1
|
Su F, Su M, Wei W, Wu J, Chen L, Sun X, Liu M, Sun S, Mao R, Bourgonje AR, Hu S. Integrating multi-omics data to reveal the host-microbiota interactome in inflammatory bowel disease. Gut Microbes 2025; 17:2476570. [PMID: 40063366 PMCID: PMC11901428 DOI: 10.1080/19490976.2025.2476570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Numerous studies have accelerated the knowledge expansion on the role of gut microbiota in inflammatory bowel disease (IBD). However, the precise mechanisms behind host-microbe cross-talk remain largely undefined, due to the complexity of the human intestinal ecosystem and multiple external factors. In this review, we introduce the interactome concept to systematically summarize how intestinal dysbiosis is involved in IBD pathogenesis in terms of microbial composition, functionality, genomic structure, transcriptional activity, and downstream proteins and metabolites. Meanwhile, this review also aims to present an updated overview of the relevant mechanisms, high-throughput multi-omics methodologies, different types of multi-omics cohort resources, and computational methods used to understand host-microbiota interactions in the context of IBD. Finally, we discuss the challenges pertaining to the integration of multi-omics data in order to reveal host-microbiota cross-talk and offer insights into relevant future research directions.
Collapse
Affiliation(s)
- Fengyuan Su
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Meng Su
- The First Clinical Medical School, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Wenting Wei
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jiayun Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Leyan Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiqiao Sun
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Moyan Liu
- Amsterdam UMC location Academic Medical Center, Department of Experimental Vascular Medicine, Amsterdam, The Netherlands
| | - Shiqiang Sun
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
2
|
Jiang ST, Wang MQ, Gao L, Zhang QC, Tang C, Dong YF. Adjusting the composition of gut microbiota prevents the development of post-stroke depression by regulating the gut-brain axis in mice. J Affect Disord 2025; 381:242-259. [PMID: 40189067 DOI: 10.1016/j.jad.2025.03.195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/12/2025]
Abstract
Disturbances in gut microbiota contribute to an imbalanced gut-brain axis, which is critical for post-stroke depression (PSD), while the underlying mechanisms remain unclear. The objective of this study was to examine the effects of modifying gut microbiota through antibiotic treatment (ABX) and fecal microbiota transplantation (FMT) on the progression of PSD. The PSD model was established by occluding the middle cerebral artery (MCAO), followed by a four-week isolated housing and restraint stress initiated three days after MCAO. For ABX, the PSD mice received antibiotic water for four weeks. While another group of PSD mice underwent FMT or fluoxetine (FLX) for four weeks. At day 35 post-MCAO, behavioral tests were conducted. Results indicated ABX and FMT significantly altered the composition of intestinal flora caused by PSD, all the treatments markedly attenuated anxiety- and depressive-like behaviors and inflammation in the gut and brain. ABX obviously alleviated PSD-induced disorder of intestinal barrier, decreased mRNA levels of TNF-α, IL-1β and IL-6, and decreased CD4+ cells in the colon. While FMT significantly decreased CD8+ cells and increased the goblet cells in colon. Furthermore, both ABX and FMT could reduce activated microglia and pro-inflammatory cytokines in the brain, alleviate decreased Nissl's bodies in the hippocampus, and reverse the decreases in 5-HT, Glu and DA in the striatum caused by PSD. Unlike ABX, FMT was similar to FLX. These findings suggest homeostasis of gut microbiota is indispensable for the development of PSD; adjusting the gut microbiota significantly improves PSD with enhanced functions of gut-brain axis.
Collapse
Affiliation(s)
- Su-Ting Jiang
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Meng-Qing Wang
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Gao
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qi-Chun Zhang
- Department of Clinical Pharmacy and Toxicity, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chao Tang
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yin-Feng Dong
- Department of Pathology and Pathophysiology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Chen C, Qi M, Xu Z, Wen J, Tang W, Diao H, Li Z, Chu Y, Feng F, Tang Z. Sesamin improved growth and overall health in young animals by enhancing gut-liver axis function. Food Funct 2025. [PMID: 40351157 DOI: 10.1039/d4fo05933f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
The immature gastrointestinal development of infants and young animals made them more vulnerable to stress-related damage, which affected the gut-liver axis and consequently impaired their health and growth. This study used weaned piglets as a model to investigate how dietary sesamin regulated the gut-liver axis and impacted young animal health. We assessed gut-liver tissue morphology, measured key indicators of intestinal barrier damage, mucosal repair, antioxidant and immune pathways in the gut-liver system and serum, and analyzed microbial composition. We further explored the interactions between sesamin and the gut-liver axis through PLS-PM and molecular docking analysis. Results showed that sesamin enhanced intestinal barrier function, reduced liver damage, decreased oxidative stress, promoted anti-inflammatory immune responses, and enriched beneficial microbes, thereby promoting overall growth. Sesamin can enhance the health of young animals by regulating the function of the gut-liver axis.
Collapse
Affiliation(s)
- Chen Chen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Min Qi
- Yunnan Animal Husbandry Station, Kunming 650225, China
| | - Zhiran Xu
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Jincheng Wen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Hui Diao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Zhangcheng Li
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Yunyun Chu
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Fu Feng
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Zhiru Tang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
4
|
Gao Z, Gao Y, Wang X, Wang T, Zhou H, Liu C, Mai K, He G. Administration of EPA improves lipopolysaccharide-induced intestinal dysfunction in turbot (Scophthalmus maximus L.) through modulation of TORC1 signaling. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110391. [PMID: 40345275 DOI: 10.1016/j.fsi.2025.110391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/20/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
Intestinal health is crucial for digestive and absorptive functions, which are associated with fish growth performance. Searching for nutraceuticals and bioactive ingredients to improve intestinal health has become urgent for the aquaculture industry. In the present study, the effects of eicosapentaenoic acid (EPA) on intestinal function were investigated in turbot with induced intestinal damage caused by lipopolysaccharide (LPS). Juvenile turbot (initial weight 100 ± 5 g) were subjected to a 10-day enforced feeding regimen. Each fish was fed twice daily with either a 100 mg amino acid mixture (CON), an additional 1 mg of LPS (LPS), or an additional combination of 1 mg LPS and 1.7 mg EPA (LE). The results indicated that EPA supplementation restored intestinal villi integrity and the mucosal barrier. The number of goblet cells and the gene expression of MUC-2 were increased by EPA. Additionally, EPA suppressed the expression of inflammatory factors (MyD88 and NF-κB p65) and pro-inflammatory cytokines (TNFα, IL-1β, and IFN-γ). Meanwhile, post-feeding plasma free amino acid levels as well as intestinal protein synthesis were improved by EPA supplementation. The target of rapamycin (TOR) signaling pathway was significantly activated by EPA. Furthermore, EPA supplementation positively influenced intestinal microbiota, reducing the abundance of prevalent pathogens while enhancing the abundance of probiotics. Collectively, the administration of EPA effectively mitigates LPS-induced damage to the intestinal barrier, inflammatory response, and dysbiosis of microbiota through modulation of the TOR signaling pathway.
Collapse
Affiliation(s)
- Zongyu Gao
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Ya Gao
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Xuan Wang
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Tingting Wang
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Huihui Zhou
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Chengdong Liu
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Kangsen Mai
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China
| | - Gen He
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, 266003, China; Key Laboratory of Aquaculture Nutrition (Ministry of Agriculture), Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
5
|
Wei L, Bai J, Zhang Y, Suo H, Wang C. Comparison of in vitro fermentation characteristics of carob gum and guar gum. Int J Biol Macromol 2025; 311:143886. [PMID: 40319966 DOI: 10.1016/j.ijbiomac.2025.143886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 04/27/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Carob gum (CG) and guar gum (GG) are widely used as additives in food processing; however, their fermentation properties in the human gut and potential effects on the human body are unclear. This study used an in vitro fermentation model to evaluate the interaction of CG and GG with intestinal flora and compare their fermentation characteristics. The results showed that CG and GG could be degraded and utilized by the intestinal flora. GG can increase the production of acetic acid, propionic acid, and butyric acid and the relative abundance of beneficial microorganisms such as Prevotella and Faecalibacterium. CG promotes propionic acid production and the relative abundance of beneficial microorganisms such as Bifidobacterium and Lactobacillus. Metabolomic studies have shown that fermented CG and GG mainly affect human metabolic pathways. GG promotes amino acid and lipid metabolism, and CG promotes amino acid metabolism and biosynthesis of secondary metabolites. This research shows that despite significant differences in how CG and GG interact with the gut microbiota, both may affect the human body.
Collapse
Affiliation(s)
- Li Wei
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing 400700, China
| | - Yuyan Zhang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China.
| |
Collapse
|
6
|
Kamdougha H, Taminiau B, Fall PA, Ben Amor S, Trigui A, Daube G, Mnif B. Alterations of ocular surface microbiome in glaucoma and its association with dry eye. J Med Microbiol 2025; 74:002013. [PMID: 40359128 PMCID: PMC12075858 DOI: 10.1099/jmm.0.002013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction. Alterations in ocular surface microbiota (OSM) have been noted in both dry eye disease (DED) and glaucoma. However, the combined effects of these conditions on OSM have not been explored.Hypothesis. We hypothesized that patients with both glaucoma and dry eye would exhibit distinct changes in OSM composition and diversity compared to those with only glaucoma, only dry eye or healthy individuals.Aim. We employed amplicon sequencing to investigate OSM profiles in patients with glaucoma and/or dry eye disease.Methods. Swab samples from the conjunctiva of both eyes were collected from 28 glaucomatous patients [13 without dry eye syndrome (G-only) and 15 with dry eye syndrome (G-DED)], 13 DED patients without glaucoma (DED-only) and 31 age-matched healthy controls (HCs). After V3-V4 16S rRNA sequencing, MOTHUR tools and R language were used to elucidate and compare OSM composition and diversity between groups.Results. Our data revealed very diverse bacterial communities with 28 phyla and 785 genera. All the groups shared the three most abundant phyla, Actinobacteria (67.47%), Firmicutes (17.14%) and Proteobacteria (13.73%). Corynebacterium (54.75%), Staphylococcus (10.71%), Cutibacterium (8.77%) and Streptococcus (3.20%) were the most abundant genera. Only the G-DED group showed higher alpha diversity than the HC group (P<0.05). However, significant differences in beta diversity were observed between all three patient groups and the HC group. The Differential Expression for Sequencing 2 (DESeq2) analysis unveiled an increased presence of opportunistic bacteria across all pathological groups, with the G-DED group demonstrating the most pronounced alterations.Conclusions. Our findings confirm the predominance of Gram-positive bacteria in normal OSM and the rise of opportunistic Gram-negative bacteria in glaucoma and dry eye disease. This is the first study to characterize OSM in glaucoma patients with DED.
Collapse
Affiliation(s)
- Houyem Kamdougha
- Laboratory of Microbiology, Habib Bourguiba University Hospital, Sfax University, Sfax, Tunisia
- Laboratory of Microbiology, Department of Food Sciences, Fundamental and Applied Research for Animal & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Bernard Taminiau
- Laboratory of Microbiology, Department of Food Sciences, Fundamental and Applied Research for Animal & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Papa Abdoulaye Fall
- Laboratory of Microbiology, Department of Food Sciences, Fundamental and Applied Research for Animal & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Saloua Ben Amor
- Department of Ophthalmology, Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Amira Trigui
- Department of Ophthalmology, Habib Bourguiba University Hospital, University of Sfax, Sfax, Tunisia
| | - Georges Daube
- Laboratory of Microbiology, Department of Food Sciences, Fundamental and Applied Research for Animal & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Basma Mnif
- Laboratory of Microbiology, Habib Bourguiba University Hospital, Sfax University, Sfax, Tunisia
- Research Laboratory Microorganisms and Human Disease "MPH LR03SP03", Sfax University, Sfax, Tunisia
| |
Collapse
|
7
|
de Assis BBT, Cabral L, Silva FA, de Araújo Bezerra J, Noronha MF, Vidal H, Dos Santos Lima M, Pimentel TC, Magnani M. Fermentation of Amazonian fruit pulp (bacaba) with distinct probiotics: Impacts on chemical composition, bioaccessibility, and effects on human intestinal microbiota. Food Res Int 2025; 209:116326. [PMID: 40253160 DOI: 10.1016/j.foodres.2025.116326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/21/2025]
Abstract
Bacaba (Oenocarpus bacaba Mart.) is an underexplored Amazonian fruit rich in polyphenols that can serve as a substrate for probiotic survival and may positively impact on the composition and metabolism of the intestinal microbiota. This study aimed to evaluate the bacaba pulp fermented with probiotics Lactobacillus acidophilus 05 (LA-05) and Lacticaseibacillus casei 01 (LC1) regarding the chemical composition and probiotics survivability during fermentation (48 h), and the effect on the modulation of the intestinal microbiota of healthy adults through 16S rRNA sequencing. The probiotic-fermented bacaba pulps showed decreased pH and total soluble solids values and sugar content (maltose, glucose, fructose, and rhamnose), and increased titratable acidity values, organic acid content (lactic and tartaric acids), and phenolic compounds concentration compared to the control pulp. Furthermore, it presented adequate probiotic viability after fermentation and simulated gastrointestinal conditions. The bacaba pulp fermented with LC1 showed a higher concentration of butyric acid and phenolic compounds concentration (trans-resveratrol, cis resveratrol, catechin, procyanidin B2, and pelargonidin 3-glucoside) and bioaccessibility compared to the control pulp. The bacaba pulp fermented with LA-5 showed a higher concentration of pelargonidin 3-glucoside and procyanidin B2 compared to the control pulp and the highest bioaccessibility of some phenolic compounds (trans-resveratrol, cis-resveratrol, catechin, epicatechin, procyanidin B1, procyanidin B2, myricetin, and isorhamnetin). In vitro fecal fermentation reduced the pH and increased the abundance of Desulfovibrionales, Lactobacillales, and Peptostreptococcales-Tissierellales for all treatments. Bacaba pulp with LC1 resulted in the lowest pH values, and increased production of organic acids and concentration of phenolic compounds. Furthermore, both probiotic pulps increased the abundance of Lactobacillales and Acidaminococcales and decreased the abundance of Clostridiales. These findings provide new information about the potential of using bacaba in a functional pulp that may benefit human health through colonic microbiota changes.
