Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Affiliation country
Publication year range
1.
Clin Immunol ; 261: 110167, 2024 04.
Article in English | MEDLINE | ID: mdl-38453127

ABSTRACT

Excessive inflammatory response and increased oxidative stress play an essential role in the pathophysiology of ischemia/reperfusion (I/R)-induced acute kidney injury (IRI-AKI). Emerging evidence suggests that lipoxin A4 (LXA4), as an endogenous negative regulator in inflammation, can ameliorate several I/R injuries. However, the mechanisms and effects of LXA4 on IRI-AKI remain unknown. In this study, A bilateral renal I/R mouse model was used to evaluate the role of LXA4 in wild-type, IRG1 knockout, and IRAK-M knockout mice. Our results showed that LXA4, as well as 5-LOX and ALXR, were quickly induced, and subsequently decreased by renal I/R. LXA4 pretreatment improved renal I/R-induced renal function impairment and renal damage and inhibited inflammatory responses and oxidative stresses in mice kidneys. Notably, LXA4 inhibited I/R-induced the activation of TLR4 signal pathway including decreased phosphorylation of TAK1, p36, and p65, but did not affect TLR4 and p-IRAK-1. The analysis of transcriptomic sequencing data and immunoblotting suggested that innate immune signal molecules interleukin-1 receptor-associated kinase-M (IRAK-M) and immunoresponsive gene 1 (IRG1) might be the key targets of LXA4. Further, the knockout of IRG1 or IRAK-M abolished the beneficial effects of LXA4 on IRI-AKI. In addition, IRG1 deficiency reversed the up-regulation of IRAK-M by LXA4, while IRAK-M knockout had no impact on the IRG1 expression, indicating that IRAK-M is a downstream molecule of IRG1. Mechanistically, we found that LXA4-promoted IRG1-itaconate not only enhanced Nrf2 activation and increased HO-1 and NQO1, but also upregulated IRAK-M, which interacted with TRAF6 by competing with IRAK-1, resulting in deactivation of TLR4 downstream signal in IRI-AKI. These data suggested that LXA4 protected against IRI-AKI via promoting IRG1/Itaconate-Nrf2 and IRAK-M-TRAF6 signaling pathways, providing the rationale for a novel strategy for preventing and treating IRI-AKI.


Subject(s)
Acute Kidney Injury , Lipoxins , Reperfusion Injury , Succinates , Mice , Animals , NF-E2-Related Factor 2/metabolism , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/pharmacology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/pharmacology , Signal Transduction , Kidney/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Acute Kidney Injury/prevention & control
2.
BMC Musculoskelet Disord ; 24(1): 597, 2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37474941

ABSTRACT

Osteoarthritis (OA) is a common disease characterized by cartilage degradation. Growing evidence showed that glucose metabolism impacts joint homeostasis and an imbalance between glycolysis and oxidative phosphorylation (OXPHOS) may exacerbate OA progression, however, a definitive link is yet to be established. Here, we report that pyruvate metabolism and oxidative phosphorylation pathway is enriched in OA cartilage through gene set enrichment analysis (GSEA) and expression of Pyruvate Dehydrogenase Kinase 1 (PDK1), an enzyme that can phosphorylate Pyruvate Dehydrogenase (PDH), and inhibit pyruvate fluxes into the tricarboxylic acid (TCA) cycle and to OXPHOS, in articular cartilage is notably reduced through destabilization of medial meniscus (DMM). Moreover, by inhibiting PDK1, cartilage loss is markedly accelerated in DMM-induced OA through extracellular matrix (ECM) degradation and apoptosis of chondrocytes. These results indicate that PDK1 is involved in the progression of OA through accelerating cartilage matrix degradation and synovium inflammation to ameliorate cartilage degeneration.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Animals , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Oxidative Phosphorylation , Chondrocytes/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Disease Models, Animal
3.
Medicina (Kaunas) ; 59(9)2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37763758

