Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.838
Filter
1.
J Cancer Res Clin Oncol ; 150(6): 287, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833016

ABSTRACT

BACKGROUND: Butyrate is a common short-chain fatty acids (SCFA), and it has been demonstrated to regulate the development of breast cancer (BC), while the underlying mechanism is still unreported. METHODS: Gas chromatography was used to measure the amounts of SCFA (acetate, propionate, and butyrate) in the feces. Cell viability was measured by the CCK-8 assay. The wound healing assay demonstrated cell migration, and the transwell assay demonstrated cell invasion. The levels of protein and gene were determined by western blot assay and RT-qPCR assay, respectively. RESULTS: The levels of SCFA were lower in the faecal samples from BC patients compared to control samples. In cellular experiments, butyrate significantly suppressed the cell viability, migration and invasion of T47D in a dose-dependent manner. In animal experiments, butyrate effectively impeded the growth of BC tumors. Toll like receptor 4 (TLR4) was highly expressed in the tumors from BC patients. Butyrate inhibited the expression of TLR4. In addition, butyrate promoted the expression of cuproptosis-related genes including PDXK (pyridoxal kinase) and SLC25A28 (solute carrier family 25 member 28), which was lowly expressed in BC tumors. Importantly, overexpression of TLR4 can reverses the promotion of butyrate to PDXK and SLC25A28 expression and the prevention of butyrate to the malignant biological behaviors of T47D cells. CONCLUSION: In summary, butyrate inhibits the development of BC by facilitating the expression of PDXK and SLC25A28 through inhibition of TLR4. Our investigation first identified a connection among butyrate, TLR4 and cuproptosis-related genes in BC progression. These findings may provide novel target for the treatment of BC.


Subject(s)
Breast Neoplasms , Butyrates , Humans , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Butyrates/pharmacology , Animals , Mice , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Cell Line, Tumor , Mice, Nude , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Cell Survival/drug effects , Mice, Inbred BALB C
2.
Mol Biol Rep ; 51(1): 767, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38878205

ABSTRACT

BACKGROUND: Esterases (EC 3.1.1.X) are enzymes that catalyze the hydrolysis ester bonds. These enzymes have large potential for diverse applications in fine industries, particularly in pharmaceuticals, cosmetics, and bioethanol production. METHODS AND RESULTS: In this study, a gene encoding an esterase from Thermobifida fusca YX (TfEst) was successfully cloned, and its product was overexpressed in Escherichia coli and purified using affinity chromatography. The TfEst kinetic assay revealed catalytic efficiencies of 0.58 s-1 mM-1, 1.09 s-1 mM-1, and 0.062 s-1 mM-1 against p-Nitrophenyl acetate, p-Nitrophenyl butyrate, and 1-naphthyl acetate substrates, respectively. Furthermore, TfEst also exhibited activity in a pH range from 6.0 to 10.0, with maximum activity at pH 8.0. The enzyme demonstrated a half-life of 20 min at 70 °C. Notably, TfEst displayed acetyl xylan esterase activity as evidenced by the acetylated xylan assay. The structural prediction of TfEst using AlphaFold indicated that has an α/ß-hydrolase fold, which is consistent with other esterases. CONCLUSIONS: The enzyme stability over a broad pH range and its activity at elevated temperatures make it an appealing candidate for industrial processes. Overall, TfEst emerges as a promising enzymatic tool with significant implications for the advancement of biotechnology and biofuels industries.


Subject(s)
Acetylesterase , Esterases , Thermobifida , Acetylesterase/metabolism , Acetylesterase/genetics , Acetylesterase/chemistry , Hydrogen-Ion Concentration , Kinetics , Substrate Specificity , Thermobifida/enzymology , Thermobifida/genetics , Esterases/metabolism , Esterases/genetics , Esterases/chemistry , Enzyme Stability , Temperature , Escherichia coli/genetics , Escherichia coli/metabolism , Cloning, Molecular/methods , Hydrolysis , Xylans/metabolism , Butyrates/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Nitrophenols
3.
Gut Microbes ; 16(1): 2363020, 2024.
Article in English | MEDLINE | ID: mdl-38841892

ABSTRACT

CD4+ T cells play a critical role in regulating autoimmune diseases, and intestinal microbial metabolites control various immune responses. Granzyme B (GzmB)-producing CD4+ T cells have been recently reported to participate in the pathogenesis of autoimmune diseases. Here, we found that GzmbB-deficient CD4+ T cells induced more severe colitis in Rag1-/- mice than wild-type (WT) CD4+ T cells. Germ-free (GF) mice exhibited a lower expression of GzmB in intestinal CD4+ T cells compared to specific pathogen-free (SPF) mice. Intestinal microbial metabolite butyrate increased GzmB expression in CD4+ T cells, especially in IL-10-producing Th1 cells, through HDAC inhibition and GPR43, but not GPR41 and GPR109a. Butyrate-treated GzmB-deficient CD4+ T cells demonstrated more severe colitis compared to butyrate-treated WT CD4+ T cells in the T cell transfer model. Butyrate altered intestinal microbiota composition, but altered microbiota did not mediate butyrate induction of intestinal CD4+ T cell expression of GzmB in mice. Blimp1 was involved in the butyrate induction of GzmB in IL-10-producing Th1 cells. Glucose metabolism, including glycolysis and pyruvate oxidation, mediated butyrate induction of GzmB in Th1 cells. In addition, we found that IKZF3 and NR2F6 regulated GzmB expression induced by butyrate. Together, our studies underscored the critical role of GzmB in mediating gut bacterial metabolite butyrate regulation of T cell tolerance at the mucosal surface.


Subject(s)
Butyrates , Colitis , Gastrointestinal Microbiome , Granzymes , Interleukin-10 , Mice, Inbred C57BL , Th1 Cells , Animals , Interleukin-10/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Th1 Cells/immunology , Mice , Gastrointestinal Microbiome/drug effects , Butyrates/metabolism , Butyrates/pharmacology , Granzymes/metabolism , Colitis/immunology , Colitis/microbiology , Colitis/metabolism , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immune Tolerance , Homeodomain Proteins
4.
Front Endocrinol (Lausanne) ; 15: 1329294, 2024.
Article in English | MEDLINE | ID: mdl-38828415

ABSTRACT

Aim: Short-term use of pemafibrate (PEM), a selective modulator of peroxisome proliferator-activated receptor alpha, has been reported to improve abnormal liver function in patients with nonalcoholic fatty liver disease with hypertriglyceridemia (HTG-NAFLD). This study aimed to clarify the effects and predictive factors of long-term 72-week PEM administration on body composition, and laboratory tests in HTG-NAFLD patients. Methods: Fifty-three HTG-NAFLD patients receiving a 72-week PEM regimen were retrospectively enrolled. Routine blood and body composition results were analyzed immediately before and at the end of the study period. Results: PEM treatment significantly improved liver enzyme levels such as aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase, and gamma-glutamyl transferase, along with lipid profiles including triglyceride, total cholesterol, and low-density lipoprotein cholesterol. PEM did not have any detectable impact on body composition parameters. The factors of female, higher AST (≥ 46 U/L) and fat mass (≥ 31.9%), as well as lower soft lean mass (< 61.6%), skeletal muscle mass (< 36%), and skeletal muscle mass index (< 6.9 kg/m2) were significantly associated with the treatment response status of a > 30% decrease in ALT. All patients completed the treatment without any adverse effects. Conclusions: Long-term PEM treatment had a positive impact on liver enzymes and lipid profiles, but it did not result in significant changes in body composition among HTG-NAFLD patients. In predicting the response to PEM treatment, the evaluation of AST and body composition may be useful.


Subject(s)
Body Composition , Hypertriglyceridemia , Non-alcoholic Fatty Liver Disease , Humans , Female , Male , Middle Aged , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/complications , Hypertriglyceridemia/blood , Retrospective Studies , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/blood , Body Composition/drug effects , Benzoxazoles/therapeutic use , Benzoxazoles/administration & dosage , Adult , Butyrates/therapeutic use , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Aged , Hypolipidemic Agents/therapeutic use , Hypolipidemic Agents/administration & dosage
5.
Sci Total Environ ; 943: 173835, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38851345

ABSTRACT

OBJECTIVE: Chronic exposure to cold temperature is known to elevate blood pressure, leading to a condition known as cold-induced hypertension (CIH). Our previous research suggested correlations between alterations in gut microbiota, decrease in butyrate level, and the onset and progression of CIH. However, the role of butyrate in CIH and the underlying mechanisms need further investigation. METHODS: We exposed Specific Pathogen Free (SPF) rats to continuous cold temperature (4 ± 1 °C) for 6 weeks to establish a CIH rat model. Rats were divided into different groups by dose and duration, and the rats under cold were administered with butyrate (0.5 or 1 g/kg/day) daily. We assessed hypertension-associated phenotypes, pathological morphological changes, and endocrine-related phenotypes of brown adipose tissue (BAT). The effects of butyrate on gut microbiota and intestinal content metabolism were evaluated by 16s RNA sequencing and non-targeted metabolomics, respectively. RESULTS: The systolic blood pressure (SBP) of rats exposed to cold after supplemented with butyrate were significantly lower than that of the Cold group. Butyrate may increase the species, abundance, and diversity of gut microbiota in rats. Specifically, butyrate intervention enriched beneficial bacterial genera, such as Lactobacillaceae, and decreased the levels of harmful bacteria genera, such as Actinobacteriota and Erysipeiotrichaceae. Cold exposure significantly increased BAT cells and the number of mitochondria. After butyrate supplementation, the levels of peroxisome proliferator-activated receptor gamma coactivator 1a and fibroblast growth factor 21 in BAT were significantly elevated (P < 0.05), and the volume and number of lipid droplets increased. The levels of ANG II and high-density lipoprotein were elevated in the Cold group but decreased after butyrate supplementation. CONCLUSION: Butyrate may attenuate blood pressure in CIH by promoting the growth of beneficial bacteria and the secretion of beneficial derived factors produced by BAT, thus alleviating the elevation of blood pressure induced by cold. This study demonstrates the anti-hypertensive effects of butyrate and its potential therapeutic mechanisms, offering novel insights to the prevention and treatment of CIH in populations living or working in cold environments.


Subject(s)
Adipose Tissue, Brown , Butyrates , Cold Temperature , Gastrointestinal Microbiome , Hypertension , Animals , Gastrointestinal Microbiome/drug effects , Adipose Tissue, Brown/drug effects , Rats , Cold Temperature/adverse effects , Male
6.
Appl Microbiol Biotechnol ; 108(1): 372, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874789

ABSTRACT

Methanol is a promising feedstock for the bio-based economy as it can be derived from organic waste streams or produced electrochemically from CO2. Acetate production from CO2 in microbial electrosynthesis (MES) has been widely studied, while more valuable compounds such as butyrate are currently attracting attention. In this study, methanol was used as a co-substrate with CO2 to enhance butyrate production in MES. Feeding with CO2 and methanol resulted in the highest butyrate production rates and titres of 0.36 ± 0.01 g L-1 d-1 and 8.6 ± 0.2 g L-1, respectively, outperforming reactors with only CO2 feeding (0.20 ± 0.03 g L-1 d-1 and 5.2 ± 0.1 g L-1, respectively). Methanol acted as electron donor and as carbon source, both of which contributed ca. 50% of the carbon in the products. Eubacterium was the dominant genus with 52.6 ± 2.5% relative abundance. Thus, we demonstrate attractive route for the use of the C1 substrates, CO2 and methanol, to produce mainly butyrate. KEY POINTS: • Butyrate was the main product from methanol and CO2 in MES • Methanol acted as both carbon and electron source in MES • Eubacterium dominating microbial culture was enriched in MES.


Subject(s)
Butyrates , Carbon Dioxide , Methanol , Methanol/metabolism , Carbon Dioxide/metabolism , Butyrates/metabolism , Bioreactors/microbiology , Carbon/metabolism , Acetates/metabolism
7.
Gut Microbes ; 16(1): 2350151, 2024.
Article in English | MEDLINE | ID: mdl-38715346

ABSTRACT

The extreme environmental conditions of a plateau seriously threaten human health. The relationship between gut microbiota and human health at high altitudes has been extensively investigated. However, no universal gut microbiota biomarkers have been identified in the plateau population, limiting research into gut microbiota and high-altitude adaptation. 668 16s rRNA samples were analyzed using meta-analysis to reduce batch effects and uncover microbiota biomarkers in the plateau population. Furthermore, the robustness of these biomarkers was validated. Mendelian randomization (MR) results indicated that Tibetan gut microbiota may mediate a reduced erythropoietic response. Functional analysis and qPCR revealed that butyrate may be a functional metabolite in high-altitude adaptation. A high-altitude rat model showed that butyrate reduced intestinal damage caused by high altitudes. According to cell experiments, butyrate may downregulate hypoxia-inducible factor-1α (HIF-1α) expression and blunt cellular responses to hypoxic stress. Our research found universally applicable biomarkers and investigated their potential roles in promoting human health at high altitudes.


Subject(s)
Altitude , Biomarkers , Butyrates , Gastrointestinal Microbiome , Hypoxia-Inducible Factor 1, alpha Subunit , Humans , Tibet , Butyrates/metabolism , Butyrates/analysis , Biomarkers/analysis , Animals , Rats , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , RNA, Ribosomal, 16S/genetics , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Bacteria/isolation & purification , Male , Adaptation, Physiological , Mendelian Randomization Analysis
8.
J Med Case Rep ; 18(1): 239, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38725071

ABSTRACT

BACKGROUND: Radiation proctitis (RP) is a significant complication of pelvic radiation. Effective treatments for chronic RP are currently lacking. We report a case where chronic RP was successfully managed by metformin and butyrate (M-B) enema and suppository therapy. CASE PRESENTATION: A 70-year-old Asian male was diagnosed with prostate cancer of bilateral lobes, underwent definitive radiotherapy to the prostate of 76 Gy in 38 fractions and six months of androgen deprivation therapy. Despite a stable PSA nadir of 0.2 ng/mL for 10 months post-radiotherapy, he developed intermittent rectal bleeding, and was diagnosed as chronic RP. Symptoms persisted despite two months of oral mesalamine, mesalamine enema and hydrocortisone enema treatment. Transition to daily 2% metformin and butyrate (M-B) enema for one week led to significant improvement, followed by maintenance therapy with daily 2.0% M-B suppository for three weeks, resulting in continued reduction of rectal bleeding. Endoscopic examination and biopsy demonstrated a good therapeutic effect. CONCLUSIONS: M-B enema and suppository may be an effective treatment for chronic RP.


Subject(s)
Enema , Metformin , Proctitis , Prostatic Neoplasms , Radiation Injuries , Humans , Male , Proctitis/drug therapy , Proctitis/etiology , Aged , Metformin/therapeutic use , Metformin/administration & dosage , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/drug therapy , Radiation Injuries/drug therapy , Chronic Disease , Treatment Outcome , Butyrates/therapeutic use , Gastrointestinal Hemorrhage/drug therapy , Gastrointestinal Hemorrhage/therapy , Gastrointestinal Hemorrhage/etiology , Suppositories
9.
Microbiol Res ; 285: 127739, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38763016

ABSTRACT

Clostridioides difficile may have a negative impact on gut microbiota composition in terms of diversity and abundance, thereby triggering functional changes supported by the differential presence of genes involved in significant metabolic pathways, such as short-chain fatty acids (SCFA). This work has evaluated shotgun metagenomics data regarding 48 samples from four groups classified according to diarrhea acquisition site (community- and healthcare facility-onset) and positive or negative Clostridioides difficile infection (CDI) result. The metagenomic-assembled genomes (MAGs) obtained from each sample were taxonomically assigned for preliminary comparative analysis concerning differences in composition among groups. The predicted genes involved in metabolism, transport, and signaling remained constant in microbiota members; characteristic patterns were observed in MAGs and genes involved in SCFA butyrate and acetate metabolic pathways for each study group. A decrease in genera and species, as well as relative MAG abundance with the presence of the acetate metabolism-related gene, was evident in the HCFO/- group. Increased antibiotic resistance markers (ARM) were observed in MAGs along with the genes involved in acetate metabolism. The results highlight the need to explore the role of acetate in greater depth as a potential protector of the imbalances produced by CDI, as occurs in other inflammatory intestinal diseases.


Subject(s)
Acetates , Clostridioides difficile , Clostridium Infections , Gastrointestinal Microbiome , Metagenome , Metagenomics , Clostridioides difficile/genetics , Acetates/metabolism , Humans , Clostridium Infections/microbiology , Fatty Acids, Volatile/metabolism , Genome, Bacterial , Butyrates/metabolism , Metabolic Networks and Pathways/genetics , Feces/microbiology , Diarrhea/microbiology
10.
Nutrients ; 16(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38794746

ABSTRACT

BACKGROUND: Cytokine storm and oxidative stress are present in chronic obstructive pulmonary disease (COPD). Individuals with COPD present high levels of NF-κB-associated cytokines and pro-oxidant agents as well as low levels of Nrf2-associated antioxidants. This condition creates a steroid-resistant inflammatory microenvironment. Lacticaseibacillus rhamnosus (Lr) is a known anti-cytokine in lung diseases; however, the effect of Lr on lung inflammation and oxidative stress in steroid-resistant COPD mice remains unknown. OBJECTIVE: Thus, we investigated the Lr effect on lung inflammation and oxidative stress in mice and macrophages exposed to cigarette smoke extract (CSE) and unresponsive to steroids. METHODS: Mice and macrophages received dexamethasone or GLPG-094 (a GPR43 inhibitor), and only the macrophages received butyrate (but), all treatments being given before CSE. Lung inflammation was evaluated from the leukocyte population, airway remodeling, cytokines, and NF-κB. Oxidative stress disturbance was measured from ROS, 8-isoprostane, NADPH oxidase, TBARS, SOD, catalase, HO-1, and Nrf2. RESULTS: Lr attenuated cellularity, mucus, collagen, cytokines, ROS, 8-isoprostane, NADPH oxidase, and TBARS. Otherwise, SOD, catalase, HO-1, and Nrf2 were upregulated in Lr-treated COPD mice. Anti-cytokine and antioxidant effects of butyrate also occurred in CSE-exposed macrophages. GLPG-094 rendered Lr and butyrate less effective. CONCLUSIONS: Lr attenuates lung inflammation and oxidative stress in COPD mice, suggesting the presence of a GPR43 receptor-dependent mechanism also found in macrophages.


Subject(s)
Lacticaseibacillus rhamnosus , Macrophages , Oxidative Stress , Pulmonary Disease, Chronic Obstructive , Receptors, G-Protein-Coupled , Animals , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Oxidative Stress/drug effects , Receptors, G-Protein-Coupled/metabolism , Mice , Humans , Macrophages/drug effects , Macrophages/metabolism , Male , Cytokines/metabolism , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Smoke/adverse effects , Dexamethasone/pharmacology , Butyrates/pharmacology , Lung/drug effects , Lung/metabolism
11.
Anim Sci J ; 95(1): e13955, 2024.
Article in English | MEDLINE | ID: mdl-38769748

ABSTRACT

This study was conducted to assess the effects of fermented rice bran (FRB) with Ligilactobacillus equi on ruminal fermentation using an in vitro system. Oat hay, corn starch, and wheat bran were used as substrate for control. Ten percent of wheat bran was replaced with rice bran (RB), rice bran fermented with distilled water, and rice bran fermented with L. equi for T1, T2, and T3, respectively. The experimental diets were mixed with buffered rumen fluid from wethers under nitrogen gas and incubated for 24 h at 39°C. The fermentation profile and microbial population were analyzed after the incubations. The results revealed that the RB and FRB (with or without L. equi) significantly reduced the gas, methane (CH4), and CH4 per dry matter digested (p < 0.001). Total short-chain fatty acid was also reduced in T1 and T2 in comparison with the control (p < 0.001). Propionate proportion was increased while butyrate proportion was reduced in response to treatment addition in cultures (p < 0.001). Anaerobic fungi and Fibrobacter succinogenes abundance were decreased in treatments (p < 0.001). Overall, CH4 production in vitro can be reduced by RB and FRB supplementation as a result of the reduction of fiber-degrading microorganisms and a decrease in gas production.


Subject(s)
Dietary Fiber , Fatty Acids, Volatile , Fermentation , Methane , Oryza , Rumen , Animals , Rumen/microbiology , Rumen/metabolism , Dietary Fiber/metabolism , Methane/metabolism , Fatty Acids, Volatile/metabolism , In Vitro Techniques , Animal Feed , Fibrobacter/metabolism , Propionates/metabolism , Butyrates/metabolism
12.
Anim Sci J ; 95(1): e13954, 2024.
Article in English | MEDLINE | ID: mdl-38797605

ABSTRACT

This study investigated the physiological characteristics and carcass performance associated with residual methane emissions (RME), and the effects of bull differences on CH4-related traits in Japanese Black cattle. Enteric methane (CH4) emissions from 156 Japanese Black cattle (111 heifers and 45 steers) were measured during early fattening using the sniffer method. Various physiological parameters were investigated to clarify the physiological traits between the high, middle, and low RME groups. CH4-related traits were examined to determine whether bull differences affected progeny CH4 emissions. Ruminal butyrate and NH3 concentrations were significantly higher in the high-RME group than in the low-RME group, whereas the propionate content was significantly higher in the low-RME group. Blood urea nitrogen, ß-hydroxybutyric acid, and insulin concentrations were significantly higher, and blood amino acids were lower in the high-RME group than in the other groups. No significant differences were observed in the carcass traits and beef fat composition between RME groups. CH4-related traits were significantly different among bull herds. Our results show that CH4-related traits are heritable, wherein bull differences affect progeny CH4 production capability, and that the above-mentioned rumen fermentations and blood metabolites could be used to evaluate enteric methanogenesis in Japanese Black cattle.


Subject(s)
Butyrates , Methane , Rumen , Animals , Methane/metabolism , Cattle/metabolism , Cattle/physiology , Male , Rumen/metabolism , Female , Butyrates/metabolism , Ammonia/metabolism , Ammonia/blood , Ammonia/analysis , Fermentation , 3-Hydroxybutyric Acid/blood , Propionates/metabolism , Blood Urea Nitrogen , Insulin/blood , Insulin/metabolism
13.
Chem Pharm Bull (Tokyo) ; 72(5): 498-506, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38735699

ABSTRACT

Using (S)-decursinol isolated from root of Angelica gigas Nakai (AGN), we semi-synthesized and evaluated a series of both enantiomerically pure decursin derivatives for their antiproliferative activities against A549 human lung cancer cells. All synthesized compounds showed a broad spectrum of inhibitory activities against the growth of A549 cells. Especially, compound (S)-2d with (E)-(furan-3-yl)acryloyl group showed the most potent activity (IC50: 14.03 µM) against A549 cancer cells as compared with the reference compound, decursin (IC50: 43.55 µM) and its enantiomer, (R)-2d (IC50: 151.59 µM). Western blotting assays indicated that (S)-2d more strongly inhibited Janus kinase 1 (JAK1) and signal transducer and activator of transcription activation 3 (STAT3) phosphorylation than decursin in a dose-dependent manner, while having no effect on CXCR7 overexpression and total STAT3 level. In addition, (S)-2d induced cell cycle arrest at G1 phase and subsequent apoptotic cell death in A549 cancer cells. Our combined analysis of molecular docking studies and biological data suggests that the inhibition of JAK1 with (S)-2d resulted in loss of STAT3 phosphorylation and inhibition of cell growth in A549 cancer cells. These overall results strongly suggest that (S)-2d (MRC-D-004) as a novel JAK1 inhibitor may have therapeutic potential in the treatment of A549 human lung cancers by targeting the JAK1/STAT3 signaling pathway.


Subject(s)
Apoptosis , Benzopyrans , Butyrates , Cell Proliferation , Drug Screening Assays, Antitumor , Molecular Docking Simulation , STAT3 Transcription Factor , Humans , Cell Proliferation/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Benzopyrans/pharmacology , Benzopyrans/chemistry , Benzopyrans/chemical synthesis , Butyrates/pharmacology , Butyrates/chemistry , Butyrates/chemical synthesis , Apoptosis/drug effects , A549 Cells , Stereoisomerism , Dose-Response Relationship, Drug , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Molecular Structure , Angelica/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry
14.
Article in English | MEDLINE | ID: mdl-38739685

ABSTRACT

An oval to rod-shaped, Gram-stain-positive, strictly anaerobic bacterium, designated LFL-14T, was isolated from the faeces of a healthy Chinese woman. Cells of the strain were non-spore-forming, grew optimally at 37 °C (growth range 30-45 °C) and pH 7.0 (growth range 6.0-9.0) under anaerobic conditions in the liquid modified Gifu anaerobic medium (mGAM). The result of 16S rRNA gene-based analysis indicated that LFL-14T shared an identity of 94.7 0% with Eubacterium ventriosum ATCC 27560T, indicating LFL-14T represented a novel taxon. The results of genome-based analysis revealed that the average nucleotide identity (ANI), the digital DNA-DNA hybridisation (dDDH) and average amino acid identity (AAI) between LFL-14T and its phylogenetically closest neighbour, Eubacterium ventriosum ATCC 27560T, were 77.0 %, 24.6 and 70.9 %, respectively, indicating that LFL-14T represents a novel species of the genus Eubacterium. The genome size of LFL-14T was 2.92 Mbp and the DNA G+C content was 33.14 mol%. We analysed the distribution of the genome of LFL-14T in cohorts of healthy individuals, type 2 diabetes patients (T2D) and patients with non-alcoholic fatty liver disease (NAFLD). We found that its abundance was higher in the T2D cohort, but it had a low average abundance of less than 0.2 % in all three cohorts. The percentages of frequency of occurrence in the T2D, healthy and NAFLD cohorts were 48.87 %, 16.72 % and 13.10 % respectively. The major cellular fatty acids of LFL-14T were C16 : 0 (34.4 %), C17 : 0 2-OH (21.4 %) and C14 : 0 (11.7 %). Additionally, the strain contained diphosphatidylglycerol (DPG) and phosphatidylethanolamine (PE), as well as unidentified phospholipids and unidentified glycolipids. The glucose fermentation products of LFL-14T were acetate and butyrate. In summary, On the basis of its chemotaxonomic, phenotypic, phylogenetic and phylogenomic properties, strain LFL-14T (= CGMCC 1.18005T = KCTC 25580T) is identified as representing a novel species of the genus Eubacterium, for which the name Eubacterium album sp. nov. is proposed.


Subject(s)
Bacterial Typing Techniques , Base Composition , DNA, Bacterial , Eubacterium , Fatty Acids , Feces , Nucleic Acid Hybridization , Phylogeny , RNA, Ribosomal, 16S , Sequence Analysis, DNA , Humans , RNA, Ribosomal, 16S/genetics , DNA, Bacterial/genetics , Female , Eubacterium/genetics , Eubacterium/isolation & purification , Eubacterium/classification , Feces/microbiology , Butyrates/metabolism , Genome, Bacterial , China , Adult
15.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732048

ABSTRACT

HIV infection results in marked alterations in the gut microbiota (GM), such as the loss of microbial diversity and different taxonomic and metabolic profiles. Despite antiretroviral therapy (ART) partially ablating gastrointestinal alterations, the taxonomic profile after successful new ART has shown wide variations. Our objective was to determine the GM composition and functions in people living with HIV (PLWHIV) under ART in comparison to seronegative controls (SC). Fecal samples from 21 subjects (treated with integrase strand-transfer inhibitors, INSTIs) and 18 SC were included. We employed 16S rRNA amplicon sequencing, coupled with PICRUSt2 and fecal short-chain fatty acid (SCFA) quantification by gas chromatography. The INSTI group showed a decreased α-diversity (p < 0.001) compared to the SC group, at the expense of increased amounts of Pseudomonadota (Proteobacteria), Segatella copri, Lactobacillus, and Gram-negative bacteria. Concurrently, we observed an enrichment in Megasphaera and Butyricicoccus, both SCFA-producing bacteria, and significant elevations in fecal butyrate in this group (p < 0.001). Interestingly, gut dysbiosis in PLWHIV was characterized by a proinflammatory environment orchestrated by Pseudomonadota and elevated levels of butyrate associated with bacterial metabolic pathways, as well as the evident presence of butyrogenic bacteria. The role of this unique GM in PLWHIV should be evaluated, as well as the use of butyrate-based supplements and ART regimens that contain succinate, such as tenofovir disoproxil succinate. This mixed profile is described for the first time in PLWHIV from Mexico.


Subject(s)
Feces , Gastrointestinal Microbiome , HIV Infections , RNA, Ribosomal, 16S , Humans , HIV Infections/microbiology , HIV Infections/drug therapy , Mexico , Female , Male , Adult , Middle Aged , Feces/microbiology , RNA, Ribosomal, 16S/genetics , Dysbiosis/microbiology , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Butyrates/metabolism
16.
Nutrients ; 16(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732615

ABSTRACT

Adequate diet, physical activity, and dietary supplementation with muscle-targeted food for special medical purposes (FSMP) or dietary supplement (DS) are currently considered fundamental pillars in sarcopenia treatment. The aim of this study is to evaluate the effectiveness of a DS (containing hydroxy-methyl-butyrate, carnosine, and magnesium, for its action on muscle function and protein synthesis and butyrate and lactoferrin for their contribution to the regulation of gut permeability and antioxidant/anti-inflammation activity) on muscle mass (assessed by dual X-ray absorptiometry (DXA)), muscle function (by handgrip test, chair test, short physical performance battery (SPPB) test, and walking speed test), inflammation (tumor necrosis factor-alpha (TNF-a), C-reactive protein (CRP), and visceral adipose tissue (VAT)) and gut axis (by zonulin). A total of 59 participants (age 79.7 ± 4.8 years, body mass index 20.99 ± 2.12 kg/m2) were enrolled and randomly assigned to intervention (n = 30) or placebo (n = 28). The skeletal muscle index (SMI) significantly improved in the supplemented group compared to the placebo one, +1.02 (CI 95%: -0.77; 1.26), p = 0.001; a significant reduction in VAT was observed in the intervention group, -70.91 g (-13.13; -4.70), p = 0.036. Regarding muscle function, all the tests significantly improved (p = 0.001) in the supplemented group compared to the placebo one. CRP, zonulin, and TNF-alpha significantly decreased (p = 0.001) in intervention, compared to placebo, -0.74 mg/dL (CI 95%: -1.30; -0.18), -0.30 ng/mL (CI 95%: -0.37; -0.23), -6.45 pg/mL (CI 95%: -8.71; -4.18), respectively. This DS improves muscle mass and function, and the gut muscle has emerged as a new intervention target for sarcopenia.


Subject(s)
Carnosine , Dietary Supplements , Lactoferrin , Magnesium , Muscle, Skeletal , Permeability , Sarcopenia , Humans , Male , Aged , Female , Sarcopenia/drug therapy , Sarcopenia/prevention & control , Carnosine/administration & dosage , Lactoferrin/administration & dosage , Lactoferrin/pharmacology , Magnesium/administration & dosage , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Permeability/drug effects , Aged, 80 and over , Valerates/administration & dosage , Valerates/pharmacology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Butyrates , Double-Blind Method , Haptoglobins , C-Reactive Protein/metabolism , C-Reactive Protein/analysis , Protein Precursors
17.
Environ Microbiol Rep ; 16(3): e13276, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733087

ABSTRACT

Syntrophic interactions are key in anaerobic food chains, facilitating the conversion of complex organic matter into methane. A typical example involves acetogenic bacteria converting fatty acids (e.g., butyrate and propionate), a process thermodynamically reliant on H2 consumption by microorganisms such as methanogens. While most studies focus on H2-interspecies transfer between these groups, knowledge on acetate cross-feeding in anaerobic systems is lacking. This study investigated butyrate oxidation by co-cultures of Syntrophomonas wolfei and Methanospirillum hungatei, both with and without the addition of the acetate scavenger Methanothrix soehngenii. Growth and gene expression patterns of S. wolfei and M. hungatei were followed in the two conditions. Although butyrate consumption rates remained constant, genes in the butyrate degradation pathway of S. wolfei were less expressed in the presence of M. soehngenii, including genes involved in reverse electron transport. Higher expression of a type IV-pili operon in S. wolfei hints to the potential for direct interspecies electron transfer between S. wolfei and M. soehngenii and an energetically advantageous relationship between the two microorganisms. Overall, the presence of the acetate scavenger M. soehngenii positively influenced the energy metabolism of S. wolfei and highlighted the relevance of including acetate scavengers when investigating syntrophic fatty acid degradation.


Subject(s)
Methanospirillum , Methanospirillum/metabolism , Methanospirillum/genetics , Butyrates/metabolism , Transcriptome , Anaerobiosis , Oxidation-Reduction , Acetates/metabolism , Microbial Interactions , Methane/metabolism , Coculture Techniques , Electron Transport
18.
J Agric Food Chem ; 72(21): 12171-12183, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38748640

ABSTRACT

Ulcerative colitis (UC) is a complex chronic inflammatory disease closely associated with gut homeostasis dysfunction. The previous studies have shown that stachyose, a functional food additive, has the potential to enhance gut health and alleviate UC symptoms. However, the underlying mechanism of its effects remains unknown. In this study, our findings showed that dietary supplements of stachyose had a significant dose-dependent protective effect on colitis symptoms, regulation of gut microbiota, and restoration of the Treg/Th17 cell balance in dextran sulfate sodium (DSS) induced colitis mice. To further validate these findings, we conducted fecal microbiota transplantation (FMT) to treat DSS-induced colitis in mice. The results showed that microbiota from stachyose-treated mice exhibited a superior therapeutic effect against colitis and effectively regulated the Treg/Th17 cell balance in comparison to the control group. Moreover, both stachyose supplementation and FMT resulted in an increase in butyrate production and the activation of PPARγ. However, this effect was partially attenuated by PPARγ antagonist GW9662. These results suggested that stachyose alleviates UC symptoms by modulating gut microbiota and activating PPARγ. In conclusion, our work offers new insights into the benefical effects of stachyose on UC and its potential role in modulating gut microbiota.


Subject(s)
Butyrates , Colitis, Ulcerative , Gastrointestinal Microbiome , Mice, Inbred C57BL , PPAR gamma , Signal Transduction , T-Lymphocytes, Regulatory , Th17 Cells , Animals , PPAR gamma/metabolism , PPAR gamma/genetics , Mice , Th17 Cells/immunology , T-Lymphocytes, Regulatory/immunology , Gastrointestinal Microbiome/drug effects , Humans , Male , Signal Transduction/drug effects , Colitis, Ulcerative/immunology , Colitis, Ulcerative/therapy , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/drug therapy , Oligosaccharides/administration & dosage , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Dextran Sulfate/adverse effects
19.
Aging (Albany NY) ; 16(9): 7961-7978, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38709282

ABSTRACT

BACKGROUND: This study combined bioinformatics and experimental verification in a mouse model of intestinal ischemia-reperfusion injury (IRI) to explore the protection mechanism exerted by butyrate against IRI. METHODS: GeneCards, Bioinformatics Analysis Tool for Molecular Mechanisms of Traditional Chinese Medicine and GSE190581 were used to explore the relationship between butyrate and IRI and aging. Protein-protein interaction networks involving butyrate and IRI were constructed via the STRING database, with hub gene analysis performed through Cytoscape. Functional enrichment analysis was conducted on intersection genes. A mouse model of IRI was established, followed by direct arterial injection of butyrate. The experiment comprised five groups: normal, sham, model, vehicle, low-dose butyrate, and high-dose butyrate. Intestinal tissue observation was done via transmission electron microscopy (TEM), histological examination via hematoxylin and eosin (H&E) staining, tight junction proteins detection via immunohistochemistry, and Western blot analysis of hub genes. Drug-target interactions were evaluated through molecular docking. RESULTS: Butyrate protected against IRI by targeting 458 genes, including HMGB1 and TLR4. Toll-like receptor pathway was implicated. Butyrate improved intestinal IRI by reducing mucosal damage, increasing tight junction proteins, and lowering levels of HMGB1, TLR4, and MyD88. Molecular docking showed strong binding energies between butyrate and HMGB1 (-3.7 kcal/mol) and TLR4 (-3.8 kcal/mol). CONCLUSIONS: According to bioinformatics predictions, butyrate mitigates IRI via multiple-target and multiple-channel mechanisms. The extent of IRI can be reduced by butyrate through the inhibition of the HMGB1-TLR4-MyD88 signaling pathway, which is related to senescence.


Subject(s)
Butyrates , HMGB1 Protein , Myeloid Differentiation Factor 88 , Reperfusion Injury , Signal Transduction , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , HMGB1 Protein/drug effects , Mice , Signal Transduction/drug effects , Butyrates/pharmacology , Male , Molecular Docking Simulation , Intestines/drug effects , Intestines/pathology , Disease Models, Animal , Mice, Inbred C57BL , Protein Interaction Maps
20.
Curr Nutr Rep ; 13(2): 294-313, 2024 06.
Article in English | MEDLINE | ID: mdl-38656688

ABSTRACT

PURPOSE OF REVIEW: Global health concerns persist in the realm of cardiovascular diseases (CVDs), necessitating innovative strategies for both prevention and treatment. This narrative review aims to explore the potential of short-chain fatty acids (SCFAs)-namely, acetate, propionate, and butyrate-as agents in the realm of postbiotics for the management of CVDs. RECENT FINDINGS: We commence our discussion by elucidating the concept of postbiotics and their pivotal significance in mitigating various aspects of cardiovascular diseases. This review centers on a comprehensive examination of diverse SCFAs and their associated receptors, notably GPR41, GPR43, and GPR109a. In addition, we delve into the intricate cellular and pharmacological mechanisms through which these receptors operate, providing insights into their specific roles in managing cardiovascular conditions such as hypertension, atherosclerosis, heart failure, and stroke. The integration of current information in our analysis highlights the potential of both SCFAs and their receptors as a promising path for innovative therapeutic approaches in the field of cardiovascular health. The idea of postbiotics arises as an optimistic and inventive method, presenting new opportunities for preventing and treating cardiovascular diseases.


Subject(s)
Cardiovascular Diseases , Fatty Acids, Volatile , Receptors, G-Protein-Coupled , Humans , Fatty Acids, Volatile/metabolism , Receptors, G-Protein-Coupled/metabolism , Gastrointestinal Microbiome/drug effects , Propionates , Animals , Butyrates , Receptors, Cell Surface
SELECTION OF CITATIONS
SEARCH DETAIL
...