Collapse
Affiliation(s)
- Bianca Beatriz Torres de Assis
- Laboratory of Microbial Process in Foods, Department of Food Engineering, Federal University of Paraíba, João Pessoa, Brazil
| | - Lucélia Cabral
- Institute of Biological Sciences, University of Brasília-UnB, Brasília, DF, Brazil
| | - Francyeli Araújo Silva
- Laboratory of Microbial Process in Foods, Department of Food Engineering, Federal University of Paraíba, João Pessoa, Brazil
| | | | - Melline Fontes Noronha
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Hubert Vidal
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon-1, Pierre Bénite, France
| | - Marcos Dos Santos Lima
- Department of Food Technology, Federal Institute of Sertão de Pernambuco, Petrolina, Brazil
| | | | - Marciane Magnani
- Laboratory of Microbial Process in Foods, Department of Food Engineering, Federal University of Paraíba, João Pessoa, Brazil.
| |
Collapse
|
8
|
Inan S, Wilson RP, Tükel Ç. IUPHAR review: From gut to brain: The role of gut dysbiosis, bacterial amyloids, and metabolic disease in Alzheimer's disease. Pharmacol Res 2025; 215:107693. [PMID: 40086611 DOI: 10.1016/j.phrs.2025.107693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Gut microbial dysbiosis, or altered gut microbial communities, in Alzheimer's Disease suggests a pathogenic role for gut inflammation and microbial products in shaping a neuroinflammatory environment. Similarly, metabolic diseases, such as obesity and diabetes, are also associated with an increased risk of Alzheimer's Disease. As the metabolic landscape shifts during gut inflammation, and gut inflammation in turn impacts metabolic processes, we explore how these interconnected pathways may contribute to the progression of Alzheimer's Disease. Additionally, we discuss the role of bacterial amyloids produced by gut microbes, which may exacerbate amyloid aggregation in the brain and contribute to neurodegenerative processes. Furthermore, we highlight potential therapeutic strategies aimed at reducing gut inflammation, improving metabolic health, and decreasing amyloid content as a means to mitigate Alzheimer's Disease progression. These approaches, targeting the gut-brain-metabolic axis, could offer promising avenues for delaying or preventing cognitive decline in affected individuals.
Collapse
Affiliation(s)
- Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| | - R Paul Wilson
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Manoria P, Noor MT. Correlation of serum vitamin D levels with serum interleukin-23 levels in patients of ulcerative colitis. Hum Immunol 2025; 86:111305. [PMID: 40199019 DOI: 10.1016/j.humimm.2025.111305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
Ulcerative Colitis (UC) is a chronic inflammatory condition resulting from an abnormal immune response to gut microbiota, leading to cytokine dysregulation, including elevated interleukin-23 (IL-23) levels. Emerging evidence suggests that vitamin D (VD) plays a crucial role in immune modulation. However, its correlation with IL-23 in UC is not well addressed. This study aims to elucidate the relationship between serum VD and IL-23 levels in UC patients. We included forty-four UC patients and forty-four healthy controls. VD insufficiency was more common in UC patients (n = 14) compared to controls (n = 5). Significant increases in IL-23 levels were observed from remission (46.6 ± 4.3 pg/mL) to severe stages (218.5 ± 62.41 pg/mL), while VD levels did not show a similar trend. IL-23 levels also rose significantly with disease extent, from proctitis to pancolitis. A significant negative correlation was found between VD and IL-23 levels (r = -0.3175; P = 0.035). IL-23 and pulse rate were significant predictors of UC in our cohort. Our findings highlight VD insufficiency to be prevalent in UC patients, with VD levels negatively correlating with IL-23 levels, which increase with disease severity and extent. Further, understanding the interplay between VD and IL-23 will help design therapeutic interventions to modulate immune response and disease progression.
Collapse
Affiliation(s)
- Piyush Manoria
- Department of Gastroenterology and Hepatology, Manoria Hospital, Bhopal, Madhya Pradesh, India.
| | - Mohd T Noor
- Department of Gastroenterology, Sri Aurobindo Medical College and PG Institute, Indore, Madhya Pradesh, India.
| |
Collapse
|
10
|
Ren L, Cao Q, Ye H, Dong Z, Zhang C, Yan F, Zhou Y, Zhou H, Zuo J, Wang W. The single degree of polymerization influences the efficacy of xylooligosaccharides in shaping microbial and metabolite profiles in chicken gut to combat avian pathogenic Escherichia coli. BMC Microbiol 2025; 25:227. [PMID: 40264018 PMCID: PMC12013008 DOI: 10.1186/s12866-025-03948-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Avian pathogenic Escherichia coli (APEC) threatens both poultry production and human health. Xylooligosaccharides (XOS) may suppress pathogenic bacteria through prebiotic actions. However, the influences of single degree of polymerization (DP) on the inhibition of APEC by XOS remain unknown. This study aimed to probe if XOS and their major monomers (xylobiose, xylotriose and xylotetraose) could differentially combat APEC via prebiotic actions using an in vitro fermentation model with chicken cecal microbiota. METHODS Microbiota were randomly divided into 7 groups (5 replicate tubes/group). Control group (CON) received no treatment; XOS group received commercial XOS mixtures; APEC group received APEC; XA, X2, X3 and X4 groups received APEC combined with commercial XOS mixtures, xylobiose, xylotriose and xylotetraose, respectively. RESULTS XOS and their major monomers mitigated APEC-induced decline (p < 0.05) in gut microbial α-diversity, with xylotetrose showing the least effect. Gut microbiota in XA, X2, X3 and X4 groups clustered together, with a relative separation observed in X4 group. XOS and their monomers elevated (p < 0.05) the abundances of Firmicutes, Bacteroidota and several probiotics (Lactobacillus, Bacteroides and Megamonas), but reduced (p < 0.05) the abundances of Proteobacteria and Escherichia-Shigella, with xylotetraose exhibiting the least efficacy. Besides, xylotriose and xylotetrose had an advantage over xylotetraose in promoting microbial production of short-chain fatty acids. Metabolomics analysis revealed that APEC challenge mainly downregulated (p < 0.05) several amino acids metabolism pathways of gut microbiota, while xylotriose had an inferiority to XOS in upregulating (p < 0.05) histidine metabolism pathway. Furthermore, microbial fermentation metabolites of all XOS monomers lowered (p < 0.05) certain virulence genes expression in APEC, with xylotriose being the most advantageous. CONCLUSIONS XOS and their major monomers differentially improved gut microbiota and metabolite profiles in chicken gut against APEC challenge. Overall, xylotriose exhibited the greatest inhibition against APEC abundance and virulence. Our findings underscore the role of single DP in influencing the prebiotic actions of XOS against APEC, providing a basis for the reasonable application of XOS in diets to combat bacterial challenge.
Collapse
Affiliation(s)
- Lulu Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qingyun Cao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Hui Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zemin Dong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Changming Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Fei Yan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuping Zhou
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Huiyun Zhou
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jianjun Zuo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Weiwei Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
11
|
Song H, Mun SH, Han DW, Kang JH, An JU, Hwang CY, Cho S. Probiotics ameliorate atopic dermatitis by modulating the dysbiosis of the gut microbiota in dogs. BMC Microbiol 2025; 25:228. [PMID: 40264044 PMCID: PMC12012994 DOI: 10.1186/s12866-025-03924-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Canine atopic dermatitis (cAD) is a chronic inflammatory disease that significantly reduces the quality of life in dogs. Dysbiosis of the gut microbiota affects skin diseases through the gut-skin axis. Therefore, microbiota-targeted therapy may potentially serve as a new management strategy for cAD. The present study aimed to investigate the association between gut microbiota and cAD and to evaluate the effect of probiotics on the clinical symptoms of cAD and gut microbiota in dogs. RESULTS Gut microbiota was analyzed at baseline and after 8 and 16 weeks. Baseline analysis revealed significantly lower (p < 0.05) gut microbial diversity in dogs with cAD than in healthy dogs. Differential abundance analysis showed that Fusobacterium, Megamonas, Collinsella, unclassified Clostridiales, Bacillus, Helicobacter, and Caproiciproducens were significantly more abundant in healthy dogs. In contrast, Clostridioides, Erysipelatoclostridium, Clostridium, Terrisporobacter, and unclassified Ruminococcaceae were significantly more abundant in dogs with cAD, In addition, differential abundance analysis showed that the abundance of 46 metabolic pathways were significantly different between healthy dogs and dogs with cAD indicating the dysbiosis of the gut microbiota in cAD. Moreover, the clinical severity of cAD was negatively correlated (p < 0.05) with alpha diversity and the abundance of Fusobacterium and Megamonas. Notably, daily probiotic administration for 16 weeks significantly decreased the clinical severity (p < 0.05). Dogs with good prognoses exhibited significantly increased alpha diversity, whereas those with poor prognoses did not, suggesting that the therapeutic effects of probiotics may be mediated by changes in gut microbial diversity. CONCLUSIONS This study highlights the association between gut microbiota dysbiosis and cAD in dogs and demonstrates that probiotic administration can effectively ameliorate cAD by improving gut microbial dysbiosis. These findings provide a basis for novel microbiota-based therapies in cAD treatment.
Collapse
Affiliation(s)
- Hyokeun Song
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, South Korea
| | - Seung-Hyun Mun
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Dae-Woong Han
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Jung-Hun Kang
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Jae-Uk An
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Cheol-Yong Hwang
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Seongbeom Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea.
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, South Korea.
- Center for Veterinary Integrated Medicine Research, Seoul National University, Seoul, South Korea.
| |
Collapse
|
12
|
Luo D, Xu R, Jiang L, Zhu Y, Li H, Cao Y, Su Z, Chen Y. Unraveling the protective mechanisms and bioactive components of litchi polysaccharides in intestinal health. Int J Biol Macromol 2025; 310:143383. [PMID: 40268031 DOI: 10.1016/j.ijbiomac.2025.143383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/07/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
In recent years, the rise in intestinal disease has driven the hunt for safer, cost-effective alternatives to traditional, side-effect-laden medications. Litchi polysaccharide (LP), derived from litchi pulp, has emerged as a potential intestinal protector, but its efficacy has not been well-established. Our study have demonstrated LP significantly preserves the integrity of the intestinal barrier in both Caenorhabditis elegans model and antibiotic-exposed mice. Furthermore, LP regulates the gut microbiota, promoting the dominance of beneficial bacteria such as Anaerostipes and Lachnoclostridium in antibiotic-exposed mice and elevating the levels of short-chain fatty acids (SCFAs). LP2-a, a key component making up 11.13 % of LP and with a molecular weight of 72,477 Da, has been isolated and identified as the main active agent. Its molecular structure, featuring galactose and arabinose and possessing a main chain composed of specific sugar units and side chains, is crucial for its protective effects. In C. elegans, LP2-a regulates the expression of intestinal structure-related genes, including up-regulating the expression of act-5 and down-regulating the levels of ajm-1, erm-1, and zoo-1, protecting the integrity of the intestinal barrier. This study provides a theoretical foundation for the potential use of LP, particularly LP2-a, in the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Danxian Luo
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510640, Guangdong, China
| | - Ruina Xu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510640, Guangdong, China
| | - Li Jiang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510640, Guangdong, China
| | - Yi Zhu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510640, Guangdong, China
| | - Huangbo Li
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510640, Guangdong, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510640, Guangdong, China
| | - Zuanxian Su
- College of Horticulture, South China Agricultural University, Guangzhou 510640, Guangdong, China.
| | - Yunjiao Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510640, Guangdong, China.
| |
Collapse
|
13
|
Umar S, Yu W, Xuan H, Ahmed I, Zhong C, Morowitz M, Rogers MB, Attard MI, Sampath V. Neonatal gut microbiota succession in mice mapped over time, site, injury and single immunoglobulin interleukin-1 related receptor genotype. iScience 2025; 28:112243. [PMID: 40248118 PMCID: PMC12005339 DOI: 10.1016/j.isci.2025.112243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/23/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Microbial succession during postnatal gut development in mice is likely impacted by site of sampling, time, intestinal injury, and host genetics. We investigated this in wild-type and Sigirr transgenic mice that encode the p.Y168X mutation identified in a neonate with necrotizing enterocolitis (NEC). Temporal profiling of the ileal and colonic microbiome after birth to weaning revealed a clear pattern of progression from a less diverse, Proteobacteria/Escherichia_Shigella dominant community to a more diverse, Firmicutes/Bacteroidetes dominant community. Formula milk feeding, a risk factor for necrotizing enterocolitis, decreased Firmicutes and increased Proteobacteria leading to enrichment of bacterial genes denoting exaggerated glycolysis and increased production of acetate and lactate. Sigirr transgenic mice exhibited modest baseline differences in microbiota composition but exaggerated formula feeding-induced dysbiosis, mucosal inflammation, and villus injury. Postnatal intestinal microbiota succession in mice resembles human neonates and is shaped by developmental maturity, ileal vs. colonic sampling, formula feeding, and Sigirr genotype.
