Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Crit Rev Microbiol ; : 1-18, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38488586

RESUMO

The uterine environment provides necessary conditions for the existence of endometrial microbiota, which in turn plays an important role in maintaining the homeostasis of the uterine environment. The endometrial microbiome is highly susceptible to external factors such as age, hormones, menstrual, pregnancy, etc. When the microbiota is imbalanced, it will further promote the occurrence of uterine diseases such as endometritis and endometrial cancer. Regulating the microbiome of the endometrium is of positive significance for promoting uterine health. Among them, antibiotics, probiotics, prebiotics, and microbial transplantation may be important pathways for regulating endometrial microbiota in the future. However, there is currently no unified plan for evaluating the endometrial microbiota. In addition, due to the small sample size, it is easy to be contaminated by exogenous bacterial DNA, which poses great challenges for studying the mechanism of microbial community regulating uterine health. Therefore, there are still many areas worth exploring for the future of endometrial microbiome.

2.
PLoS Pathog ; 19(11): e1011764, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37948460

RESUMO

Subacute ruminal acidosis (SARA) has been demonstrated to promote the development of mastitis, one of the most serious diseases in dairy farming worldwide, but the underlying mechanism is unclear. Using untargeted metabolomics, we found hexadecanamide (HEX) was significantly reduced in rumen fluid and milk from cows with SARA-associated mastitis. Herein, we aimed to assess the protective role of HEX in Staphylococcus aureus (S. aureus)- and SARA-induced mastitis and the underlying mechanism. We showed that HEX ameliorated S. aureus-induced mastitis in mice, which was related to the suppression of mammary inflammatory responses and repair of the blood-milk barrier. In vitro, HEX depressed S. aureus-induced activation of the NF-κB pathway and improved barrier integrity in mouse mammary epithelial cells (MMECs). In detail, HEX activated PPARα, which upregulated SIRT1 and subsequently inhibited NF-κB activation and inflammatory responses. In addition, ruminal microbiota transplantation from SARA cows (S-RMT) caused mastitis and aggravated S. aureus-induced mastitis, while these changes were reversed by HEX. Our findings indicate that HEX effectively attenuates S. aureus- and SARA-induced mastitis by limiting inflammation and repairing barrier integrity, ultimately highlighting the important role of host or microbiota metabolism in the pathogenesis of mastitis and providing a potential strategy for mastitis prevention.


Assuntos
Mastite , Staphylococcus aureus , Humanos , Feminino , Animais , Camundongos , Bovinos , Staphylococcus aureus/metabolismo , NF-kappa B/metabolismo , Leite , Mastite/metabolismo
3.
Front Cell Infect Microbiol ; 13: 1282431, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37868345

RESUMO

The microbiota gut brain (MGB) axis has been shown to play a significant role in the regulation of inflammatory and infective diseases. Exploring the structure and communication mode of MGB axis is crucial for understanding its role in diseases, and studying the signaling pathways and regulatory methods of MGB axis regulation in diseases is also of profound significance for future clinical research. This article reviews the composition, communication mechanism of MGB axis and its role in inflammatory and infective diseases, including Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), autism spectrum disorder (ASD), depression, psoriasis, irritable bowel syndrome (IBS), and inflammatory bowel diseases (IBD). In addition, our investigation delved into the regulatory functions of the inflammasome, IFN-I, NF-κB, and PARK7/DJ-1 innate immune signaling pathway in the context of inflammatory and infective diseases. Ultimately, we discussed the efficacy of various interventions, including fecal microbiota transplantation (FMT), antibiotics, probiotics, prebiotics, synbiotics, and postbiotics, in the management of inflammatory and infective diseases. Understanding the role and mechanism of the MGB axis might make positive effects in the treatment of inflammatory and infective diseases.


