Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Crit Rev Food Sci Nutr ; : 1-21, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38920093

ABSTRACT

Limosillactobacillus reuteri (L. reuteri), a type of Lactobacillus spp., stands out as the most extensively researched probiotic. Its remarkable intestinal adhesion has led to widespread applications in both the food and medical sectors. Notably, recent research highlights the probiotic efficacy of L. reuteri sourced from breast milk, particularly in influencing social behavior and mitigating atopic dermatitis. In this review, our emphasis is on surveying recent literature regarding the promotion of host's health by L. reuteri. We aim to provide a concise summary of the latest regulatory effects and potential mechanisms attributed to L. reuteri in the realms of metabolism, brain- and immune-related functions. The mechanism through which L. reuteri promotes host health by modulating the intestinal microenvironment primarily involves promoting intestinal epithelial renewal, bolstering intestinal barrier function, regulating gut microbiota and its metabolites, and suppressing inflammation and immune responses. Additionally, this review delves into new technologies, identifies shortcomings, and addresses challenges in current L. reuteri research. Finally, the application prospects of L. reuteri are provided. Therefore, a better understanding of the role and mechanisms of L. reuteri will contribute significantly to the development of new probiotic functional foods and enable precise, targeted interventions for various diseases.

2.
Heliyon ; 10(9): e30612, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38742057

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and healthcare burden worldwide. The progression of COPD is a combination of genetic predisposition and environmental factors, primarily cigarette smoking, and the underlying mechanisms are still unknown. Intestinal microecology impacts host immunity, metabolism, and resistance to pathogenic infections, which may be involved in pulmonary disease. Moreover, substantial interaction occurs between the intestinal and respiratory immune niches. After reviewing nearly 500 articles, we found the gut-lung axis plays an important role in the development of COPD. COPD patients often have dysbiosis of the intestinal microenvironment, which can affect host immunity through a series of mechanisms, exacerbating or protecting against COPD progression. This paper summarizes how the gut-lung axis influences COPD, including the alterations of intestinal microecology, the pathological mechanisms, and the involved immune responses. Finally, we summarize the latest research advances in COPD treatment from the perspective of regulating the gut-lung axis and intestinal immunity and evaluate the potential value of the gut-lung axis in improving COPD prognosis.

3.
Sci Rep ; 14(1): 9903, 2024 04 30.
Article in English | MEDLINE | ID: mdl-38688964

ABSTRACT

The edible fungus industry is one of the pillar industries in the Yunnan-Guizhou Plateau, China. The expansion of the planting scale has led to the release of various mushroom residues, such as mushroom feet, and other wastes, which are not treated adequately, resulting in environmental pollution. This study investigated the ability of black soldier fly (Hermetia illucens L.) larvae (BSFL) to degrade mushroom waste. Moreover, this study analyzed changes in the intestinal bacterial community and gene expression of BSFL after feeding on mushroom waste. Under identical feeding conditions, the remaining amount of mushroom waste in Pleurotus ostreatus treatment group was reduced by 18.66%, whereas that in Flammulina velutipes treatment group was increased by 31.08%. Regarding gut microbial diversity, compared with wheat bran-treated control group, Dysgonomonas, Providencia, Enterococcus, Pseudochrobactrum, Actinomyces, Morganella, Ochrobactrum, Raoultella, and Ignatzschineria were the most abundant bacteria in the midgut of BSFL in F. velutipes treatment group. Furthermore, Dysgonomonas, Campylobacter, Providencia, Ignatzschineria, Actinomyces, Enterococcus, Morganella, Raoultella, and Pseudochrobactrum were the most abundant bacteria in the midgut of BSFL in P. ostreatus treatment group. Compared with wheat bran-treated control group, 501 upregulated and 285 downregulated genes were identified in F. velutipes treatment group, whereas 211 upregulated and 43 downregulated genes were identified in P. ostreatus treatment group. Using Kyoto Encyclopedia of Genes and Genomes and Gene Ontology enrichment analyses, we identified 14 differentially expressed genes (DEGs) related to amino sugar and nucleotide sugar metabolism in F. velutipes treatment group, followed by 12 DEGs related to protein digestion and absorption. Moreover, in P. ostreatus treatment group, two DEGs were detected for fructose and mannose metabolism, and two were noted for fatty acid metabolism. These results indicate that feeding on edible mushroom waste can alter the intestinal microbial community structure of BSFL; moreover, the larval intestine can generate a corresponding feedback. These changes contribute to the degradation of edible mushroom waste by BSFL and provide a reference for treating edible mushroom waste using BSFL.


Subject(s)
Agaricales , Gastrointestinal Microbiome , Larva , Pleurotus , Animals , Larva/microbiology , Pleurotus/metabolism , Agaricales/metabolism , Agaricales/genetics , Biodegradation, Environmental , Diptera/microbiology , Diptera/metabolism , Flammulina/metabolism , Flammulina/genetics , Bacteria/metabolism , Bacteria/genetics , Bacteria/classification
4.
Nutr Rev ; 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553410

ABSTRACT

Context: Research regarding the treatment of inflammatory bowel disease (IBD) with probiotics has not yielded consistent results. OBJECTIVE: The aim of this meta-analysis was to evaluate the efficacy of probiotics supplementation in patients with IBD. DATA SOURCES: Randomized controlled trials (RCTs) evaluating the efficacy of probiotics in patients with IBD were searched in PubMed, the Google Scholar database, Web of Science, and CrossRef for the period July 2003 to June 2023. DATA EXTRACTION: The RCTs were extracted, independently by 2 authors, according to the PICOS criteria. DATA ANALYSIS: Seven studies, including a total of 795 patients, met the study criteria. Five end points were selected to evaluate the efficacy. Of these, 3 indicators showed a statistically significant difference in efficacy: C-reactive protein (odds ratio [OR]: -2.45, 95% confidence interval [CI]: -3.16, -1.73, P < .01), the number of fecal Bifidobacterium (OR: 3.37, 95% CI: 3.28, 3.47, P < .01), and Lactobacillus(OR: 2.00, 95% CI: 1.91, 2.09, P < .01). The other 2 indicators (disease activity for Crohn's disease and for ulcerative colitis) showed no statistically significant difference, while the OR reflected a positive correlation. CONCLUSION: Probiotics supplementation may have a positive effect on IBD by reducing clinical symptoms, reducing the serological inflammatory markers, and increasing favorable gut flora in patients with IBD. Additional RCTs are needed to evaluate the therapeutic effect of probiotics in IBD.

5.
Cell Commun Signal ; 22(1): 176, 2024 03 12.
Article in English | MEDLINE | ID: mdl-38475799

ABSTRACT

BACKGROUND: The impact of antidepressants on Inflammatory bowel diseases (IBD) has been extensively studied. However, the biological effects and molecular mechanisms of antidepressants in alleviating colitis remain unclear. METHODS: We systematically assessed how antidepressants (fluoxetine, fluvoxamine and venlafaxine) affected IBD and chose fluoxetine, the most effective one, for mechanism studies. We treated the C56BL/6 mice of the IBD model with fluoxetine and their controls. We initially assessed the severity of intestinal inflammation in mice by body weight loss, disease Activity Index scores and the length of the colon. The H&E staining and immunohistochemical staining of MUC2 of colon sections were performed to observe the pathological changes. RT-qPCR and western blot were conducted to assess the expression level of the barrier and inflammation-associated genes. Then, single-cell RNA sequencing was performed on mouse intestinal mucosa. Seurat was used to visualize the data. Uniform Manifold Approximation and Projection (UMAP) was used to perform the dimensionality reduction. Cell Chat package was used to perform cell-cell communication analysis. Monocle was used to conduct developmental pseudotime analysis. Last, RT-qPCR, western blot and immunofluorescence staining were conducted to test the phenomenon discovered by single-cell RNA sequencing in vitro. RESULTS: We found that fluoxetine treatment significantly alleviated colon inflammation. Notably, single-cell RNA sequencing analysis revealed that fluoxetine affected the distribution of different cell clusters, cell-cell communication and KEGG pathway enrichment. Under the treatment of fluoxetine, enterocytes, Goblet cells and stem cells became the dominating cells. The pseudotime analysis showed that there was a trend for M1 macrophages to differentiate into M2 macrophages. Lastly, we tested this phenomenon in vitro, which exhibited anti-inflammatory effects on enterocytes. CONCLUSIONS: Fluoxetine exhibited anti-inflammatory effects on intestinal mucosa via remodeling of the intestinal cells and macrophages, which reveals that fluoxetine is a promising therapeutic drug for the treatment of IBD and psychiatric comorbidities.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Mice , Fluoxetine/metabolism , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Cytokines/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Inflammation/metabolism , Intestinal Mucosa/metabolism , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Mice, Inbred C57BL
6.
Poult Sci ; 103(5): 103655, 2024 May.
Article in English | MEDLINE | ID: mdl-38537402

ABSTRACT

To develop effective antibiotics alternatives is getting more and more important to poultry healthy production. The study investigated the effects of a microencapsulated essential oils and organic acids preparation (EOA) on growth performance, slaughter performance, nutrient digestibility and intestinal microenvironment of broilers. A total of 624 1-day-old male Arbor Acres broilers were randomly divided into 6 groups including the control group (T1) fed with basal diet, the antibiotic group (T2) supplemented with basal diet with 45 mg/kg bacitracin methylene disalicylate (BMD), and 4 inclusion levels of EOA-treated groups (T3, T4, T5, T6 groups) chickens given basal diet with 200, 400, 600, and 800 mg EOA/kg of diet, respectively. Results showed that compared with the control, the 200 mg/kg EOA group increased average daily gain (ADG) and average body weight (ABW) during the early stage (P < 0.05). EOA addition decreased crypt depth of the ileum (P < 0.05), but villus height to crypt depth ratio was increased by EOA addition at 200 and 400 mg/kg at d 21 (P < 0.05). Compared with the control, dietary addition EOA at 200, 400 and 600 mg/kg increased the lipase activity in the duodenum at d 21 (P < 0.05). Increased lactic acid bacteria population was found in cecal digesta of the 400 mg/kg EOA group at d 21 (P < 0.05), and higher concentration of butyric acid level was observed in cecal digesta at d 21 and d 42 in the 200 mg/kg EOA group compared with the control (P < 0.05). RT-PCR analysis found that dietary EOA addition decreased the gene expression of IL-1ß, COX-2 and TGF-ß4 in the ileum at d 21 (P < 0.05), while only the 200 mg/kg EOA increased the gene expression of IL-10, TGF-ß4, Claudin-1, ZO-1, CATH-1, CATH-3, AvBD-1, AvBD-9 and AvBD-12 in the ileum at d 42 (P < 0.05) compared with the control. In summary, adding 200 mg/kg and 400 mg/kg of the EOA to the diet could improve the growth performance and intestinal microenvironment through improving intestinal morphology, increasing digestive enzymes activity and cecal lactic acid bacteria abundance and butyric acid content, improving intestinal barrier function as well as maintaining intestinal immune homeostasis. The improving effect induced by EOA addition in the early growth stage was better than that in the later growth stage. Overall, the EOA product might be an effective antibiotic alternative for broiler industry.


Subject(s)
Animal Feed , Animal Nutritional Physiological Phenomena , Chickens , Diet , Digestion , Oils, Volatile , Animals , Chickens/growth & development , Chickens/physiology , Animal Feed/analysis , Male , Diet/veterinary , Oils, Volatile/administration & dosage , Oils, Volatile/pharmacology , Digestion/drug effects , Animal Nutritional Physiological Phenomena/drug effects , Dietary Supplements/analysis , Intestines/drug effects , Intestines/anatomy & histology , Random Allocation , Dose-Response Relationship, Drug , Drug Compounding/veterinary , Nutrients/metabolism
7.
Bioact Mater ; 36: 203-220, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38463553

ABSTRACT

Ulcerative colitis (UC) is characterized by chronic inflammatory processes of the intestinal tract of unknown origin. Current treatments lack understanding on how to effectively alleviate oxidative stress, relieve inflammation, as well as modulate gut microbiota for maintaining intestinal homeostasis synchronously. In this study, a novel drug delivery system based on a metal polyphenol network (MPN) was constructed via metal coordination between epigallocatechin gallate (EGCG) and Fe3+. Curcumin (Cur), an active polyphenolic compound, with distinguished anti-inflammatory activity was assembled and encapsulated into MPN to generate Cur-MPN. The obtained Cur-MPN could serve as a robust reactive oxygen species modulator by efficiently scavenging superoxide radical (O2•-) as well as hydroxyl radical (·OH). By hitchhiking yeast microcapsule (YM), Cur-MPN was then encapsulated into YM to obtain CM@YM. Our findings demonstrated that CM@YM was able to protect Cur-MPN to withstand the harsh gastrointestinal environment and enhance the targeting and retention abilities of the inflamed colon. When administered orally, CM@YM could alleviate DSS-induced colitis with protective and therapeutic effects by scavenging ROS, reducing pro-inflammatory cytokines, and regulating the polarization of macrophages to M1, thus restoring barrier function and maintaining intestinal homeostasis. Importantly, CM@YM also modulated the gut microbiome to a favorable state by improving bacterial diversity and transforming the compositional structure to an anti-inflammatory phenotype as well as increasing the content of short-chain fatty acids (SCFA) (such as acetic acid, propionic acid, and butyric acid). Collectively, with excellent biocompatibility, our findings indicate that synergistically regulating intestinal microenvironment will be a promising approach for UC.

8.
Nano Lett ; 24(7): 2289-2298, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38341876

ABSTRACT

Antibiotic therapeutics to combat intestinal pathogen infections often exacerbate microbiota dysbiosis and impair mucosal barrier functions. Probiotics are promising strategies, because they inhibit pathogen colonization and improve intestinal microbiota imbalance. Nevertheless, their limited targeting ability and susceptibility to oxidative stress have hindered their therapeutic potential. To tackle these challenges, Ces3 is synthesized by in situ growth of CeO2 nanozymes with positive charges on probiotic spores, facilitating electrostatic interactions with negatively charged pathogens and possessing a high reactive oxygen species (ROS) scavenging activity. Importantly, Ces3 can resist the harsh environment of the gastrointestinal tract. In mice with S. Typhimurium-infected acute gastroenteritis, Ces3 shows potent anti-S. Typhimurium activity, thereby alleviating the dissemination of S. Typhimurium into other organs. Additionally, owing to its O2 deprivation capacity, Ces3 promotes the proliferation of anaerobic probiotics, reshaping a healthy intestinal microbiota. This work demonstrates the promise of combining antibacterial, anti-inflammatory, and O2 content regulation properties for acute gastroenteritis therapy.


Subject(s)
Gastroenteritis , Probiotics , Animals , Mice , Intestines , Gastroenteritis/drug therapy , Gastroenteritis/microbiology , Anti-Bacterial Agents/therapeutic use , Probiotics/therapeutic use , Spores
9.
ACS Nano ; 18(3): 2131-2148, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38198697

ABSTRACT

Endotoxemia is a life-threatening multiple organ failure disease caused by bacterial endotoxin infection. Unfortunately, current single-target therapy strategies have failed to prevent the progression of endotoxemia. Here, we reported that alanine fullerene redox modulator (AFRM) remodeled the intestinal microenvironment for multiple targets endotoxemia mitigation by suppressing inflammatory macrophages, inhibiting macrophage pyroptosis, and repairing epithelial cell barrier integrity. Specifically, AFRM exhibited broad-spectrum and self-cascade redox regulation properties with superoxide dismutase (SOD)-like enzyme, peroxidase (POD)-like enzyme activity, and hydroxyl radical (•OH) scavenging ability. Guided by proteomics, we demonstrated that AFRM regulated macrophage redox homeostasis and down-regulated LPS/TLR4/NF-κB and MAPK/ERK signaling pathways to suppress inflammatory hyperactivation. Of note, AFRM could attenuate inflammation-induced macrophage pyroptosis via inhibiting the activation of gasdermin D (GSDMD). In addition, our results revealed that AFRM could restore extracellular matrix and cell-tight junction proteins and protect the epithelial cell barrier integrity by regulating extracellular redox homeostasis. Consequently, AFRM inhibited systemic inflammation and potentiated intestinal epithelial barrier damage repair during endotoxemia in mice. Together, our work suggested that fullerene based self-cascade redox modulator has the potential in the management of endotoxemia through synergistically remodeling the inflammation and epithelial barriers in the intestinal microenvironment.


Subject(s)
Endotoxemia , Fullerenes , Mice , Animals , Endotoxemia/chemically induced , Endotoxemia/metabolism , Intestines , NF-kappa B/metabolism , Inflammation , Oxidation-Reduction , Lipopolysaccharides/pharmacology
10.
Front Immunol ; 14: 1306375, 2023.
Article in English | MEDLINE | ID: mdl-38077358

ABSTRACT

Inflammatory bowel disease (IBD) is a complex group of chronic intestinal diseases, the cause of which has not yet been clarified, but it is widely believed that the disorder of the intestinal microenvironment and its related functional changes are key factors in the development of the disease. Houttuynia cordata thunb. is a traditional plant with abundant resources and long history of utilization in China, which has attracted widespread attention in recent years due to its potential in the treatment of IBD. However, its development and utilization are limited owing to the aristolochic acid alkaloids contained in it. Therefore, based on the relationship between the intestinal microenvironment and IBD, this article summarizes the potential mechanisms by which the main active ingredients of Houttuynia cordata thunb., such as volatile oils, polysaccharides, and flavonoids, and related traditional Chinese medicine preparations, such as Xiezhuo Jiedu Formula, alleviate IBD by regulating the intestinal microenvironment. At the same time, combined with current reports, the medicinal and edible safety of Houttuynia cordata thunb. is explained for providing ideas for further research and development of Houttuynia chordate thunb. in IBD disease, more treatment options for IBD patients, and more insights into the therapeutic potential of plants with homology of medicine and food in intestinal diseases, and even more diseases.


Subject(s)
Alkaloids , Drugs, Chinese Herbal , Houttuynia , Inflammatory Bowel Diseases , Humans , Plant Extracts , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Inflammatory Bowel Diseases/drug therapy
11.
Hum Exp Toxicol ; 42: 9603271231211894, 2023.
Article in English | MEDLINE | ID: mdl-38116628

ABSTRACT

OBJECTIVE: This study aimed to investigate the impact of esketamine on the intestinal flora and microenvironment in mice using mRNA transcriptome sequencing and 16S rRNA sequencing. METHODS: Ten female mice were randomly assigned to two groups. One group received daily intramuscular injections of sterile water, while the other group received esketamine. After 24 days, the mice were sacrificed, and their intestinal tissues and contents were collected for 16S rRNA sequencing and mRNA transcriptome sequencing. The intergroup differences in the mouse intestinal flora were analyzed. Differentially expressed genes were utilized to construct ceRNA networks and transcription factor regulatory networks to assess the effects of esketamine on the intestinal flora and intestinal tissue genes. RESULTS: Esketamine significantly altered the abundance of intestinal microbiota, including Adlercreutzia equolifaciens and Akkermansia muciniphila. Differential expression analysis revealed 301 significantly upregulated genes and 106 significantly downregulated genes. The ceRNA regulatory network consisted of 6 lncRNAs, 44 miRNAs, and 113 mRNAs, while the regulatory factor network included 13 transcription factors and 53 target genes. Gene Ontology enrichment analysis indicated that the differentially expressed genes were primarily associated with immunity, including B-cell activation and humoral immune response mediation. The biological processes in the ceRNA regulatory network primarily involved transport, such as organic anion transport and monocarboxylic acid transport. The functional annotation of target genes in the TF network was mainly related to epithelial cells, including epithelial cell proliferation and regulation. CONCLUSION: Esketamine induces changes in gut microbiota and the intestinal microenvironment, impacting the immune environment and transport modes.


Subject(s)
Gastrointestinal Microbiome , MicroRNAs , RNA, Long Noncoding , Female , Animals , Mice , RNA, Ribosomal, 16S/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Long Noncoding/genetics
12.
Am J Chin Med ; 51(8): 1927-1955, 2023.
Article in English | MEDLINE | ID: mdl-37930334

ABSTRACT

Depression is a highly heterogeneous mental illness. Drug treatment is currently the main therapeutic strategy used in the clinic, but its efficacy is limited by the modulation of a single target, slow onset, and side effects. The gut-brain axis is of increasing interest because intestinal microenvironment disorders increase susceptibility to depression. In turn, depression affects intestinal microenvironment homeostasis by altering intestinal tissue structure, flora abundance and metabolism, hormone secretion, neurotransmitter transmission, and immune balance. Depression falls into the category of "stagnation syndrome" according to Traditional Chinese Medicine (TCM), which further specifies that "the heart governs the spirit and is exterior-interior with the small intestine". However, the exact mechanisms of the means by which the disordered intestinal microenvironment affects depression are still unclear. Here, we present an overview of how the Chinese materia medica (CMM) protects against depression by repairing intestinal microenvironment homeostasis. We review the past five years of research progress in classical antidepressant TCM formulae and single CMMs on regulating the intestinal microenvironment for the treatment of depression. We then analyze and clarify the multitarget functions of CMM in repairing intestinal homeostasis and aim to provide a new theoretical basis for CMM clinical application in the treatment of depression.


Subject(s)
Drugs, Chinese Herbal , Materia Medica , Materia Medica/therapeutic use , Depression/drug therapy , Medicine, Chinese Traditional , Biological Transport , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use
13.
J Agric Food Chem ; 71(33): 12558-12573, 2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37581333

ABSTRACT

Recent research consensus has highlighted the role of intestinal luminal factors in the association between intestinal microenvironment homeostasis and food allergy. However, the association between intestinal immune homeostasis and food allergy-related proteomic features remains elusive. In this study, we aimed to investigate the changes in gluten allergy (GA)-defined phenotypes and endotypes and intestinal microenvironment factors in BALB/c mice and linked GA to colonic proteomic signatures. Combined with increased allergy and diarrhea scores, intense antibody responses and abnormalities in T-cell cytokine production were induced in mice. GA-associated disruption of intestinal microenvironment homeostasis was underlined by the increased colonic pH, decreased intestinal antioxidant capacity, impaired intestinal barrier function, and decreased production and imbalanced proportions of short-chain fatty acids. 16S rRNA amplicon sequencing showed that the gut microbiota dysbiosis in mice was characterized by significant enrichment of six bacterial taxonomic units, including Prevotellaceae, Escherichia Shigella, Alloprevotella, Escherichia coli, Bacteroides vulgatus, and Lachnospiraceae bacterium DW59, which was correlated with immune end points. Using a label-free proteomics quantitative approach, 24 differentially expressed proteins linking GA-induced gut dysbiosis were identified, with four of them enriched in the serine endopeptidase inhibitor activity pathway. The development of GA in mice was associated with changes in specific intestinal luminal factors and may be mediated by serine protease activity-associated metabolic routes.


Subject(s)
Food Hypersensitivity , Gastrointestinal Microbiome , Animals , Mice , Dysbiosis/microbiology , RNA, Ribosomal, 16S/genetics , Proteomics , Mice, Inbred BALB C , Glutens
14.
World J Gastrointest Oncol ; 15(5): 776-786, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37275451

ABSTRACT

The relevance of constipation to the development and progression of colorectal cancer (CRC) is currently a controversial issue. Studies have shown that changes in the composition of the gut microbiota, a condition known as ecological imbalance, are correlated with an increasing number of common human diseases, including CRC and constipation. CRC is the second leading cause of cancer-related deaths worldwide, and constipation has been receiving widespread attention as a risk factor for CRC. Early colonoscopy screening of constipated patients, with regular follow-ups and timely intervention, can help detect early intestinal lesions and reduce the risks of developing colorectal polyps and CRC. As an important regulator of the intestinal microenvironment, the gut microbiota plays a critical role in the onset and progression of CRC. An increasing amount of evidence supports the thought that gut microbial composition and function are key determinants of CRC development and progression, with alterations inducing changes in the expression of host genes, metabolic regulation, and local and systemic immunological responses. Furthermore, constipation greatly affects the composition of the gut microbiota, which in turn influences the susceptibility to intestinal diseases such as CRC. However, the crosstalk between the gut microbiota, constipation, and CRC is still unclear.

15.
CNS Neurosci Ther ; 29 Suppl 1: 185-199, 2023 06.
Article in English | MEDLINE | ID: mdl-37309254

ABSTRACT

BACKGROUND: Stroke is not only a major cause of disability but also the third leading cause of death, following heart disease and cancer. It has been established that stroke causes permanent disability in 80% of survivors. However, current treatment options for this patient population are limited. Inflammation and immune response are major features that are well-recognized to occur after a stroke. The gastrointestinal tract hosts complex microbial communities, the largest pool of immune cells, and forms a bidirectional regulation brain-gut axis with the brain. Recent experimental and clinical studies have highlighted the importance of the relationship between the intestinal microenvironment and stroke. Over the years, the influence of the intestine on stroke has emerged as an important and dynamic research direction in biology and medicine. AIMS: In this review, we describe the structure and function of the intestinal microenvironment and highlight its cross-talk relationship with stroke. In addition, we discuss potential strategies aiming to target the intestinal microenvironment during stroke treatment. CONCLUSION: The structure and function of the intestinal environment can influence neurological function and cerebral ischemic outcome. Improving the intestinal microenvironment by targeting the gut microbiota may be a new direction in treating stroke.


Subject(s)
Gastrointestinal Microbiome , Stroke , Humans , Stroke/drug therapy , Brain , Gastrointestinal Microbiome/physiology , Intestines , Inflammation
16.
Food Res Int ; 169: 112840, 2023 07.
Article in English | MEDLINE | ID: mdl-37254413

ABSTRACT

This study aimed to investigate the dynamic changes in intestinal microenvironment factors in the development of gluten-induced allergy (GA). Our results showed that GA provoked increasingly severe allergic phenotypes such as allergic and diarrheal symptoms with the gluten sensitization frequency, which was accompanied by dynamically rising levels of gluten-specific immunoglobulin (Ig) E, IgG2a and IgA, serum histamine, T cell-related inflammatory cytokines, and intestinal indexes. An increase in luminal pH was more significant in the large intestine versus the small intestine, which was due to a dynamic decline in colonic short-chain fatty acid levels. Both antioxidant capacity and intestinal permeability in the large intestine varied with the GA severity, as evidenced by a dynamic increase in the malondialdehyde content and a decrease in the superoxide dismutase activity and total antioxidant capacity. Moreover, we demonstrated that intestinal microenvironment dysbiosis occurred before a true allergy reaction began. Spearman correlation analysis suggested that the characteristic bacterial cluster, namely Alistipes, Desulfovibrio, Ileibacterium, Parabacteroides, and Ruminococcus torques group, are essential in the association between GA and intestinal microenvironment homeostasis.


Subject(s)
Antioxidants , Food Hypersensitivity , Humans , Intestines , Intestine, Small , Glutens/adverse effects , Immunoglobulin E
17.
Front Nutr ; 10: 1103463, 2023.
Article in English | MEDLINE | ID: mdl-37006920

ABSTRACT

Background: Constipation is common gastrointestinal disorder with high prevalence and recurrence, making people suffering. However, the treatment for constipation remains ineffectual. We aimed to the study the effects and mechanisms of postbiotic of hawthorn-probiotic on loperamide modeled old KM mice. Methods: Constipated mice were grouped and treated with 10% lactulose (Y), hawthorn group (S), probiotic group (F) and postbiotic of hawthorn-probiotic (FS). Fecal changes were observed. AQP3 and Enac-γ were measured by RT-qPCR and Western blotting, intestinal barrier by H&E and immunofluorescence staining, cell proliferation and apoptosis by CCK8 and flow cytometry. Gut microbiota was further determined by 16 s rRNA sequence of feces. Results: Postbiotic of hawthorn-probiotic improved intestinal movement and pathomorphology, elevated AQP3, Enac-γ and mucin-2 expression, accompanied by decreased serum TNF-α and cell apoptosis, but increased proliferation. Furthermore, it modified the gut microbiota of constipated mice, featured by upregulation of Lactobacillaceae. Conclusion: Postbiotic of hawthorn-probiotic relieved constipation by combined effects of regulating intestinal water and sodium metabolism, maintain intestinal barrier and gut microflora.Graphical Abstract.

18.
Chemosphere ; 322: 138209, 2023 May.
Article in English | MEDLINE | ID: mdl-36822518

ABSTRACT

Triazole antifungal pesticides work by inhibiting the activity of lanosterol-14-α-demethylase, a member of cytochrome P450 enzymes (CYPs), but this effect is non-specific. Bile acids (BAs) are important physical surfactants for lipids absorption in intestine, and synthesized by CYPs 7A1/27A1. Thus, we presume that triazole exposure might influence the therapeutic effect or safety of oral medication through disturbing the BAs pool, even though the exposure is under an acceptable daily intake (ADI) dose. Short- and long-term of ADI dose tebuconazole (TEB) exposure animal models were established through various routes, and statins with different hydrophilic and lipophilic properties were gavaged. It exhibited that the activity of CYP7A1/27A1 was indeed inhibited but the expression was up-regulated, the BAs pool was changed either the content and the composition, and the absorption behavior of statins with strong and medium degree of lipid-solubility were significantly changed. A series of experiments performed on models of intestinal mucus, Caco-2 cell monolayer and Caco-2/HT29 co-culture system revealed that the TEB-exposure induced BAs disturbance made impacts on drug absorption in many aspects, including drug solubility and the structure of intestinal barriers. This study suggests us to be more alert about the hazard of pesticides residues for elderly and chronically ill groups.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Pesticides , Humans , Animals , Bile Acids and Salts , Caco-2 Cells , No-Observed-Adverse-Effect Level
19.
Precis Clin Med ; 6(4): pbad031, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38163004

ABSTRACT

Inflammatory bowel diseases (IBD) are a group of chronic relapsing gastrointestinal inflammatory diseases with significant global incidence. Although the pathomechanism of IBD has been extensively investigated, several aspects of its pathogenesis remain unclear. Long non-coding RNAs (lncRNAs) are transcripts with more than 200 nucleotides in length that have potential protein-coding functions. LncRNAs play important roles in biological processes such as epigenetic modification, transcriptional regulation and post-transcriptional regulation. In this review, we summarize recent advances in research on IBD-related lncRNAs from the perspective of the overall intestinal microenvironment, as well as their potential roles as immune regulators, diagnostic biomarkers and therapeutic targets or agents for IBD.

20.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-962645

ABSTRACT

Ulcerative colitis (UC), a chronic inflammatory bowel disease affecting the colorectum with high morbidity and prevalence, has become a global burden. However, the causes and pathogenesis are still unclear. Available studies have verified that the imbalance of intestinal microenvironment is crucial in the occurrence and development of UC. Intestinal microenvironment is mainly composed of intestinal microbiota and intestinal mucosal cells, which are involved in the physiological and pathological activities of the body through the intestinal microbial barrier, chemical barrier, mechanical barrier, and immune barrier. Thus, probiotic agents, 5-aminosalicylic acid preparations, corticosteroids, immunosuppressants, biological preparations and other drugs are commonly used in western medicine for the treatment of UC, which, however, have limitations. Therefore, it is the key task for the prevention and treatment of UC to find new therapies. In recent years, it has been found that traditional Chinese medicine (TCM) has unique advantages in the prevention and treatment of UC. Chinese medicine compounds and Chinese medicine monomers can regulate the intestinal microenvironment through multiple pathways and targets, thereby intervening the occurrence and development of UC. It has gradually become a hot spot in the prevention and treatment of UC and attracted extensive attention. Therefore, this study first discussed the correlation between intestinal microenvironment imbalance and UC and then summarized the mechanisms of TCM against UC from the aspects of regulating intestinal flora, improving chemical barrier, protecting mechanical barrier, and inhibiting immune inflammatory response, in order to provide new ideas for the research on TCM in the treatment of UC.

SELECTION OF CITATIONS
SEARCH DETAIL
...