Collapse
Affiliation(s)
- Shahid Umar
- Department of Surgery, University of Kansas Medical Center, USA
| | - Wei Yu
- Department of Pediatrics/Neonatology, Children’s Mercy Hospital, Kansas City, USA
| | - Hao Xuan
- Department of Electrical Engineering and Computer Science, University of Kansas, USA
| | - Ishfaq Ahmed
- Department of Math, Science and Computer Technology, Kansas City Community College, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, USA
| | - Michael Morowitz
- Division of Pediatric General and Thoracic Surgery, University of Pittsburgh Children’s Hospital, Pittsburgh, PA, USA
| | - Mathew Brian Rogers
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | - Mark Ivan Attard
- Neonatal Unit, Aberdeen Maternity Hospital, Aberdeen AB25 2ZL, UK
| | - Venkatesh Sampath
- Department of Electrical Engineering and Computer Science, University of Kansas, USA
| |
Collapse
|
14
|
Luo Y, Luo L, Xia M, Liu Q, Zhang G. Studies on the changes in rectal permeability and intestinal microbiota with developmental age in young rats. Front Microbiol 2025; 16:1551693. [PMID: 40336831 PMCID: PMC12058081 DOI: 10.3389/fmicb.2025.1551693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/31/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction The gut contains a diverse array of commensal microorganisms, forming a vital biological barrier within the intestine that contributes to the overall intestinal mucosal barrier. However, research on the rectal barrier during early development remains limited. This study aims to investigate the relationship between intestinal microbiota and rectal barrier function in young rats. Methods We evaluated the rectal barrier structure and function in rats at 2-, 4-, and 10-week-old. Methodology included histological analysis, Muc2 expression quantification, immunofluorescence localization of tight junction proteins (ZO-1, Occludin, Claudins), blood glucose monitoring after rectal insulin administration, and 16S rDNA sequencing of rectal microbiota. Spearman correlation analysis was used to explore mechanisms linking age-dependent changes in rectal permeability to microbiota dynamics. Results Physiological rectal permeability was significantly higher in 2-week-old rats compared to 4- and 10-week-old rats (p < 0.01), although systemic biomarkers (LPS, D-LA, and LBP) showed no significant differences. The rectal microbiota exhibited marked age-dependent shifts in composition, α/β-diversity, and metabolic pathways, with increased abundance of beneficial taxa (e.g., Muribaculaceae, Akkermansia) in older rats. Correlation analysis revealed strong associations between reduced permeability, elevated Occludin expression, and microbiota maturation (R = 0.65, p < 0.001). Conclusion This study demonstrates that age-dependent maturation of the rectal barrier is closely linked to microbiota composition and tight junction protein expression, providing insights into developmental mechanisms and potential strategies for pediatric rectal drug delivery.
Collapse
Affiliation(s)
- Yunfeng Luo
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Liangming Luo
- Yudu County Hospital of Traditional Chinese Medicine, Ganzhou, China
| | - Mengle Xia
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qian Liu
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Guosong Zhang
- Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
15
|
D’Auria E, Ferrigno C, Pellicani S, Di Gallo A, Zuccotti GV, Agosti M, Baldassarre ME, Salvatore S. Neonatal Food Protein-Induced Enterocolitis: Current Insights and Knowledge Gaps. J Clin Med 2025; 14:2461. [PMID: 40217910 PMCID: PMC11989300 DOI: 10.3390/jcm14072461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/14/2025] Open
Abstract
Acute and chronic Food Protein-Induced Enterocolitis Syndrome (FPIES) has been well characterized in children; otherwise, neonatal FPIES (N-FPIES) remains poorly understood. In terms of pathophysiology, neonatal FPIES appears to have a more prevalent TH2 response and is characterized by specific clinical features that make the diagnosis challenging. Genetic and environmental risk factors may predispose to the development of FPIES. Recent evidence indicates that a characteristic microbiota signature may lead to barrier dysfunction, reduced regulatory T cells, and abnormal intestinal production of serotonin, responsible for the symptoms of FPIES. Regarding clinical presentation, newborns with FPIES may not fully meet the current guideline's diagnostic criteria at disease onset, being more similar to clinical entity specific of neonatal age than to acute FPIES in infants and children. Hence, differentiation from other neonatal medical and surgical conditions-particularly necrotizing enterocolitis (NEC)-remains a critical challenge for clinicians. This present review highlights our current understanding of N-FPIES, in term of pathophysiology, clinical presentation diagnosis, and treatment strategies. Refining diagnostic criteria for N-FPIES represents a clinical priority to help physicians in diagnosing and managing this challenging condition. Last, but not least, larger clinical trials are needed to optimize treatment practices in term and preterm newborns with FPIES.
Collapse
Affiliation(s)
- Enza D’Auria
- Allergy Unit-Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (C.F.); (A.D.G.)
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy;
| | - Cristina Ferrigno
- Allergy Unit-Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (C.F.); (A.D.G.)
| | - Stefano Pellicani
- Department of Biomedical Science and Human Oncology, Aldo Moro University of Bari, 70124 Bari, Italy;
| | - Anna Di Gallo
- Allergy Unit-Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (C.F.); (A.D.G.)
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy;
- Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy;
| | - Massimo Agosti
- Department of Medicine and Technical Innovation, Pediatrics, Hospital “F. Del Ponte”, University of Insubria, 21100 Varese, Italy; (M.A.); (S.S.)
| | | | - Silvia Salvatore
- Department of Medicine and Technical Innovation, Pediatrics, Hospital “F. Del Ponte”, University of Insubria, 21100 Varese, Italy; (M.A.); (S.S.)
| |
Collapse
|
16
|
Wei S, Ma X, Chen Y, Wang J, Hu L, Liu Z, Mo L, Zhou N, Chen W, Zhu H, Yan S. Alzheimer's Disease-Derived Outer Membrane Vesicles Exacerbate Cognitive Dysfunction, Modulate the Gut Microbiome, and Increase Neuroinflammation and Amyloid-β Production. Mol Neurobiol 2025; 62:5109-5132. [PMID: 39514171 DOI: 10.1007/s12035-024-04579-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Although our understanding of the molecular biology of Alzheimer's disease (AD) continues to improve, the etiology of the disease, particularly the involvement of gut microbiota disturbances, remains a challenge. Outer membrane vesicles (OMVs) play a key role in central nervous system diseases, but the impact of OMVs on AD progression remains unclear. In this study, we hypothesized that AD-derived OMVs (OMVsAD) were a risk factor in AD pathology. To test our hypothesis, young APP/PS1 mice (AD mice) were given OMVsAD by gavage. Young AD mice were euthanized 120 days after gavage to assess the intestinal barrier, gut microbiota diversity, mediators of neuroinflammation, glial markers, amyloid burden, and short-chain fatty acid (SCFA) levels. Our results showed that OMVsAD accelerated cognitive dysfunction after 120 days of intragastric administration. Morris water maze experiment and new object recognition test showed that OMVsAD caused significantly poorer spatial ability learning and memory of the AD mice. We observed the OMVsAD-treated APP/PS1 mice display OMVs disrupting the intestinal barrier compared with controls of normal human-derived OMVs. Compared with the OMVsHC group, claudin-5 and ZO-1 related to the intestinal barrier were significantly downregulated in the OMVsAD group. The OMVsAD activate microglia in the cerebral cortex and hippocampus of AD mice, and the levels of IL-1β, IL-6, TNF-α, and NF-Κb were upregulated. We also found that OMVsAD increased Aβ production. 16S rRNA sequencing showed that OMVsAD negatively regulated the α- and β-diversity index of intestinal flora and reduced the levels of SCFA. OMVsAD may change the intestinal flora of young AD, damage the intestinal mucosa and blood-brain barrier, and accelerate AD neuropathological damage.
Collapse
Affiliation(s)
- Shouchao Wei
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Xiaochen Ma
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Yating Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junjun Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Basic Medicine College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Li Hu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Basic Medicine College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhou Liu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Lang Mo
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Ning Zhou
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Wenrong Chen
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - He Zhu
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.
| | - Shian Yan
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.
| |
Collapse
|
17
|
Chen L, Wang X, Wang S, Liu W, Song Z, Liao H. The impact of gut microbiota on the occurrence, treatment, and prognosis of ischemic stroke. Neurobiol Dis 2025; 207:106836. [PMID: 39952411 DOI: 10.1016/j.nbd.2025.106836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025] Open
Abstract
Ischemic stroke (IS) is a cerebrovascular disease that predominantly affects middle-aged and elderly populations, exhibiting high mortality and disability rates. At present, the incidence of IS is increasing annually, with a notable trend towards younger affected individuals. Recent discoveries concerning the "gut-brain axis" have established a connection between the gut and the brain. Numerous studies have revealed that intestinal microbes play a crucial role in the onset, progression, and outcomes of IS. They are involved in the entire pathophysiological process of IS through mechanisms such as chronic inflammation, neural regulation, and metabolic processes. Although numerous studies have explored the relationship between IS and intestinal microbiota, comprehensive analyses of specific microbiota is relatively scarce. Therefore, this paper provides an overview of the typical changes in gut microbiota following IS and investigates the role of specific microorganisms in this context. Additionally, it presents a comprehensive analysis of post-stroke microbiological therapy and the relationship between IS and diet. The aim is to identify potential microbial targets for therapeutic intervention, as well as to highlight the benefits of microbiological therapies and the significance of dietary management. Overall, this paper seeks to provide key strategies for the treatment and management of IS, advocating for healthy diets and health programs for individuals. Meanwhile, it may offer a new perspective on the future interdisciplinary development of neurology, microbiology and nutrition.
Collapse
Affiliation(s)
- Liying Chen
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xi Wang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Shiqi Wang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Weili Liu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | | | - Huiling Liao
- Neurology Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
18
|
Go EJ, Ryu BR, Gim GJ, Shin YR, Kang MJ, Kim MJ, Baek JS, Lim JD. Regulation of Intestinal Barrier Function and Gut Microbiota by Hot Melt Extrusion-Drug Delivery System-Prepared Mulberry Anthocyanin in an Inflammatory Bowel Disease Model. Pharmaceuticals (Basel) 2025; 18:475. [PMID: 40283912 PMCID: PMC12030684 DOI: 10.3390/ph18040475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Anthocyanins (ACNs) derived from mulberry (Morus alba L.) exhibit potent antioxidant and anti-inflammatory activities. However, their low stability and bioavailability in physiological environments limit their therapeutic potential. This study aimed to enhance the stability and controlled release ACNs using a hot-melt extrusion drug delivery system (HME-DDS) formulation, HME-MUL-F2, and evaluate its effects on gut barrier function and microbiota composition in a DSS-induced colitis model. Methods: The anthocyanin content of HME-MUL-F2 was quantified and compared with that of raw mulberry extract. The formulation's protective effects were assessed in Caco-2 and RAW 264.7 cells, confirming its biocompatibility and anti-inflammatory properties. The therapeutic efficacy was further evaluated in a dextran sulfate sodium (DSS)-induced inflammatory bowel disease (IBD) model, focusing on gut barrier integrity, inflammatory cytokine modulation, and gut microbiota composition. Results: HME-MUL-F2 significantly improved gut barrier function by upregulating tight junction proteins and reducing inflammatory cytokine levels in the colitis model. Moreover, the formulation modulated gut microbiota composition, promoting beneficial bacteria while suppressing pathogenic strains. HME-MUL-F2 administration led to a significant increase in the Bacteroidetes-to-Firmicutes ratio, which is associated with improved gut health. These results indicate that HME-MUL-F2 significantly enhances anthocyanin bioavailability, leading to improved gut health and potential therapeutic applications for inflammatory conditions. Conclusions: This study highlights the potential of HME technology for improving the stability, bioavailability, and therapeutic efficacy of anthocyanins. HME-MUL-F2 is a sustained-release formulation that enhances gut barrier function and modulates intestinal microbial balance in a DSS-induced inflammatory bowel disease model. These findings strongly suggest that the observed therapeutic effects of HME-MUL-F2 are primarily due to enhanced anthocyanin bioavailability and targeted delivery to the colon, although further clinical studies will provide more definitive confirmation.
Collapse
Affiliation(s)
- Eun-Ji Go
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (E.-J.G.); (B.R.R.); (Y.R.S.); (M.J.K.); (M.J.K.); (J.-S.B.)
| | - Byeong Ryeol Ryu
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (E.-J.G.); (B.R.R.); (Y.R.S.); (M.J.K.); (M.J.K.); (J.-S.B.)
- Institute of Cannabis Research, Colorado State University-Pueblo, 2200 Bonforte Blvd, Pueblo, CO 81001-4901, USA
| | - Gyeong Ju Gim
- National Agrobiodiversity Center, National Academy of Agricultural Science, Rural Development Administration, Jeonju 54874, Republic of Korea;
| | - Ye Rim Shin
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (E.-J.G.); (B.R.R.); (Y.R.S.); (M.J.K.); (M.J.K.); (J.-S.B.)
| | - Min Ji Kang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (E.-J.G.); (B.R.R.); (Y.R.S.); (M.J.K.); (M.J.K.); (J.-S.B.)
| | - Min Jun Kim
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (E.-J.G.); (B.R.R.); (Y.R.S.); (M.J.K.); (M.J.K.); (J.-S.B.)
| | - Jong-Suep Baek
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (E.-J.G.); (B.R.R.); (Y.R.S.); (M.J.K.); (M.J.K.); (J.-S.B.)
- Department of Bio-Functional Material, Kangwon National University, Samcheok 25949, Republic of Korea
| | - Jung Dae Lim
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (E.-J.G.); (B.R.R.); (Y.R.S.); (M.J.K.); (M.J.K.); (J.-S.B.)
- Department of Bio-Functional Material, Kangwon National University, Samcheok 25949, Republic of Korea
| |
Collapse
|
19
|
Rodrigues FG, Ormanji MS, Meca R, Montenegro H, Cuppari L, de Borst MH, Heilberg IP. Effects of a high-fat diet on gut microbiota and possible implications for bone health in male Wistar rats. Lipids 2025. [PMID: 40103344 DOI: 10.1002/lipd.12440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/20/2025]
Abstract
Diet plays an important role in the composition of gut microbiota. Emerging research suggests that bone homeostasis can also be influenced by the gut microbiota. The aim of this study was to assess possible alterations in gut microbiota in an experimental obesity model induced by a high-fat diet (HFD) and the possible effects on parameters of bone metabolism and remodeling. Male Wistar rats were fed a HFD (60% lipids) or standard (control) diet for 14 weeks. Biochemical and hormonal parameters, bone histomorphometry, bone protein levels, and gut microbiota composition were analyzed. HFD animals exhibited a greater gut microbiota α-diversity represented by the Shannon Index and an increased relative abundance of the Proteobacteria phylum. Histomorphometry detected lower bone formation in the HFD group, accompanied by increased levels of serum and bone leptin and FGF-23 (fibroblast growth factor-23). The Shannon Index was correlated directly with bone FGF-23 (R 0.96, p = 0.04) and inversely with the osteoblastic surface (R -0.95, p = 0.04). The present study disclosed a significant increase in gut microbiota α-diversity and relative abundance of Proteobacteria phylum in obese animals fed a high-fat diet in parallel with increased levels of bone and serum leptin and FGF-23 and lower bone formation. The associations of Shannon Index with bone levels of FGF-23 and reduced osteoblastic surface suggest a link between HFD-induced higher gut microbiota diversity and low bone formation.
Collapse
Affiliation(s)
- Fernanda Guedes Rodrigues
- Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Renata Meca
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | | | - Lilian Cuppari
- Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Martin H de Borst
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ita Pfeferman Heilberg
- Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
20
|
Ding W, Cheng Y, Liu X, Zhu Z, Wu L, Gao J, Lei W, Li Y, Zhou X, Wu J, Gao Y, Ling Z, Jiang R. Harnessing the human gut microbiota: an emerging frontier in combatting multidrug-resistant bacteria. Front Immunol 2025; 16:1563450. [PMID: 40165964 PMCID: PMC11955657 DOI: 10.3389/fimmu.2025.1563450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Antimicrobial resistance (AMR) has become a major and escalating global health threat, undermining the effectiveness of current antibiotic and antimicrobial therapies. The rise of multidrug-resistant bacteria has led to increasingly difficult-to-treat infections, resulting in higher morbidity, mortality, and healthcare costs. Tackling this crisis requires the development of novel antimicrobial agents, optimization of current therapeutic strategies, and global initiatives in infection surveillance and control. Recent studies highlight the crucial role of the human gut microbiota in defending against AMR pathogens. A balanced microbiota protects the body through mechanisms such as colonization resistance, positioning it as a key ally in the fight against AMR. In contrast, gut dysbiosis disrupts this defense, thereby facilitating the persistence, colonization, and dissemination of resistant pathogens. This review will explore how gut microbiota influence drug-resistant bacterial infections, its involvement in various types of AMR-related infections, and the potential for novel microbiota-targeted therapies, such as fecal microbiota transplantation, prebiotics, probiotics, phage therapy. Elucidating the interactions between gut microbiota and AMR pathogens will provide critical insights for developing novel therapeutic strategies to prevent and treat AMR infections. While previous reviews have focused on the general impact of the microbiota on human health, this review will specifically look at the latest research on the interactions between the gut microbiota and the evolution and spread of AMR, highlighting potential therapeutic strategies.
Collapse
Affiliation(s)
- Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingbin Wu
- Department of Intensive Care Unit, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| | - Jie Gao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Yating Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Center for Genomics and Personalized Medicine, Stanford, CA, United States
- Stanford Diabetes Research Center, Stanford, CA, United States
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - Jian Wu
- Department of Clinical Laboratory, Suzhou Municipal Hospital, Suzhou, Jiangsu, China
| | - Yongtao Gao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ruilai Jiang
- Department of Intensive Care Unit, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| |
Collapse
|
21
|
Chavez-Arroyo A, Radlinski LC, Bäumler AJ. Principles of gut microbiota assembly. Trends Microbiol 2025:S0966-842X(25)00071-X. [PMID: 40089422 DOI: 10.1016/j.tim.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
The gut microbiota plays a critical role in human health, yet its taxonomic complexity, interpersonal variability, and resistance to change in adulthood present challenges for understanding the factors driving shifts in its composition and function. Here, we propose a hierarchy of ecological factors governing gut microbiota assembly, stability, and resilience. At the apex of this hierarchy is habitat filtering by host-derived electron acceptors, which dictates the ecological guilds that dominate distinct gut regions. Host dietary behavior shapes niche availability within these ecological guilds by regulating nutrient availability. Priority effects preserve taxonomic stability whereas microbial antagonism governs competition for open ecological positions. This framework highlights how host control over microbial energy metabolism directs microbiota self-assembly and maintains gut homeostasis.
Collapse
Affiliation(s)
- Alfredo Chavez-Arroyo
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Lauren C Radlinski
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
22
|
Huang H, Zhao T, Ma W. Omega-3 polyunsaturated fatty acids attenuates cognitive impairment via the gut-brain axis in diabetes-associated cognitive dysfunction rats. Brain Behav Immun 2025; 127:147-169. [PMID: 40068791 DOI: 10.1016/j.bbi.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 02/11/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Diabetes-related cognitive dysfunction (DACD) is a comorbidity of type 2 diabetes that has a negative effect on patients' quality of life. Research has indicated that disruption of the gut microbiota (GM) may be linked to dementia with altered cognitive performance. Conversely, omega-3 polyunsaturated fatty acids (n-3 PUFAs) may reverse DACD. The present study aimed to assess the effects of an n-3 PUFA intervention and fecal microbiota transplantation (FMT) on high-fat and streptozotocin-induced DACD model rats. In DACD rats, n-3 PUFA treatment restored fasting blood glucose (FBG) levels and cognitive function, increased the expression of anti-inflammatory cytokines and downregulated the expression of proinflammatory cytokines in the cortex and colon. Additionally, the expression of the postsynaptic density protein-95 mRNA and protein varied with n-3 PUFA treatment. Treatment with n-3 PUFAs also increased the expression of tight junction proteins. Beneficial and short-chain fatty acid-producing bacteria were more abundant when rats were exposed to n-3 PUFAs. After FMT from the rats with DACD symptoms that were improved by the n-3 PUFA dietary intervention into another batch of DACD rats, we observed recovery in recipient DACD rats. These results indicated that the alleviation of DACD symptoms by n-3 PUFAs was attributed to gut microbiota remodeling.
Collapse
Affiliation(s)
- Hongying Huang
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Nanchang Institute of Disease Control and Prevention, China Railway Nanchang Bureau Group Co., Ltd., Nanchang, 330003, People's Republic of China
| | - Tong Zhao
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Weiwei Ma
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China.
| |
Collapse
|
23
|
Davison S, Mascellani Bergo A, Ward Z, Sackett A, Strykova A, Jaimes JD, Travis D, Clayton JB, Murphy HW, Danforth MD, Smith BK, Blekhman R, Fuh T, Niatou Singa FS, Havlik J, Petrzelkova K, Gomez A. Cardiometabolic disease risk in gorillas is associated with altered gut microbial metabolism. NPJ Biofilms Microbiomes 2025; 11:33. [PMID: 39984469 PMCID: PMC11845621 DOI: 10.1038/s41522-025-00664-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/03/2025] [Indexed: 02/23/2025] Open
Abstract
Cardiometabolic disease is the leading cause of death in zoo apes; yet its etiology remains unknown. Here, we investigated compositional and functional microbial markers in fecal samples from 57 gorillas across U.S. zoos, 20 of which are diagnosed with cardiovascular disease, in contrast with 17 individuals from European zoos and 19 wild gorillas from Central Africa. Results show that zoo-housed gorillas in the U.S. exhibit the most diverse gut microbiomes and markers of increased protein and carbohydrate fermentation, at the expense of microbial metabolic traits associated with plant cell-wall degradation. Machine learning models identified unique microbial traits in U.S. gorillas with cardiometabolic distress; including reduced metabolism of sulfur-containing amino acids and hexoses, increased abundance of potential enteric pathogens, and low fecal butyrate and propionate production. These findings show that cardiometabolic disease in gorillas is potentially associated with altered gut microbial function, influenced by zoo-specific diets and environments.
Collapse
Affiliation(s)
- Samuel Davison
- Department of Animal Science, University of Minnesota, Saint Paul, MN, USA
- Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Anna Mascellani Bergo
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Zoe Ward
- Department of Animal Science, University of Minnesota, Saint Paul, MN, USA
| | - April Sackett
- Department of Animal Science, University of Minnesota, Saint Paul, MN, USA
| | - Anna Strykova
- Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - José Diógenes Jaimes
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Dominic Travis
- The Marine Mammal Center, Sausalito, CA, USA
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jonathan B Clayton
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Hayley W Murphy
- HWM and MDD: Great Ape Heart Project, Detroit Zoological Society, Royal Oak, MI, USA
| | - Marietta D Danforth
- HWM and MDD: Great Ape Heart Project, Detroit Zoological Society, Royal Oak, MI, USA
| | | | - Ran Blekhman
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Terence Fuh
- WWF Central African Republic, Bayanga, Central African Republic
| | | | - Jaroslav Havlik
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czech Republic.
| | - Klara Petrzelkova
- Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic.
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Brno, Czech Republic.
- Liberec Zoo, Liberec, Czech Republic.
| | - Andres Gomez
- Department of Animal Science, University of Minnesota, Saint Paul, MN, USA.
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
24
|
Chen L, Tu Z, Zhong Z, Wei S, Hu M, Wang Y. Differential effects of polyvinyl chloride microplastics and kaolin particles on gut immunity of mussels at environmental concentrations. JOURNAL OF HAZARDOUS MATERIALS 2025; 484:136711. [PMID: 39647329 DOI: 10.1016/j.jhazmat.2024.136711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
Both microplastics (MPs) and kaolin are marine suspended particles capable of influencing the physiology of bivalve mollusks. However, the current research on MPs lacks the analysis of their own physical and chemical toxicity, and the comparative study of the toxicity of microplastics and natural suspended particles (NSPs) in aquatic environment. In this work, three experiments are layered, with Experiment 1 directly comparing polyvinyl chloride MPs (PVC MPs) and kaolin and showing that MPs have greater deleterious effects on thick-shelled mussels than kaolin, with the exception of physical damage and effects on gut microorganisms. As the presence or absence of chemicals may be the main difference between MPs and kaolin, in Experiment 2 the toxicity drivers of PVC MPs itself were investigated, demonstrating that the chemicals in MPs are indeed toxic and that the harmful effects of MPs on mussels may be due to the superposition of their own physical and chemical toxicity. Finally, in Experiment 3 mussels were exposed to the chemicals in PVC MPs and kaolin in a composite and found that the toxicity of the composite exposure was greater than that of the single exposure to kaolin, suggesting that the chemicals may be the main factor contributing to the difference in toxicity between PVC MPs and kaolin. In conclusion, this work addresses the lack of a natural particle control group in current studies of MPs, confirms that the toxicity drivers of MPs are due to both physical and chemical factors, highlights the role of NSPs in the environment, and provides new insights for evaluating the toxic effects of MPs in the natural marine environment.
Collapse
Affiliation(s)
- Liming Chen
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China
| | - Zhihan Tu
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Zhen Zhong
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China
| | - Shuaishuai Wei
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China
| | - Menghong Hu
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China; State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200241, China
| | - Youji Wang
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China.
| |
Collapse
|
25
|
Tjaden NEB, Liou MJ, Sax SE, Lassoued N, Lou M, Schneider S, Beigel K, Eisenberg JD, Loeffler E, Anderson SE, Yan G, Litichevskiy L, Dohnalová L, Zhu Y, Jin DMJC, Raab J, Furth EE, Thompson Z, Rubenstein RC, Pilon N, Thaiss CA, Heuckeroth RO. Dietary manipulation of intestinal microbes prolongs survival in a mouse model of Hirschsprung disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637436. [PMID: 39990395 PMCID: PMC11844371 DOI: 10.1101/2025.02.10.637436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Enterocolitis is a common and potentially deadly manifestation of Hirschsprung disease (HSCR) but disease mechanisms remain poorly defined. Unexpectedly, we discovered that diet can dramatically affect the lifespan of a HSCR mouse model ( Piebald lethal , sl/sl ) where affected animals die from HAEC complications. In the sl/sl model, diet alters gut microbes and metabolites, leading to changes in colon epithelial gene expression and epithelial oxygen levels known to influence colitis severity. Our findings demonstrate unrecognized similarity between HAEC and other types of colitis and suggest dietary manipulation could be a valuable therapeutic strategy for people with HSCR. Abstract Hirschsprung disease (HSCR) is a birth defect where enteric nervous system (ENS) is absent from distal bowel. Bowel lacking ENS fails to relax, causing partial obstruction. Affected children often have "Hirschsprung disease associated enterocolitis" (HAEC), which predisposes to sepsis. We discovered survival of Piebald lethal ( sl/sl ) mice, a well-established HSCR model with HAEC, is markedly altered by two distinct standard chow diets. A "Protective" diet increased fecal butyrate/isobutyrate and enhanced production of gut epithelial antimicrobial peptides in proximal colon. In contrast, "Detrimental" diet-fed sl/sl had abnormal appearing distal colon epithelium mitochondria, reduced epithelial mRNA involved in oxidative phosphorylation, and elevated epithelial oxygen that fostered growth of inflammation-associated Enterobacteriaceae . Accordingly, selective depletion of Enterobacteriaceae with sodium tungstate prolonged sl/sl survival. Our results provide the first strong evidence that diet modifies survival in a HSCR mouse model, without altering length of distal colon lacking ENS. Highlights Two different standard mouse diets alter survival in the Piebald lethal ( sl/sl ) mouse model of Hirschsprung disease, without impacting extent of distal colon aganglionosis (the region lacking ENS). Piebald lethal mice fed the "Detrimental" diet had many changes in colon epithelial transcriptome including decreased mRNA for antimicrobial peptides and genes involved in oxidative phosphorylation. Detrimental diet fed sl/sl also had aberrant-appearing mitochondria in distal colon epithelium, with elevated epithelial oxygen that drives lethal Enterobacteriaceae overgrowth via aerobic respiration. Elimination of Enterobacteriaceae with antibiotics or sodium tungstate improves survival of Piebald lethal fed the "Detrimental diet". Graphical abstract
Collapse
|
26
|
Xie C, Cheng J, Chen P, Yan X, Luo C, Qu H, Shu D, Ji J. Integrating gut and IgA-coated microbiota to identify Blautia as a probiotic for enhancing feed efficiency in chickens. IMETA 2025; 4:e264. [PMID: 40027490 PMCID: PMC11865324 DOI: 10.1002/imt2.264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 03/05/2025]
Abstract
This study explores the role of IgA-coated bacteria in improving feed efficiency in chickens, offering a novel perspective for probiotic screening. Chickens with high feed efficiency were found to have a greater abundance of Gram-positive bacteria, while low feed efficiency chickens exhibited higher levels of Gram-negative bacteria and potential pathogens. Through fecal microbiota transplantation (FMT) and integrating analysis of cecal and IgA-coated microbiota, we precisely identified Blautia as a key genus linked to improved feed efficiency. Further validation demonstrated that Blautia coccoides, a representative species of this genus, enhances feed efficiency and activates B cells to produce Immunoglobulin A (IgA), both in vivo and in vitro. Our findings provide new insights into the potential of IgA-coated bacteria as functional probiotics, offering a promising strategy for enhancing feed efficiency in animal production.
Collapse
Affiliation(s)
- Chunlin Xie
- State Key Laboratory of Swine and Poultry Breeding IndustryGuangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural SciencesGuangzhouChina
| | - Jiaheng Cheng
- State Key Laboratory of Swine and Poultry Breeding IndustryGuangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural SciencesGuangzhouChina
| | - Peng Chen
- State Key Laboratory of Swine and Poultry Breeding IndustryGuangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural SciencesGuangzhouChina
| | - Xia Yan
- State Key Laboratory of Swine and Poultry Breeding IndustryGuangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural SciencesGuangzhouChina
| | - Chenglong Luo
- State Key Laboratory of Swine and Poultry Breeding IndustryGuangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural SciencesGuangzhouChina
| | - Hao Qu
- State Key Laboratory of Swine and Poultry Breeding IndustryGuangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural SciencesGuangzhouChina
| | - Dingming Shu
- State Key Laboratory of Swine and Poultry Breeding IndustryGuangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural SciencesGuangzhouChina
| | - Jian Ji
- State Key Laboratory of Swine and Poultry Breeding IndustryGuangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural SciencesGuangzhouChina
| |
Collapse
|
27
|
Radlinski LC, Bäumler AJ. Microbiome science needs more microbiologists. Nat Microbiol 2025; 10:263-264. [PMID: 39843550 DOI: 10.1038/s41564-024-01922-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Affiliation(s)
- Lauren C Radlinski
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA, USA.
| |
Collapse
|
28
|
Atanasova K, Knödler LL, Reindl W, Ebert MP, Thomann AK. Role of the gut microbiome in psychological symptoms associated with inflammatory bowel diseases. Semin Immunopathol 2025; 47:12. [PMID: 39870972 PMCID: PMC11772462 DOI: 10.1007/s00281-025-01036-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/02/2025] [Indexed: 01/29/2025]
Abstract
The brain-gut axis constitutes the basis for the bidirectional communication between the central nervous system and the gastrointestinal tract driven by neural, hormonal, metabolic, immunological, and microbial signals. Alterations in the gut microbiome composition as observed in inflammatory bowel diseases can modulate brain function and emerging empirical evidence has indicated that interactions among the brain-gut microbiome-axis seem to play a significant role in the pathogenesis of both inflammatory bowel diseases and psychiatric disorders and their comorbidity. Yet, the immunological and molecular mechanisms underlying the co-occurrence of inflammatory bowel diseases and psychological symptoms are still poorly understood. The aim of this narrative review is to highlight contemporary empirical findings supporting a pivotal role of the gut microbiome in the pathophysiology of highly prevalent neuropsychiatric symptoms in inflammatory bowel diseases such as fatigue, depression, and anxiety. Finally, we focus on microbiome modulation as potential treatment option for comorbid neuropsychiatric symptoms in immune-mediated diseases and especially in inflammatory bowel diseases. High-quality clinical trials are required to clarify how microbiome modulation through dietary interventions or probiotic, prebiotic or synbiotic treatment can be used clinically to improve mental health and thus quality of life of patients with inflammatory bowel diseases.
Collapse
Affiliation(s)
- Konstantina Atanasova
- Department of Medicine II, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany.
- Department of Psychosomatic Medicine, Medical Faculty Mannheim, Central Institute for Mental Health Mannheim, Heidelberg University, Mannheim, Germany.
| | - Laura-Louise Knödler
- Department of Medicine II, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Wolfgang Reindl
- Department of Medicine II, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Philip Ebert
- Department of Medicine II, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Anne Kerstin Thomann
- Department of Medicine II, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
29
|
Guo F, Qiao J, Hu Z, Huang J, Bi R, Abbas W, Zhen W, Guo Y, Wang Z. Yeast cell wall polysaccharides accelerate yet in-feed antibiotic delays intestinal development and maturation via modulating gut microbiome in chickens. J Anim Sci Biotechnol 2025; 16:14. [PMID: 39856758 PMCID: PMC11763161 DOI: 10.1186/s40104-024-01145-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND It is important to promote intestinal development and maturation of chicks for feed digestion and utilization, intestinal health, and disease resistance. This study aimed to investigate the effects of dietary yeast cell wall polysaccharides (YCWP) addition on intestinal development and maturation of chickens and its potential action mechanism. METHODS 180 one-day-old male Arbor Acres broilers were randomly assigned to three groups containing control (basal diets without any antibiotics or anticoccidial drug), bacitracin methylene disalicylate (BMD)-treated group (50 mg/kg) and YCWP-supplemented group (100 mg/kg). RESULTS Compared with control group, in-feed antibiotic BMD continuous administration significantly decreased crypt depth (d 21) and villus height (d 42) along with mucosal maltase activity (d 42) in the ileum (P < 0.05). Also, BMD markedly downregulated gene expression levels of β-catenin, lysozyme, occludin and FABP-2 (d 21) and innate immune related genes CD83 and MHC-I mRNA levels (d 42, P < 0.05), and decreased goblet cell counts in the ileum of chickens (d 21 and d 42, P < 0.05). While, TLR-2, TLR-6 and iNOS mRNA abundances were notably upregulated by BMD treatment (d 42, P < 0.05). Nevertheless, dietary YCWP addition significantly increased the ratio of villus height to crypt depth (d 21), villus surface area (d 21 and d 42), ileal alkaline phosphatase and maltase activities as well as goblet cell (d 21 and d 42) and IgA-producing plasma cell numbers as compared to BMD treatment (d 21, P < 0.05). YCWP addition also upregulated gene expression levels of Lgr5, Wnt/β-catenin signaling pathway related gene (Wnt3, β-catenin, d 21; β-catenin, d 42), intestinal cells proliferation marker Ki-67 and barrier function related genes (occludin, d 21 and d 42, P < 0.05). Moreover, YCWP significantly increased antigen presenting cell marker related genes (MHC-II, d 21; CD83 and MHC-I, d 42), TLR-1, TLR-2 and TLR-6 mRNA levels (d 21, P < 0.05). Cecal microbiome analysis showed that YCWP addition obviously improved cecal microbial composition, as indicated by increasing relative abundance of Fournierella, Psychrobacter and Ruminiclostridium on d 21, and Alistipes and Lactobacillus on d 42, which were positively related with gut development and maturation related indexes (P < 0.05). CONCLUSION Collectively, YCWP promoted yet antibiotic BMD delayed intestinal morphological and immunological development linked with modulating gut microbiome in chickens.
Collapse
Affiliation(s)
- Fangshen Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Jianing Qiao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Zeqiong Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Jia Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Ruichen Bi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Waseem Abbas
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Wenrui Zhen
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, People's Republic of China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Zhong Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China.
| |
Collapse
|
30
|
Li L, Zhong S, Ye J, Hu S, Xiong X, Chen G, Hu Z. Shenmai injection revives cardiac function in rats with hypertensive heart failure: involvement of microbial-host co-metabolism. BMC Complement Med Ther 2025; 25:24. [PMID: 39856640 PMCID: PMC11761217 DOI: 10.1186/s12906-024-04737-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
Heart failure (HF) is a complex syndrome marked by considerable expenditures and elevated mortality and morbidity rates globally. Shenmai injection (SMI), a form of Traditional Chinese Medicine-based therapy, has demonstrated effectiveness in treating HF. Recent research suggests that Traditional Chinese Medicine (TCM) may induce beneficial changes in microbial-host co-metabolism, potentially providing cardiovascular protection. This study used a rat model of hypertensive heart failure (H-HF) to explore the mechanism of SMI. The possible compounds and key targets of SMI against H-HF were investigated using network pharmacology. The pharmacodynamics of SMI were validated using the H-HF animal model, with analysis of fecal gut microbiota integrating metabolomics and 16S rRNA sequencing. Metorigin metabolite traceability analysis and the MetaboAnalyst platform were utilized to explore the action mechanism. To evaluate changes in serum TMAO levels, targeted metabolomics was performed. Finally, the study looked at the intrinsic relationships among modifications in the intestinal flora, metabolite profile changes, and the targets of SMI compounds to clarify how they might be used to treat H-HF. According to metabolomics and 16S rRNA sequencing, by reestablishing homeostasis in the gut microbiota, SMI affects vital metabolic pathways, such as energy metabolism, amino acid metabolism, and bile acid metabolism. Increased serum TMAO levels were identified to be a risk factor for H-HF, and SMI was able to downregulate the levels of TMAO-related metabolites. Network pharmacology analysis identified 13 active components of SMI targeting 46 proteins, resulting in differential expression changes in 8 metabolites and 24 gut microbes. In conclusion, this study highlights the effectiveness of SMI in alleviating H-HF and its potential to modulate microbial-host co-metabolism. Through a comprehensive discussion of the interconnected relationships among the components, targets, metabolites, and gut microbiota, it provided fresh light on the therapeutic mechanism of SMI on H-HF.
Collapse
Affiliation(s)
- Lin Li
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Senjie Zhong
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiahao Ye
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyuan Hu
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiajun Xiong
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Guangyu Chen
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Engineering Technology Research Center For Medicinal and Functional Food, Changsha, Hunan, China
| | - Zhixi Hu
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
31
|
Zheng ZL, Zheng QF, Wang LQ, Liu Y. Bowel preparation before colonoscopy: Consequences, mechanisms, and treatment of intestinal dysbiosis. World J Gastroenterol 2025; 31:100589. [PMID: 39811511 PMCID: PMC11684204 DOI: 10.3748/wjg.v31.i2.100589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/22/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
The term "gut microbiota" primarily refers to the ecological community of various microorganisms in the gut, which constitutes the largest microbial community in the human body. Although adequate bowel preparation can improve the results of colonoscopy, it may interfere with the gut microbiota. Bowel preparation for colonoscopy can lead to transient changes in the gut microbiota, potentially affecting an individual's health, especially in vulnerable populations, such as patients with inflammatory bowel disease. However, measures such as oral probiotics may ameliorate these adverse effects. We focused on the bowel preparation-induced changes in the gut microbiota and host health status, hypothesized the factors influencing these changes, and attempted to identify measures that may reduce dysbiosis, thereby providing more information for individualized bowel preparation for colonoscopy in the future.
Collapse
Affiliation(s)
- Ze-Long Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Qing-Fan Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Li-Qiang Wang
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Yi Liu
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
32
|
Heckmann ND, Culler MW, Mont MA, Lieberman JR, Parvizi J. Emerging Concepts in Periprosthetic Joint Infection Research: The Human Microbiome. J Arthroplasty 2025:S0883-5403(25)00001-4. [PMID: 39798621 DOI: 10.1016/j.arth.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/26/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
Microorganisms, including bacteria, fungi, and viruses, that reside on and within the human body are collectively known as the human microbiome. Dysbiosis, or disruption in the microbiome, has been implicated in several disease processes, including asthma, obesity, autoimmune diseases, and numerous other conditions. While the Human Microbiome Project and the generation of descriptive studies it inspired established correlations between characteristic patterns in the composition of the microbiome and specific disease phenotypes, current research has begun to focus on elucidating the causal role of the microbiome in disease pathogenesis. Within the field of orthopaedic surgery, researchers have proposed the concept of a "gut-joint axis," whereby the intestinal microbiome influences joint health and the development of diseases, such as osteoarthritis and periprosthetic joint infection (PJI). It is theorized that intestinal dysbiosis increases gut permeability, leading to the translocation of bacteria and their metabolic products into the systemic circulation and the stimulation of proinflammatory response cascades throughout the body, including within the joints. While correlative studies have identified patterns of dysbiotic derangement associated with osteoarthritis and PJI, translational research is needed to clarify the precise mechanisms by which these changes influence disease processes. Additionally, an emerging body of literature has challenged the previously held belief that certain body sites are sterile and do not possess a microbiome, with studies identifying distinct microbial genomic signatures and a core microbiome that varies between anatomic sites. A more thorough characterization of the joint microbiome may have profound implications for our understanding of PJI pathogenesis and our ability to stratify patients based on risk. The purpose of this review was to outline our current understanding of the human microbiome to describe the gut-joint axis and its role in specific pathologies, including PJI, and to highlight the potential of microbiome-based therapeutic interventions in the field of orthopaedics.
Collapse
Affiliation(s)
- Nathanael D Heckmann
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - McKenzie W Culler
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Michael A Mont
- LifeBridge Health, Sinai Hospital of Baltimore, The Rubin Institute for Advanced Orthopaedics, Baltimore, Maryland, United States
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Javad Parvizi
- International Joint Center, Acibadem University Hospital, Istanbul, Turkey
| |
Collapse
|
33
|
Rong Y, Zhu M, Wang N, Zhang F, Liu T. Photodynamic therapy with a novel photosensitizer inhibits DSS-induced ulcerative colitis in rats via the NF-κB signaling pathway. Front Pharmacol 2025; 15:1539363. [PMID: 39845801 PMCID: PMC11750845 DOI: 10.3389/fphar.2024.1539363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Ulcerative colitis (UC) is an inflammatory bowel disease characterized by inflammation and ulceration of the digestive tract. Methods Photodynamic therapy (PDT) with a novel photosensitizer LD4 was used to treat UC rat models to explore the therapeutic effect and mechanism of LD4-PDT on UC. 16S ribosomal RNA was used to detect the composition of Gut microbiota. Results Our findings indicate that LD4-PDT could protect the integrity of the colonic mucosa, alleviate the inflammatory response and promote the healing of colonic mucosa. Mechanism studies demonstrated that LD4-PDT could inhibit the NF-κB signaling pathway, downregulated the expression of the inflammatory factors' tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and myeloperoxidase (MPO), increased the contents of glutathione (GSH) and superoxide dismutase (SOD) and decreased the content of malondialdehyde (MDA). Additionally, analysis of gut microbiota revealed that LD4-PDT treatment could decrease the abundance of the Proteobacteria phylum in fecal samples, while no significant differences were observed in the Firmicutes, Bacteroidetes, or Actinobacteria phyla among the three groups using 16S rRNA analysis. Discussion In summary, our data suggested that LD4-PDT could inhibit DSS-induced UC in rats via the NF-κB signaling pathway, indicating its potential as a novel photosensitizer for the treatment of UC.
Collapse
Affiliation(s)
- Yumei Rong
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
- Tianjin Key Laboratory of Biomedical Material, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Minghui Zhu
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Nan Wang
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Feiyu Zhang
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Tianjun Liu
- Tianjin Key Laboratory of Biomedical Material, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
34
|
Smith MZ, York M, Townsend KS, Martin LM, Gull T, Coghill LM, Ericsson AC, Johnson PJ. Effects of orally administered clioquinol on the fecal microbiome of horses. J Vet Intern Med 2025; 39:e17276. [PMID: 39709594 PMCID: PMC11663420 DOI: 10.1111/jvim.17276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Whereas restoration of fecal consistency after treatment with clioquinol for chronic diarrhea and free fecal water syndrome has been attributed to its antiprotozoal properties, actions of clioquinol on the colonic bacterial microbiota have not been investigated. OBJECTIVES Characterize the dynamics of fecal microbial diversity before, during, and after PO administration of clioquinol to healthy horses. STUDY DESIGN Experimental prospective cohort study using a single horse group. METHODS Eight healthy adult horses received PO clioquinol (10 g, daily) for 7 days. Feces were obtained daily for 7 days before, during, and after conclusion of treatment, and again 3 months later. Libraries of 16S rRNA V4 region amplicons generated from fecal DNA were sequenced using the Illumina sequencing platform. Bioinformatic analysis was undertaken with QIIME2 and statistical analyses included analysis of variance (ANOVA) and permutational multivariate ANOVA (PERMANOVA). RESULTS The richness and composition of the fecal microbiome was altered after administration of clioquinol, reaching a maximum effect by the fifth day of administration. Changes included a 90% decrease in richness, and compensatory expansion of facultative anaerobes including Streptococcaceae, Enterococcaceae, and Enterobacteriaceae. Multiple horses had Salmonella cultured from feces. MAIN LIMITATIONS Limitations including lack of control group and modest sample size are obviated by robust longitudinal study design and strong effect size associated with drug exposure. CONCLUSIONS Clioquinol has broad-spectrum antibacterial effects on the fecal microbiome of horses, but spares certain bacterial families including several pathogens and pathobionts. Clioquinol should be used with caution in horses, in an environment free of contamination with fecal pathogens.
Collapse
Affiliation(s)
- Mikaila Z. Smith
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
| | - Mary York
- University of Missouri (MU) Bioinformatics and Analytics CoreBond Life Sciences CenterColumbia, Missouri 65201USA
- Institute for Data Science and InformaticsUniversity of MissouriColumbia, Missouri 65211USA
| | - Kile S. Townsend
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65211USA
| | - Lynn M. Martin
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65211USA
| | - Tamara Gull
- MU Veterinary Medical Diagnostic Laboratory (VMDL), College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65211USA
| | - Lyndon M. Coghill
- University of Missouri (MU) Bioinformatics and Analytics CoreBond Life Sciences CenterColumbia, Missouri 65201USA
- Department of Veterinary Pathobiology, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65201USA
| | - Aaron C. Ericsson
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
- Department of Veterinary Pathobiology, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65201USA
- University of Missouri Metagenomics CenterColumbia, Missouri 65201USA
| | - Philip J. Johnson
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65211USA
| |
Collapse
|
35
|
Hackstein CP. Liver damage and immune responses. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025; 63:56-64. [PMID: 39793602 DOI: 10.1055/a-2365-3796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2025]
Abstract
Chronic liver disease (CLD) has massive systemic repercussions including major impacts on the body's immune system. Abnormalities in phenotype, function and numbers of various immune cell subsets have been established by a large number of clinical and pre-clinical studies. The loss of essential immune functions renders CLD-patients exceptionally susceptible to bacterial and viral infections and also impairs the efficacy of vaccination. Consequently, infections represent a major clinical issue causing significant morbidity and mortality in these patients. Mechanistically, the immune dysfunction associated with CLD results from the increased translocation of bacteria and bacterial cues from the intestine. These trigger a signaling axis around the cytokines IFN I and IL-10 in hepatic myeloid cells, which aside from impairing the function of the myeloid cells themselves, also has notable negative impacts on the functionality of other immune cells. T cells in CLD-patients and -models are especially affected by this signaling axis and display a variety of quantitative and qualitative defects. Due to the high clinical relevance, understanding the mechanisms underlaying CED-associated immune dysfunction is of critical importance to discover and develop new therapeutic targets.
Collapse
Affiliation(s)
- Carl-Philipp Hackstein
- Institut für Molekulare Immunologie, Technische Universität München, München, Germany
- Zentrum für Infektionsprävention (ZIP), Technische Universität München, Freising, Germany
| |
Collapse
|
36
|
Li H, Wu Z, Yu B, Chen J, Yang C, Guo Y, Sun B. Dietary Capsaicin Supplementation Mitigates Calving-Induced Stress and Enhances Antioxidant Capacity, Immune Function, and Gut Microbiota in Periparturient Dairy Cows. Antioxidants (Basel) 2024; 14:28. [PMID: 39857362 PMCID: PMC11762672 DOI: 10.3390/antiox14010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
This study investigated the effects of dietary capsaicin supplementation on antioxidant capacity, immune function, and gut microbiota in periparturient dairy cows. Twenty Holstein cows with an average parity of 2.5 ± 0.76, milk production of 31.30 ± 2.39 kg, and 36.10 ± 2.38 days to calving were randomly assigned to either a control group fed a basal diet or a treatment group supplemented with 1.2 g/head/day of capsaicin. The supplementation was administered during an evaluation period spanning from 28 days before delivery to 21 days after delivery using a randomized block experimental design. Results showed that capsaicin significantly reduced milk somatic cell count and pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) while enhancing serum antioxidant enzymes (SOD, GSH-Px, and CAT) and immunoglobulin levels (IgG, IgA, and IgM). Moreover, capsaicin altered gut microbiota composition, increasing the relative abundance of beneficial genera. These findings suggest that dietary capsaicin supplementation during the transition period improves lactation performance and supports immune function, as well as alleviates oxidative stress. This study highlights the potential of capsaicin as a practical dietary strategy for enhancing productivity in dairy farming.
Collapse
Affiliation(s)
| | | | | | | | | | - Yongqing Guo
- Herbivore Research Laboratory, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| | - Baoli Sun
- Herbivore Research Laboratory, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou 510642, China
| |
Collapse
|
37
|
Jørgensen AB, Almer L, Samaniego Castruita JA, Pedersen MS, Kirkby NS, Jensen EA, Alfaro-Núñez A, Friis-Hansen L, Brandstrup B. The baseline fecal microbiome differs in patients with and without anastomotic leakage after colorectal cancer surgery. Heliyon 2024; 10:e40616. [PMID: 39687136 PMCID: PMC11647948 DOI: 10.1016/j.heliyon.2024.e40616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Background Anastomotic leakage (AL) is a severe complication of colorectal surgery. The risk of AL is affected by both surgery and patient factors. Gut microbiomes can be generated from the residual material from the fecal immunochemical test (FIT). We, therefore, examined if AL after colorectal cancer surgery could be associated with specific baseline microbiomes in the FIT screening sampling tubes collected weeks before surgery. Methods Samples from patients participating in the Danish colorectal cancer screening program were biobanked from 2016 to 2018, and samples from patients who had surgery for screening-detected cancer were included. They were matched with patients without AL in a 1:2 ratio based on age, sex, location of anastomosis (colonic/rectal), ASA classification, and smoking habits. Bacterial DNA was extracted from the sampling tubes, and the fecal microbiomes were analyzed with targeted 16S ribosomal RNA third-generation sequencing. Results 18 patients who developed AL after surgery were matched with 36 without AL. The alpha diversity was lower in the AL group (p = 0.035), and the AL group separated from the Controls in the PCoA plot (p < 0.001). This was due to the patients undergoing rectal resections, with significant differences in alpha- and beta diversity (p = 0.025 and p = 0.002, respectively). The prevalence of bacteria with the potential to produce collagenase was higher in patients who developed AL (odds ratio 1.29 (95% CI 1.28-1.30), p < 0.001). Conclusions We found differences in the baseline microbiome profile associated with subsequent development of AL after surgery for screening-detected rectal cancer.
Collapse
Affiliation(s)
- Anders Bech Jørgensen
- Department of Surgery, Part of Copenhagen University Hospitals - Holbæk, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Louise Almer
- Center for Translational Research, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| | | | - Martin Schou Pedersen
- Department of Clinical Microbiology, Copenhagen University Hospital - Rigshospitalet, Denmark
| | - Nikolai Søren Kirkby
- Department of Clinical Microbiology, Copenhagen University Hospital - Rigshospitalet, Denmark
| | - Esther Agnete Jensen
- Department of Clinical Biochemistry, Part of Copenhagen University Hospitals - Næstved, Denmark
- The Secretariat for Colorectal Cancer Screening, Region Zealand, Næstved Hospital, Denmark
| | - Alonzo Alfaro-Núñez
- Department of Clinical Biochemistry, Part of Copenhagen University Hospitals - Næstved, Denmark
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Campus Copenhagen, A.C. Meyers Vænge 15, 2450 Copenhagen, Denmark
| | - Lennart Friis-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Denmark
| | - Birgitte Brandstrup
- Department of Surgery, Part of Copenhagen University Hospitals - Holbæk, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
38
|
García G, Soto J, Netherland M, Hasan NA, Buchaca E, Martínez D, Carlin M, de Jesus Cano R. Evaluating the Effects of Sugar Shift ® Symbiotic on Microbiome Composition and LPS Regulation: A Double-Blind, Placebo-Controlled Study. Microorganisms 2024; 12:2525. [PMID: 39770729 PMCID: PMC11678924 DOI: 10.3390/microorganisms12122525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
(1) Background: This study evaluated the effects of BiotiQuest® Sugar Shift®, a novel probiotic formulation, for its impact on gut microbiome composition and metabolic health in type 2 diabetes mellitus (T2D). T2D is characterized by chronic inflammation and gut microbiome imbalances, yet the therapeutic potential of targeted probiotics remains underexplored. (2) Methods: In a 12-week randomized, double-blind, placebo-controlled trial, 64 adults with T2D received either Sugar Shift or placebo capsules twice daily. Each dose provided 18 billion CFU of eight GRAS-certified bacterial strains and prebiotics. Clinical samples were analyzed for metabolic markers, and microbiome changes were assessed using 16S rRNA sequencing and metagenomics. (3) Results: Sugar Shift significantly reduced serum lipopolysaccharide (LPS) levels, improved insulin sensitivity (lower HOMA-IR scores), and increased short-chain fatty acid (SCFA)-producing genera, including Bifidobacterium, Faecalibacterium, Fusicatenibacter, and Roseburia. Pro-inflammatory taxa like Enterobacteriaceae decreased, with reduced LPS biosynthesis genes and increased SCFA production genes. The Lachnospiraceae:Enterobactericeae ratio emerged as a biomarker of reduced inflammation. (4) Conclusions: These findings demonstrate the potential of Sugar Shift to restore gut homeostasis, reduce inflammation, and improve metabolic health in T2D. Further studies are warranted to explore its long-term efficacy and broader application in metabolic disease management.
Collapse
Affiliation(s)
- Gissel García
- Pathology Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | - Josanne Soto
- Clinical Laboratory Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | | | - Nur A. Hasan
- EzBiome, 704 Quince Orchard Rd, Gaithersburg, MD 20878, USA (N.A.H.)
| | - Emilio Buchaca
- Internal Medicine Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | - Duniesky Martínez
- Research and Development Department, Center for Genetic Engineering and Biotechnology of Sancti Spíritus (CIGBSS), Circunvalante Norte S/N, Olivos 3, Apartado Postal 83, Sancti Spíritus 60200, Cuba;
| | - Martha Carlin
- The BioCollective, LLC, 4800 Dahlia Street, G8, Denver, CO 80216, USA;
| | - Raúl de Jesus Cano
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| |
Collapse
|
39
|
Voziki A, Deda O, Kachrimanidou M. The Efficacy of Fecal Microbiota Transplantation in Mouse Models Infected with Clostridioides difficile from the Perspective of Metabolic Profiling: A Systematic Review. Metabolites 2024; 14:677. [PMID: 39728458 DOI: 10.3390/metabo14120677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/07/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Objectives: This systematic review evaluates the effectiveness of fecal microbiota transplantation (FMT) in treating Clostridioides difficile infection (CDI) in mouse models using a metabolomics-based approach. Methods: A comprehensive search was conducted in three databases (PubMed, Scopus, Google Scholar) from 10 April 2024 to 17 June 2024. Out of the 460 research studies reviewed and subjected to exclusion criteria, only 5 studies met all the inclusion criteria and were analyzed. Results: These studies consistently showed that FMT effectively restored gut microbiota and altered metabolic profiles, particularly increasing short-chain fatty acids (SCFAs) and secondary bile acids, which inhibited C. difficile growth. FMT proved superior to antibiotic and probiotic treatments in re-establishing a healthy gut microbiome, as evidenced by significant changes in the amino acid and carbohydrate levels. Despite its promise, variability in the outcomes-due to factors such as immune status, treatment protocols, and donor microbiome differences-underscores the need for standardization. Rather than pursuing immediate standardization, the documentation of factors such as donor and recipient microbiome profiles, preparation methods, and administration details could help identify optimal configurations for specific contexts and patient needs. In all the studies, FMT was successful in restoring the metabolic profile in mice. Conclusions: These findings align with the clinical data from CDI patients, suggesting that FMT holds potential as a therapeutic strategy for gut health restoration and CDI management. Further studies could pave the way for adoption in clinical practice.
Collapse
Affiliation(s)
- Anna Voziki
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Olga Deda
- Laboratory of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Biomic AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi Rd., 57001 Thessaloniki, Greece
| | - Melania Kachrimanidou
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
40
|
Xin Z, Xin C, Huo J, Liu Q, Dong H, Li R, Liu Y. Neuroprotective Effect of a Multistrain Probiotic Mixture in SOD1 G93A Mice by Reducing SOD1 Aggregation and Targeting the Microbiota-Gut-Brain Axis. Mol Neurobiol 2024; 61:10051-10071. [PMID: 38349516 PMCID: PMC11584480 DOI: 10.1007/s12035-024-03988-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/25/2024] [Indexed: 11/24/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the selective loss of motor neurons. A bidirectional communication system known as the "microbiota-gut-brain" axis has a regulatory function in neurodegenerative disorders. The impact of probiotics on ALS through the "microbiota-gut-brain" axis remains uncertain. A longitudinal investigation was conducted to examine the alterations in the structure of the ileum and colon in mutant superoxide dismutase 1 (SOD1G93A) transgenic mice models of ALS by using immunofluorescence and Western blotting. Subsequently, the mice were administered a multistrain probiotic mixture (LBE) or vehicle orally, starting from 60 days of age until the terminal stage of the disease. The effects of these agents on the behavior, gut microbiota, microbial metabolites, and pathological processes of the spinal and intestine of SOD1G93A mice were analyzed, with a focus on exploring potential protective mechanisms. SOD1G93A mice exhibit various structural abnormalities in the intestine. Oral administration of LBE improved the proinflammatory response, reduced aberrant superoxide dismutase 1 (SOD1) aggregation, and protected neuronal cells in the intestine and spinal cord of SOD1G93A mice. Furthermore, LBE treatment resulted in a change in intestinal microbiota, an increase in short-chain fatty acid levels, and an enhancement in autophagy flux. SOD1G93A mice exhibited various structural abnormalities in the intestine. LBE can improve the proinflammatory response, reduce aberrant SOD1 aggregation, and protect neuronal cells in the spinal cord and intestine of SOD1G93A mice. The positive effect of LBE can be attributed to increased short-chain fatty acids and enhanced autophagy flux.
Collapse
Affiliation(s)
- Zikai Xin
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, People's Republic of China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Cheng Xin
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, People's Republic of China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Jia Huo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, People's Republic of China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Qi Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, People's Republic of China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Hui Dong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, People's Republic of China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Rui Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, People's Republic of China.
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China.
| | - Yaling Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, People's Republic of China.
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China.
| |
Collapse
|
41
|
Liu S, Dai X, Zhao J, Zhang X, Zhu M, Wang K, Fu D. Blended Tea Ameliorates T2DM via Modulation of Gut Microflora. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2024; 79:851-859. [PMID: 39153160 DOI: 10.1007/s11130-024-01222-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
Increasing evidences suggest that type 2 diabetes mellitus (T2DM) is closely related to gut microflora dysbiosis, which can be improved by dietary intervention. Four natural plant products, including Cyclocarya paliurus, Fu brick tea, Ampelopsis grossedentata, and Lithocarpus litseifolius, were blended to form a blended tea product for obtaining the better flavor. The blended tea was also expected to have excellent pharmacological activity. Therefore, the ameliorative effect of blended tea on T2DM and underlying mechanisms were studied in this study. The results showed that the blended tea extract effectively attenuated the symptoms of glucose and lipid metabolism-related disorders in T2DM mice fed by high-fat and high-sucrose diet. Furthermore, blended tea extract intervention significantly attenuated gut microbiota dysbiosis, the abundance of bacteria such as Bacteroidetes and Firmicutes, which aid in the hydrolysis and utilization of carbohydrates, significantly increased, while the abundance of pathogenic bacteria such as Proteobacteria significantly decreased. Certain core microorganisms involved in energy metabolism, including Ruminococcaceae_UCG-005, Butyricimonas, Roseburia, Oscillibacter, [Eubacterium]_nodatum_group, Muribaculaceae, Prevotellaceae UCG 001, were also found to be improved by blended tea extract. Collectively, our results demonstrated that the blended tea may ameliorate T2DM through modulation of gut microflora. The blended tea may serve as novel functional drink for the treatment of T2DM and dysbiosis of gut microbiota.
Collapse
Affiliation(s)
- Sui Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China
| | - Xinyue Dai
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China
| | - Jinqi Zhao
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China
| | - Xuwen Zhang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China
| | - Mingzhi Zhu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China.
- National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, Hunan, 410128, China.
| | - Kunbo Wang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China.
- National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, Hunan, 410128, China.
| | - Donghe Fu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan, China.
- National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, Hunan, 410128, China.
| |
Collapse
|
42
|
Zhang Y, Zhao T, Zhang Y, Song Q, Meng Q, Zhou S, Wei L, Qi Y, Guo Y, Cong J. Accumulation and depuration of tire wear particles in zebrafish (Danio rerio) and toxic effects on gill, liver, and gut. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175625. [PMID: 39163933 DOI: 10.1016/j.scitotenv.2024.175625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/15/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
The toxic effects of tire wear particles (TWPs) in the environment are a growing concern for a variety of aquatic organisms. However, studies about TWPs toxicity on aquatic organisms are limited. This study investigated the accumulation and depuration of TWPs in zebrafish at three different concentrations (5 mg/L, 10 mg/L, and 20 mg/L), as well as the toxic effects on the gill, liver, and gut. We found that TWPs could accumulate in the gill and gut for a long time, and the number of TWPs at the high-concentration (20 mg/L) was higher than at the low-concentration (5 mg/L). TWPs induced oxidative stress in the gill and liver. The liver transcriptome profiles indicated that the high concentration of TWPs tended to up-regulate metabolic processes, whereas the low concentration of TWPs was inclined to down-regulate cellular processes. The high-concentration treatment significantly increased xenobiotic biodegradation and metabolism, and lipid metabolism-related pathways, whereas the low-concentration treatment distinctly altered amino acid metabolism-related pathways. The expression of gstt1b, ugt1a1, mgst3b, miox, hsd17b3, and cyp8b1 gene was up-regulated in all TWPs treatments. In addition, Gemmobacter and Shinella enriched in the high-concentration treatment were closely correlated with the degradation of TWPs. These findings provided objective evidence for the toxicity evaluation of TWPs on zebrafish.
Collapse
Affiliation(s)
- Yun Zhang
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Tianyu Zhao
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Yanan Zhang
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266000, China
| | - Qianqian Song
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Qingxuan Meng
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Siyu Zhou
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Lijuan Wei
- School of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Yinuo Qi
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Yinyuan Guo
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Jing Cong
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China.
| |
Collapse
|
43
|
Lee JY, Bays DJ, Savage HP, Bäumler AJ. The human gut microbiome in health and disease: time for a new chapter? Infect Immun 2024; 92:e0030224. [PMID: 39347570 PMCID: PMC11556149 DOI: 10.1128/iai.00302-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024] Open
Abstract
The gut microbiome, composed of the colonic microbiota and their host environment, is important for many aspects of human health. A gut microbiome imbalance (gut dysbiosis) is associated with major causes of human morbidity and mortality. Despite the central part our gut microbiome plays in health and disease, mechanisms that maintain homeostasis and properties that demarcate dysbiosis remain largely undefined. Here we discuss that sorting taxa into meaningful ecological units reveals that the availability of respiratory electron acceptors, such as oxygen, in the host environment has a dominant influence on gut microbiome health. During homeostasis, host functions that limit the diffusion of oxygen into the colonic lumen shelter a microbial community dominated by primary fermenters from atmospheric oxygen. In turn, primary fermenters break down unabsorbed nutrients into fermentation products that support host nutrition. This symbiotic relationship is disrupted when host functions that limit the luminal availability of host-derived electron acceptors become weakened. The resulting changes in the host environment drive alterations in the microbiota composition, which feature an elevated abundance of facultatively anaerobic microbes. Thus, the part of the gut microbiome that becomes imbalanced during dysbiosis is the host environment, whereas changes in the microbiota composition are secondary to this underlying cause. This shift in our understanding of dysbiosis provides a novel starting point for therapeutic strategies to restore microbiome health. Such strategies can either target the microbes through metabolism-based editing or strengthen the host functions that control their environment.
Collapse
Affiliation(s)
- Jee-Yon Lee
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
| | - Derek J. Bays
- Department of Internal Medicine, Division of Infectious Diseases, School of Medicine, University of California Davis, Sacramento, California, USA
| | - Hannah P. Savage
- Department of Pathology Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
| |
Collapse
|
44
|
Wang J, Du J, Gou X, Huang Y, He J, Lu X, Xie M. Propyl acetate protects intestinal barrier during parenteral nutrition in mice and Caco-2 cells. JPEN J Parenter Enteral Nutr 2024; 48:917-926. [PMID: 39187914 DOI: 10.1002/jpen.2681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Gut microbiota dysbiosis induces intestinal barrier damage during parenteral nutrition (PN). However, the underlying mechanisms remain unclear. This study aimed to investigate gut microbiota dysbiosis, luminal short-chain fatty acids, and autophagy in a mouse model and how these short-chain fatty acids regulate autophagy. METHODS Eight-week-old male specific-pathogen-free mice were randomly divided into a Chow group (standard diet and intravenous normal saline infusion) and a PN group (continuous infusion of PN nutrient solution) for 7 days. Caco-2 cells were also treated with intestinal rinse solutions from Chow and PN mouse models. RESULTS Compared with the Chow group, the PN group exhibited increased Proteobacteria and decreased Firmicutes, correlating with decreased propyl acetate. In the PN group, intestinal tissue exhibited elevated adenosine monophosphate-activated protein kinase (AMPK) phosphorylation, LC3II protein levels, and Atg3 and Atg7 messenger RNA levels. P62 protein levels were decreased, indicating an increase of autophagy flux in the PN group. In the Caco-2 cell model, cells treated with PN solution plus propyl acetate exhibited increased Claudin-1 and occluding along with decreased interleukin-6 and tumor necrosis factor α compared with those treated with PN solution alone. Propyl acetate addition inhibited the AMPK-mammalian target of rapamycin (mTOR) pathway, mitigating the excessive autophagy induced by the PN intestinal rinse solution in Caco-2 cells. CONCLUSION PN led to a significant reduction in propyl acetate levels in the intestine, excessive activation of autophagy, and barrier dysfunction. Propyl acetate inhibited excessive autophagy via the AMPK/mTOR signaling pathway and protected the intestinal barrier during PN.
Collapse
Affiliation(s)
- Jiwei Wang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jing Du
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaomei Gou
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yong Huang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jixin He
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaoyun Lu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ming Xie
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
45
|
Jessop E, Renaud DL, Verbrugghe A, Obregon D, Macnicol J, McMahon A, Li L, Gamsjäger L, Gomez DE. Fecal microbiota of diarrheic calves: Before, during, and after recovering from disease. J Vet Intern Med 2024; 38:3358-3366. [PMID: 39340403 PMCID: PMC11586557 DOI: 10.1111/jvim.17201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND It is unknown if gastrointestinal dysbiosis in diarrheic calves causes disease or is a consequence of the disease. OBJECTIVES Describe the fecal microbiota of calves before, during, and after recovering from diarrhea. ANIMALS Fifteen female Holstein calves of 0 to 21 days old from a single farm. Seven calves remained healthy throughout the study, and 8 developed diarrhea on Day 14. METHODS Longitudinal cohort study. Microbiota composition was characterized by amplifying the V4 region of the 16S rRNA gene. RESULTS Diversity (Shannon index) increased with age in healthy and diarrheic calves from Day 3 to 21, but diarrheic calves had a lower diversity on the day diarrhea was first observed (Day 14). By Day 21, diversity increased in calves that recovered from diarrhea and was not significantly different from that of their healthy counterparts (P > .05). Weighted UniFrac distance showed significant differences in the fecal microbiota between diarrheic and healthy calves at Day 14 of age (PERMANOVA, P < .05), but not before or after diarrhea (PERMANOVA, P > .05). Lactobacillus, Clostridium Sensu Stricto 1, and Collinsella were differentially abundant on Day 10 in calves that developed diarrhea on Day 14 (P < .05). CONCLUSION AND CLINICAL IMPORTANCE The fecal microbiota of healthy and diarrheic calves evolved similarly during the first 10 days of age but differed significantly on the day of onset of diarrhea. Enriching Lactobacillus, Clostridium Sensu Stricto 1, and Collinsella before diarrhea onset could have been contributed to the development of diarrhea.
Collapse
Affiliation(s)
- Emma Jessop
- Department of Clinical StudiesUniversity of Guelph, Ontario Veterinary CollegeGuelphOntarioCanada
| | - David L. Renaud
- Department of Population MedicineUniversity of Guelph, Ontario Veterinary CollegeGuelphOntarioCanada
| | - Adronie Verbrugghe
- Department of Clinical StudiesUniversity of Guelph, Ontario Veterinary CollegeGuelphOntarioCanada
| | - Dasiel Obregon
- School of Environmental SciencesUniversity of GuelphGuelphOntarioCanada
| | - Jenniffer Macnicol
- Department of PathobiologyUniversity of Guelph, Ontario Veterinary CollegeGuelphOntarioCanada
| | - Aoife McMahon
- Department of Clinical StudiesUniversity of Guelph, Ontario Veterinary CollegeGuelphOntarioCanada
| | - Lynna Li
- Department of Clinical StudiesUniversity of Guelph, Ontario Veterinary CollegeGuelphOntarioCanada
| | - Lisa Gamsjäger
- Department of Population Health and PathobiologyCollege of Veterinary Medicine, North Carolina State UniversityRaleighNorth CarolinaUSA
| | - Diego E. Gomez
- Department of Clinical StudiesUniversity of Guelph, Ontario Veterinary CollegeGuelphOntarioCanada
| |
Collapse
|
46
|
Varghese S, Rao S, Khattak A, Zamir F, Chaari A. Physical Exercise and the Gut Microbiome: A Bidirectional Relationship Influencing Health and Performance. Nutrients 2024; 16:3663. [PMID: 39519496 PMCID: PMC11547208 DOI: 10.3390/nu16213663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: The human gut microbiome is a complex ecosystem of microorganisms that can influence our health and exercise habits. On the other hand, physical exercise can also impact our microbiome, affecting our health. Our narrative review examines the bidirectional relationship between physical activity and the gut microbiome, as well as the potential for targeted probiotic regimens to enhance sports performance. Methods: We conducted a comprehensive literature review to select articles published up till January 2024 on the topics of physical exercise, sports, probiotics, and gut microbiota from major scientific databases, incorporating over 100 studies. Results: We found that the impact of physical activity on the gut microbiome varies with the type and intensity of exercise. Moderate exercise promotes a healthy immune system, while high-intensity exercise for a long duration can cause a leaky gut and consequent systemic inflammation, which may disrupt the microbial balance. Combining aerobic and resistance training significantly affects bacterial diversity, linked to a lower prevalence of chronic metabolic disorders. Furthermore, exercise enhances gut microbiome diversity, increases SCFA production, improves nutrient utilization, and modulates neural and hormonal pathways, improving gut barrier integrity. Our findings also showed probiotic supplementation is associated with decreased inflammation, enhanced sports performance, and fewer gastrointestinal disturbances, suggesting that the relationship between the gut microbiome and physical activity is mutually influential. Conclusions: The bidirectional relationship between physical activity and the gut microbiome is exemplified by how exercise can promote beneficial bacteria while a healthy gut microbiome can potentially enhance exercise ability through various mechanisms. These findings underscore the importance of adding potential tailored exercise regimens and probiotic supplementation that consider individual microbiome profiles into exercise programs.
Collapse
Affiliation(s)
| | | | | | | | - Ali Chaari
- Department of Biochemistry, Premedical Division, Weill Cornell Medicine–Qatar, Qatar Foundation, Education City, Doha P.O. Box 24144, Qatar; (S.V.); (S.R.); (A.K.); (F.Z.)
| |
Collapse
|
47
|
Zhang Z, Yang Y, Zhang Y, Xie G. Co-frequency or contrary? The effects of Qiwei Baizhu Powder and its bioactive compounds on mucosa-associated microbiota of mice with antibiotic-associated diarrhea. Front Cell Infect Microbiol 2024; 14:1483048. [PMID: 39529635 PMCID: PMC11551125 DOI: 10.3389/fcimb.2024.1483048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Qiwei Baizhu Powder (QWBZP) has been proven effective in treating antibiotic-associated diarrhea (AAD), and the mechanism is associated with regulating the gut microbiota. However, the role of the bioactive compounds of QWBZP in regulating the gut microbiota is still unclear. In this study, 24 mice were divided into a normal control group (N), a model group (R), a QWBZP decoction group (TW), and a QWBZP-TG group (TG). AAD mouse models were established by mixed antibiotic administration. After modeling, mice in the TW group and TG group were treated with QWBZP decoction and QWBZP-TG, respectively. Mice in the N group and R group were gavaged with sterile water. 16S rRNA gene sequencing was used to investigate the changes of mucosa-associated microbiota (MAM) in the small intestine of mice. Moreover, the levels of diamine oxidase (DAO), D-Lactate, secretory immunoglobulin A (sIgA), interleukin 6 (IL-6), IL-10, and tumor necrosis factor-α (TNF-α) were detected using enzyme-linked immunosorbent assay (ELISA) kits. The results showed that QWBZP-TG significantly altered the diversity, structure, and abundance of MAM in the AAD mice. QWBZP-TG exerted a stronger suppression effect on Escherichia and Clostridium compared with QWBZP decoction. Meanwhile, QWBZP-TG downregulated the abundance of Lactobacillus, which elicited an opposite effect to QWBZP decoction. Prevotella was the signature bacteria that responded to the QWBZP-TG intervention. Furthermore, both QWBZP decoction and QWBZP-TG decreased the levels of DAO, D-Lactate, sIgA, IL-6, and TNF-α in the AAD mice. The role of glycosides is to help QWBZP ameliorate diarrhea symptoms by inhibiting the proliferation of diarrhea-associated bacteria, reducing inflammation and regulating immunity.
Collapse
|
48
|
Caruso R, Lo BC, Chen GY, Núñez G. Host-pathobiont interactions in Crohn's disease. Nat Rev Gastroenterol Hepatol 2024:10.1038/s41575-024-00997-y. [PMID: 39448837 DOI: 10.1038/s41575-024-00997-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
The mammalian intestine is colonized by trillions of microorganisms that are collectively referred to as the gut microbiota. The majority of symbionts have co-evolved with their host in a mutualistic relationship that benefits both. Under certain conditions, such as in Crohn's disease, a subtype of inflammatory bowel disease, some symbionts bloom to cause disease in genetically susceptible hosts. Although the identity and function of disease-causing microorganisms or pathobionts in Crohn's disease remain largely unknown, mounting evidence from animal models suggests that pathobionts triggering Crohn's disease-like colitis inhabit certain niches and penetrate the intestinal tissue to trigger inflammation. In this Review, we discuss the distinct niches occupied by intestinal symbionts and the evidence that pathobionts triggering Crohn's disease live in the mucus layer or near the intestinal epithelium. We also discuss how Crohn's disease-associated mutations in the host disrupt intestinal homeostasis by promoting the penetration and accumulation of pathobionts in the intestinal tissue. Finally, we discuss the potential role of microbiome-based interventions in precision therapeutic strategies for the treatment of Crohn's disease.
Collapse
Affiliation(s)
- Roberta Caruso
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Bernard C Lo
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Grace Y Chen
- Department of Internal Medicine and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Caulat LC, Lotoux A, Martins MC, Kint N, Anjou C, Teixeira M, Folgosa F, Morvan C, Martin-Verstraete I. Physiological role and complex regulation of O 2-reducing enzymes in the obligate anaerobe Clostridioides difficile. mBio 2024; 15:e0159124. [PMID: 39189748 PMCID: PMC11481553 DOI: 10.1128/mbio.01591-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/22/2024] [Indexed: 08/28/2024] Open
Abstract
Clostridioides difficile, the major cause of antibiotic-associated diarrhea, is a strict anaerobic, sporulating Firmicutes. However, during its infectious cycle, this anaerobe is exposed to low oxygen (O2) tensions, with a longitudinal decreasing gradient along the gastrointestinal tract and a second lateral gradient with higher O2 tensions in the vicinity of the cells. A plethora of enzymes involved in oxidative stress detoxication has been identified in C. difficile, including four O2-reducing enzymes: two flavodiiron proteins (FdpA and FdpF) and two reverse rubrerythrins (revRbr1 and revRbr2). Here, we investigated the role of the four O2-reducing enzymes in the tolerance to increasing physiological O2 tensions and air. The four enzymes have different, yet overlapping, spectra of activity. revRbr2 is specific to low O2 tensions (<0.4%), FdpA to low and intermediate O2 tensions (0.4%-1%), revRbr1 has a wider spectrum of activity (0.1%-4%), and finally FdpF is more specific to tensions > 4% and air. These different O2 ranges of action partly arise from differences in regulation of expression of the genes encoding those enzymes. Indeed, we showed that revrbr2 is under the dual control of σA and σB. We also identified a regulator of the Spx family that plays a role in the induction of fdp and revrbr genes upon O2 exposure. Finally, fdpF is regulated by Rex, a regulator sensing the NADH/NAD+ ratio. Our results demonstrate that the multiplicity of O2-reducing enzymes of C. difficile is associated with different roles depending on the environmental conditions, stemming from a complex multi-leveled network of regulation. IMPORTANCE The gastrointestinal tract is a hypoxic environment, with the existence of two gradients of O2 along the gut, one longitudinal anteroposterior decreasing gradient and one proximodistal increasing from the lumen to the epithelial cells. O2 is a major source of stress for an obligate anaerobe such as the enteropathogen C. difficile. This bacterium possesses a plethora of enzymes capable of scavenging O2 and reducing it to H2O. In this work, we identified the role of the four O2-reducing enzymes in the tolerance to the physiological O2 tensions faced by C. difficile during its infectious cycle. These four enzymes have different spectra of action and protect the vegetative cells over a large range of O2 tensions. These differences are associated with a distinct regulation of each gene encoding those enzymes. The complex network of regulation is crucial for C. difficile to adapt to the various O2 tensions encountered during infection.
Collapse
Affiliation(s)
- Léo C. Caulat
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Aurélie Lotoux
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Maria C. Martins
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Nicolas Kint
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Cyril Anjou
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Miguel Teixeira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Filipe Folgosa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Claire Morvan
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Isabelle Martin-Verstraete
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
50
|
Castro AM, Sabater C, Gutiérrez-Díaz I, Navarro S, Rodriguez S, Molinos C, Jiménez S, Claver A, Espin B, Domínguez G, Coronel C, Toyos P, Sariego L, Fernández P, Perez D, Margolles A, Díaz JJ, Delgado S. The intestinal microbiome of infants with cow's milk-induced FPIES is enriched in taxa and genes of enterobacteria. J Pediatr Gastroenterol Nutr 2024; 79:841-849. [PMID: 39175183 DOI: 10.1002/jpn3.12356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024]
Abstract
OBJECTIVES Food protein-induced enterocolitis syndrome (FPIES) is a severe type of non-IgE (immunoglobulin E)-mediated (NIM) food allergy, with cow's milk (CM) being the most common offending food. The relationship between the gut microbiota and its metabolites with the inflammatory process in infants with CM FPIES is unknown, although evidence suggests a microbial dysbiosis in NIM patients. This study was performed to contribute to the knowledge of the interaction between the gut microbiota and its derived metabolites with the local immune system in feces of infants with CM FPIES at diagnosis. METHODS Twelve infants with CM FPIES and a matched healthy control group were recruited and the gut microbiota was investigated by 16S amplicon and shotgun sequencing. Fatty acids (FAs) were measured by gas chromatography, while immune factors were determined by enzyme-linked immunosorbent assay and Luminex technology. RESULTS A specific pattern of microbiota in the gut of CM FPIES patients was found, characterized by a high abundance of enterobacteria. Also, an intense excretion of FAs in the feces of these infants was observed. Furthermore, correlations were found between fecal bifidobacteria and immune factors. CONCLUSION These fecal determinations may be useful to gain insight into the pathophysiology of this syndrome and should be taken in consideration for future studies of FPIES patients.
Collapse
Affiliation(s)
- Ana M Castro
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Carlos Sabater
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Isabel Gutiérrez-Díaz
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Sandra Navarro
- Primary Care Center Teatinos-Corredoria, Oviedo, Asturias, Spain
| | - Silvia Rodriguez
- Paediatrics Service, Hospital Universitario de San Agustín, Avilés, Asturias, Spain
| | - Cristina Molinos
- Paediatrics Department, Hospital Universitario de Cabueñes, Gijón, Asturias, Spain
| | | | - Angela Claver
- Allergology, Hospital Universitario Dexeus, Barcelona, Spain
| | - Beatriz Espin
- Paediatric Gastroenterology Unit, Hospital Universitario Virgen del Rocío de Sevilla, Sevilla, Spain
| | - Gloria Domínguez
- Gastroenterology and Nutrition Section, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | | | - Paula Toyos
- Paediatric Group, ISPA, Oviedo, Asturias, Spain
| | - Lydia Sariego
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | | | - David Perez
- Paediatrics Service, Hospital Universitario de San Agustín, Avilés, Asturias, Spain
| | - Abelardo Margolles
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| | - Juan J Díaz
- Paediatric Group, ISPA, Oviedo, Asturias, Spain
| | - Susana Delgado
- MicroHealth Group, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas (IPLA-CSIC)/Instituto Biosanitario del Principado de Asturias (ISPA), Villaviciosa, Asturias, Spain
| |
Collapse
|