Assuntos
Transtorno do Espectro Autista , Doenças Transmissíveis , Microbioma Gastrointestinal , Probióticos , Humanos , Eixo Encéfalo-Intestino , Microbioma Gastrointestinal/fisiologia , Probióticos/uso terapêutico , Imunidade Inata , Encéfalo
4.
Microb Pathog ; 182: 106225, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37419220

RESUMO

Mastitis is one of the common diseases in dairy cows which threatens the health of cows and impacts on economic benefits seriously. Recent studies have been showed that Subacute Ruminal Acidosis (SARA) increased the susceptibility of cow mastitis. SARA leads the disturbance of the rumen microbiota, and the rumen bacterial disordered community is an important endogenous factor of cow mastitis. That is to say, cows which suffer from SARA have a disordered rumen microbiota, a prolonged decline in ruminal PH and a high level of lipopolysaccharide (LPS) in the rumen, blood. Therefore, ruminal metabolism is closely related to the rumen microbiota. However, the specific mechanism of SARA and mastitis still not clear. We found an intestinal metabolite according to the metabonomics, which is correlated to inflammation. Phytophingosine (PS), a product from rumen fluid and milk of the cows which suffer from SARA and mastitis. It has the effect of killing bacteria and anti-inflammatory. Emerging evidences indicate that PS can alleviate inflammatory diseases. However, how PS affects mastitis is largely unknown. In this study, we explored the concrete role of PS on Staphylococcus aureus (S. aureus) -induced mastitis in mice. We found that PS obviously decreased the level of the proinflammatory cytokines. Meanwhile, PS also significantly relieved the mammary gland inflammation caused by S. aureus and restored the function of the blood-milk barrier. Here, we showed that PS increased the expression of the classic Tight-junctions (TJs) proteins including ZO-1, Occludin and Claudin-3. Moreover, PS improves S. aureus-induced mastitis by inhibiting the activation of the NF-κB and NLRP3 signaling pathways. These data indicated that PS relieved S. aureus-induced mastitis effectively. This also provides a reference for exploring the correlation between the intestinal metabolism and inflammation.


Assuntos
Doenças dos Bovinos , Mastite , Humanos , Feminino , Animais , Bovinos , Camundongos , Leite/metabolismo , Staphylococcus aureus , Rúmen/metabolismo , Mastite/tratamento farmacológico , Inflamação/metabolismo , Concentração de Íons de Hidrogênio , Dieta/veterinária , Lactação , Doenças dos Bovinos/metabolismo
5.
Microbiome ; 11(1): 78, 2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37069691

RESUMO

BACKGROUND: Mastitis is one of the most severe diseases in humans and animals, especially on dairy farms. Mounting evidence indicates that gastrointestinal dysbiosis caused by induction of subacute ruminal acidosis (SARA) by high-grain diet consumption and low in dietary fiber is associated with mastitis initiation and development, however, the underlying mechanism remains unknown. RESULTS: In the present study, we found that cows with SARA-associated mastitis have altered metabolic profiles in the rumen, with increased sialic acids level in particular. Consumption of sialic acid (SA) in antibiotic-treated mice, but not healthy mice, induced marked mastitis. SA treatment of antibiotic-treated mice also induced mucosal and systemic inflammatory responses, as evidenced by increased colon and liver injuries and several inflammatory markers. In addition, gut dysbiosis caused by antibiotic impaired gut barrier integrity, which was aggravated by SA treatment. SA potentiated serum LPS level caused by antibiotic treatment, leading to increased activation of the TLR4-NF-κB/NLRP3 pathways in the mammary gland and colon. Moreover, SA facilitated gut dysbiosis caused by antibiotic, and especially enhanced Enterobacteriaceae and Akkermansiaceae, which correlated with mastitis parameters. Fecal microbiota transplantation from SA-antibiotic-treated mice mimicked mastitis in recipient mice. In vitro experiments showed that SA prompted Escherichia coli growth and virulence gene expression, leading to higher proinflammatory cytokine production in macrophages. Targeting the inhibition of Enterobacteriaceae by sodium tungstate or treating with the commensal Lactobacillus reuteri alleviated SA-facilitated mastitis. In addition, SARA cows had distinct ruminal microbial structure by the enrichment of SA-utilizing opportunistic pathogenic Moraxellaceae and the depletion of SA-utilizing commensal Prevotellaceae. Treating mice with the specific sialidase inhibitor zanamivir reduced SA production and Moraxellaceae abundance, and improved mastitis in mice caused by ruminal microbiota transplantation from cows with SARA-associated mastitis. CONCLUSIONS: This study, for the first time, indicates that SA aggravates gut dysbiosis-induced mastitis by promoting gut microbiota disturbance and is regulated by commensal bacteria, indicating the important role of the microbiota-gut-mammary axis in mastitis pathogenesis and suggesting a potential strategy for mastitis intervention based on gut metabolism regulation. Video Abstract.


