1
|
Shibahara T, Temizoz B, Egashira S, Hosomi K, Park J, Surucu N, Björk A, Sag E, Doi T, Kisla Ekinci RM, Balci S, Versnel MA, Kunisawa J, Yamamoto M, Hayashi T, Ito S, Kamiyama Y, Kobiyama K, Katsikis PD, Coban C, Gursel M, Ozen S, Nishida S, Kumanogoh A, Ishii KJ. Microbial dysbiosis fuels STING-driven autoinflammation through cyclic dinucleotides. J Autoimmun 2025; 154:103434. [PMID: 40334619 DOI: 10.1016/j.jaut.2025.103434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/17/2025] [Accepted: 05/01/2025] [Indexed: 05/09/2025]
Abstract
Aberrant activation of the stimulator of interferon genes (STING) pathway is a hallmark of autoinflammatory disorders such as STING-associated vasculopathy with onset in infancy (SAVI), characterized by systemic inflammation affecting blood vessels, skin, and lungs. Despite its clinical significance, the mechanisms linking STING activation to disease pathology remain poorly defined. In this study, we demonstrated that SAVI mice harboring the N153S STING mutation exhibit diverse disease phenotypes, with a subset developing severe colitis and diarrhea alongside exacerbated systemic inflammation. These diarrheal SAVI mice showed pronounced dysbiosis, marked by reduced short-chain fatty acid-producing bacteria and an enrichment of segmented filamentous bacteria. This microbial imbalance was accompanied by elevated levels of both microbial and host-derived cyclic dinucleotides (CDNs), potent activators of the STING pathway. Notably, antibiotic treatment ameliorated inflammation, underscoring the role of dysbiosis in driving STING-mediated autoinflammation. Furthermore, in SAVI patients, elevated systemic microbial and host-derived CDNs were observed. In conditions such as systemic lupus erythematosus (SLE)-a heterogeneous autoimmune disease with potential STING involvement-systemic microbial CDNs were significantly correlated with disease biomarkers, including type I interferon scores and anti-dsDNA antibodies. In contrast, no such correlations were observed in STING-independent conditions like rheumatoid arthritis (RA). Importantly, this study highlights that both microbial and host-derived CDNs are key drivers of STING activation, suggesting that personalized treatment strategies could target cGAS or the microbiome based on a patient's specific CDN profile. These findings position systemic CDNs as valuable biomarkers and therapeutic targets for STING-driven diseases.
Collapse
Affiliation(s)
- Takayuki Shibahara
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Burcu Temizoz
- Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan; Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center (VDesC), The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo, Japan
| | - Shiori Egashira
- Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Jonguk Park
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Naz Surucu
- Department of Biological Sciences, Middle East Technical University (METU), Ankara, Turkey
| | - Albin Björk
- Division of Rheumatology, Department of Medicine, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden; Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Erdal Sag
- Department of Pediatric Rheumatology, Hacettepe University, Ankara, Turkey
| | - Takehiko Doi
- Department of Pediatrics, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | | | - Sibel Balci
- Department of Pediatric Rheumatology, Cukurova University, Adana, Turkey
| | - Marjan A Versnel
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Division of Infectious Disease, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tomoya Hayashi
- Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan; Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center (VDesC), The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo, Japan
| | - Shuichi Ito
- Department of Pediatrics, School of Medicine, Yokohama City University, Kanagawa, Japan
| | - Yuji Kamiyama
- Department of Pediatrics, School of Medicine, Yokohama City University, Kanagawa, Japan
| | - Kouji Kobiyama
- Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan; Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center (VDesC), The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo, Japan
| | - Peter D Katsikis
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Cevayir Coban
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Immunopathology, WPI, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan; International Vaccine Design Center (VDesC), The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo, Japan
| | - Mayda Gursel
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Seza Ozen
- Department of Pediatric Rheumatology, Hacettepe University, Ankara, Turkey
| | - Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Immunopathology, WPI, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
| | - Ken J Ishii
- Laboratory of Mockup Vaccine, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan; Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; International Vaccine Design Center (VDesC), The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
2
|
Sessa L, Malavolta E, Sodero G, Cipolla C, Rigante D. The conspiring role of gut microbiota as primer of autoimmune thyroid diseases: A scoping focus. Autoimmun Rev 2025; 24:103780. [PMID: 39971108 DOI: 10.1016/j.autrev.2025.103780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
The thyroid gland is the body's largest single organ specialized for endocrine hormone production, and still unraveled mechanisms regulate its interaction between the hypothalamic-pituitary-thyroid axis and composition of the gut microbiota: in particular, a disrupted integrity of the intestinal barrier, causing dysbiosis and increasing detrimental substances or reducing beneficial metabolites, such as short-chain fatty acids (SCFAs) with proinflammatory effects, may be crucial for the induction of an autoimmune thyroid disease. More specifically, Lactobacilli and Bifidobacteria have a role in this partnership through a "molecular mimicry" mechanism, as their protein sequences share structural similarity with thyroid peroxidase and thyroglobulin. Lactobacilli can also increase T helper 17 cells, modifying the number of colonic regulatory T cells, largely implicated in the maintenance of immunological tolerance at the gut barrier. Additionally, Blautia and Anaerostipes work beneficially with butyric acid, one of the SCFAs, promoting antimicrobial peptide synthesis from the intestinal cells and bolstering the innate immune system's ability to struggle against pathogens, which can also influence thyroid hormone levels by regulating iodine uptake and metabolism. This review aims to summarize the current knowledge about the contribution of gut microbiota changes in triggering immune abnormalities leading to autoimmune thyroid diseases.
Collapse
Affiliation(s)
- Linda Sessa
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elena Malavolta
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Sodero
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Clelia Cipolla
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Donato Rigante
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Università Cattolica Sacro Cuore, Rome, Italy.
| |
Collapse
|
3
|
Yang B, Xu Y, Zhang W, Zhu D, Huang B, Yang Y, Jia X, Feng L. Oral absorption mechanisms of polysaccharides and potential as carriers for the construction of nano-delivery systems: A review. Int J Biol Macromol 2025; 310:143184. [PMID: 40253019 DOI: 10.1016/j.ijbiomac.2025.143184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Polysaccharides have garnered increasing attention in recent years for their potential in oral drug delivery within biomaterials and pharmaceuticals, owing to their excellent physicochemical properties, bioactivity, and low toxicity. However, the absorption of polysaccharides encounters multiple challenges posed by the biological, chemical, mechanical, and immune barriers of the intestinal mucosa. Therefore, elucidating the mechanisms by which polysaccharides traverse the intestinal mucosa for oral absorption is essential for their further development and application. Current studies have identified several polysaccharide absorption pathways, including transcellular transport, paracellular transport, M cell and Peyer's patches mediated transport, and intestinal flora mediated transport. Furthermore, numerous studies have demonstrated that polysaccharides can enhance the solubility, gastrointestinal stability, and permeability of small molecule components, which significantly improves their bioavailability. More importantly, nano-delivery systems utilizing polysaccharides as carriers have shown great promise in enhancing the targeting of small molecule components, thereby opening new avenues for drug delivery applications. We hope this review will provide theoretical support and inspiration for a deeper understanding of oral absorption mechanisms and the potential of polysaccharides in the development of nano-delivery systems.
Collapse
Affiliation(s)
- Bing Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Jiangning Hospital of Chinese Medicine, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yan Xu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Weiye Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Dandan Zhu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Bin Huang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yanjun Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Liang Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Jiangning Hospital of Chinese Medicine, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
4
|
Xu R, Yu Y, Chen T. Exploring the dark side of probiotics to pursue light: Intrinsic and extrinsic risks to be opportunistic pathogens. Curr Res Food Sci 2025; 10:101044. [PMID: 40235735 PMCID: PMC11999689 DOI: 10.1016/j.crfs.2025.101044] [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: 01/26/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Probiotics, live microorganisms with multiple health benefits, have gained popularity for their roles in maintaining daily health and treating a variety of diseases. However, they have the potential to be opportunistic pathogens in some conditions. This review delves into the intrinsic and extrinsic risks associated with probiotics. Intrinsic risks involve the production of harmful substances, such as toxins and invasive factors, biofilm formation, bacteria emboli, antibiotic resistance with relevant genetic materials, genetic plasticity, and metabolic issues, while extrinsic risks include problems in regulatory oversight and public awareness, host health status and appropriately administration. It emphasizes the need for a balanced view of their therapeutic benefits and potential hazards, advocating for further research to understand the complex interactions between probiotics and the human microbiome, to optimize the safety and efficacy of probiotics.
Collapse
Affiliation(s)
- Ruiyan Xu
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yifeng Yu
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tingtao Chen
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
5
|
Młynarska E, Barszcz E, Budny E, Gajewska A, Kopeć K, Wasiak J, Rysz J, Franczyk B. The Gut-Brain-Microbiota Connection and Its Role in Autism Spectrum Disorders. Nutrients 2025; 17:1135. [PMID: 40218893 PMCID: PMC11990867 DOI: 10.3390/nu17071135] [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: 01/16/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Autism spectrum disorder (ASD) is a group of complex neurodevelopmental conditions with a heterogeneous and multifactorial etiology that is not yet fully understood. Among the various factors that may contribute to ASD development, alterations in the gut microbiota have been increasingly recognized. Microorganisms in the gastrointestinal tract play a crucial role in the gut-brain axis (GBA), affecting nervous system development and behavior. Dysbiosis, or an imbalance in the microbiota, has been linked to both behavioral and gastrointestinal (GI) symptoms in individuals with ASD. The microbiota interacts with the central nervous system through mechanisms such as the production of short-chain fatty acids (SCFAs), the regulation of neurotransmitters, and immune system modulation. Alterations in its composition, including reduced diversity or an overabundance of specific bacterial taxa, have been associated with the severity of ASD symptoms. Dietary modifications, such as gluten-free or antioxidant-rich diets, have shown potential for improving gut health and alleviating behavioral symptoms. Probiotics, with their anti-inflammatory properties, may support neural health and reduce neuroinflammation. Fecal microbiota transplantation (FMT) is being considered, particularly for individuals with persistent GI symptoms. It has shown promising outcomes in enhancing microbial diversity and mitigating GI and behavioral symptoms. However, its limitations should be considered, as discussed in this narrative review. Further research is essential to better understand the long-term effects and safety of these therapies. Emphasizing the importance of patient stratification and phenotype characterization is crucial for developing personalized treatment strategies that account for individual microbiota profiles, genetic predispositions, and coexisting conditions. This approach could lead to more effective interventions for individuals with ASD. Recent findings suggest that gut microbiota may play a key role in innovative therapeutic approaches to ASD management.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Barszcz
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Emilian Budny
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Kacper Kopeć
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
6
|
Huang HJ, Liu Y, Li DW, Wang X, Feng NX, Li HY, Mo CH, Yang WD. Polystyrene Microplastics Can Aggravate the Damage of the Intestinal Microenvironment Caused by Okadaic Acid: A Prevalent Algal Toxin. Mar Drugs 2025; 23:129. [PMID: 40137315 PMCID: PMC11943709 DOI: 10.3390/md23030129] [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: 02/05/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
As emerging contaminants, microplastics (MPs) may pose a threat to human health. Their co-exposure with the widespread phycotoxin okadaic acid (OA), a marine toxin known to cause gastrointestinal toxicity, may exacerbate health risk and raise public safety concern. In this study, the toxicity mechanisms of MPs and OA on intestinal microenvironment was explored using human Caco-2 cells as the model, which was combined with an in vitro fecal fermentation experiment. Our results showed that co-exposure to MPs (80 μg/mL) and OA (20 ng/mL) significantly decreased cell viability, increased intracellular reactive oxygen species (ROS) production, elevated lactate dehydrogenase release, impaired ABC transporter activity, promoted OA accumulation, and triggered inflammatory response compared to the control, MPs, and OA groups, indicating that co-exposure directly compromises intestinal epithelial integrity. In vitro fermentation experiments revealed that co-exposure disrupted gut microbial composition, decreasing the relative abundance of some bacteria, such as Parasutterella and Adlercreutzia, while increasing opportunistic pathogens, such as Escherichia-Shigella, increased. These findings provide new insights into the impact and underlying mechanisms of MPs and OA co-exposure on intestinal homeostasis, highlighting the potential health risks associated with MPs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ce-Hui Mo
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (H.-J.H.)
| | - Wei-Dong Yang
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (H.-J.H.)
| |
Collapse
|
7
|
Pardali EC, Gkouvi A, Gkouskou KK, Manolakis AC, Tsigalou C, Goulis DG, Bogdanos DP, Grammatikopoulou MG. Autoimmune protocol diet: A personalized elimination diet for patients with autoimmune diseases. Metabol Open 2025; 25:100342. [PMID: 39850611 PMCID: PMC11755016 DOI: 10.1016/j.metop.2024.100342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/25/2025] Open
Abstract
The autoimmune protocol diet (AIP) is a personalized elimination diet that aims to determine and exclude the foods that might trigger immune responses, leading to inflammation and symptomatology associated with autoimmune diseases. Focusing on gut health and the importance of the gut microbiome in immune regulation and overall well-being, the AIP starts by eliminating foods that might create negative effects on the patients and continues by developing a personalized and tailored diet plan for them. This comprehensive approach aims to mitigate symptoms and improve quality of life of individuals with autoimmune conditions. This review presents and critically appraises current knowledge on the AIP protocol, highlight any oversights, and discuss findings from relevant clinical trials.
