Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Biochem Biophys Res Commun ; 720: 150118, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38776757

ABSTRACT

Tectorigenin (TEC) as a plant extract has the advantage of low side effects on metabolic dysfunction-associated steatohepatitis (MASH) treatment. Our previous study have shown that tRNA-derived RNA fragments (tRFs) associated with autophagy and pyroptosis in MASH, but whether TEC can mitigate MASH through tRFs-mediated mitophagy is not fully understood. This study aims to investigate whether TEC relies on tRFs to adjust the crosstalk of hepatocyte mitophagy with pyroptosis in MASH. Immunofluorescence results of PINK1 and PRKN with MitoTracker Green-labeled mitochondria verified that TEC enhanced mitophagy. Additionally, TEC inhibited pyroptosis, as reflected by the level of GSDME, NLRP3, IL-1ß, and IL-18 decreased after TEC treatment, while the effect of pyroptosis inhibition by TEC was abrogated by Pink1 silencing. We found that the upregulation expression of tRF-3040b caused by MASH was suppressed by TEC. The promotion of mitophagy and the suppression of pyroptosis induced by TEC were abrogated by tRF-3040b mimics. TEC reduced lipid deposition, inflammation, and pyroptosis, and promoted mitophagy in mice, but tRF-3040b agomir inhibited these effects. In summary, our findings provided that TEC significantly reduced the expression of tRF-3040b to enhance mitophagy, thereby inhibiting pyroptosis in MASH. We elucidated a powerful theoretical basis and provided safe and effective potential drugs for MASH with the prevention and treatment.


Subject(s)
Down-Regulation , Isoflavones , Mice, Inbred C57BL , Mitophagy , Pyroptosis , Pyroptosis/drug effects , Mitophagy/drug effects , Animals , Mice , Male , Isoflavones/pharmacology , Down-Regulation/drug effects , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/drug therapy , Fatty Liver/genetics , Humans
2.
World J Gastroenterol ; 30(9): 1189-1212, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38577195

ABSTRACT

BACKGROUND: Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) plays a crucial role in metabolizing and detoxifying endogenous and exogenous substances. However, its contribution to the progression of liver damage remains unclear. AIM: To determine the role and mechanism of UGT1A1 in liver damage progression. METHODS: We investigated the relationship between UGT1A1 expression and liver injury through clinical research. Additionally, the impact and mechanism of UGT1A1 on the progression of liver injury was analyzed through a mouse model study. RESULTS: Patients with UGT1A1 gene mutations showed varying degrees of liver damage, while patients with acute-on-chronic liver failure (ACLF) exhibited relatively reduced levels of UGT1A1 protein in the liver as compared to patients with chronic hepatitis. This suggests that low UGT1A1 levels may be associated with the progression of liver damage. In mouse models of liver injury induced by carbon tetrachloride (CCl4) and concanavalin A (ConA), the hepatic levels of UGT1A1 protein were found to be increased. In mice with lipopolysaccharide or liver steatosis-mediated liver-injury progression, the hepatic protein levels of UGT1A1 were decreased, which is consistent with the observations in patients with ACLF. UGT1A1 knockout exacerbated CCl4- and ConA-induced liver injury, hepatocyte apoptosis and necroptosis in mice, intensified hepatocyte endoplasmic reticulum (ER) stress and oxidative stress, and disrupted lipid metabolism. CONCLUSION: UGT1A1 is upregulated as a compensatory response during liver injury, and interference with this upregulation process may worsen liver injury. UGT1A1 reduces ER stress, oxidative stress, and lipid metabolism disorder, thereby mitigating hepatocyte apoptosis and necroptosis.


Subject(s)
Glucuronosyltransferase , Liver , Animals , Humans , Mice , Disease Models, Animal , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Liver/metabolism
3.
J Basic Microbiol ; 64(3): e2300424, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38175847

ABSTRACT

A series of pot trials were undertaken to examine the impact of four arbuscular mycorrhizal fungi (AMF), namely Glomus mosseae (G.m), Glomus etunicatum (G.e), Corymbiglomus tortuosum (C.t), and the combined application of Glomus etunicatum and Corymbiglomus tortuosum (G.e + C.t), on the energy metabolism of amaranth plants grown in soil enriched with selenite at a concentration of 0.5 mg kg-1 . The inoculation of four AMFs resulted in an increase in both amaranth biomass and selenium (Se) content in leaves. The activities of phosphoglucose isomerase (PGI) and glucose-6-phosphate dehydrogenase + 6-phosphogluconate dehydrogenase were observed to decrease when AMFs were inoculated, as compared with the absence of AMF inoculation. The inoculation with G.m, C.t, and G.e + C.t resulted in an increase in succinate dehydrogenase activity; however, the inoculation with G.m, G.e, and G.e + C.t led to an increase in ascorbate oxidase activity. Furthermore, the inoculation of all four AMFs resulted in an increase in cytochrome c oxidase activity and the concentrations of oxidized coenzyme I (NAD) and reduced coenzyme I (NADH). The polyphenol oxidase activity of amaranth plants increased when inoculated with G.m and G.e, whereas it decreased when inoculated with C.t and G.e + C.t. Furthermore, the application of all four AMF treatments resulted in a reduction in adenosine triphosphate (ATP) levels and energy charge. It was worth mentioning that there was a clear inverse relationship between the energy charge and the biomass, Se concentration in the leaves. The findings presented in this research indicated that AMF may have an impact on energy metabolism and ultimately the biomass of amaranth by influencing the uptake of Se.