Assuntos
Microbioma Gastrointestinal , Mastite , Microbiota , Humanos , Feminino , Animais , Bovinos , Camundongos , Microbioma Gastrointestinal/fisiologia , Ácido N-Acetilneuramínico , Disbiose/induzido quimicamente , Enterobacteriaceae , Escherichia coli
7.
PLoS Pathog ; 19(1): e1011108, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36656870

RESUMO

Mounting evidence suggests that the gut microbiota plays an important role in the pathogenesis of mastitis, an important disease affecting the health of lactating women and the development of the dairy industry. However, the effect of the regulation of the gut microbiota by dietary components on mastitis development remains unknown. In this study, we found that a fiber-enriched diet alleviated Staphylococcus aureus (S. au)-induced mastitis in mice, which was dependent on the gut microbiota as depletion of the gut microbiota by antibiotics abolished this protective effect. Likewise, fecal microbiota transplantation (FMT) from high-inulin (HI)-treated mice (HIF) to recipient mice improved S. au-induced mastitis in mice. Consumption of an HI diet and HIF increased fecal short-chain fatty acid (SCFA) levels compared with the control group. Moreover, treatment with SCFAs, especially butyrate, alleviated S. au-induced mastitis in mice. Mechanistically, consumption of an HI diet enhanced the host antimicrobial program in macrophages through inhibiting histone deacetylase 3 by the production of butyrate. Collectively, our results suggest that modulation of the gut microbiota and its metabolism by dietary components is a potential strategy for mastitis intervention and serve as a basis for other infectious diseases.


Assuntos
Butiratos , Mastite , Animais , Feminino , Camundongos , Antibacterianos/farmacologia , Dieta , Lactação , Macrófagos , Mastite/terapia , Staphylococcus aureus , Fibras na Dieta
8.
Microbiome ; 10(1): 205, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36451232

RESUMO

BACKGROUND: Mounting experimental evidence has shown that the gut microbiota plays a significant role in the pathogenesis of mastitis, and clinical investigations have found that the occurrence of mastitis is correlated with ruminal dysbiosis. However, the underlying mechanism by which the ruminal microbiota participates in the development of mastitis remains unknown. RESULTS: In the present study, we found that cows with clinical mastitis had marked systemic inflammation, which was associated with significant ruminal dysbiosis, especially enriched Proteobacteria in the rumen. Ruminal microbiota transplantation from mastitis cows (M-RMT) to mice induced mastitis symptoms in recipient mice along with increased mammary proinflammatory signature activation of the TLR4-cGAS-STING-NF-κB/NLRP3 pathways. M-RMT also induced mucosal inflammation and impaired intestinal barrier integrity, leading to increased endotoxemia and systemic inflammation. Moreover, we showed that M-RMT mirrored ruminal microbiota disruption in the gut of recipient mice, as evidenced by enriched Proteobacteria and similar bacterial functions, which were correlated with most proinflammatory parameters and serum lipopolysaccharide (LPS) levels in mice. Recurrent low-grade LPS treatment mirrored gut dysbiosis-induced endotoxemia and caused severe mastitis in mice. Furthermore, we found that gut dysbiosis-derived LPS reduced host alkaline phosphatase activity by activating neuraminidase (Neu), which facilitates low-grade LPS exposure and E. coli-induced mastitis in mice. Conversely, treatment with calf intestinal alkaline phosphatase or the Neu inhibitor zanamivir alleviated low-grade LPS exposure and E. coli-induced mastitis in mice. CONCLUSIONS: Our results suggest that ruminal dysbiosis-derived low-grade endotoxemia can cause mastitis and aggravate pathogen-induced mastitis by impairing host anti-inflammatory enzymes, which implies that regulating the ruminal or gut microbiota to prevent low-grade systemic inflammation is a potential strategy for mastitis intervention. Video Abstract.


Assuntos
Endotoxemia , Mastite , Feminino , Humanos , Animais , Bovinos , Camundongos , Disbiose , Lipopolissacarídeos , Fosfatase Alcalina , Escherichia coli , Anti-Inflamatórios , Inflamação , Proteobactérias
9.
Cell Rep ; 41(8): 111681, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36417859

RESUMO

The precise mechanism by which gut dysbiosis contributes to the pathogenesis of extraintestinal diseases and how commensal microbes mediate these processes remain unclear. Here, we show that cows with mastitis had marked gut dysbiosis, characterized by the enrichment of opportunistic pathogenic Escherichia_Shigella and the depletion of commensal Roseburia. Fecal microbiota transplantation from donor cows with mastitis (M-FMT) to recipient mice significantly caused mastitis and changed the gut and mammary microbiota in mice. Notably, M-FMT facilitated the translocation of pathobiont from the gut into the mammary gland, and the depletion of Enterobacteriaceae alleviated M-FMT-induced mastitis in mice. In contrast, commensal Roseburia intestinalis improved M-FMT-induced mastitis and microbial dysbiosis in the gut and mammary gland and limited bacterial translocation by producing butyrate, which was associated with inflammatory signaling inhibition and barrier repair. Our research suggests that commensal Roseburia alleviates gut-dysbiosis-induced mastitis, although further studies in dairy cows and humans are needed.


Assuntos
Microbioma Gastrointestinal , Mastite , Feminino , Bovinos , Camundongos , Animais , Humanos , Disbiose/complicações , Translocação Bacteriana , Butiratos/farmacologia , Microbioma Gastrointestinal/fisiologia , Mastite/complicações
10.
Ecotoxicol Environ Saf ; 245: 114123, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36183427

RESUMO

Cadmium (Cd) is a type of high-risk heavy metal that can damage organs such as the liver, but its mechanism is not yet clear. Ferroptosis is a newly discovered mode of regulatory cell death. We explored whether ferroptosis is involved in Cd-induced liver damage and the underlying mechanism. Our research showed that Cd induced liver damage by inducing ferroptosis, and the use of ferroptosis inhibitors reduced the degree of liver damage. Moreover, the occurrence of ferroptosis was accompanied by the activation of the PERK-eIF2α-ATF4-CHOP signaling pathway, and inhibiting endoplasmic reticulum (ER) stress reduced ferroptosis demonstrating that ferroptosis induced by Cd is dependent on ER stress. In addition, chloroquine, a common autophagy inhibitor, mitigated ferroptosis caused by Cd exposure. Then, the iron chelator deferoxamine reduced Cd-induced lipid peroxidation and cell death, demonstrating that the iron regulation disorder caused by ferritin phagocytosis contributes to the Cd-induced ferroptosis. In conclusion, our results show that Cd-induced liver toxicity is accompanied by ferroptosis, which contributes to Cd inducing oxidative stress to trigger autophagy and ER stress to promote the process of ferroptosis.


Assuntos
Ferroptose , Hepatopatias , Autofagia , Cádmio/metabolismo , Cádmio/toxicidade , Cloroquina , Desferroxamina , Estresse do Retículo Endoplasmático , Ferritinas , Humanos , Ferro/metabolismo , Quelantes de Ferro
11.
FEMS Microbiol Ecol ; 98(7)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35648454

RESUMO

The colonization and virulence production of Staphylococcus aureus (S. aureus), a known pathogen that induces mastitis, depend on its quorum-sensing (QS) system and biofilm formation. It has been reported that Bacillus can inhibit the QS system of S. aureus, thereby reducing S. aureus colonization in the intestine. However, whether Bacillus affects S. aureus biofilm formation and consequent colonization during mastitis is still unknown. In this study, the differences in the colonization of S. aureus and Bacillus were first analyzed by isolating and culturing bacteria from milk samples. It was found that the colonization of Bacillus and S. aureus in cow mammary glands was negatively correlated. Secondly, we found that although Bacillus did not affect S. aureus growth, it inhibited the biofilm formation of S. aureus by interfering its QS signaling. The most significant anti-biofilm effect was found in Bacillus subtilis H28 (B. subtilis H28). Finally, we found that B. subtilis H28 treatment alleviated S. aureus-induced mastitis in a mice model. Our results rerealed that bovine milk derived commensal Bacillus inhibited S. aureus colonization and alleviated S. aureus-induced mastitis by influencing biofilm formation, suggesting a potential targeted strategy to limit the colonization of S. aureus in vivo.


Assuntos
Bacillus , Mastite Bovina , Infecções Estafilocócicas , Animais , Bacillus subtilis , Biofilmes , Bovinos , Feminino , Humanos , Mastite Bovina/microbiologia , Mastite Bovina/prevenção & controle , Camundongos , Leite/microbiologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus
12.
Microbiol Spectr ; 10(4): e0081122, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-35727038

RESUMO

Intestinal microbiota-mediated aryl hydrocarbon receptor (AhR) activation plays an important role in host-microbiota interactions and disease development. However, whether AhR activation mediates infection-induced inflammation in remote organs is not clear. The purpose of this study is to assess the effects and underlying mechanism of AhR activation and gut microbiota-mediated dietary tryptophan (Trp) metabolism on infection-induced inflammation using an Escherichia coli (E. coli)-induced endometritis model in mice. We found that AhR activation by 6-formylindolo (3,2-b) carbazole (Ficz), which is an AhR agonist derived from the photooxidation of Trp, alleviated E. coli-induced endometritis by repairing barrier function and inhibiting inflammatory responses, while inhibition of AhR by CH223191, which is a synthetic AhR antagonist, aggravated E. coli-induced endometritis. Gut dysbiosis damaged AhR activation and exacerbated E. coli-induced endometritis in mice, which responded to the reduced abundance of AhR ligand producers, such as Lactobacillus spp. Supplementation with dietary Trp ameliorated E. coli-induced endometritis in a microbiota-dependent manner, which was associated with the production of AhR ligands. Administration of AhR ligands, including indole and indole aldehyde, but not indole-3-propionic acid, rescued the protective effect of Trp on E. coli-induced endometritis in dysbiotic mice. Moreover, consumption of Lactobacillus reuteri (L. reuteri) containing AhR ligand-producing capability also alleviated E. coli-induced endometritis in mice in an AhR-dependent manner. Our results demonstrate that microbiota-mediated AhR activation is a key factor in fighting pathogen-caused inflammation, which leads to a potential strategy to regulate the gut microbiota and metabolism by dietary Trp or probiotics for the intervention of infectious diseases and reproductive health. IMPORTANCE Infection-induced endometritis is a common and frequently occurring disease in humans and animals. Accumulating evidence suggests an important role of the gut microbiota in the development of infection-induced inflammation. Whether and how gut microbiota-mediated AhR activation regulates the pathogenesis of pathogen-induced endometritis remains unknown. The current study found that AhR activation ameliorated E. coli-induced endometritis, and inhibition of AhR produced negative results. Gut dysbiosis reduced the abundance of AhR ligand producers including Lactobacillus spp., damaged AhR activation, and exacerbated E. coli-induced endometritis. Supplementation with dietary Trp, AhR ligands, and L. reuteri containing AhR ligand-producing capability alleviated E. coli-induced endometritis in mice. Our results suggest an important role of microbiota-mediated AhR activation in the pathogenesis of endometritis and provide potential strategies for the intervention of infectious diseases and reproductive health by regulating the gut microbiota and metabolism.


Assuntos
Endometrite , Microbioma Gastrointestinal , Limosilactobacillus reuteri , Animais , Disbiose/terapia , Endometrite/terapia , Escherichia coli/metabolismo , Feminino , Humanos , Inflamação , Limosilactobacillus reuteri/metabolismo , Ligantes , Camundongos , Receptores de Hidrocarboneto Arílico/metabolismo , Triptofano/metabolismo
13.
Anal Chem ; 94(22): 7944-7951, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35616317

RESUMO

High-quality oxygen isotope analysis of composition-variable minerals (e.g., ubiquitous carbonates) using secondary ion mass spectrometry (SIMS) is extremely challenging. The classical off-line procedure, which requires additional electron probe microanalyzer (EPMA) chemical compositions for calibrating instrumental mass fractionation (IMF), is inherently inaccurate and analytically inefficient. In this study, the first accurate and paired SIMS analysis of δ18O and Fe# [molar Fe/(Mg + Fe)] in dolomite is reported. Based on five newly developed dolomite O-isotopic standards with an Fe# range of 0.01-0.35 obtained by SIMS, a novel accurate and rapid online matrix effect calibration method for dolomite O-isotope analysis was developed using concurrent SIMS 18O-16O-56Fe16O-24Mg16O measurements without additional chemical electron probe microanalysis. A logistic equation was proposed as the best-fit curve to represent the δ18O matrix effect based on the 56Fe16O/24Mg16O ratios. For CTD-4 carbonatitic dolomite with variable Fe# but homogeneous oxygen isotopes, the off-line method exhibited highly variable apparent δ18O values in the range of 5.74-10.11‰. The online method yielded a homogeneous δ18O value of 7.94 ± 0.34‰ (2SD, n = 40), which is comparable with that of bulk analysis (7.94 ± 0.20‰; 2SD). Comprehensive analyses validated the online method as the best strategy for performing accurate δ18O analysis of samples with highly heterogeneous compositions. Based on its accuracy, simplicity, and economic feasibility, this method has potential applications in the analysis of composition-complex dolomites, detrital dolomites, and other precious terrestrial and extraterrestrial materials.


Assuntos
Carbonato de Cálcio , Minerais , Carbonato de Cálcio/química , Calibragem , Magnésio , Isótopos de Oxigênio/química
14.
Sci Adv ; 8(19): eabn6045, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35559677

RESUMO

Biosilicification-the formation of biological structures composed of silica-has a wide distribution among eukaryotes; it plays a major role in global biogeochemical cycles, and has driven the decline of dissolved silicon in the oceans through geological time. While it has long been thought that eukaryotes are the only organisms appreciably affecting the biogeochemical cycling of Si, the recent discoveries of silica transporter genes and marked silicon accumulation in bacteria suggest that prokaryotes may play an underappreciated role in the Si cycle, particularly in ancient times. Here, we report a previously unidentified magnetotactic bacterium that forms intracellular, amorphous silica globules. This bacterium, phylogenetically affiliated with the phylum Nitrospirota, belongs to a deep-branching group of magnetotactic bacteria that also forms intracellular magnetite magnetosomes and sulfur inclusions. This contribution reveals intracellularly controlled silicification within prokaryotes and suggests a previously unrecognized influence on the biogeochemical Si cycle that was operational during early Earth history.


Assuntos
Magnetossomos , Silício , Bactérias/genética , Eucariotos , Óxido Ferroso-Férrico , Magnetossomos/genética , Dióxido de Silício
15.
Front Chem ; 10: 840473, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35464206

RESUMO

Hydrogen isotope analysis of coal is an important tool in the geochemical analysis of coal. The traditional method of hydrogen isotope analysis of coal requires the oxidation of organic matter bound hydrogen in coal to water by an oxidizing agent and then its reduction to hydrogen by a reducing agent. This method is time-consuming and laborious, and makes it difficult to cope with the rapid detection of large numbers of samples. The recent development of continuous flow IRMS systems (CF-IRMS) has solved the problem of inefficient analysis, but does not guarantee the quantitative conversion of organic bound H to H2, resulting in inaccurate measured hydrogen isotope values. In this study, for the hydrogen isotope analysis of coal, an alternative continuous flow system (Cr-EA-IRMS) was used to achieve high precision hydrogen isotope measurements of coal samples by filling a quartz reaction tube with Cr. The results obtained by this method (-121.3 ± 1.1‰) for the reference material (GBW11104) are consistent with those obtained by the conventional method (-121.4 ± 0.6‰). Using this method, hydrogen isotope measurements for a variety of imported coals revealed significant differences in the hydrogen isotopes of coals from different coal producing regions including Russia, South East Asia, and Australia. Therefore, the use of hydrogen isotope testing analysis of coal could be a potential means of tracing the origin of coal.

16.
Microb Pathog ; 166: 105487, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35429585

RESUMO

Endometritis is a common obstetric disease that occurs most frequently after parturition in a variety of animals. Animal infertility due to endometritis severely hinders animal husbandry and often causes serious economic losses to the dairy farming industry. According to reports, Bacillus subtilis (B. subtilis) can prevent pathogenic colonization of epithelial cells and exert immunostimulatory effects. The present study aimed to reveal the protective effect of B. subtilis on endometritis induced by Escherichia coli (E. coli) in mice. The experimental model required in this experiment was established by injecting E. coli intrauterinely, and different concentrations of B. subtilis H28 were administered 10 days before E. coli injection. The pathological changes in the uterine tissue of mice were assessed by haematoxylin-eosin (H&E) staining. Myeloperoxidase (MPO) activity measurements and enzyme-linked immunosorbent assay (ELISA) based measurement of pro-inflammatory cytokines levels were performed. Activation of NF-κB signaling pathway were detected by Western blot, and the changes in the levels of tight junction proteins (TJPs) was analyzed using Western blot detection and quantitative real-time polymerase chain reaction (qRT-PCR). As seen from the results, B. subtilis H28 pretreatment decreased uterine neutrophil infiltration, IL-1ß and TNF-α production, and the NF-κB activation during endometritis induced by E. coli. In addition, B. subtilis H28 significantly increased the expression of the tight junction proteins ZO-1, claudin-3 and occludin in uterine infected with E. coli. In conclusion, in the present study, we found that B. subtilis H28 ameliorated E. coli-induced endometritis by maintaining the endometrial barrier and inhibiting the inflammatory response.


Assuntos
Bacillus subtilis , Endometrite , Infecções por Escherichia coli , Animais , Citocinas/metabolismo , Endometrite/microbiologia , Endometrite/terapia , Escherichia coli/metabolismo , Infecções por Escherichia coli/terapia , Feminino , Camundongos , NF-kappa B/metabolismo , Proteínas de Junções Íntimas
17.
Food Chem Toxicol ; 163: 112909, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35292335

RESUMO

Bisphenol A (BPA) is a common environmental contaminant, whose exposure is associated with the progression of various kidney diseases. BPA exposure has turned out to be associated with cytotoxicity to renal tubular epithelial cells, but its underlying mechanism remains unknown. Herein, we found that BPA induced ferroptosis in kidney and renal tubular epithelial cells, as showed by increased intracellular iron accumulation, lipid peroxidation and cells death upon BPA exposure. Additionally, utilization of ferrostatin-1 and desferrioxamine, typical ferroptosis inhibitors, can fundamentally diminish cells death. Intriguingly, we discovered that autophagy inhibitor chloroquine can shield renal tubular epithelial cells from BPA-caused ferroptosis. Furthermore, we found that ferritinophagy, a phenomenon that degradation of ferritin and inducing subsequent iron overload, occurred after BPA exposure and excessive iron promoted ferroptosis through Fenton reaction. We next demonstrated that BPA activated the AMPK-mTOR-ULK1 signaling pathway. In turn, AMPK, mTOR, and ULK1 knockdown dramatically mitigated BPA-induced TCMK-1 cells death, and decreased MDA and LC3 levels, but increased FTH protein content. These results indicate that activation of the AMPK-mTOR-ULK1 signaling is involved in BPA-induced ferritinophagy. In conclusion, renal dysfunction and renal tubular epithelial damage induced by BPA are linked to ferroptosis, which depends on the activation of ferritinophagy through AMPK-mTOR-ULK1 axis.


Assuntos
Ferroptose , Proteínas Quinases Ativadas por AMP , Autofagia , Compostos Benzidrílicos , Células Epiteliais/metabolismo , Ferro/metabolismo , Fenóis , Serina-Treonina Quinases TOR/genética
18.
Microb Pathog ; 164: 105414, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35093486

RESUMO

Mastitis is part of the aggressive diseases that affecting the development of dairy farming. Lactic acid bacteria (LAB), an important microbiological agent of gastrointestinal flora, can effectively promote the development of the immune system. Herein, the objectives of this study is to explore the protective role of LAB on Staphylococcus aureus(S. aureus)-induced mastitis in mice. 88 strains of suspected LAB were isolated from the milk of healthy dairy cows. Antibacterial activity was screened, and the 16S rRNA sequence analysis showed that the bacteria were Enterococcus mundtii H81 (E. mundtii H81). Furthermore, the model of mastitis has been established by nipple duct injection of S. aureus in mice, while E. mundtii H81 was treated 2 h before S. aureus injection. Twenty-four hours later of S. aureus infection, the mammary gland tissues were collected. The pathological changes of the mammary gland were observed by H&E staining. The levels of TNF-α and IL-1ß were measured by ELISA and the myeloperoxidase (MPO) activity was measured by the MPO assay kit. We also observed the changes of nuclear transcription factor kappa B (NF-κB) by using western blotting. The results showed that E. mundtii H81 pretreatment reduced neutrophil infiltration, and significantly reduce the secretion of TNF-α and IL-1ß, down-regulate the phosphorylation of p65 NF-κB and IκB, and the expression of tight junction protein Claudin 3 and ZO-1 was up-regulated. Collectively, our findings showed that E. mundtii H81 protects mammary gland from S. aureus-induced mastitis, which may be a candidate of treatment for mastitis infected by S. aureus.


Assuntos
Mastite , Probióticos , Infecções Estafilocócicas , Animais , Bovinos , Enterococcus , Feminino , Humanos , Sistema de Sinalização das MAP Quinases , Glândulas Mamárias Animais , Mastite/microbiologia , Camundongos , NF-kappa B/metabolismo , RNA Ribossômico 16S/genética , Transdução de Sinais , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus , Receptor 2 Toll-Like/metabolismo
19.
Free Radic Biol Med ; 175: 236-248, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34520822

RESUMO

Acute cadmium (Cd) exposure is a significant risk factor for renal injury and lacks effective treatment strategies. Ferroptosis is a recently identified iron-dependent form of nonapoptotic cell death mediated by membrane damage resulting from lipid peroxidation, and it is implicated in many diseases. However, whether ferroptosis is involved in Cd-induced renal injury and, if so, how it operates. Here, we show that Cd can induce ferroptosis in kidney and renal tubular epithelial cells, as demonstrated by elevation of intracellular iron levels and lipid peroxidation, as well as impaired antioxidant production. Treatment with a ferroptosis inhibitor alleviated Cd-induced cell death. Intriguingly, we established that Cd-induced ferroptosis depended on endoplasmic reticulum (ER) stress, by demonstrating that Cd activated the PERK-eIF2α-ATF4-CHOP pathway and that inhibition of ER stress reduced ferroptosis caused by Cd. We further found that autophagy was required for Cd-induced ferroptosis because the inhibition of autophagy by chloroquine mitigated Cd-induced ferroptosis. Furthermore, we showed that iron dysregulation by ferritinophagy contributed to Cd-induced ferroptosis, by showing that the iron chelator desferrioxamine alleviated Cd-induced cell death and lipid peroxidation. In addition, ER stress is likely activated by MitoROS which trigger autophagy and ferroptosis. Collectively, our results indicate that ferroptosis is involved in Cd-induced renal toxicity and regulated by the MitoROS-ER stress-ferritinophagy axis.


Assuntos
Estresse do Retículo Endoplasmático , Ferroptose , Apoptose , Autofagia , Cádmio/toxicidade , Células Epiteliais
20.
Mol Immunol ; 137: 134-144, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34247099

RESUMO

Mastitis is one of the most serious diseases in humans and animals, especially in the modern dairy industry. Seeking safe and effective mastitis prevention strategies is urgent since food safety and drug residues in milk remain an enormous concern, despite the contribution of antibiotics to control mastitis. Kynurenic acid (KYNA), derived from the kynurenine pathway of tryptophan metabolism, has been shown to exhibit anti-inflammatory and immunomodulatory effects in many diseases. Recently, it was reported that impaired KYNA levels were associated with mastitis. However, the physiological role of KYNA in mastitis has not yet been elucidated. Therefore, the aim of this study was to investigate the protective role of KYNA in pathogen-induced mastitis in mice, as well as the underlying mechanism of this effect. We first evaluated the effects of KYNA on LPS-induced mastitis in mice. Additionally, the underlying anti-inflammatory mechanism of KYNA was investigated in mammary epithelial cells (MMECs). Furthermore, we examined the effects of KYNA on S. aureus and E. coli induced mastitis in mice. Our results demonstrated that KYNA alleviated LPS-induced mastitis by reducing inflammatory responses and enhancing blood-milk barrier integrity. The fundamental mechanisms involved the inhibition of NF-κB and activation of Nrf2/Ho-1, which is probably mediated by G protein-coupled receptor 35 but not aryl hydrocarbon receptor. Notably, KYNA also protected against S. aureus and E. coli induced mastitis in mice. In conclusion, our results highlight the role of KYNA in mastitis and serve as a basis for using endogenous metabolite as a novel preventative or therapeutic strategy for disease intervention.


Assuntos
Inflamação/tratamento farmacológico , Ácido Cinurênico/farmacologia , Mastite/tratamento farmacológico , Leite/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Escherichia coli/patogenicidade , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Feminino , Heme Oxigenase-1/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/microbiologia , Mastite/metabolismo , Mastite/microbiologia , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA