1
|
Zhu F, Ying H, Siadat SD, Fateh A. The gut-lung axis and microbiome dysbiosis in non-tuberculous mycobacterial infections: immune mechanisms, clinical implications, and therapeutic frontiers. Gut Pathog 2025; 17:40. [PMID: 40481550 PMCID: PMC12144820 DOI: 10.1186/s13099-025-00718-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/30/2025] [Indexed: 06/11/2025] Open
Abstract
Non-tuberculous mycobacteria (NTM) are emerging pathogens of global concern, particularly in regions with declining tuberculosis rates. This review synthesizes current evidence on the epidemiology, immune pathogenesis, and microbiome interactions underlying NTM infections. The rising incidence of NTM is driven by environmental factors, immunocompromised populations, and advanced diagnostics. Clinically, NTM manifests as pulmonary, lymphatic, skin/soft tissue, or disseminated disease, with Mycobacterium avium complex (MAC) and M. abscessus being predominant pathogens. Host immunity, particularly Th1 responses mediated by IL-12/IFN-γ and TLR2 signaling, is critical for controlling NTM, while dysregulated immunity (e.g., elevated Th2 cytokines, PD-1/IL-10 pathways) exacerbates susceptibility. Emerging research highlights the gut-lung axis as a pivotal mediator of disease, where microbiome dysbiosis-marked by reduced Prevotella and Bifidobacterium-impairs systemic immunity and promotes NTM progression. Short-chain fatty acids (SCFAs) and microbial metabolites like inosine modulate macrophage and T-cell responses, offering therapeutic potential. Studies reveal distinct airway microbiome signatures in NTM patients, characterized by enriched Streptococcus and Prevotella, and reduced diversity linked to worse outcomes. Despite advances, treatment remains challenging due to biofilm formation, antibiotic resistance, and relapse rates. This review underscores the need for microbiome-targeted therapies, personalized medicine, and longitudinal studies to unravel causal relationships between microbial ecology and NTM pathogenesis.
Collapse
Affiliation(s)
- Fangfang Zhu
- Pinghu Traditional Chinese Medicine Hospital, Pinghu, Zhejiang, 314200, China
| | - Hao Ying
- Zhuji People´s Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang, 311800, China.
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
Lindsell HB, Williams NC, Magistro D, Corsetti M, Walton GE, Hunter KA. Could the Therapeutic Effect of Physical Activity on Irritable Bowel Syndrome Be Mediated Through Changes to the Gut Microbiome? A Narrative and Hypothesis Generating Review. Neurogastroenterol Motil 2025; 37:e70004. [PMID: 40026117 PMCID: PMC12075915 DOI: 10.1111/nmo.70004] [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: 10/25/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is one of the most prevalent gastrointestinal (GI) disorders worldwide. Defined as a disorder of gut-brain interaction, its pathophysiology is still not completely clear. Consequently, current treatments primarily target symptoms rather than addressing the cause of the condition. The gut microbiome is increasingly acknowledged as central to IBS pathophysiology and, thus, may have therapeutic potential. Several national treatment guidelines recommend increasing physical activity for IBS management. AIMS This review summarises the evidence about the relationship between physical activity, IBS symptoms, and the gut microbiome, investigating the hypothesis that physical activity's therapeutic effects on IBS may be explained via modulation of the gut microbiome. RESULTS This review revealed that routine exercise was associated with a 15%-66% reduction in symptom severity and up to 41% enhanced QoL in IBS participants, and modulates the gut microbiome in healthy controls. DISCUSSION This review generates the hypothesis that routine physical activity may favorably alter gut microbiome composition in IBS to improve IBS symptomology. While a plausible hypothesis, research needs to confirm whether gut microbiome modulation is involved in physical activity associated IBS symptom relief. CONCLUSION Furthermore, the establishment of the most effective mode, duration, and intensity of physical activity for each sex and IBS-subtype is needed, with patient input during this process crucial to successfully translate science into practice.
Collapse
Affiliation(s)
- Hannah B. Lindsell
- Department of Sport, Health and Performance Enhancement (SHAPE) Research Centre, Department of Sport ScienceNottingham Trent UniversityNottinghamUK
| | - Neil C. Williams
- Department of Sport, Health and Performance Enhancement (SHAPE) Research Centre, Department of Sport ScienceNottingham Trent UniversityNottinghamUK
| | - Daniele Magistro
- Department of Sport, Health and Performance Enhancement (SHAPE) Research Centre, Department of Sport ScienceNottingham Trent UniversityNottinghamUK
| | - Maura Corsetti
- NIHR Nottingham Biomedical Research CentreNottingham University Hospitals NHS Trust UK, School of MedicineNottinghamUK
| | - Gemma E. Walton
- Department of Food and Nutritional SciencesThe University of ReadingReadingUK
| | - Kirsty A. Hunter
- Department of Sport, Health and Performance Enhancement (SHAPE) Research Centre, Department of Sport ScienceNottingham Trent UniversityNottinghamUK
| |
Collapse
|
3
|
Saini S, Sabaeifard P, Coughlin L, Poulides N, Gan S, Zhan X, Dang M, Koh AY, Zia A. Identifying Microbiota and Immune Host Factors Associated With Bleeding Risk in Children With Immune Thrombocytopenia. Am J Hematol 2025; 100:1090-1093. [PMID: 40110651 DOI: 10.1002/ajh.27669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Affiliation(s)
- Shelly Saini
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Parastoo Sabaeifard
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Laura Coughlin
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nicole Poulides
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shuheng Gan
- Department of Population and Data Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiaowei Zhan
- Department of Population and Data Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mary Dang
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Andrew Y Koh
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ayesha Zia
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
4
|
Klak K, Maciuszek M, Michalik A, Mazur M, Zawisza M, Pecio A, Nowak B, Chadzinska M. Fire in the belly: Stress and antibiotics induce dysbiosis and inflammation in the gut of common carp. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110301. [PMID: 40157582 DOI: 10.1016/j.fsi.2025.110301] [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: 09/27/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Fish are exposed to numerous stressors which negatively affect their immune response and increase infection susceptibility. The risk of bacterial infections results in the excessive and preventive use of antibiotics. Therefore, we aimed to study how antibiotic treatment and restraint stress will affect the stress response, microbiota composition, gut morphology, and inflammatory reaction in common carp. Both restraint stress and antibiotic treatment increased cortisol level. Moreover, antibiotics induced dysbiosis in fish gut, manifested by a decrease in the total abundance of bacteria, and a shift in bacteria diversity, including a reduced number of Aeromonas, Bacteroides, Barnesiellaceae, Cetobacterium and Shewanella and an increased abundance of Flavobacterium. To a lesser extent, stress modified gut microbiota, as it decreased bacteria number and slightly changed the microbiota composition by decreasing Cetobacterium abundance and increasing Vibrio abundance. Microbiota of the antibiotic-treated and stressed fish shifted from the beneficial bacterial genera - Cetobacterium and Bacteroides, to the increased presence of unfavorable bacteria such as Brevinema, Flavobacterium and Desulfovibrionaceae. Stress and antibiotic-induced changes in the gut microbiota were related to the changes in the gut morphology when the higher abundance of goblet and rodlet cells and increased secretion activity of goblet cells were observed. Moreover, up-regulation of the expression of genes encoding pro-inflammatory mediators and cytokines involved in the Th17 immune response was present in the gut of the antibiotic-treated and stressed fish. We conclude that in carp antibiotics and stress alter the abundance and composition of the microbiota and induce Th17-dependent inflammatory reaction in the gut. Moreover, our results strongly suggest the interplay of the stress axis and the brain-gut-microbiota axis.
Collapse
Affiliation(s)
- Katarzyna Klak
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Magdalena Maciuszek
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Anna Michalik
- Department of Invertebrate Development and Morphology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Mikolaj Mazur
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Maria Zawisza
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland.
| | - Anna Pecio
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| | - Barbara Nowak
- Institute for Marine and Antarctic Studies - Launceston, University of Tasmania, Launceston, Tasmania, Australia.
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
5
|
Cai J, Zhang W, Zhu S, Lin T, Mao R, Wu N, Zhang P, Kang M. Gut and Intratumoral microbiota: Key to lung Cancer development and immunotherapy. Int Immunopharmacol 2025; 156:114677. [PMID: 40279944 DOI: 10.1016/j.intimp.2025.114677] [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: 02/03/2025] [Revised: 03/25/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
Lung cancer is a common malignant tumor worldwide with high incidence and mortality rates. Previous studies have claimed that the microbial community plays an integral role in the development and progression of lung cancer. Emerging evidence reveals that gut flora plays a key role in cancer formation and evolution by participating in mechanisms such as metabolism, regulation of inflammation and immune response. Not only the gut flora, but also the presence of intratumoral microbiota may influence lung cancer progression through multiple mechanisms. These research advances suggest that intestinal flora and intratumoral microbiota may not only serve as potential biomarkers for lung cancer, but may also be targets for therapy. However, current studies on both in lung cancer are still limited. Given this, this study aimed to systematically review the latest findings on the major bacterial species of the intestinal flora and their possible protective or harmful roles in the formation, progression, and metastasis of lung cancer. In addition, we analyzed the potential mechanisms by which the intratumoral microbiota affected lung cancer and elaborated on the potential roles of the gut flora and its metabolites, as well as the intratumoral microbiota, in modulating the efficacy of immunotherapy in lung cancer.
Collapse
Affiliation(s)
- Junlan Cai
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Weiguang Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Shujing Zhu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Tianxin Lin
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Renyan Mao
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ningzi Wu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China; Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China.
| |
Collapse
|
6
|
Belloni S, Arrigoni C, Magon A, Giacon C, Ceruso MH, Arcidiacono MA, Conte G, Caruso R. Symptomatologic outcomes of gut microbiota modifiers (probiotics, prebiotics and synbiotics) in cancer care: A scoping review of randomized controlled trials. Crit Rev Oncol Hematol 2025; 212:104779. [PMID: 40412575 DOI: 10.1016/j.critrevonc.2025.104779] [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: 06/10/2024] [Revised: 05/19/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Microbiota modifiers offer potential benefits for improving the wide spectrum of symptoms and clinical outcomes in individuals with cancer. However, there is a lack of comprehensive literature mapping to determine which specific cancer and treatment-related symptoms have been investigated as potential targets for gut microbiota modifiers. This scoping review aims to systematically analyze clinical trials on microbiota modifiers in managing cancer and treatment-related symptoms in adults. METHODS We conducted a scoping review of randomized controlled trials (RCTs) across four databases up to May 2025, following our published protocol and JBI principles with PRISMA 2020 guidelines. RESULTS The literature review identified 33 eligible studies, primarily involving patients with pelvic cancers. The most common outcomes examined in the clinical trials were gastrointestinal symptoms. Other studies focused on patients with head, neck, and breast cancer, examining quality of life, mucositis, fatigue, anxiety, depression, and the use of rescue drugs. CONCLUSION Despite evidence of potential benefits for gastrointestinal symptoms, inconsistent findings across studies warrant further well-designed, large-scale research to understand probiotics' effectiveness and mechanisms.
Collapse
Affiliation(s)
- Silvia Belloni
- Department of Public Health, Experimental and Forensic Medicine, Section of Hygiene, University of Pavia, Pavia, Italy.
| | - Cristina Arrigoni
- Department of Public Health, Experimental and Forensic Medicine, Section of Hygiene, University of Pavia, Pavia, Italy.
| | - Arianna Magon
- Health Professions Research and Development Unit, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Chiara Giacon
- Haematology Unit, Fondazione I.R.C.C.S. Policlinico San Matteo, Pavia, Italy
| | | | | | - Gianluca Conte
- Health Professions Research and Development Unit, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Rosario Caruso
- Health Professions Research and Development Unit, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy; Department of Biomedical Sciences for Health, University of Milan, Italy
| |
Collapse
|
7
|
Jia L, Yuan J, Chen Y, Liang P, Wu J, Xie Y, Bai X, Wang Y, Geng W. Lactobacillus kefiranofaciens and its enhancement effect on the anti-inflammatory function of CII/Gln in MIA-induced osteoarthritis by protecting the intestinal barrier and gut microecology. Int J Food Sci Nutr 2025:1-14. [PMID: 40401730 DOI: 10.1080/09637486.2025.2508173] [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: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Osteoarthritis (OA) is a globally chronic disease affecting middle-aged and elderly individuals, with growing evidences implicating gut microbiota in its pathogenesis. Lactobacillus kefiranofaciens ZW3 (ZW3) shows potential in modulating gut microbiota, protecting intestinal barrier, and regulating immunity. This study explored the therapeutic effects of ZW3, alone and combined with CII/Gln, using a monoiodoacetate (MIA)-induced OA model. Results indicated that ZW3 significantly mitigated cartilage damage and inflammation alone or combined with CII/Gln, possibly by improving intestinal integrity, reduced oxidative stress, and regulated MMPs expression. 16S rDNA sequencing showed ZW3, especially with CII/Gln, increased beneficial bacteria of Ruminococcaceae and Lachnospiraceae abundances. Furthermore, ZW3 or with CII/Gln elevated SCFAs levels in intestinal contents in OA rats. These findings propose a novel probiotic-based strategy, potentially combined with functional foods, for OA intervention and treatment.
Collapse
Affiliation(s)
- Longgang Jia
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Jiahu Yuan
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Yu Chen
- Binhai New Area Hospital of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Peixin Liang
- Tangshan Normal University, Tangshan, Hebei, P. R. China
| | - Jiangtao Wu
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Yufeng Xie
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
- College of Food Science and Engineering, Harbin University, Harbin, P. R. China
| | - Xiaojia Bai
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Yanping Wang
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Weitao Geng
- Engineering Research Center of Food Biotechnology, Ministry of Education, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| |
Collapse
|
8
|
Roman-Pepine D, Serban AM, Capras RD, Cismaru CM, Filip AG. A Comprehensive Review: Unraveling the Role of Inflammation in the Etiology of Heart Failure. Heart Fail Rev 2025:10.1007/s10741-025-10519-w. [PMID: 40360833 DOI: 10.1007/s10741-025-10519-w] [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] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
Heart failure (HF) remains a leading cause of morbidity and mortality worldwide, with inflammation playing a pivotal role in its pathogenesis. This comprehensive review aims to elucidate the intricate mechanisms by which inflammation contributes to the development and progression of HF. The review synthesizes current research on the involvement of both innate and adaptive immune responses in HF, highlighting the roles of cytokines, chemokines, and other inflammatory mediators. Recent studies have demonstrated that chronic inflammation, driven by factors such as oxidative stress, neurohormonal activation, and metabolic disturbances, leads to adverse cardiac remodeling and impaired myocardial function. The review explores how systemic inflammation, characterized by elevated levels of inflammatory biomarkers like C-reactive protein (CRP) and interleukin-6 (IL-6), correlates with HF severity and outcomes. Additionally, it discusses the impact of comorbid conditions such as diabetes, obesity, and hypertension on inflammatory pathways and HF risk. The review also delves into the therapeutic implications of targeting inflammation in HF. Despite mixed results from early clinical trials, emerging evidence suggests that anti-inflammatory therapies offer benefits in specific HF phenotypes. The potential of novel therapeutic strategies, including the use of biologics and small molecule inhibitors, is examined in the context of their ability to modulate inflammatory responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Diana Roman-Pepine
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania.
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001, Cluj-Napoca- Napoca, Romania.
| | - Adela Mihaela Serban
- Cardiology Department, Heart Institute Niculae Stăncioiu, 19-21 Motilor Street, 400001, Cluj-Napoca- Napoca, Romania
- 5 Th Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Roxana-Denisa Capras
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Cristina Mihaela Cismaru
- Department of Infectious Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| | - Adriana Gabriela Filip
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400348, Cluj-Napoca-Napoca, Romania
| |
Collapse
|
9
|
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
|
10
|
Van Pee T, Engelen L, De Boevre M, Derrien M, Hogervorst J, Pero-Gascon R, Plusquin M, Poma G, Vich I Vila A, Covaci A, Vanhaecke L, De Saeger S, Raes J, Nawrot TS. Sex differences in the association between long-term ambient particulate air pollution and the intestinal microbiome composition of children. ENVIRONMENT INTERNATIONAL 2025; 199:109457. [PMID: 40273556 PMCID: PMC12086174 DOI: 10.1016/j.envint.2025.109457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
The intestinal microbiome is essential for gastrointestinal and overall health, yet its response to air pollution in children remains underexplored. In a study involving 412 young children from the ENVIRONAGE cohort, stool samples were analysed via Illumina Miseq sequencing to assess microbiome alpha diversity (observed richness, species evenness, and Shannon diversity) and composition. Exposure to previous year particulate air pollution (black carbon, PM2.5, coarse PM, and PM10) was modeled using high-resolution spatial-temporal interpolation models. Multiple linear regression models were adjusted for a priori selected covariables and stratified by sex. Furthermore, we performed a differential relative abundance analysis at family and genus level, while accounting for the same covariables. Statistically significant effect modification by sex was apparent for several intestinal alpha diversity indices and air pollutants. In boys, we observed negative associations between particulate air pollution exposure and intestinal microbiome richness (estimates ranging from -5.55 to -9.06 per interquartile range (IQR) increase in particulate air pollution exposure) and Shannon diversity (estimates ranging from -0.058 to -0.095 per IQR increase). Differently, in girls non-significant positive associations were observed with species evenness (estimates ranging from 0.019 to 0.020 per IQR increase) and Shannon diversity (estimate 0.065 per IQR increase in black carbon). After multiple testing correction, we reported several bacterial families and genera (Streptococcaceae, Clostridiales Incertae Sedis XIII, Coriobacteriaceae, Streptococcus, and Paraprevotella) to be oppositely associated with particulate air pollution exposure in boys and girls. Our findings show a sex-dependent association between particulate air pollution exposure and intestinal microbiome composition, highlighting boys as potentially more vulnerable to diversity loss associated with childhood exposure to particulate pollution.
Collapse
Affiliation(s)
- Thessa Van Pee
- Centre for Environmental Sciences, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium
| | - Liesa Engelen
- Centre for Environmental Sciences, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium
| | - Marthe De Boevre
- Centre of Excellence in Mycotoxicology and Public Health, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Muriel Derrien
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Katholieke Universiteit Leuven 3000 Leuven, Belgium
| | - Janneke Hogervorst
- Centre for Environmental Sciences, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium
| | - Roger Pero-Gascon
- Centre of Excellence in Mycotoxicology and Public Health, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Michelle Plusquin
- Centre for Environmental Sciences, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium
| | - Giulia Poma
- Toxicological Centre, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Arnau Vich I Vila
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Katholieke Universiteit Leuven 3000 Leuven, Belgium
| | - Adrian Covaci
- Toxicological Centre, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Lynn Vanhaecke
- Laboratory of Integrative Metabolomics (LIMET), Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Sarah De Saeger
- Centre of Excellence in Mycotoxicology and Public Health, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Department of Biotechnology and Food Technology, Faculty of Sciences, University of Johannesburg, South Africa
| | - Jeroen Raes
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Katholieke Universiteit Leuven 3000 Leuven, Belgium
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium; Department of Public Health and Primary Care, Leuven University, Herestraat 49-box 706, 3000 Leuven, Belgium.
| |
Collapse
|
11
|
Yu S, Zhu X, Zhao X, Li Y, Niu X, Chen Y, Ying J. Improvement of chronic metabolic inflammation and regulation of gut homeostasis: Tea as a potential therapy. Pharmacol Ther 2025; 269:108828. [PMID: 40020787 DOI: 10.1016/j.pharmthera.2025.108828] [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: 04/30/2024] [Revised: 12/27/2024] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
Chronic metabolic inflammation is a common mechanism linked to the development of metabolic disorders such as obesity, diabetes, and cardiovascular disease (CVD). Chronic metabolic inflammation often related to alterations in gut homeostasis, and pathological processes involve the activation of endotoxin receptors, metabolic reprogramming, mitochondrial dysfunction, and disruption of intestinal nuclear receptor activity. Recent investigations into homeostasis and chronic metabolic inflammation have revealed a novel mechanism which is characterized by a timing interaction involving multiple components and targets. This article explores the positive impact of tea consumption on metabolic health of populations, with a special focus on the improvement of inflammatory indicators and the regulation of gut microbiota. Studies showed that tea consumption is related to the enrichment of gut microbiota. The relative proportion of Firmicutes/Bacteroidetes (F/B) is altered, while the abundance of Lactobacillus, Bifidobacterium, and A. muciniphila increased significantly in most of the studies. Thus, tea consumption could provide potential protection from the development of chronic diseases by improving gut homeostasis and reducing chronic metabolic inflammation. The direct impact of tea on intestinal homeostasis primarily targets lipopolysaccharide (LPS)-related pathways. This includes reducing the synthesis of intestinal LPS, inhibiting LPS translocation, and preventing the binding of LPS to TLR4 receptors to block downstream inflammatory pathways. The TLR4/MyD88/NF-κB p65 pathway is crucial for anti-metaflammatory responses. The antioxidant properties of tea are linked to enhancing mitochondrial function and mitigating mitochondria-related inflammation by eliminating free radicals, inhibiting NLRP3 inflammasomes, and modulating Nrf2/ARE activity. Tea also contributes to safeguarding the intestinal barrier through various mechanisms, such as promoting the synthesis of short-chain fatty acids in the intestine, activating intestinal aryl hydrocarbon receptor (AhR) and farnesoid X receptor (FXR), and improving enteritis. Functional components that improve chronic metabolic inflammation include tea polyphenols, tea pigments, TPS, etc. Tea metabolites such as 4-Hydroxyphenylacetic acid and 3,4-Dihydroxyflavan derivatives, etc., also contribute to anti-chronic metabolic inflammation effects of tea consumption. The raw materials and processing technologies affect the functional component compositions of tea; therefore, consuming different types of tea may result in varying action characteristics and mechanisms. However, there is currently limited elaboration on this aspect. Future research should conduct in-depth studies on the mechanism of tea and its functional components in improving chronic metabolic inflammation. Researchers should pay attention to whether there are interactions between tea and other foods or drugs, explore safe and effective usage and dosage, and investigate whether there are individual differences in the tea-drinking population leading to different effects of tea intervention. Ultimately, the application of tea drinking could be a universal therapy for regulating intestinal homeostasis, anti-chronic metabolic inflammatory responses, and promoting metabolic health.
Collapse
Affiliation(s)
- Shiyi Yu
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China
| | - Xuan Zhu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Xiayu Zhao
- National Institute of Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yan Li
- National Institute of Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xinghe Niu
- Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; COFCO Nutrition and Health Research Institute, Beijing 102209, China
| | - Yinghua Chen
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China
| | - Jian Ying
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China.
| |
Collapse
|
12
|
Nadeem-Tariq A, Kazemeini S, Michelberger M, Fang C, Maitra S, Nelson K. The Role of Gut Microbiota in Orthopedic Surgery: A Systematic Review. Microorganisms 2025; 13:1048. [PMID: 40431221 PMCID: PMC12113667 DOI: 10.3390/microorganisms13051048] [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: 03/20/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
The human gut microbiome represents a complex ecosystem comprising numerous microorganisms critical to basic physiological processes. The gut microbiome's composition and functionality influence surgical outcomes following orthopedic procedures. The purpose of this study was to evaluate the gut microbiota on critical aspects of orthopedic surgical outcomes. A comprehensive literature search was conducted via PubMed, the Cumulative Index for Nursing and Allied Health Literature (CINAHL), Google Scholar, Cochrane Library, Medline, and Web of Science. A total of 18 research articles of the 599 retrieved results were included in this study. Significant correlations were identified between microbial composition and surgical outcomes, including infection rates, inflammatory responses, and postoperative complications. Bacterial genera like Alistipes and Helicobacter increased postoperative cognitive dysfunction (POCD) risk, while short-chain fatty acid (SCFA)-producing bacteria showed negative correlations with inflammatory markers. Probiotic interventions reduced POCD incidence from 16.4% to 5.1% and modulated inflammatory responses. Additionally, bacterial composition was associated with critical surgical parameters such as bone healing, infection rate, and recovery trajectory. Inflammation, healing processes, and recovery trajectories are influenced by microbial composition in surgical settings. Targeted interventions, such as probiotics, show promise in reducing surgical risks and improving patient recovery.
Collapse
Affiliation(s)
- Ahmed Nadeem-Tariq
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA
| | | | | | | | | | | |
Collapse
|
13
|
Lawrence DA, O'Sullivan B, Graf J, Hogan A, Herbest KW, Salazar JC. The biological and sociological implications of diversity, equity, and inclusion (DEI): life within microbiomes and on earth. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2025:1-9. [PMID: 40298084 DOI: 10.1080/10937404.2025.2497826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
From a biological point of view, Diversity, Equity, and Inclusion (DEI) are important at multiple levels, which include our genetics, microbiomes, diets, and all organ system interactions. Considering only DEI's sociological aspects is equivalent to the error of "throwing out the baby with the bath water." Variances in microbial diversity within our microbiomes might affect our health through systemic interactions affecting metabolites, maintaining immune homeostasis, and wound healing of cellular damage from an infection, physical stress, or psychological trauma. An imbalance of our immune cell subsets, both innate and adaptive, and the microbes in any of our microbiomes might lead to more cellular damage from excessive inflammation and oxidative stress and less immune regulation. The immune dysregulation may occur due to the loss of endometrial barriers enabling the spread of microbes, environmental pollutants, and allergens. Heat waves, sleep deprivation, and increased prevalence of pollutants such as polychlorinated biphenyls, which weaken endothelial barriers, may be responsible for the enhanced prevalence of physical and psychological stresses. Leakage of our useful gut microbiota into the periphery might initiate inflammatory responses, and an altered gut microbiome might affect the gut-brain axis that influences physical and mental health.
Collapse
Affiliation(s)
- David A Lawrence
- Department of Environmental Health, University at Albany, Albany, NY, USA
- Department of Environmental Health, New York State Department of Health, Albany, NY, USA
| | - Brandon O'Sullivan
- Department of Environmental Health, University of Hawaii Manoa, Honolulu, HI, USA
| | - Joerg Graf
- Department of Environmental Health, University of Hawaii Manoa, Honolulu, HI, USA
| | - Alex Hogan
- Pediatrics, Connecticut Children's Medical Center, Hartford, USA
- Pediatrics, UConn Health, Farmington, USA
| | - Katherine W Herbest
- Pediatrics, Connecticut Children's Medical Center, Hartford, USA
- Pediatrics, UConn Health, Farmington, USA
| | - Juan C Salazar
- Pediatrics, Connecticut Children's Medical Center, Hartford, USA
- Pediatrics, UConn Health, Farmington, USA
| |
Collapse
|
14
|
Viggiano D, Iulianiello P, Mancini A, Iacuzzo C, Apicella L, Di Pietro RA, Hamzeh S, Cacciola G, Lippiello E, Gigliotti A, Secondulfo C, Bilancio G, Gigliotti G. Immunological Avalanches in Renal Immune Diseases. Biomedicines 2025; 13:1003. [PMID: 40299571 PMCID: PMC12024534 DOI: 10.3390/biomedicines13041003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
The complex nature of immune system behavior in both autoimmune diseases and transplant rejection can be understood through the lens of avalanche dynamics in critical-point systems. This paper introduces the concept of the "immunological avalanche" as a framework for understanding unpredictable patterns of immune activity in both contexts. Just as avalanches represent sudden releases of accumulated potential energy, immune responses exhibit periods of apparent stability followed by explosive flares triggered by seemingly minor stimuli. The model presented here draws parallels between immune system behavior and other complex systems such as earthquakes, forest fires, and neuronal activity, where localized events can propagate into large-scale disruptions. In autoimmune conditions like systemic lupus erythematosus (SLE), which affects multiple organ systems including the kidneys in approximately 50% of patients, these dynamics manifest as alternating periods of remission and flares. Similarly, in transplant recipients, the immune system exhibits metastable behavior under constant allograft stimulation. This critical-point dynamics framework is characterized by threshold-dependent activation, positive feedback loops, and dynamic non-linearity. In autoimmune diseases, triggers such as UV light exposure, infections, or stress can initiate cascading immune responses. In transplant patients, longitudinal analysis reveals how monitoring oscillatory patterns in blood parameters and biological age markers can predict rejection risk. In a preliminary study on kidney transplant, all measured variables showed temporal instability. Proteinuria exhibited precise log-log linearity in power law analysis, confirming near-critical-point system behavior. Two distinct dynamic patterns emerged: large oscillations in eGFR, proteinuria, or biological age predicted declining function, while small oscillations indicated stability. During avalanche events, biological age increased dramatically, with partial reversal leaving persistent elevation after acute episodes. Understanding these dynamics has important implications for therapeutic approaches in both contexts. Key findings suggest that monitoring parameter oscillations, rather than absolute values, better indicates system instability and potential avalanche events. Additionally, biological age calculations provide valuable prognostic information, while proteinuria measurements offer efficient sampling for system dynamics assessment. This conceptual model provides a unifying framework for understanding the pathogenesis of both autoimmune and transplant-related immune responses, potentially leading to new perspectives in disease management and rejection prediction.
Collapse
Affiliation(s)
- Davide Viggiano
- Department Translational Medical Sciences, University of Campania, 80131 Naples, Italy; (P.I.); (G.C.)
| | - Pietro Iulianiello
- Department Translational Medical Sciences, University of Campania, 80131 Naples, Italy; (P.I.); (G.C.)
| | - Antonio Mancini
- Department of Nephrology and Dialysis, Eboli Hospital, 84025 Eboli, Italy; (A.M.); (A.G.); (G.G.)
| | - Candida Iacuzzo
- Unit of Nephrology, Dialysis and Transplantation, Salerno University Hospital, 84131 Salerno, Italy; (C.I.); (L.A.); (R.A.D.P.)
| | - Luca Apicella
- Unit of Nephrology, Dialysis and Transplantation, Salerno University Hospital, 84131 Salerno, Italy; (C.I.); (L.A.); (R.A.D.P.)
| | - Renata Angela Di Pietro
- Unit of Nephrology, Dialysis and Transplantation, Salerno University Hospital, 84131 Salerno, Italy; (C.I.); (L.A.); (R.A.D.P.)
| | - Sarah Hamzeh
- Department of Public Health, Federico II University of Naples, 80131 Naples, Italy;
| | - Giovanna Cacciola
- Department Translational Medical Sciences, University of Campania, 80131 Naples, Italy; (P.I.); (G.C.)
| | - Eugenio Lippiello
- Department Mathematics and Physics, University of Campania, 81100 Caserta, Italy;
| | - Andrea Gigliotti
- Department of Nephrology and Dialysis, Eboli Hospital, 84025 Eboli, Italy; (A.M.); (A.G.); (G.G.)
| | - Carmine Secondulfo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (C.S.)
| | - Giancarlo Bilancio
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (C.S.)
| | - Giuseppe Gigliotti
- Department of Nephrology and Dialysis, Eboli Hospital, 84025 Eboli, Italy; (A.M.); (A.G.); (G.G.)
| |
Collapse
|
15
|
Deng Y, Zhang Y, Xiao J, Cao Y, Ho CT, Lu M. Allicin Improves Diet-Induced Nonalcoholic Steatohepatitis and Gut Microbiota Dysbiosis in Mice via the Involvement of the Circadian Clock Gene Rev-erbα. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9019-9032. [PMID: 40168418 DOI: 10.1021/acs.jafc.4c12566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Nonalcoholic Steatohepatitis (NASH) is a progressive liver disease characterized by inflammation and liver damage. Allicin, a bioactive compound derived from garlic, has demonstrated anti-inflammatory and antioxidant properties. This study explores the effects of allicin on NASH and gut microbiota dysbiosis induced by a high-fat, high-fructose diet (HFFD) in mice. Allicin supplementation significantly alleviated hepatic inflammation, improved glucose metabolism, and modulated the circadian rhythm gene Rev-erbα, which plays a critical role in regulating inflammation. The anti-inflammatory effects of allicin were diminished in Si-Rev-erbα-treated HepG2 cells, highlighting the importance of circadian regulation in mediating these effects. Allicin's anti-inflammatory effects were associated with increased levels of short-chain fatty acids (SCFAs) and the restoration of diurnal oscillations in proinflammatory cytokines and gut microbiota composition, particularly in genera, such as Akkermansia, Bacteroidetes, and Lactobacillus. These findings suggest that allicin could be a promising therapeutic approach for managing NASH, liver dysfunction, and related metabolic disorders through the modulation of circadian rhythms and the gut microbiome.
Collapse
Affiliation(s)
- Yupei Deng
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yiyi Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
16
|
Kurhaluk N, Kamiński P, Bilski R, Kołodziejska R, Woźniak A, Tkaczenko H. Role of Antioxidants in Modulating the Microbiota-Gut-Brain Axis and Their Impact on Neurodegenerative Diseases. Int J Mol Sci 2025; 26:3658. [PMID: 40332186 PMCID: PMC12027284 DOI: 10.3390/ijms26083658] [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: 03/14/2025] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
This narrative review presents the role of antioxidants in regulating the gut microbiota and the impact on the gut-brain axis, with a particular focus on neurodegenerative diseases, such as Alzheimer's (AD) and Parkinson's disease (PD). These diseases are characterised by cognitive decline, motor dysfunction, and neuroinflammation, all of which are significantly exacerbated by oxidative stress. This review elucidates the contribution of oxidative damage to disease progression and explores the potential of antioxidants to mitigate these pathological processes through modulation of the gut microbiota and associated pathways. Based on recent studies retrieved from reputable databases, including PubMed, Web of Science, and Scopus, this article outlines the mechanisms by which antioxidants influence gut health and exert neuroprotective effects. Specifically, it discusses how antioxidants, including polyphenols, vitamins, and flavonoids, contribute to the reduction in reactive oxygen species (ROS) production and neuroinflammation, thereby promoting neuronal survival and minimising oxidative damage in the brain. In addition, the article explores the role of antioxidants in modulating key molecular pathways involved in oxidative stress and neuroinflammation, such as the NF-κB, Nrf2, MAPK, and PI3K/AKT pathways, which regulate ROS generation, inflammatory cytokine expression, and antioxidant responses essential for maintaining cellular homeostasis in both the gut and the central nervous system. In addition, this review explores the complex relationship between gut-derived metabolites, oxidative stress, and neurodegenerative diseases, highlighting how dysbiosis-an imbalance in the gut microbiota-can exacerbate oxidative stress and contribute to neuroinflammation, thereby accelerating the progression of such diseases as AD and PD. The review also examines the role of short-chain fatty acids (SCFAs) produced by beneficial gut bacteria in modulating these pathways to attenuate neuroinflammation and oxidative damage. Furthermore, the article explores the therapeutic potential of microbiota-targeted interventions, including antioxidant delivery by probiotics and prebiotics, as innovative strategies to restore microbial homeostasis and support brain health. By synthesising current knowledge on the interplay between antioxidants, the gut-brain axis, and the molecular mechanisms underlying neurodegeneration, this review highlights the therapeutic promise of antioxidant-based interventions in mitigating oxidative stress and neurodegenerative disease progression. It also highlights the need for further research into antioxidant-rich dietary strategies and microbiota-focused therapies as promising avenues for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22 B, 76-200 Słupsk, Poland;
| | - Piotr Kamiński
- Department of Medical Biology and Biochemistry, Division of Ecology and Environmental Protection, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, 85-094 Bydgoszcz, Poland;
- Department of Biotechnology, Institute of Biological Sciences, Faculty of Biological Sciences, University of Zielona Góra, Prof. Z. Szafran St. 1, 65-516 Zielona Góra, Poland
| | - Rafał Bilski
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Halina Tkaczenko
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22 B, 76-200 Słupsk, Poland;
| |
Collapse
|
17
|
He Y, Hu H, Liang X, Liang J, Li F, Zhou X. Gut microbes-muscle axis in muscle function and meat quality. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2885-4. [PMID: 40220074 DOI: 10.1007/s11427-024-2885-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/12/2025] [Indexed: 04/14/2025]
Abstract
The concept of the gut microbes-muscle axis underscores the impact of intestinal microbiota on the muscular system, an area that is increasingly coming to light. However, current interpretations and applications of this concept remain underdeveloped. In this review, we concluded and discussed factors, such as short-chain fatty acids, amino acids, vitamins, bile acids, antibiotics, cytokines, hormones, and extracellular vesicles that mediate gut microbes-muscle crosstalk and influence the gut microbes-muscle axis. Additionally, we examined how the gut microbes-muscle axis affects muscle mass, muscle strength, muscle metabolism, as well as muscle oxidative and immune status. Furthermore, we reviewed the influence of the microbes-muscle axis on muscle fiber type transition, muscle fat deposition, and meat quality. These insights illuminate the potential mechanisms by which the gut microbes-muscle axis operates in humans and animals. Thus, this review provides a theoretical foundation for future research and offers practical guidance for its application in biomedical and livestock industries.
Collapse
Affiliation(s)
- Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Hong Hu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| | - Xuqing Liang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Jing Liang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fengna Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, 410219, China.
| |
Collapse
|
18
|
Liu HJ, Wu MC, Gau SY. Role of gut microbiota and mesenteric adipose tissue in the pathology of Crohn's disease: Potential therapeutic targets. World J Gastroenterol 2025; 31:102291. [PMID: 40248060 PMCID: PMC12001166 DOI: 10.3748/wjg.v31.i13.102291] [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: 10/14/2024] [Revised: 03/01/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
This editorial comments on the article by Wu et al in the World Journal of Gastroenterology. The article explored the relationship between mesenteric adipose tissue, creeping fat, inflammation, and gut microbiota in Crohn's disease (CD). We discussed three key aspects of the interaction between gut microbiota and inflammatory bowel disease (IBD): The physiological functions of the gut microbiota, the potential role of probiotics in IBD treatment; and the effect of fecal microbiota transplantation (FMT) in combating IBD. IBD, comprising CD and ulcerative colitis (UC), is influenced by the gut microbiota. Changes in gut microbiota composition disrupt intestinal function and promote chronic inflammation, but the exact mechanisms remain unclear. Probiotics have demonstrated some efficacy in inducing remission in UC, though their effectiveness in CD is still debated. FMT shows promise in treating IBD, especially UC, by restoring gut microbiota diversity and inducing clinical remission. As for CD, FMT has potential, but more studies are needed to confirm its long-term effectiveness and safety. Dietary approaches may help manage IBD symptoms or disease activity, but patient adherence is crucial. Clinicians and researchers must recognize the importance of the gut microbiota and the need for personalized therapies targeting microbial imbalances.
Collapse
Affiliation(s)
- Han-Jung Liu
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Meng-Che Wu
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Division of Pediatric Gastroenterology, Children's Medical Center, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Shuo-Yan Gau
- Department and Graduate Institute of Business Administration, National Taiwan University, Taipei 106319, Taiwan
| |
Collapse
|
19
|
Zarimeidani F, Rahmati R, Mostafavi M, Darvishi M, Khodadadi S, Mohammadi M, Shamlou F, Bakhtiyari S, Alipourfard I. Gut Microbiota and Autism Spectrum Disorder: A Neuroinflammatory Mediated Mechanism of Pathogenesis? Inflammation 2025; 48:501-519. [PMID: 39093342 PMCID: PMC12053372 DOI: 10.1007/s10753-024-02061-y] [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: 01/31/2024] [Revised: 04/28/2024] [Accepted: 05/21/2024] [Indexed: 08/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social communication and behavior, frequently accompanied by restricted and repetitive patterns of interests or activities. The gut microbiota has been implicated in the etiology of ASD due to its impact on the bidirectional communication pathway known as the gut-brain axis. However, the precise involvement of the gut microbiota in the causation of ASD is unclear. This study critically examines recent evidence to rationalize a probable mechanism in which gut microbiota symbiosis can induce neuroinflammation through intermediator cytokines and metabolites. To develop ASD, loss of the integrity of the intestinal barrier, activation of microglia, and dysregulation of neurotransmitters are caused by neural inflammatory factors. It has emphasized the potential role of neuroinflammatory intermediates linked to gut microbiota alterations in individuals with ASD. Specifically, cytokines like brain-derived neurotrophic factor, calprotectin, eotaxin, and some metabolites and microRNAs have been considered etiological biomarkers. We have also overviewed how probiotic trials may be used as a therapeutic strategy in ASD to reestablish a healthy balance in the gut microbiota. Evidence indicates neuroinflammation induced by dysregulated gut microbiota in ASD, yet there is little clarity based on analysis of the circulating immune profile. It deems the repair of microbiota load would lower inflammatory chaos in the GI tract, correct neuroinflammatory mediators, and modulate the neurotransmitters to attenuate autism. The interaction between the gut and the brain, along with alterations in microbiota and neuroinflammatory biomarkers, serves as a foundational background for understanding the etiology, diagnosis, prognosis, and treatment of autism spectrum disorder.
Collapse
Affiliation(s)
- Fatemeh Zarimeidani
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Rahem Rahmati
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mehrnaz Mostafavi
- Faculty of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Darvishi
- School of Aerospace and Subaquatic Medicine, Infectious Diseases & Tropical Medicine Research Center (IDTMC), AJA University of Medical Sciences, Tehran, Iran
| | - Sanaz Khodadadi
- Student Research Committee, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farid Shamlou
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Salar Bakhtiyari
- Feinberg Cardiovascular and Renal Research Institute, North Western University, Chicago. Illinois, USA
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcin Kasprzaka 44/52, 01-224, Warsaw, Poland.
| |
Collapse
|
20
|
Parekh Z, Xiao J, Mani A, Evans Q, Phung C, Barba HA, Xie B, Sidebottom AM, Sundararajan A, Lin H, Ramaswamy R, Dao D, Gonnah R, Yehia M, Hariprasad SM, D'Souza M, Sulakhe D, Chang EB, Skondra D. Fecal Microbial Profiles and Short-Chain Fatty Acid/Bile Acid Metabolomics in Patients With Age-Related Macular Degeneration: A Pilot Study. Invest Ophthalmol Vis Sci 2025; 66:21. [PMID: 40202735 PMCID: PMC11993127 DOI: 10.1167/iovs.66.4.21] [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: 04/11/2024] [Accepted: 02/25/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose Age-related macular degeneration (AMD) is a multifactorial disease, and studies have implicated the role of gut microbiota in its pathogenesis. However, characterization of microbiome dysbiosis and associated microbial-derived metabolomic profiles across AMD stages remains unknown. In this pilot study, we explored how gut microbiome composition and gut-derived metabolites differ in AMD. Methods Our pilot study analyzed fasted stool samples that were collected from 22 patients at a tertiary academic center. Subjects were classified as control, intermediate AMD, or advanced AMD based on clinical presentation. 16S rRNA amplicon sequencing and standard chromatography-mass spectrometry methods were used to identify bacterial taxonomy composition and abundance of short-chain fatty acids (SCFAs) and bile acids (BAs), respectively. Genetic testing was used to investigate the frequency of 14 high-risk single nucleotide polymorphisms (SNPs) associated with AMD in the AMD cohort. Results Forty-three differentially abundant genera were present among the control, intermediate, and advanced groups. Taxa with known roles in immunologic pathways, such as Desulfovibrionales (q = 0.10) and Terrisporobacter (q = 1.16e-03), were in greater abundance in advanced AMD patients compared to intermediate. Advanced AMD patients had decreased abundance of 12 SCFAs, including acetate (P = 0.002), butyrate (P = 0.04), and propionate (P = 0.01), along with 12 BAs, including taurocholic acid (P = 0.02) and tauroursodeoxycholic acid (P = 0.04). Frequencies of high-risk SNPs were not significantly different between the intermediate and advanced AMD groups. Conclusions This pilot study identifies distinct gut microbiome compositions and metabolomic profiles associated with AMD and its stages, providing preliminary evidence of a potential link between gut microbiota and AMD pathogenesis. To validate these findings and elucidate the underlying mechanisms, future research with larger cohorts and more comprehensive sampling is strongly recommended.
Collapse
Affiliation(s)
- Zaid Parekh
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Jason Xiao
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Amir Mani
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Quadis Evans
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Christopher Phung
- Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Hugo A. Barba
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Bingqing Xie
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Ashley M. Sidebottom
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Anitha Sundararajan
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Huaiying Lin
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Ramanujam Ramaswamy
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - David Dao
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Reem Gonnah
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Madeleine Yehia
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Seenu M. Hariprasad
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| | - Mark D'Souza
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Dinanath Sulakhe
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Eugene B. Chang
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States
- Duchossois Family Institute, The University of Chicago, Chicago, Illinois, United States
| | - Dimitra Skondra
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
21
|
Wu W, Li S, Ye Z. Targeting the gut microbiota-inflammation-brain axis as a potential therapeutic strategy for psychiatric disorders: A Mendelian randomization analysis. J Affect Disord 2025; 374:150-159. [PMID: 39809351 DOI: 10.1016/j.jad.2025.01.050] [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: 07/23/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND Extensive research indicates a link between gut microbiota dysbiosis and psychiatric disorders. However, the causal relationships between gut microbiota and different types of psychiatric disorders, as well as whether inflammatory factors mediate these relationships, remain unclear. METHODS We utilized summary statistics from the largest genome-wide association studies to date for gut microbiota (n = 18,340 in MiBioGen consortium), circulating inflammatory factors (n = 8293 for 41 factors and n = 14,824 for 91 factors in GWAS catalog), and six major psychiatric disorders from the Psychiatric Genomics Consortium (PGC): attention deficit hyperactivity disorder (ADHD, n = 38,691), anxiety disorder (ANX, n = 2248), bipolar disorder (BIP, n = 41,917), anorexia nervosa (AN, n = 16,992), schizophrenia (SCZ, n = 36,989), and autism spectrum disorder (ASD, n = 18,381). We conducted bidirectional Mendelian randomization (MR) analysis to explore the causal relationships between gut microbiota and psychiatric disorders. Additionally, we performed two-step MR and multivariable MR (MVMR) analyses to identify potential mediating inflammatory factors. RESULTS We found significant causal relationships between 11 gut microbiota and ADHD, 2 gut microbiota and ANX, 11 gut microbiota and BIP, 8 gut microbiota and AN, 15 gut microbiota and SCZ, and 5 gut microbiota and ASD. There were 16 positive and 15 negative causal effects between inflammatory factors and psychiatric disorders. Furthermore, MVMR analysis results indicated that the correlation between genus Roseburia and ADHD was mediated by MCSF, with a mediation proportion of 3.3 %; the correlation between genus Erysipelotrichaceae UCG003 and BIP was mediated by GDNF, with a mediation proportion of 3.7 %; and the correlation between family Prevotellaceae and SCZ was mediated by CD40, with a mediation proportion of 8.2 %. CONCLUSIONS The MR analysis results supported causal relationships between gut microbiota and six psychiatric disorders, as well as the potential mediating role of inflammatory factors. This study highlights the potential role of the gut microbiota-inflammation-brain axis in psychiatric disorders.
Collapse
Affiliation(s)
- Wenjing Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China
| | - Shuhan Li
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Zengjie Ye
- School of Nursing, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
22
|
Sun Y, Xu B. A critical review on effects of artificial sweeteners on gut microbiota and gastrointestinal health. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:2737-2747. [PMID: 39878083 DOI: 10.1002/jsfa.14148] [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] [Received: 05/02/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
Artificial sweeteners have emerged as popular alternatives to traditional sweeteners, driven by the growing concern over sugar consumption and its associated rise in obesity and metabolic disorders. Despite their widespread use, the safety and health implications of artificial sweeteners remain a topic of debate, with conflicting evidence contributing to uncertainty about their long-term effects. This review synthesizes current scientific evidence regarding the impact of artificial sweeteners on gut microbiota and gastrointestinal health. Our analysis of in vitro experiments, animal models, and clinical trials reveals that artificial sweeteners can alter the composition and abundance of gut microbes. These changes raise concerns about their potential to affect overall gut health and contribute to gastrointestinal disorders. Additionally, artificial sweeteners have been shown to influence the production of metabolites by gut bacteria, further impacting systemic health. The findings suggest that artificial sweeteners may have complex and sometimes contradictory effects on gut microbiota. While some studies indicate potential benefits, such as reduced caloric intake and weight management, others highlight detrimental effects on microbial balance and metabolic functions. The inconsistent results underscore the need for further research to comprehensively understand the physiological impacts of various artificial sweeteners on human health. Future studies should aim for long-term, well-controlled investigations to clarify these relationships, ensuring evidence-based guidelines for the safe use of artificial sweeteners in diet management. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yizhe Sun
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, China
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, China
| |
Collapse
|
23
|
Singlitico A, Grassa D, Kaplan R, Smimmo A, Maccauro G, Vitiello R. The hidden connection between gut microbiota and periprosthetic joint infections: a scoping review. J Bone Jt Infect 2025; 10:85-92. [PMID: 40271508 PMCID: PMC12015178 DOI: 10.5194/jbji-10-85-2025] [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] [Received: 10/17/2024] [Accepted: 02/04/2025] [Indexed: 04/25/2025] Open
Abstract
Background: Periprosthetic joint infections (PJIs) pose a significant challenge in orthopedic surgery, and emerging evidence suggests that the gut microbiome may play a crucial role in their development and management. Despite the rarity of these infections, the continuous increase in prosthetic joint arthroplasties has made understanding how to prevent them more pressing. A stronger comprehension of the disruption of the gut microbiome and how this can lead to more of these infections and other pre-surgical risks may be crucial in preventing them. Objective: This article aims to provide a stronger understanding of the topic through the analysis of different pieces of already existing literature to help draw new conclusions and raise potential questions that need answering. Methods: A comprehensive search strategy without filters was employed, and multiple papers were thoroughly analyzed, understood, and compiled into this paper. Conclusions: Despite the limitations of some of the analyzed studies and finite evidence, this paper suggests that there could be a connection between periprosthetic joint infections and a compromised gut microbiome. However, further research is required to draw a definitive conclusion.
Collapse
Affiliation(s)
- Alessandro Singlitico
- Department of Orthopaedics, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Daniele Grassa
- Department of Orthopaedics, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Rami Kaplan
- Department of Orthopaedics, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alessandro Smimmo
- Department of Orthopaedic and Traumatology, Aurelia Hospital Garofalo Healthcare, 00165 Rome, Italy
| | - Giulio Maccauro
- Department of Orthopaedics, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Raffaele Vitiello
- Department of Orthopaedics, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
24
|
Cercamondi CI, Bendik I, Eckhardt E, Mak T, Seifert N, Kuratli K, Richard N, Tamasi B, Mussler B, Wintergerst E. A Postbiotic Derived from Lactobacillaceae Protects Intestinal Barrier Function in a Challenge Model Using Colon Organoid Tubules. Foods 2025; 14:1173. [PMID: 40238399 PMCID: PMC11988720 DOI: 10.3390/foods14071173] [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: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Postbiotics may help strengthen intestinal barrier function. This study assessed the effects of a postbiotic derived from Limosilactobacillus fermentum and Lactobacillus delbrueckii subsp. lactis on epithelial barrier and cytokine production. Human-derived colon tubules were cultured on chips for 15 days. On day 8, the epithelial barrier was disrupted with 0.7 μM afatinib. Postbiotic doses of 5, 10, or 20 mg/mL were added on days 6, 8, 11, and 13. Trans-epithelial electrical resistance (TEER) was measured on days 6, 8, 11, 13, and 15, along with phase contrast imaging. Cytokine levels were measured on day 13. All three postbiotic concentrations resulted in better TEER recovery on day 15 vs. the control (p < 0.001). On day 13, 10 and 20 mg/mL increased TEER (p < 0.001), but only 20 mg/mL did on day 11 (p < 0.05). Phase imaging confirmed the dose-dependent effect. The 20 mg/mL dose more effectively reduced CCL2, CX3CL1, CXCL1, CXCL5, IL-8, IL-11, and IL-4 than the other doses (p < 0.01), and 10 mg/mL more effectively reduced CCL2, CXCL1, CXCL10, IL-10, IL-11, and IL-23 than 5 mg/mL (p < 0.01). In a colonic organoid model, the Lactobacillaceae-derived postbiotic prevented drug-induced epithelial damage, enhanced recovery, and modulated cytokine secretion towards a more anti-inflammatory profile in a dose-dependent manner.
Collapse
Affiliation(s)
- Colin I. Cercamondi
- DSM-Firmenich AG, Wurmisweg 576, 4303 Kaiseraugst, Switzerland; (I.B.); (E.E.); (T.M.); (N.S.); (K.K.); (N.R.); (B.T.); (B.M.); (E.W.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Vemulapalli V, Shirwaikar Thomas A. The Role of Vitamin D in Gastrointestinal Homeostasis and Gut Inflammation. Int J Mol Sci 2025; 26:3020. [PMID: 40243631 PMCID: PMC11988781 DOI: 10.3390/ijms26073020] [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: 02/13/2025] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Gastrointestinal homeostasis describes a delicate state of equilibrium in which various systems cooperate to maintain digestive health, support microbial activity, and regulate immune responses. There is growing evidence that Vitamin D is one of the many factors that influences gastrointestinal homeostasis through its effects on gut barrier integrity, regulating microbial diversity and modulating immune responses. Given these effects of Vitamin D, there may be potential for it as both a preventative and a therapeutic intervention for a variety of conditions, but especially for inflammatory conditions of the gastrointestinal tract. This article will summarize the role of Vitamin D in a state of equilibrium, as well as its role in a pro-inflammatory state in the gastrointestinal tract.
Collapse
Affiliation(s)
- Varun Vemulapalli
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anusha Shirwaikar Thomas
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
26
|
Hauser G, Benjak Horvat I, Rajilić-Stojanović M, Krznarić-Zrnić I, Kukla M, Aljinović-Vučić V, Mikolašević I. Intestinal Microbiota Modulation by Fecal Microbiota Transplantation in Nonalcoholic Fatty Liver Disease. Biomedicines 2025; 13:779. [PMID: 40299326 PMCID: PMC12024620 DOI: 10.3390/biomedicines13040779] [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: 01/04/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Numerous factors are involved in the pathogenesis of nonalcoholic fatty liver disease (NAFLD), which are responsible for its development and progression as an independent entity, but also thanks to their simultaneous action. This is explained by the hypothesis of multiple parallel hits. These factors are insulin resistance, lipid metabolism alteration, oxidative stress, endoplasmic reticulum stress, inflammatory cytokine liberation, gut microbiota dysbiosis or gut-liver axis activation. This is a systematic review which has an aim to show the connection between intestinal microbiota and the role of its disbalance in the development of NAFLD. The gut microbiota is made from a wide spectrum of microorganisms that has a systemic impact on human health, with a well-documented role in digestion, energy metabolism, the stimulation of the immune system, synthesis of essential nutrients, etc. It has been shown that dysbiosis is associated with all three stages of chronic liver disease. Thus, the modulation of the gut microbiota has attracted research interest as a novel therapeutic approach for the management of NAFLD patients. The modification of microbiota can be achieved by substantial diet modification and the application of probiotics or prebiotics, while the most radical effects are observed by fecal microbiota transplantation (FMT). Given the results of FMT in the context of metabolic syndrome (MetS) and NAFLD in animal models and scarce pilot studies on humans, FMT seems to be a promising treatment option that could reverse intestinal dysbiosis and thereby influence the course of NAFLD.
Collapse
Affiliation(s)
- Goran Hauser
- Department of Gastroenterology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (G.H.); (I.K.-Z.); (I.M.)
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| | - Indira Benjak Horvat
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
- County Hospital Varaždin, 42000 Varaždin, Croatia
| | - Mirjana Rajilić-Stojanović
- Department of Biochemical Engineering & Biotechnology, Faculty of Technology and Metallurgy, University of Belgrade, 11000 Belgrade, Serbia;
| | - Irena Krznarić-Zrnić
- Department of Gastroenterology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (G.H.); (I.K.-Z.); (I.M.)
| | - Michail Kukla
- Department of Internal Medicine and Geriatrics, Jagiellonian University Medical College, 31-121 Cracow, Poland;
- Department of Endoscopy, University Hospital in Cracow, 30-688 Cracow, Poland
- 1st Infectious Diseases Ward, Gromkowski Regional Specialist Hospital, Wroclaw, 5 Koszarowa St., 50-149 Wroclaw, Poland
| | - Vedrana Aljinović-Vučić
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
- Medical Affairs Department, Jadran Galenski Laboratorij d.d., 51000 Rijeka, Croatia
| | - Ivana Mikolašević
- Department of Gastroenterology, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia; (G.H.); (I.K.-Z.); (I.M.)
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| |
Collapse
|
27
|
Alzahrani AJ, Al-Hebshi BM, Yahia ZA, Al-Judaibi EA, Alsaadi KH, Al-Judaibi AA. Impact of Microbiota Diversity on Inflammatory Bowel Disease. Microorganisms 2025; 13:710. [PMID: 40284547 PMCID: PMC12029714 DOI: 10.3390/microorganisms13040710] [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: 02/07/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic condition that includes two main types, Crohn's disease (CD) and ulcerative colitis (UC), involving inflammation of the gastrointestinal (GI) tract. The exact cause of IBD is unknown but could be a combination of genetic, environmental, and immune system factors. This study investigated the impact of IBD on microbiota diversity by evaluating the differences in microbial composition and the microbiota of a control group (A) of healthy individuals and a group (B) of IBD patients. Sixty biopsies were collected from participants recruited from hospitals in Makkah, Saudi Arabia. Biopsy specimens were taken during colonoscopy examination, and bacterial identification was performed by extracting ribosomal DNA from sigmoid colon biopsies using a DNeasy Blood & Tissue Kit. Metagenomics and bioinformatics analyses were then conducted to analyze and compare the microbiota in the two groups. The results showed that the varieties of core microbiome species were 3.81% greater in the IBD patients than in the members of the control group. Furthermore, the differences between the groups were significantly greater than the variations within each group. Differences between the two groups were detected in the relative abundance of Clostridium nexile, Ruminococcus gnavus, Ruminococcus faecis, and Escherichia coli. These results indicate that microbiota could play a role in the pathogenesis of IBD and suggest that microbial diversity can serve as a biomarker for diagnosing the disease and monitoring its progression.
Collapse
Affiliation(s)
- Ashwag J. Alzahrani
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| | - Basma M. Al-Hebshi
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| | - Zolfekar A. Yahia
- Department of Internal Medicine, Al Noor Specialist Hospital, Ministry of Health, Makkah 24242, Saudi Arabia;
| | - Effat A. Al-Judaibi
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| | - Khloud H. Alsaadi
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| | - Awatif A. Al-Judaibi
- Department of Biological Sciences, Microbiology Section, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia; (A.J.A.); (B.M.A.-H.); (E.A.A.-J.); (K.H.A.)
| |
Collapse
|
28
|
Mamun MAA, Rakib A, Mandal M, Singh UP. Impact of a High-Fat Diet on the Gut Microbiome: A Comprehensive Study of Microbial and Metabolite Shifts During Obesity. Cells 2025; 14:463. [PMID: 40136712 PMCID: PMC11940932 DOI: 10.3390/cells14060463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Over the last few decades, the prevalence of metabolic diseases such as obesity, diabetes, non-alcoholic fatty liver disease, hypertension, and hyperuricemia has surged, primarily due to high-fat diet (HFD). The pathologies of these metabolic diseases show disease-specific alterations in the composition and function of their gut microbiome. How HFD alters the microbiome and its metabolite to mediate adipose tissue (AT) inflammation and obesity is not well known. Thus, this study aimed to identify the changes in the gut microbiome and metabolomic signatures induced by an HFD to alter obesity. To explore the changes in the gut microbiota and metabolites, 16S rRNA gene amplicon sequencing and metabolomic analyses were performed after HFD and normal diet (ND) feeding. We noticed that, at taxonomic levels, the number of operational taxonomic units (OTUs), along with the Chao and Shannon indexes, significantly shifted in HFD-fed mice compared to those fed a ND. Similarly, at the phylum level, an increase in Firmicutes and a decrease in Bacteroidetes were noticed in HFD-fed mice. At the genus level, an increase in Lactobacillus and Ruminococcus was observed, while Allobaculum, Clostridium, and Akkermansia were markedly reduced in the HFD group. Many bacteria from the Ruminococcus genus impair bile acid metabolism and restrict weight loss. Firmicutes are efficient in breaking down complex carbohydrates into short-chain fatty acids (SCFAs) and other metabolites, whereas Bacteroidetes are involved in a more balanced or efficient energy extraction. Thus, an increase in Firmicutes over Bacteroidetes enhances the absorption of more calories from food, which may contribute to obesity. Taken together, the altered gut microbiota and metabolites trigger AT inflammation, which contributes to metabolic dysregulation and disease progression. Thus, this study highlights the potential of the gut microbiome in the development of therapeutic strategies for obesity and related metabolic disorders.
Collapse
Affiliation(s)
| | | | | | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA; (M.A.A.M.); (A.R.); (M.M.)
| |
Collapse
|
29
|
McDonnell KJ. Operationalizing Team Science at the Academic Cancer Center Network to Unveil the Structure and Function of the Gut Microbiome. J Clin Med 2025; 14:2040. [PMID: 40142848 PMCID: PMC11943358 DOI: 10.3390/jcm14062040] [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: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Oncologists increasingly recognize the microbiome as an important facilitator of health as well as a contributor to disease, including, specifically, cancer. Our knowledge of the etiologies, mechanisms, and modulation of microbiome states that ameliorate or promote cancer continues to evolve. The progressive refinement and adoption of "omic" technologies (genomics, transcriptomics, proteomics, and metabolomics) and utilization of advanced computational methods accelerate this evolution. The academic cancer center network, with its immediate access to extensive, multidisciplinary expertise and scientific resources, has the potential to catalyze microbiome research. Here, we review our current understanding of the role of the gut microbiome in cancer prevention, predisposition, and response to therapy. We underscore the promise of operationalizing the academic cancer center network to uncover the structure and function of the gut microbiome; we highlight the unique microbiome-related expert resources available at the City of Hope of Comprehensive Cancer Center as an example of the potential of team science to achieve novel scientific and clinical discovery.
Collapse
Affiliation(s)
- Kevin J McDonnell
- Center for Precision Medicine, Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
30
|
Ghosh AN, Walsh CJ, Maiden MJ, Stinear TP, Deane AM. Effect of dietary fibre on the gastrointestinal microbiota during critical illness: A scoping review. World J Crit Care Med 2025; 14:98241. [DOI: 10.5492/wjccm.v14.i1.98241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 12/11/2024] Open
Abstract
The systemic effects of gastrointestinal (GI) microbiota in health and during chronic diseases is increasingly recognised. Dietary strategies to modulate the GI microbiota during chronic diseases have demonstrated promise. While changes in dietary intake can rapidly change the GI microbiota, the impact of dietary changes during acute critical illness on the microbiota remain uncertain. Dietary fibre is metabolised by carbohydrate-active enzymes and, in health, can alter GI microbiota. The aim of this scoping review was to describe the effects of dietary fibre supplementation in health and disease states, specifically during critical illness. Randomised controlled trials and prospective cohort studies that include adults (> 18 years age) and reported changes to GI microbiota as one of the study outcomes using non-culture methods, were identified. Studies show dietary fibres have an impact on faecal microbiota in health and disease. The fibre, inulin, has a marked and specific effect on increasing the abundance of faecal Bifidobacteria. Short chain fatty acids produced by Bifidobacteria have been shown to be beneficial in other patient populations. Very few trials have evaluated the effect of dietary fibre on the GI microbiota during critical illness. More research is necessary to establish optimal fibre type, doses, duration of intervention in critical illness.
Collapse
Affiliation(s)
- Angajendra N Ghosh
- Department of Intensive Care, The Northern Hospital, Epping 3076, Victoria, Australia
| | - Calum J Walsh
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, Melbourne 3052, Victoria, Australia
| | - Matthew J Maiden
- Department of Intensive Care, The Royal Melbourne Hospital, The University of Melbourne, Parkville 3050, Victoria, Australia
| | - Tim P Stinear
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, Melbourne 3052, Victoria, Australia
| | - Adam M Deane
- Department of Intensive Care Medicine, The Royal Melbourne Hospital, Parkville 3050, Victoria, Australia
| |
Collapse
|
31
|
Manfredi JN, Gupta SK, Vyavahare S, Deak F, Lu X, Buddha L, Wankhade U, Lohakare J, Isales C, Fulzele S. Gut microbiota dysbiosis in Alzheimer's disease (AD): Insights from human clinical studies and the mouse AD models. Physiol Behav 2025; 290:114778. [PMID: 39672482 DOI: 10.1016/j.physbeh.2024.114778] [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: 08/20/2024] [Revised: 11/19/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Alzheimer's Disease (AD) is a debilitating neurocognitive disorder with an unclear underlying mechanism. Recent studies have implicated gut microbiota dysbiosis with the onset and progression of AD. The connection between gut microbiota and AD can significantly affect the prevention and treatment of AD patients. This systematic review summarizes primary outcomes of human and mouse AD models concerning gut microbiota alterations. A systematic literature search in February through March 2023 was conducted on PubMed, Embase, and Web of Science. We identified 711 as potential manuscripts of which 672 were excluded because of irrelevance to the identified search criteria. Primary outcomes include microbiota compositions of control and AD models in humans and mice. In total, 39 studies were included (19 mouse and 20 human studies), published between 2017 and 2023. We included studies involving well-established mice models of AD (5xFAD, 3xTg-AD, APP/PS1, Tg2576, and APPPS2) which harbor mutations and genes that drive the formation of Aß plaques. All human studies were included on those with AD or mild cognitive impairment. Among alterations in gut microbiota, most studies found a decreased abundance of the phyla Firmicutes and Bifidobacteria, a genus of the phylum Actinomycetota. An increased abundance of the phyla Bacteroidetes and Proteobacteria were identified in animal and human studies. Studies indicated that gut microbiota alter the pathogenesis of AD through its impact on neuroinflammation and permeability of the gastrointestinal tract. The ensuing increase in blood-brain barrier permeability may accelerate Aβ penetrance and formation of neuritic plaques that align with the amyloid hypothesis of AD pathogenesis. Further studies should assess the relationship between gut microbiota and AD progression and therapy preserving beneficial gut microbiota.
Collapse
Affiliation(s)
- John N Manfredi
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sonu Kumar Gupta
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sagar Vyavahare
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ferenc Deak
- Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA
| | - Xinyun Lu
- Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA
| | - Lasya Buddha
- Arkansas Children's Nutrition Center, Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Umesh Wankhade
- Arkansas Children's Nutrition Center, Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jayant Lohakare
- College of Agriculture, Food, and Natural Resources, Prairie View A&M University, Prairie View, TX 77446, USA
| | - Carlos Isales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA; College of Agriculture, Food, and Natural Resources, Prairie View A&M University, Prairie View, TX 77446, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, GA, USA; Department of Orthopedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
32
|
Kiouri DP, Batsis GC, Mavromoustakos T, Giuliani A, Chasapis CT. Structure-Based Modeling of the Gut Bacteria-Host Interactome Through Statistical Analysis of Domain-Domain Associations Using Machine Learning. BIOTECH 2025; 14:13. [PMID: 40227324 PMCID: PMC11940256 DOI: 10.3390/biotech14010013] [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: 01/08/2025] [Revised: 02/16/2025] [Accepted: 02/21/2025] [Indexed: 04/15/2025] Open
Abstract
The gut microbiome, a complex ecosystem of microorganisms, plays a pivotal role in human health and disease. The gut microbiome's influence extends beyond the digestive system to various organs, and its imbalance is linked to a wide range of diseases, including cancer and neurodevelopmental, inflammatory, metabolic, cardiovascular, autoimmune, and psychiatric diseases. Despite its significance, the interactions between gut bacteria and human proteins remain understudied, with less than 20,000 experimentally validated protein interactions between the host and any bacteria species. This study addresses this knowledge gap by predicting a protein-protein interaction network between gut bacterial and human proteins. Using statistical associations between Pfam domains, a comprehensive dataset of over one million experimentally validated pan-bacterial-human protein interactions, as well as inter- and intra-species protein interactions from various organisms, were used for the development of a machine learning-based prediction method to uncover key regulatory molecules in this dynamic system. This study's findings contribute to the understanding of the intricate gut microbiome-host relationship and pave the way for future experimental validation and therapeutic strategies targeting the gut microbiome interplay.
Collapse
Affiliation(s)
- Despoina P. Kiouri
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Georgios C. Batsis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
| | - Thomas Mavromoustakos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Alessandro Giuliani
- Environment and Health Department, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
| |
Collapse
|
33
|
Peña-Durán E, García-Galindo JJ, López-Murillo LD, Huerta-Huerta A, Balleza-Alejandri LR, Beltrán-Ramírez A, Anaya-Ambriz EJ, Suárez-Rico DO. Microbiota and Inflammatory Markers: A Review of Their Interplay, Clinical Implications, and Metabolic Disorders. Int J Mol Sci 2025; 26:1773. [PMID: 40004236 PMCID: PMC11854938 DOI: 10.3390/ijms26041773] [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: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The human microbiota, a complex ecosystem of microorganisms, plays a pivotal role in regulating host immunity and metabolism. This review investigates the interplay between microbiota and inflammatory markers, emphasizing their impact on metabolic and autoimmune disorders. Key inflammatory biomarkers, such as C-reactive protein (CRP), interleukin-6 (IL-6), lipopolysaccharides (LPS), zonulin (ZO-1), and netrin-1 (Ntn1), are discussed in the context of intestinal barrier integrity and chronic inflammation. Dysbiosis, characterized by alterations in microbial composition and function, directly modulates the levels and activity of these biomarkers, exacerbating inflammatory responses and compromising epithelial barriers. The disruption of microbiota is further correlated with increased intestinal permeability and chronic inflammation, serving as a precursor to conditions like type 2 diabetes (T2D), obesity, and non-alcoholic fatty liver disease. Additionally, this review examines therapeutic strategies, including probiotics and prebiotics, designed to restore microbial balance, mitigate inflammation, and enhance metabolic homeostasis. Emerging evidence positions microbiota-targeted interventions as critical components in the advancement of precision medicine, offering promising avenues for diagnosing and treating inflammatory and metabolic disorders.
Collapse
Affiliation(s)
- Emiliano Peña-Durán
- Licenciatura en Médico Cirujano y Partero, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Jesús Jonathan García-Galindo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas II, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Luis Daniel López-Murillo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas I, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Alfredo Huerta-Huerta
- Hospital Medica de la Ciudad, Santa Catalina, Calle. Pablo Valdez 719, La Perla, Guadalajara 44360, Mexico
| | - Luis Ricardo Balleza-Alejandri
- Doctorado en Farmacología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Alberto Beltrán-Ramírez
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas I, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Elsa Janneth Anaya-Ambriz
- Departamento de Ciencias de la Salud, Centro Universitario de los Valles, Universidad de Guadalajara, Ameca 46708, Mexico
| | - Daniel Osmar Suárez-Rico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas II, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), Universidad de Guadalajara, Guadalajara 44430, Mexico
| |
Collapse
|
34
|
An J, Kwon H, Oh SY, Kim YJ. Association between breast cancer risk factors and blood microbiome in patients with breast cancer. Sci Rep 2025; 15:6115. [PMID: 39972005 PMCID: PMC11840066 DOI: 10.1038/s41598-025-90180-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
This study investigated the relationship between risk factors for breast cancer (BC) and the microbiome by comparing the microbiomes of BC patients with fatty liver disease to those with a normal liver. Bacterial extracellular vesicles were collected from each blood sample, and next-generation sequencing was performed. The analysis identified specific microbiome profiles shared among groups with hyperglycaemia, hyperlipidaemia, and high body mass index (BMI), which were then compared with functional biomarkers. In particular, the genus Faecalibacterium was a specific bacterium found in the groups with high concentrations of low-density lipoprotein cholesterol, high BMI, and fatty liver disease. Therefore, when the prognosis of patients with BC was analysed based on Faecalibacterium presence, it was confirmed that patients' prognoses tended to deteriorate. In this study, BC risk factors, such as hyperglycaemia, hyperlipidaemia, fatty liver, and high BMI, were interconnected through the microbiome. This provides insights into how the risk factors for BC are linked and their impact on the microbiome and human health.
Collapse
Affiliation(s)
- Jeongshin An
- Institute of Convergence Medicine Research, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea.
- Department of Surgery, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea.
| | - Hyungju Kwon
- Department of Surgery, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea
| | - Se-Young Oh
- Institute of Convergence Medicine Research, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea
| | - Young Ju Kim
- Department of Obstetrics and Gynecology, Ewha Medical Institute and College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea
| |
Collapse
|
35
|
Nel NH, Marafie A, Bassis CM, Sugino KY, Nzerem A, Knickmeyer RR, McKee KS, Comstock SS. Edinburgh postpartum depression scores are associated with vaginal and gut microbiota in pregnancy. J Affect Disord 2025; 371:22-35. [PMID: 39481687 DOI: 10.1016/j.jad.2024.10.086] [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: 02/15/2024] [Revised: 09/26/2024] [Accepted: 10/20/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND Prenatal and postpartum depression may be influenced by the composition of host associated microbiomes. As such, the objective of this study was to elucidate the relationship between the human gut or vaginal microbiomes in pregnancy with prenatal or postpartum depression. METHODS 140 female participants were recruited at their first prenatal visit and completed the Edinburgh Postnatal Depression Scale (EPDS) to screen for depression and anxiety, in addition the EPDS was completed one month postpartum. Vaginal and stool biospecimens were collected in the third trimester, analyzed using 16S rRNA gene sequencing, and assessed for alpha and beta diversity. Individual taxa differences and clustering using the k-medoids algorithm enabled community state type classification. RESULTS Participants with higher postpartum EPDS scores had higher species richness and lower abundance of L. crispatus in the vaginal microbiota compared to those with lower EPDS scores. Participants with a higher prenatal EPDS score had lower species richness of the gut microbiome. Participants with a vaginal community state type dominated by L. iners had the highest mean prenatal EPDS scores, whereas postpartum EPDS scores were similar regardless of prenatal vaginal state type. LIMITATIONS Our small sample size and participant's self-report bias limits generalizability of results. CONCLUSIONS Depression in the prenatal and postpartum period is associated with the composition and diversity of the gut and vaginal microbiomes in the third trimester of pregnancy. These results provide a foundational understanding of the microbial relationships between maternal health and depression for identifying potential therapeutic treatments.
Collapse
Affiliation(s)
- Nikita H Nel
- Department of Food Science and Human Nutrition, Michigan State University, 204 Trout, 469 Wilson Rd, East Lansing, MI 48824, United States of America
| | - Anfal Marafie
- College of Human Medicine, Michigan State University, United States of America
| | - Christine M Bassis
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, United States of America
| | - Kameron Y Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Adannaya Nzerem
- Department of Food Science and Human Nutrition, Michigan State University, 204 Trout, 469 Wilson Rd, East Lansing, MI 48824, United States of America
| | | | - Kimberly S McKee
- Department of Family Medicine, University of Michigan Medical School, United States of America
| | - Sarah S Comstock
- Department of Food Science and Human Nutrition, Michigan State University, 204 Trout, 469 Wilson Rd, East Lansing, MI 48824, United States of America.
| |
Collapse
|
36
|
Markus MRP, Weiss FU, Hertel J, Weiss S, Rühlemann M, Bang C, Franke A, Völker U, Homuth G, Kocher T, Völzke H, Lerch MM, Ittermann T, Felix SB, Ewert R, Bahls M, Dörr M, Frost F. Lower cardiorespiratory fitness is associated with an altered gut microbiome. The Study of Health in Pomerania (SHIP). Sci Rep 2025; 15:5171. [PMID: 39939328 PMCID: PMC11822121 DOI: 10.1038/s41598-025-88415-4] [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: 05/24/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
Sedentarism is characterized by low levels of physical activity, a risk factor for obesity and cardio-metabolic diseases. It can also adversely affect the composition and diversity of the gut microbiome which may result in harmful consequences for human health. While cardiorespiratory fitness (CRF) is inversely and independently associated with cardiovascular risk factors and diseases and all-cause mortality, the relationship between low CRF and the gut microbiome is not well known. A total of 3,616 individuals from two independent population-based cohorts of the Study of Health in Pomerania (SHIP-START and SHIP-TREND) performed standardized, symptom-limited cardiopulmonary exercise testing (CPET) and had faecal samples collected to determine gut microbiota profiles (16S rRNA gene sequencing). We analysed cross-sectional associations of CRF with the gut microbiome composition controlling for confounding factors. Lower CRF was associated with reduced microbial diversity, loss of beneficial short-chain fatty acid producing bacteria (i.e. Butyricoccus, Coprococcus, unclassified Ruminococcaceae or Lachnospiraceae) and an increase in opportunistic pathogens such as Escherichia/Shigella, or Citrobacter. Decreased cardiorespiratory performance was associated with a gut microbiota pattern that has been previously related to a proinflammatory state. These associations were independent of body weight or glycemic control.
Collapse
Affiliation(s)
- Marcello Ricardo Paulista Markus
- Department of Internal Medicine B, Cardiology, Angiology, Pneumology and Internal Intensive Care Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner site Greifswald, Greifswald, Germany.
- German Center for Diabetes Research (DZD), Partner site Greifswald, Greifswald, Germany.
| | - Frank-Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Johannes Hertel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Malte Rühlemann
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel, Kiel, Germany
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel, Kiel, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Kocher
- Unit of Periodontology, Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner site Greifswald, Greifswald, Germany
- Department of Study of Health in Pomerania/Clinical-Epidemiological Research, Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Till Ittermann
- German Centre for Cardiovascular Research (DZHK), Partner site Greifswald, Greifswald, Germany
- Department of Study of Health in Pomerania/Clinical-Epidemiological Research, Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Burkhard Felix
- Department of Internal Medicine B, Cardiology, Angiology, Pneumology and Internal Intensive Care Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Greifswald, Greifswald, Germany
| | - Ralf Ewert
- Department of Internal Medicine B, Cardiology, Angiology, Pneumology and Internal Intensive Care Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Martin Bahls
- Department of Internal Medicine B, Cardiology, Angiology, Pneumology and Internal Intensive Care Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Greifswald, Greifswald, Germany
| | - Marcus Dörr
- Department of Internal Medicine B, Cardiology, Angiology, Pneumology and Internal Intensive Care Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Greifswald, Greifswald, Germany
| | - Fabian Frost
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
37
|
da Silva LE, Martins DF, de Oliveira MP, Stenier MR, Fernandes BB, Willemann SDS, de Souza G, Vieira WF, Hewitson A, Cidral-Filho FJ, Rezin GT. Photobiomodulation of gut microbiota with low-level laser therapy: a light for treating neuroinflammation. Lasers Med Sci 2025; 40:64. [PMID: 39903307 DOI: 10.1007/s10103-025-04319-9] [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: 08/08/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
The gut microbiota is known to interact with various organs in the body, including the central nervous system, through the gut-brain axis. Intestinal dysbiosis can lead to increased peripheral inflammation and, consequently, affect the brain, resulting in neuroinflammation. Photobiomodulation (PBM) has demonstrated positive regulatory effects on the imbalance of certain body functions, including pain, inflammation, immunity, wound healing, and gut microbiota dysbiosis. Therefore, PBM at the intestinal level could help improve intestinal dysbiosis and reestablish cerebral homeostasis. In this context, this study aimed to conduct a narrative review of the literature on the effects of PBM at the intestinal level on intestinal dysbiosis and neuroinflammation. Overall, the findings highlight that PBM modulates the gut microbiota, suggesting it could serve as a therapy for neurological conditions affecting the gut-brain axis. Future research should focus on further elucidating the molecular mechanisms underlying this therapy.
Collapse
Affiliation(s)
- Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes (Neuroimet), Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Brazil.
| | - Daniel Fernandes Martins
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes (Neuroimet), Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Brazil
| | - Mariella Reinol Stenier
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes (Neuroimet), Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Brazil
| | - Bruna Barros Fernandes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes (Neuroimet), Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Brazil
| | - Stefanny da Silva Willemann
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes (Neuroimet), Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Brazil
| | - Gabriela de Souza
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
| | - Willians Fernando Vieira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Structural and Functional Biology, State University of Campinas, Campinas, Brazil
| | | | - Francisco J Cidral-Filho
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
- Integrative Wellbeing Institute, Orlando, USA
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes (Neuroimet), Graduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Brazil
| |
Collapse
|
38
|
Mondal R, Ritu RB, Kitaoka K, Azahar NM, Moniruzzaman M, Ogata S, Kiyoshige E, Tohara H, Kobayashi Y, Kashihara N, Naito T, Nakashima N, Tamura K, Nishimura K, Viera AJ, Yano Y. Oral microbiome alpha diversity and all-cause, cardiovascular, and non-cardiovascular mortality in US adults: Evidence from the NHANES 2009-2019. Atherosclerosis 2025; 401:119074. [PMID: 39644613 DOI: 10.1016/j.atherosclerosis.2024.119074] [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: 07/15/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND AND AIMS Knowledge about the association between oral microbiome diversity within individuals and cardiovascular disease (CVD) and non-CVD mortality is scarce. Besides, variation by sex and racial and ethnic groups, and the potential mediators of these associations remain unclear. We aimed to investigate the associations of oral microbiome alpha diversity with all-cause, CVD, and non-CVD mortality, and the interaction effects of sex and racial and ethnic groups and potential mediators in the associations. METHODS The National Health and Nutrition Examination Survey (NHANES) is a population-based observational study, conducted periodically in Mexican American, Other Hispanic, Non-Hispanic (NH) White, NH Black, and other racial/ethnic participants. We linked 2009-12 survey data of 8199 adults to the mortality data until 2019. By analyzing RNA gene sequences from oral rinse samples, microbiome alpha diversity within individuals was assessed using operational taxonomic unit (OTU) richness. Potential mediators included obesity, diabetes mellitus, dyslipidemia, hypertension, and periodontitis. Multivariable Cox proportional hazards regression and causal mediation analysis were used. RESULTS Baseline mean ± standard deviation (SD) age was 42.1 ± 15.1 years. Over a median follow-up of 9.1 years, 405 all-cause mortality occurred (CVD, 105; non-CVD, 300). Each 1-SD increment in OTU richness was inversely associated with all-cause mortality (hazard ratio [HR] 0.92, 95 % confidence interval [CI] 0.90-0.95), CVD mortality (HR, 0.92; 95 % CI, 0.90-0.95), and non-CVD mortality (HR, 0.92; 95 % CI, 0.90-0.95). With evidence of significant racial and ethnic groups-interaction (p <0.05), these associations were evident in Mexican American, NH White, and others racial/ethnic participants. None of the potential mediators significantly mediated the associations of OTU richness with all-cause, CVD, and non-CVD mortality. CONCLUSIONS Lower oral microbiome alpha diversity is associated with higher risk for all-cause, CVD, and non-CVD mortality, and the associations are varied by racial and ethnic groups.
Collapse
Affiliation(s)
- Rajib Mondal
- Department of Preventive Medicine, NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan; Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Rani Baroi Ritu
- Department of Preventive Medicine, NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Kaori Kitaoka
- Department of Advanced Epidemiology, NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Nazar Mohd Azahar
- NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan; Faculty of Health Sciences, Universiti Teknologi MARA, Cawangan Pulau Pinang, Kampus Bertam, Pulau Pinang, Malaysia
| | - Mohammad Moniruzzaman
- NCD Epidemiology Research Center, Shiga University of Medical Science, Shiga, Japan; Socio-Spatial Determinants of Health (SSDH) Laboratory, Population and Community Health Sciences Branch, Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Soshiro Ogata
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Eri Kiyoshige
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Haruka Tohara
- Department of Dysphagia Rehabilitation, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Yusuke Kobayashi
- YCU Co-Creation Innovation Center, Yokohama City University, Yokohama, Japan
| | | | - Toshio Naito
- Department of General Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Naoki Nakashima
- Medical Information Center, Kyushu University Hospital, Japan
| | - Kosuke Tamura
- Socio-Spatial Determinants of Health (SSDH) Laboratory, Population and Community Health Sciences Branch, Division of Intramural Research, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Kunihiro Nishimura
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Anthony J Viera
- Department of Family Medicine and Community Health, Duke University, NC, USA
| | - Yuichiro Yano
- Department of General Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan; Department of Family Medicine and Community Health, Duke University, NC, USA.
| |
Collapse
|
39
|
Luo Y, Li M, Luo D, Tang B. Gut Microbiota: An Important Participant in Childhood Obesity. Adv Nutr 2025; 16:100362. [PMID: 39733798 PMCID: PMC11786877 DOI: 10.1016/j.advnut.2024.100362] [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: 09/04/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024] Open
Abstract
Increasing prevalence of childhood obesity has emerged as a critical global public health concern. Recent studies have challenged the previous belief that obesity was solely a result of excessive caloric intake. Alterations in early-life gut microbiota can contribute to childhood obesity through their influence on nutrient absorption and metabolism, initiation of inflammatory responses, and regulation of gut-brain communication. The gut microbiota is increasingly acknowledged to play a crucial role in human health, as certain beneficial bacteria have been scientifically proven to possess the capacity to reduce body fat content and enhance intestinal barrier function and their metabolic products to exhibit anti-inflammatory effect. Examples of such microbes include bifidobacteria, Akkermansia muciniphila, and Lactobacillus reuteri. In contrast, an increase in Enterobacteriaceae and propionate-producing bacteria (Prevotellaceae and Veillonellaceae) has been implicated in the induction of low-grade systemic inflammation and disturbances in lipid metabolism, which can predispose individuals to obesity. Studies have demonstrated that modulating the gut microbiota through diet, lifestyle changes, prebiotics, probiotics, or fecal microbiota transplantation may contribute to gut homeostasis and the management of obesity and its associated comorbidities. This review aimed to elucidate the impact of alterations in gut microbiota composition during early life on childhood obesity and explores the mechanisms by which gut microbiota contributes to the pathogenesis of obesity and specifically focused on recent advances in using short-chain fatty acids for regulating gut microbiota and ameliorating obesity. Additionally, it aimed to discuss the therapeutic strategies for childhood obesity from the perspective of gut microbiota, aiming to provide a theoretical foundation for interventions targeting pediatric obesity based on gut microbiota.
Collapse
Affiliation(s)
- Yu Luo
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Maojun Li
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dan Luo
- Department of Pediatrics, School of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Binzhi Tang
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Department of Pediatrics, School of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
40
|
Hu Y, Schnabl B, Stärkel P. Origin, Function, and Implications of Intestinal and Hepatic Macrophages in the Pathogenesis of Alcohol-Associated Liver Disease. Cells 2025; 14:207. [PMID: 39936998 PMCID: PMC11816606 DOI: 10.3390/cells14030207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
Macrophages are members of the human innate immune system, and the majority reside in the liver. In recent years, they have been recognized as essential players in the maintenance of liver and intestinal homeostasis as well as key guardians of their respective immune systems, and they are increasingly being recognized as such. Paradoxically, they are also likely involved in chronic pathologies of the gastrointestinal tract and potentially in the alteration of the gut-liver axis in alcohol use disorder (AUD) and alcohol-associated liver disease (ALD). To date, the causal relationship between macrophages, the pathogenesis of ALD, and the immune dysregulation of the gut remains unclear. In this review, we will discuss our current understanding of the heterogeneity of intestinal and hepatic macrophages, their ontogeny, the potential factors that regulate their origin, and the evidence of how they are associated with the manifestation of chronic inflammation. We will also illustrate how the micro-environment of the intestine shapes the phenotypes and functionality of the macrophage compartment in both the intestines and liver and how they change during chronic alcohol abuse. Finally, we highlight the obstacles to current research and the prospects for this field.
Collapse
Affiliation(s)
- Yifan Hu
- Laboratory of Hepato-Gastroenterology, Institute of Clinical and Experimental Research, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA 92161, USA;
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Peter Stärkel
- Laboratory of Hepato-Gastroenterology, Institute of Clinical and Experimental Research, Université Catholique de Louvain, 1200 Brussels, Belgium;
- Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
41
|
Scala M, Tabone M, Paolini M, Salueña A, Iturra RA, Ferreiro VR, Alvarez-Mon MÁ, Serretti A, Soltero MDRG, Rodriguez-Jimenez R. Unlocking the Link Between Gut Microbiota and Psychopathological Insights in Anorexia Nervosa: A Systematic Review. EUROPEAN EATING DISORDERS REVIEW 2025. [PMID: 39887544 DOI: 10.1002/erv.3179] [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: 11/07/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
OBJECTIVE This systematic review explores the associations between qualitative/quantitative changes in gut microbiota and psychopathological symptoms or other clinical features in patients with eating disorders (EDs). Secondary outcomes include exploring gut microbiota changes in EDs and potential relationships with psychotropic drug use. METHOD A systematic search was conducted across biomedical databases from inception to June 2024 according to PRISMA guidelines. The risk of bias was assessed, and a narrative synthesis was performed due to the heterogeneity of the outcomes. RESULTS Only findings related to anorexia nervosa (AN) were identified. Ten studies, of which seven were longitudinal, two cross-sectional, and one interventional (N = 350 patients with AN, and 304 HCs), were included. Despite no clear links between diversity metrics and clinical symptoms being observed, specific taxa belonging to phylum Firmicutes, such as Clostridium, Roseburia, Lactobacillus, Faecalibacterium, and Bifidobacterium belonging to Actinobacteriota correlated with ED psychopathology, including anxiety and depressive symptoms. CONCLUSIONS Changes in microbiota were related to anxiety and depressive symptoms, as well as altered eating behaviours by modulating inflammation and insulin pathways through short-chain fatty acids (SCFAs), that also lead to neurotransmitter imbalances. Further studies are required to replicate these finding and to explore whether similar patterns are observed in other EDs.
Collapse
Affiliation(s)
- Mauro Scala
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy
- Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
- Division of Neuroscience, Health Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Legal Medicine, Pathology, and Psychiatry, Complutense University of Madrid (UCM), Madrid, Spain
| | - Mariangela Tabone
- Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
| | - Marco Paolini
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Salueña
- Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
| | - Rocío Arroyo Iturra
- Department of Legal Medicine, Pathology, and Psychiatry, Complutense University of Madrid (UCM), Madrid, Spain
| | - Veronica Romero Ferreiro
- Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
- Division of Neuroscience, Health Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- CIBERSAM/ISCIII (Biomedical Research Networking Centre in Mental Health), Madrid, Spain
| | - Miguel Ángel Alvarez-Mon
- CIBERSAM/ISCIII (Biomedical Research Networking Centre in Mental Health), Madrid, Spain
- Department of Medicine and Medical Specialities, University of Alcala, Alcala de Henares, Spain
- Department of Psychiatry and Mental Health, Infanta Leonor University Hospital, Madrid, Spain
| | - Alessandro Serretti
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| | - Maria Del Rocío Gonzalez Soltero
- Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
- Molecular Microbiology Group, Health Research Institute of the University Hospital La Paz (IdiPAZ), Hospital Universitario La Paz, Madrid, Spain
| | - Roberto Rodriguez-Jimenez
- Division of Neuroscience, Health Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Legal Medicine, Pathology, and Psychiatry, Complutense University of Madrid (UCM), Madrid, Spain
- CIBERSAM/ISCIII (Biomedical Research Networking Centre in Mental Health), Madrid, Spain
| |
Collapse
|
42
|
Guodong W, Yinhang W, Xinyue W, Hong S, Jian C, Zhanbo Q, Shuwen H. Fecal occult blood affects intestinal microbial community structure in colorectal cancer. BMC Microbiol 2025; 25:34. [PMID: 39833681 PMCID: PMC11745023 DOI: 10.1186/s12866-024-03721-7] [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: 03/17/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Gut microbes have been used to predict CRC risk. Fecal occult blood test (FOBT) has been recommended for population screening of CRC. OBJECTIVE To analyze the effects of fecal occult blood test (FOBT) on gut microbes. METHODS Fecal samples from 107 healthy individuals (FOBT-negative) and 111 CRC patients (39 FOBT-negative and 72 FOBT-positive) were included for 16 S ribosomal RNA sequencing. Based on the results of different FOBT, the community structure and diversity of intestinal bacteria in healthy individuals and CRC patients were analyzed. Characteristic gut bacteria were screened, and various machine learning algorithms were applied to construct CRC risk prediction models. RESULTS The gut microbiota of healthy people and CRC patients with different fecal occult blood were mapped. There was no statistical difference in diversity between CRC patients with negative FOBT and positive FOBT. Bacteroides, Blautia and Escherichia-Shigella were more correlated to healthy individuals, while Streptococcus showed higher correlation with CRC patients with negative FOBT. The accuracy of CRC risk prediction model based on the support vector machines (SVM) algorithm was the highest (89.71%). Subsequently, FOBT was included as a characteristic element in the model construction, and the prediction accuracy of the model was all increased. Similarly, the CRC risk prediction model based on SVM algorithm had the highest accuracy (92%). CONCLUSION FOB affects the community composition of gut microbes. When predicting CRC risk based on gut microbiome, considering the influence of FOBT is expected to improve the accuracy of CRC risk prediction.
Collapse
Affiliation(s)
- Wu Guodong
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Wu Yinhang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Wu Xinyue
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Shen Hong
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China.
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China.
- ASIR (Institute - Association of intelligent systems and robotics), 14B rue Henri Sainte Claire, Deville, Rueil-Malmaison, 92500, France.
| |
Collapse
|
43
|
Koh YC, Hsu HW, Ho PY, Lin WS, Hsu KY, Majeed A, Ho CT, Pan MH. Feruloylacetone and Its Analog Demethoxyferuloylacetone Mitigate Obesity-Related Muscle Atrophy and Insulin Resistance in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:1231-1243. [PMID: 39754576 PMCID: PMC11741112 DOI: 10.1021/acs.jafc.4c07798] [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] [Received: 08/24/2024] [Revised: 12/07/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
Obesity-induced muscle alterations, such as inflammation, metabolic dysregulation, and myosteatosis, lead to a decline in muscle mass and function, often resulting in sarcopenic obesity. Currently, there are no definitive treatments for sarcopenic obesity beyond lifestyle changes and dietary supplementation. Feruloylacetone (FER), a thermal degradation product of curcumin, and its analog demethoxyferuloylacetone (DFER), derived from the thermal degradation of bisdemethoxycurcumin, have shown potential antiobesity effects in previous studies. This study investigates the impact of FER and DFER on obesity-related glucose intolerance and muscle atrophy. High-fat diet (HFD) feeding resulted in muscle mass reduction and increased intramuscular triglyceride accumulation, both of which were mitigated by FER and DFER supplementation. The supplements activated the PI3K/Akt/mTOR signaling pathway, enhanced muscle protein synthesis, and decreased markers of muscle protein degradation. Additionally, FER and DFER supplementation improved glucose homeostasis in HFD-fed mice. The supplements also promoted the formation of a gut microbial consortium comprising Blautia intestinalis, Dubosiella newyorkensis, Faecalicatena fissicatena, Waltera intestinalis, Clostridium viride, and Caproiciproducens galactitolivorans, which contributed to the reduction of obesity-induced chronic inflammation. These findings suggest, for the first time, that FER and DFER may prevent obesity-related complications, including muscle atrophy and insulin resistance, thereby warranting further research into their long-term efficacy and safety.
Collapse
Affiliation(s)
- Yen-Chun Koh
- Institute
of Food Sciences and Technology, National
Taiwan University, 10617 Taipei, Taiwan
| | - Han-Wen Hsu
- Institute
of Food Sciences and Technology, National
Taiwan University, 10617 Taipei, Taiwan
| | - Pin-Yu Ho
- Institute
of Food Sciences and Technology, National
Taiwan University, 10617 Taipei, Taiwan
| | - Wei-Sheng Lin
- Institute
of Food Sciences and Technology, National
Taiwan University, 10617 Taipei, Taiwan
- Department
of Food Science, National Quemoy University, 89250 Quemoy, Taiwan
| | - Kai-Yu Hsu
- Institute
of Food Sciences and Technology, National
Taiwan University, 10617 Taipei, Taiwan
| | - Anju Majeed
- Sami-Sabinsa
Group Limited, Bengaluru 560058, Karnataka, India
| | - Chi-Tang Ho
- Department
of Food Science, Rutgers University, New Brunswick 08901, New Jersey, United
States
| | - Min-Hsiung Pan
- Institute
of Food Sciences and Technology, National
Taiwan University, 10617 Taipei, Taiwan
- Department
of Medical Research, China Medical University Hospital, China Medical University, 40402 Taichung, Taiwan
| |
Collapse
|
44
|
Liu Z, Wang M, Hu Y, Li J, Gong W, Guo X, Song S, Zhu B. Ulva lactuca polysaccharides combined with fecal microbiota transplantation ameliorated dextran sodium sulfate-induced colitis in C57BL/6J mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:422-432. [PMID: 39212113 DOI: 10.1002/jsfa.13839] [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] [Received: 03/22/2024] [Revised: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) of healthy donors improves ulcerative colitis (UC) patients by restoring the balance of the gut microbiota. However, donors vary in microbial diversity and composition, often resulting in weak or even ineffective FMT. Improving the efficacy of FMT through combination treatment has become a promising strategy. Ulva lactuca polysaccharides (ULP) have been found to benefit host health by regulating gut microbiota. The effect of the combination of ULP and FMT in ameliorating UC has not yet been evaluated. RESULTS The present study found that supplementation with ULP combined with FMT showed better effects in ameliorating UC than supplementation with FMT alone. Results suggested that FMT or ULP combined with FMT alleviated the symptoms of UC in mice, as evidenced by prevention of body weight loss, improvement of disease activity index and protection of the intestinal mucus. Notably, ULP in combination with FMT was more effective than FMT in reducing levels of cytokines and related inflammatory enzymes. In addition, ULP combined with FMT effectively restored the dysbiosis induced by dextran sulfate sodium (DSS) and further enriched probiotics (such as Bifidobacterium). The production of short-chain fatty acids, especially acetic acid, was also significantly enriched by ULP combined with FMT. CONCLUSION Supplementation of ULP combined with FMT could significantly ameliorate DSS-induced colitis in mice by inhibiting inflammation and restoring dysbiosis of gut microbiota. These results suggested that ULP combined with FMT has potential application in ameliorating UC. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhengqi Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
- National Engineering Research Center of Seafood, National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, School of Food Science and Technology, Dalian Polytechnic University, Dalian, PR China
| | - Menghui Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Yuanyuan Hu
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Jinjin Li
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Wei Gong
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Xiaoming Guo
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Shuang Song
- National Engineering Research Center of Seafood, National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, School of Food Science and Technology, Dalian Polytechnic University, Dalian, PR China
| | - Beiwei Zhu
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
- National Engineering Research Center of Seafood, National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, School of Food Science and Technology, Dalian Polytechnic University, Dalian, PR China
| |
Collapse
|
45
|
Xu X, Zhang H, Meng K, Cai H, Liu W, Song L, Zhang Z, Zhu Q, Han X, Han Y, Yang P. Limosilactobacillus reuteri ZY15 Alleviates Intestinal Inflammation and Barrier Dysfunction via AKT/mTOR/HIF-1α/RORγt/IL-17 Signaling and the Gut Microbiota in ETEC K88-Challenged Mice. Antioxidants (Basel) 2025; 14:58. [PMID: 39857392 PMCID: PMC11763039 DOI: 10.3390/antiox14010058] [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: 11/22/2024] [Revised: 12/27/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Limosilactobacillus reuteri, a recognized probiotic, improves intestinal health in animals, but the mechanism remains unclear. This study investigates the mechanisms by which L. reuteri ZY15, isolated from healthy pig feces, mitigates intestinal barrier damage and inflammation caused by oxidative stress in Enterotoxigenic Escherichia coli (ETEC) K88-challenged mice. The results indicated that L. reuteri ZY15 increased antioxidant capacity by reducing serum reactive oxygen species (ROS) and superoxide dismutase (SOD) levels. L. reuteri ZY15 enhanced the intestinal barrier by upregulating mucin 1, mucin 2, occludin, zonula occludens-1 (ZO-1), and claudin-1 expressions in protein and mRNA levels. It significantly alleviated intestinal inflammation by reducing the proinflammatory cytokines interleukin-1β (IL-1β), interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and interleukin-17 (IL-17) mRNA and protein levels. Notably, L. reuteri ZY15 suppressed intestinal inflammation by inhibiting AKT/mTOR/HIF-1α/RORγt/IL-17 pathway activation. Additionally, it significantly altered the structure of gut microorganisms by enriching Akkermansia and Clostridia_UCG.014, and thereby re-establishing colonization resistance and alleviating ETEC K88-induced intestinal barrier damage and inflammation in mice. Taken together, our findings reveal the protective mechanism of L. reuteri ZY15 in mice challenged with ETEC K88 by regulating AKT/mTOR/HIF-1α/RORγt/IL-17 signaling and microbial imbalance. Leveraging these properties, live L. reuteri ZY15 offers a promising alternative treatment for Escherichia coli-induced diarrhea in weaned piglets.
Collapse
Affiliation(s)
- Xin Xu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Hongwei Zhang
- Chengde Academy of Agriculture and Forestry Sciences, Chengde 067000, China;
| | - Kun Meng
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Hongying Cai
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Weiwei Liu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Liye Song
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Zihan Zhang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Qijun Zhu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Xiling Han
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Yunsheng Han
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| | - Peilong Yang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.X.); (K.M.); (H.C.); (W.L.); (L.S.); (Z.Z.); (Q.Z.); (X.H.)
| |
Collapse
|
46
|
Li S, Ma X, Mei H, Chang X, He P, Sun L, Xiao H, Wang S, Li R. Association between gut microbiota and short-chain fatty acids in children with obesity. Sci Rep 2025; 15:483. [PMID: 39748068 PMCID: PMC11695941 DOI: 10.1038/s41598-024-84207-4] [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: 03/19/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
The gut microbiome and its metabolites may be important role in regulating the pathogenesis of obesity. This study aimed to characterize the gut microbiome and short-chain fatty acid (SCFA) metabolome in obese children. This case-control study recruited children aged 7‒14 years and divided them into a normal group (NG) and an obese group (OG) based on their body mass index. Whole-genome shotgun metagenomic analysis was performed on fecal samples from the OG and NG groups to characterize the signatures and functional potential of the gut microbiota. Serum metabolite profiles were analyzed using high-performance liquid chromatography/mass spectrometry (LC/MS). The Statistical Package for the Social Sciences (SPSS, version 26) and R software were used for data analysis. A total of 99 children were recruited, with 49 in the OG and 50 in the NG. At the phylum level, Proteobacteria were significantly more abundant in children in the OG than those in the NG. At the genus level, Oscillibacter and Alistipes were significantly lower in children in the OG than those in the NG. Caproate levels significantly increased, whereas butyrate and isobutyrate levels decreased in children in the OG than those in the NG. Kyoto encyclopedia of genes and genomes (KEGG) functional analysis revealed 28 enriched KEGG pathways, of which/with the phosphotransferase system (PTS) and enhanced biofilm formation by Escherichia coli were particularly significant in the OG. Spearman's correlation analysis indicated that the genus Oscillibacter and species Clostridium_sp._CAG:302 connect serum metabolites and the gut microbiota in childhood obesity. Childhood obesity is correlated with the symbiotic status of the gut microbiota. The microbiota influences human metabolism via specific pathways, particularly butyrate, caproate, and the genus Oscillibacter, all closely associated with obesity.
Collapse
Affiliation(s)
- Shihan Li
- Department of Child Healthcare, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, 100 Hongkong Road, Wuhan, 430016, Hubei, China
| | - Xinyu Ma
- Department of Radiology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Mei
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuening Chang
- Department of Child Healthcare, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, 100 Hongkong Road, Wuhan, 430016, Hubei, China
| | - Peiling He
- Department of Child Healthcare, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, 100 Hongkong Road, Wuhan, 430016, Hubei, China
| | - Lingli Sun
- Department of Child Healthcare, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, 100 Hongkong Road, Wuhan, 430016, Hubei, China
| | - Han Xiao
- Department of Child Healthcare, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, 100 Hongkong Road, Wuhan, 430016, Hubei, China.
| | - Shiqiong Wang
- Department of Child Healthcare, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, 100 Hongkong Road, Wuhan, 430016, Hubei, China.
| | - Ruizhen Li
- Department of Child Healthcare, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, 100 Hongkong Road, Wuhan, 430016, Hubei, China.
| |
Collapse
|
47
|
Wu R, Hong G, Cheng X, Zhu Y. The association between use of proton pump inhibitors and frailty index among middle-aged and older adults. Br J Clin Pharmacol 2025; 91:210-219. [PMID: 39305011 DOI: 10.1111/bcp.16260] [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: 05/31/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 12/28/2024] Open
Abstract
AIMS This study aimed to investigate the association between use of proton pump inhibitors (PPI) and frailty index (FI), and to assess the causality relationship using Mendelian randomization (MR). METHODS A total of 9756 middle-aged and older adults from the National Health and Nutrition Examination Survey were included. The FI was evaluated using a previously validated 49-item deficit model to assess frailty status, which is one of the common approaches to measure overall health burden. We performed weighted multivariable-adjusted linear regression to assess the association between PPI use and FI, and conducted a two-sample MR to evaluate causality, employing various sensitivity analyses for robustness. The inverse variance weighted (IVW) method was used as the primary analysis. RESULTS Multiple linear regression analysis revealed a positive association between PPI use and FI (β = 0.048, 95% confidence interval [CI]: 0.042-0.054, P < .001). This association was observed in both short-term (≤ 1 year) and long-term (> 1 year) PPI users (P for trend < 0.001). The MR study also revealed a positive association between PPI use and FI based on the IVW method (β = 1.183, 95% CI: 0.474-1.892, P = .001). CONCLUSIONS While our findings suggest a potential link between PPI use and FI, they should be interpreted with caution due to the study's limitations. Although the MR analysis suggests a causal relationship, further research, particularly longitudinal studies, is needed to confirm these findings and better establish temporality.
Collapse
Affiliation(s)
- Rui Wu
- Hefei Ion Medical Center, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Guojun Hong
- Department of Pharmacy, Nanjing Gaochun People's Hospital, Nanjing, Jiangsu Province, China
| | - Xiankun Cheng
- Department of Pharmacy, Maanshan People's Hospital, Maanshan, Anhui Province, China
| | - Yue Zhu
- Hefei Ion Medical Center, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
48
|
Darvishi M, Rafsanjani SMRH, Nouri M, Abbaszadeh S, Heidari-Soureshjani S, Kasiri K, Rahimian G. Biological Mechanisms of Polyphenols against Clostridium Difficile: A Systematic Review. Infect Disord Drug Targets 2025; 25:e18715265313944. [PMID: 39234903 DOI: 10.2174/0118715265313944240726115600] [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: 03/02/2024] [Revised: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Clostridium difficile is an opportunistic infection that can lead to antibiotic- associated diarrhea and toxic megacolon. OBJECTIVE This systematic review study aimed to investigate polyphenols' antibacterial and antitoxin properties and their effects on reducing complications related to C. difficile Infections (CDI). METHODS This systematic review was conducted following the PRISMA guideline 2020. Multiple databases, including Web of Science, PubMed, Cochrane Library, EMBASE, and Scopus, were searched thoroughly for existing literature. After considering the inclusion and exclusion criteria for the review, 18 articles were included. Data were collected and registered into an Excel file for further investigations and conclusions. RESULTS Polyphenols by reducing Reactive Oxygen Species (ROS) levels, increasing inflammatory factor Interleukin 10 (IL-10), reducing Nuclear Factor kappa B (NF-κB) and Tumour Necrosis Factor- α (TNF-α), IL-6, IL-1α, IL-1β, Granulocyte Colony-stimulating Factor (G-CSF), and Monocyte Chemoattractant Protein-1 (MCP-1) and Macrophage Inflammatory Protein-1 alpha (MIP-1α) levels, and regulating the expression of Bcl-2 and Bax, make the growth and replication conditions of C. difficile more difficult and prevent it from producing toxins. Furthermore, polyphenols can exhibit prebiotic properties, promoting the growth of beneficial Bifidobacterium and Lactobacillus species and consequently regulating gut microbiota, exerting antimicrobial activities against C. difficile. They also induce their beneficial effects by inhibiting the production of C. difficile TcdA and TcdB. CONCLUSION Polyphenols have been reported to inhibit C. difficile growth and toxin production by several mechanisms in preclinical studies. However, more clinical studies are needed to investigate their safety in humans.
Collapse
Affiliation(s)
- Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), School of Aerospace and Subaquatic Medicine, Aja University of Medical Sciences, Tehran, Iran
| | | | - Majid Nouri
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), Aja University of Medical Sciences, Tehran, Iran
| | - Saber Abbaszadeh
- Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Karamali Kasiri
- Department of Pediatrics, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ghorbanali Rahimian
- Department of Internal Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
49
|
Hairul Hisham HI, Lim SM, Neoh CF, Abdul Majeed AB, Shahar S, Ramasamy K. Effects of non-pharmacological interventions on gut microbiota and intestinal permeability in older adults: A systematic review: Non-pharmacological interventions on gut microbiota/barrier. Arch Gerontol Geriatr 2025; 128:105640. [PMID: 39305569 DOI: 10.1016/j.archger.2024.105640] [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: 06/23/2024] [Revised: 08/21/2024] [Accepted: 09/13/2024] [Indexed: 11/03/2024]
Abstract
This systematic review appraised previous findings of non-pharmacological interventions on gut microbiota and/ or intestinal permeability in older adults. A literature search was performed using PubMed, Scopus, ScienceDirect and the Cochrane Library. Relevant studies were shortlisted based on the inclusion and exclusion criteria, and evaluated for risks of bias using the "Cochrane Collaboration's Risk of Bias 2" and the "NIH Quality Assessment Tool for Before-After (Pre-Post) Studies with No Control Group". The primary outcomes were the effects of non-pharmacological interventions on gut microbiota diversity and composition, and intestinal permeability in older adults. Out of 85,114 studies, 38 were shortlisted. Generally, the non-pharmacological interventions were beneficial against dysbiosis and the leaky gut in older adults. Considering specific interventions with two or more studies that reported consistent outcomes, a pattern was observed amongst the Mediterranean diet (MD), polyphenol-rich (PR) diet and supplements (i.e., probiotics, prebiotics and synbiotics). As for the other interventions, the very few studies that have been conducted did not allow a strong conclusion to be made just yet. The MD (single and multidomain interventions) restored gut microbiota by increasing species richness (alpha diversity) and reduced intestinal permeability (zonulin) and inflammation (CRP). The PR diet only showed slight changes in the gut microbiota but improved the gut barrier by reducing zonulin, CRP and IL-6. Probiotics, prebiotics and synbiotics increased the genus Bifidobacterium spp. which are considered beneficial bacteria. This review has uncovered insights into the relationship between gut microbiota and intestinal epithelial barriers of specific non-pharmacological interventions in older adults.
Collapse
Affiliation(s)
- Hazwanie Iliana Hairul Hisham
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Chin Fen Neoh
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Abu Bakar Abdul Majeed
- Brain Degeneration and Therapeutics Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| | - Suzana Shahar
- Centre of Healthy Aging and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, 50300 Kuala Lumpur, Malaysia
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
50
|
Tursi A, Mocci G, Satta PU, Elisei W. Impact of a Symbiotic Mixture on Moderate-to-severe Diverticular Disease of the Colon. Rev Recent Clin Trials 2025; 20:27-35. [PMID: 39051586 DOI: 10.2174/0115748871308652240712101604] [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: 03/04/2024] [Revised: 05/08/2024] [Accepted: 06/12/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Microbial imbalance is thought to play a role in the pathogenesis of Diverticular Disease (DD). OBJECTIVE We aimed to assess the efficacy of a symbiotic mixture (Prolactis GG Plus®) in the treatment of moderate to severe DD, scored according to the Diverticular Inflammation and Complication Assessment (DICA) classification. METHODS A retrospective study was conducted enrolling the following patients: at the first diagnosis of DD; in whom DD was diagnosed with colonoscopy and scored according to DICA classification; treated with Prolactis GG Plus® two times/daily for 2 consecutive months; in whom the severity of the abdominal pain was scored with a 10-points visual-analogue scale (VAS) at baseline and the end of follow-up; in whom fecal calprotectin (FC) was assessed at baseline and the end of follow-up as μg/g. RESULTS Twenty-four patients were identified (10 males, 14 females; 16 as DICA 2, and 8 as DICA 3). Prolactis GG Plus® decreased the severity of abdominal pain both in DICA 2 (p =0.02) and DICA 3 patients (p =0.01), while FC decreased significantly in DICA 2 (p <0.02) but not in DICA 3 (p =0.123) patients. Acute diverticulitis occurred during the follow-up in two DICA 3 patients but none DICA 2 patients. Add-on therapy was required by eight DICA 2 (50%) and six DICA 3 patients (75%). CONCLUSION In newly diagnosed patients with DD, the symbiotic mixture Prolactis GG Plus® can be a potential treatment for moderate (DICA 2) DD as a single treatment.
Collapse
Affiliation(s)
- Antonio Tursi
- Department of Territorial Gastroenterology Service, ASL BAT, Andria (BT), Italy
- Department of Medical and Surgical Sciences, School of Medicine, Catholic University, Rome, Italy
| | - Giammarco Mocci
- Division of Gastroenterology, "Brotzu" Hospital, Cagliari, Italy
| | - Paolo Usai Satta
- Division of Gastroenterology, "Brotzu" Hospital, Cagliari, Italy
| | - Walter Elisei
- Division of Gastroenterology, "S. Camillo" Hospital, Rome, Italy
| |
Collapse
|