Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nutr Diabetes ; 14(1): 45, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886355

ABSTRACT

BACKGROUND/OBJECTIVES: Increased free fatty acid (FFA) promotes adiponectin secretion in healthy subjects and induces inflammation in diabetes. Given the potential pro-inflammatory role of adiponectin in "adiponectin paradox", we performed this study in patients with type 2 diabetes mellitus (T2DM) to assess the association of FFA with adiponectin and to investigate whether adiponectin mediates FFA-related inflammation. METHODS: This cross-sectional study consisted of adult patients with T2DM. FFA, adiponectin, and tumor necrosis factor-α (TNF-α) were assayed from fasting venous blood after overnight fasting for at least 8 h. Multivariable linear regression analysis and restricted cubic splines (RCS) analysis were performed to identify the association between FFA and adiponectin. Mediation analysis was performed to determine the mediating effect of adiponectin on the association between FFA and TNF-α. RESULTS: This study included 495 participants, with 332 males (67.1%) and a mean age of 47.0 ± 11.2 years. FFA was positively associated with adiponectin (b = 0.126, 95%CI: 0.036-0.215, P = 0.006) and was the main contributor to the increase of adiponectin (standardized b = 0.141). The RCS analysis demonstrated that adiponectin increased with FFA when FFA was less than 0.7 mmol/L but did not further increase thereafter (Poverall < 0.001 and Pnon-linear < 0.001). In addition, adiponectin mediated the association between FFA and TNF-α. The mediating effect was 0.08 (95%CI: 0.03-0.13, P = 0.003) and the mediating effect percentage was 26.8% (95%CI: 4.5-49.2, P = 0.02). CONCLUSIONS: In patients with T2DM, FFA was positively associated with adiponectin when FFA was less than 0.7 mmol/L. Elevated adiponectin mediated FFA-related inflammation. This study may provide insights into the pro-inflammatory effect of adiponectin in T2DM.


Subject(s)
Adiponectin , Diabetes Mellitus, Type 2 , Fatty Acids, Nonesterified , Tumor Necrosis Factor-alpha , Humans , Adiponectin/blood , Male , Fatty Acids, Nonesterified/blood , Female , Middle Aged , Tumor Necrosis Factor-alpha/blood , Cross-Sectional Studies , Diabetes Mellitus, Type 2/blood , Adult , Inflammation/blood
2.
Comput Biol Med ; 157: 106777, 2023 05.
Article in English | MEDLINE | ID: mdl-36924737

ABSTRACT

BACKGROUND: This study aims to evaluate the efficacy and therapeutic mechanism of bufalin on lung adenocarcinoma (LUAD) through a comprehensive strategy integrating network pharmacology, metabolomics and molecular biology verification. METHODS: The putative targets of bufalin were discerned from PharmMapper and Swiss Target Prediction database. LUAD-related targets were obtained by target filtering of GeneCard database and data mining of GEO database. PPI network was constructed to screen the core targets, and their clinical significance was assessed through several public databases. GO and KEGG pathway analyses were performed to identify possible enrichment of genes with specific biological themes. Molecular docking and molecular dynamics (MD) simulation were employed to determine the correlation and binding pattern between bufalin and core targets. The potential mechanisms of bufalin acting on LUAD, as predicted by network pharmacology analyses, were experimentally validated using in-vitro and in-vivo models. Finally, the effects of bufalin intervention on metabolite profile and metabolic pathway in LUAD nude mice were investigated by non-targeted metabolomics. RESULTS: 209 bufalin targets and 1082 LUAD-associated targets were harvested, of which 51 intersection targets were identified. 10 core targets including Akt1, STAT3, EGFR, CASP3 and SRC were picked out through network topology analysis, and they had a potent binding activity with bufalin as indicated by molecular docking and MD simulation. Hub module of PPI network was closely related to cell proliferation and apoptosis. GO and KEGG enrichment analyses suggested that bufalin exerted therapeutic effects on LUAD possibly by inhibiting proliferation and promoting apoptosis via PI3K/Akt, FoxO1 and MAPK/ERK pathways, which were confirmed by a series of in-vitro studies as well as HE, TUNEL and Ki-67 staining of tumor tissues. Further metabolomics analysis revealed that bufalin mainly regulated ABC transporter and remodeled AA metabolism, thereby contributing to the treatment of LUAD. CONCLUSION: From molecular and metabolic perspective, the present study not only provided a unique insight into the possible mechanisms of bufalin against LUAD after successfully filtering out associated key target genes, differential endogenous metabolites, and signaling pathways, but also proposed a novel promising therapeutic strategy for LUAD.