Subject(s)
Amaranthus , Fungi , Mycorrhizae , NAD , Energy Metabolism
4.
Dig Dis Sci ; 68(11): 4186-4195, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37679574

ABSTRACT

BACKGROUND: Hepatic stellate cell hyperactivation is a central link in liver fibrosis development, transforming growth factor ß1 (TGF-ß1) is a key activator of HSCs. AIMS: This study investigated whether anlotinib attenuates CCl4 induced liver fibrosis in mice and explored its antifibrotic mechanism. METHODS: We used the human hepatic stellate cell line LX-2 for in vitro assays and used TGF-ß1 to induce hepatic fibrosis in LX-2 cells. We analyzed cytotoxicity using a cell-counting kit-8 and transwell chambers to detect the migratory ability of LX-2 cells. Western blotting was used to detect the protein levels of collagen type I, α-smooth muscle actin, and p-Smad3. In addition, mice with CCl4-induced hepatic fibrosis were used as in vivo models. Histopathological examination was performed using H&E staining, Masson's trichrome staining, and immunohistochemistry. RESULTS: Anlotinib significantly reversed TGF-ß1-induced protein levels of Col I, α-SMA and p-Smad3 and inhibits migratory and proliferative abilities in vitro using LX-2 cells. CCl4 cause F4 grade (Ishak) hepatic fibrosis, liver inflammatory scores ranged from 12 to 14 (Ishak), a mean ALT measurement of 130 U/L and a mean measurement AST value of 119 U/L in mice. However, the CCl4-induced changes were markedly attenuated by anlotinib treatment, which returned to F2 grade (Ishak) hepatic fibrosis, liver inflammatory scores ranged from 4 to 6 (Ishak), a mean ALT measurement of 40 U/L and a mean measurement AST value of 56 U/L in mice. CONCLUSIONS: Our results suggest that anlotinib-mediated suppression of liver fibrosis is related to the inhibition of TGF-ß1 signaling pathway. Hepatic stellate cell hyper activation is a central link in liver fibrosis development, transforming growth factor ß1 is a key activator of HSCs. Anlotinib is a multi-targeted tyrosine kinase inhibitor that has similar targets to nintedanib, a clinically used anti-pulmonary fibrosis drug. Our study demonstrates an FDA-approved drug-anlotinib-that could prevent liver fibrosis and inflammation. Experiments in cell cultures and mice show that anlotinib can inhibit the activation of hepatic stellate cells by down-regulating the TGFß1/smad3 pathway, thereby reversing liver fibrosis. In animal experiments, anlotinib showed protective effects on the CCl4-induced liver damage, including ameliorating liver inflammation, reversing liver fibrosis and reducing liver enzymes. This is a very good signal, anlotinib may be useful for halting or reversing the progression of liver fibrosis and could be employed in the development of novel therapeutic drugs for the management of chronic liver diseases.

5.
Clin Infect Dis ; 74(11): 1925-1932, 2022 06 10.
Article in English | MEDLINE | ID: mdl-34487151

ABSTRACT

BACKGROUND: Pradefovir is a liver-targeted prodrug of adefovir, a nucleoside/nucleotide analogue with antiviral activity against hepatitis B virus (HBV) DNA polymerase. This phase 2 study compared the efficacy and safety of oral pradefovir (30, 45, 60, or 75 mg) versus tenofovir disoproxil fumarate (TDF; 300 mg) and aimed to identify the most appropriate dose of pradefovir for the forthcoming phase 3 study. METHODS: Treatment-naive and experienced (not on treatment >6 months) patients with chronic hepatitis B were eligible. RESULTS: A total of 240 participants were randomized and treated in the study (48 per group). Approximately 80% were hepatitis B e antigen (HBeAg) positive, and 10% had liver cirrhosis. The reductions from baseline in HBV DNA levels achieved at week 24 were 5.40, 5.34, 5.33, and 5.40 log10 IU/mL, with pradefovir doses of 30-, 45-, 60-, and 75-mg, respectively, compared with 5.12 log10 IU/mL with TDF. However, HBeAg loss was attained by more participants who received 45-, 60-, or 75-mg pradefovir than by those receiving TDF (12%, 6%, and 9% vs 3%). The TDF group exhibited a more significant increase in serum creatinine than the pradefovir 30- and 45-mg groups, and serum phosphate levels were comparable among all groups. Most adverse events (AEs) were mild (grade 1). No treatment-related severe AEs were reported. Overall, AEs and laboratory abnormalities were comparable to those in the TDF group. CONCLUSIONS: Pradefovir and TDF exhibited comparable reductions in HBV DNA levels. All treatments were safe and well tolerated. CLINICAL TRIALS REGISTRATION: NCT00230503 and China Drug Trials CTR2018042.