Collapse
Affiliation(s)
- Eleni C. Pardali
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | - Arriana Gkouvi
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | - Kalliopi K. Gkouskou
- Department of Biology, Medical School, National and Kapodistrian University of Athens, Goudi Campus, Athens, Greece
- GENOSOPHY P.C., Athens, Greece
| | | | - Christina Tsigalou
- Laboratory of Hygiene and Environmental Protection, Medical School, Democritus University of Thrace, University Hospital, Alexandroupolis, Greece
| | - Dimitrios G. Goulis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios P. Bogdanos
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | - Maria G. Grammatikopoulou
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
8
|
Parhizkar E, Vosough P, Baneshi M, Keshavarzi A, Lohrasbi P, Taghizadeh S, Savardashtaki A. Probiotics and gut microbiota modulation: implications for skin health and disease management. Arch Microbiol 2025; 207:68. [PMID: 39988585 DOI: 10.1007/s00203-025-04267-6] [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: 09/23/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
The gut microbiota, consisting of a varied population of microorganisms in the digestive tract, is essential for sustaining overall human health, encompassing skin health. This review explored the intricate relationship between gut microbiota and various skin disorders, investigating the pathways through which gut dysbiosis may have impacted the development and progression of these conditions. We focused on the impact of gut microbiota on atopic dermatitis, psoriasis, acne vulgaris, acne rosacea, and melanoma. The review highlighted the potential of probiotics as a therapeutic strategy for modulating gut microbiota composition and, consequently, improving skin health. We discussed the evidence supporting the use of probiotics in managing these skin disorders and explored the mechanisms by which probiotics delivered their positive effects. Finally, we discussed the potential role of gut microbiota in other skin diseases, emphasizing the need for further research to unravel the complex interplay between the gut and the skin. Significant gaps remain in understanding the gut-skin axis, how microbial interactions contribute to skin disorders, and how to effectively manipulate the microbiome for therapeutic purposes. This review provided extensive research on the gut-skin axis, highlighting the promising prospects of modulating gut microbiota as a therapeutic strategy for various dermatological conditions.
Collapse
Affiliation(s)
- Elahe Parhizkar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Baneshi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Parvin Lohrasbi
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Holt BM, Stine JM, Beardslee LA, Ayansola H, Jin Y, Pasricha PJ, Ghodssi R. An ingestible bioimpedance sensing device for wireless monitoring of epithelial barriers. MICROSYSTEMS & NANOENGINEERING 2025; 11:24. [PMID: 39915452 PMCID: PMC11802857 DOI: 10.1038/s41378-025-00877-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/03/2024] [Accepted: 01/04/2025] [Indexed: 02/09/2025]
Abstract
Existing gastrointestinal (GI) diagnostic tools are unable to non-invasively monitor mucosal tight junction integrity in vivo beyond the esophagus. In the GI tract, local inflammatory processes induce alterations in tight junction proteins, enhancing paracellular ion permeability. Although transepithelial electrical resistance (TEER) may be used in the laboratory to assess mucosal barrier integrity, there are no existing methodologies for characterizing tight junction dilation in vivo. Addressing this technology gap, intraluminal bioimpedance sensing may be employed as a localized, non-invasive surrogate to TEER electrodes used in cell cultures. Thus far, bioimpedance has only been implemented in esophagogastroduodenoscopy (EGD) due to the need for external electronics connections. In this work, we develop a novel, noise-resilient Bluetooth-enabled ingestible device for the continuous, non-invasive measurement of intestinal mucosal "leakiness." As a proof-of-concept, we validate wireless impedance readout on excised porcine tissues in motion. Through an animal study, we demonstrate how the device exhibits altered impedance response to tight junction dilation induced on mice colonic tissue through calcium-chelator exposure. Device measurements are validated using standard benchtop methods for assessing mucosal permeability.
Collapse
Affiliation(s)
- Brian M Holt
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, 20742, USA
- Institute for Systems Research, University of Maryland, College Park, MD, 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA
| | - Justin M Stine
- Matrix Lab at A. James Clark School of Engineering, University of Maryland, California, MD, 20619, USA
| | - Luke A Beardslee
- Institute for Systems Research, University of Maryland, College Park, MD, 20742, USA
| | - Hammed Ayansola
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Younggeon Jin
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | | | - Reza Ghodssi
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, 20742, USA.
- Institute for Systems Research, University of Maryland, College Park, MD, 20742, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA.
- Matrix Lab at A. James Clark School of Engineering, University of Maryland, California, MD, 20619, USA.
| |
Collapse
|
10
|
Xie Q, Yang M, Duanmu Q, Kang M, Wang J, Tan BE. Ningxiang pig-derived Lactobacillus reuteri improves the gut health of weaned piglets by regulating intestinal barrier function and cytokine profiles. Sci Rep 2025; 15:3993. [PMID: 39893246 PMCID: PMC11787358 DOI: 10.1038/s41598-025-87105-5] [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: 11/23/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
Weaning stress in piglets can induce intestinal damage, resulting in impaired growth performance. Probiotics have emerged as significant contributors to enhancing gut health in piglets. This study aimed to evaluate the effects of Lactobacillus reuteri (L. reuteri) supplementation on growth performance, intestinal immunity, and intestinal barrier integrity in weaned piglets. In this investigation, fourteen healthy weaned piglets of similar age and weight, were randomly assigned to two groups (n = 7), receiving either normal saline (Control group) or L. reuteri (L-treatment group) over a 16-day period. The findings revealed no significant impact of L. reuteri on growth performance compared to controls. However, it lowered serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels (P < 0.05) and elevated serum concentrations of Immunoglobulin A (IgA), immunoglobulin G (IgG), and secretory immunoglobulin A (sIgA) (P < 0.05). Additionally, L. reuteri notably enhanced the villus height-to-crypt depth ratio in the ileum (P < 0.05) and increased mRNA expression of zonula occludens-1 (ZO-1), Claudin-1, and ZO-1 in ileal tissue (P < 0.05). Furthermore, L. reuteri reduced mRNA expression of pro-inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) (P < 0.05) in the jejunum and colon while enhancing the expression of the anti-inflammatory cytokine interleukin-10 (IL-10) (P < 0.05) in both the ileum and colon. This study demonstrates that L. reuteri isolated from Ningxiang pigs can improve the intestinal health of weaned piglets by modulating gut barrier function and cytokine levels.
Collapse
Affiliation(s)
- Qian Xie
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Mei Yang
- Huizhou Engineering Vocational College, Huizhou, 516023, Guangdong, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Qing Duanmu
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Meng Kang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Jing Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China
| | - Bi E Tan
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and Poultry, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, Hunan, People's Republic of China.
- Yuelushan Laboratory, Changsha, 410128, Hunan, People's Republic of China.
| |
Collapse
|
11
|
Ünal S, Kartal SP, Özsoy A, Aydin FN. Evaluation of Serum Zonulin Level and Intestinal Permeability in Patients with Chronic Spontaneous Urticaria and the Relationship Between Serum Zonulin Level and Disease Severity. Dermatol Pract Concept 2025; 15:dpc.1501a4237. [PMID: 40117602 PMCID: PMC11928137 DOI: 10.5826/dpc.1501a4237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 03/23/2025] Open
Abstract
INTRODUCTION An emerging hypothesis suggests a potential link between enhanced intestinal permeability and the advancement of chronic spontaneous urticaria (CSU). OBJECTIVE This study aimed to investigate the role of intestinal permeability in the etiopathogenesis of CSU by measuring serum zonulin levels, a marker of intestinal permeability, in both CSU patients and control subjects. Additionally, the study sought to explore the correlation between the severity of the illness and zonulin levels. METHODS The study involved 61 patients with CSU and 59 healthy control individuals. For the CSU patients, comprehensive data were collected encompassing various aspects: age at onset of the condition, duration of the most recent attack, presence of any comorbid conditions, dosage of antihistamines being used, and urticaria activity score as well as detailed personal and family medical histories. Additionally, demographic information for these patients was also meticulously documented. RESULT The study revealed a statistically significant difference in zonulin levels between the CSU patient group and the control group, with a p-value of 0.000, indicating a highly significant disparity. Furthermore, among the CSU patients, those who presented with angioedema exhibited considerably higher zonulin levels compared to those without angioedema. This variation in zonulin levels based on the presence of angioedema was also statistically significant, with a p-value of 0.023. CONCLUSION The observed results suggest that increased intestinal permeability, as indicated by elevated zonulin levels, may play a crucial role in the pathophysiology of both CSU and angioedema. This association highlights the potential significance of intestinal permeability in the development and manifestation of these conditions.
Collapse
Affiliation(s)
- Simge Ünal
- Dışkapı Yıldırım Beyazıt Hospital, Department of Dermatology and Venereology, Ankara, Turkey
| | - Selda Pelin Kartal
- Dışkapı Yıldırım Beyazıt Hospital, Department of Dermatology and Venereology, Ankara, Turkey
| | - Ahmet Özsoy
- Dışkapı Yıldırım Beyazıt Hospital, Department of Clinic Biochemistry, Ankara, Turkey
| | - Fevzi Nuri Aydin
- Dışkapı Yıldırım Beyazıt Hospital, Department of Clinic Biochemistry, Ankara, Turkey
| |
Collapse
|
12
|
Guerrero Aznar MD, Villanueva Guerrero MD, Beltrán García M, Hernández Cruz B. Specific Composition Diets and Improvement of Symptoms of Immune-Mediated Inflammatory Diseases in Adulthood-Could the Comparison Between Diets Be Improved? Nutrients 2025; 17:493. [PMID: 39940351 PMCID: PMC11819864 DOI: 10.3390/nu17030493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Diet is considered a possible cofactor, which affects the immune system and potentially causes dysregulation of intestinal homeostasis and inflammation. This study aimed to review the quality of evidence on the effects of specific diet composition on symptoms of immune-mediated inflammatory diseases (IMIDs), including rheumatoid arthritis (RA), spondyloarthritis, multiple sclerosis (MS), inflammatory bowel disease (IBD) [remission maintenance of Crohn's disease and ulcerative colitis], psoriasis and psoriatic arthritis in adult patients. We conducted a review of meta-analyses and Cochrane systematic reviews using PubMed and EMBASE, from inception to September 2024, and Google Scholar. The methodological quality of the meta-analyses was assessed using the AMSTAR 2 rating system. Three Cochrane systematic reviews and eight meta-analyses were evaluated. Some specific composition diets have been shown to reduce the symptoms of RA, IBD, and MS and improve activity parameters in IBD and RA, with critically low or low levels of evidence. The reduction in inflammatory biomarker levels is unclear. This review summarizes the global evidence for specific dietary interventions, mostly with anti-inflammatory properties due to their components, to improve IMID symptoms, clarifying the weaknesses of clinical trials and dietary meta-analyses with critically low or low levels of evidence; and shows the need to use indices such as the Dietary Inflammatory Index, which allows diets to be classified by their pro-inflammatory or anti-inflammatory food content, to better compare diet groups in clinical trials. The difficulty of obtaining high-level evidence from dietary studies is apparent and may delay the application of the results. Clinicians should be aware of the role of diets with anti-inflammatory properties as a complement to pharmacological treatments in IMIDs.
Collapse
Affiliation(s)
- M. Dolores Guerrero Aznar
- Pharmacy Health Management Unit, Virgen Macarena University Hospital, 41009 Seville, Spain; (M.D.V.G.); (M.B.G.)
| | | | - Margarita Beltrán García
- Pharmacy Health Management Unit, Virgen Macarena University Hospital, 41009 Seville, Spain; (M.D.V.G.); (M.B.G.)
| | - Blanca Hernández Cruz
- Rheumatology Health Management Unit, Virgen Macarena University Hospital, 41009 Seville, Spain;
| |
Collapse
|
13
|
Liao W, Zhang X, Jia C, Chen W, Cai Y, Zhang H, Wei J, Chen T. Lactobacillus rhamnosus LC-STH-13 ameliorates the progression of SLE in MRL/lpr mice by inhibiting the TLR9/NF-κB signaling pathway. Food Funct 2025; 16:475-486. [PMID: 39744924 DOI: 10.1039/d4fo03966a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease often treated with glucocorticoids, which can lead to complications such as osteoporosis and an increased infection risk. Hence, identifying safe and effective treatment strategies is crucial. Lactobacillus has shown promise in improving immune disorders. We investigated Lactobacillus rhamnosus LC-STH-13 for its probiotic properties. Female MRL/lpr mice, prone to lupus, were used to assess its impact on SLE development. The results showed that the intervention with L. rhamnosus LC-STH-13 significantly reduced the level of circulating anti-autoantibodies (p < 0.05) and rebalanced Th17/Treg cells (p < 0.05). Kidney tissue analysis revealed reduced immune cell infiltration and immune complex deposition in glomeruli. L. rhamnosus LC-STH-13 mitigated kidney inflammation via the TLR9/NF-κB pathway (p < 0.05) and attenuated complement-induced renal damage (p < 0.05). Furthermore, 16S rRNA sequencing data analysis indicated that L. rhamnosus LC-STH-13 can restore intestinal microecological imbalance caused by the development of SLE. These findings suggested that L. rhamnosus LC-STH-13 improves the development of SLE by regulating the TLR9/NF-κB pathway and intestinal microbiota, offering a foundation for exploring safe and effective treatments.