ABSTRACT

Background and Objectives: Non-alcoholic steatohepatitis (NASH) is a significant risk factor for hepatocellular carcinoma (HCC) development. Timely treatment during the NASH stage is essential to minimize the possibility of disease progression to HCC. Cuproptosis is a newly identified form of cellular death that could impact the progression of various diseases and cancers. Materials and Methods: Transcriptome and single-cell sequencing datasets were utilized to investigate the role of cuproptosis-related genes (CRGs) in NASH progression to HCC. FDX1, LIPT1, and PDHP were identified as CRGs in NASH patients, and FDX1, DBT, GCSH, SLC31A1, and DLAT were identified as CRGs in patients with NASH progressing to HCC. FDX1 was found to play a significant role in both NASH patients and patients with NASH progressing to HCC. This study constructed cuproptosis-related clusters (CRCs) using the Nonnegative Matrix Factorization algorithm, and they were linked to fatty acid metabolism and the PPAR signaling pathway in both NASH CRCs and HCC CRCs. The Weighted Correlation Network Analysis algorithm identified CRP, CRC, TAT, CXCL10, and ACTA1 as highly relevant genes in NASH CRCs and HCC CRCs. The expression of FDX1 was validated in both mouse models and human NASH samples. Results: The investigation highlights FDX1 as a pivotal CRG in both NASH and NASH progression to HCC. The comprehensive characterization of CRGs sheds light on their potential biofunctional importance in the context of NASH and HCC. Our experimental results show that FDX1 expression was significantly increased in NASH patients. Conclusions: The present study identified key CRGs, revealing their potential impact on NASH and HCC. Meanwhile, targeting FDX1 may prevent the progression of NASH to HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Mice , Humans , Carcinoma, Hepatocellular/genetics , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/genetics , Liver Neoplasms/genetics , Risk Factors , Sequence Analysis, RNA , Apoptosis
4.
Medicina (Kaunas) ; 59(3)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36984424

ABSTRACT

Background and Objectives. The prognostic role of adjacent nontumor tissue in patients with breast cancer (BC) is still unclear. The activity changes in immunologic and hallmark gene sets in normal tissues adjacent to BC may play a crucial role in predicting the prognosis of BC patients. The aim of this study was to identify BC subtypes and ribosome-associated prognostic genes based on activity changes of immunologic and hallmark gene sets in tumor and adjacent nontumor tissues to improve patient prognosis. Materials and Methods. Gene set variation analysis (GSVA) was applied to assess immunoreactivity changes in the overall sample and three immune-related BC subtypes were identified by non-negative matrix factorization (NMF). KEGG (Kyoto Encyclopedia of Genes and Genomes) and GO (Gene Ontology) analyses were after determining the prognostic gene set using the least absolute shrinkage and selection operator (LASSO) method. Ribosome-related genes were identified by PPI (protein-protein interaction) analysis, and finally a prognostic risk model was constructed based on the expression of five ribosomal genes (RPS18, RPL11, PRLP1, RPL27A, and RPL38). Results. A comprehensive analysis of immune and marker genomic activity changes in normal breast tissue and BC tissue identified three immune-related BC subtypes. BC subtype 1 has the best prognosis, and subtype 3 has the worst overall survival rate. We identified a prognostic gene set in nontumor tissue by the least absolute shrinkage and selection operator (LASSO) method. We found that the results of both KEGG and GO analyses were indistinguishable from those of ribosome-associated genes. Finally, we determined that genes associated with ribosomes exhibit potential as a reliable predictor of overall survival in breast cancer patients. Conclusions. Our research provides an important guidance for the treatment of BC. After a mastectomy, the changes in gene set activity of both BC tissues and the nontumor tissues adjacent to it should be thoroughly evaluated, with special attention to changes in ribosome-related genes in the nontumor tissues.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Prognosis , Mastectomy , Ribosomes/genetics , Breast
5.
Mediators Inflamm ; 2022: 5791915, 2022.
Article in English | MEDLINE | ID: mdl-35769208

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is the common liver disease characterized by hepatic steatosis, inflammation, and fibrosis; there are no approved drugs to treat this disease because of incomplete understanding of pathophysiological mechanisms of NASH. Milk fat globule-epidermal growth factor-factor 8 (MFG-E8), a multifunctional glycoprotein, has shown anti-inflammation and antifibrosis. Here, MFG-E8 was shown to play a key role in NASH progression. Using methionine and choline deficient (MCD) diet-fed mice, we found MFG-E8 knockout exacerbated hepatic damage and steatosis as indicated by increased plasma transaminases activities and hepatic histopathologic change, higher hepatic triglycerides (TGs), and lipid accumulation. Moreover, liver fibrosis and inflammation elicited by MCD were aggravated in MFG-E8 knockout mice. Mechanistically, MFG-E8 knockout facilitated activation of hepatic toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway in MCD-fed mice. In vitro experiment, the TLR4 specific antagonist TAK-242 rescued palmitic acid- (PA-) primed lipid formation and inflammation in MFG-E8 knockout primary murine hepatocytes. These findings indicated that MFG-E8 is involved in the progression of NASH and the possible mechanism by which MFG-E8 knockout exacerbated NASH in mice is associated with activation of the TLR4/NF-κB signaling pathway.