Subject(s)
Hepatitis B, Chronic , Prodrugs , Adenine/analogs & derivatives , Antiviral Agents/adverse effects , DNA, Viral , Hepatitis B e Antigens , Hepatitis B virus/genetics , Humans , Organophosphorus Compounds , Prodrugs/adverse effects , Tenofovir/adverse effects , Treatment Outcome , Viral Load
6.
Sci Rep ; 11(1): 21854, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750395

ABSTRACT

Exploring the expression of miR-571 in patients with liver fibrosis and its role in the progression of liver fibrosis. A total of 74 patients with liver fibrosis in our institution from September to December 2018 were collected for study, and the expression of miR-571, Notch3 and Jagged1 in patients with different progressions of liver fibrosis was determined by RT-PCR and Western blot analysis. Set up Notch3 up group and Notch3 down regulated group, RT-PCR and Western blot were used to determine the effect of Notch signaling on the expression of fibrogenic factors. CCK-8, cell scratch assays, Transwell assays, flow cytometry were used to determine the effect of miR-571 on LX-2 proliferation, migration, apoptosis in human stem stellate cells, and RT-PCR, Western blot assays were performed to determine the effect of miR-571 on the Notch3 signaling pathway and the expression of profibrogenic factors. miR-571, Notch3 and Jagged1 are up-regulated in patients with liver fibrosis and is associated with the progression of liver fibrosis. Notch3 signaling pathway can promote the expression of fibroblast in human hepatic stellate cells; miR-571 can inhibit the apoptosis of human hepatic stellate cells, promote cell proliferation and migration; up regulation of miR-571 can promote the expression of Notch3 and Jagged1, and up-regulation of miR-571 also promoted the expression of related fibroblasts. MiR-571 can promote the activation of human stem cell stellate cells and the expression of fibroblast related factors through Notch3 signaling pathway.


Subject(s)
Liver Cirrhosis/genetics , MicroRNAs/genetics , Receptor, Notch3/genetics , Adolescent , Adult , Apoptosis , Case-Control Studies , Cell Line , Cell Movement , Cell Proliferation , Disease Progression , Female , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatitis B, Chronic/complications , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Male , MicroRNAs/metabolism , Middle Aged , Receptor, Notch3/metabolism , Signal Transduction , Up-Regulation , Young Adult
7.
Eur J Pharmacol ; 876: 172946, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-31996320

ABSTRACT

Liver fibrosis is involved in the progression of most chronic liver diseases. Even though we have made a huge progress in order to understand the pathogenesis of liver fibrosis, however, there is still a lack of productive treatments. Being a traditional Chinese medicine, Platycodin D (PD), an oleanane kind of triterpenoid saponin has been put to extensive use for treating different kinds of illnesses that include not just anti-nociceptive, but also antiviral, anti-inflammatory, and anti-cancer for thousands of years. Nonetheless, there has been no clarification made for its effects on the progression of liver fibrosis. In this manner, we carried out in vitro studies for the purpose of investigating the anti-fibrosis impact of PD. Activation of hepatic stellate cells was evaluated by means of the detection of the proliferation of HSCs and the expression of specific proteins. We discovered the fact that PD had the potential of activating HSCs. Thereafter, we detected the apoptosis and autophagy of the HSCs; as the results suggested, PD induced apoptosis and autophagy of the HSCs. It augmented the expression level of apoptotic proteins that included Bax, Cytochrome C (cyto-c), cleaved caspase3 and cleaved caspase9, in addition to the autophagy relevant proteins, for instance, LC3II, beclin1, Atg5 and Atg9. Further research was carried out for the investigation of the underlying molecular mechanism, and discovered that PD promoted the phosphorylation of JNK and c-Jun. Treating the JNK inhibitor P600125 inhibited the effect of PD, confirming the impact of PD on the regulation of JNK/c-Jun pathway. Thus, we speculated that PD alleviates liver fibrosis and activation of hepatic stellate via promoting phosphorylation of JNK and c-Jun and further altering the autophagy along with apoptosis of HSCs.


Subject(s)
Hepatic Stellate Cells/drug effects , Liver Cirrhosis/prevention & control , Liver/drug effects , MAP Kinase Signaling System/drug effects , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Cell Proliferation/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Phosphorylation , Saponins/administration & dosage , Saponins/therapeutic use , Triterpenes/administration & dosage , Triterpenes/therapeutic use
8.
World J Gastroenterol ; 25(30): 4222-4234, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31435175

ABSTRACT

BACKGROUND: Liver fibrosis is a refractory disease whose persistence can eventually induce cirrhosis or even liver cancer. Early liver fibrosis is reversible by intervention. As a member of the transforming growth factor-beta (TGF-ß) superfamily, bone morphogenetic protein 7 (BMP7) has anti-liver fibrosis functions. However, little is known about BMP7 expression changes and its potential regulatory mechanism as well as the relationship between BMP7 and TGF-ß during liver fibrosis. In addition, the mechanism underlying the anti-liver fibrosis function of BMP7 needs to be further explored. AIM: To investigate changes in the dynamic expression of BMP7 during liver fibrosis, interactions between BMP7 and TGF-ß1, and possible mechanisms underlying the anti-liver fibrosis function of BMP7. METHODS: Changes in BMP7 expression during liver fibrosis and the interaction between BMP7 and TGF-ß1 in mice were observed. Exogenous BMP7 was used to treat mouse primary hepatic stellate cells (HSCs) to observe its effect on activation, migration, and proliferation of HSCs and explore the possible mechanism underlying the anti-liver fibrosis function of BMP7. Mice with liver fibrosis received exogenous BMP7 intervention to observe improvement of liver fibrosis by using Masson's trichrome staining and detecting the expression of the HSC activation indicator alpha-smooth muscle actin (α-SMA) and the collagen formation associated protein type I collagen (Col I). Changes in the dynamic expression of BMP7 during liver fibrosis in the human body were further observed. RESULTS: In the process of liver fibrosis induced by carbon tetrachloride (CCl4) in mice, BMP7 protein expression first increased, followed by a decrease; there was a similar trend in the human body. This process was accompanied by a sustained increase in TGF-ß1 protein expression. In vitro experiment results showed that TGF-ß1 inhibited BMP7 expression in a time- and dose-dependent manner. In contrast, high doses of exogenous BMP7 inhibited TGF-ß1-induced activation, migration, and proliferation of HSCs; this inhibitory effect was associated with upregulation of pSmad1/5/8 and downregulation of phosphorylation of Smad3 and p38 by BMP7. In vivo experiment results showed that exogenous BMP7 improved liver fibrosis in mice. CONCLUSION: During liver fibrosis, BMP7 protein expression first increases and then decreases. This changing trend is associated with inhibition of BMP7 expression by sustained upregulation of TGF-ß1 in a time- and dose-dependent manner. Exogenous BMP7 could selectively regulate TGF-ß/Smad pathway-associated factors to inhibit activation, migration, and proliferation of HSCs and exert anti-liver fibrosis functions. Exogenous BMP7 has the potential to be used as an anti-liver fibrosis drug.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/pathology , Liver/pathology , Administration, Oral , Animals , Bone Morphogenetic Protein 7/administration & dosage , Carbon Tetrachloride/toxicity , Cells, Cultured , Down-Regulation , Hepatic Stellate Cells/drug effects , Humans , Liver/cytology , Liver/drug effects , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Mice , Phosphorylation , Primary Cell Culture , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation
9.
Zhongguo Zhong Yao Za Zhi ; 44(11): 2231-2235, 2019 Jun.
Article in Chinese | MEDLINE | ID: mdl-31359647

ABSTRACT

In order to reveal the relationship between the amount of soil microorganisms and the quality of Fritillaria taipaiensis, both cultivated and wild F. taipaiensis were collected from Chongqing, Wuxi at different stages of their growth as objects of the research. The mycorrhizal infection rate and colonization intensity, peimisine and total alkaloid content in bulbs, the amount of microorganisms and biomass carbon content in rhizospheric soil were all determined using common methods. The results showed that the typical arbuscular-vesicle roots were formed after the AM fungi infected the F. taipaiensis roots which were collected from different origins. The mycorrhizal infection rates were ranged from 78.74% to 98.68% and the colonization intensities were ranged from 13.29% to 37.06%. The rhizospheric microorganisms of F. taipaiensis showed abundant resources. The distribution rule of them in the rhizospheric soil was as follows: the amount of bacteria>the amount of actinomycetes>the amount of fungi. The rhizospheric bacteria, decomposition inorganic phosphorus bacteria, decomposition organic phosphorus bacteria, actinomycetes amount and the total number of microbes increased first and then decreased with the increase of years, while decomposition potassium bacteria showed decreasing trend and fungi showed gradual increasing trend. The soil microbial flora content in the soil changed from "bacterial type" with a high fertility to "fungal type" with a low fertility. The mass fraction of peimisine and total alkaloid content increased first and then decreased with the increase of over the years, the same trend of culturable rhizosphere soil bacteria and actinomycetes indicated that the growth years affected the quality of soil and medicinal materials on different levels. Therefore, the diversity of microbial communities in rhizosphere soil reduced with the increase of years leading to the continuous cropping obstacles and the destruction of medicinal quality of F. taipaiensis.


