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA