Gut MicrobesPub Date : 2024-01-01Epub Date: 2024-01-04DOI: 10.1080/19490976.2023.2298246
Giorgio Gargari, Giacomo Mantegazza, Cesare Cremon, Valentina Taverniti, Alice Valenza, Maria Raffaella Barbaro, Giovanni Marasco, Robin Duncan, Walter Fiore, Roberto Ferrari, Valerio De Vitis, Giovanni Barbara, Simone Guglielmetti
{"title":"<i>Collinsella aerofaciens</i> as a predictive marker of response to probiotic treatment in non-constipated irritable bowel syndrome.","authors":"Giorgio Gargari, Giacomo Mantegazza, Cesare Cremon, Valentina Taverniti, Alice Valenza, Maria Raffaella Barbaro, Giovanni Marasco, Robin Duncan, Walter Fiore, Roberto Ferrari, Valerio De Vitis, Giovanni Barbara, Simone Guglielmetti","doi":"10.1080/19490976.2023.2298246","DOIUrl":"10.1080/19490976.2023.2298246","url":null,"abstract":"<p><p>Probiotics are exploited for adjuvant treatment in IBS, but reliable guidance for selecting the appropriate probiotic to adopt for different forms of IBS is lacking. We aimed to identify markers for recognizing non-constipated (NC) IBS patients that may show significant clinical improvements upon treatment with the probiotic strain <i>Lacticaseibacillus paracasei</i> DG (LDG). To this purpose, we performed a post-hoc analysis of samples collected during a multicenter, double-blind, parallel-group, placebo-controlled trial in which NC-IBS patients were randomized to receive at least 24 billion CFU LDG or placebo capsules <i>b.i.d</i>. for 12 weeks. The primary clinical endpoint was the composite response based on improved abdominal pain and fecal type. The fecal microbiome and serum markers of intestinal (PV1 and zonulin), liver, and kidney functions were investigated. We found that responders (R) in the probiotic arm (25%) differed from non-responders (NR) based on the abundance of 18 bacterial taxa, including the families <i>Coriobacteriaceae</i>, <i>Dorea</i> spp. and <i>Collinsella aerofaciens</i>, which were overrepresented in R patients. These taxa also distinguished R (but not NR) patients from healthy controls. Probiotic intervention significantly reduced the abundance of these bacteria in R, but not in NR. Analogous results emerged for <i>C. aerofaciens</i> from the analysis of data from a previous trial on IBS with the same probiotic. Finally, <i>C. aerofaciens</i> was positively correlated with the plasmalemmal vesicle associated protein-1 (PV-1) and the markers of liver function. In conclusion, LDG is effective on NC-IBS patients with NC-IBS with a greater abundance of potential pathobionts. Among these, <i>C. aerofaciens</i> has emerged as a potential predictor of probiotic efficacy.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2298246"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10773624/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139097653","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Probiotic <i>Lactobacillus</i> spp. improves <i>Drosophila</i> memory by increasing lactate dehydrogenase levels in the brain mushroom body neurons.","authors":"Shuk-Man Ho, Wan-Hua Tsai, Chih-Ho Lai, Meng-Hsuan Chiang, Wang-Po Lee, Hui-Yu Wu, Pei-Yi Bai, Tony Wu, Chia-Lin Wu","doi":"10.1080/19490976.2024.2316533","DOIUrl":"10.1080/19490976.2024.2316533","url":null,"abstract":"<p><p>Probiotics are live microorganisms that offer potential benefits to their hosts and can occasionally influence behavioral responses. However, the detailed mechanisms by which probiotics affect the behavior of their hosts and the underlying biogenic effects remain unclear. Lactic acid bacteria, specifically <i>Lactobacillus</i> spp. are known probiotics. <i>Drosophila melanogaster</i>, commonly known as the fruit fly, is a well-established model organism for investigating the interaction between the host and gut microbiota in translational research. Herein, we showed that 5-day administration of <i>Lactobacillus acidophilus</i> (termed GMNL-185) or <i>Lacticaseibacillus rhamnosus</i> (termed GMNL-680) enhances olfactory-associative memory in <i>Drosophila</i>. Moreover, a combined diet of GMNL-185 and GMNL-680 demonstrated synergistic effects on memory functions. Live brain imaging revealed a significant increase in calcium responses to the training odor in the mushroom body β and γ lobes of flies that underwent mixed feeding with GMNL-185 and GMNL-680. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and whole-mount brain immunohistochemistry revealed significant upregulation of lactate dehydrogenase (LDH) expression in the fly brain following the mixed feeding. Notably, the genetic knockdown of <i>Ldh</i> in neurons, specifically in mushroom body, ameliorated the beneficial effects of mixed feeding with GMNL-185 and GMNL-680 on memory improvement. Altogether, our results demonstrate that supplementation with <i>L. acidophilus</i> and <i>L. rhamnosus</i> enhances memory functions in flies by increasing brain LDH levels.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2316533"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10877976/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139899703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gut MicrobesPub Date : 2024-01-01Epub Date: 2024-02-23DOI: 10.1080/19490976.2024.2319889
Won Jun Kim, Bum Ju Kil, Chaewon Lee, Tae Young Kim, Goeun Han, Yukyung Choi, Kyunghwan Kim, Chang Hun Shin, Seung-Young Park, Heebal Kim, Myunghoo Kim, Chul Sung Huh
{"title":"<i>B. longum</i> CKD1 enhances the efficacy of anti-diabetic medicines through upregulation of IL- 22 response in type 2 diabetic mice.","authors":"Won Jun Kim, Bum Ju Kil, Chaewon Lee, Tae Young Kim, Goeun Han, Yukyung Choi, Kyunghwan Kim, Chang Hun Shin, Seung-Young Park, Heebal Kim, Myunghoo Kim, Chul Sung Huh","doi":"10.1080/19490976.2024.2319889","DOIUrl":"10.1080/19490976.2024.2319889","url":null,"abstract":"<p><p>The gut microbiota plays a pivotal role in metabolic disorders, notably type 2 diabetes mellitus (T2DM). In this study, we investigated the synergistic potential of combining the effects of <i>Bifidobacterium longum</i> NBM7-1 (CKD1) with anti-diabetic medicines, Lobeglitazone<sup>Ⓡ</sup> (LO), Sitagliptin<sup>Ⓡ</sup> (SI), and Metformin<sup>Ⓡ</sup> (Met), to alleviate hyperglycemia in a diabetic mouse model. CKD1 effectively mitigated insulin resistance, hepatic steatosis, and enhanced pancreatic β-cell function, as well as fortifying gut-tight junction integrity. In the same way, SI-CKD1 and Met- CKD1 synergistically improved insulin sensitivity and prevented hepatic steatosis, as evidenced by the modulation of key genes associated with insulin signaling, β-oxidation, gluconeogenesis, adipogenesis, and inflammation by qRT-PCR. The comprehensive impact on modulating gut microbiota composition was observed, particularly when combined with Metformin<sup>Ⓡ</sup>. This combination induced an increase in the abundance of <i>Rikenellaceae</i> and <i>Alistipes</i> related negatively to the T2DM incidence while reducing the causative species of <i>Cryptosporangium, Staphylococcaceae</i>, and <i>Muribaculaceae</i>. These alterations intervene in gut microbiota metabolites to modulate the level of butyrate, indole-3-acetic acid, propionate, and inflammatory cytokines and to activate the IL-22 pathway. However, it is meaningful that the combination of <i>B. longum</i> NBM7-1(CKD1) reduced the medicines' dose to the level of the maximal inhibitory concentrations (IC50). This study advances our understanding of the intricate relationship between gut microbiota and metabolic disorders. We expect this study to contribute to developing a prospective therapeutic strategy modulating the gut microbiota.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2319889"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10896159/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139930916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gut MicrobesPub Date : 2024-01-01Epub Date: 2024-02-23DOI: 10.1080/19490976.2024.2314201
Sirena C Tran, Kaeli N Bryant, Timothy L Cover
{"title":"The <i>Helicobacter pylori cag</i> pathogenicity island as a determinant of gastric cancer risk.","authors":"Sirena C Tran, Kaeli N Bryant, Timothy L Cover","doi":"10.1080/19490976.2024.2314201","DOIUrl":"10.1080/19490976.2024.2314201","url":null,"abstract":"<p><p><i>Helicobacter pylori</i> strains can be broadly classified into two groups based on whether they contain or lack a chromosomal region known as the <i>cag</i> pathogenicity island (<i>cag</i> PAI). Colonization of the human stomach with <i>cag</i> PAI-positive strains is associated with an increased risk of gastric cancer and peptic ulcer disease, compared to colonization with <i>cag</i> PAI-negative strains. The <i>cag</i> PAI encodes a secreted effector protein (CagA) and components of a type IV secretion system (Cag T4SS) that delivers CagA and non-protein substrates into host cells. Animal model experiments indicate that CagA and the Cag T4SS stimulate a gastric mucosal inflammatory response and contribute to the development of gastric cancer. In this review, we discuss recent studies defining structural and functional features of CagA and the Cag T4SS and mechanisms by which <i>H. pylori</i> strains containing the <i>cag</i> PAI promote the development of gastric cancer and peptic ulcer disease.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2314201"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10896142/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139930917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gut MicrobesPub Date : 2024-01-01Epub Date: 2024-03-04DOI: 10.1080/19490976.2024.2323234
Ye Peng, Hein M Tun, Siew C Ng, Hogan Kok-Fung Wai, Xi Zhang, Jaclyn Parks, Catherine J Field, Piush Mandhane, Theo J Moraes, Elinor Simons, Stuart E Turvey, Padmaja Subbarao, Jeffrey R Brook, Tim K Takaro, James A Scott, Francis Kl Chan, Anita L Kozyrskyj
{"title":"Maternal smoking during pregnancy increases the risk of gut microbiome-associated childhood overweight and obesity.","authors":"Ye Peng, Hein M Tun, Siew C Ng, Hogan Kok-Fung Wai, Xi Zhang, Jaclyn Parks, Catherine J Field, Piush Mandhane, Theo J Moraes, Elinor Simons, Stuart E Turvey, Padmaja Subbarao, Jeffrey R Brook, Tim K Takaro, James A Scott, Francis Kl Chan, Anita L Kozyrskyj","doi":"10.1080/19490976.2024.2323234","DOIUrl":"10.1080/19490976.2024.2323234","url":null,"abstract":"<p><p>Childhood obesity is linked to maternal smoking during pregnancy. Gut microbiota may partially mediate this association and could be potential targets for intervention; however, its role is understudied. We included 1,592 infants from the Canadian Healthy Infants Longitudinal Development Cohort. Data on environmental exposure and lifestyle factors were collected prenatally and throughout the first three years. Weight outcomes were measured at one and three years of age. Stool samples collected at 3 and 12 months were analyzed by sequencing the V4 region of 16S rRNA to profile microbial compositions and magnetic resonance spectroscopy to quantify the metabolites. We showed that quitting smoking during pregnancy did not lower the risk of offspring being overweight. However, exclusive breastfeeding until the third month of age may alleviate these risks. We also reported that maternal smoking during pregnancy significantly increased Firmicutes abundance and diversity. We further revealed that Firmicutes diversity mediates the elevated risk of childhood overweight and obesity linked to maternal prenatal smoking. This effect possibly occurs through excessive microbial butyrate production. These findings add to the evidence that women should quit smoking before their pregnancies to prevent microbiome-mediated childhood overweight and obesity risk, and indicate the potential obesogenic role of excessive butyrate production in early life.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2323234"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10913716/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140021581","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gut MicrobesPub Date : 2024-01-01Epub Date: 2024-03-04DOI: 10.1080/19490976.2024.2323220
Wenjie Tang, Yusen Wei, Zhixiang Ni, Kangwei Hou, Xin M Luo, Haifeng Wang
{"title":"IgA-mediated control of host-microbial interaction during weaning reaction influences gut inflammation.","authors":"Wenjie Tang, Yusen Wei, Zhixiang Ni, Kangwei Hou, Xin M Luo, Haifeng Wang","doi":"10.1080/19490976.2024.2323220","DOIUrl":"10.1080/19490976.2024.2323220","url":null,"abstract":"<p><p>The mechanisms of how host-microbe mutualistic relationships are established at weaning contingently upon B-cell surveillance remain inadequately elucidated. We found that <i>CD138</i><sup>+</sup> plasmacyte (PC)-mediated promotion of IgA response regulates the symbiosis between <i>Bacteroides uniformis</i> (<i>B. uniformis</i>) and the host during the weaning period. The IgA-skewed response of <i>CD138</i><sup>+</sup> PCs is essential for <i>B. uniformis</i> to occupy a defined gut luminal niche, thereby fostering stable colonization. Furthermore, <i>B. uniformis</i> within the natural gut niche was perturbed in the absence of IgA, resulting in exacerbated gut inflammation in IgA-deficient mice and weaned piglets. Thus, we propose that the priming and maintenance of intestinal IgA response from <i>CD138</i><sup>+</sup> PCs are required for host-microbial symbiosis, whereas the perturbation of which would enhance inflammation in weaning process.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2323220"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10936605/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140027963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"<i>B. thetaiotaomicron</i>-derived acetic acid modulate immune microenvironment and tumor growth in hepatocellular carcinoma.","authors":"Hongbin Ma, Liang Yang, Yingchao Liang, Fenghua Liu, Jinxiang Hu, Rui Zhang, Yong Li, Lei Yuan, Feiling Feng","doi":"10.1080/19490976.2023.2297846","DOIUrl":"10.1080/19490976.2023.2297846","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, and emerging evidence suggests that the gut microbiota may play a role in its development and progression. In this study, the association between <i>B. thetaiotaomicron</i>, a gut microbiota species, and HCC recurrence, as well as patient clinical outcomes, was investigated. It was observed that <i>B. thetaiotaomicron</i>-derived acetic acid has the potential to modulate the polarization of <b>pro-pro-inflammatory macrophagess</b>, which promotes the function of cytotoxic CD8+ T cells. The increased biosynthesis of fatty acids was implicated in the modulation of <b>pro-inflammatory macrophages</b> polarization by <i>B. thetaiotaomicron</i>-derived acetic acid. Furthermore, <i>B. thetaiotaomicron</i>-derived acetic acid was found to facilitate the transcription of ACC1, a key enzyme involved in fatty acid biosynthesis, through histone acetylation modification in the ACC1 promoter region. Curcumin, an acetylation modification inhibitor, significantly blocked the inhibitory effects of <i>B. thetaiotaomicron</i> and acetic acid on HCC tumor growth. These findings highlight the potential role of gut microbiota-derived acetic acid in HCC recurrence and patient clinical outcomes, and suggest a complex interplay between gut microbiota, immune modulation, fatty acid metabolism, and epigenetic regulation in the context of HCC development. Further research in this area may provide insights into novel strategies for HCC prevention and treatment by targeting the gut microbiota and its metabolites.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2297846"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10813637/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139546120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gut MicrobesPub Date : 2024-01-01Epub Date: 2024-03-14DOI: 10.1080/19490976.2024.2328868
Lingxi Li, Xiaowen Huang, Haoyan Chen
{"title":"Unveiling the hidden players: exploring the role of gut mycobiome in cancer development and treatment dynamics.","authors":"Lingxi Li, Xiaowen Huang, Haoyan Chen","doi":"10.1080/19490976.2024.2328868","DOIUrl":"10.1080/19490976.2024.2328868","url":null,"abstract":"<p><p>The role of gut fungal species in tumor-related processes remains largely unexplored, with most studies still focusing on fungal infections. This review examines the accumulating evidence suggesting the involvement of commensal and pathogenic fungi in cancer biological process, including oncogenesis, progression, and treatment response. Mechanisms explored include fungal influence on host immunity, secretion of bioactive toxins/metabolites, interaction with bacterial commensals, and migration to other tissues in certain types of cancers. Attempts to utilize fungal molecular signatures for cancer diagnosis and fungal-derived products for treatment are discussed. A few studies highlight fungi's impact on the responsiveness and sensitivity to chemotherapy, radiotherapy, immunotherapy, and fecal microbiota transplant. Given the limited understanding and techniques in fungal research, the studies on gut fungi are still facing great challenges, despite having great potentials.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2328868"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10950292/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140131329","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gut MicrobesPub Date : 2024-01-01Epub Date: 2024-05-05DOI: 10.1080/19490976.2024.2347722
Li-Wei Xie, Shang Cai, Hai-Yan Lu, Feng-Ling Tang, Rui-Qiu Zhu, Ye Tian, Ming Li
{"title":"Microbiota-derived I3A protects the intestine against radiation injury by activating AhR/IL-10/Wnt signaling and enhancing the abundance of probiotics.","authors":"Li-Wei Xie, Shang Cai, Hai-Yan Lu, Feng-Ling Tang, Rui-Qiu Zhu, Ye Tian, Ming Li","doi":"10.1080/19490976.2024.2347722","DOIUrl":"10.1080/19490976.2024.2347722","url":null,"abstract":"<p><p>The intestine is prone to radiation damage in patients undergoing radiotherapy for pelvic tumors. However, there are currently no effective drugs available for the prevention or treatment of radiation-induced enteropathy (RIE). In this study, we aimed at investigating the impact of indole-3-carboxaldehyde (I3A) derived from the intestinal microbiota on RIE. Intestinal organoids were isolated and cultivated for screening radioprotective tryptophan metabolites. A RIE model was established using 13 Gy whole-abdominal irradiation in male C57BL/6J mice. After oral administration of I3A, its radioprotective ability was assessed through the observation of survival rates, clinical scores, and pathological analysis. Intestinal stem cell survival and changes in the intestinal barrier were observed through immunofluorescence and immunohistochemistry. Subsequently, the radioprotective mechanisms of I3A was investigated through 16S rRNA and transcriptome sequencing, respectively. Finally, human colon cancer cells and organoids were cultured to assess the influence of I3A on tumor radiotherapy. I3A exhibited the most potent radioprotective effect on intestinal organoids. Oral administration of I3A treatment significantly increased the survival rate in irradiated mice, improved clinical and histological scores, mitigated mucosal damage, enhanced the proliferation and differentiation of Lgr5<sup>+</sup> intestinal stem cells, and maintained intestinal barrier integrity. Furthermore, I3A enhanced the abundance of probiotics, and activated the AhR/IL-10/Wnt signaling pathway to promote intestinal epithelial proliferation. As a crucial tryptophan metabolite, I3A promotes intestinal epithelial cell proliferation through the AhR/IL-10/Wnt signaling pathway and upregulates the abundance of probiotics to treat RIE. Microbiota-derived I3A demonstrates potential clinical application value for the treatment of RIE.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"16 1","pages":"2347722"},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11086037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140854883","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}