Subject(s)
Alkaloids/analysis , Fritillaria/chemistry , Mycorrhizae , Rhizosphere , Soil Microbiology , Fritillaria/microbiology , Plant Roots
11.
PLoS One ; 14(4): e0215616, 2019.
Article in English | MEDLINE | ID: mdl-30986272

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0201136.].

12.
Drug Des Devel Ther ; 12: 4107-4115, 2018.
Article in English | MEDLINE | ID: mdl-30584275

ABSTRACT

PURPOSE: Liver fibrosis is a worldwide health issue. Development of effective new drugs for treatment of this disease is of great importance. This study investigated the therapeutic effects of ferulic acid on liver fibrosis in vitro and in vivo. MATERIALS AND METHODS: Human hepatic stellate cell line (HSC) LX-2 was used for in vitro assays. Transforming growth factor ß1 (TGF-ß1) was used to induce hepatic fibrosis in LX-2 cells. Western blot was used to detect protein levels of collagen I, fibronectin, α-smooth muscle actin (SMA), p-Smad2, p-Smad3, p-p38, and p-JNK. Gene expression was measured by RT-qPCR. Fluorescence staining was used to determine localization of Smad4. CCl4-induced hepatic fibrosis in SD rats was used as an in vivo model. Histological features were detected by hematoxylin and eosin staining. Levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), hexadecenoic acid (HA), and hydroxyproline (Hyp) were measured by ELISA. RESULTS: TGF-ß1 treatment significantly increased levels of collagen I, fibronectin, α-SMA, p-Smad2, p-Smad3, and Smad4 in LX-2 cells. Ferulic acid improved TGF-ß1-induced hepatic fibrosis via regulation of the TGF-ß1/Smad pathway. Consistent with in vitro data, CCl4 caused severe hepatic fibrosis in SD rats, as determined by ALT, AST, HA, and Hyp upregulation. Protein levels of p-Smad2 and p-Smad3 in liver tissues were significantly increased following treatment with CCl4. All CCL4-induced changes were markedly attenuated by ferulic acid treatment. CONCLUSION: Ferulic acid potently improved hepatic fibrosis via inhibition of the TGF-ß1/Smad pathway in vitro and in vivo. These findings provided evidence for potential use of ferulic acid to treat or prevent liver fibrosis.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Coumaric Acids/pharmacology , Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Animals , Carbon Tetrachloride , Cell Line , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Collagen Type I/metabolism , Cytoprotection , Fibronectins/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Phosphorylation , Rats, Wistar , Signal Transduction/drug effects
13.
Food Funct ; 9(12): 6298-6306, 2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30411754

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent diseases worldwide. Blueberry, combined with probiotics (BP), might be a potential candidate for NAFLD treatment, due to its anti-inflammatory and anti-apoptotic properties. Here, we investigated whether the anti-inflammatory cytokine, IL-22, was involved in the therapeutic process of BP, using cell and rat models of NAFLD. Results indicated that BP significantly reduced lipid droplets and triglyceride (TG) accumulation in NAFLD cells. However, when IL-22 was deficient, the lipid droplets and TG content were significantly increased. In vivo, the serum parameters and pathological degree of NAFLD rats were significantly improved by BP, while IL-22 silencing significantly abolished the BP effect. Immunohistochemistry, immunofluorescence, qRT-PCR, and western blotting showed that the NAFLD group expressed significantly lower levels of IL-22, JAK1, and STAT3, and higher levels of BAX, than the normal group. Furthermore, BP significantly elevated the levels of IL-22, JAK1 and STAT3, and reduced the level of BAX in NAFLD, while IL-22 silencing prevented BP from restoring the expression of JAK1, STAT3, and BAX. We conclude that IL-22 is vital for the therapeutic effect of BP, and acts via activation of JAK1/STAT3 signaling and inhibition of the apoptosis factor BAX, which makes IL-22 a promising target for therapy of NAFLD.


Subject(s)
Blueberry Plants/metabolism , Interleukins/metabolism , Janus Kinase 1/metabolism , Non-alcoholic Fatty Liver Disease/diet therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Probiotics/administration & dosage , STAT3 Transcription Factor/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Blueberry Plants/chemistry , Fruit and Vegetable Juices/analysis , Humans , Interleukins/genetics , Janus Kinase 1/genetics , Male , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Rats , Rats, Wistar , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , bcl-2-Associated X Protein/genetics , Interleukin-22
14.
Cell Physiol Biochem ; 51(3): 1389-1398, 2018.
Article in English | MEDLINE | ID: mdl-30481761

ABSTRACT

BACKGROUND/AIMS: Hepatic stellate cells (HSCs) are the primary cell type responsible for liver fibrosis. Our study proved that thymosin beta 4 (Tß4) has anti-fibrogenic effects in HSCs through PI3K/AKT pathway. However, the underlying mechanisms are not fully elucidated. Circular RNAs (circRNAs) play important roles in fine-tuning gene expression and are often deregulated in cancers. However, the expression profile and clinical significance of in liver fibrosis is still unknown. Therefore, we hypothesize that Tß4 influences circRNAs in liver fibrosis. METHODS: Circular RNA microarray was conducted to identify Tß4-related circRNAs. Pathway analysis and miRNA response elements analysis was conducted to predict the potential roles of differentially expressed circRNAs in liver fibrosis. CCK8 assays and flow cytometric assays were conducted to clarify the role of circRNA in liver fibrosis. Bioinformatics analysis and in vitro experiments were conducted to clarify the mechanism of circRNA-mediated gene regulation in liver fibrosis. RESULTS: A total of 644 differentially expressed circRNAs were identified between the Tß4-depleted LX-2 cells and the control LX2 cells. The expression of circRNA-0067835 was significantly increased in the Tß4-depleted LX-2 cells compared with control. Knockdown of circRNA-0067835 observably decreased LX-2 cell proliferation by causing G1 arrest and promoting apoptosis. Bioinformatics online programs predicted that circRNA-0067835 acted as miR-155 sponge to regulate FOXO3a expression, which was validated using luciferase reporter assay. CONCLUSION: Our experiments showed that circRNA-0067835 regulated liver fibrosis progression by acting as a sponge of miR-155 to promote FOXO3a expression, indicating that circRNA-0067835 may serve as a potential therapeutic target for patients with liver fibrosis.


Subject(s)
Forkhead Box Protein O3/genetics , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , MicroRNAs/genetics , RNA/genetics , Signal Transduction , Thymosin/genetics , Animals , Cell Line , Cells, Cultured , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , RNA, Circular , Transcriptome
15.
PLoS One ; 13(8): e0201136, 2018.
Article in English | MEDLINE | ID: mdl-30071053

ABSTRACT

Cyclophosphamide (CTX) has immunosuppressive effects and has been wildly used as one anti-cancer drug in clinical. Significant toxicity has been noticed particularly in the reproductive system. CTX promotes the maturation of ovarian follicles, decreases follicular reserve, and ultimately lead to ovarian failure or even premature ovarian failure (POF). The placental extract (HPE) has been shown to have some beneficial impact on reproductive system; however, little is known regarding to the effect of HPE on protecting CTX-induced ovarian injury and the mechanism involved. Whether human placental extracts (HPE) has a protective effect on CTX-induced toxicity on ovarian was studied by using a CTX-induced ovarian injury animal model. The effects of HEP on histopathology, the number of atretic follicles, the weight of the ovary, serum hormone levels, and apoptosis in granulosa cells were studied in mice with CTX or control vehicle. Our results have demonstrated that HPE inhibited p-Rictor, reduced the expression of Bad, Bax and PPAR, and activated Akt and Foxo3a (increased their phosphorylation). Mice treated with HPE showed higher ovarian weight, lower number of atretic follicles, higher serum levels of the hormones E2 and progesterone, and lower apoptosis and serum levels of LH and FSH in granulosa cells, than that in the control animal group. Our data show that ovarian injury can be attenuated by HPE. HPE likely protects follicular granulosa cells from undergoing significant apoptosis and reduce atresia follicle formation, therefore, alleviates CTX-induced ovarian injury.


Subject(s)
Cyclophosphamide/toxicity , Forkhead Box Protein O3/metabolism , Ovary/drug effects , Placental Extracts/pharmacology , Protective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antineoplastic Agents, Alkylating/toxicity , Apoptosis/drug effects , Apoptosis/physiology , Dose-Response Relationship, Drug , Female , Hormones/blood , Humans , Mice, Inbred C57BL , Organ Size , Ovary/metabolism , Ovary/pathology , Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors , Peroxisome Proliferator-Activated Receptors/metabolism , Phosphorylation/drug effects , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/prevention & control , Rapamycin-Insensitive Companion of mTOR Protein/antagonists & inhibitors , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/antagonists & inhibitors , bcl-Associated Death Protein/metabolism
16.
World J Gastroenterol ; 23(40): 7253-7264, 2017 Oct 28.
Article in English | MEDLINE | ID: mdl-29142472

ABSTRACT

AIM: To investigate the protective effect of prostaglandin E1 (PGE1) against endoplasmic reticulum (ER) stress-induced hepatocyte apoptosis, and to explore its underlying mechanisms. METHODS: Thapsigargin (TG) was used to induce ER stress in the human hepatic cell line L02 and hepatocarcinoma-derived cell line HepG2. To evaluate the effects of PGE1 on TG-induced apoptosis, PGE1 was used an hour prior to TG treatment. Activation of unfolded protein response signaling pathways were detected by western blotting and quantitative real-time RT-PCR. Apoptotic index and cell viability of L02 cells and HepG2 cells were determined with flow cytometry and MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay. RESULTS: Pretreatment with 1 µmol/L PGE1 protected against TG-induced apoptosis in both L02 cells and HepG2 cells. PGE1 enhanced the TG-induced expression of C/EBP homologous protein (CHOP), glucose-regulated protein (GRP) 78 and spliced X box-binding protein 1 at 6 h. However, it attenuated their expressions after 24 h. PGE1 alone induced protein and mRNA expressions of GRP78; PGE1 also induced protein expression of DNA damage-inducible gene 34 and inhibited the expressions of phospho-PKR-like ER kinase, phospho-eukaryotic initiation factor 2α and CHOP. Treatment with protein kinase A (PKA)-inhibitor H89 or KT5720 blocked PGE1-induced up-regulation of GRP78. Further, the cytoprotective effect of PGE1 on hepatocytes was not observed after blockade of GRP78 expression by H89 or small interfering RNA specifically targeted against human GRP78. CONCLUSION: Our study demonstrates that PGE1 protects against ER stress-induced hepatocyte apoptosis via PKA pathway-dependent induction of GRP78 expression.


Subject(s)
Alprostadil/pharmacology , Apoptosis/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , Heat-Shock Proteins/metabolism , Carbazoles/pharmacology , Cell Survival/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Heat-Shock Proteins/genetics , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/physiology , Humans , Isoquinolines/pharmacology , Phosphorylation , Pyrroles/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Thapsigargin/pharmacology , Transcription Factor CHOP/metabolism , Unfolded Protein Response/drug effects , X-Box Binding Protein 1/metabolism
17.
Sci Rep ; 7(1): 4032, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28642549

ABSTRACT

Understanding the underlying molecular mechanisms of liver fibrosis is important to develop effective therapy. Herein, we show that focal-adhesion-kinse (FAK) plays a key role in promoting hepatic stellate cells (HSCs) activation in vitro and liver fibrosis progression in vivo. FAK activation is associated with increased expression of α-smooth muscle actin (α-SMA) and collagen in fibrotic live tissues. Transforming growth factor beta-1 (TGF-ß1) induces FAK activation in a time and dose dependent manner. FAK activation precedes the α-SMA expression in HSCs. Inhibition of FAK activation blocks the α-SMA and collagen expression, and inhibits the formation of stress fibers in TGF-ß1 treated HSCs. Furthermore, inhibition of FAK activation significantly reduces HSC migration and small GTPase activation, and induces apoptotic signaling in TGF-ß1 treated HSCs. Importantly, FAK inhibitor attenuates liver fibrosis in vivo and significantly reduces collagen and α-SMA expression in an animal model of liver fibrosis. These data demonstrate that FAK plays an essential role in HSC activation and liver fibrosis progression, and FAK signaling pathway could be a potential target for liver fibrosis.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Actins/genetics , Actins/metabolism , Animals , Becaplermin/metabolism , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Enzyme Activation , Female , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Male , Mice , Protein Kinase Inhibitors/pharmacology , Transforming Growth Factor beta1/metabolism
18.
Sci Rep ; 7(1): 1359, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28465509

ABSTRACT

Growing evidence has shown that gut microbiome is a key factor involved in liver health. Therefore, gut microbiota modulation with probiotic bacteria, such as Saccharomyces boulardii, constitutes a promising therapy for hepatosis. In this study, we aimed to investigate the protective effects of S. boulardii on D-Galactosamine-induced liver injury in mice. Liver function test and histopathological analysis both suggested that the liver injury can be effectively attenuated by S. boulardii administration. In the meantime, S. boulardii induced dramatic changes in the gut microbial composition. At the phylum level, we found that S. boulardii significantly increased in the relative abundance of Bacteroidetes, and decreased the relative abundance of Firmicutes and Proteobacteria, which may explain the hepatic protective effects of S. boulardii. Taken together, our results demonstrated that S. boulardii administration could change the gut microbiota in mice and alleviate acute liver failure, indicating a potential protective and therapeutic role of S. boulardii.


Subject(s)
Chemical and Drug Induced Liver Injury/microbiology , Galactosamine/toxicity , Gastrointestinal Microbiome , Saccharomyces boulardii , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , BALB 3T3 Cells , Chemical and Drug Induced Liver Injury/diet therapy , Chemical and Drug Induced Liver Injury/pathology , Galactosamine/analogs & derivatives , Mice , Probiotics
19.
PLoS One ; 11(12): e0167451, 2016.
Article in English | MEDLINE | ID: mdl-27911957

ABSTRACT

RATIONALE: Tumor necrosis factor-alpha (TNF-α) is a potent pro-inflammatory mediator and its expression is up-regulated in chronic obstructive pulmonary disease (COPD). Tristetraprolin (TTP) is implicated in regulation of TNF-α expression; however, whether TTP is involved in cigarette smoke-induced TNF-α expression has not been determined. METHODS: TTP expression was examined by western blot analysis in murine alveolar macrophages and alveolar epithelial cells challenged without or with cigarette smoke extract (CSE). TNF-α mRNA stability, and the decay of TNF-α mRNA, were determined by real-time quantitative RT-PCR. TNF-α protein levels were examined at the same time in these cells. To identify the molecular mechanism involved, a construct expressing the human beta-globin reporter mRNA containing the TNF-α 3'-untranslated region was generated to characterize the TTP targeted site within TNF-α mRNA. RESULTS: CSE induced TTP down-regulation in alveolar macrophages and alveolar epithelial cells. Reduced TTP expression resulted in significantly increased TNF-α mRNA stability. Importantly, increased TNF-α mRNA stability due to impaired TTP function resulted in significantly increased TNF-α levels in these cells. Forced TTP expression abrogated the increased TNF-α mRNA stability and expression induced by CSE. By using the globin reporter construct containing TNF-α mRNA 3'-untranslated region, the data indicate that TTP directly targets the adenine- and uridine-rich region (ARE) of TNF-α mRNA and negatively regulates TNF-α expression at the post-transcriptional level. CONCLUSION: The data demonstrate that cigarette smoke exposure reduces TTP expression and impairs TTP function, resulting in significantly increased TNF-α mRNA stability and excessive TNF-α expression in alveolar macrophages and epithelial cells. The data suggest that TTP is a novel post-transcriptional regulator and limits excessive TNF-α expression and inflammatory response induced by cigarette smoke.


Subject(s)
Complex Mixtures/toxicity , Down-Regulation/drug effects , Epithelial Cells/metabolism , Macrophages, Alveolar/metabolism , RNA Stability/drug effects , RNA, Messenger/metabolism , Respiratory Mucosa/metabolism , Smoking/metabolism , Tristetraprolin/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cells, Cultured , Epithelial Cells/pathology , Humans , Macrophages, Alveolar/pathology , Mice , RNA, Messenger/genetics , Respiratory Mucosa/pathology , Smoking/genetics , Smoking/pathology , Tristetraprolin/genetics , Tumor Necrosis Factor-alpha/genetics
20.
Hum Gene Ther ; 27(11): 880-891, 2016 11.
Article in English | MEDLINE | ID: mdl-27136973

ABSTRACT

Liver fibrosis results in cirrhosis, liver cancer, and liver failure, which is a major cause of mortality worldwide. Gene therapy is a relatively new paradigm in medicine, with enormous therapeutic potential. The development of an efficient and safe delivery system is essential for clinical gene therapy. In the present study, we evaluated augmenter of liver regeneration/growth factor ERV1-like (ALR/GFER) gene therapeutic effect mediated by a novel minicircle vector (MC-hALR). The results in liver fibrotic rats that received MC-hALR through hydrodynamics-based transfection (HBT) for 8 weeks indicated that the minicircle DNA vector produced a more effective gene therapy effect than traditional plasmids (pcDNA3.1-hALR). Even when we reduced the treatment dose of MC-hALR to 30% (w/w) and the treatment frequency from weekly to biweekly, the in vitro and in vivo results still demonstrated that higher ALR gene expression significantly blocked increases in transforming growth factor-ß1 (TGF-ß1), platelet derived growth factor-BB (PDGF-BB), and α-smooth muscle aorta (α-SMA) levels; effectively suppressed the production of collagens, especially collagen I; and effectively alleviated liver injury and fibrosis in rats, thereby improving the survival rate of liver fibrotic rats. It is preliminarily concluded that the relative overexpression of MC-hALR inhibits the activation of hepatic stellate cells (HSCs), thereby alleviating liver fibrosis in rats.


Subject(s)
DNA, Circular/genetics , DNA-Binding Proteins/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Hepatic Stellate Cells/cytology , Liver Cirrhosis/genetics , Liver Cirrhosis/therapy , Neoplasm Proteins/genetics , Animals , Liver Regeneration , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...