Subject(s)
Antigens, Surface , Milk Proteins , NF-kappa B , Non-alcoholic Fatty Liver Disease , Toll-Like Receptor 4 , Animals , Antigens, Surface/metabolism , Lipid Metabolism , Methionine/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Milk Proteins/metabolism , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism
6.
Pharmacol Res ; 172: 105838, 2021 10.
Article in English | MEDLINE | ID: mdl-34425230

ABSTRACT

Accumulating evidence indicates that metabolic events profoundly modulate the progression of various diseases. Pyruvate is a central metabolic intermediate in glucose metabolism. In the present study, the metabolic status of pyruvate and its pharmacological significance has been investigated in mice with lipopolysaccharide/D-galactosamine (LPS/D-Gal)-induced fulminant liver injury. Our results indicated that LPS/D-Gal exposure decreased the activity of pyruvate kinase and the content of pyruvate, which were reversed by the PKM2 activator TEPP-46. Pretreatment with TEPP-46 or supplementation with the cell-permeable pyruvate derivate ethyl pyruvate (EP) attenuated LPS/D-Gal-induced liver damage. Interestingly, post-insult intervention of pyruvate metabolism also resulted in beneficial outcomes. The phospho-antibody microarray analysis and immunoblot analysis found that the inhibitory phosphorylation of cyclin dependent kinase 1 (CDK1) was reversed by TEPP-46, DASA-58 or EP. In addition, the therapeutic benefits of PKM2 activator or EP were blunted by the CDK1 inhibitor Ro 3306. Our data suggests that LPS/D-Gal exposure-induced decline of pyruvate might be a novel metabolic mechanism underlies the development of LPS/D-Gal-induced fulminant liver injury, PKM2 activator or pyruvate derivate might have potential value for the pharmacological intervention of fulminant liver injury.


Subject(s)
CDC2 Protein Kinase/metabolism , Liver Diseases/metabolism , Pyruvate Kinase/metabolism , Pyruvic Acid/metabolism , Animals , Apoptosis/drug effects , Galactosamine , Hepatocytes/drug effects , Lipopolysaccharides , Liver/metabolism , Liver/pathology , Liver Diseases/etiology , Liver Diseases/pathology , Male , Mice , Mice, Inbred BALB C , Pyridazines/pharmacology , Pyrroles/pharmacology , Pyruvates/pharmacology
7.
Adv Exp Med Biol ; 1316: 117-131, 2021.
Article in English | MEDLINE | ID: mdl-33740247

ABSTRACT

Tumor- or cancer-associated fibroblasts (TAFs), one of the most abundant stromal cell types in various carcinomas, consist of a heterogeneous cell population. Typically, TAFs are assigned with pro-tumor activities to promote tumor growth and progression. One of the key features of solid tumors is the metabolic reprogramming that induces alterations of bioenergetics and biosynthesis in both tumor cells and TAFs. Therefore, this review emphasizes TAFs lipid metabolism related to both TAFs differentiation process and TAFs crosstalk with cancer cells. We hope that this review will help understand lipid metabolism in tumor microenvironment, and support the rational design of metabolism-based approaches to improve the efficacy of cancer therapy.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms , Fibroblasts , Humans , Lipid Metabolism , Stromal Cells , Tumor Microenvironment
8.
Drug Dev Res ; 82(8): 1111-1123, 2021 12.
Article in English | MEDLINE | ID: mdl-33733518

ABSTRACT

CQMUH-011 is a modified adamantane sulfonamide compound, that inhibits macrophage proliferation and possesses anti-inflammatory properties. Here, fresh mouse splenocytes were obtained and stimulated with concanavalin A (ConA, 5 µg/ml) in vitro; and experimental autoimmune hepatitis (AIH) was induced by ConA (20 mg/kg, iv) in vivo, to clarify the protective effects of CQMUH-011 against AIH and its possible mechanisms. Our results demonstrated that CQMUH-011 pretreatment can dose-dependently inhibit the proliferation of splenocytes in vitro. In vivo, CQMUH-011 administration reduced the hepatic histopathological score and the infiltration of lymphocytes in the liver parenchyma; additionally, it downregulated the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and pro-inflammatory cytokines interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and interleukin (IL)-6 in serum, as well as those of methane dicarboxylic aldehyde and myeloperoxidase in the liver tissues. It also down-regulated the expression of p-NF-κB and related proteins in the liver tissues. Furthermore, CQMUH-011 could maintain the balance of CD3+ CD4+ /CD3+ CD8+ and decrease the percentages of CD8+ CD69+ and CD4+ CD25+/- CD69+ T-cells in the splenocytes of ConA-challenged mice. Moreover, we found thatCD4+ CD25+/- CD69+ T-cells were significantly correlated with ALT levels, especially CD4+ CD25- CD69+ T-cells. In conclusion, CQMUH-011 exerts potential protective effects against ConA-induced hepatitis, which may be partially attributed to its inhibition of T cells, especially the suppression of the proliferation of CD4+ CD25- CD69+ and CD8+ CD69+ subsets in the spleen. CQMUH-011 also reduced the early apoptosis of lymphocytes in the thymus.