Collapse
Affiliation(s)
- Wen Liao
- School of Life Sciences, Nanchang University, Nanchang 330031, China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Xinyi Zhang
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Chunjian Jia
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Wenjing Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yujie Cai
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Hongyan Zhang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Nanchang, 330006, China.
| | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Tingtao Chen
- School of Life Sciences, Nanchang University, Nanchang 330031, China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
14
|
Elbehiry A, Marzouk E, Abalkhail A, Sindi W, Alzahrani Y, Alhifani S, Alshehri T, Anajirih NA, ALMutairi T, Alsaedi A, Alzaben F, Alqrni A, Draz A, Almuzaini AM, Aljarallah SN, Almujaidel A, Abu-Okail A. Pivotal role of Helicobacter pylori virulence genes in pathogenicity and vaccine development. Front Med (Lausanne) 2025; 11:1523991. [PMID: 39850097 PMCID: PMC11756510 DOI: 10.3389/fmed.2024.1523991] [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: 11/06/2024] [Accepted: 12/13/2024] [Indexed: 01/25/2025] Open
Abstract
One of the most prevalent human infections is Helicobacter pylori (H. pylori), which affects more than half of the global population. Although H. pylori infections are widespread, only a minority of individuals develop severe gastroduodenal disorders. The global resistance of H. pylori to antibiotics has reached concerning levels, significantly impacting the effectiveness of treatment. Consequently, the development of vaccines targeting virulence factors may present a viable alternative for the treatment and prevention of H. pylori infections. This review aims to provide a comprehensive overview of the current understanding of H. pylori infection, with a particular focus on its virulence factors, pathophysiology, and vaccination strategies. This review discusses various virulence factors associated with H. pylori, such as cytotoxin-associated gene A (cagA), vacuolating cytotoxin gene (vacA), outer membrane proteins (OMPs), neutrophil-activated protein (NAP), urease (ure), and catalase. The development of vaccines based on these virulence characteristics is essential for controlling infection and ensuring long-lasting protection. Various vaccination strategies and formulations have been tested in animal models; however, their effectiveness and reproducibility in humans remain uncertain. Different types of vaccines, including vector-based vaccines, inactivated whole cells, genetically modified protein-based subunits, and multiepitope nucleic acid (DNA) vaccines, have been explored. While some vaccines have demonstrated promising results in murine models, only a limited number have been successfully tested in humans. This article provides a thorough evaluation of recent research on H. pylori virulence genes and vaccination methods, offering valuable insights for future strategies to address this global health challenge.
Collapse
Affiliation(s)
- Ayman Elbehiry
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Eman Marzouk
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Wael Sindi
- Department of Population, Public and Environmental Health, General Administration of Health Services, Ministry of Defense, Riyadh, Saudi Arabia
| | - Yasir Alzahrani
- Department of Psychiatry, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Salem Alhifani
- Department of Psychiatry, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Turki Alshehri
- Department of Dental, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| | - Nuha Abdulaziz Anajirih
- Department of Medical Emergency Services, Faculty of Health Sciences, Umm Al-Qura University, Al-Qunfudah, Saudi Arabia
| | - Turki ALMutairi
- Department of Education and Training, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - Ahmad Alsaedi
- Department of Education and Training, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - Feras Alzaben
- Department of Food Service, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Abdullah Alqrni
- Department of Preventive Medicine, King Fahad Armed Hospital, Jeddah, Saudi Arabia
| | - Abdelmaged Draz
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Abdulaziz M. Almuzaini
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Sahar N. Aljarallah
- Department of Pharmacy Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Abdulrahman Almujaidel
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Akram Abu-Okail
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
15
|
Schoenaker JM, Nelson VS, Henderickx JGE, Terveer EM, Jansen AJG, Porcelijn L, Netelenbos T, Schipperus MR, Kapur R. The intestinal flora: The key to unraveling heterogeneity in immune thrombocytopenia? Blood Rev 2025; 69:101252. [PMID: 39672701 DOI: 10.1016/j.blre.2024.101252] [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: 10/16/2024] [Revised: 11/22/2024] [Accepted: 12/01/2024] [Indexed: 12/15/2024]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder characterized by enhanced platelet destruction and impaired platelet production, due to a loss of immune tolerance that leads to targeting of platelets and megakaryocytes by glycoprotein-autoantibodies and/or cytotoxic T cells. There is a high degree of heterogeneity in ITP patients signified by unpredictable disease trajectories and treatment responses. Initial studies in humans have identified intestinal microbiota perturbance in ITP. Recently, gut microbial perturbance has been linked to other autoimmune diseases. Based on these findings, we hypothesize that intestinal microbiota may influence ITP pathophysiology through several mechanisms, including induction of platelet-autoantibody production, increasing complement-dependent platelet cytotoxicity, disturbing T cell homeostasis, impairing megakaryocyte function, and increasing platelet-desialylation and -clearance. The pathophysiological heterogeneity of ITP may, at least in part, be attributed to a perturbed intestinal microbiota. Therefore, a better understanding of intestinal microbiota in ITP may result in a more personalized therapeutic approach.
Collapse
MESH Headings
- Humans
- Gastrointestinal Microbiome/immunology
- Purpura, Thrombocytopenic, Idiopathic/microbiology
- Purpura, Thrombocytopenic, Idiopathic/etiology
- Purpura, Thrombocytopenic, Idiopathic/immunology
- Purpura, Thrombocytopenic, Idiopathic/metabolism
- Purpura, Thrombocytopenic, Idiopathic/pathology
- Blood Platelets/immunology
- Blood Platelets/metabolism
- Blood Platelets/pathology
- Animals
- Disease Susceptibility
- Autoantibodies/immunology
Collapse
Affiliation(s)
- Jente M Schoenaker
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, the Netherlands.
| | - Vivianne S Nelson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, the Netherlands; Department of Hematology, HagaZiekenhuis, 2545 AA The Hague, the Netherlands; Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Jannie G E Henderickx
- Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Department of Medical Microbiology, Leiden University Center of Infectious Diseases (LU-CID) Research, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Elisabeth M Terveer
- Department of Medical Microbiology, Leiden University Center of Infectious Diseases (LU-CID) Research, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Netherlands Donor Feces Bank, LUCID Medical Microbiology & Infection Prevention, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - A J Gerard Jansen
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, the Netherlands.
| | - Leendert Porcelijn
- Sanquin Diagnostic Services, Department of Immunohematology Diagnostics, Sanquin, 1066 CX Amsterdam, the Netherlands.
| | - Tanja Netelenbos
- Department of Hematology, HagaZiekenhuis, 2545 AA The Hague, the Netherlands.
| | | | - Rick Kapur
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Wang X, Cao Z, Su J, Ge X, Zhou Z. Oral barriers to food-derived active peptides and nano-delivery strategies. J Food Sci 2025; 90:e17672. [PMID: 39828408 DOI: 10.1111/1750-3841.17672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/04/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
Food-derived bioactive peptides are a class of peptides from natural protein. It may have biological effects on the human body and play a significant role in protecting human physiological health and regulating physiological metabolism, such as lowering blood pressure, lowering cholesterol, antioxidant, antibacterial, regulating immune activity, and so on. However, most of the natural food-derived functional peptides need to overcome a variety of barriers in the body to enter the blood circulation system and target to specific tissues to generate physiological activity. During this process, the bioavailability of the functional peptides will be reduced. The nano-delivery system can offer the feasibility to overcome these obstacles and improve the stability and bioavailability of food-derived active peptides by nanoencapsulation. This work summarizes the application of food-derived bioactive peptides and the obstacles during the delivery pathway in vivo. Moreover, the different nano-delivery systems used for bioactive peptides and their application were summarized, which could provide ideas for oral delivery of food-derived bioactive peptides.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, P. R. China
| | - Zhaoxin Cao
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, P. R. China
| | - Jingyi Su
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, P. R. China
| | - Xuemei Ge
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, P. R. China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Zhiyong Zhou
- College of Medicine and Health Sciences, China Three Gorges University, Yichang, P. R. China
| |
Collapse
|
17
|
Chasov V, Gilyazova E, Ganeeva I, Zmievskaya E, Davletshin D, Valiullina A, Bulatov E. Gut Microbiota Modulation: A Novel Strategy for Rheumatoid Arthritis Therapy. Biomolecules 2024; 14:1653. [PMID: 39766360 PMCID: PMC11674688 DOI: 10.3390/biom14121653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that leads to joint inflammation, progressive tissue damage and significant disability, severely impacting patients' quality of life. While the exact mechanisms underlying RA remain elusive, growing evidence suggests a strong link between intestinal microbiota dysbiosis and the disease's development and progression. Differences in microbial composition between healthy individuals and RA patients point to the role of gut microbiota in modulating immune responses and promoting inflammation. Therapies targeting microbiota restoration have demonstrated promise in improving treatment efficacy, enhancing patient outcomes and slowing disease progression. However, the complex interplay between gut microbiota and autoimmune pathways in RA requires further investigation to establish causative relationships and mechanisms. Here, we review the current understanding of the gut microbiota's role in RA pathogenesis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Vitaly Chasov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Elvina Gilyazova
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Irina Ganeeva
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Ekaterina Zmievskaya
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Damir Davletshin
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Aygul Valiullina
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Emil Bulatov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
18
|
Li H, Fan L, Yang S, Tan P, Lei W, Yang H, Gao Z. Lactobacillus acidophilus 6074 Fermented Jujube Juice Ameliorated DSS-induced Colitis via Repairing Intestinal Barrier, Modulating Inflammatory Factors, and Gut Microbiota. Mol Nutr Food Res 2024:e202400568. [PMID: 39676427 DOI: 10.1002/mnfr.202400568] [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: 07/22/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
Lactobacillus acidophilus L. acidophilus Lactobacillus, Bifidobacterium, and Akkermansia, This study aimed to explore the ameliorative effects and underlying mechanisms of oral administration Lactobacillus acidophilus 6074 fermented jujube juice (LAFJ) on dextran sulfate sodium (DSS)-induced colitis in mice. In this study, jujube juice was used as a substrate and fermented by L. acidophilus 6074 to investigate its effects on gut microbiota, intestinal barrier function, oxidative stress, inflammatory factors, and short-chain fatty acids (SCFAs) in mice with colitis and to reveal its potential mechanism for alleviating colitis. The results demonstrated that fermentation caused significant changes in the nutrients and nonnutrients of jujube juice, mainly in organic acids (malic acid, lactic acid, citric acid, and succinic acid) and free amino acids (Thr, Met, Ser, Ile, and Lys). High-dose LAFJ (20 mL/kg/day) significantly reduced the disease activity index (DAI), improved histopathological morphology, and increased colon length in colitis mice. LAFJ alleviated colon damage and preserved the integrity of the colonic mucosal barrier by promoting the expression of colonic tight junction proteins occludin, claudin-1, and zonula occluden-1 (ZO-1). Furthermore, LAFJ inhibited the production of proinflammatory factors and attenuated oxidative stress. Gut microbiota of mice revealed that LAFJ increased beneficial bacteria such as Lactobacillus, Bifidobacterium, and Akkermansia, promoted the production of SCFAs, and inhibited the growth of harmful microorganisms. Overall, LAFJ could reshape and restore gut microbiota imbalance caused by intestinal inflammation and alleviate the development of colitis, which may become a novel dietary intervention.
Collapse
Affiliation(s)
- Hongcai Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Lingjia Fan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Siqi Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Pei Tan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Wenzhi Lei
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Haihua Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Zhenpeng Gao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| |
Collapse
|
19
|
Wang H, Li S, Zhang L, Zhang N. The role of fecal microbiota transplantation in type 2 diabetes mellitus treatment. Front Endocrinol (Lausanne) 2024; 15:1469165. [PMID: 39735647 PMCID: PMC11671274 DOI: 10.3389/fendo.2024.1469165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/22/2024] [Indexed: 12/31/2024] Open
Abstract
In contemporary microbial research, the exploration of interactions between microorganisms and multicellular hosts constitutes a burgeoning field. The gut microbiota is increasingly acknowledged as a pivotal contributor to various disorders within the endocrine system, encompassing conditions such as diabetes and thyroid diseases. A surge in research activities has been witnessed in recent years, elucidating the intricate interplay between the gut microbiota and disorders of the endocrine system. Simultaneously, fecal microbiota transplantation (FMT) has emerged as a focal point, garnering substantial attention in both biomedical and clinical spheres. Research endeavors have uncovered the remarkable therapeutic efficacy of FMT across diverse diseases, with particular emphasis on its application in addressing type 2 diabetes mellitus (T2DM) and associated com-plications. Consequently, this manuscript accentuates the intimate connection between the gut microbiota and disorders within the endocrine system, with a specific focus on exploring the potential of FMT as an intervention in the therapeutic landscape of T2DM and its complications. Furthermore, the article scrutinizes concerns inherent in treatment modalities centered around the gut microbiota, proposing viable solutions to address these issues.
Collapse
Affiliation(s)
| | | | | | - Nan Zhang
- *Correspondence: Nan Zhang, ; Luping Zhang,
| |
Collapse
|
20
|
Zhao R, Gu J, Zhao H, Wang Z, Liu X, Yuan C, Zheng X, Yang T, Xu X, Cai Y. Expression of integrin α4β1 and α4β7 on B cells correlates with autoimmune responses in Graves' disease. Int Immunopharmacol 2024; 142:113218. [PMID: 39317053 DOI: 10.1016/j.intimp.2024.113218] [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: 04/12/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Integrins are upregulated on endothelial cells and T-lymphocytes in autoimmune thyroid disease (AITD), potentially contributing to immune response localization. The role of integrins on B-cells in AITD remains unclear. METHODS Peripheral blood samples were collected from healthy controls (n = 56), patients with Graves' disease (GD) (n = 37) and Hashimoto's thyroiditis (HT) (n = 52). Ultrasound-guided fine-needle aspiration (FNA) of the thyroid was performed in patients with non-autoimmune thyroid disease (nAITD) (n = 19), GD (n = 11), and HT (n = 40). Integrins α4β7, α4β1, and αEβ7 in B cells were measured by flow cytometry. Serum zonulin levels were quantified via ELISA. Associations of integrins on B cells with thyroid hormones, thyroid autoantibodies, AITD duration, and zonulin were analyzed. RESULTS HT patients exhibited lower α4β7 and higher α4β1 expression on B cells compared to healthy controls and GD patients. While α4β7 was predominant on circulating B cells, the dominant integrin expressed on intrathyroidal B cells varied with specific thyroid diseases. In GD patients, α4β7 and α4β1 expression on circulating B cells correlated positively and negatively with thyroid function and thyroid stimulating immunoglobulins (TSI) levels, respectively. Intrathyroidal α4β1+ B cells positively correlated with TSH levels in HT patients. Additionally, serum zonulin was elevated in HT patients, and intrathyroidal α4β7+ B cells and α4β1+ B cells correlated negatively and positively with zonulin levels, respectively. Integrin αEβ7 on B cells showed no significant association with AITD. CONCLUSION Integrins expressed on B cells potentially play a role in the pathogenesis of AITD and might serve as immune biomarkers for the disease.
Collapse
Affiliation(s)
- Ruiling Zhao
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Junjie Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hang Zhao
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhixiao Wang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaoyun Liu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Cuiping Yuan
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xuqin Zheng
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xinyu Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Yun Cai
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
21
|
Luo Y, Sheikh TMM, Li X, Yuan Y, Yao F, Wang M, Guo X, Wu J, Shafiq M, Xie Q, Jiao X. Exploring the dynamics of gut microbiota, antibiotic resistance, and chemotherapy impact in acute leukemia patients: A comprehensive metagenomic analysis. Virulence 2024; 15:2428843. [PMID: 39620486 PMCID: PMC11622590 DOI: 10.1080/21505594.2024.2428843] [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: 05/07/2024] [Revised: 08/24/2024] [Accepted: 11/06/2024] [Indexed: 12/08/2024] Open
Abstract
Leukemia poses significant challenges to its treatment, and understanding its complex pathogenesis is crucial. This study used metagenomic sequencing to investigate the interplay between chemotherapy, gut microbiota, and antibiotic resistance in patients with acute leukemia (AL). Pre- and post-chemotherapy stool samples from patients revealed alterations in microbial richness, taxa, and antibiotic resistance genes (ARGs). The analysis revealed a decreased alpha diversity, increased dispersion in post-chemotherapy samples, and changes in the abundance of specific bacteria. Key bacteria such as Enterococcus, Klebsiella, and Escherichia coli have been identified as prevalent ARG carriers. Correlation analysis between gut microbiota and blood indicators revealed potential links between microbial species and inflammatory biomarkers, including C-reactive protein (CRP) and adenosine deaminase (ADA). This study investigated the impact of antibiotic dosage on microbiota and ARGs, revealing networks connecting co-occurring ARGs with microbial species (179 nodes, 206 edges), and networks associated with ARGs and antibiotic dosages (50 nodes, 50 edges). Antibiotics such as cephamycin and sulfonamide led to multidrug-resistant Klebsiella colonization. Our analyses revealed distinct microbial profiles with Salmonella enterica elevated post-chemotherapy in NF patients and Akkermansia muciniphila elevated pre-chemotherapy. These microbial signatures could inform strategies to modulate the gut microbiome, potentially mitigating the risk of neutropenic fever in patients undergoing chemotherapy. Finally, a comprehensive analysis of KEGG modules shed light on disrupted metabolic pathways after chemotherapy, providing insights into potential targets for managing side effects. Overall, this study revealed intricate relationships between gut microbiota, chemotherapy, and antibiotic resistance, providing new insights into improving therapy and enhancing patient outcomes.
Collapse
Affiliation(s)
- Ying Luo
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | | - Xin Li
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - YuMeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Fen Yao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Meimei Wang
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoling Guo
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Jilong Wu
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Shantou University Medical College, Shantou, China
| | - Qingdong Xie
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| |
Collapse
|
22
|
Gautam R, Maan P, Patel AK, Vasudevan S, Arora T. Unveiling the complex interplay between gut microbiota and polycystic ovary syndrome: A narrative review. Clin Nutr 2024; 43:199-208. [PMID: 39481287 DOI: 10.1016/j.clnu.2024.10.028] [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: 04/04/2024] [Revised: 07/11/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND & AIM Polycystic Ovary Syndrome (PCOS) is a complex endocrine disorder that affects women throughout their reproductive age and characterised via polycystic ovaries, hyperandrogenism, and irregular menstruation. There is rising evidence that the pathophysiology of PCOS is significantly affected via the gut microbiota and its metabolic products. METHODS This narrative review synthesizes current literature exploring the relationship between gut microbiota and PCOS. A comprehensive search of electronic databases was conducted to identify relevant studies. Further this review also analysed therapeutic options of probiotics, prebiotics, Fecal Microbiota Transplant (FMT), high fiber and poly phenol rich diet and novel therapeutic agents in treatment of PCOS. RESULTS Emerging evidence suggests alterations in the composition and diversity of gut microbiota in women with PCOS. The current literature showed a complex relationship of gut microbiota, short chain fatty acids (SCFAs) metabolism, intestinal permeability and LPS (Lipid Polysaccharide) metabolism, gut-brain axis and bile acid (BA) pathway within etiology and pathophysiology of PCOS. Additionally, the factors such as diet, lifestyle, genetics, and environmental influences may all contribute to alterations in gut microbiota that could potentially exacerbate or mitigate PCOS symptoms. CONCLUSION The review provides valuable insights into the intricate interplay between the gut and female reproductive health. The present evidence suggested that alterations in diversity and function of the gut microbiota may lead to specific pathogenic changes that lead to development of PCOS. A comprehensive understanding of these microbial dynamics may lead to new therapeutic approaches that target the gut micro biome.
Collapse
Affiliation(s)
- Rohit Gautam
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Pratibha Maan
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Arbind Kumar Patel
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sudharsan Vasudevan
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Taruna Arora
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India.
| |
Collapse
|
23
|
Soriano-Lerma A, García-Burgos M, Barton W, Alférez MJM, Crespo-Pérez JV, Soriano M, López-Aliaga I, Cotter PD, García-Salcedo JA. Comprehensive insight into the alterations in the gut microbiome and the intestinal barrier as a consequence of iron deficiency anaemia. Biomed J 2024; 47:100701. [PMID: 38281699 PMCID: PMC11550200 DOI: 10.1016/j.bj.2024.100701] [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: 05/30/2023] [Revised: 11/09/2023] [Accepted: 01/19/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Iron deficiency is the top leading cause of anaemia, whose treatment has been shown to deteriorate gut health. However, a comprehensive analysis of the intestinal barrier and the gut microbiome during iron deficiency anemia (IDA) has not been performed to date. This study aims to delve further into the analysis of these two aspects, which will mean a step forward minimising the negative impact of iron supplements on intestinal health. METHODS IDA was experimentally induced in an animal model. Shotgun sequencing was used to analyse the gut microbiome in the colonic region, while the intestinal barrier was studied through histological analyses, mRNA sequencing (RNA-Seq), qPCR and immunofluorescence assays. Determinations of lipopolysaccharide (LPS) and bacteria-specific immunoglobulins were performed to assess microbial translocation. RESULTS Microbial metabolism in the colon shifted towards an increased production of certain amino acids, short chain fatty acids and nucleotides, with Clostridium species being enriched during IDA. Structural alterations of the colonic epithelium were shown by histological analysis. RNA-Seq revealed a downregulation of extracellular matrix-associated genes and proteins and an overall underdeveloped epithelium. Increased levels of serum LPS and an increased immune response against dysbiotic bacteria support an impairment in the integrity of the gut barrier during IDA. CONCLUSIONS IDA negatively impacts the gut microbiome and the intestinal barrier, triggering an increased microbial translocation. This study emphasizes the deterioration of gut health during IDA and the fact that it should be addressed when treating the disease.
Collapse
Affiliation(s)
- Ana Soriano-Lerma
- Department of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja), Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain; GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, Granada, Spain; Instituto de Investigación Biosanitaria ibs, Granada, Spain
| | - María García-Burgos
- Department of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja), Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain; GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, Granada, Spain
| | - Wiley Barton
- VistaMilk SFI Research Centre, Cork, Ireland; Teagasc Food Research Centre, Carlow, Ireland
| | - María José M Alférez
- Department of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja), Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain
| | - Jorge Valentín Crespo-Pérez
- Service of Anatomical pathology, Intercenter Regional Unit Granada, University Hospital Virgen de las Nieves, Granada, Spain
| | - Miguel Soriano
- Center for Intensive Mediterranean Agrosystems and Agri-food Biotechnology (CIAIMBITAL), University of Almeria, Almeria, Spain.
| | - Inmaculada López-Aliaga
- Department of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja), Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Granada, Spain.
| | - Paul D Cotter
- VistaMilk SFI Research Centre, Cork, Ireland; Teagasc Food Research Centre, Carlow, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - José A García-Salcedo
- GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, Granada, Spain; Instituto de Investigación Biosanitaria ibs, Granada, Spain; Microbiology Unit, University Hospital Virgen de las Nieves, Granada, Spain
| |
Collapse
|
24
|
Xu C, Liu Y, Li K, Zhang J, Wei B, Wang H. Absorption of food-derived peptides: Mechanisms, influencing factors, and enhancement strategies. Food Res Int 2024; 197:115190. [PMID: 39593400 DOI: 10.1016/j.foodres.2024.115190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 11/28/2024]
Abstract
Food-derived peptides (FPs) are bioactive molecules produced from dietary proteins through enzymatic hydrolysis or fermentation. These peptides exhibit various biological activities. However, their efficacy largely depends on bioavailability, the ability to cross absorption barriers, and reach target sites within the body. This review addresses key issues in FP absorption, including barriers, pathways, influencing factors, and strategies to enhance absorption. The biochemical and physical barriers to FP absorption include pH variations, enzymes, unstirred water layer, mucus layer, and intestinal epithelial cells. FPs enter the bloodstream via four main pathways: carrier-mediated transport, endocytosis, paracellular, and passive diffusion. The barrier-crossing efficiency depends on the structural properties and state of FPs and coexisting substances. Absorption efficiency can be significantly improved with permeability enhancers, nano-delivery systems, and chemical modifications. These insights provide a scientific basis and practical guidance for optimizing the bioactivity and health benefits of food-derived peptides.
Collapse
Affiliation(s)
- Chengzhi Xu
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Yuting Liu
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Ke Li
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Juntao Zhang
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Benmei Wei
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan, Hubei, China.
| | - Haibo Wang
- College of Life Science and Technology, Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, Hubei Engineering University, Xiaogan, Hubei, China.
| |
Collapse
|
25
|
Młynarska E, Wasiak J, Gajewska A, Bilińska A, Steć G, Jasińska J, Rysz J, Franczyk B. Gut Microbiota and Gut-Brain Axis in Hypertension: Implications for Kidney and Cardiovascular Health-A Narrative Review. Nutrients 2024; 16:4079. [PMID: 39683474 DOI: 10.3390/nu16234079] [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: 09/17/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
INTRODUCTION Arterial hypertension is a major contributor to a wide range of health complications, with cardiac hypertrophy and chronic kidney disease being among the most prevalent. Consequently, novel strategies for the treatment and prevention of hypertension are actively being explored. Recent research has highlighted a potential link between hypertension and the gut-brain axis. A bidirectional communication between the microbiota and the brain via the vagus nerve, enteric nervous system, hypothalamus-pituitary-adrenal axis, secreted short-chain fatty acids, and neurotransmitter metabolism. MATERIALS AND METHODS A comprehensive literature search was conducted using databases such as PubMed to identify studies exploring the relationship between gut microbiota and hypertension, along with the effects of dietary interventions and probiotics on blood pressure regulation. DISCUSSION Studies in both animal models and human subjects have demonstrated a strong correlation between alterations in gut microbiota composition and the development of hypertension. By influencing blood pressure, the gut microbiota can potentially affect the progression of cardiovascular and kidney disorders. Modulating gut microbiota through dietary interventions and probiotics has shown promise in regulating blood pressure and reducing systemic inflammation, offering a novel approach to managing hypertension. Diets such as the Mediterranean diet, which is rich in polyphenols and omega-3 fatty acids and low in sodium, promote the growth of beneficial gut bacteria that support cardiovascular health. Additionally, probiotics have been found to enhance gut barrier function, reduce inflammation, and modulate the Renin-Angiotensin System, all of which contribute to lowering blood pressure. CONCLUSIONS Further research is needed to determine the mechanisms of action of the microbiota in hypertension. The aim of this study was to evaluate the influence of gut microbiota on blood pressure regulation and the progression of hypertension-related complications, such as cardiovascular and kidney disorders.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Aleksandra Bilińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Greta Steć
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Jasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
26
|
Zhou M, Chen Y, Jin W, Li P, Hu J, Guo X. Traditional Chinese Medicine: A Promising Treatment Option for Intestinal Fibrosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2107-2129. [PMID: 39581857 DOI: 10.1142/s0192415x24500812] [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: 11/26/2024]
Abstract
Intestinal fibrosis, a common complication of inflammatory bowel disease, in particular in Crohn's disease, arises from chronic inflammation, leading to intestinal narrowing, structural damage, and functional impairment that significantly impact patients' quality of life. Current treatment options for intestinal fibrosis are limited, with surgery being the primary intervention. Traditional Chinese Medicine (TCM) has emerged as a promising approach in preventing and treating intestinal fibrosis. However, there is a scarcity of literature summarizing the mechanisms underlying TCM's efficacy in this context. To address this gap, we conducted a comprehensive review, uncovering multiple mechanisms through which TCM mitigates intestinal fibrosis. These mechanisms include immune cell balance regulation, suppression of inflammatory responses, reduction of inflammatory mediators, alleviation of colon tissue damage, restoration of intestinal function, modulation of growth factors to inhibit fibroblast activation, dynamic regulation of TIMPs and MMPs to reduce extracellular matrix deposition, inhibition of epithelial-mesenchymal transition and endothelial-mesenchymal transition, autophagy modulation, maintenance of the intestinal mucosal barrier, prevention of tissue damage by harmful factors, and regulation of cell proliferation and apoptosis. This study aims to bridge existing knowledge gaps by presenting recent evidence supporting the utilization of TCM in both clinical and experimental research settings.
Collapse
Affiliation(s)
- Meng'en Zhou
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Yan Chen
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Wenqi Jin
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Peng Li
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Jie Hu
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Xiutian Guo
- Department of Anorectal, Shanghai Municipal, Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| |
Collapse
|
27
|
Zhang Y, Song M, Fan J, Guo X, Tao S. Impact of probiotics-derived extracellular vesicles on livestock gut barrier function. J Anim Sci Biotechnol 2024; 15:149. [PMID: 39506860 PMCID: PMC11542448 DOI: 10.1186/s40104-024-01102-8] [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: 06/03/2024] [Accepted: 09/05/2024] [Indexed: 11/08/2024] Open
Abstract
Probiotic extracellular vesicles (pEVs) are biologically active nanoparticle structures that can regulate the intestinal tract through direct or indirect mechanisms. They enhance the intestinal barrier function in livestock and poultry and help alleviate intestinal diseases. The specific effects of pEVs depend on their internal functional components, including nucleic acids, proteins, lipids, and other substances. This paper presents a narrative review of the impact of pEVs on the intestinal barrier across various segments of the intestinal tract, exploring their mechanisms of action while highlighting the limitations of current research. Investigating the mechanisms through which probiotics operate via pEVs could deepen our understanding and provide a theoretical foundation for their application in livestock production.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Mengzhen Song
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Jinping Fan
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Xuming Guo
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China.
| |
Collapse
|
28
|
Guarner F, Bustos Fernandez L, Cruchet S, Damião A, Maruy Saito A, Riveros Lopez JP, Rodrigues Silva L, Valdovinos Diaz MA. Gut dysbiosis mediates the association between antibiotic exposure and chronic disease. Front Med (Lausanne) 2024; 11:1477882. [PMID: 39568738 PMCID: PMC11576192 DOI: 10.3389/fmed.2024.1477882] [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: 08/08/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Antibiotics are safe, effective drugs and continue to save millions of lives and prevent long-term illness worldwide. A large body of epidemiological, interventional and experimental evidence shows that exposure to antibiotics has long-term negative effects on human health. We reviewed the literature data on the links between antibiotic exposure, gut dysbiosis, and chronic disease (notably with regard to the "developmental origins of health and disease" ("DOHaD") approach). Molecular biology studies show that the systemic administration of antibiotic to infants has a rapid onset but also often a long-lasting impact on the microbial composition of the gut. Along with other environmental factors (e.g., an unhealthy "Western" diet and sedentary behavior), antibiotics induce gut dysbiosis, which can be defined as the disruption of a previously stable, functionally complete microbiota. Gut dysbiosis many harmful long-term effects on health. Associations between early-life exposure to antibiotics have been reported for chronic diseases, including inflammatory bowel disease, celiac disease, some cancers, metabolic diseases (obesity and type 2 diabetes), allergic diseases, autoimmune disorders, atherosclerosis, arthritis, and neurodevelopmental, neurodegenerative and other neurological diseases. In mechanistic terms, gut dysbiosis influences chronic disease through direct effects on mucosal immune and inflammatory pathways, plus a wide array of direct or indirect effects of short-chain fatty acids, the enteric nervous system, peristaltic motility, the production of hormones and neurotransmitters, and the loss of intestinal barrier integrity (notably with leakage of the pro-inflammatory endotoxin lipopolysaccharide into the circulation). To mitigate dysbiosis, the administration of probiotics in patients with chronic disease is often (but not always) associated with positive effects on clinical markers (e.g., disease scores) and biomarkers of inflammation and immune activation. Meta-analyses are complicated by differences in probiotic composition, dose level, and treatment duration, and large, randomized, controlled clinical trials are lacking in many disease areas. In view of the critical importance of deciding whether or not to prescribe antibiotics (especially to children), we suggest that the DOHaD concept can be logically extended to "gastrointestinal origins of health and disease" ("GOHaD") or even "microbiotic origins of health and disease" ("MOHaD").
Collapse
Affiliation(s)
| | - Luis Bustos Fernandez
- Centro Medico Bustos Fernandez, Instituto de Gastroenterologia, Buenos Aires, Argentina
| | - Sylvia Cruchet
- Institute of Nutrition and Food Technology, Universidad de Chile, Santiago, Chile
| | - Adérson Damião
- Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Aldo Maruy Saito
- Catedra de Pediatria, Hospital Cayetano Heredia, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | |
Collapse
|
29
|
Beck DL, Gilbert ER, Cline MA. Embryonic thermal challenge is associated with increased stressor resiliency later in life: Molecular and morphological mechanisms in the small intestine. Comp Biochem Physiol A Mol Integr Physiol 2024; 297:111724. [PMID: 39111617 DOI: 10.1016/j.cbpa.2024.111724] [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: 06/10/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024]
Abstract
Developing chick embryos that are subjected to increased incubation temperature are more stressor-resilient later in life, but the underlying process is poorly understood. The potential mechanism may involve changes in small intestine function. In this study, we determined behavioral, morphological, and molecular effects of increased embryonic incubation temperatures and post-hatch heat challenge in order to understand how embryonic heat conditioning (EHC) affects gut function. At 4 days post-hatch, duodenum, jejunum, and ileum samples were collected at 0, 2, and 12 h relative to the start of heat challenge. In EHC chicks, we found that markers of heat and oxidative stress were generally lower while those of nutrient transport and antioxidants were higher. Temporally, gene expression changes in response to the heat challenge were similar in control and EHC chicks for markers of heat and oxidative stress. Crypt depth was greater in control than EHC chicks at 2 h post-challenge, and the villus height to crypt depth ratio increased from 2 to 12 h in both control and EHC chicks. Collectively, these results suggest that EHC chicks might be more energetically efficient at coping with thermal challenge, preferentially allocating nutrients to other tissues while protecting the mucosal layer from oxidative damage. These results provide targets for future studies aimed at understanding the molecular mechanisms underlying effects of embryonic heat exposure on intestinal function and stressor resiliency later in life.
Collapse
Affiliation(s)
- David L Beck
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Elizabeth R Gilbert
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Mark A Cline
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
30
|
Młynarska E, Jakubowska P, Frąk W, Gajewska A, Sornowska J, Skwira S, Wasiak J, Rysz J, Franczyk B. Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases. Nutrients 2024; 16:3570. [PMID: 39458564 PMCID: PMC11510709 DOI: 10.3390/nu16203570] [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: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Recent research highlights the growing interest in the impact of nutrition on cognitive health and function in disease, as dietary habits are increasingly recognized as crucial factors in relation to brain function. This focus is especially important given the rising prevalence of neurodegenerative diseases and the cognitive decline associated with poor dietary choices. Links are now being sought between brain function and the microbiota and gut-brain axis. Mechanisms are proposed that include low-grade chronic neuroinflammation, the influence of short-chain fatty acids, or the disruption of glial cells and transmitters in the brain. METHODS We reviewed the articles on pubmed. This is not a systematic review, but of the narrative type. We wanted to outline the issue and summarise the latest information. RESULTS The axis in question has its foundation in nutrition. It has been reported that diet, particularly the components and the timing of food intake, has an impact on cognitive processes. The Mediterranean diet is most often cited in the literature as being beneficial to health. In order to obtain a more complete view, it is worth considering other dietary patterns, even those that impair our health. CONCLUSIONS Determining what is beneficial and what is not will allow us to develop a speronized strategy for the prevention of, and fight against, cognitive impairment. Appropriately selected supplements, the functions of which we have also discussed, may prove supportive.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Paulina Jakubowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Weronika Frąk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Sornowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Sylwia Skwira
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
31
|
Złotkowska D, Markiewicz LH, Ogrodowczyk AM, Wróblewska B, Wasilewska E. Enhanced Effect of β-Lactoglobulin Immunization in Mice with Mild Intestinal Deterioration Caused by Low-Dose Dextran Sulphate Sodium: A New Experimental Approach to Allergy Studies. Nutrients 2024; 16:3430. [PMID: 39458426 PMCID: PMC11510979 DOI: 10.3390/nu16203430] [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: 08/28/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Cow's milk allergy is one of the most common food allergies in children, and its pathomechanism is still under investigation. Recently, an increasing number of studies have linked food allergy to intestinal barrier dysfunction. The present study aimed to investigate changes in the intestinal microenvironment during the development of β-lactoglobulin (β-lg) allergy under conditions of early intestinal dysfunction. METHODS BALB/c mice received intraperitoneal β-lg with Freund's adjuvant, followed by oral β-lg while receiving dextran sulphate sodium salt (DSS) in their drinking water (0.2% w/v). The immunized group without DSS and the groups receiving saline, oral β-lg, or DSS served as controls. RESULTS The study showed that the immunization effect was greater in mice with mild intestinal barrier dysfunction. Although DSS did not affect the mice's humoral response to β-lg, in combination with β-lg, it significantly altered their cellular response, affecting the induction and distribution of T cells in the inductive and peripheral tissues and the activation of immune mediators. Administration of β-lg to sensitized mice receiving DSS increased disease activity index (DAI) scores and pro-inflammatory cytokine activity, altered the distribution of claudins and zonulin 1 (ZO-1) in the colonic tissue, and negatively affected the balance and activity of the gut microbiota. CONCLUSIONS The research model used appears attractive for studying food allergen sensitization, particularly in relation to the initial events leading to mucosal inflammation and the development of food hypersensitivity.
Collapse
Affiliation(s)
| | | | | | | | - Ewa Wasilewska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10 Str., 10-748 Olsztyn, Poland
| |
Collapse
|
32
|
Chen Y, Li Y, Li X, Fang Q, Li F, Chen S, Chen W. Indole‑3‑propionic acid alleviates intestinal epithelial cell injury via regulation of the TLR4/NF‑κB pathway to improve intestinal barrier function. Mol Med Rep 2024; 30:189. [PMID: 39219265 PMCID: PMC11350629 DOI: 10.3892/mmr.2024.13313] [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: 03/29/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Indole‑3‑propionic acid (IPA), a product of Clostridium sporogenes metabolism, has been shown to improve intestinal barrier function. In the present study, in vitro experiments using NCM460 human colonic epithelial cells were performed to investigate how IPA alleviates lipopolysaccharide (LPS)‑induced intestinal epithelial cell injury, with the aim of improving intestinal barrier function. In addition, the underlying mechanism was explored. NCM460 cell viability and apoptosis were measured using the Cell Counting Kit‑8 assay and flow cytometry, respectively. The integrity of the intestinal epithelial barrier was evaluated by measuring transepithelial electrical resistance (TEER). The underlying molecular mechanism was explored using western blotting, immunofluorescence staining, a dual luciferase reporter gene assay and quantitative PCR. The results showed that 10 µg/ml LPS induced the most prominent decrease in cell viability after 24 h of treatment. By contrast, IPA effectively inhibited LPS‑induced apoptosis in the intestinal epithelial cells. Additionally, >0.5 mM IPA improved intestinal barrier function by increasing TEER and upregulating the expression of tight junction proteins (zonula occludens‑1, claudin‑1 and occludin). Furthermore, IPA inhibited the release of pro‑inflammatory cytokines (IL‑1β, IL‑6 and TNF‑α) in a dose‑dependent manner and this was achieved via regulation of the Toll‑like receptor 4 (TLR4)/myeloid differentiation factor 88/NF‑κB and TLR4/TRIF/NF‑κB pathways. In conclusion, IPA may alleviate LPS‑induced inflammatory injury in human colonic epithelial cells. Taken together, these results suggest that IPA may be a potential therapeutic approach for the management of diseases characterized by LPS‑induced intestinal epithelial cell injury and intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Ying Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
| | - Yu Li
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
| | - Xiaojuan Li
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
| | - Qingqing Fang
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
| | - Feng Li
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Shiyao Chen
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
33
|
Macura B, Kiecka A, Szczepanik M. Intestinal permeability disturbances: causes, diseases and therapy. Clin Exp Med 2024; 24:232. [PMID: 39340718 PMCID: PMC11438725 DOI: 10.1007/s10238-024-01496-9] [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: 08/20/2024] [Accepted: 09/22/2024] [Indexed: 09/30/2024]
Abstract
Nowadays, a pathological increase in the permeability of the intestinal barrier (the so-called leaky gut) is increasingly being diagnosed. This condition can be caused by various factors, mainly from the external environment. Damage to the intestinal barrier entails a number of adverse phenomena: dysbiosis, translocation of microorganisms deep into the intestinal tissue, immune response, development of chronic inflammation. These phenomena can ultimately lead to a vicious cycle that promotes the development of inflammation and further damage to the barrier. Activated immune cells in mucosal tissues with broken barriers can migrate to other organs and negatively affect their functioning. Damaged intestinal barrier can facilitate the development of local diseases such as irritable bowel disease, inflammatory bowel disease or celiac disease, but also the development of systemic inflammatory diseases such as rheumatoid arthritis, ankylosing spondylitis, hepatitis, and lupus erythematosus, neurodegenerative or psychiatric conditions, or metabolic diseases such as diabetes or obesity. However, it must be emphasized that the causal links between a leaky gut barrier and the onset of certain diseases often remain unclear and require in-depth research. In light of recent research, it becomes crucial to prevent damage to the intestinal barrier, as well as to develop therapies for the barrier when it is damaged. This paper presents the current state of knowledge on the causes, health consequences and attempts to treat excessive permeability of the intestinal barrier.
Collapse
Affiliation(s)
- Barbara Macura
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland.
| | - Aneta Kiecka
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland
| | - Marian Szczepanik
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland
| |
Collapse
|
34
|
Katimbwa DA, Kim Y, Kim MJ, Jeong M, Lim J. Solubilized β-Glucan Supplementation in C57BL/6J Mice Dams Augments Neurodevelopment and Cognition in the Offspring Driven by Gut Microbiome Remodeling. Foods 2024; 13:3102. [PMID: 39410136 PMCID: PMC11476385 DOI: 10.3390/foods13193102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
A maternal diet rich in dietary fiber, such as β-glucan, plays a crucial role in the offspring's acquisition of gut microbiota and the subsequent shaping of its microbiome profile and metabolome. This in turn has been shown to aid in neurodevelopmental processes, including early microglial maturation and immunomodulation via metabolites like short chain fatty acids (SCFAs). This study aimed to investigate the effects of oat β-glucan supplementation, solubilized by citric acid hydrolysis, from gestation to adulthood. Female C57BL/6J mice were orally supplemented with soluble oat β-glucan (ObG) or carboxymethyl cellulose (CMC) via drinking water at 200 mg/kg body weight during breeding while the control group received 50 mg/kg body weight of carboxymethyl cellulose. ObG supplementation increased butyrate production in the guts of both dams and 4-week-old pups, attributing to alterations in the gut microbiota profile. One-week-old pups from the ObG group showed increased neurodevelopmental markers similar to four-week-old pups that also exhibited alterations in serum markers of metabolism and anti-inflammatory cytokines. Notably, at 8 weeks, ObG-supplemented pups exhibited the highest levels of spatial memory and cognition compared to the control and CMC groups. These findings suggest a potential enhancement of neonatal neurodevelopment via shaping of early-life gut microbiome profile, and the subsequent increased later-life cognitive function.
Collapse
Affiliation(s)
- Dorsilla A. Katimbwa
- Department of Food Biomaterials, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Yoonsu Kim
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Min Jeong Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Minsoo Jeong
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jinkyu Lim
- Department of Food Biomaterials, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
35
|
Qi P, Chen X, Tian J, Zhong K, Qi Z, Li M, Xie X. The gut homeostasis-immune system axis: novel insights into rheumatoid arthritis pathogenesis and treatment. Front Immunol 2024; 15:1482214. [PMID: 39391302 PMCID: PMC11464316 DOI: 10.3389/fimmu.2024.1482214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Rheumatoid arthritis is a widely prevalent autoimmune bone disease that imposes a significant burden on global healthcare systems due to its increasing incidence. In recent years, attention has focused on the interaction between gut homeostasis and the immune system, particularly in relation to bone health. Dysbiosis, which refers to an imbalance in the composition and function of the gut microbiota, has been shown to drive immune dysregulation through mechanisms such as the release of pro-inflammatory metabolites, increased gut permeability, and impaired regulatory T cell function. These factors collectively contribute to immune system imbalance, promoting the onset and progression of Rheumatoid arthritis. Dysbiosis induces both local and systemic inflammatory responses, activating key pro-inflammatory cytokines such as tumor necrosis factor-alpha, Interleukin-6, and Interleukin-17, which exacerbate joint inflammation and damage. Investigating the complex interactions between gut homeostasis and immune regulation in the context of Rheumatoid arthritis pathogenesis holds promise for identifying new therapeutic targets, revealing novel mechanisms of disease progression, and offering innovative strategies for clinical treatment.
Collapse
Affiliation(s)
- Peng Qi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xin Chen
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jiexiang Tian
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Kexin Zhong
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhonghua Qi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Menghan Li
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xingwen Xie
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
36
|
Chatterjee P, Spalinger MR, Acevedo C, Gries CM, Manz SM, Canale V, Santos AN, Shawki A, Sayoc-Becerra A, Lei H, Crawford MS, Eckmann L, Borneman J, McCole DF. Intestinal Epithelial PTPN2 Limits Pathobiont Colonization by Immune-Directed Antimicrobial Responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614848. [PMID: 39386684 PMCID: PMC11463449 DOI: 10.1101/2024.09.24.614848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Background and Aims Loss of activity of the inflammatory bowel disease (IBD) susceptibility gene, protein tyrosine phosphatase non-receptor type 2 (PTPN2), is associated with altered microbiome composition in both human subjects and mice. Further, expansion of the bacterial pathobiont, adherent-invasive E. coli (AIEC), is strongly linked to IBD pathogenesis. The mechanism by which intestinal epithelial cells (IEC) maintain equilibrium between commensal microbiota and immune cells to restrict invading pathobionts is poorly understood. Here, we investigated the role of IEC-specific PTPN2 in regulating AIEC colonization. Methods Tamoxifen-inducible, intestinal epithelial cell-specific Ptpn2 knockout mice (Ptpn2 ΔIEC) and control Ptpn2 fl/fl mice were infected with either non-invasive E. coli K12, or fluorescent-tagged mAIEC (mAIECred) for four consecutive days or administered PBS. Subsequently, bacterial colonization in mouse tissues was quantified. mRNA and protein expression were assayed in intestinal epithelial cells (IECs) or whole tissue lysates by PCR and Western blot. Tissue cytokine expression was determined by ELISA. Intestinal barrier function was determined by in vivo administration of 4 kDa FITC-dextran (FD4) or 70kDa Rhodamine-B dextran (RD70) fluorescent probes. Confocal microscopy was used to determine the localization of tight-junction proteins. Results Ptpn2 ΔIEC mice exhibited increased mAIECred - but not K12 - bacterial load in the distal colon compared to infected Ptpn2 fl/fl mice. The higher susceptibility to mAIECred infection was associated with altered levels of antimicrobial peptide (AMPs). Ileal RNA expression of the alpha-defensin AMPs, Defa5 and Defa6, as well as MMP7, was significantly lower in Ptpn2 ΔIEC vs. Ptpn2 fl/fl mice, after mAIECred but not K12 infection. Further, we observed increased tight junction-regulated permeability determined by elevated in vivo FD4 but not RD70 permeability in Ptpn2 ΔIEC-K12 mice compared to their respective controls. This effect was further exacerbated in Ptpn2 ΔIEC mAIEC-infected mice. Further, Ptpn2 ΔIEC mice displayed lower IL-22, IL-6, IL-17A cytokine expression post mAIEC infection compared to Ptpn2 fl/fl controls. Recombinant IL-22 reversed the FD4 permeability defect and reduced bacterial burden in Ptpn2 ΔIEC mice post mAIEC challenge. Conclusion Our findings highlight that intestinal epithelial PTPN2 is crucial for mucosal immunity and gut homeostasis by promoting anti-bacterial defense mechanisms involving coordinated epithelial-immune responses to restrict pathobiont colonization.
Collapse
Affiliation(s)
- Pritha Chatterjee
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Marianne R. Spalinger
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Charly Acevedo
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Casey M. Gries
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Salomon M. Manz
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Vinicius Canale
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Alina N. Santos
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Ali Shawki
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Anica Sayoc-Becerra
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Hillmin Lei
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Meli’sa S. Crawford
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| | - Lars Eckmann
- Division of Gastroenterology, University of California, San Diego, La Jolla, California
| | - James Borneman
- Department of Microbiology and Plant Pathology, University of California, Riverside, Riverside, California
| | - Declan F. McCole
- Division of Biomedical Sciences, University of California, Riverside, Riverside, California
| |
Collapse
|
37
|
Walker AC, Bhargava R, Bucher MJ, Argote YM, Brust AS, Czyż DM. Identification of proteotoxic and proteoprotective bacteria that non-specifically affect proteins associated with neurodegenerative diseases. iScience 2024; 27:110828. [PMID: 39310761 PMCID: PMC11414702 DOI: 10.1016/j.isci.2024.110828] [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: 10/24/2023] [Revised: 04/05/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
There are no cures for neurodegenerative protein conformational diseases (PCDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Emerging evidence suggests the gut microbiota plays a role in their pathogenesis, though the influences of specific bacteria on disease-associated proteins remain elusive. Here, we reveal the effects of 229 human bacterial isolates on the aggregation and toxicity of Aβ1-42, α-synuclein, and polyglutamine tracts in Caenorhabditis elegans expressing these culprit proteins. Our findings demonstrate that bacterial effects on host protein aggregation are consistent across different culprit proteins, suggesting that microbes affect protein stability by modulating host proteostasis rather than selectively targeting disease-associated proteins. Furthermore, we found that feeding C. elegans proteoprotective Prevotella corporis activates the heat shock response, revealing an unexpected discovery of a microbial influence on host proteostasis. Insight into how individual bacteria affect PCD proteins could open new strategies for prevention and treatment by altering the abundance of microbes.
Collapse
Affiliation(s)
- Alyssa C. Walker
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Rohan Bhargava
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Michael J. Bucher
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Yoan M. Argote
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Amanda S. Brust
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Daniel M. Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
38
|
Chen H, Cao J, Zhang F, Xiong W. Significance of Gut Microbiota on Graves' Disease. Int J Gen Med 2024; 17:3967-3974. [PMID: 39281039 PMCID: PMC11402343 DOI: 10.2147/ijgm.s467888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/13/2024] [Indexed: 09/18/2024] Open
Abstract
Growing research proves gut microbiota and thyroid autoimmunity are linked. Graves' disease (GD), as an autoimmune thyroid disease (AITD), is attributed to the production of thyroid-stimulating hormone receptor (TSHR) autoantibodies that bind to the thyroid follicular endothelial cells. It is well known that genetic factors, environmental factors, and immune disorders count for much in the development of GD. So far, the pathogenesis of GD is not elucidated. Emerging research reveals that the change in gut microbiota composition and its metabolites are related to GD. The gut microbial diversity is reduced in GDs compared with healthy controls (HCs). Firmicutes and Bacteroidetes account for a large proportion at the genus level. It is found that phyla Bacteroidetes increased while phyla Firmicutes decreased in Graves' Disease patients (GD patients). Moreover, gut microbiota modulates the immune system to produce cytokines through bacterial metabolites. This article aims to find out the relation between gut microbiota dysbiosis and the development of GD. As more molecular pathways of bacterial metabolites are revealed, targeting microbiota is expected to the treatment of GD.
Collapse
Affiliation(s)
- Haiyan Chen
- Wuzhou Workers Hospital, Wuzhou, Guangxi Zhuang, People's Republic of China
| | - Jiamin Cao
- Department of Ophthalmology, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Feng Zhang
- Department of Ophthalmology, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Wei Xiong
- Department of Ophthalmology, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| |
Collapse
|
39
|
Retuerto M, Al-Shakhshir H, Herrada J, McCormick TS, Ghannoum MA. Analysis of Gut Bacterial and Fungal Microbiota in Children with Autism Spectrum Disorder and Their Non-Autistic Siblings. Nutrients 2024; 16:3004. [PMID: 39275319 PMCID: PMC11396985 DOI: 10.3390/nu16173004] [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: 08/02/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a multifactorial disorder involving genetic and environmental factors leading to pathophysiologic symptoms and comorbidities including neurodevelopmental disorders, anxiety, immune dysregulation, and gastrointestinal (GI) abnormalities. Abnormal intestinal permeability has been reported among ASD patients and it is well established that disturbances in eating patterns may cause gut microbiome imbalance (i.e., dysbiosis). Therefore, studies focusing on the potential relationship between gut microbiota and ASD are emerging. We compared the intestinal bacteriome and mycobiome of a cohort of ASD subjects with their non-ASD siblings. Differences between ASD and non-ASD subjects include a significant decrease at the phylum level in Cyanobacteria (0.015% vs. 0.074%, p < 0.0003), and a significant decrease at the genus level in Bacteroides (28.3% vs. 36.8%, p < 0.03). Species-level analysis showed a significant decrease in Faecalibacterium prausnitzii, Prevotella copri, Bacteroides fragilis, and Akkermansia municiphila. Mycobiome analysis showed an increase in the fungal Ascomycota phylum (98.3% vs. 94%, p < 0.047) and an increase in Candida albicans (27.1% vs. 13.2%, p < 0.055). Multivariate analysis showed that organisms from the genus Delftia were predictive of an increased odds ratio of ASD, whereas decreases at the phylum level in Cyanobacteria and at the genus level in Azospirillum were associated with an increased odds ratio of ASD. We screened 24 probiotic organisms to identify strains that could alter the growth patterns of organisms identified as elevated within ASD subject samples. In a preliminary in vivo preclinical test, we challenged wild-type Balb/c mice with Delftia acidovorans (increased in ASD subjects) by oral gavage and compared changes in behavioral patterns to sham-treated controls. An in vitro biofilm assay was used to determine the ability of potentially beneficial microorganisms to alter the biofilm-forming patterns of Delftia acidovorans, as well as their ability to break down fiber. Downregulation of cyanobacteria (generally beneficial for inflammation and wound healing) combined with an increase in biofilm-forming species such as D. acidovorans suggests that ASD-related GI symptoms may result from decreases in beneficial organisms with a concomitant increase in potential pathogens, and that beneficial probiotics can be identified that counteract these changes.
Collapse
Affiliation(s)
- Mauricio Retuerto
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hilmi Al-Shakhshir
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Janet Herrada
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thomas S McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mahmoud A Ghannoum
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
- Center for Medical Mycology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
40
|
Safdar M, Ullah M, Hamayun S, Wahab A, Khan SU, Abdikakhorovich SA, Haq ZU, Mehreen A, Naeem M, Mustopa AZ, Hasan N. Microbiome miracles and their pioneering advances and future frontiers in cardiovascular disease. Curr Probl Cardiol 2024; 49:102686. [PMID: 38830479 DOI: 10.1016/j.cpcardiol.2024.102686] [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: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024]
Abstract
Cardiovascular diseases (CVDs) represent a significant global health challenge, underscoring the need for innovative approaches to prevention and treatment. Recent years have seen a surge in interest in unraveling the complex relationship between the gut microbiome and cardiovascular health. This article delves into current research on the composition, diversity, and impact of the gut microbiome on CVD development. Recent advancements have elucidated the profound influence of the gut microbiome on disease progression, particularly through key mediators like Trimethylamine-N-oxide (TMAO) and other microbial metabolites. Understanding these mechanisms reveals promising therapeutic targets, including interventions aimed at modulating the gut microbiome's interaction with the immune system and its contribution to endothelial dysfunction. Harnessing this understanding, personalized medicine strategies tailored to individuals' gut microbiome profiles offer innovative avenues for reducing cardiovascular risk. As research in this field continues to evolve, there is vast potential for transformative advancements in cardiovascular medicine, paving the way for precision prevention and treatment strategies to address this global health challenge.
Collapse
Affiliation(s)
- Mishal Safdar
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Punjab, Pakistan
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Shah Hamayun
- Department of Cardiology, Pakistan Institute of Medical Sciences (PIMS), Islamabad, 04485 Punjab, Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Shahid Ullah Khan
- Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, 22080, Khyber Pakhtunkhwa, Pakistan
| | | | - Zia Ul Haq
- Department of Public Health, Institute of Public Health Sciences, Khyber Medical University, Peshawar 25120, Pakistan
| | - Aqsa Mehreen
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Punjab, Pakistan
| | - Muhammad Naeem
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Punjab, Pakistan
| | - Apon Zaenal Mustopa
- Research Center for Genetic Engineering, National Research, and Innovation Agency (BRIN), Bogor 16911, Indonesia
| | - Nurhasni Hasan
- Faculty of Pharmacy, Universitas Hasanuddin, Jl. Perintis Kemerdekaan Km 10, Makassar 90245, Republic of Indonesia.
| |
Collapse
|
41
|
Yang C, Xiao J, Xu Z, Wang Z. Gut Microbiota Changes and Its Potential Relations with Thyroid Disorders: From Composition to Therapeutic Targets. Int J Gen Med 2024; 17:3719-3731. [PMID: 39219667 PMCID: PMC11363920 DOI: 10.2147/ijgm.s481183] [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: 06/03/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Composed of over 1200 species of anaerobes and aerobes bacteria along with bacteriophages, viruses, and fungal species, the human gut microbiota (GM) is vital to health, including digestive equilibrium, immunologic, hormonal, and metabolic homeostasis. Micronutrients, usually refer to trace elements (copper, iodine, iron, selenium, zinc) and vitamins (A, C, D, E), interact with the GM to influence host immune metabolism. So far, microbiome studies have revealed an association between disturbances in the microbiota and various pathological disorders, such as anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis, anxiety, depression, early-onset cancers, type 1 diabetes (T1D) and type 2 diabetes (T2D). As common conditions, thyroid diseases, encompassing Graves' disease (GD), Graves' orbitopathy (GO), Hashimoto's thyroiditis (HT), benign nodules, and papillary thyroid cancer (TC), have negative impacts on the health of all populations. Following recent studies, GM might play an integral role in triggering diseases of the thyroid gland. Not only do environmental triggers and genetic predisposing background lead to auto-aggressive damage, involving cellular and humoral networks of the immune system, but the intestinal microbiota interacts with distant organs by signals that may be part of the bacteria themselves or their metabolites. The review aims to describe the current knowledge about the GM in the metabolism of thyroid hormones and the pathogenesis of thyroid diseases and its involvement in the appearance of benign nodules and papillary TC. We further focused on the reciprocal interaction between GM composition and the most used treatment drugs for thyroid disorders. However, the exact etiology has not yet been known. To elucidate more precisely the mechanism for GM involvement in the development of thyroid diseases, future work is needed.
Collapse
Affiliation(s)
- Cai Yang
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Jiafeng Xiao
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Zibei Xu
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Zehong Wang
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| |
Collapse
|
42
|
Gangaiah D, Gu M, Zaparte A, Will O, Dolan LC, Goering A, Pillai J, Mane SP, Plata G, Helmes EB, Welsh DA, Mahajan AK. Effects of Limosilactobacillus reuteri strains PTA-126787 and PTA-126788 on intestinal barrier integrity and immune homeostasis in an alcohol-induced leaky gut model. Sci Rep 2024; 14:19584. [PMID: 39179898 PMCID: PMC11344072 DOI: 10.1038/s41598-024-70549-6] [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: 06/17/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
Intestinal barrier is a first line of defense that prevents entry of various harmful substances from the lumen into the systemic environment. Impaired barrier function with consequent translocation of harmful substances into systemic circulation ("leaky gut") is a central theme in many gastrointestinal, autoimmune, mental, and metabolic diseases. Probiotics have emerged as a promising strategy to maintain intestinal integrity and address "leaky gut". Using in silico, in vitro and avian in vivo analyses, we previously showed that two novel L. reuteri strains, PTA-126787 (L. reuteri 3630) and PTA-126788 (L. reuteri 3632), isolated from broiler chickens possess favorable safety profiles. Consistent with a recent study, here we show that L. reuteri 3630 and 3632 are phylogenetically similar to human L. reuteri strains. Daily administration of high doses of L. reuteri 3630 and 3632 to Sprague Dawley rats for 28 days was found to be safe with no adverse effects. More importantly, administration of L. reuteri 3630 and 3632 significantly reduced markers associated with alcohol-induced leaky gut, by downregulating inflammatory cytokines and upregulating anti-inflammatory cytokines in an alcohol model of leaky gut in mice. While L. reuteri 3630 cells and supernatant showed no activation, L. reuteri 3632 cells but not supernatant showed activation of AhR, a key transcription factor that regulates gut and immune homeostasis. L. reuteri 3630 is creamish white in morphology typical of Lactobacillus species and L. reuteri 3632 displays a unique orange pigmentation, which was stable even after passaging for 480 generations. We identified a rare polyketide biosynthetic gene cluster in L. reuteri 3632 that likely encodes for the orange-pigmented secondary metabolite. Similar to L. reuteri 3632 cells, the purified orange metabolite activated AhR. All together, these data provide evidence on the phylogenetic relatedness, safety, efficacy, and one of the likely mechanisms of action of L. reuteri 3630 and 3632 for potential probiotic applications to address "leaky gut" and associated pathologies in humans.
Collapse
Affiliation(s)
| | - Min Gu
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Aline Zaparte
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Olaf Will
- Elanco Animal Health, Inc., Alfred-Nobel-Strasse 50, 40789, Monheim Am Rhein, Germany
| | - Laurie C Dolan
- GRAS Associates, 1180 Grand Park Avenue, North Bethesda, MD, 20852, USA
| | | | - Jason Pillai
- MicroMGx, Inc., 3440 S Dearborn St, Chicago, IL, 60616, USA
| | | | - German Plata
- BiomEdit, LLC, 2710 Innovation Way, Greenfield, IN, 46140, USA
| | - Emily B Helmes
- BiomEdit, LLC, 2710 Innovation Way, Greenfield, IN, 46140, USA
| | - David A Welsh
- Department of Medicine, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | | |
Collapse
|
43
|
Sikdar S, Mitra D, Das O, Bhaumik M, Dutta S. The functional antagonist of sphingosine-1-phosphate, FTY720, impairs gut barrier function. Front Pharmacol 2024; 15:1407228. [PMID: 39224783 PMCID: PMC11366638 DOI: 10.3389/fphar.2024.1407228] [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: 03/26/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
FTY720 or fingolimod is a known functional antagonist of sphingosine-1-phosphate (S1P), and it is effective in treating multiple sclerosis and preventing inflammatory bowel disease (IBD). Evidence shows that its use in mice can increase the susceptibility to mucosal infections. Despite the significant contribution of S1P to barrier function, the effect of the administration of FTY720 on the mucosal barrier has never been investigated. In this study, we looked into how FTY720 therapy affected the function of the gut barrier susceptibility. Administration of FTY720 to C57BL/6 mice enhances the claudin-2 expression and reduces the expression of claudin-4 and occludin, as studied by qPCR, Western blot, and immunofluorescence. FTY720 inhibits the Akt-mTOR pathway to decrease occludin and claudin-4 expression and increase claudin-2 expression. FTY720 treatment induced increased colonic inflammation, with notably greater immune cell infiltration, colon histopathology, and increased production of TNF-α, IFN-γ, CXCL-1, and CXCL-2 than that in control mice. Taking into account the close association of "the leaky gut" and gut dysbiosis among the major diseases, we therefore can infer that the vigilance of gut pathology should be maintained, where FTY720 is used as a treatment option.
Collapse
Affiliation(s)
- Sohini Sikdar
- Division of Immunology , ICMR-National Institute for Research in Bacterial Infections (NIRBI), Kolkata, India
| | - Debmalya Mitra
- Center of Radiological Research, Columbia University Irving Medical Center, New York, NY, United States
| | - Oishika Das
- Division of Immunology , ICMR-National Institute for Research in Bacterial Infections (NIRBI), Kolkata, India
| | - Moumita Bhaumik
- Division of Immunology , ICMR-National Institute for Research in Bacterial Infections (NIRBI), Kolkata, India
| | - Shanta Dutta
- Division of Immunology , ICMR-National Institute for Research in Bacterial Infections (NIRBI), Kolkata, India
| |
Collapse
|
44
|
Zhu G, Yan L, Fang L, Fan C, Sun H, Zhou X, Zhang Y, Shi Z. Possible immune mechanisms of gut microbiota and its metabolites in the occurrence and development of immune thrombocytopenia. Front Microbiol 2024; 15:1426911. [PMID: 39171254 PMCID: PMC11335631 DOI: 10.3389/fmicb.2024.1426911] [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: 05/02/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease characterized by increased platelet destruction and impaired production, leading to an elevated bleeding tendency. Recent studies have demonstrated an important link between the gut microbiota and the onset and progression of several immune diseases in humans, emphasizing that gut microbiota-derived metabolites play a non-negligible role in autoimmune diseases. The gut microbiota and its metabolites, such as short-chain fatty acids, oxidized trimethylamine, tryptophan metabolites, secondary bile acids and lipopolysaccharides, can alter intestinal barrier permeability by modulating immune cell differentiation and cytokine secretion, which in turn affects the systemic immune function of the host. It is therefore reasonable to hypothesize that ecological dysregulation of the gut microbiota may be an entirely new factor in the triggering of ITP. This article reviews the potential immune-related mechanisms of the gut microbiota and representative metabolites in ITP, as well as the important influence of leaky gut on the development of ITP, with a view to enriching the theoretical system of ITP-related gut microecology and providing new ideas for the study of ITP.
Collapse
Affiliation(s)
- Gengda Zhu
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lixiang Yan
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lijun Fang
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Hematology and Blood Diseases Hospital, National Clinical Medical Research Center for Blood Diseases, Tianjin, China
| | - Chenyang Fan
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Sun
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinli Zhou
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yucheng Zhang
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhexin Shi
- National Medical Research Center of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
45
|
Kinashi Y, Tanaka K, Kimura S, Hirota M, Komiyama S, Shindo T, Hashiguchi A, Takahashi D, Shibata S, Karaki SI, Ohno H, Hase K. Intestinal epithelium dysfunctions cause IgA deposition in the kidney glomeruli of intestine-specific Ap1m2-deficient mice. EBioMedicine 2024; 106:105256. [PMID: 39059316 PMCID: PMC11338063 DOI: 10.1016/j.ebiom.2024.105256] [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: 02/01/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Intestinal epithelial cells (IECs) serve as robust barriers against potentially hostile luminal antigens and commensal microbiota. Epithelial barrier dysfunction enhances intestinal permeability, leading to leaky gut syndrome (LGS) associated with autoimmune and chronic inflammatory disorders. However, a causal relationship between LGS and systemic disorders remains unclear. Ap1m2 encodes clathrin adaptor protein complex 1 subunit mu 2, which facilitates polarized protein trafficking toward the basolateral membrane and contributes to the establishment of epithelial barrier functions. METHODS We generated IEC-specific Ap1m2-deficient (Ap1m2ΔIEC) mice with low intestinal barrier integrity as an LSG model and examined the systemic impact. FINDINGS Ap1m2ΔIEC mice spontaneously developed IgA nephropathy (IgAN)-like features characterized by the deposition of IgA-IgG immune complexes and complement factors in the kidney glomeruli. Ap1m2 deficiency markedly enhanced aberrantly glycosylated IgA in the serum owing to downregulation and mis-sorting of polymeric immunoglobulin receptors in IECs. Furthermore, Ap1m2 deficiency caused intestinal dysbiosis by attenuating IL-22-STAT3 signaling. Intestinal dysbiosis contributed to the pathogenesis of IgAN because antibiotic treatment reduced aberrantly glycosylated IgA production and renal IgA deposition in Ap1m2ΔIEC mice. INTERPRETATION IEC barrier dysfunction and subsequent dysbiosis by AP-1B deficiency provoke IgA deposition in the mouse kidney. Our findings provide experimental evidence of a pathological link between LGS and IgAN. FUNDING AMED, AMED-CREST, JSPS Grants-in-Aid for Scientific Research, JST CREST, Fuji Foundation for Protein Research, and Keio University Program for the Advancement of Next Generation Research Projects.
Collapse
Affiliation(s)
- Yusuke Kinashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Keisuke Tanaka
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Shunsuke Kimura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan.
| | - Masato Hirota
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Seiga Komiyama
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Akinori Hashiguchi
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan; Depatment of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Takahashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan; Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shin-Ichiro Karaki
- Laboratory of Physiology, Department of Environmental and Life Sciences, University of Shizuoka, Shizuoka, Japan
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan; Laboratory for Immune Regulation, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan; Institute of Fermentation Sciences (IFeS), Faculty of Food and Agricultural Sciences, Fukushima University, Fukushima, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan.
| |
Collapse
|
46
|
Mahran A, Hosni AM, Farag NG, Elkhawaga AA, Mageed AAA. Role of Claudin- 3 as a biomarker of gut-skin axis integrity in patients with psoriasis. Arch Dermatol Res 2024; 316:476. [PMID: 39023797 DOI: 10.1007/s00403-024-03071-4] [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: 02/16/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 07/20/2024]
Abstract
Increased intestinal permeability and gut dysbiosis are important factors in the pathophysiology of psoriasis and its associated conditions. Claudin-3 is a protein that is found in tight junctions and may be used to assess the integrity of the gut barrier. The aim of this study was to investigate serum concentration of Claudin- 3 (CLDN3) in patients with psoriasis. Exploring its possible relations with patients' demographic, clinical and laboratory findings was another objective. Fifty psoriatic patients and thirty-five age- and sex-matched healthy volunteers served as the study's control group in this case-control, hospital-based research. The amount of serum CLDN3 was determined by means of an enzyme-linked immunosorbent test (ELISA). Concentration of serum CLDN3 was found to be significantly higher in patients with psoriasis. (p = 0.002). There was no statistically significant correlation between CLDN3 and patient's clinical & laboratory variables. We demonstrated that gut permeability is dysfunctional in patients with psoriasis as indicated by reduction of serum CLDN3. Further investigations are needed to determine whether modulation of gut barrier may represent a new therapeutic approach for psoriasis.
Collapse
Affiliation(s)
- Ayman Mahran
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amal Mohammed Hosni
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Nesma G Farag
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amal A Elkhawaga
- Department of Medical Microbiology and immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed A Abdel Mageed
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
47
|
Młynarska E, Wasiak J, Gajewska A, Steć G, Jasińska J, Rysz J, Franczyk B. Exploring the Significance of Gut Microbiota in Diabetes Pathogenesis and Management-A Narrative Review. Nutrients 2024; 16:1938. [PMID: 38931292 PMCID: PMC11206785 DOI: 10.3390/nu16121938] [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: 05/17/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Type 2 diabetes is a disease with significant health consequences for the individual. Currently, new mechanisms and therapeutic approaches that may affect this disease are being sought. One of them is the association of type 2 diabetes with microbiota. Through the enteric nervous system and the gut-microbiota axis, the microbiota affects the functioning of the body. It has been proven to have a real impact on influencing glucose and lipid metabolism and insulin sensitivity. With dysbiosis, there is increased bacterial translocation through the disrupted intestinal barrier and increased inflammation in the body. In diabetes, the microbiota's composition is altered with, for example, a more abundant class of Betaproteobacteria. The consequences of these disorders are linked to mechanisms involving short-chain fatty acids, branched-chain amino acids, and bacterial lipopolysaccharide, among others. Interventions focusing on the gut microbiota are gaining traction as a promising approach to diabetes management. Studies are currently being conducted on the effects of the supply of probiotics and prebiotics, as well as fecal microbiota transplantation, on the course of diabetes. Further research will allow us to fully develop our knowledge on the subject and possibly best treat and prevent type 2 diabetes.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Greta Steć
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Jasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
48
|
Kimilu N, Gładyś-Cieszyńska K, Pieszko M, Mańkowska-Wierzbicka D, Folwarski M. Carrageenan in the Diet: Friend or Foe for Inflammatory Bowel Disease? Nutrients 2024; 16:1780. [PMID: 38892712 PMCID: PMC11174395 DOI: 10.3390/nu16111780] [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: 05/13/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
While the exact pathogenesis of IBD remains unclear, genetic, environmental and nutritional factors as well as the composition of the gut microbiome play crucial roles. Food additives, which are increasingly consumed in the Western diet, are being investigated for their potential effects on IBD. These additives can affect gut health by altering the composition of the microbiota, immune responses, and intestinal permeability, contributing to autoimmune diseases and inflammation. Despite the growing number of studies on food additives and IBD, the specific effects of carrageenan have not yet been sufficiently researched. This review addresses this gap by critically analyzing recent studies on the effects of carrageenan on the gut microbiota, intestinal permeability, and inflammatory processes. We searched the MEDLINE and SCOPUS databases using the following terms: carrageenan, carrageenan and inflammatory bowel disease, carrageenan and cancer, food additives and microbiome, food additives and intestinal permeability, and food additives and autoimmune diseases. In animal studies, degraded carrageenan has been shown to trigger intestinal ulceration and inflammation, highlighting its potential risk for exacerbating IBD. It can affect the gut microbiota, reduce bacterial diversity, and increase intestinal permeability, contributing to "leaky gut" syndrome. Some studies suggest that carrageenan may inhibit the growth of cancer cells by influencing the progression of the cell cycle, but the anti-cancer effect is still unclear. Carrageenan may also increase glucose intolerance and insulin resistance. Further research is needed to determine whether carrageenan should be excluded from the diet of individuals with IBD.
Collapse
Affiliation(s)
- Nina Kimilu
- Students’ Scientific Circle of Clinical Nutrition, Medical University of Gdansk, 80-211 Gdansk, Poland
| | | | - Magdalena Pieszko
- Department of Clinical Nutrition and Dietetics, Medical University of Gdansk, 80-210 Gdansk, Poland (M.P.)
| | - Dorota Mańkowska-Wierzbicka
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marcin Folwarski
- Department of Clinical Nutrition and Dietetics, Medical University of Gdansk, 80-210 Gdansk, Poland (M.P.)
- Home Enteral and Parenteral Nutrition Unit, Nicolaus Copernicus Hospital, 80-803 Gdansk, Poland
| |
Collapse
|
49
|
Gorini F, Tonacci A. Vitamin D: An Essential Nutrient in the Dual Relationship between Autoimmune Thyroid Diseases and Celiac Disease-A Comprehensive Review. Nutrients 2024; 16:1762. [PMID: 38892695 PMCID: PMC11174782 DOI: 10.3390/nu16111762] [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: 05/11/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Autoimmune thyroid diseases (AITD) are among the most frequent autoimmune disorders, with a multifactorial etiology in which both genetic and environmental determinants are probably involved. Celiac disease (CeD) also represents a public concern, given its increasing prevalence due to the recent improvement of screening programs, leading to the detection of silent subtypes. The two conditions may be closely associated due to common risk factors, including genetic setting, changes in the composition and diversity of the gut microbiota, and deficiency of nutrients like vitamin D. This comprehensive review discussed the current evidence on the pivotal role of vitamin D in modulating both gut microbiota dysbiosis and immune system dysfunction, shedding light on the possible relevance of an adequate intake of this nutrient in the primary prevention of AITD and CeD. While future technology-based strategies for proper vitamin D supplementation could be attractive in the context of personalized medicine, several issues remain to be defined, including standardized assays for vitamin D determination, timely recommendations on vitamin D intake for immune system functioning, and longitudinal studies and randomized controlled trials to definitely establish a causal relationship between serum vitamin D levels and the onset of AITD and CeD.
Collapse
Affiliation(s)
- Francesca Gorini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | | |
Collapse
|
50
|
Chang CC, Wang CT, Shih HM, Ho CH, Hsu CC, Lin HJ, Chiu YW, Huang CC. Alcohol abuse may increase the risk of autoimmune connective tissue disease: a nationwide population-based cohort study. Front Psychiatry 2024; 14:1308245. [PMID: 38883846 PMCID: PMC11178937 DOI: 10.3389/fpsyt.2023.1308245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/26/2023] [Indexed: 06/18/2024] Open
Abstract
Objectives Altered immune and inflammatory responses resulting from alcohol abuse have been implicated in increasing the risk of autoimmune connective tissue disease (ACTD). However, limited research has been conducted on this topic in the Asian population. Therefore, this study was undertaken to investigate and address this knowledge gap. Methods Using data from Taiwan's National Health Insurance Research Database, we identified all patients with alcohol abuse between 2000 and 2017. We selected a comparison cohort without alcohol abuse, matching them in terms of age, sex, and index date at a 3:1 ratio. We collected information on common underlying comorbidities for analysis. Both cohorts were followed up until the diagnosis of ACTD or the end of 2018. Results A total of 57,154 patients with alcohol abuse and 171,462 patients without alcohol abuse were included in the study. The age and sex distributions were similar in both cohorts, with men accounting for 89.8% of the total. After adjusting for underlying comorbidities, patients with alcohol abuse had a higher risk of developing ACTD [adjusted hazard ratio (AHR): 1.12, 95% confidence interval (CI): 1.01-1.25]. The stratified analysis revealed that this increased risk was specific to the male population. Additionally, besides alcohol abuse, liver disease, renal disease, coronary artery disease, and chronic obstructive pulmonary disease were identified as independent predictors for ACTD. Conclusion This study demonstrates that alcohol abuse increases the risk of developing ACTD in the Asian population, particularly among men. Therefore, it is important to implement alcohol cessation, especially in individuals with liver disease, renal disease, coronary artery disease, and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Chi-Chen Chang
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chia-Ti Wang
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Hong-Mo Shih
- Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Department of Public Health, China Medical University, Taichung, Taiwan
| | - Chung-Han Ho
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Department of Information Management, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chien-Chin Hsu
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hung-Jung Lin
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Emergency Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Wei Chiu
- Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Public Health, China Medical University, Taichung, Taiwan
| | - Chien-Cheng Huang
- Department of Emergency Medicine, Chi Mei Medical Center, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Emergency Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|