Subject(s)
Adenocarcinoma of Lung , Drugs, Chinese Herbal , Lung Neoplasms , Animals , Mice , Mice, Nude , Molecular Docking Simulation , Network Pharmacology , Phosphatidylinositol 3-Kinases , Molecular Biology , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics
3.
J Ethnopharmacol ; 282: 114672, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-34560213

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Presently, insulin resistance has been a growing concern that urgently needs to be addressed, because it not only places patients at risk of developing type 2 diabetes mellitus but also results in metabolic syndrome and different aspects of cardiovascular diseases. Shenqi Jiangtang Granule (SJG) is a classic traditional Chinese medicine (TCM) prescription that is widely used to treat diabetes mellitus and its complications in clinical practice. While studies have revealed that SJG with multi-ingredients and multi-targets characteristics possesses potential anti-insulin resistance pharmacological properties, its mechanisms of action and molecular targets for the treatment of insulin resistance are still obscure, which prompt us to conduct an in-depth research. AIM OF THE STUDY: This study was purposed to uncover the pharmacological mechanism of SJG against insulin resistance through integrating network pharmacology and experimental validation. MATERIALS AND METHODS: The putative ingredients of SJG and its related targets were discerned from the TCMSP database. Subsequently, insulin resistance-associated targets were retrieved from GeneCard, OMIM, and GEO database. Compound-target, protein-protein interaction (PPI), and compound-target-pathway networks were established using Cytoscape software. GO and KEGG pathway analyses were performed to identify possible enrichment of genes with specific biological themes. Molecular docking was used to verify the correlation between the main active ingredients and hub targets. Optimal docking conformation was further analyzed by molecular dynamics (MD) simulation. Finally, the potential molecular mechanisms of SJG acting on insulin resistance, as predicted by the network pharmacology analyses, were validated experimentally in insulin-resistant rat model. RESULTS: 136 active compounds, 211 corresponding targets in addition to 1463 disease-related targets were collected, of which 94 intersection targets were obtained. 29 key targets including AKT1, VEGFA, IL-6, CASP3, and PTGS2 were identified through PPI network analysis. Hub module of PPI network was closely associated with inflammation. GO and KEGG analyses also revealed that inflammation-related pathways may be a central factor for SJG to modulate insulin resistance. Molecular docking test showed a good binding potency between primary active ingredients and core targets, and the binding mode of optimal docking conformation was stable in MD simulation. A rat model of insulin resistance was successfully induced by chronic high-fat diet (HFD) consumption. Through a series of in vivo studies, including HEC, ITT, and HOMA-IR measurement, it was revealed that SJG exhibited a beneficial effect on ameliorating insulin resistance, as demonstrated by a significant increase of GIR and a significant decrease of AUCITT and HOMA-IR index value. Further molecular biological analysis showed that SJG can decrease the mRNA expression level and serum concentration of inflammatory cytokines (TNF-α, IL-6, and IL-1ß), along with suppressing the p-NFκB protein overexpression, indicating its anti-inflammatory activity. Also, it can contribute to the reversal of the impaired hepatic insulin signaling pathway, as evidenced by up-regulated protein expression of p-Akt and GLUT2. CONCLUSIONS: Through in silico and in vivo approaches, the present study not only provides a unique insight into the possible mechanism of SJG in insulin resistance after successfully filtering out associated key target genes and signaling pathways, but also suggests a novel promising therapeutic strategy for curing insulin resistance.


Subject(s)
Computer Simulation , Drugs, Chinese Herbal , Insulin Resistance , Animals , Rats , Blood Glucose/drug effects , Body Weight , Drugs, Chinese Herbal/pharmacology , Glucose Clamp Technique , Insulin/blood , Models, Molecular , Molecular Docking Simulation , Network Pharmacology , Protein Conformation , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Acta Pharmacol Sin ; 42(5): 735-743, 2021 May.
Article in English | MEDLINE | ID: mdl-32770172

ABSTRACT

Insulin resistance (IR) is a major metabolic risk factor even before the onset of hyperglycemia. Recently, berberine (BBR) is found to improve hyperglycemia and IR. In this study, we investigated whether BBR could improve IR independent of hyperglycemia. Acute insulin-resistant state was induced in rats by systemic infusion of intralipid (6.6%). BBR was administered via different delivery routes before or after the beginning of a 2-h euglycemic-hyperinsulinemic clamp. At the end of experiment, rats were sacrificed, gastrocnemius muscle was collected for detecting mitochondrial swelling, phosphorylation of Akt and AMPK, as well as the mitochondrial permeability regulator cyclophilin D (CypD) protein expression. We showed that BBR administration markedly ameliorated intralipid-induced IR without affecting blood glucose, which was accompanied by alleviated mitochondrial swelling in skeletal muscle. We used human skeletal muscle cells (HSMCs), AML12 hepatocytes, human umbilical vein endothelial cells, and CypD knockout mice to investigate metabolic and molecular alternations. In either HSMCs or AML12 hepatocytes, BBR (5 µM) abolished palmitate acid (PA)-induced increase of CypD protein levels. In CypD-deficient mice, intralipid-induced IR was greatly attenuated and the beneficial effect of BBR was diminished. Furthermore, we demonstrated that the inhibitory effect of BBR on intralipid-induced IR was mainly mediated by skeletal muscle, but not by intestine, liver, or microvasculature; BBR administration suppressed intralipid-induced upregulation of CypD expression in skeletal muscle. These results suggest that BBR alleviates intralipid-induced IR, which is related to the inhibition of CypD protein expression in skeletal muscle.


Subject(s)
Berberine/therapeutic use , Hyperinsulinism/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin Resistance/physiology , Animals , Cell Line , Cyclophilins/metabolism , Emulsions , Humans , Hyperinsulinism/chemically induced , Hyperinsulinism/metabolism , Male , Mice , Muscle Cells/drug effects , Muscle Cells/metabolism , Phospholipids , Rats, Sprague-Dawley , Soybean Oil
5.
J Endocrinol ; 246(1): 41-55, 2020 07.
Article in English | MEDLINE | ID: mdl-32420901

ABSTRACT

Subclinical hyperthyroidism, a condition characterized by decreased thyroid-stimulating hormone (TSH) and normal concentration of thyroid hormone, is associated with an elevated risk for cognitive impairment. TSH is the major endogenous ligand of the TSH receptor (TSHR) and its role is dependent on signal transduction of TSHR. It has not, however, been established whether TSHR signaling is involved in the regulation of cognition. Here, we utilized Tshr knockout mice and found that Tshr deletion led to significantly compromised performance in learning and memory tests. Reduced dendritic spine density and excitatory synaptic density as well as altered synaptic structure in CA1 subfield of the hippocampus were also noted. Furthermore, the synapse-related gene expression was altered in the hippocampus of Tshr -/- mice. These findings suggest that TSHR signaling deficiency impairs spatial learning and memory, which discloses a novel role of TSHR signaling in brain function.


Subject(s)
Memory/physiology , Receptors, Thyrotropin/metabolism , Spatial Learning/physiology , Animals , Brain/metabolism , Brain/physiology , Cognition/physiology , Hippocampus/metabolism , Mice , Receptors, Thyrotropin/genetics
6.
Nutr Metab (Lond) ; 15: 86, 2018.
Article in English | MEDLINE | ID: mdl-30555521

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a chronic and progressive liver disease with an increased risk of morbidity and mortality. However, so far no specific pharmacotherapy has been approved. Gynostemma pentaphylla (Thunb.) Makino (GP) is a traditional Chinese medicine that is widely used against hyperlipemia as well as hyperglycemia. This study aims to evaluate the effect of GP on NAFLD and explore the possible mechanism. METHODS: High-fat-diet induced NAFLD mice model were orally administrated with GP at dose of 11.7 g/kg or equivalent volume of distilled water once a day for 16 weeks. Body weight, food intake and energy expenditure were assessed to evaluate the general condition of mice. The triglycerides, total cholesterol content in the liver and liver histopathology, serum lipid profile and serum insulin level, fecal microbiome, hepatic microRNAs and relative target genes were analyzed. RESULTS: Mice in GP treatment group displayed improved hepatic triglycerides content with lower lipid droplet in hepatocyte and NAFLD activity score. Besides, GP treatment altered the composition of gut microbiota and the relative abundance of some of the key components that are implicated in metabolic disorders, especially phylum Firmicutes (Eubacterium, Blautia, Clostridium and Lactobacillus). Several hepatic microRNAs were downregulated by GP treatment such as miR-130a, miR-34a, miR-29a, miR-199a, among which the expression miR-34a was altered by more than four-fold compared to that of HFD group (3:14). The correlation analysis showed that miR-34a was strongly related to the change of gut microbiota especially phylum Firmicutes (R = 0.796). Additionally, the target genes of miR-34a (HNF4α, PPARα and PPARα) were restored by GP both in mRNA and protein levels. CONCLUSION: Our results suggested that GP modulated the gut microbiota and suppressed hepatic miR-34a, which was associated with the amelioration of hepatic steatosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...