Subject(s)
Adamantane , Anti-Inflammatory Agents , Hepatitis, Autoimmune , Sulfonamides , T-Lymphocytes , Animals , Female , Mice , Adamantane/analogs & derivatives , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cytokines/blood , Germinal Center/drug effects , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/immunology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Mice, Inbred BALB C , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
9.
Biomed Chromatogr ; 34(3): e4767, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31785600

ABSTRACT

Disorders of certain branched-chain amino acids may be associated with the occurrence and development of non-alcoholic fatty liver disease. Measurement of related branched-chain amino acid levels could provide a reference for the clinical and scientific research of the non-alcoholic fatty liver disease. An established HPLC-FLD method was used to quantify aspartic acid, glutamate, glutamine, glycine, taurine, tyrosine, 4-amino butanoic acid, tryptophan, methionine, valine, phenylalanine, isoleucine and leucine in mouse brain tissue. Brain tissue samples mixed with internal standard (3-aminobutyric acid) were processed, then derivatized with 2-O-phthaldialdehyde, and finally separated on an ODS2 column through gradient elution at a flow rate of 1.0 ml·min-1 . The excitation and emission wavelengths were set at 340 and 455 nm, respectively. The mobile phase A was 100% methanol and the mobile phase B consisted of 30 mmol·L-1 sodium acetate (pH 6.8). The injection volume was 20 µl and the single run time was 45 min. Several parameters, accuracy, precision, and stability, were verified and the results showed the established method had good sensitivity and resolution for all of the 13 compounds and internal standard in mouse brain.


Subject(s)
Amino Acids/analysis , Aminobutyrates/analysis , Brain/metabolism , Chromatography, High Pressure Liquid/methods , Non-alcoholic Fatty Liver Disease/metabolism , Amino Acids/metabolism , Animals , Brain Chemistry/physiology , Limit of Detection , Linear Models , Male , Mice , Mice, Inbred C57BL , Reproducibility of Results , Spectrometry, Fluorescence
10.
Exp Dermatol ; 28(1): 45-52, 2019 01.
Article in English | MEDLINE | ID: mdl-30372793

ABSTRACT

Cutaneous squamous cell carcinoma (SCC) is one of the most common non-melanoma skin cancers worldwide. While its exact tumorigenesis mechanisms is far from well-established and less satisfied therapeutic strategy can be clinically used nowadays. In this study, we intended to investigate the role of DNA damage-inducible transcript 4 (DDIT4) in human SCC. Firstly, we identified DDIT4 is significantly suppressed in human SCC tissue and cultured A431 cell line, and reduced DDIT4 accelerates keratinocytes proliferation but impedes the autophagy flux through mTORC1 pathway by affecting the downstream S6 Kinase1, 4E-BP1, Beclin1 and LC3 II/I. While 1,25(OH)2 D3 enhanced DDIT4 expression and activated autophagy and inhibit mTORC1 to take the effect of anti-proliferation and activating autophagy. Further, formation of direct vitamin D receptor (VDR)-DDIT4 transcription complex was verified by ChIP-qPCR, which showed the molecular mechanism of how 1,25(OH)2 D3 promotes DDIT4 transcription. Thirdly, xenograft tumor-bearing mice model treated by gradient concentrations of 1,25(OH)2 D3 revealed the obvious anti-carcinoma effect of 1,25(OH)2 D3 in vivo and DDIT4 acted the molecular vector of 1,25(OH)2 D3 through mTORC1. Lastly, elevated DDIT4 expression was verified in human actinic keratoses tissue, and chronic long-term ultraviolet (UV) irradiation on mouse disclosed UV could promote DDIT4 expression inside epidermis. Conclusively, our research suggested a novel molecular mechanism about the human SCC tumorigenesis and the pharmacological mechanism about how 1,25(OH)2 D3 take its anti-carcinoma role on human SCC, as well as a striking paradoxes that how UV irradiation plays the tumorigenesis effect but synchronously take a protective role in the early stage of SCC carcinogenesis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Calcitriol/pharmacology , Carcinoma, Squamous Cell/metabolism , Transcription Factors/metabolism , Animals , Autophagy , Cell Line, Tumor , Cell Proliferation , Cholecalciferol/metabolism , DNA Damage , Female , Humans , Keratinocytes/cytology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Receptors, Calcitriol/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Ultraviolet Rays
11.
Immunopharmacol Immunotoxicol ; 41(3): 438-445, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31119954

ABSTRACT

Objective: Paeonol is a natural phenolic component isolated from the root bark of peony with multiple pharmacological activities. We investigated the anti-fibrotic effect and underlying mechanism of paeonol. Methods: Twenty-four male C57BL/6J mice were divided into 4 groups (n = 6 in each group), injected with CCl4 to induce liver fibrosis and administrated with paeonol according to the regimen. The serum activity of ALT and AST, and H&E staining were to assess liver injury. Sirius and Masson staining, and hydroxyproline content were to evaluate the degree of liver fibrosis. TNF-α, IL-6, TGF-ß, MDA, GSH-PX, SOD, and CAT were detected to reflect inflammation and oxidative stress. RT-qPCR and Western blot analysis to assess the activation of HSCs and TGF-ß/Smad3 signaling. Results: Paeonol ameliorated liver injury and liver fibrosis, reflected by the decrease of ALT, AST, less lesion in H&E staining, mitigated fibrosis in Sirius and Masson staining, lessened content of hydroxyproline. Paeonol attenuated the level of IL-6 and TNF-α, and elevated the activity of GSH-PX, SOD, and CAT with reducing the level of MDA. The expression of col 1a, α-SMA, vimentin, and desmin were down-regulated and TGF-ß/Smad3 signaling pathway was inhibited. Conclusion: These data demonstrated that paeonol could alleviate CCl4-induced liver fibrosis through suppression of hepatic stellate cells activation via inhibiting the TGF-ß/Smad3 signaling.


Subject(s)
Acetophenones/pharmacology , Carbon Tetrachloride Poisoning/drug therapy , Hepatic Stellate Cells/immunology , Liver Cirrhosis/drug therapy , Signal Transduction/drug effects , Smad3 Protein/immunology , Transforming Growth Factor beta/immunology , Animals , Carbon Tetrachloride Poisoning/immunology , Carbon Tetrachloride Poisoning/pathology , Hepatic Stellate Cells/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Male , Mice , Signal Transduction/immunology
12.
Phytother Res ; 32(11): 2247-2255, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30047580

ABSTRACT

Hepatic ischemia/reperfusion (I/R) injury is a major cause of high morbidity and mortality after liver resection, transplantation, and hemorrhagic shock. Paeoniflorin (PF), the main substance of glucosides in Radix Paeoniae Alba, has been widely used to treat various hepatic inflammatory diseases including I/R injury. However, the underlying mechanisms of PF on hepatic I/R injury remain further investigated. In this study, the liver I/R model was performed by clamping the portal vein and hepatic artery with an atraumatic clamp for 90 min followed by 6 hr reperfusion. PF (100 mg/kg) was given three times a day by gavage before I/R. The blood and hepatic samples were collected to evaluate liver injury and molecular indexes. The results showed that PF pretreatment significantly inhibited I/R-induced serum ALT and AST activities (40.3% and 53.8% those of I/R group, respectively), hepatic pathological damages and hepatic apoptosis (P < 0.01), and infiltration of neutrophils into liver. In addition, PF suppressed the production of pro-inflammatory cytokines (P < 0.01), decreased the expression of high mobility group box-1 (HMGB1), and down-regulated toll-like receptors 4 (TLR4) and phosphorylated ERK1/2, JNK1/2, p38, and NF-κB signal molecules expression in the I/R-operated mice. These findings indicated that PF played a protective role in liver I/R injury, and this protection was associated with inhibition of I/R-activated HMGB1-TLR4 signaling pathway to attenuate hepatic inflammation responses.


Subject(s)
Glucosides/pharmacology , HMGB1 Protein/antagonists & inhibitors , Liver/drug effects , Monoterpenes/pharmacology , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Apoptosis , Caspase 3/metabolism , Down-Regulation , Interleukin-1beta/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Paeonia/chemistry , Protective Agents/pharmacology , Reperfusion Injury/prevention & control , Tumor Necrosis Factor-alpha/metabolism
13.
Toxicol Appl Pharmacol ; 320: 8-16, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28213090

ABSTRACT

Glycyrrhetinic acid (GA), the main active ingredient of licorice, reportedly has anti-inflammatory and hepatoprotective properties, but its molecular mechanisms remain be elusive. In the present study, Balb/c mice were pretreated with GA (10, 30, or 100mg/kg) 1h before lipopolysaccharide (LPS)/d-galactosamine (D-GalN) administration. In other in vitro experiment, RAW264.7 macrophages were pretreated with GA before LPS exposure. The mortality, hepatic tissue histology, serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were analyzed. Toll like receptor 4 (TLR4), interleukin-1 receptor-associated kinases (IRAKs), activation of mitogen-activated protein kinases (MAPKs) and NF-κB, and production of TNF-α were assessed by flow cytometry, western blotting, and enzyme-linked immunosorbent assay (ELISA), respectively. Our results showed that pretreatment with GA protected mice against LPS/D-GalN-induced fulminant hepatic failure (FHF), including a dose-dependent alleviation of mortality and ALT/AST elevation, ameliorating hepatic pathological damage, and decreasing TNF-α release. Moreover, GA inhibited LPS-induced activation of MAPKs and NF-κB in response to LPS, but the expression of TLR4 was not affected in vivo and in vitro. Notably, GA pretreatment in vivo suppressed IRAK-1 activity while inducing IRAK-M expression. Silencing of IRAK-M expression with siRNA blocked these beneficial effects of GA on the activation of MAPKs and NF-κB as well as TNF-α production in LPS-primed macrophages. Taken together, we conclude that GA could prevent LPS/D-GalN-induced FHF. The underlying mechanisms may be related to up-regulation of IRAK-M, which in turn caused deactivation of IRAK-1 and subsequent MAPKs and NF-κB, resulting in inhibiting TNF-α production.


Subject(s)
Galactosamine/toxicity , Glycyrrhetinic Acid/therapeutic use , Interleukin-1 Receptor-Associated Kinases/biosynthesis , Lipopolysaccharides/toxicity , Liver Failure, Acute/metabolism , Liver Failure, Acute/prevention & control , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Line , Dose-Response Relationship, Drug , Galactosamine/antagonists & inhibitors , Glycyrrhetinic Acid/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Liver Failure, Acute/chemically induced , Male , Mice , Mice, Inbred BALB C , Up-Regulation/drug effects , Up-Regulation/physiology
14.
Immunopharmacol Immunotoxicol ; 38(2): 61-7, 2016.
Article in English | MEDLINE | ID: mdl-26630551

ABSTRACT

Acute hepatic failure involves in excessive oxidative stress and inflammatory responses, leading to a high mortality due to lacking effective therapy. Resolvin D1 (RvD1), an endogenous lipid mediator derived from polyunsaturated fatty acids, has been shown anti-inflammatory and anti-oxidative actions, however, whether RvD1 has protective effects on hepatic failure remains elusive. In this study, the roles and molecular mechanisms of RvD1 were explored in carbon tetrachloride (CCl4)-induced acute liver injury. Our results showed that RvD1 protected mice against CCl4-induced hepatic damage, as evaluated by reduced aminotransferase activities and malondialdehyde content, elevated glutathione and superoxide dismutase activities, and alleviated hepatic pathological damage. Moreover, RvD1 significantly attenuated serum tumor necrosis factor-α and interleukin-6 levels as well as hepatic myeloperoxidase activity, whereas enhanced serum IL-10 level in CCl4-administered mice. Further, RvD1 markedly up-regulated the expression and activity of heme oxygenase-1 (HO-1). However, inhibition of HO-1 activity reversed the protective effects of RvD1 on CCl4-induced liver injury. These results suggest that RvD1 could effectively prevent CCl4-induced liver injury by inhibition of oxidative stress and inflammation, and the underlying mechanism may be related to up-regulation of HO-1.


Subject(s)
Carbon Tetrachloride Poisoning , Docosahexaenoic Acids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/biosynthesis , Liver Failure, Acute , Membrane Proteins/biosynthesis , Up-Regulation/drug effects , Animals , Carbon Tetrachloride Poisoning/enzymology , Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/prevention & control , Cytokines/biosynthesis , Liver Failure, Acute/enzymology , Liver Failure, Acute/pathology , Liver Failure, Acute/prevention & control , Male , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects
15.
Biochem Biophys Res Commun ; 461(2): 230-6, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25866179

ABSTRACT

Sesamin has been described to exert anti-oxidant and anti-inflammatory properties. In present study, we investigated the potential effects and mechanisms of sesamin on lipopolysaccharide (LPS)-induced fulminant hepatic failure (FHF) in d-galactosamine (D-GalN)-sensitized mice. Our results showed that pretreatment with sesamin dose-dependently improved LPS/D-GalN-induced mortality and liver injury as indicated by reduced serum levels of aminotransferases and alleviated pathological damage as well as hepatocyte apoptosis in mice. Additionally, sesamin markedly attenuated LPS/D-GalN-induced adhesion molecules expression, and decreased neutrophils recruitment. Furthermore, sesamin inhibited LPS-induced tumor necrosis factor-alpha (TNF-α) production, p38 mitogen-activated protein kinases (MAPK) and NF-κB activation, and Toll like receptor (TLR) 4 expression in mice and in RAW264.7 macrophage cells. In summary, these results demonstrate that sesamin protects mice from LPS-induced FHF and the molecular mechanisms may down-regulate the expression of TLR4, block MAPK and NF-κB activation, decrease the production of TNF-α.


Subject(s)
Dioxoles/therapeutic use , Lignans/therapeutic use , Liver Failure, Acute/prevention & control , Liver/drug effects , Liver/pathology , Protective Agents/therapeutic use , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Antioxidants/therapeutic use , Cell Line , Galactosamine , Lipopolysaccharides , Liver/immunology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/immunology , Liver Failure, Acute/pathology , Male , Mice , Mice, Inbred BALB C , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/immunology
16.
Biochem Biophys Res Commun ; 459(3): 541-6, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25749337

ABSTRACT

The glycolytic inhibitor 2-deoxyglucose (2-DG) is a calorie restriction (CR) mimetic produces CR-like beneficial effects in both acute and chronic pathological processes, but whether 2-DG is also helpful in critical and life-threatening situation is not known. In the present study, the potential benefits of 2-DG in lipopolysaccharide/D-galactosamine (LPS/D-Gal)-induced lethal liver injury were investigated. The results indicated that treatment with 2-DG suppressed the elevation of plasma aminotransferases, alleviated the histopathological abnormalities and improved the survival rate of LPS/D-Gal-exposed mice. Treatment with 2-DG also suppressed the production of pro-apoptotic cytokine TNF-α, the phosphorylation of JNK, the activation of caspase cascade and the count of TUNEL-positive apoptotic hepatocytes. These data suggested that the CR mimetic 2-DG could also provide beneficial effects in lethal pathological process such as LPS/D-Gal-induced fulminant liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Deoxyglucose/pharmacology , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Biomimetics , Caloric Restriction , Caspases/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Deoxyglucose/administration & dosage , Galactosamine/toxicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/biosynthesis
17.
Crit Care ; 18(5): 517, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25212718

ABSTRACT

INTRODUCTION: Sodium bicarbonate (SBIC) was reported to be a promising approach to prevent cardiac surgery-associated acute kidney injury (CSA-AKI). However, the results remain controversial. We conducted a systematic review and meta-analysis to evaluate the efficacy and safety of SBIC on the prevention of CSA-AKI in adult patients undergoing cardiac surgery. METHODS: PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases were systematically searched. Randomized controlled trials (RCTs) assessing the effect of SBIC versus placebo on the prevention of CSA-AKI in adult patients undergoing cardiac surgery were included. Two investigators independently searched articles, extracted data, and assessed the quality of included studies. The primary outcome was the incidence of CSA-AKI. Meta-analysis was performed using random-effects models. RESULTS: Five RCTs involving 1079 patients were included in the meta-analysis. Overall, compared with placebo, SBIC was not associated with a reduced risk of CSA-AKI (relative risk [RR] 0.99; 95% confidence interval [CI] 0.78 to 1.24; P = 0.911). SBIC failed to alter the clinical outcomes of hospital length of stay (weighted mean difference [WMD] 0.23 days; 95%CI -0.88 to 1.33 days; P = 0.688), renal replacement therapy (RR 0.94; 95%CI 0.49 to 1.82; P = 0.861), hospital mortality (RR 1.37; 95%CI 0.46 to 4.13; P = 0.572), postoperative atrial fibrillation (RR 1.02; 95%CI 0.65 to 1.61; P = 0.915). However, SBIC was associated with significant increased risks in longer duration of ventilation (WMD 0.64 hours; 95%CI 0.16 to 1.11 hours; P = 0.008), longer ICU length of stay (WMD 2.06 days; 95%CI 0.54 to 3.58 days; P = 0.008), and increased incidence of alkalemia (RR 2.21; 95%CI 1.42 to 3.42; P <0.001). CONCLUSIONS: SBIC could not reduce the incidence of CSA-AKI. Contrarily, SBIC prolongs the duration of ventilation and ICU length of stay, and increases the risk of alkalemia. Thus, SBIC should not be recommended for the prevention of CSA-AKI and perioperative SBIC infusion should be administrated with caution.


Subject(s)
Acute Kidney Injury/prevention & control , Cardiac Surgical Procedures , Sodium Bicarbonate/therapeutic use , Adult , Atrial Fibrillation , Humans , Length of Stay , Postoperative Complications , Randomized Controlled Trials as Topic , Renal Replacement Therapy
18.
Hepatol Res ; 44(11): 1151-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23941578

ABSTRACT

AIM: Lipopolysaccharide (LPS)-induced liver injury in D-galactosamine (D-Gal)-sensitized mice is a well-established animal model widely used in exploring the pathogenesis of fulminant hepatitis. Increasing evidence has indicated that reactive oxygen species (ROS)-induced oxidative injury may be involved in LPS/D-Gal-induced hepatitis. Catalase (CAT) is a major antioxidant enzyme while aminotriazole (ATZ) is commonly used as a CAT inhibitor. In the present study, the effects of ATZ on LPS/D-Gal-induced liver injury were investigated. METHODS: Fuliminant liver injury was induced by intraperitoneal injection of LPS combined with D-Gal, ATZ was administrated 0.5 h prior to LPS/D-Gal challenge. The degree of liver injury, the level of hepatic oxidative stress, the grade of hepatic apoptosis and the survival of experimental animals were determined. RESULTS: Our experimental data showed that treatment with ATZ significantly enhanced LPS/D-Gal-induced elevation of serum aspartate transaminase (AST) and alanine transaminase (ALT), exacerbated the hepatic histopathological abnormality and decreased the survival rate of experimental animals. ATZ inhibited the activity of CAT, increased the content of H2 O2 and the levels of malondialdehyde (MDA) in liver tissues. In addition, treatment with ATZ also enhanced LPS/D-Gal-induced hepatic apoptosis as evidenced by increased caspases activities in liver tissues and increased number of terminal deoxynucleotidyl transferase dUTP nick end labeling positive cells in liver sections. CONCLUSION: These findings suggested that CAT might be involved in the progression of LPS/D-Gal-induced fulminant liver injury.

19.
J Gastroenterol Hepatol ; 29(6): 1308-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24476051

ABSTRACT

BACKGROUND AND AIM: Fulminant hepatic failure (FHF) is a serious clinic syndrome with extremely poor prognosis and no effective treatment except for liver transplantation. Synthetic RGDS peptide, an inhibitor of integrins, was proved to suppress integrin signals. In this study, we investigated the protection effects of RGDS peptide on lipopolysaccharide/D-galactosamine (LPS/D-GalN)-induced FHF and the underlying molecular mechanisms. METHODS: Synthetic RGDS peptide was given intraperitoneally 30 min before LPS/D-GalN injection. Liver function and the extent of liver injury were analyzed biochemically and pathologically respectively. Enzyme-linked immunosorbent assay, real-time polymerase chain reaction and Western blotting were used to detect effectors and signaling molecules. RESULTS: Pretreatment with synthetic RGDS peptide significantly improved LPS/D-GalN-induced mortality, and liver injury as determined by alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, as well as pathological analysis. In addition, RGDS peptide significantly reduced tumor necrosis factor (TNF)-α and macrophage inflammatory protein (MIP)-2 production, and decreased myeloperoxidase (MPO) and NF-κB activity. Furthermore, Western blotting indicated that the levels of phospho-integrin ß3, phospho-focal adhesion kinase (FAK) and phospho-p38 mitogen-activated protein kinases (MAPK) decreased with RGDS peptide pretreatment. CONCLUSION: Together, these data suggest that synthetic RGDS peptide protect against LPS/D-GalN-induced FHF by inhibiting inflammatory cells migration and blocking the integrin αVß3-FAK-p38 MAPK and NF-κB signaling.


Subject(s)
Galactosamine , Lipopolysaccharides , Liver Failure, Acute/chemically induced , Liver Failure, Acute/drug therapy , Oligopeptides/administration & dosage , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Biomarkers/metabolism , Chemokine CXCL2/metabolism , Focal Adhesion Kinase 1/metabolism , Injections, Intraperitoneal , Integrins/antagonists & inhibitors , Liver Failure, Acute/diagnosis , Liver Failure, Acute/metabolism , Mice, Inbred BALB C , NF-kappa B/metabolism , Oligopeptides/pharmacology , Peroxidase/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
20.
J Appl Toxicol ; 34(3): 265-71, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23620140

ABSTRACT

The prognosis for fulminant hepatic failure (FHF) still remains extremely poor with a high mortality and, therefore, better treatments are urgently needed. Syringin, a main active substance isolated from Eleutherococcus senticosus, has been reported to exhibit immunomodulatory and anti-inflammatory properties. In this study, we investigated the effects and underlying mechanisms of syringin on lipopolysaccharide (LPS) and D-galactosamine (D-GalN)-induced FHF in mice. Mice were administered syringin (10, 30 and 100 mg kg(-1), respectively) intraperitoneally (i.p) 30 min before LPS/D-GalN then mortality and liver injury were evaluated subsequently. We found that syringin dose-dependently attenuated LPS/D-GalN-induced FHF, as indicated by reduced mortality, inhibited aminotransferase and malondialdehyde (MDA) content, an increased glutathione (GSH) concentration and alleviated pathological liver injury. In addition, syringin inhibited LPS/D-GalN-induced hepatic caspase-3 activation and hepatocellular apoptosis, myeloperoxidase (MPO) activity and intercellular adhesion molecule-1 (ICAM-1) expression, as well as hepatic tissues tumor necrosis factor-alpha (TNF-α) production and NF-κB activation in a dose-dependent manner. These experimental data indicate that syringin might alleviate the FHF induced by LPS/D-GalN through inhibiting NF-κB activation to reduce TNF-α production.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Galactosamine/toxicity , Glucosides/therapeutic use , Lipopolysaccharides/toxicity , Liver Failure, Acute/prevention & control , Phenylpropionates/therapeutic use , Protective Agents/therapeutic use , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Eleutherococcus/chemistry , Glucosides/administration & dosage , Glucosides/isolation & purification , Liver Failure, Acute/etiology , Liver Failure, Acute/pathology , Liver Function Tests , Mice , Mice, Inbred BALB C , Phenylpropionates/administration & dosage , Phenylpropionates/isolation & purification , Protective Agents/administration & dosage , Protective Agents/isolation & purification , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL