Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 181(12): 1768-1792, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38355288

ABSTRACT

BACKGROUND AND PURPOSE: Panax ginseng is widely applied in the adjuvant treatment of cardiometabolic diseases in clinical practice without clear mechanisms. This study aims to clearly define the efficacy and underlying mechanism of P. ginseng and its active components in protecting against atherosclerosis. EXPERIMENTAL APPROACH: The anti-atherogenic efficacy of total ginseng saponin extract (TGS) and its components was evaluated on Ldlr-/- mice. Gut microbial structure was analysed by 16S rRNA sequencing and PCR. Bile acid profiles were revealed using targeted metabolomics with LC-MS/MS analysis. The contribution of gut microbiota to atherosclerosis was assessed by co-housing experiments. KEY RESULTS: Ginsenoside Rb1, representing protopanaxadiol (PPD)-type saponins, increased intestinal Lactobacillus abundance, resulting in enhanced bile salt hydrolase (BSH) activity to promote intestinal conjugated bile acid hydrolysis and excretion, followed by suppression of enterohepatic farnesoid X receptor (FXR)-fibroblast growth factor 15 (FGF15) signal, and thereby increased cholesterol 7α-hydroxylase (CYP7A1) transcriptional expression and facilitated metabolic elimination of cholesterol. Synergistically, protopanaxatriol (PPT)-type saponins, represented by ginsenoside Rg1, protected against atherogenesis-triggered gut leak and metabolic endotoxaemia. Ginsenoside Rg1 directly induced mucin production to nutritionally maintain Akkermansia muciniphila, which reciprocally inhibited gut permeation. Rb1/Rg1 combination, rather than a single compound, can largely mimic the holistic efficacy of TGS in protecting Ldlr-/- mice from atherogenesis. CONCLUSION AND IMPLICATIONS: Our study provides strong evidence supporting TGS and ginsenoside Rb1/Rg1 combinations as effective therapies against atherogenesis, via targeting different signal nodes by different components and may provide some elucidation of the holistic mode of herbal medicines.


Subject(s)
Atherosclerosis , Gastrointestinal Microbiome , Ginsenosides , Homeostasis , Mice, Knockout , Panax , Animals , Ginsenosides/pharmacology , Gastrointestinal Microbiome/drug effects , Homeostasis/drug effects , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Male , Mice , Panax/chemistry , Mice, Inbred C57BL , Bile Acids and Salts/metabolism , Receptors, LDL/metabolism , Fibroblast Growth Factors/metabolism , Amidohydrolases/metabolism , Cholesterol 7-alpha-Hydroxylase/metabolism
2.
Eur J Pharmacol ; 945: 175618, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-36841284

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is the hepatic manifestation of metabolic syndrome. Non-resolving inflammation, triggered by sustained accumulation of lipids, is an important driving force of NASH. Thus, unveiling metabolic immune regulation could help better understand the pathology and intervention of NASH. In this study, we found the recruitment of neutrophils is an early inflammatory event in NASH mice, following the formation of neutrophil extracellular traps (NETs). NET is an initiating factor which exacerbates inflammatory responses in macrophages. Inhibition of NETs using DNase I significantly alleviated inflammation in NASH mice. We further carried out a metabolomic study to identify possible metabolic triggers of NETs, and linoleic acid (LA) metabolic pathway was the most altered pathway. We re-analyzed published clinical data and validated that LA metabolism was highly correlated with NASH. Consistently, both LA and γ-linolenic acid (GLA) were active in triggering NETs formation by oxidative burst. Furthermore, we identified silybin, a hepatoprotective agent, as a potent NETosis inhibitor, which effectively blocked NETs formation both in vitro and in vivo. Together, this study not only provide new insights into metabolism-immune causal link in NASH progression, but also demonstrate silybin as an important inhibitor of NETs and its therapeutical potential in treating NETosis-related diseases.


Subject(s)
Extracellular Traps , Non-alcoholic Fatty Liver Disease , Animals , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Extracellular Traps/metabolism , Silybin/pharmacology , Disease Models, Animal , Neutrophils , Fatty Acids, Unsaturated/pharmacology , Inflammation/metabolism
3.
Cell Rep ; 42(1): 112011, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36656708

ABSTRACT

Systemic metabolites serving as danger-associated molecular patterns play crucial roles in modulating the development, differentiation, and activity of innate immune cells. Yet, it is unclear how innate immune cells detect systemic metabolites for signal transmission. Here, we show that bile acids function as endogenous mitofusin 2 (MFN2) ligands and differentially modulate innate immune response to bacterial infection under cholestatic and physiological conditions. Bile acids at high concentrations promote mitochondrial tethering to the endoplasmic reticulum (ER), leading to calcium overload in the mitochondrion, which activates NLRP3 inflammasome and pyroptosis. By contrast, at physiologically relevant low concentrations, bile acids promote mitochondrial fusion, leading to enhanced oxidative phosphorylation and thereby strengthening infiltrated macrophages mediated phagocytotic clearance of bacteria. These findings support that bile acids, as endogenous activators of MFN2, are vital for tuning innate immune responses against infections, representing a causal link that connects systemic metabolism with mitochondrial dynamics in shaping innate immunity.


Subject(s)
Bile Acids and Salts , Immunity, Innate , Bile Acids and Salts/metabolism , Inflammasomes/metabolism , Mitochondria/metabolism , Macrophages/metabolism , Hydrolases/metabolism
4.
FASEB J ; 36(9): e22468, 2022 09.
Article in English | MEDLINE | ID: mdl-35913801

ABSTRACT

Cholestasis is a common complication of hepatitis B virus (HBV) infection, characterized by increased intrahepatic and plasma bile acid levels. Cholestasis was found negatively associated with hepatitis outcome, however, the exact mechanism by which cholestasis impacts anti-viral immunity and impedes HBV clearance remains elusive. Here, we found that cholestatic mice are featured with dysfunctional T cells response, as indicated by decreased sub-population of CD25+ /CD69+ CD4+ and CD8+ cells, while CTLA-4+ CD4+ and CD8+ subsets were increased. Mechanistically, bile acids disrupt intracellular calcium homeostasis via inhibiting mitochondria calcium uptake and elevating cytoplasmic Ca2+ concentration, leading to STIM1 and ORAI1 decoupling and impaired store-operated Ca2+ entry which is essential for NFAT signaling and T cells activation. Moreover, in a transgenic mouse model of HBV infection, we confirmed that cholestasis compromised both CD4+ and CD8+ T cells activation resulting in poor viral clearance. Collectively, our results suggest that bile acids play pivotal roles in anti-HBV infection via controlling T cells activation and metabolism and that targeting the regulation of bile acids may be a therapeutic strategy for host-virus defense.


Subject(s)
Cholestasis , Hepatitis B , Animals , Bile Acids and Salts , CD8-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Cholestasis/complications , Hepatitis B/complications , Hepatitis B virus/metabolism , Mice
5.
Eur J Pharmacol ; 928: 174976, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35487253

ABSTRACT

Gastritis is a common inflammation of stomach with multiple pathogenesis. This study was designed to investigate the protective effects of oral octreotide (OCT) against ethanol-induced acute gastric injury and H. pylori-induced chronic gastritis via promoting gastric mucosa restoration, reducing gastric acid secretion and inflammation. Male C57BL/6J mice were randomly divided and treated with three doses of OCT (0.5, 2.5, 10 mg/kg) alone or combined respectively with 10 mg/kg omeprazole (OME), 0.2 g/L metronidazole (MTZ)/0.1 g/L clarithromycin (CLR) in drinking water. Oxidative stress analysis, bacterial load analysis, qPCR, gastric histopathology examinations were performed in our study. Ethanol-induced acute gastric ulcer was restored by OCT alone at doses of 2.5 mg/kg, or combined with OME as indicated by markedly reducing Gastrin, Il-6 and Il1b expression through induction of Muc5ac and Occludin, significantly improving hyperacidity and gastric bleeding. As well, OCT combined with MTZ/CLR restored the integrity of gastric mucosa damaged by H. pylori via elevating the expression of Muc5ac and somatostatin receptor 2, decreasing inflammation and increasing the number of chorionic or glands. Besides, OCT is more suitable for long-term medication in the treatment of chronic gastritis than OME. In conclusion, our results proved that the newly developed oral OCT-based therapies were more effective to reverse gastric mucosa damage and inflammation in ethanol and H. pylori infection-induced gastric injury, it is of great significance for supplementing new clinical regimens for the treatment of acute and chronic gastritis.


Subject(s)
Gastritis, Atrophic , Gastritis , Helicobacter Infections , Helicobacter pylori , Animals , Clarithromycin/metabolism , Clarithromycin/pharmacology , Clarithromycin/therapeutic use , Ethanol/pharmacology , Gastric Mucosa , Gastritis/drug therapy , Gastritis/prevention & control , Gastritis, Atrophic/drug therapy , Helicobacter Infections/complications , Helicobacter Infections/drug therapy , Inflammation/drug therapy , Male , Mice , Mice, Inbred C57BL , Octreotide/pharmacology , Octreotide/therapeutic use , Omeprazole/pharmacology , Omeprazole/therapeutic use
6.
J Ethnopharmacol ; 277: 114223, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34044080

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The traditional Chinese medicine (TCM) preparation, Shengmai Yin (SMY), is widely applied in cardiovascular disease treatments. However, the pharmacological mechanism of its therapeutic effects has not been fully clarified. AIM OF THIS STUDY: This study aimed to clearly define the efficacy and underlying mechanism of SMY and its active components in protecting against atherosclerosis. MATERIALS AND METHODS: The pharmacological effects of SMY and its components were evaluated upon a mouse hypercholesteremia model induced by a high cholesterol diet (HCD) for 12 weeks and Apoe-/- mice, a mouse atherosclerosis model. Pathological indicators including serum cholesterol levels, cytokines and histological changes in aortic root plaques were assessed. Untargeted metabolomic, untargeted lipidomic and targeted lipidomic changing profiles were investigated to clarify pharmacological mechanisms. RESULTS: SMY and red ginseng crude extracts (GE) significantly decreased the serum cholesterol levels in hypercholesteremia mice and reduced the aortic root plaque areas and exerted antiatherogenic efficacy in Apoe-/- mice. Moreover, total red ginseng saponin extracts (TGS) showed the most apparent improvement on maintaining lipid homeostasis, representing the effects of red ginseng in SMY on atherosclerosis treatment. Mechanically, TGS inhibited serum secreted phospholipase A2 (sPLA2) activity and lowered the serum levels of lysophosphatidylcholine (lysoPC), which is a risk factor for atherosclerosis. CONCLUSIONS: Our findings revealed that ginsenosides from SMY exerted therapeutic effects on atherosclerosis by maintaining lipid homeostasis including cholesterol and lysoPCs.


Subject(s)
Atherosclerosis/prevention & control , Drugs, Chinese Herbal/pharmacology , Ginsenosides/pharmacology , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Cholesterol/blood , Cholesterol, Dietary , Cytokines/blood , Disease Models, Animal , Drug Combinations , Ginsenosides/isolation & purification , Lysophosphatidylcholines/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Acta Pharmacol Sin ; 42(1): 68-76, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32457417

ABSTRACT

Programmed cell death (PCD), including apoptosis, apoptotic necrosis, and pyroptosis, is involved in various organ dysfunction syndromes. Recent studies have revealed that a substrate of caspase-3, gasdermin E (GSDME), functions as an effector for pyroptosis; however, few inhibitors have been reported to prevent pyroptosis mediated by GSDME. Here, we developed a class of GSDME-derived inhibitors containing the core structure of DMPD or DMLD. Ac-DMPD-CMK and Ac-DMLD-CMK could directly bind to the catalytic domains of caspase-3 and specifically inhibit caspase-3 activity, exhibiting a lower IC50 than that of Z-DEVD-FMK. Functionally, Ac-DMPD/DMLD-CMK substantially inhibited both GSDME and PARP cleavage by caspase-3, preventing apoptotic and pyroptotic events in hepatocytes and macrophages. Furthermore, in a mouse model of bile duct ligation that mimics intrahepatic cholestasis-related acute hepatic failure, Ac-DMPD/DMLD-CMK significantly alleviated liver injury. Together, this study not only identified two specific inhibitors of caspase-3 for investigating PCD but also, more importantly, shed light on novel lead compounds for treating liver failure and organ dysfunctions caused by PCD.


Subject(s)
Amino Acid Chloromethyl Ketones/therapeutic use , Caspase 3/metabolism , Caspase Inhibitors/therapeutic use , Liver Diseases/prevention & control , Oligopeptides/therapeutic use , Protective Agents/therapeutic use , Amino Acid Chloromethyl Ketones/chemistry , Animals , Apoptosis/drug effects , Bile Ducts/surgery , Caspase Inhibitors/chemistry , Cell Line, Tumor , Humans , Ligation , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Oligopeptides/chemistry , Peptide Fragments/chemistry , Protective Agents/chemistry , Pyroptosis/drug effects , Receptors, Estrogen/chemistry
8.
J Ethnopharmacol ; 266: 113394, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-32941971

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disease of the gastrointestinal tract, consisting of ulcerative colitis (UC) and Crohn's disease (CD). Gut microbiota and their metabolites may play a role in the pathogen of IBD, especially of the UC. Qingchang Huashi Formula (QHF), a traditional Chinese medicine formula, has shown therapeutic effect on treating UC based on the clinical practice without clear pharmacological mechanism. AIM OF THE STUDY: The aim of this study was to clearly define the effect of QHF and its components, Baitouweng (PBR) and Baizhi (ADR) on treating UC. MATERIALS AND METHODS: Pharmacodynamic effects of QHF and single herb were evaluated in dextran sulfate sodium (DSS) induced acute or chronic colitis mice. Body weight loss, disease activity index (DAI) and colon length were estimated. Histological changes were observed by H&E staining. The number and abundance of gut microbiota were measured with 16S rRNA sequencing. LC-MS and GC-MS were used to detect the concentration of metabolites (e.g., bile acids (BAs) and short chain fatty acids (SCFAs)). The goblet cell was observed by Alcian blue/periodic acid-Schiff (AB/PAS) straining and the crypt stem cell was estimated by immunohistochemical analyses. The colorectal tissues were used to detect levels of IL-1ß, IL-6 and TNF-α by ELISA or qRT-PCR. The expression of NLRP3, Caspase 1 and IL-1ß were examined by western blotting. RESULTS: QHF significantly inhibited colitis, protected mice from the loss of body weight and colon shorten. Comparatively, ADR and PBR showed strong efficacy in inhibiting DSS-induced colitis. We verified that while ADR was responsible for QHF's effect on maintaining gut microbiota homeostasis and metabolism, PBR was more prominent in keeping crypt stem cells proliferation and colonic goblet cells function. Moreover, we demonstrated that the alleviation of colitis by QHF was associated with the restoration of gut microbiota-metabolism homeostasis, protection of intestinal epithelial barrier and regulation of NLRP3/IL-1ß pathway. CONCLUSIONS: The finding of the present study suggested that QHF is curative in DSS-induced colitis by restoring gut microbiota-metabolism homeostasis and goblet cells function. An optimized QHF was constituted by ADR and PBR, which showed comparable efficacy on colitis to that of QHF. Our work probed out the active constitutes as well as the relevant pharmacological mechanisms of QHF, shedding light on potential new drug combination for the treatment of IBD.


Subject(s)
Colitis, Ulcerative/drug therapy , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Goblet Cells/drug effects , Animals , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/microbiology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Goblet Cells/pathology , Homeostasis/drug effects , Male , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S
9.
Cell Metab ; 33(2): 424-436.e10, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33308446

ABSTRACT

Caspase-4 is an intracellular sensor for cytosolic bacterial lipopolysaccharide (LPS) and underlies infection-elicited pyroptosis. It is unclear whether and how caspase-4 detects host-derived factors to trigger pyroptosis. Here we show that mitochondrial permeability transition (MPT) activates caspase-4 by promoting the assembly of a protein complex, which we term the Apaf-1 pyroptosome, for the execution of facilitated pyroptosis. MPT, when induced by bile acids, calcium overload, or an adenine nucleotide translocator 1 (ANT1) activator, triggers assembly of the pyroptosome comprised of Apaf-1 and caspase-4 with a stoichiometry ratio of 7:2. Unlike the direct cleavage of gasdermin D (GSDMD) by caspase-4 upon LPS ligation, caspase-4 activated in the Apaf-1 pyroptosome proceeds to cleave caspase-3 and thereby GSDME to induce pyroptosis. Caspase-4-initiated and GSDME-executed pyroptosis underlies cholestatic liver failure. These findings identify Apaf-1 pyroptosome as a pivotal machinery for cells sensing MPT signals and may shed light on understanding how cells execute intrinsic pyroptosis under sterile conditions.


Subject(s)
Apoptotic Protease-Activating Factor 1/metabolism , Inflammasomes/metabolism , Mitochondrial Transmembrane Permeability-Driven Necrosis , Animals , Cell Line , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Mol Cell Proteomics ; 17(8): 1531-1545, 2018 08.
Article in English | MEDLINE | ID: mdl-29739823

ABSTRACT

Butyrate is a short chain fatty acid present in a high concentration in the gut lumen. It has been well documented that butyrate, by serving as an energetic metabolite, promotes the proliferation of normal colonocytes while, by serving as a histone deacetylase inhibitor, epigenetically suppressing the proliferation of cancerous counterparts undergoing the Warburg effect. However, how butyrate interrupts the metabolism of colorectal cancer cells and ultimately leads to the suppression of cell proliferation remains unclear. Here, we employed a metabolomics-proteomics combined approach to explore the link between butyrate-mediated proliferation arrest and cell metabolism. A metabolomics study revealed a remodeled metabolic profile with pronounced accumulation of pyruvate, decreased glycolytic intermediates upstream of pyruvate and reduced levels of nucleotides in butyrate-treated HCT-116 cells. Supplementation of key metabolite intermediates directly affected cancer-cell metabolism and modulated the suppressive effect of butyrate in HCT-116 cells. By a Drug Affinity Responsive Target Stability (DARTS)-based quantitative proteomics approach, we revealed the M2 isoform of a pyruvate kinase, PKM2, as a direct binding target of butyrate. Butyrate activates PKM2 via promoting its dephosphorylation and tetramerization and thereby reprograms the metabolism of colorectal cancer cells, inhibiting the Warburg effect while favoring energetic metabolism. Our study thus provides a mechanistic link between PKM2-induced metabolic remodeling and the antitumorigenic function of butyrate and demonstrates a widely applicable approach to uncovering unknown protein targets for small molecules with biological functions.


Subject(s)
Butyrates/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Pyruvate Kinase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/enzymology , Enzyme Activation/drug effects , Glycolysis/drug effects , Humans , Models, Biological , Phosphorylation/drug effects , Protein Multimerization , Proteomics
11.
J Pharmacol Sci ; 135(4): 148-155, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29233468

ABSTRACT

Butyrate is a typical short chain fatty acid produced by gut microbiota of which the dysmetabolism has been consistently associated with colorectal diseases. However, whether butyrate affects metastatic colorectal cancer is not clear. In this study we investigated in vitro the effect of butyrate on motility, a significant metastatic factor of colorectal cancer cells and explored the potential mechanism. By using wound healing and transwell-based invasion models, we demonstrated that pretreatment of butyrate significantly inhibited motility of HCT116, HT29, LOVO and HCT8 cells, this activity was further attributed to deactivation of Akt1 and ERK1/2. Suberanilohydroxamic acid (SAHA), another HDAC inhibitor, mimicked the inhibitory effect of butyrate on cell motility and deactivation of Akt/ERK. Furthermore, by silencing of HDAC3 with siRNA, we confirmed dependence of butyrate's effect on HDAC3, the similar reduced cell motility observed under HDAC3 silencing also indicates the significance of HDAC itself in cell motility. In conclusion, we confirmed the HDAC3-relied activity of butyrate on inhibiting motility of colorectal cancer cells via deactivating Akt/ERK signaling. Our data indicate that modulating butyrate metabolism is an effective therapeutic strategy of metastatic colorectal cancer; and HDAC3 might be a novel target for management of colorectal cancer metastasis.


Subject(s)
Butyrates/pharmacology , Cell Movement/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Histone Deacetylases/metabolism , MAP Kinase Signaling System/drug effects , Butyrates/metabolism , Butyrates/therapeutic use , Cell Line, Tumor , Histone Deacetylase Inhibitors , Histone Deacetylases/physiology , Humans , Hydroxamic Acids/pharmacology , Molecular Targeted Therapy , Neoplasm Metastasis , Vorinostat
SELECTION OF CITATIONS
SEARCH DETAIL
...