Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 654
Filtrer
1.
Cell Mol Life Sci ; 81(1): 293, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38976012

RÉSUMÉ

The function of astrocytes in response to gut microbiota-derived signals has an important role in the pathophysiological processes of central nervous system (CNS) diseases. However, the specific effects of microbiota-derived metabolites on astrocyte activation have not been elucidated yet. Experimental autoimmune encephalomyelitis (EAE) was induced in female C57BL/6 mice as a classical MS model. The alterations of gut microbiota and the levels of short-chain fatty acids (SCFAs) were assessed after EAE induction. We observed that EAE mice exhibit low levels of Allobaculum, Clostridium_IV, Clostridium_XlVb, Lactobacillus genera, and microbial-derived SCFAs metabolites. SCFAs supplementation suppressed astrocyte activation by increasing the level of tryptophan (Trp)-derived AhR ligands that activating the AhR. The beneficial effects of SCFAs supplementation on the clinical scores, histopathological alterations, and the blood brain barrier (BBB)-glymphatic function were abolished by intracisterna magna injection of AAV-GFAP-shAhR. Moreover, SCFAs supplementation suppressed the loss of AQP4 polarity within astrocytes in an AhR-dependent manner. Together, SCFAs potentially suppresses astrocyte activation by amplifying Trp-AhR-AQP4 signaling in EAE mice. Our study demonstrates that SCFAs supplementation may serve as a viable therapy for inflammatory disorders of the CNS.


Sujet(s)
Aquaporine-4 , Astrocytes , Encéphalomyélite auto-immune expérimentale , Acides gras volatils , Souris de lignée C57BL , Récepteurs à hydrocarbure aromatique , Transduction du signal , Tryptophane , Animaux , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/métabolisme , Astrocytes/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Acides gras volatils/pharmacologie , Acides gras volatils/métabolisme , Récepteurs à hydrocarbure aromatique/métabolisme , Souris , Tryptophane/métabolisme , Tryptophane/pharmacologie , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Aquaporine-4/métabolisme , Aquaporine-4/génétique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques
2.
Anim Sci J ; 95(1): e13972, 2024.
Article de Anglais | MEDLINE | ID: mdl-38923622

RÉSUMÉ

High grain feeding or weaning, which could compromise the rumen epithelium by increasing ruminal short-chain fatty acid (SCFA) concentrations with pH reduction, is associated with high levels of ruminal toll-like receptor 5 (TLR5). This study aimed to determine the role of TLR5 in the rumen epithelium. Immunohistochemistry revealed that TLR5 was localized in cells on the basal side (i.e., basal and spinous layers) rather than in the granular layer in the rumen epithelium, where tight junctions are most potent, in pre- and post-weaning calves (n = 9). Primary bovine rumen epithelial cells (BRECs) obtained from Holstein cows (n = 3) were cultured to investigate the factors that upregulate TLR5; however, SCFA, low pH (pH 5.6), BHBA, L-lactate, D-lactate, and LPS did not upregulate TLR5 gene expression in BREC. Primary BREC treated with flagellin (TLR5 ligand) had higher expression of interleukin-1ß (IL-1ß) (P < 0.05) than BREC treated with vehicle. In addition, BREC treated with IL-1ß had higher expression of antimicrobial peptides and C-X-C motif chemokine ligand 8 than BREC treated with vehicle (P < 0.05). These results suggest that ruminal TLR5 may recognize epithelial disruption via flagellin and mediate the immune response via IL-1ß during high-grain feeding or weaning.


Sujet(s)
Cellules épithéliales , Expression des gènes , Interleukine-1 bêta , Interleukine-8 , Rumen , Récepteur de type Toll-5 , Animaux , Récepteur de type Toll-5/génétique , Récepteur de type Toll-5/métabolisme , Rumen/métabolisme , Bovins/métabolisme , Cellules épithéliales/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Cellules cultivées , Interleukine-8/métabolisme , Interleukine-8/génétique , Sevrage , Peptides antimicrobiens/génétique , Peptides antimicrobiens/métabolisme , Flagelline/pharmacologie , Acides gras volatils/métabolisme , Acides gras volatils/pharmacologie , Ligands , Régulation positive
3.
Molecules ; 29(9)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38731399

RÉSUMÉ

The antibacterial effects of a selection of volatile fatty acids (acetic, propionic, butyric, valeric, and caproic acids) relevant to anaerobic digestion were investigated at 1, 2 and 4 g/L. The antibacterial effects were characterised by the dynamics of Enterococcus faecalis NCTC 00775, Escherichia coli JCM 1649 and Klebsiella pneumoniae A17. Mesophilic anaerobic incubation to determine the minimum bactericidal concentration (MBC) and median lethal concentration of the VFAs was carried out in Luria Bertani broth at 37 °C for 48 h. Samples collected at times 0, 3, 6, 24 and 48 h were used to monitor bacterial kinetics and pH. VFAs at 4 g/L demonstrated the highest bactericidal effect (p < 0.05), while 1 g/L supported bacterial growth. The VFA cocktail was the most effective, while propionic acid was the least effective. Enterococcus faecalis NCTC 00775 was the most resistant strain with the VFAs MBC of 4 g/L, while Klebsiella pneumoniae A17 was the least resistant with the VFAs MBC of 2 g/L. Allowing a 48 h incubation period led to more log decline in the bacterial numbers compared to earlier times. The VFA cocktail, valeric, and caproic acids at 4 g/L achieved elimination of the three bacteria strains, with over 7 log10 decrease within 48 h.


Sujet(s)
Antibactériens , Enterococcus faecalis , Acides gras volatils , Klebsiella pneumoniae , Tests de sensibilité microbienne , Acides gras volatils/métabolisme , Acides gras volatils/pharmacologie , Antibactériens/pharmacologie , Antibactériens/composition chimique , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecalis/croissance et développement , Klebsiella pneumoniae/effets des médicaments et des substances chimiques , Klebsiella pneumoniae/croissance et développement , Anaérobiose , Escherichia coli/effets des médicaments et des substances chimiques , Escherichia coli/croissance et développement , Propionates/pharmacologie , Concentration en ions d'hydrogène , Acides pentanoïques/pharmacologie
4.
Sci Rep ; 14(1): 11325, 2024 05 17.
Article de Anglais | MEDLINE | ID: mdl-38760458

RÉSUMÉ

The low response rate of immune checkpoint inhibitors (ICIs) is a challenge. The efficacy of ICIs is influenced by the tumour microenvironment, which is controlled by the gut microbiota. In particular, intestinal bacteria and their metabolites, such as short chain fatty acids (SCFAs), are important regulators of cancer immunity; however, our knowledge on the effects of individual SCFAs remains limited. Here, we show that isobutyric acid has the strongest effect among SCFAs on both immune activity and tumour growth. In vitro, cancer cell numbers were suppressed by approximately 75% in humans and mice compared with those in controls. Oral administration of isobutyric acid to carcinoma-bearing mice enhanced the effect of anti-PD-1 immunotherapy, reducing tumour volume by approximately 80% and 60% compared with those in the control group and anti-PD-1 antibody alone group, respectively. Taken together, these findings may support the development of novel cancer therapies that can improve the response rate to ICIs.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Microenvironnement tumoral , Animaux , Souris , Humains , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Lignée cellulaire tumorale , Femelle , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Immunothérapie/méthodes , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/anatomopathologie , Acides gras volatils/métabolisme , Acides gras volatils/pharmacologie , Synergie des médicaments
5.
PLoS One ; 19(5): e0304551, 2024.
Article de Anglais | MEDLINE | ID: mdl-38814895

RÉSUMÉ

Coronary microvascular dysfunction (CMD) is a critical pathogenesis of cardiovascular diseases. Lower endothelial nitric oxide synthase (eNOS) phosphorylation leads to reduced endothelium-derived relaxing factor nitric oxide (NO) generation, causing and accelerating CMD. Endoplasmic reticulum stress (ER stress) has been shown to reduce NO production in umbilical vein endothelial cells. Oxidized low-density lipoprotein (ox-LDL) damages endothelial cell function. However, the relationship between ox-LDL and coronary microcirculation has yet to be assessed. Short-chain fatty acid (SCFA), a fermentation product of the gut microbiome, could improve endothelial-dependent vasodilation in human adipose arterioles, but the effect of SCFA on coronary microcirculation is unclear. In this study, we found ox-LDL stimulated expression of ER chaperone GRP78. Further, we activated downstream PERK/eIF2a, IRE1/JNK, and ATF6 signaling pathways, decreasing eNOS phosphorylation and NO production in human cardiac microvascular endothelial. Furthermore, SCFA-propionate can inhibit ox-LDL-induced eNOS phosphorylation reduction and raise NO production; the mechanism is related to the inhibition of ER stress and downstream signaling pathways PERK/eIF2a, IRE1/JNK, and ATF6. In summary, we demonstrate that ox-LDL induced CMD by activating ER stress, propionate can effectively counteract the adverse effects of ox-LDL and protect coronary microcirculation function via inhibiting ER stress.


Sujet(s)
Chaperonne BiP du réticulum endoplasmique , Stress du réticulum endoplasmique , Lipoprotéines LDL , Nitric oxide synthase type III , Monoxyde d'azote , Propionates , Transduction du signal , Humains , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Lipoprotéines LDL/métabolisme , Nitric oxide synthase type III/métabolisme , Propionates/pharmacologie , Monoxyde d'azote/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Vaisseaux coronaires/effets des médicaments et des substances chimiques , Vaisseaux coronaires/métabolisme , Acides gras volatils/métabolisme , Acides gras volatils/pharmacologie , eIF-2 Kinase/métabolisme , Facteur de transcription ATF-6/métabolisme , Microcirculation/effets des médicaments et des substances chimiques , Protéines du choc thermique/métabolisme
6.
ACS Chem Neurosci ; 15(8): 1712-1727, 2024 04 17.
Article de Anglais | MEDLINE | ID: mdl-38581382

RÉSUMÉ

Short-chain fatty acids (SCFAs) are gut microbial metabolic derivatives produced during the fermentation of ingested complex carbohydrates. SCFAs have been widely regarded to have a potent anti-inflammatory and neuro-protective role and have implications in several disease conditions, such as, inflammatory bowel disease, type-2 diabetes, and neurodegenerative disorders. Japanese encephalitis virus (JEV), a neurotropic flavivirus, is associated with life threatening neuro-inflammation and neurological sequelae in infected hosts. In this study, we hypothesize that SCFAs have potential in mitigating JEV pathogenesis. Postnatal day 10 BALB/c mice were intraperitoneally injected with either a SCFA mixture (acetate, propionate, and butyrate) or PBS for a period of 7 days, followed by JEV infection. All mice were observed for onset and progression of symptoms. The brain tissue was collected upon reaching terminal illness for further analysis. SCFA-supplemented JEV-infected mice (SCFA + JEV) showed a delayed onset of symptoms, lower hindlimb clasping score, and decreased weight loss and increased survival by 3 days (p < 0.0001) upon infection as opposed to the PBS-treated JEV-infected animals (JEV). Significant downregulation of inflammatory cytokines TNF-α, MCP-1, IL-6, and IFN-Υ in the SCFA + JEV group relative to the JEV-infected control group was observed. Inflammatory mediators, phospho-NF-kB (P-NF-kB) and iba1, showed 2.08 ± 0.1 and 3.132 ± 0.43-fold upregulation in JEV versus 1.19 ± 0.11 and 1.31 ± 0.11-fold in the SCFA + JEV group, respectively. Tissue section analysis exhibited reduced glial activation (JEV group─42 ± 2.15 microglia/ROI; SCFA + JEV group─27.07 ± 1.8 microglia/ROI) in animals that received SCFA supplementation prior to infection as seen from the astrocytic and microglial morphometric analysis. Caspase-3 immunoblotting showed 4.08 ± 1.3-fold upregulation in JEV as compared to 1.03 ± 0.14-fold in the SCFA + JEV group and TUNEL assay showed a reduced cellular death post-JEV infection (JEV-6.4 ± 1.5 cells/ROI and SCFA + JEV-3.7 ± 0.73 cells/ROI). Our study critically contributes to the increasing evidence in support of SCFAs as an anti-inflammatory and neuro-protective agent, we further expand its scope as a potential supplementary intervention in JEV-mediated neuroinflammation.


Sujet(s)
Encéphalite japonaise , Acides gras volatils , Microbiome gastro-intestinal , Maladies neuro-inflammatoires , Microbiome gastro-intestinal/physiologie , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/microbiologie , Microglie/effets des médicaments et des substances chimiques , Microglie/immunologie , Encéphalite japonaise/traitement médicamenteux , Encéphalite japonaise/immunologie , Encéphalite japonaise/microbiologie , Encéphalite japonaise/prévention et contrôle , Encéphalite japonaise/virologie , Acides gras volatils/pharmacologie , Acides gras volatils/usage thérapeutique , Virus de l'encéphalite japonaise (sous-groupe)/effets des médicaments et des substances chimiques , Virus de l'encéphalite japonaise (sous-groupe)/immunologie , Virus de l'encéphalite japonaise (sous-groupe)/pathogénicité , Analyse de survie , Chimiokines/immunologie , Chimiokines/métabolisme , Médiateurs de l'inflammation/immunologie , Médiateurs de l'inflammation/métabolisme , Syndrome de libération de cytokines/immunologie , Syndrome de libération de cytokines/métabolisme , Syndrome de libération de cytokines/prévention et contrôle , Humains , Femelle , Animaux , Souris , Apoptose/effets des médicaments et des substances chimiques , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/virologie , Charge virale/effets des médicaments et des substances chimiques , Facteurs temps
7.
Front Immunol ; 15: 1328263, 2024.
Article de Anglais | MEDLINE | ID: mdl-38650948

RÉSUMÉ

Background: Despite therapy advances, one of the leading causes of cancer deaths still remains lung cancer. To improve current treatments or prevent non-small cell lung cancer (NSCLC), the role of the nutrition in cancer onset and progression needs to be understood in more detail. While in colorectal cancer, the influence of local microbiota derived SCFAs have been well investigated, the influence of SCFA on lung cancer cells via peripheral blood immune system should be investigated more deeply. In this respect, nutrients absorbed via the gut might affect the tumor microenvironment (TME) and thus play an important role in tumor cell growth. Objective: This study focuses on the impact of the short-chain fatty acid (SCFA) Sodium Butyrate (SB), on lung cancer cell survival. We previously described a pro-tumoral role of glucose on A549 lung adenocarcinoma cell line. In this study, we wanted to know if SB would counteract the effect of glucose and thus cultured A549 and H520 in vitro with and without SB in the presence or absence of glucose and investigated how the treatment with SB affects the survival of lung cancer cells and its influence on immune cells fighting against lung cancer. Methods: In this study, we performed cell culture experiments with A549, H520 and NSCLC-patient-derived epithelial cells under different SB levels. To investigate the influence on the immune system, we performed in vitro culture of peripheral mononuclear blood cells (PBMC) from control, smoker and lung cancer patients with increasing SB concentrations. Results: To investigate the effect of SB on lung tumor cells, we first analyzed the effect of 6 different concentrations of SB on A549 cells at 48 and 72 hours cell culture. Here we found that, SB treatment reduced lung cancer cell survival in a concentration dependent manner. We next focused our deeper analysis on the two concentrations, which caused the maximal reduction in cell survival. Here, we observed that SB led to cell cycle arrest and induced early apoptosis in A549 lung cancer cells. The expression of cell cycle regulatory proteins and A549 lung cancer stem cell markers (CD90) was induced. Additionally, this study explored the role of interferon-gamma (IFN-γ) and its receptor (IFN-γ-R1) in combination with SB treatment, revealing that, although IFN-γ-R1 expression was increased, IFN-γ did not affect the efficacy of SB in reducing tumor cell viability. Furthermore, we examined the effects of SB on immune cells, specifically CD8+ T cells and natural killer (NK) cells from healthy individuals, smokers, and NSCLC patients. SB treatment resulted in a decreased production of IFN-γ and granzyme B in CD8+ T cells and NK cells. Moreover, SB induced IFN-γ-R1 in NK cells and CD4+ T cells in the absence of glucose both in PBMCs from controls and NSCLC subjects. Conclusion: Overall, this study highlights the potential of SB in inhibiting lung cancer cell growth, triggering apoptosis, inducing cell cycle arrest, and modulating immune responses by activating peripheral blood CD4+ T cells while selectively inducing IFN-γ-R1 in NK cells in peripheral blood and inhibiting peripheral blood CD8+ T cells and NK cells. Thus, understanding the mechanisms of action of SB in the TME and its influence on the immune system provide valuable insights of potentially considering SB as a candidate for adjunctive therapies in NSCLC.


Sujet(s)
Lymphocytes T CD4+ , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Acides gras volatils/pharmacologie , Acides gras volatils/métabolisme , Mâle , Femelle , Cellules A549 , Adulte d'âge moyen , Sujet âgé , Microenvironnement tumoral/immunologie , Acide butyrique/pharmacologie , Lignée cellulaire tumorale , Mort cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Interféron gamma/métabolisme
8.
Food Funct ; 15(7): 3640-3652, 2024 Apr 02.
Article de Anglais | MEDLINE | ID: mdl-38482709

RÉSUMÉ

This study aimed to investigate the effects of gac fruit juice and its probiotic fermentation (FGJ) utilizing Lactobacillus paracasei on the modulation of the gut microbiota and the production of short-chain fatty acids (SCFAs). We conducted a comparison between FGJ, non-fermented gac juice (GJ), and control samples through in vitro digestion and colonic fermentation using the human gut microbiota derived from fecal inoculum. Our findings revealed that both GJ and FGJ led to an increase in the viability of Lactobacilli, with FGJ exhibiting even higher levels compared to the control. The results from the 16S rDNA amplicon sequencing technique showed that both GJ and FGJ exerted positive impact on the gut microbiota by promoting beneficial bacteria, notably Lactobacillus mucosae and Bacteroides vulgatus. Additionally, both GJ and FGJ significantly elevated the levels of SCFAs, particularly acetic, propionic, and n-butyric acids, as well as lactic acid, in comparison to the control. Notably, FGJ exhibited a more pronounced effect on the gut microbiota compared to GJ. This was evident in its ability to enhance species richness, reduce the Firmicutes to Bacteroidetes (F/B) ratio, promote Akkermansia, and inhibit pathogenic Escherichia coli. Moreover, FGJ displayed enhanced production of SCFAs, especially acetic and lactic acids, in contrast to GJ. Our findings suggest that the probiotic fermentation of gac fruit enhances its functional attributes in promoting a balanced gut microbiota. This beverage demonstrates potential as a functional food with potential advantages for sustaining intestinal health.


Sujet(s)
Microbiome gastro-intestinal , Humains , Jus de fruits et de légumes , Fermentation , Acides gras volatils/pharmacologie , Fruit
9.
J Oral Sci ; 66(2): 125-129, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38494703

RÉSUMÉ

PURPOSE: Bacterial cells in mature dental plaque produce a high concentration of short-chain fatty acids (SCFAs) such as butyrate and propionate. SCFA-treatment on human gingival epithelial Ca9-22 cells induced cell death. However, the exact mechanism underlying cell death remains unclear. In this study, the relationship between reactive oxygen species (ROS) and autophagy induction during SCFA-induced cell death was examined. METHODS: Human gingival epithelial Ca9-22 cells were treated with butyrate or propionate to induce cell death and the number of dead cells were measured using SYTOX-green dye. A siRNA for ATG5 and N-acetylcysteine (NAC) were used for autophagy reduction and ROS-scavenging, respectively. Release of damage-associated molecular patterns (DAMPs) such as Sin3A-associated protein 130 (SAP130) and high-mobility group box 1 (HMGB1) were detected using western blot. RESULTS: Reducing autophagy significantly suppressed SCFA-induced Ca9-22 cell death. ROS generation was observed upon SCFA treatment, and scavenging ROS with NAC decreased cell death. NAC also reduced the SCFA-induced increase in microtubule-associated protein 1 light chain 3B (LC3B)-I and LC3B-II, and mitigated the release of DAMPs. CONCLUSION: The findings suggest that ROS generation is necessary for autophagy, which is required for SCFA-induced cell death and accompanying DAMP release.


Sujet(s)
Butyrates , Propionates , Humains , Butyrates/pharmacologie , Propionates/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Acides gras volatils/pharmacologie , Autophagie/physiologie
10.
Zhongguo Zhong Yao Za Zhi ; 49(1): 208-215, 2024 Jan.
Article de Chinois | MEDLINE | ID: mdl-38403353

RÉSUMÉ

This study aimed to investigate the regulatory effects of Zuogui Jiangtang Jieyu Formula(ZJJ) on the intestinal flora, short chain fatty acids(SCFAs), and neuroinflammation in rats with diabetes mellitus complicated depression(DD). The DD model was established in rats and model rats were randomly divided into a model group, a positive drug(metformin + fluoxetine) group, a ZJJ low-dose group, and a ZJJ high-dose group, with eight rats in each group. Another eight rats were assigned to the blank group. Subsequently, depressive-like behavior test was conducted on the rats, and cerebrospinal fluid samples were collected to measure pro-inflammatory cytokines [interleukin-1ß(IL-1ß), interleukin-6(IL-6), and tumor necrosis factor-α(TNF-α)]. Blood serum samples were collected to measure proteins related to the hypothalamic-pituitary-adrenal axis(HPA axis), including corticotropin-releasing hormone(CRH), adrenocorticotropic hormone(ACTH), and cortisol(CORT), as well as glucose metabolism. Gut contents were collected from each group for 16S rRNA sequencing analysis of intestinal flora and SCFAs sequencing. The results indicated that ZJJ not only improved glucose metabolism in DD rats(P<0.01) but also alleviated depressive-like behavior(P<0.05) and HPA axis hyperactivity(P<0.05 or P<0.01). Besides, it also improved the neuroinflammatory response in the brain, as evidenced by a significant reduction in pro-inflammatory cytokines in cerebrospinal fluid(P<0.05 or P<0.01). Additionally, ZJJ improved the intestinal flora, causing the intestinal flora in DD rats to resemble that of the blank group, characterized by an increased Firmicutes abundance. ZJJ significantly increased the levels of SCFAs(acetic acid, butyric acid, valeric acid, and isovaleric acid)(P<0.01). Therefore, it is deduced that ZJJ can effectively ameliorate intestinal flora dysbiosis, regulate SCFAs, and thereby improve both glucose metabolism disturbances and depressive-like behavior in DD.


Sujet(s)
Diabète , Médicaments issus de plantes chinoises , Microbiome gastro-intestinal , Rats , Animaux , Axe hypothalamohypophysaire/métabolisme , Dépression/traitement médicamenteux , ARN ribosomique 16S , Axe hypophyso-surrénalien/métabolisme , Corticolibérine/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Glucose/métabolisme , Acides gras volatils/métabolisme , Acides gras volatils/pharmacologie
11.
J Oral Sci ; 66(2): 102-106, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38417878

RÉSUMÉ

PURPOSE: Alveolar osteitis (dry sockets) is a painful condition characterized by a limited immune response. It is typically caused by the removal of blood clots from extracted tooth sockets, which leads to the fermentation of trapped food remnants by oral bacteria in the cavities, producing high concentrations of short-chain fatty acids (SCFAs). This study examined the effects of SCFAs on immunity and bone metabolism. METHODS: Mouse macrophage Raw264.7 cells were treated with oral bacteria supernatants or SCFA mixtures, and inducible nitric oxide synthase (iNOS) levels were determined by western blot. The same cells were treated with SCFA mixtures in the presence of receptor activator of nuclear factor-kappa B ligand (RANKL), and osteoclast-like cells were counted. MC3T3-E1 cells were treated with SCFA mixtures and stained with alizarin red S. RESULTS: Raw264.7 cells treated with oral bacterial culture supernatants of Porphyromonas gingivalis and Fusobacterium nucleatum inhibited lipopolysaccharide (LPS)-induced iNOS production, likely due to SCFA content. SCFA mixtures mimicking these supernatants inhibited the number of RANKL-induced tartrate-resistant acid phosphatase (TRAP)-positive cells and MC3T3-E1 cell mineralization. CONCLUSION: These data suggest that SCFAs produced by P. gingivalis and F. nucleatum may reduce the inflammatory response and mildly induce mineralization of the alveolar walls. These results may contribute to the understanding of alveolar osteitis.


Sujet(s)
Alvéolite , Souris , Animaux , Alvéolite/métabolisme , Ostéoclastes , Porphyromonas gingivalis , Tartrate-resistant acid phosphatase/métabolisme , Acides gras volatils/métabolisme , Acides gras volatils/pharmacologie
12.
Gut Microbes ; 16(1): 2297872, 2024.
Article de Anglais | MEDLINE | ID: mdl-38165200

RÉSUMÉ

Hyperbaric oxygen (HBO) therapy is a well-established method for improving tissue oxygenation and is typically used for the treatment of various inflammatory conditions, including infectious diseases. However, its effect on the intestinal mucosa, a microenvironment known to be physiologically hypoxic, remains unclear. Here, we demonstrated that daily treatment with hyperbaric oxygen affects gut microbiome composition, worsening antibiotic-induced dysbiosis. Accordingly, HBO-treated mice were more susceptible to Clostridioides difficile infection (CDI), an enteric pathogen highly associated with antibiotic-induced colitis. These observations were closely linked with a decline in the level of microbiota-derived short-chain fatty acids (SCFAs). Butyrate, a SCFA produced primarily by anaerobic microbial species, mitigated HBO-induced susceptibility to CDI and increased epithelial barrier integrity by improving group 3 innate lymphoid cell (ILC3) responses. Mice displaying tissue-specific deletion of HIF-1 in RORγt-positive cells exhibited no protective effect of butyrate during CDI. In contrast, the reinforcement of HIF-1 signaling in RORγt-positive cells through the conditional deletion of VHL mitigated disease outcome, even after HBO therapy. Taken together, we conclude that HBO induces intestinal dysbiosis and impairs the production of SCFAs affecting the HIF-1α-IL-22 axis in ILC3 and worsening the response of mice to subsequent C. difficile infection.


Sujet(s)
Clostridioides difficile , Infections à Clostridium , Microbiome gastro-intestinal , Oxygénation hyperbare , Souris , Animaux , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires , Immunité innée , Oxygénation hyperbare/effets indésirables , , Dysbiose/thérapie , Lymphocytes , Butyrates/pharmacologie , Acides gras volatils/pharmacologie , Antibactériens/pharmacologie
13.
Ocul Surf ; 32: 48-57, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38224777

RÉSUMÉ

PURPOSE: Short chain fatty acids (SCFAs) produced by gut microbiota are known to play primary roles in gut homeostasis by immunomodulation partially through G-protein coupled receptors (GPR) 43. Using mouse models of TLR ligand induced keratitis, we investigated whether SCFAs and GPR43 play any regulatory roles in the pathogenesis of inflammatory responses in the eye. METHODS: Both human and mouse eyes were labeled with a specific antibody for GPR43 and imaged by a laser scanning confocal microscope. Corneal cups from naïve C57BL/6J (B6) and GPR43 knockout (KO) mice were stimulated with TLR ligands in the presence or absence of sodium butyrate overnight and then processed for RT-PCR assay for expression of GPR43 and cytokines. Keratitis was induced by Poly I:C in wild type (WT) B6, GPR43KO and chimeric mice and the disease severity was evaluated by the corneal fluorescein staining test, and infiltrating cell staining and calculating in corneal whole mount. RESULTS: GPR43 is expressed in both human and mouse eyes and the expression is bidirectionally regulated by TLR ligands and butyrate. Butyrate significantly inhibited inflammation caused by several TLR ligands such as Poly I:C, Flagellin, and CpG-ODN (TLR-3, 5 and 9 agonists, respectively) in WT, but not GPR43KO, mice. Butyrate inhibition of TLR-induced keratitis is mediated by the GPR43 expressed in tissue but not hematopoietic, cells. CONCLUSIONS: This is the first report to demonstrate of the protective effect of SCFAs on microbial keratitis, and the dynamic expression and anti-inflammatory function of GPR43 in the eye. SCFAs can modulate inflammation and immunity in the eye through GPR43.


Sujet(s)
Modèles animaux de maladie humaine , Acides gras volatils , Kératite , Récepteurs couplés aux protéines G , Animaux , Humains , Souris , Cornée/métabolisme , Cornée/anatomopathologie , Cytokines/métabolisme , Acides gras volatils/métabolisme , Acides gras volatils/pharmacologie , Kératite/métabolisme , Kératite/anatomopathologie , Ligands , Souris de lignée C57BL , Souris knockout , Microscopie confocale , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/génétique
14.
Am J Physiol Cell Physiol ; 326(2): C317-C330, 2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38073487

RÉSUMÉ

Small organic molecules in the intestinal lumen, particularly short-chain fatty acids (SCFAs) and glucose, have long been postulated to enhance calcium absorption. Here, we used 45Ca radioactive tracer to determine calcium fluxes across the rat intestine after exposure to glucose and SCFAs. Confirming previous reports, glucose was found to increase the apical-to-basolateral calcium flux in the cecum. Under apical glucose-free conditions, SCFAs (e.g., butyrate) stimulated the cecal calcium fluxes by approximately twofold, while having no effect on proximal colon. Since SCFAs could be absorbed into the circulation, we further determined whether basolateral SCFA exposure rendered some positive actions. It was found that exposure of duodenum and cecum on the basolateral side to acetate or butyrate increased calcium fluxes. Under butyrate-rich conditions, cecal calcium transport was partially diminished by Na+/H+ exchanger 3 (NHE3) inhibitor (tenapanor) and nonselective transient receptor potential vanilloid subfamily 6 (TRPV6) inhibitor (miconazole). To confirm the contribution of TRPV6 to SCFA-stimulated calcium transport, we synthesized another TRPV6 inhibitor that was demonstrated by in silico molecular docking and molecular dynamics to occlude TRPV6 pore and diminish the glucose- and butyrate-induced calcium fluxes. Therefore, besides corroborating the importance of luminal molecules in calcium absorption, our findings provided foundation for development of more effective calcium-rich nutraceuticals in combination with various absorptive enhancers, e.g., glucose and SCFAs.NEW & NOTEWORTHY Organic molecules in the intestinal lumen, e.g., glucose and short-chain fatty acids (SCFAs), the latter of which are normally produced by microfloral fermentation, can stimulate calcium absorption dependent on transient receptor potential vanilloid subfamily 6 (TRPV6) and Na+/H+ exchanger 3 (NHE3). A selective TRPV6 inhibitor synthesized and demonstrated by in silico docking and molecular dynamics to specifically bind to the pore domain of TRPV6 was used to confirm a significant contribution of this channel. Our findings corroborate physiological significance of nutrients and SCFAs in enhancing calcium absorption.


Sujet(s)
Calcium , Acides gras volatils , Rats , Animaux , Échangeur-3 de sodium-hydrogène/métabolisme , Calcium/métabolisme , Simulation de docking moléculaire , Acides gras volatils/pharmacologie , Acides gras volatils/métabolisme , Butyrates/pharmacologie , Protéines de transport/métabolisme , Duodénum/métabolisme , Glucose/métabolisme , Absorption intestinale
15.
J Nutr Biochem ; 124: 109534, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-37977404

RÉSUMÉ

Protein is the most important macro-nutrient when it comes to maximizing health, body composition, muscle growth, and recovery of body tissue. In recent years, it has been found that protein also plays an important role in metabolism and gut microbiota. This study was performed to investigate the effects of an isocaloric diet with different crude protein contents on the energy metabolism of Sprague-Dawley (SD) rats. Results revealed that compared with the 20% crude protein (CP; control) diet, the 38% CP diet improved serum parameters that are associated with dyslipidemia and glucose metabolic disorders in SD rats, whereas the 50% CP diet increased liver injury indicators and fatty acid synthesis-related genes and protein expression in the liver. Compared with the control diet, the 14% CP diet increased the abundance of colonic short-chain fatty acid-producing bacteria (Lachnospiraceae_NK4A136_group and Ruminiclostridium_9) and promoted colonic microbial cysteine and methionine metabolism, the 38% CP diet up-regulated colonic microbial lysine biosynthesis and degradation pathways, and the 50% CP diet down-regulated colonic mucosal cholesterol metabolism. Furthermore, the increase of multiple colonic enteropathogenic bacteria in the 50% CP group was associated with higher palmitic acid and stearic acid concentrations in the colonic microbes and lower cholesterol and arachidonic acid concentrations in the colonic mucosa. These findings revealed that the 14% CP and 38% CP diets improved rats' energy metabolism, while the 50% CP diet was accompanied by lipid metabolism imbalances and an increase in the abundance of multiple enteropathogenic bacteria.


Sujet(s)
Microbiome gastro-intestinal , Rats , Animaux , Rat Sprague-Dawley , Régime alimentaire , Acides gras volatils/pharmacologie , Cholestérol/pharmacologie , Métabolisme énergétique , Métabolisme lipidique
16.
Arthritis Rheumatol ; 76(4): 647-659, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-37994265

RÉSUMÉ

OBJECTIVE: The objective for this study was to evaluate the effects of short chain fatty acids (SCFAs) on arthritic bone remodeling. METHODS: We treated a recently described preclinical murine model of psoriatic arthritis (PsA), R26STAT3Cstopfl/fl CD4Cre mice, with SCFA-supplemented water. We also performed in vitro osteoclast differentiation assays in the presence of serum-level SCFAs to evaluate the direct impact of these microbial metabolites on maturation and function of osteoclasts. We further characterized the molecular mechanism of SCFAs by transcriptional analysis. RESULTS: The osteoporosis condition in R26STAT3Cstopfl/fl CD4Cre animals is attributed primarily to robust osteoclast differentiation driven by an expansion of osteoclast progenitor cells (OCPs), accompanied by impaired osteoblast development. We show that SCFA supplementation can rescue the osteoporosis phenotype in this model of PsA. Our in vitro experiments revealed an inhibitory effect of the SCFAs on osteoclast differentiation, even at very low serum concentrations. This suppression of osteoclast differentiation enabled SCFAs to impede osteoporosis development in R26STAT3Cstopfl/fl CD4Cre mice. Further interrogation revealed that bone marrow-derived OCPs from diseased mice expressed a higher level of SCFA receptors than those of control mice and that the progenitor cells in the bone marrow of SCFA-treated mice presented a modified transcriptomic landscape, suggesting a direct impact of SCFAs on bone marrow progenitors in the context of osteoporosis. CONCLUSION: We demonstrated how gut microbiota-derived SCFAs can regulate distal pathology (ie, osteoporosis) and identified a potential therapeutic option for restoring bone density in rheumatic disease, further highlighting the critical role of the gut-bone axis in these disorders.


Sujet(s)
Arthrite psoriasique , Ostéoporose , Souris , Animaux , Ostéoclastes/métabolisme , Arthrite psoriasique/métabolisme , Remodelage osseux , Différenciation cellulaire , Ostéoporose/métabolisme , Acides gras volatils/métabolisme , Acides gras volatils/pharmacologie
17.
Pharmacol Res ; 199: 107041, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38128856

RÉSUMÉ

Short-chain fatty acids (SCFAs), the main metabolites of gut microbiota, have been associated with lower blood glucose and lipid levels in diabetic mice. However, a comprehensive summary and comparison of the effects of different SCFA interventions on blood glucose and lipid levels in diabetic mice is currently unavailable. This study aims to compare and rank the effects of different types of SCFAs on blood glucose and lipid levels by collecting relevant animal research. A systematic search through PubMed, Embase, Cochrane Library, and Web of Science database was conducted to identify relevant studies from inception to March 17, 2023. Both pairwise meta-analysis and Bayesian network meta-analysis were used for statistical analyses. In total, 18 relevant studies involving 5 interventions were included after screening 3793 citations and 53 full-text articles. Notably, butyrate therapy (mean difference [MD] = -4.52, 95% confidence interval [-6.29, -2.75]), acetate therapy (MD = -3.12, 95% confidence interval [-5.79, -0.46]), and propionate therapy (MD = -2.96, 95% confidence interval [-5.66, -0.26]) significantly reduced the fasting blood glucose levels compared to the control group; butyrate therapy was probably the most effective intervention, with a surface under the cumulative ranking curve (SUCRA) value of 85.5%. Additionally, acetate plus propionate therapy was probably the most effective intervention for reducing total cholesterol (SUCRA = 85.8%) or triglyceride levels (SUCRA = 88.1%). These findings underscore the potential therapeutic implications of SCFAs for addressing metabolic disorders, particularly in type 2 diabetes mellitus.


Sujet(s)
Glycémie , Diabète expérimental , Diabète de type 2 , Acides gras volatils , Animaux , Souris , Acétates , Théorème de Bayes , Glycémie/effets des médicaments et des substances chimiques , Butyrates/pharmacologie , Butyrates/usage thérapeutique , Diabète expérimental/traitement médicamenteux , Diabète de type 2/traitement médicamenteux , Acides gras volatils/pharmacologie , Acides gras volatils/usage thérapeutique , Méta-analyse en réseau , Propionates
18.
Int J Mol Sci ; 24(23)2023 Nov 27.
Article de Anglais | MEDLINE | ID: mdl-38069141

RÉSUMÉ

With the increasing prevalence of sleep deprivation (SD)-related disorders, the effective treatment of sleep disorders has become a critical health research topic. Thus, we hypothesized and investigated the effectiveness of a 3-week melatonin intervention on neuropsychiatric behavioral responses mediated throughout melatonin receptors, gut microbiota, and lipid metabolites in rats with chronic SD. Eighteen 6-week-old Wistar rats were used and divided into the control grup (C, n = 6), SD group (n = 6), and melatonin-supplemented group (SDM, n = 6). During weeks 0 to 6, animals were provided with the AIN-93M diet and free access to water. Four-week chronic SD was conducted from weeks 7 to 10. Exogenous melatonin administration (10 mg/kg BW) was injected intraperitoneally 1 h before the daily administration of SD for 3 weeks in the SDM group. SD rats exhibited anxiety-like behavior, depression-like behavior, and cognitive impairment. Exogenous melatonin administration ameliorated neuropsychiatric behaviors induced by chronic SD. Analysis of fecal metabolites indicated that melatonin may influence brain messaging through the microbiota-gut-brain axis by increasing the production of short-chain fatty acids (SCFA) and decreasing the production of secondary bile acids (SBA). Four-week SD reduced the cerebral cortex expression of MT1, but not in the colon. Chronic SD led to anxiety and depression-like behaviors and cognitive decline, as well as the reduced intestinal level of SCFAs and the enhanced intestinal level of SBAs in rats. In this work, we confirmed our hypothesis that a 3-week melatonin intervention on neuropsychiatric behavioral response mediated throughout melatonin receptors, gut microbiota, and lipid metabolites in rats with chronic SD.


Sujet(s)
Microbiome gastro-intestinal , Mélatonine , Microbiote , Rats , Animaux , Privation de sommeil/traitement médicamenteux , Privation de sommeil/complications , Mélatonine/pharmacologie , Mélatonine/usage thérapeutique , Récepteurs à la mélatonine , Rat Wistar , Acides gras volatils/pharmacologie
19.
Food Funct ; 14(23): 10329-10346, 2023 Nov 27.
Article de Anglais | MEDLINE | ID: mdl-37955225

RÉSUMÉ

Maca is a functional food with anti-inflammatory activity, and it is rich in protein. Currently, inflammatory bowel disease (IBD) is a common gastrointestinal disease. However, there is little research focusing on the effect of maca protein (MCP) on IBD. In this study, we extracted MCP from maca root and explored its effect and mechanism on improving dextran sodium sulfate (DSS)-induced IBD in mice. The results indicated that MCP intervention alleviated the clinical symptoms and colon tissue damage of mice with DSS-induced colitis and inhibited the expression of inflammatory factors. Moreover, it can modulate the gut microbiota composition in mice with DSS-induced colitis. The regulation is achieved by reducing the relative abundance of the IBD-exacerbating key bacterial genera: Lachnospiraceae_NK4A136_group, Bacteroides, Desulfovibrio, Prevotella, Helicobacter and Sutterella, while increasing the relative abundance of the IBD-alleviating key bacterial genera: norank_f_Muribaculaceae, Lactobacillus, Oscillospira, Akkermansia and Bifidobacterium. MCP can also promote the production of short-chain fatty acids (SCFAs). The further western blotting results indicated that MCP can regulate the Treg/Th17 immune balance in mice with colitis via the SCFAs-GPR41/43/HDAC1 signaling pathway. Overall, MCP can alleviate colitis by comprehensively regulating the gut microbiota and inflammatory response. It may be a promising functional component that reduces the risk of colitis by maintaining intestinal health.


Sujet(s)
Colite , Microbiome gastro-intestinal , Maladies inflammatoires intestinales , Animaux , Souris , Colite/induit chimiquement , Colite/traitement médicamenteux , Colite/métabolisme , Côlon , Acides gras volatils/pharmacologie , Sulfate dextran/effets indésirables , Souris de lignée C57BL , Modèles animaux de maladie humaine
20.
Food Funct ; 14(23): 10401-10417, 2023 Nov 27.
Article de Anglais | MEDLINE | ID: mdl-37955584

RÉSUMÉ

Gut microbiota are closely related to lipopolysaccharide (LPS)-induced acute lung injury (ALI). Akkermansia muciniphila (A. muciniphila) maintains the intestinal barrier function and regulates the balance of reduced glutathione/oxidized glutathione. However, it may be useful as a treatment strategy for LPS-induced lung injury. Our study aimed to explore whether A. muciniphila could improve lung injury by affecting the gut microbiota. The administration of A. muciniphila effectively attenuated lung injury tissue damage and significantly decreased the oxidative stress and inflammatory reaction induced by LPS, with lower levels of myeloperoxidase (MDA), enhanced superoxide dismutase (SOD) activity, decreased pro-inflammatory cytokine levels, and reduced macrophage and neutrophil infiltration. Moreover, A. muciniphila maintained the intestinal barrier function, reshaped the disordered microbial community, and promoted the secretion of short-chain fatty acids (SCFAs). A. muciniphila significantly downregulated the expression of TLR2, MyD88 and NF-kappa B (P < 0.05). Butyrate supplementation demonstrated a significant improvement in the inflammatory response (P < 0.05) and mitigation of histopathological damage in mice with ALI, thereby restoring the intestinal butyric acid concentration. In conclusion, our findings indicate that A. muciniphila inhibits the accumulation of inflammatory cytokines and attenuates the activation of the TLR2/Myd88/NF-κB pathway due to exerting anti-inflammatory effects through butyrate. This study provides an experimental foundation for the potential application of A. muciniphila and butyrate in the prevention and treatment of ALI.


Sujet(s)
Lésion pulmonaire aigüe , Microbiome gastro-intestinal , Animaux , Souris , Lipopolysaccharides/effets indésirables , Facteur de différenciation myéloïde-88/métabolisme , Récepteur de type Toll-2/métabolisme , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/traitement médicamenteux , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Cytokines/métabolisme , Acides gras volatils/pharmacologie , Acide butyrique/pharmacologie , Poumon
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...