1
|
Gawlik-Kotelnicka O, Gabryelska A, Sochal M, Czarnecka-Chrebelska K, Pikus E, Brzeziańska-Lasota E, Białasiewicz P, Strzelecki D. Lipopolysaccharide-binding protein levels, obstructive sleep apnea, and depression: A cross-sectional study of adults. Brain Res 2025; 1856:149575. [PMID: 40101844 DOI: 10.1016/j.brainres.2025.149575] [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: 07/01/2024] [Revised: 01/20/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Obstructive sleep apnea (OSA) and depression are highly comorbid. Increased intestinal permeability has been hypothesized to play a role in the pathogenesis of both. The current study aimed to assess the severity of OSA symptoms, comorbid depressive symptoms, and lipopolysaccharide-binding protein (LBP) levels in adult patients being diagnosed for OSA syndrome. The study population consisted of 176 subjects. An apnea-hypopnea index (AHI) ≥ 5/hour was used for the diagnosis of OSA syndrome. Depressive symptoms were assessed with the Beck Depression Inventory-2. LBP levels were measured in the blood serum by enzyme-linked immunosorbent assay (ELISA). Associations between clinical symptom profiles or severity and LBP as an intestinal permeability biomarker marker were tested. LBP levels were not different between patients with different OSA severity, as assessed with AHI or daily sleepiness. Nor were LBP levels different in subjects with different depressiveness severity. Daily sleepiness was weakly positively correlated with depression score, and LBP levels correlated positively with a neutrophils-to-lymphocytes ratio. Finally, LBP levels were not explained by multiple linear regression models, including sleep-related parameter values and depression score. Intestinal permeability, as measured with LBP level, may not explain the comorbidity of depression and daily sleepiness in the course of OSA syndrome.
Collapse
Affiliation(s)
- Oliwia Gawlik-Kotelnicka
- Department of Affective and Psychotic Disorders, Medical University of Lodz 90-419 Lodz, Poland.
| | - Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz 90-419 Lodz, Poland.
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz 90-419 Lodz, Poland.
| | | | - Ewa Pikus
- Department of Biomedicine and Genetics, Medical University of Lodz 90-419 Lodz, Poland.
| | | | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz 90-419 Lodz, Poland.
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz 90-419 Lodz, Poland.
| |
Collapse
|
2
|
Zheng Y, Yu Y, Chen M, Zhang H, Wang W, Fan X, Sun L, Tang L, Ta D. Abdominal LIPUS Stimulation Prevents Cognitive Decline in Hind Limb Unloaded Mice by Regulating Gut Microbiota. Mol Neurobiol 2025; 62:7313-7329. [PMID: 39878866 DOI: 10.1007/s12035-025-04709-8] [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] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
Weightlessness usually causes disruption of the gut microbiota and impairs cognitive function. There is a close connection between gut microbiota and neurological diseases. Low-intensity pulsed ultrasound (LIPUS) has a beneficial effect on reducing intestinal inflammation. So we wondered if abdominal LIPUS stimulation can have a positive impact on weightlessness induced cognitive decline by reducing intestinal dysfunction. The findings revealed that the hind limb unloaded mice exhibited evident disruption in intestinal structure and gut microbial homeostasis, along with impairment in their learning and memory capabilities. However, 4-week abdominal LIPUS treatment improved intestinal function in hind limb unloaded mice, characterized by upregulation of tight junction proteins ZO-1 and Occludin expression in the colon, increased diversity and abundance of intestinal microbiota, decreased serum lipopolysaccharide (LPS), and increased short chain fatty acids in colon contents. The hind limb unloaded mice treated with LIPUS exhibited heightened activity levels, improved exploratory tendencies, and significantly enhanced learning and memory faculties, and elevated expression of neuroadaptation-related proteins such as PSD95, GAP43, P-CREB, BDNF, and its receptor TRKB in the hippocampus. Furthermore, the hind limb unloaded mice receiving fecal transplants from the mice whose abdomens were irradiated with LIPUS displayed enhanced cognitive abilities and improved intestinal structure, akin to the outcomes observed in hind limb unloaded mice who received LIPUS abdominal treatment directly. The above results indicate that LIPUS enhances intestinal structure and microbiota, which helps alleviate cognitive impairment caused by weightlessness. LIPUS could be a potential strategy to simultaneously improve gut dysfunction and cognitive decline in astronauts or bedridden patients.
Collapse
Affiliation(s)
- Yumei Zheng
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yanan Yu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Mengyao Chen
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Huiyuan Zhang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Wanzhao Wang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Dean Ta
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, 200433, China.
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
3
|
Wang Y, Li T, Dong Z, Zhang Q, Mi J, Wang Q, Lin G, Ma Q, Jia R, Huang S. Extracellular Vesicles From Lactobacillus fermentum Enhance Intestinal Barrier Integrity and Restore Gut Microbial Homeostasis in Experimental Murine Colitis. J Nutr 2025; 155:1311-1323. [PMID: 40058701 DOI: 10.1016/j.tjnut.2025.03.001] [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: 10/08/2024] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Lactobacillus fermentum has been shown to improve intestinal health and treat colitis; however, its precise efficacy and mechanisms in inflammatory bowel disease remain unclear. OBJECTIVES This study aimed to evaluate whether L fermentum and its metabolites, extracellular vesicles, and other components could modulate intestinal barrier function and gut microbiota to alleviate dextran sulfate sodium (DSS)-induced colitis in mice. METHODS Forty-eight mice were randomly assigned to 6 groups: control, DSS, L fermentum+DSS group (LF+DSS), heat-inactivated L fermentum+DSS group (LHF+DSS), L fermentum supernatant solution+DSS group (LSF+DSS), and L fermentum extracellular vesicles+DSS group (LEV+DSS). After a 1-wk acclimation, mice were gavaged daily for 3 wk. Fresh cultures, including live (LF+DSS), heat-inactivated (LHF+DSS), supernatant (LSF+DSS), and extracellular vesicles (LEV+DSS), were prepared daily. During the final 7 d, the control group received normal water, and the other groups received 3% DSS. Data were collected daily, followed by sample collection from the mice. RESULTS In this study, significant reductions (P < 0.05) in body weight changes, disease activity index, intestinal damage, and histology scores were observed in the treatment groups, especially LEV+DSS and LF+DSS. Additionally, compared with the DSS group, colonic mucus secretion, as well as claudin-1 and occludin expression, increased significantly (P < 0.05) in the LEV+DSS and LF+DSS groups, whereas proinflammatory cytokines IL-1β and TNF-α decreased (P < 0.05) and IL-10 increased (P < 0.05) in the LEV+DSS group. L fermentum and its components significantly regulated gut microbiota α-diversity and β-diversity, affecting overall composition. Linear discriminant analysis effect size analysis revealed an enrichment of beneficial bacteria including Prevotellaceae_UCG-001, Romboutsia, and Ruminococcus species in the LF+DSS group and Akkermansia, Odoribacter, and Marvinbryantia species in the LEV+DSS group. Both L fermentum and its extracellular vesicles significantly downregulated the gene expression of TNF-α and IL-1β, whereas the expression of IL-10 was upregulated, thereby contributing to the alleviation of colitis symptoms. CONCLUSIONS This study reveals that L fermentum alleviates colitis through modulation of the gut microbiota and reinforcement of the intestinal mucosal barrier, with its extracellular vesicles potentially playing a key role in this regulatory process.
Collapse
Affiliation(s)
- Yanwei Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; School of Life Science, Shanxi University, Taiyuan, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Zhuo Dong
- Hubei International Travel Healthcare Center, Hubei, China
| | - Qiyue Zhang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jingqiu Mi
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Qingfeng Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Gang Lin
- Institute of Quality Standards and Testing Technology for Agricultural Products, Chinese Academy of Agricultural Science, Beijing, China
| | - Qiugang Ma
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Ru Jia
- School of Life Science, Shanxi University, Taiyuan, China.
| | - Shimeng Huang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China.
| |
Collapse
|
4
|
Akerele CA, Koralnik LR, Lafont E, Gilman C, Walsh-Messinger J, Malaspina D. Nutrition and brain health: Implications of Mediterranean diet elements for psychiatric disorders. Schizophr Res 2025; 281:30-44. [PMID: 40315757 DOI: 10.1016/j.schres.2025.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 04/03/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
The Mediterranean diet is an anti-inflammatory diet now recognized for prevention and intervention against cardiometabolic disorders, although emerging literature also shows its benefits for mental health. This paper surveys literature pertinent to the Mediterranean diet with respect to schizophrenia, bipolar and unipolar depression and cognition. The National Library of Medicine database of literature was searched for publications relating to our topic through June 2024. Our results highlight the Mediterranean diet's potential role in mediating inflammation, potentially through the gut-brain axis, and its neuroprotective role against cognitive decline. Moreover, individual components of the Mediterranean diet are potentially therapeutic as well as protective, particularly fruits, vegetables, fatty fish, and whole grains. These dietary components reduce systemic inflammation, modulate gut microbiota and influence critical pathways such as moderating oxidative stress. Overall, this survey of recent literature highlights the potential of the Mediterranean diet to promote mental health and reduce the risk or severity of psychiatric and neurodegenerative disorders. The analysis underscores broader implications for the Mediterranean diet in advancing mental health outcomes and emphasizes the need for further studies and increased public education to encourage healthier eating habits and dietary interventions.
Collapse
Affiliation(s)
| | | | - Ezequiel Lafont
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY 10029, USA
| | - Caitlin Gilman
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Dolores Malaspina
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY 10029, USA.
| |
Collapse
|
5
|
Cole LD, Kuhn KA. It Takes a Village: Juvenile Idiopathic Arthritis and the Microbiome. Rheum Dis Clin North Am 2025; 51:233-282. [PMID: 40246440 DOI: 10.1016/j.rdc.2025.01.005] [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] [Indexed: 04/19/2025]
Abstract
Multiple risk factors for juvenile idiopathic arthritis (JIA) influence the microbiome, and various differences in the oral and fecal microbiome have been described to date in JIA. This review summarizes what is known and discusses potential implications for future research on the microbiome in JIA.
Collapse
Affiliation(s)
- Lyndsey D Cole
- Sections of Rheumatology & Infectious Diseases, Department of Pediatrics, University of Colorado Anschutz Medical Campus, 13123 East 16th Avenue, B311, Aurora, CO 80045, USA.
| | - Kristine A Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Mingels S. Paediatric pain and malnutrition in low-income countries: A narrative review. Nutr Health 2025:2601060251336823. [PMID: 40304640 DOI: 10.1177/02601060251336823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Background: Despite its omnipresence, paediatric pain remains poorly understood and documented, especially in low-income countries. Such pain can be a symptom of long-term subclinical conditions such as systemic chronic inflammation (SCI). The latter can be related to malnutrition. Aim: To explore a potential association between paediatric pain and malnutrition in low-income countries. Methods: Narrative review, including a literature search in the PubMed, EMBASE, Web of Science and Scopus databases (update 24 March 2025). The search query comprised controlled terminology and free text words relating to 'Malnutrition', 'Pain', 'SCI' and 'Paediatric'. Results: To comprehend the complex relation between malnutrition and paediatric pain, associations between (1a) malnutrition, and nociceptive brain development, (1b) malnutrition, the gut microbiome and SCI, and (2) SCI and pain were explored. (1a) Early noxious exposure (e.g. malnutrition-related SCI) can cause long-term alterations in pain perception, brain function and structures. The consequences of malnutrition on the nociceptive brain depend on the life-cycle. (1b) Moderate acute malnutrition causes chronic inflammation and exaggerated inflammatory responses. Such responses could indicate hyper-inflammatory phenotypes. (2) Systemic-induced inflammation causes a widespread increase in musculoskeletal pain sensitivity. Conclusion: Malnutrition could contribute to the development of a nociceptive brain and SCI. Malnutrition-related SCI could induce changes in pain perception/thresholds, and predispose to developing chronic pain. If a relation between malnutrition and SCI predisposes children to develop pain, the prevailing biophysical approach needs revision. A multidimensional interdisciplinary approach seems more relevant. Such approach includes social, cognitive, socioeconomic, lifestyle, nutritional (e.g. integrating nutritional and microbiome-targeted interventions) and environmental dimensions.
Collapse
Affiliation(s)
- Sarah Mingels
- Musculoskeletal Research Unit, Department of Rehabilitation Sciences, Faculty of Movement and Rehabilitation Sciences, Leuven University, Leuven, Belgium
| |
Collapse
|
7
|
Xie X, Chen X, Wang X, Wang S, Qi P. Dual regulatory effects of gut microbiota and their metabolites in rheumatoid arthritis: balancing pathogenic and protective mechanisms. Front Immunol 2025; 16:1584023. [PMID: 40370449 PMCID: PMC12075411 DOI: 10.3389/fimmu.2025.1584023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disorder characterized by destructive, symmetric joint inflammation and synovitis, resulting in substantial disability that profoundly compromises patients' quality of life. Its pathogenesis encompasses complex interactions between genetic and environmental factors. Recent advances in bacterial DNA sequencing technologies have uncovered a significant correlation between the human gut microbiota composition and rheumatoid arthritis progression. Growing clinical and experimental evidence establishes the gut-joint axis as a crucial mediator in rheumatoid arthritis pathogenesis. Comprehensive investigation of gut microbial communities and their metabolites' influence on rheumatoid arthritis mechanisms, coupled with the elucidation of microbiome's bidirectional regulatory effects in disease development, not only deepens our understanding of pathological processes but also establishes a theoretical framework for developing novel diagnostic biomarkers and personalized therapeutic interventions to enhance patient outcomes.
Collapse
Affiliation(s)
- Xingwen Xie
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xin Chen
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xuetao Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Sunli Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Peng Qi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
8
|
Chancharoenthana W, Kamolratanakul S, Udompornpitak K, Wannigama DL, Schultz MJ, Leelahavanichkul A. Alcohol-induced gut permeability defect through dysbiosis and enterocytic mitochondrial interference causing pro-inflammatory macrophages in a dose dependent manner. Sci Rep 2025; 15:14710. [PMID: 40289168 PMCID: PMC12034794 DOI: 10.1038/s41598-025-97593-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Although toxicity of alcohol toward the intestines and immunity is mentioned, there might be different effect of alcohol in a low and a high dose and the rodent model development using a simple SHIRPA binary score night be useful. Hence, a low and high dose of alcohol (6.30 and 1.26 g/kg/day) were administered in might for 16 weeks before determination of several parameters. As such, the peak blood alcohol concentration (BAC) of low and high dose of alcohol were approximately at 0.05 and 0.15%, respectively, at 1 h post-administration, which correlated with SHIRPA score at 1.8 ± 0.8 and 7.2 ± 0.6, respectively. After 16 wk of administration, a significant liver injury in high-dose alcohol was indicated by liver enzymes, liver weight, histology score, apoptosis, and hepatic accumulation of triglyceride (TG) and oxidative stress (malondialdehyde; MDA) with reduced anti-oxidant (glutathione). Meanwhile, low-dose alcohol demonstrated only elevated apoptosis with increased TG and MDA in liver tissue. Leaky gut from both dose of alcohol was also demonstrated by FITC-dextran, endotoxemia, serum beta glucan, and reduced occludin. However, bacterial abundance (microbiome analysis) of the feces from small bowel of high-dose alcohol, but not the low dose, was different from the control (increased Alitipes spp. with reduced Lachnospiraceae). In conclusion, both low- and high-dose alcohol induced leaky gut, while only the high-dose caused gut dysbiosis and alcohol damaged mitochondria but enhanced glycolysis in enterocytes and macrophages. Leaky gut might be more sensitive than dysbiosis to determine alcohol-induced intestinal injury.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 16/F Ratchanakarin Building 420/6 Rajvithi Rd., Ratchathewi, Bangkok, 10400, Thailand.
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| | - Supitcha Kamolratanakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 16/F Ratchanakarin Building 420/6 Rajvithi Rd., Ratchathewi, Bangkok, 10400, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Kanyarat Udompornpitak
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, 10500, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10500, Thailand
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases, Faculty of Medicine Yamagata University and Yamagata University Hospital, Yamagata, Japan
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Pathogen Hunter's Research Collaborative Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- Yamagata Prefectural University of Health Sciences, Kamiyanagi, Yamagata, 990-2212, Japan
- Biofilms and Antimicrobial Resistance Consortium of ODA receiving countries, The University of Sheffield, Sheffield, UK
| | - Marcus J Schultz
- Department of Intensive Care and Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, 10500, Thailand.
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10500, Thailand.
- Immunology Unit, Department of Microbiology, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
9
|
Zou T, Tang X, Wang H, Shang X, Liang X, Ma X. Nanocrystalline cellulose-geniposide complex enhances gut-brain axis modulation for depression treatment. Commun Biol 2025; 8:667. [PMID: 40287572 PMCID: PMC12033350 DOI: 10.1038/s42003-025-07934-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Depression, a major global health issue, is closely associated with imbalances in gut microbiota and altered intestinal functions. This study investigates the antidepressant potential of a composite of Geniposide (GP) and Nanocrystalline Cellulose (NCC), focusing on its effects on the gut-brain axis. Utilizing network pharmacology, GP was identified as a key compound targeting the BCL2 gene in depression management. Experimental approaches, including a chronic unpredictable mild stress (CUMS) model in mice, cellular assays, and fecal microbiota transplantation (FMT), were used to evaluate the composite's effectiveness. Results indicate that GP activates the adenosine monophosphate-activated protein kinase (AMPK) pathway by upregulating BCL2, enhancing intestinal barrier integrity, and balancing gut flora. These mechanisms contribute to its positive effects on hippocampal function and depressive-like behaviors in mice, suggesting that the GP-NCC composite could be a promising avenue for developing depression therapies that target gut health.
Collapse
Affiliation(s)
- Tianyu Zou
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China.
| | - Xiang Tang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Haiping Wang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaolong Shang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xiaoyu Liang
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| | - Xuemiao Ma
- Department of Encephalopathy, Shenzhen Luohu District Hospital of Traditional Chinese Medicine (Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine), Shenzhen, 518000, Guangdong, PR China
| |
Collapse
|
10
|
Hafeez E, Du D, Ni H, Zhu K, Hu F, Zhou J, Chen D. Regulation and mechanism of Bletilla striata polysaccharide on delaying aging in Drosophila melanogaster. Int J Biol Macromol 2025; 310:143382. [PMID: 40268010 DOI: 10.1016/j.ijbiomac.2025.143382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/12/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Bletilla striata polysaccharide (BSP) is a natural bioactive compound known for its promising health benefits, including antioxidant, immunomodulatory, and anti-inflammatory effects. However, its potential in combating aging remains largely unexplored. This study aims to investigate the anti-aging effects of BSP in the Drosophila melanogaster model. The results show that BSP supplementation significantly extends the lifespan of flies in a concentration-dependent manner, with the most pronounced effects observed at a concentration of 3 mg/mL. Lifespan extension is associated with enhanced antioxidative capacities, as evidenced by increased SOD and CAT activities, and decreased MDA content. Additionally, BSP ameliorates age-related symptoms, including improved climbing ability and enhanced intestinal barrier function. Furthermore, BSP supplementation enhances resistance to H2O2-induced oxidative and starvation stresses, attenuates the lead (Pb)-induced toxicity, and delays the onset of Alzheimer's phenotypes in flies. RNA-Seq analysis reveals that BSP supplementation leads to the differential expression of 992 genes. KEGG pathway analysis highlights significant changes in metabolic pathways, including galactose metabolism, starch and sucrose metabolism, and carbon metabolism. Key genes such as Mal-A1, Amy-d, Men-b, Pgm-1, Mdh1, and Hex-C are downregulated, while CG32026, CG11291, and Ald2 are upregulated. These findings suggest BSP exhibits significant anti-aging and protective properties, making it a potential therapeutic agent.
Collapse
Affiliation(s)
- Eqra Hafeez
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Dongsheng Du
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Hang Ni
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Kai Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Fan Hu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Jihai Zhou
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Dongsheng Chen
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China.
| |
Collapse
|
11
|
Mohamed DA, Ramadan AA, Mabrok HB, Ibrahim GE, Mohammed SE. Persea americana Peel: A Promising Source of Nutraceutical for the Mitigation of Cardiovascular Risk in Arthritic Rats Through the Gut-Joint Axis. Biomolecules 2025; 15:590. [PMID: 40305360 PMCID: PMC12025046 DOI: 10.3390/biom15040590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease characterized by the inflammation of synovial fluid. The incidence of cardiovascular diseases (CVDs) is increasing in RA patients. This research is the first report to investigate the anti-arthritic effect of avocado peel nutraceutical (APN) and its potential in mitigating the cardiovascular risk associated with RA. The antioxidant activity and phytochemical composition of APN were assessed. The potential interaction of APN's active compounds with protein tyrosine phosphatase non-receptor type 22 (PTPN22) was studied using molecular docking. The impact of APN on the plasma lipid profile, oxidative and inflammatory markers, and the indices of coronary risk and atherogenicity as CVD markers were evaluated. The gene expression of COX-2, IL-6, IL-1β, IL-10, and TNF-α in liver and spleen tissues were measured. The rat gut microbiota profile was investigated using 16S rRNA amplicon sequencing. APN exhibited high antioxidant activity, low atherogenicity and thrombogenicity indices, and a high ratio of hypocholesterolemic to hypercholesterolemic fatty acids indicating its cardioprotective potential. The administration of APN led to a reduction in oxidative stress markers, inflammatory markers, dyslipidemia, and CVD markers. APN administration downregulated the expression of COX-2, IL-6, IL-1β, and TNF-α genes, while the IL-10 gene was significantly upregulated in the liver and spleen. Treatment with APN was favorable in restoring eubiosis in the gut by modulating RA-associated bacterial taxa linked to impaired immune function and cardiometabolic diseases. In molecular docking, β-amyrin and ellagic acid showed the highest binding affinity for PTPN22. APN may represent a promising approach to ameliorating the cardiovascular risk of RA. The present results will be offering a foundation for future in-depth research in nutraceuticals from agriculture by-products. Additionally, they will be supporting the public health policies aimed at preventing and controlling rheumatoid arthritis.
Collapse
Affiliation(s)
- Doha A. Mohamed
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Cairo 12622, Egypt; (A.A.R.); (H.B.M.); (S.E.M.)
| | - Asmaa A. Ramadan
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Cairo 12622, Egypt; (A.A.R.); (H.B.M.); (S.E.M.)
| | - Hoda B. Mabrok
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Cairo 12622, Egypt; (A.A.R.); (H.B.M.); (S.E.M.)
| | - Gamil E. Ibrahim
- Chemistry of Aroma and Flavor Department, Food Industries and Nutrition Institute, National Research Centre, Cairo 12622, Egypt;
| | - Shaimaa E. Mohammed
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Cairo 12622, Egypt; (A.A.R.); (H.B.M.); (S.E.M.)
| |
Collapse
|
12
|
Kurhaluk N, Kamiński P, Bilski R, Kołodziejska R, Woźniak A, Tkaczenko H. Role of Antioxidants in Modulating the Microbiota-Gut-Brain Axis and Their Impact on Neurodegenerative Diseases. Int J Mol Sci 2025; 26:3658. [PMID: 40332186 PMCID: PMC12027284 DOI: 10.3390/ijms26083658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
This narrative review presents the role of antioxidants in regulating the gut microbiota and the impact on the gut-brain axis, with a particular focus on neurodegenerative diseases, such as Alzheimer's (AD) and Parkinson's disease (PD). These diseases are characterised by cognitive decline, motor dysfunction, and neuroinflammation, all of which are significantly exacerbated by oxidative stress. This review elucidates the contribution of oxidative damage to disease progression and explores the potential of antioxidants to mitigate these pathological processes through modulation of the gut microbiota and associated pathways. Based on recent studies retrieved from reputable databases, including PubMed, Web of Science, and Scopus, this article outlines the mechanisms by which antioxidants influence gut health and exert neuroprotective effects. Specifically, it discusses how antioxidants, including polyphenols, vitamins, and flavonoids, contribute to the reduction in reactive oxygen species (ROS) production and neuroinflammation, thereby promoting neuronal survival and minimising oxidative damage in the brain. In addition, the article explores the role of antioxidants in modulating key molecular pathways involved in oxidative stress and neuroinflammation, such as the NF-κB, Nrf2, MAPK, and PI3K/AKT pathways, which regulate ROS generation, inflammatory cytokine expression, and antioxidant responses essential for maintaining cellular homeostasis in both the gut and the central nervous system. In addition, this review explores the complex relationship between gut-derived metabolites, oxidative stress, and neurodegenerative diseases, highlighting how dysbiosis-an imbalance in the gut microbiota-can exacerbate oxidative stress and contribute to neuroinflammation, thereby accelerating the progression of such diseases as AD and PD. The review also examines the role of short-chain fatty acids (SCFAs) produced by beneficial gut bacteria in modulating these pathways to attenuate neuroinflammation and oxidative damage. Furthermore, the article explores the therapeutic potential of microbiota-targeted interventions, including antioxidant delivery by probiotics and prebiotics, as innovative strategies to restore microbial homeostasis and support brain health. By synthesising current knowledge on the interplay between antioxidants, the gut-brain axis, and the molecular mechanisms underlying neurodegeneration, this review highlights the therapeutic promise of antioxidant-based interventions in mitigating oxidative stress and neurodegenerative disease progression. It also highlights the need for further research into antioxidant-rich dietary strategies and microbiota-focused therapies as promising avenues for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22 B, 76-200 Słupsk, Poland;
| | - Piotr Kamiński
- Department of Medical Biology and Biochemistry, Division of Ecology and Environmental Protection, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Skłodowska-Curie St. 9, 85-094 Bydgoszcz, Poland;
- Department of Biotechnology, Institute of Biological Sciences, Faculty of Biological Sciences, University of Zielona Góra, Prof. Z. Szafran St. 1, 65-516 Zielona Góra, Poland
| | - Rafał Bilski
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Karłowicz St. 24, 85-092 Bydgoszcz, Poland; (R.B.); (R.K.); (A.W.)
| | - Halina Tkaczenko
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22 B, 76-200 Słupsk, Poland;
| |
Collapse
|
13
|
Wasim R, Sumaiya, Ahmad A, Anwar A, Salman A. Microbial imbalance in the gut: a new frontier in Rheumatoid arthritis research. Inflammopharmacology 2025:10.1007/s10787-025-01737-7. [PMID: 40220199 DOI: 10.1007/s10787-025-01737-7] [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: 02/26/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025]
Abstract
A chronic autoimmune illness that causes joint destruction and inflammation, rheumatoid arthritis (RA) often results in disability. Genetic, environmental, and immune system variables all have a role in the pathophysiology of RA. The complex community of bacteria that live in the gastrointestinal system, known as the gut microbiota, has been implicated in the onset and progression of RA in recent years, according to mounting data. An imbalance in the gut microbiota's composition, known as dysbiosis, has been noted in RA patients. This imbalance may impact inflammatory pathways and immunological responses, which in turn may contribute to the development and severity of the illness. Research has shown that some bacterial species, including Firmicutes, Bacteroidetes, and Proteobacteria, are either more abundant or less prevalent in RA patients than in healthy people. The gut-immune system axis may be modulated, immunological tolerance may be affected, and pro-inflammatory cytokine production may be enhanced by these microbial changes, all of which may lead to systemic inflammation linked to RA. Moreover, changes in intestinal permeability and a rise in microbial metabolite translocation may make autoimmune reactions worse. Probiotics, antibiotics, and dietary changes have also been investigated as possible treatment approaches to help RA patients regain the balance of their gut microbiota. Still up for debate, however, are the precise ways in which the gut microbiome affects RA. Comprehending the complex connection between gut microbiota and RA may give new perspectives on managing and preventing the condition, as well as future prospects for medicines that target the microbiome.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India.
| | - Sumaiya
- Career Post Graduate Institute of Dental Sciences and Hospital, Lucknow, India
| | - Asad Ahmad
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| | - Aamir Anwar
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| | - Aimen Salman
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| |
Collapse
|
14
|
Charneca S, Hernando A, Almada-Correia I, Polido-Pereira J, Vieira A, Sousa J, Almeida AS, Motta C, Barreto G, Eklund KK, Alonso-Pérez A, Gómez R, Cicci F, Mauro D, Pinho SS, Fonseca JE, Costa-Reis P, Guerreiro CS. TASTY trial: protocol for a study on the triad of nutrition, intestinal microbiota and rheumatoid arthritis. Nutr J 2025; 24:52. [PMID: 40189532 PMCID: PMC11974026 DOI: 10.1186/s12937-025-01089-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 02/06/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND The gut microbiota has been implicated in the onset and progression of Rheumatoid Arthritis (RA). It has been proposed that gut dysbiosis impairs gut barrier function, leading to alterations in mucosal integrity and immunity. This disruption allows bacterial translocation, contributing to the perpetuation of the inflammatory process. Since diet is recognised as a key environmental factor influencing the gut microbiota, nutritional interventions targeting RA activity are currently being explored. This study aims to investigate whether a dietary intervention based on a typical Mediterranean Diet enriched with fermented foods (MedDiet +) can impact the gut microbiota, intestinal permeability, and RA-related outcomes. METHODS One hundred RA patients are being recruited at Unidade Local de Saúde (ULS) Santa Maria in Lisbon, Portugal, and randomly assigned to either the intervention (MedDiet +) or the control group. The 12-week nutritional intervention includes a personalised dietary plan following the MedDiet + pattern, along with educational resources, food basket deliveries, and clinical culinary workshops, all developed and monitored weekly by registered dietitians. The control group receives standardised general healthy diet recommendations at baseline. The intervention's effects will be assessed by evaluating disease activity, functional status, quality of life, intestinal permeability, endotoxemia, inflammatory biomarkers, intestinal and oral microbiota, serum proteomics, and serum glycome profile characterisation. DISCUSSION We anticipate obtaining integrative insights into the interplay between diet, the gut, and RA, while also exploring the underlying mechanisms driving these changes. This study, conducted by a multidisciplinary research team of registered dietitians, rheumatologists, biologists, and immunologists, aims to bridge the current gap between nutrition-related knowledge and RA. TRIAL REGISTRATION Registered in ClinicalTrials.gov (NCT06758817; date of registry: January 6th 2025).
Collapse
Affiliation(s)
- Sofia Charneca
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana Hernando
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Inês Almada-Correia
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Joaquim Polido-Pereira
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Serviço de Reumatologia, ULS Santa Maria, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Adriana Vieira
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Joana Sousa
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Ana Santos Almeida
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Carla Motta
- Departamento de Alimentação e Nutrição, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
| | - Gonçalo Barreto
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, 00029, Finland.
| | - Kari K Eklund
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, 00029, Finland
- Department of Rheumatology, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Ana Alonso-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706, Santiago de Compostela, Spain
| | - Rodolfo Gómez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706, Santiago de Compostela, Spain
| | - Francesco Cicci
- Dipartimento di Medicina di Precisione, Università Della Campania L. Vanvitelli, Naples, Italy
| | - Daniele Mauro
- Dipartimento di Medicina di Precisione, Università Della Campania L. Vanvitelli, Naples, Italy
| | - Salomé S Pinho
- Institute for Research and Innovation in Health (i3s), University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - João Eurico Fonseca
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Serviço de Reumatologia, ULS Santa Maria, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Patrícia Costa-Reis
- Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- GIMM - Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Pediatric Rheumatology Unit, ULS Santa Maria, Lisbon, Portugal
| | - Catarina Sousa Guerreiro
- Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
- Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| |
Collapse
|
15
|
Makaro A, Kasprzak Z, Jaczynska M, Swierczynski M, Salaga M. Role of Cytochromes P450 in Intestinal Barrier Function: Possible Involvement in the Pathogenesis of Leaky Gut Syndrome. Dig Dis Sci 2025; 70:1293-1304. [PMID: 39971825 DOI: 10.1007/s10620-025-08873-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
The intestinal barrier constitutes the largest surface of the human body communicating with the external environment. Alterations affecting elements of intestinal wall may lead to increased intestinal permeability and resulting translocation of bacteria or its components to the bloodstream in the form of the "leaky gut syndrome" (LGS). One of the most common causes of LGS is the disruption of tight junctions (TJ) maintained by tight junction proteins (TJP). LGS and associated alterations in TJP are observed in numerous gastrointestinal (GI) diseases, including inflammatory bowel diseases (IBD) such as Crohn's disease (CD) and ulcerative colitis (UC). Current literature indicates the key role of LGS in many pathological processes, further emphasizing the need for effective pharmacological approaches to treat this syndrome. One of the potential pharmacological targets in LGS treatment are members of the cytochrome P450 (CYP450) superfamily. By affecting intestinal permeability, they may lead to LGS development. It was found that the expression of CYP8B1 synthesizing cholic acid and CYP26 degrading all-trans retinoic acid indirectly influence TJs. CYP2E1 responsible for the metabolism of a wide variety of chemicals, including ethanol, plays a crucial role in the impairment of the intestinal wall. Contrarily, the overexpression of CYP27B1 has a protective effect on the intestinal integrity. CYP1A1, CYP2A6, CYP2J2 and CYP3A were also suggested to influence the GI tract, through their capability to metabolize serotonin, nicotine, endocannabinoids and gemcitabine, respectively. This review summarizes the findings on the role of CYP450 isoforms in intestinal hyperpermeability and their potential involvement in the pathophysiology of LGS.
Collapse
Affiliation(s)
- Adam Makaro
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Zuzanna Kasprzak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Maria Jaczynska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Mikolaj Swierczynski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Maciej Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland.
| |
Collapse
|
16
|
Zwierz M, Suprunowicz M, Mrozek K, Pietruszkiewicz J, Oracz AJ, Konarzewska B, Waszkiewicz N. Vitamin B12 and Autism Spectrum Disorder: A Review of Current Evidence. Nutrients 2025; 17:1220. [PMID: 40218978 PMCID: PMC11990331 DOI: 10.3390/nu17071220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/14/2025] Open
Abstract
Vitamin B12 (cobalamin) plays a crucial role in neurodevelopment, particularly during pregnancy and early childhood. It is essential for DNA synthesis, red blood cell formation, and nervous system function. Maternal B12 levels are particularly important, as they influence fetal brain development. Inadequate maternal intake during pregnancy may lead to altered neurodevelopmental trajectories and increase the risk of ASD. Postnatally, insufficient dietary cobalamin in infants and young children could further contribute to cognitive and behavioral impairments. One potential mechanism linking low B12 levels to ASD involves its role in the gut microbiota balance. Dysbiosis, commonly observed in individuals with ASD, is associated with increased gut permeability, low-grade inflammation, and disruptions in the gut-brain axis, all of which may contribute to ASD symptoms. Additionally, B12 is essential for neurotransmitter metabolism, particularly in the synthesis of serotonin and dopamine, which regulate mood, cognition, and behavior. Cobalamin also plays a key role in neuronal myelination, which ensures efficient signal transmission in the nervous system. Disruptions in these processes could underlie some of the cognitive and behavioral features associated with ASD. Despite growing evidence, the link between B12 and ASD remains inconclusive due to inconsistent findings across studies. Research suggests that B12 levels may serve as a potential biomarker for disease progression and treatment response. However, many studies rely on single-time-point measurements, failing to account for individual variability, genetic predispositions, dietary intake, and environmental factors, all of which can influence B12 levels and ASD risk. Further longitudinal studies are needed to clarify this relationship.
Collapse
Affiliation(s)
- Mateusz Zwierz
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (M.S.); (K.M.); (J.P.); (A.J.O.); (B.K.); (N.W.)
| | | | | | | | | | | | | |
Collapse
|
17
|
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
|
18
|
Beyoğlu D, Idle JR. The Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2025; 26:2882. [PMID: 40243472 PMCID: PMC11988851 DOI: 10.3390/ijms26072882] [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: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a condition wherein excessive fat accumulates in the liver, leading to inflammation and potential liver damage. In this narrative review, we evaluate the tissue microbiota, how they arise and their constituent microbes, and the role of the intestinal and hepatic microbiota in MASLD. The history of bacteriophages (phages) and their occurrence in the microbiota, their part in the potential causation of MASLD, and conversely, "phage therapy" for antibiotic resistance, obesity, and MASLD, are all described. The microbiota metabolism of bile acids and dietary tryptophan and histidine is defined, together with the impacts of their individual metabolites on MASLD pathogenesis. Both periodontitis and intestinal microbiota dysbiosis may cause MASLD, and how individual microorganisms and their metabolites are involved in these processes is discussed. Novel treatment opportunities for MASLD involving the microbiota exist and include fecal microbiota transplantation, probiotics, prebiotics, synbiotics, tryptophan dietary supplements, intermittent fasting, and phages or their holins and endolysins. Although FDA is yet to approve phage therapy in clinical use, there are multiple FDA-approved clinical trials, and this may represent a new horizon for the future treatment of MASLD.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
| | - Jeffrey R. Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
- Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
19
|
Lv J, Jin S, Zhou Y, Fu C, Shen Y, Liu B, Li J, Li M, Zhang Y, Feng N. Gut microbiota-derived metabolite phenylacetylglutamine inhibits the progression of prostate cancer by suppressing the Wnt/β-catenin signaling pathway. Front Pharmacol 2025; 16:1528058. [PMID: 40135235 PMCID: PMC11932994 DOI: 10.3389/fphar.2025.1528058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/13/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Prostate cancer is one of the most common malignant tumors among men worldwide, and current treatments still face many challenges. Therefore, researchers are continuously seeking new therapeutic methods to improve treatment efficacy and reduce side effects. Phenylacetylglutamine (PAGln), a common metabolite of the gut microbiota, has been reported to have anti-inflammatory and anti-tumor activities. METHODS We assessed the impact of PAGln on prostate cancer using in vitro and in vivo models. Cell proliferation, migration, and invasion capabilities were evaluated through CCK8, EdU incorporation, and colony formation assays, as well as wound healing and Transwell assays. The in vivo anti-cancer effects of PAGln were evaluated using a BALB/c nude mouse xenograft model of prostate cancer and a lung metastatic tumor model established via tail vein injection. Molecular mechanisms were investigated through qRT-PCR and Western blot analysis. RESULTS PAGln inhibited the proliferation, migration, and invasion of prostate cancer (PCa) cells in vitro and suppressed the growth of prostate cancer in vivo. PAGln notably increased the mRNA levels of CCNG2 in PCa cells. Importantly, the knockdown of CCNG2 weakened the effects of PAGln on PCa cells. Mechanistic studies revealed that PAGln could promote the phosphorylation of β-catenin by upregulating CCNG2, thereby inhibiting the Wnt/β-catenin signaling pathway. CONCLUSION In summary, PAGln can effectively inhibit the proliferation, migration, and invasion of PCa by upregulating CCNG2 and suppressing the Wnt/β-catenin signaling pathway. These findings suggest that PAGln may serve as a promising therapeutic agent for prostate cancer.
Collapse
Affiliation(s)
- Jing Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Shengkai Jin
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Yuhua Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Chaowei Fu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Yang Shen
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
- Jiangnan Medical Center, Nanjing Medical University, Nanjing, China
| | - Bo Liu
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
- Medical School of Nantong University, Nantong, China
| | - Jufa Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Menglu Li
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Yuwei Zhang
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
- Medical School of Nantong University, Nantong, China
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
- Jiangnan Medical Center, Nanjing Medical University, Nanjing, China
- Medical School of Nantong University, Nantong, China
| |
Collapse
|
20
|
Liu XJ, Ye-er-tai YLY, Jia YB, Wu CH, Wang XX, Yang KM, Yao X, Ling JH. Runchangningshen paste activates NLRP6 inflammasome-mediated autophagy to stimulate colonic mucin-2 secretion and modulates mucosal microbiota in functional constipation. World J Gastroenterol 2025; 31:102256. [PMID: 40061589 PMCID: PMC11886036 DOI: 10.3748/wjg.v31.i9.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/06/2024] [Accepted: 01/02/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Runchangningshen paste (RCNSP) is a paste made of four medicinal and edible homologous Chinese medicine mixed with honey. It is known for its ability to nourish yin and blood as well as to loosen the bowel to relieve constipation. The pathophysiology of functional constipation (FC) is associated with a reduction in mucin-2 (MUC2) secretion and microbial dysbiosis. AIM To investigate the underlying mechanism of RCNSP against FC through MUC2 and the gut mucosal microbiota. METHODS Ultra-performance liquid chromatography tandem mass spectrometry characterized RCNSP composition to elucidate the material basis of action. FC model was induced via loperamide gavage (16 mg/kg) twice daily for 7 days. Applying defecation function and gastrointestinal motility to assess constipation severity. Hematoxylin and eosin and Alcian blue-periodic acid-schiff staining analyzed colonic mucosal morphology. Transmission electron microscope was used to observe the ultrastructure of goblet cells (GCs). Immunofluorescence colocalization, quantitative PCR, and western blot assessed the impact of RCNSP on gene and protein expression within the NLRP6/autophagy pathway. 16S rDNA was employed to sequence the gut mucosal microbiota. RESULTS RCNSP contained 12 components with potential laxative effects. It enhanced defecation function, accelerated gastrointestinal motility, and maintained colonic mucosal integrity. RCNSP treatment significantly increased GC abundance and MUC2 production while preserving GC ultrastructure. At the molecular level, RCNSP enhanced the colocalized expression of key regulatory proteins and modulated mRNA and protein expressions in the NLRP6/autophagy pathway. Through 16S rDNA sequencing analysis, RCNSP significantly altered the mucosal microbiota composition. Specifically, it increased beneficial bacterial strains while reducing harmful ones. Simultaneously, RCNSP reduced butyrate-producing bacteria like Proteobacteria, Enterobacteriaceae, Blautia, and Eubacterium and decreased hydrogen sulfide-producing species, such as Prevotellaceae. It also reduced bile acid-inhibiting species, such as g_Eubacter_coprostanoligenes_group and Erysipelotrichaceae while increasing bile acid-producing species, such as Colidextribacter. CONCLUSION Our findings suggested that RCNSP ameliorated constipation through a dual mechanism: It stimulated colonic MUC2 secretion by activating NLRP6 inflammasome-mediated autophagy and modulated the composition of the mucosal microbiota.
Collapse
Affiliation(s)
- Xue-Jiao Liu
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Ye-li-ya Ye-er-tai
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Yue-Bo Jia
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Chen-Heng Wu
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xiang-Xiang Wang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ke-Ming Yang
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong Province, China
| | - Xuan Yao
- Guangdong Shaxi Pharmaceutical Co., Ltd, Zhongshan 528471, Guangdong Province, China
| | - Jiang-Hong Ling
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| |
Collapse
|
21
|
Choe BH. Practical concepts and strategies for early diagnosis and management of eosinophilic gastrointestinal disorders in East-Asian children. Clin Exp Pediatr 2025; 68:185-198. [PMID: 39533717 PMCID: PMC11884954 DOI: 10.3345/cep.2024.01165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/06/2024] [Accepted: 10/06/2024] [Indexed: 11/16/2024] Open
Abstract
Eosinophilic gastrointestinal disorders (EGIDs) are emerging as significant concerns in the Korean pediatric population and transitioning from rare to more commonly diagnosed conditions. This review discusses the increasing prevalence of EGID among children and adolescents and highlights the complexities involved in its diagnosis and management. This review begins with a thorough examination of the diverse clinical presentations of EGIDs in Korean children, with a special focus on common gastrointestinal symptoms such as abdominal pain, diarrhea, and bloody stool. Additionally, we explored extraintestinal manifestations, including growth failure, malnutrition, and associated allergic comorbidities, highlighting their importance in the clinical landscape of EGIDs. Because of its subtle and overlapping symptoms with those of other gastrointestinal disorders, EGID is frequently underdiagnosed. Addressing this challenge requires maintaining a high index of suspicion and employing a comprehensive diagnostic approach to differentiating EGID from functional gastrointestinal disorders and other inflammatory or systemic diseases such as inflammatory bowel disease. The optimal management of EGID requires a collaborative multidisciplinary strategy that includes dietary management, regular monitoring, and tailored medical interventions. This review emphasizes the importance of proactive patient and caregiver education and regular follow-ups to improve long-term outcomes in affected children. Enhanced awareness among healthcare providers and better educational resources for families are critical for the early identification and effective management of EGID among pediatric patients.
Collapse
Affiliation(s)
- Byung-Ho Choe
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, Korea
| |
Collapse
|
22
|
Ma G, Chen Z, Xie Z, Liu J, Xiao X. Mechanisms underlying changes in intestinal permeability during pregnancy and their implications for maternal and infant health. J Reprod Immunol 2025; 168:104423. [PMID: 39793281 DOI: 10.1016/j.jri.2025.104423] [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: 08/31/2024] [Revised: 12/01/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Proper regulation of intestinal permeability is essential for maintaining the integrity of the intestinal mucosal barrier. An abnormal increase in permeability can significantly contribute to the onset and progression of various diseases, including autoimmune disorders, metabolic conditions, allergies, and inflammatory bowel diseases. The potential connection between intestinal permeability and maternal health during pregnancy is increasingly recognized, yet a comprehensive review remains lacking. Pregnancy triggers a series of physiological structural adaptations and significant hormonal fluctuations that collectively contribute to an increase in intestinal permeability. Although an increase in intestinal permeability is typically a normal physiological response during pregnancy, an abnormal rise is associated with immune dysregulation, metabolic disorders, and various pregnancy-related complications, such as recurrent pregnancy loss, gestational diabetes mellitus, overweight and obesity during pregnancy, intrahepatic cholestasis of pregnancy, and preeclampsia. This paper discusses the components of the intestinal mucosal barrier, the concept of intestinal permeability and its measurement methods, and the mechanisms and physiological significance of increased intestinal permeability during pregnancy. It thoroughly explores the association between abnormal intestinal permeability during pregnancy and maternal diseases, aiming to provide evidence for the pathophysiology of disease development in pregnant women. Additionally, the paper examines intervention methods, such as gut microbiota modulation and nutritional interventions, to regulate intestinal permeability during pregnancy, improve immune and metabolic states, and offer feasible strategies for the prevention and adjuvant treatment of clinical pregnancy complications.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - JinXiang Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
| |
Collapse
|
23
|
Džidić Krivić A, Begagić E, Hadžić S, Bećirović A, Bećirović E, Hibić H, Tandir Lihić L, Kadić Vukas S, Bečulić H, Kasapović T, Pojskić M. Unveiling the Important Role of Gut Microbiota and Diet in Multiple Sclerosis. Brain Sci 2025; 15:253. [PMID: 40149775 PMCID: PMC11939953 DOI: 10.3390/brainsci15030253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS), characterized by neurodegeneration, axonal damage, demyelination, and inflammation. Recently, gut dysbiosis has been linked to MS and other autoimmune conditions. Namely, gut microbiota has a vital role in regulating immune function by influencing immune cell development, cytokine production, and intestinal barrier integrity. While balanced microbiota fosters immune tolerance, dysbiosis disrupts immune regulation, damages intestinal permeability, and heightens the risk of autoimmune diseases. The critical factor in shaping the gut microbiota and modulating immune response is diet. Research shows that high-fat diets rich in saturated fats are associated with disease progression. Conversely, diets rich in fruits, yogurt, and legumes may lower the risk of MS onset and progression. Specific dietary interventions, such as the Mediterranean diet (MD) and ketogenic diet, have shown potential to reduce inflammation, support neuroprotection, and promote CNS repair. Probiotics, by restoring microbial balance, may also help mitigate immune dysfunction noted in MS. Personalized dietary strategies targeting the gut microbiota hold promise for managing MS by modulating immune responses and slowing disease progression. Optimizing nutrient intake and adopting anti-inflammatory diets could improve disease control and quality of life. Understanding gut-immune interactions is essential for developing tailored nutritional therapies for MS patients.
Collapse
Affiliation(s)
- Amina Džidić Krivić
- Department of Neurology, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (A.D.K.); (L.T.L.)
- Department of Physiology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
| | - Emir Begagić
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (E.B.)
- Department of Doctoral Studies, School of Medicine, University of Tuzla, 75000 Tuzla, Bosnia and Herzegovina
| | - Semir Hadžić
- Internal Medicine Clinic, University Clinical Center of Tuzla, Ulica prof. dr. Ibre Pašića, 75000 Tuzla, Bosnia and Herzegovina (E.B.)
- Department of Physiology, School of Medicine, University of Tuzla, Univerzitetska 1, 75000 Tuzla, Bosnia and Herzegovina
| | - Amir Bećirović
- Internal Medicine Clinic, University Clinical Center of Tuzla, Ulica prof. dr. Ibre Pašića, 75000 Tuzla, Bosnia and Herzegovina (E.B.)
| | - Emir Bećirović
- Internal Medicine Clinic, University Clinical Center of Tuzla, Ulica prof. dr. Ibre Pašića, 75000 Tuzla, Bosnia and Herzegovina (E.B.)
| | - Harisa Hibić
- Department of Maxillofacial Surgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina
| | - Lejla Tandir Lihić
- Department of Neurology, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (A.D.K.); (L.T.L.)
- Department of Neurology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
| | - Samra Kadić Vukas
- Department of Neurology, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (A.D.K.); (L.T.L.)
- Department of Neurology, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
| | - Hakija Bečulić
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina; (E.B.)
- Department of Anatomy, School of Medicine, University of Zenica, Travnička 1, 72000 Zenica, Bosnia and Herzegovina
| | - Tarik Kasapović
- Internal Medicine Clinic, University Clinical Center of Tuzla, Ulica prof. dr. Ibre Pašića, 75000 Tuzla, Bosnia and Herzegovina (E.B.)
| | - Mirza Pojskić
- Department of Neurosurgery, University Hospital Marburg, Baldingerstr., 35033 Marburg, Germany
| |
Collapse
|
24
|
Parodi E, Novi M, Bottino P, La Porta E, Merlotti G, Castello LM, Gotta F, Rocchetti A, Quaglia M. The Complex Role of Gut Microbiota in Systemic Lupus Erythematosus and Lupus Nephritis: From Pathogenetic Factor to Therapeutic Target. Microorganisms 2025; 13:445. [PMID: 40005809 PMCID: PMC11858628 DOI: 10.3390/microorganisms13020445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
The role of gut microbiota (GM) and intestinal dysbiosis in triggering the onset and/or modulating the severity and progression of lupus nephritis (LN) has been the object of intense research over the last few years. Some alterations at the phyla level, such as the abundance of Proteobacteria and reduction in Firmicutes/Bacteroidetes (F/B) ratio and in α-diversity have been consistently reported in systemic lupus erythematosus (SLE), whereas a more specific role has been ascribed to some species (Bacteroides thetaiotaomicron and Ruminococcus gnavus) in LN. Underlying mechanisms include microbial translocation through a "leaky gut" and subsequent molecular mimicry, immune dysregulation (alteration of IFNγ levels and of balance between Treg and Th17 subsets), and epigenetic interactions. Levels of bacterial metabolites, such as butyrate and other short-chain fatty acids (SCFAs), appear to play a key role in modulating LN. Beyond bacterial components of GM, virome and mycobiome are also increasingly recognized as important players in the modulation of an immune response. On the other hand, microbiota-based therapy appears promising and includes diet, prebiotics, probiotics, symbiotics, and fecal microbiota transplantation (FMT). The modulation of microbiota could correct critical alterations, such as F/B ratio and Treg/Th17 imbalance, and blunt production of autoantibodies and renal damage. Despite current limits, GM is emerging as a powerful environmental factor that could be harnessed to interfere with key mechanisms leading to SLE, preventing flares and organ damage, including LN. The aim of this review is to provide a state-of-the-art analysis of the role of GM in triggering and modulating SLE and LN on the one hand, while exploring possible therapeutic manipulation of GM to control the disease on the other hand.
Collapse
Affiliation(s)
- Emanuele Parodi
- Nephrology and Dialysis Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy;
| | - Marialuisa Novi
- Gastroenterology Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy;
| | - Paolo Bottino
- Microbiology Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy; (F.G.); (A.R.)
| | - Edoardo La Porta
- Nephrology and Dialysis Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Guido Merlotti
- Department of Primary Care, Azienda Socio Sanitaria Territoriale (ASST) of Pavia, 27100 Pavia, Italy;
| | - Luigi Mario Castello
- Internal Medicine Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy;
- Department of Translational Medicine, Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| | - Franca Gotta
- Microbiology Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy; (F.G.); (A.R.)
| | - Andrea Rocchetti
- Microbiology Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy; (F.G.); (A.R.)
| | - Marco Quaglia
- Nephrology and Dialysis Unit, SS. Antonio e Biagio e Cesare Arrigo University Hospital, 15121 Alessandria, Italy;
- Department of Translational Medicine, Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| |
Collapse
|
25
|
Zhao H, Wang Y, Ren J. Helicobacter pylori and rheumatoid arthritis: Investigation of relation from traditional Chinese medicine. Microb Pathog 2025; 199:107239. [PMID: 39708982 DOI: 10.1016/j.micpath.2024.107239] [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: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune condition that predominantly affects synovial joints, manifesting with joint swelling, pain, and stiffness. In advanced stages, unchecked inflammation can inflict damage on bone and cartilage, resulting in disabilities and deformities of the joints. Additionally, systemic and extra-articular complications may arise due to the consequences of uncontrolled inflammation. Helicobacter pylori (H. pylori) is one of the most prevalent chronic bacterial infections in humans. This microorganism is a spiral-shaped, flagellated, microaerophilic gram-negative bacterium. Prolonged exposure leads to the activation of the immune system, with infected gastric mucosa epithelial cells continuously producing cytokines. This production, in turn, triggers the generation of antibodies as well as T Helper 1 and T Helper 2 effector T cells. The persistent antigenic stimulation resulting from H. pylori infection could lead to the progression of autoimmune diseases. Numerous clinical and pharmacological trials have illustrated the efficacy of traditional Chinese medicine against H. pylori. This review aims to delve into the connection between H. pylori and rheumatoid arthritis so as understand the pathogenesis. The concluding section of this review explores the interplay of Chinese medicine and Helicobacter pylori concerning rheumatoid arthritis.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Rheumatism and Immunology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), No.4, Renmin Road, Shibei District, Qingdao, 266033, China
| | - Yige Wang
- Shandong University of Traditional Chinese Medicine, No.16369, Jingshi Road, Lixia District, Jinan, 250013, China
| | - Jiahui Ren
- Department of Rheumatism and Immunology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), No.4, Renmin Road, Shibei District, Qingdao, 266033, China
| |
Collapse
|
26
|
WU J, NING Y, TAN L, MA F, LIN Y, ZHUO Y. Difference of the gut microbiota of premature ovarian insufficiency in two traditional Chinese syndromes. J TRADIT CHIN MED 2025; 45:132-139. [PMID: 39957167 PMCID: PMC11764924 DOI: 10.19852/j.cnki.jtcm.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 04/25/2024] [Indexed: 02/18/2025]
Abstract
PURPOSE To investigate the differences in gut microbial characteristics between two traditional Chinese syndromes of premature ovarian insufficiency (POI). METHODS Forty women with POI were recruited from the Department of Traditional Chinese Medicine at Shenzhen Maternity and Child Healthcare Hospital between June and December 2020. Women with POI were divided into the kidney deficiency and blood stasis syndrome (SDBS) and Qi and blood deficiency syndrome (QBDS) groups. Gut microbial community profiles were analyzed by 16S rRNA gene sequencing using an Illumina MiSeq system. A retrospective study comparing hormone levels and gut microbiota information was performed between the SDBS and QBDS groups. RESULTS Compared with the QBDS group, the serum levels of estradiol (E2) and anti-Müllerian hormone (AMH) were significantly decreased in the SDBS group. The quantities of Adlercreutzia, Eggerthella, Klebsiella, and Paraprevotella significantly increased in the SDBS group, whereas Lactobacillus decreased significantly. Moreover, alterations in the microbiome in the SDBS and QBDS groups were closely related to the levels of E2 and AMH. The area under the receiver operating characteristic curve for the classification of the two syndromes by the gut microbiome was 0.71. CONCLUSIONS There were significant differences in the dominant microbiota between the SDBS and QBDS groups, and the change in Proteobacteria in the QBDS group was more significant. The characteristics of gut microbiota help us differentiate between the SDBS and QBDS groups, which may provide a basis for the objectification of TCM syndrome types.
Collapse
Affiliation(s)
- Jiaman WU
- 1 Department of Traditional Chinese Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
| | - Yan NING
- 1 Department of Traditional Chinese Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
| | - Liya TAN
- 2 Guangzhou University of Traditional Chinese Medicine, Guangzhou 510405, China
| | - Fei MA
- 1 Department of Traditional Chinese Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
| | - Yanting LIN
- 1 Department of Traditional Chinese Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
| | - Yuanyuan ZHUO
- 3 Department of Acupuncture and Moxibustion, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China
| |
Collapse
|
27
|
Ribeiro MGC, Kravchychyn ACP, Bressan J, Hermsdorff HHM. Adiposity and inflammation markers explain mostly part of the plasma zonulin variation in Brazilian adults with overweight/obesity: A cross-sectional analysis from Brazilian nuts study. Clin Nutr 2025; 45:22-30. [PMID: 39731881 DOI: 10.1016/j.clnu.2024.12.017] [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: 07/27/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 12/30/2024]
Abstract
OBJECTIVE This study evaluated intestinal permeability according to plasma zonulin and its association with adiposity, inflammation, cardiometabolic risk, liver function, and intestinal health markers in adults with overweight/obesity. METHODOLOGY This study is a cross-sectional analysis using baseline data from the Brazilian Nut Study, which involved 123 participants (93 women, age 33.2 ± 8.58 years, BMI 33.9 ± 4.30kg/m2). Subjects were divided into quartiles according to plasma zonulin, assessed by Elisa. Cytokines were assessed by flow cytometry; anthropometric measurements were collected by standard procedure and body composition was assessed by DXA. SCFA analysis was performed by high-performance liquid chromatography, and fecal pH, by a pH meter. Linear regression models were performed (α<5 %). RESULTS Participants included in the last quartile of plasma zonulin had higher values of body fat (%), pro-inflammatory cytokines (CRP, IL-1). According to the multivariate regression model, each one-unit increased in body fat, CRP, IL-12p70, IL-6 and IL-8 resulted correspondingly in an increment of 0.42, 0.14, 0.192, 0.250 and 0.312 ng/ml in plasma zonulin, respectively. Conversely, a one-unit decreased in IL-10 led to an increase of 0.40 ng/ml in plasma zonulin. CONCLUSION Intestinal permeability assessed by plasma zonulin is associated with adiposity, subclinical inflammation and reduced serum HDL levels adults with overweight/obesity, while adiposity and inflammation markers are independent factors for plasma zonulin variation.
Collapse
Affiliation(s)
- Madalena Geralda Cupertino Ribeiro
- Laboratory of Clinical Analysis and Genomics, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil; Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil.
| | - Ana Claudia Pelissari Kravchychyn
- Laboratory of Clinical Analysis and Genomics, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil; Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Josefina Bressan
- Laboratory of Clinical Analysis and Genomics, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil; Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Helen Hermana Miranda Hermsdorff
- Laboratory of Clinical Analysis and Genomics, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil; Laboratory of Energy Metabolism and Body Composition, Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| |
Collapse
|
28
|
Yang Y, Hong Q, Zhang X, Liu Z. Bifidobacterium animalis BD400 protects from collagen-induced arthritis through histidine metabolism. Front Immunol 2025; 16:1518181. [PMID: 39911381 PMCID: PMC11794514 DOI: 10.3389/fimmu.2025.1518181] [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: 10/28/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
Background Rheumatoid arthritis (RA) is a common chronic and systemic autoimmune disease. Numerous clinical studies have indicated a correlation between alterations in gut microbiota and the onset and progression of RA. This research aims to restore intestinal microbiota to a healthy state through the oral administration of Bifidobacterium in the early stages with the goal of delaying the onset and progression of RA. Methods Collagen-induced arthritis (CIA) rat model was constructed to assess the development of RA using arthritis clinical scores, paw thickness, pathological analysis of knee joint. The immune response was evaluated by determinating specific antibodies and cytokines in serum and synovial fluid. The expression of intestinal barrier protein was analyzed by qPCR to evaluate the intestinal barrier function. Alterations in gut microbiota and metabolites were assessed by 16S rDNA and non-targeted metabolomics. Results The findings reveal that administering Bifidobacterium animalis BD400 orally led to a significant reduction in arthritis clinical scores and paw swelling thickness in CIA rats. Additionally, there was a decrease in osteo-facial fusion and calcified cartilage thickening in the knee joint. Furthermore, the oral administration of B. animalis BD400 resulted in the down-regulation of inflammatory factors TNF-α and collagenase MMP-13 in the knee joint. Levels of specific antibodies (anti-CII IgG, anti-CII IgG1, and anti-CII IgG2a) and cytokine IL-17A in serum, as well as cytokines (TNF-α and IL-1β) in the synovial fluid of B. animalis BD400-treated CIA rats, were significantly reduced (p < 0.05). The gene expression levels of intestinal barrier proteins (occludin-1, MUC-2, and ZO-1) showed a significant increase (p < 0.05) in B. animalis BD400-treated CIA rats. The oral administration of B. animalis BD400 altered the composition of intestinal microorganisms in CIA rats at the phylum and genus levels, particularly affecting the genus HT002. B. animalis BD400 alleviates RA by down-regulating 1-methyl-L-histidine and urocanate in the histidine metabolism, laying a foundation for the RA prevention. Conclusion By affecting genus HT002 and histidine metabolism in the gut microbiota of CIA rats, B. animalis BD400 restored intestinal permeability, inhibited systemic inflammatory response, and ultimately slowed down the development of RA.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/immunology
- Arthritis, Experimental/prevention & control
- Arthritis, Experimental/microbiology
- Arthritis, Experimental/pathology
- Rats
- Gastrointestinal Microbiome
- Probiotics/administration & dosage
- Bifidobacterium animalis/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/prevention & control
- Arthritis, Rheumatoid/immunology
- Cytokines/metabolism
- Male
- Disease Models, Animal
- Administration, Oral
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Shanghai, China
- Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Qing Hong
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Shanghai, China
- Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Xuehong Zhang
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Shanghai, China
- Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China
| |
Collapse
|
29
|
Gawlik-Kotelnicka O, Czarnecka-Chrebelska K, Margulska A, Pikus E, Wasiak J, Skowrońska A, Brzeziańska-Lasota E, Strzelecki D. Associations between intestinal fatty-acid binding protein and clinical and metabolic characteristics of depression. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111170. [PMID: 39393435 DOI: 10.1016/j.pnpbp.2024.111170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
INTRODUCTION The topic of increased intestinal permeability is associated with disruption of the intestinal barrier, leading to the "leaky gut" syndrome. Depressive disorders often coexist with abdominal obesity, metabolic syndrome, or its components and complications. Intestinal permeability has been proven to relate to all of the above. METHODS In this cross-sectional study, we aimed to assess the "leaky gut" blood biomarker - intestinal fatty acid-binding protein (I-FABP) - in 114 adult patients diagnosed with depressive disorders depending on abdominal obesity comorbidity, depression, anxiety, and stress level, or antidepressant use. The corrected p-value was set at 0.02. We analyzed patients' mental state, diet, anthropometric parameters, metabolic laboratory markers and I-FABP. RESULTS There was no difference in circulating I-FABP levels between obese and non-obese patients with depressive disorders (p = 0.648). Similarly, I-FABP levels were not different in patients with different emotional symptoms severity (p = 0.829 for self-assessed depression, p = 0.164 for anxiety, and p = 0.543 for stress). But, I-FABP levels differed significantly between patients treated and not treated with antidepressants (p = 0.011). In general linear model analysis treatment with antidepressants, anxiety severity level, their interaction, along with smoking status, drinks intake, and using dietary supplements were shown to significantly explain I-FABP variance (p < 0.001, R2adj = 0.261). CONCLUSIONS Comorbid obesity did not increase intestinal permeability circulating marker, I-FABP, in the population of patients with depressive disorders. Treatment with antidepressants may be connected to higher I-FABP levels. Using dietary supplements, drinks intake, smoking status, or anxiety level may serve as explanatory factors.
Collapse
Affiliation(s)
- Oliwia Gawlik-Kotelnicka
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechosłowacka 8/10, 92-216 Lodz, Poland.
| | | | - Aleksandra Margulska
- Department of Child and Adolescent Psychiatry, Medical University of Lodz, Czechosłowacka 8/10, 92-216 Lodz, Poland.
| | - Ewa Pikus
- Department of Biomedicine and Genetics, Medical University of Lodz, 92-213 Lodz, Poland.
| | - Jakub Wasiak
- Faculty of Medicine, Medical University of Lodz, Kościuszki 4, 90-419 Lodz, Poland.
| | - Anna Skowrońska
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechosłowacka 8/10, 92-216 Lodz, Poland
| | - Ewa Brzeziańska-Lasota
- Department of Biomedicine and Genetics, Medical University of Lodz, 92-213 Lodz, Poland.
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechosłowacka 8/10, 92-216 Lodz, Poland.
| |
Collapse
|
30
|
Comi L, Giglione C, Klinaku FT, Pialorsi F, Tollemeto V, Zurlo M, Seneci A, Magni P. Valorizing Agro‐Food Waste for Nutraceutical Development: Sustainable Approaches for Managing Metabolic Dysfunction‐Associated Steatotic Liver Disease and Related Co‐Morbidities. FOOD FRONTIERS 2024. [DOI: 10.1002/fft2.535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
ABSTRACTThis comprehensive investigation delves into the interconnectedness of different features of cardiometabolic syndrome, such as metabolic dysfunction‐associated steatotic liver disease (MASLD), atherosclerotic cardiovascular disease (ASCVD), and gut dysbiosis, highlighting the crucial role of nutraceuticals in their management and prevention. Given the significant overlap in the pathophysiology of these conditions, the treatment with nutraceuticals, especially those derived from agro‐food waste, offers a promising, sustainable, and innovative approach to healthcare. The 2030 Agenda for Sustainable Development and the One Health concept are key frameworks for selecting the most interesting supply chain for the production of nutraceuticals from agro‐food waste, ensuring environmental sustainability, and innovative agricultural practices. In this review, the therapeutic potential of kiwifruit and apples has been explored, detailing how their bioactive compounds, like polyphenols, fiber, pectin, kaempferol, phloretin, and phlorizin, may contribute to the management of MASLD, ASCVD, and gut dysbiosis. Various extraction methods for active ingredients, including chemical, water, and enzyme extractions, are analyzed for their respective benefits and drawbacks. By integrating scientific research, sustainable agricultural practices, and innovative extraction methods, we can develop effective strategies to combat these pervasive health issues. This holistic approach not only enhances individual health outcomes but also supports broader environmental and societal goals, promoting a healthier future for all.
Collapse
Affiliation(s)
- Laura Comi
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Claudia Giglione
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Fationa Tolaj Klinaku
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | | | | | | | | | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
- IRCCS MultiMedica, Sesto San Giovanni Milan Italy
| |
Collapse
|
31
|
Li X, Li C, Wu P, Zhang L, Zhou P, Ma X. Recent status and trends of innate immunity and the gut-kidney aixs in IgAN: A systematic review and bibliometric analysis. Int Immunopharmacol 2024; 143:113335. [PMID: 39423662 DOI: 10.1016/j.intimp.2024.113335] [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: 05/26/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND There is a significant global demand for precise diagnosis and effective treatment of IgA nephropathy (IgAN), with innate immunity, particularly the complement system, exerting a profound influence on its pathogenesis. Additionally, the gut-kidney axis pathway is vital in the emergence and development of IgAN. METHODS We conducted a comprehensive search in the Web of Science database, spanning from January 1, 2000 to December 18, 2023. The gathered literature underwent a visual examination through CiteSpace, VOSviewer, and Scimago Graphica to delve into authors, nations, organizations, key terms, and other pertinent elements. RESULT Between 2000 and 2023, a total of 720 publications were identified, out of which 436 publications underwent screening for highly relevant literature analysis. The average annual number of articles focusing on IgAN, innate immunity, and the gut-kidney axis is approximately 31, with an upward trend observed. In terms of research impact encompassing publication count and authorship, the United States emerged as the leading contributor. Prominent keywords included "complement", "activation", "microbe", "gut-kidney axis", "C4d deposition", "alternative pathway" and "B cells" along with other prospective hot topics. CONCLUSION The correlation between IgAN and innate immunity is a focal point in current scientific research. Recent literature underscores the significance of the gut-kidney axis, where intestinal microorganisms and metabolites may influence IgAN. The complement system, a key component of innate immunity, also has a crucial function.Advancements in prevention, diagnosis, and treatment hinge on unraveling this intricate relationship.
Collapse
Affiliation(s)
- Xun Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Chengni Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Peiwen Wu
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Lifang Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Ping Zhou
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China.
| | - Xin Ma
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China.
| |
Collapse
|
32
|
Stanco D, Lipsa D, Bogni A, Bremer-Hoffmann S, Clerbaux LA. An Adverse Outcome Pathway for food nanomaterial-induced intestinal barrier disruption. FRONTIERS IN TOXICOLOGY 2024; 6:1474397. [PMID: 39776762 PMCID: PMC11703861 DOI: 10.3389/ftox.2024.1474397] [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: 08/01/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction The ingestion of nanomaterials (NMs) may impair the intestinal barrier, but the underlying mechanisms remain evasive, and evidence has not been systematically gathered or produced. A mechanistic-based approach would be instrumental in assessing whether relevant NMs disrupt the intestinal barrier, thereby supporting the NM risk assessment in the food sector. Methods In this study, we developed an adverse outcome pathway (AOP) based on biological plausibility and by leveraging information from an existing NM-relevant AOP that leads to hepatic outcomes. We then extracted the current evidence from the literature for a targeted selection of NMs with high relevance to the food sector, namely, ZnO, CuO, FeO, SiO2, and Ag NMs and nanocellulose. Results We propose a new AOP (AOP 530) that starts with endocytic lysosomal uptake, leading to lysosomal disruption inducing mitochondrial dysfunction. Mitochondrial impairments can lead to cell injury/death and disrupt the intestinal barrier. The evidence collected supports that these food-related NMs can be taken up by intestinal cells and indicates that intestinal barrier disruption may occur due to Ag, CuO, and SiO2 NMs, while only few studies support this outcome for FeO and ZnO. Lysosomal disruption and mitochondrial dysfunction are rarely evaluated. For nanocellulose, none of the studies report toxicity-related events. Conclusion The collection of existing scientific evidence supporting our AOP linking NM uptake to intestinal barrier impairments allowed us to highlight current evidence gaps and data inconsistencies. These inconsistencies could be associated with the variety of stressors, biological systems, and key event (KE)-related assays used in different studies. This underscores the need for further harmonized methodologies and the production of mechanistic evidence for the safety regulatory assessment of NMs in the food sector.
Collapse
Affiliation(s)
- Deborah Stanco
- European Commission, Joint Research Center (JRC), Ispra, Italy
| | | | - Alessia Bogni
- European Commission, Joint Research Center (JRC), Ispra, Italy
| | | | - Laure-Alix Clerbaux
- Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| |
Collapse
|
33
|
Mignini I, Galasso L, Piccirilli G, Calvez V, Termite F, Esposto G, Borriello R, Miele L, Ainora ME, Gasbarrini A, Zocco MA. Interplay of Oxidative Stress, Gut Microbiota, and Nicotine in Metabolic-Associated Steatotic Liver Disease (MASLD). Antioxidants (Basel) 2024; 13:1532. [PMID: 39765860 PMCID: PMC11727446 DOI: 10.3390/antiox13121532] [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: 09/29/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Oxidative stress has been described as one of the main drivers of intracellular damage and metabolic disorders leading to metabolic syndrome, a major health problem worldwide. In particular, free radicals alter lipid metabolism and promote lipid accumulation in the liver, existing in the hepatic facet of metabolic syndrome, the metabolic dysfunction-associated steatotic liver disease (MASLD). Recent literature has highlighted how nicotine, especially if associated with a high-fat diet, exerts a negative effect on the induction and progression of MASLD by upregulating inflammation and increasing oxidative stress, abdominal fat lipolysis, and hepatic lipogenesis. Moreover, considerable evidence shows the central role of intestinal dysbiosis in the pathogenesis of MASLD and the impact of nicotine-induced oxidative stress on the gut microbiome. This results in an intricate network in which oxidative stress stands at the intersection point between gut microbiome, nicotine, and MASLD. The aim of this review is to delve into the molecular mechanisms linking tobacco smoking and MASLD, focusing on nicotine-induced microbiota modifications and their impact on MASLD development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (L.G.); (G.P.); (V.C.); (F.T.); (G.E.); (R.B.); (L.M.); (M.E.A.); (A.G.)
| |
Collapse
|
34
|
Qi P, Xie R, Liu H, Zhang Z, Cheng Y, Ma J, Wan K, Xie X. Mechanisms of gut homeostasis regulating Th17/Treg cell balance in PMOP. Front Immunol 2024; 15:1497311. [PMID: 39735544 PMCID: PMC11671525 DOI: 10.3389/fimmu.2024.1497311] [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: 09/16/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a metabolic bone disease driven by estrogen deficiency, primarily manifesting as reduced bone mass and heightened fracture risk. Its development is intricately linked to the balance between Th17 and Treg cells. Recent studies have highlighted the significant role of gut homeostasis in PMOP. The gut microbiota profoundly impacts bone health by modulating the host's immune system, metabolic pathways, and endocrine functions. In particular, the regulation of Th17 and Treg cell balance by gut homeostasis plays a pivotal role in the onset and progression of PMOP. Th17 cells secrete pro-inflammatory cytokines that stimulate osteoclast activity, accelerating bone resorption, while Treg cells counteract this process through anti-inflammatory mechanisms, preserving bone mass. The gut microbiota and its metabolites can influence Th17/Treg equilibrium, thereby modulating bone metabolism. Furthermore, the integrity of the gut barrier is critical for systemic immune stability, and its disruption can lead to immune dysregulation and metabolic imbalances. Thus, targeting gut homeostasis to restore Th17/Treg balance offers a novel therapeutic avenue for the prevention and treatment of PMOP.
Collapse
Affiliation(s)
- Peng Qi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | | | - Hao Liu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zixuan Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yuan Cheng
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jilong Ma
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Kangwei Wan
- 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
|
35
|
He T, Qian W. Immunologic derangement caused by intestinal dysbiosis and stress is the intrinsic basis of reactive arthritis. Z Rheumatol 2024; 83:305-313. [PMID: 38403666 PMCID: PMC11655581 DOI: 10.1007/s00393-024-01480-4] [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] [Accepted: 01/02/2024] [Indexed: 02/27/2024]
Abstract
Reactive arthritis (ReA) is defined as arthritis resulting from infections in other body parts, such as the gastrointestinal and urogenital tracts. The primary clinical manifestations involve acute-onset and self-limiting asymmetric large joint inflammation in the lower limbs. Although bacterial or chlamydia infections have long been recognized as playing a pivotal role in its pathogenesis, recent studies suggest that antibiotic treatment may perpetuate rather than eradicate chlamydia within the host, indicating an involvement of other mechanisms in Reactive arthritis. Reactive arthritis is currently believed to be associated with infection, genetic marker (HLA-B27), and immunologic derangement. As an autoimmune disease, increasing attention has been given to understanding the role of the immune system in Reactive arthritis. This review focuses on elucidating how the immune system mediates reactive arthritis and explores the roles of intestinal dysbiosis-induced immune disorders and stress-related factors in autoimmune diseases, providing novel insights into understanding reactive arthritis.
Collapse
Affiliation(s)
- Tao He
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiqing Qian
- Nanjing City Hospital of Chinese Medicine, 157, Daming Road, Nanjing, Qinhuai District, China.
| |
Collapse
|
36
|
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
|
37
|
Su Z, Bian L, Zhao H, Yang C, Gu Y, Cai Y, Yang T, Xu X. KIR2DL5 +CD8 + T cells associate with dietary lipid intake and are active in type 1 diabetes. Int Immunopharmacol 2024; 141:112971. [PMID: 39178517 DOI: 10.1016/j.intimp.2024.112971] [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: 08/06/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Recent studies have shown that KIR+CD8+ T cells play a role in suppressing autoimmunity by eliminating pathogenic CD4+ T cells. However, their specific role in type 1 diabetes (T1D) remains unclear. METHODS In this study, we enrolled 108 patients diagnosed with T1D and 86 healthy individuals. We conducted flow cytometric analysis to examine the various subtypes of KIR+CD8+ T cells derived from peripheral blood mononuclear cells. Additionally, CD8+ T cells were isolated from the peripheral blood of T1D patients to assess the functions of different KIR+CD8+ T cell subtypes. To investigate the influence of lipids on the characteristics and activities of these T cell subtypes, the isolated CD8+ T cells were cultured with varying concentrations of palmitic acid (PA). Furthermore, we utilized an NSG (NOD scid gamma) mouse adoptive transfer model to assess the impact of dietary lipid intake on the functionality of KIR2DL5+CD8+ T cells in vivo. RESULTS We observed variations in circulating KIR+CD8+ T cell subtypes between patients with T1D and healthy controls. Notably, we observed a significant negative correlation between the frequencies of circulating KIR+CD8+ T cells and the titers of ZnT8 autoantibodies in individuals with T1D. Among these subtypes, KIR2DL5+CD8+ T cells demonstrated a positive association with dietary fat intake, characterized by increased perforin expression and reduced PD-1 expression. Importantly, KIR2DL5+CD8+ T cells exhibited enhanced proliferative capacity compared to other KIR+CD8+ T cell subsets. Palmitic acid (PA) was found to enhance the activation of KIR2DL5+CD8+ T cells and strengthened their ability to suppress CD4+ T cell proliferation in T1D patients. Moreover, dietary lipid intake significantly enhanced the functionality of KIR2DL5+CD8+ T cells in an NSG mouse adoptive transfer model. CONCLUSION Our findings suggest that lipid intake enhances the functionality of human KIR2DL5+CD8+ T cells and may offer implications for immunotherapy in T1D.
Collapse
Affiliation(s)
- Zhangyao Su
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lingling Bian
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Endocrinology, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Hang Zhao
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chun Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yong Gu
- 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
| | - 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.
| |
Collapse
|
38
|
Shi P, Li Y, Yang H, Li Q, Li Q, Ye M, Nian D. Lactobacillus rhamnosus ameliorates experimental autoimmune neuritis via modulation of gut microbiota and metabolites. Heliyon 2024; 10:e39126. [PMID: 39524841 PMCID: PMC11550083 DOI: 10.1016/j.heliyon.2024.e39126] [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: 06/16/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background Guillain-Barre syndrome (GBS), an autoimmune disease of the peripheral nervous system, is hallmarked by demyelination and immune cellular infiltration. Experimental autoimmune neuritis (EAN), considered a GBS prototype model, has been studied for its potential therapeutic benefits from lactobacilli. This study evaluated the protective role of Lactobacillus rhamnosus GG (GG) for treatment in EAN. T cell ratio, inflammation factors, sciatic nerve pathology, intestinal permeability, and gut inflammation were assessed on day 19 post-immunization to evaluate GG's effect on EAN. Fecal metabolomics and 16s rRNA microbiome analysis were conducted to elucidate its mechanism. Results GG dynamically balanced CD4+/CD8+T cell ratio, reduced serum IL-1β and TNF-α expression, improved sciatic nerve demyelination and inflammation, and enhanced neurological scores during peak disease period. Intestinal mucosal damage was evident in EAN rats, with downregulated Occludin and ZO-1 and upregulated IL-1β, TNF-α, and Reg3γ. GG treatment restored intestinal mucosal integrity, upregulated Occludin and ZO-1, and downregulated IL-1, TNF-α, and Reg3γ. GG partially rectified the gut microbiota and metabolite imbalance in EAN rats. Conclusion GG mitigates EAN through immune response modulation and inflammation reduction via the gut microbiota and metabolites.
Collapse
Affiliation(s)
- Peng Shi
- Department of Neurology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Yu Li
- Department of Neurology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Hui Yang
- Department of Neurology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Qiang Li
- Department of Neurology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Qianqian Li
- Department of Neurology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Ming Ye
- Department of Neurology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Di Nian
- Department of Medical Examination, Bengbu Medical College, Bengbu 233030, China
| |
Collapse
|
39
|
Xi M, Ruan Q, Zhong S, Li J, Qi W, Xie C, Wang X, Abuduxiku N, Ni J. Periodontal bacteria influence systemic diseases through the gut microbiota. Front Cell Infect Microbiol 2024; 14:1478362. [PMID: 39619660 PMCID: PMC11604649 DOI: 10.3389/fcimb.2024.1478362] [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: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Many systemic diseases, including Alzheimer disease (AD), diabetes mellitus (DM) and cardiovascular disease, are associated with microbiota dysbiosis. The oral and intestinal microbiota are directly connected anatomically, and communicate with each other through the oral-gut microbiome axis to establish and maintain host microbial homeostasis. In addition to directly, periodontal bacteria may also be indirectly involved in the regulation of systemic health and disease through the disturbed gut. This paper provides evidence for the role of periodontal bacteria in systemic diseases via the oral-gut axis and the far-reaching implications of maintaining periodontal health in reducing the risk of many intestinal and parenteral diseases. This may provide insight into the underlying pathogenesis of many systemic diseases and the search for new preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Mengying Xi
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Qijun Ruan
- Department of Periodontics, Shenzhen Longgang Otolaryngology hospital, Shenzhen, China
| | - Sulan Zhong
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Jiatong Li
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Weijuan Qi
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Congman Xie
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiaoyan Wang
- Department of Periodontics, Shenzhen Longgang Otolaryngology hospital, Shenzhen, China
| | - Nuerbiya Abuduxiku
- Department of Stomatology, The First People’s Hospital of Kashi, Kashi, China
| | - Jia Ni
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
40
|
Ren X, Xin L, Peng L, Xiao Y, Zhou Z, Luo H, Zhu Z, Wei Q, Jiang Y, He H, Xiang L, Wang Y, Tang Y, Gu H. Association between sulfur microbial diet and the risk of esophageal cancer: a prospective cohort study in 101,752 American adults. Nutr J 2024; 23:139. [PMID: 39511614 PMCID: PMC11542201 DOI: 10.1186/s12937-024-01035-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Sulfur microbial diet (SMD) is a dietary pattern closely related to the intestinal load of sulfur-metabolizing microbes in humans. Diet and microbes may play an important role in the carcinogenesis of esophagus. However, epidemiological studies on SMD and esophageal cancer (EC) risk are scarce. Here, we evaluated this association based on a large American cohort. METHODS In the cohort of the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial, a SMD score was calculated to evaluate participants' compliance of SMD pattern, with higher scores presenting greater adherence. Cox hazards regression model was used to explore the association between the SMD score and the incidence of EC, esophageal squamous cell carcinoma (ESCC), and esophageal adenocarcinoma (EA). Subgroup analyses were conducted to figure out potential modifiers interacting with SMD on EC. Sensitivity analyses were used to testify the robustness of our main result. RESULTS Among 101,752 participants, 154 EC cases, consisted of 41 ESCC cases and 97 EA cases, were identified with mean follow-up of 8.9 years. In the fully adjusted model, the highest versus the lowest quartiles of the SMD score were found to be associated with an increased risk of EC and ESCC (EC: HRQ4 vs. Q1: 1.64; 95% CI: 1.05, 2.56; P = 0.016 for trend; ESCC: HRQ4 vs. Q1: 2.37; 95% CI: 1.02, 5.47; P = 0.031 for trend), while not significantly associated with increases risk of EA (HRQ4 vs. Q1: 1.41; P = 0.144 for trend). The main result remained through a series of sensitivity analyses. Subgroup analyses showed a stronger association between SMD and EC in participants with no regular consumption of aspirin (HRQ4 vs. Q1: 1.90; 95% CI: 1.04, 3.47) than in those using aspirin regularly (HRQ4 vs. Q1: 1.37; 95% CI: 0.71, 2.66) (P = 0.008 for interaction). CONCLUSION Adherence to the SMD pattern may be associated with increased risks of EC and ESCC, particularly for EC in individuals who do not regularly consume aspirin.
Collapse
Affiliation(s)
- Xiaorui Ren
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Li Xin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Linglong Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Yi Xiao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Zhihang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haoyun Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Zhiyong Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Qi Wei
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Yahui Jiang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Hongmei He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Ling Xiang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxu Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China
| | - Yunhao Tang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China.
| | - Haitao Gu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, No.288 Tianwen Avenue, Nan'an District, Chongqing, 400010, China.
| |
Collapse
|
41
|
Biennier S, Fontaine M, Duquenoy A, Schwintner C, Doré J, Corvaia N. Narrative Review: Advancing Dysbiosis Treatment in Onco-Hematology with Microbiome-Based Therapeutic Approach. Microorganisms 2024; 12:2256. [PMID: 39597645 PMCID: PMC11596191 DOI: 10.3390/microorganisms12112256] [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: 09/30/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
This review explores the complex relationship between gut dysbiosis and hematological malignancies, focusing on graft-versus-host disease (GvHD) in allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients. We discuss how alterations in microbial diversity and composition can influence disease development, progression, and treatment outcomes in blood cancers. The mechanisms by which the gut microbiota impacts these conditions are examined, including modulation of immune responses, production of metabolites, and effects on intestinal barrier function. Recent advances in microbiome-based therapies for treating and preventing GvHD are highlighted, with emphasis on full ecosystem standardized donor-derived products. Overall, this review underscores the growing importance of microbiome research in hematology-oncology and its potential to complement existing treatments and improve outcomes for thousands of patients worldwide.
Collapse
Affiliation(s)
- Salomé Biennier
- MaaT Pharma, 69007 Lyon, France; (S.B.); (A.D.); (C.S.); (N.C.)
| | | | - Aurore Duquenoy
- MaaT Pharma, 69007 Lyon, France; (S.B.); (A.D.); (C.S.); (N.C.)
| | | | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, 78350 Jouy-en-Josas, France;
| | | |
Collapse
|
42
|
Guilliams TG, Weintraub JL, Spar M. Intestinal Permeability In Subjects With Rheumatoid Arthritis: A Critical Therapeutic Priority. Integr Med (Encinitas) 2024; 23:16-26. [PMID: 39534664 PMCID: PMC11552960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Rheumatoid arthritis is increasingly being recognized as the synovial manifestation of a group of systemic autoinflammatory conditions known as immune-mediated inflammatory diseases. While each of these conditions displays unique diagnostic signs and symptoms based on the tissue targeted by inflammation, most immune-mediated inflammatory diseases share common features, including their immune-signaling pathways. Owing to these similarities, great advances have emerged in the past few decades using therapies designed to block downstream inflammatory mediators (eg, cytokine-blocking biologics, Janus Kinase (JAK) inhibitors). Unfortunately, fewer advances have been made in therapies that have the potential to target the upstream antecedents and triggers of these complex inflammatory diseases, such as the immunologic chain of events triggered by intestinal hyperpermeability (ie, leaky gut) or gastrointestinal dysbiosis (ie, alterations in the gut microbiota). In the past few decades, intestinal hyperpermeability has emerged as an important antecedent for a wide range of chronic immunological and metabolic conditions, including celiac disease, obesity, cardiovascular disease, and a number of immune-mediated inflammatory diseases such as inflammatory bowel disease, psoriasis, and rheumatoid arthritis. In this narrative review, we discuss the growing awareness that biomarkers of intestinal permeability are frequently associated with non-gastrointestinal immune-mediated inflammatory diseases, particularly those associated with the gut-joint axis, such as rheumatoid arthritis. We suggest that measures of intestinal permeability, along with lifestyle and nutrient interventions that target gut-barrier function, may be important adjunctive clinical tools to help patients with rheumatoid arthritis achieve and maintain remission.
Collapse
Affiliation(s)
- Thomas G. Guilliams
- Scientific Director, AndHealth, Columbus, Ohio; Founder and Director of the Point Institute, Stevens Point, Wisconsin; Adjunct Associate Professor, School of Pharmacy, University of Wisconsin—Madison
| | - Jill L. Weintraub
- Rheumatology Advisor, AndHealth, Columbus, Ohio; Integrative Rheumatology Consultants, Westchester and New York, New York
| | - Myles Spar
- VP and National Director of Medical Services, AndHealth, Columbus, Ohio; Associate Professor, Andrew Weil Center for Integrative Medicine, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
43
|
Jia N, Meng Y, Li J, Cui M, Li Y, Jiang D, Chu X. Pharmacodynamic and pharmacokinetic study of Shaoyao Gancao decoction for repairing intestinal barrier damage in ulcerative colitis. Mol Immunol 2024; 175:132-142. [PMID: 39369520 DOI: 10.1016/j.molimm.2024.09.013] [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: 06/05/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE To study the therapeutic effect and mechanism of Shaoyao Gancao Decoction (SGD) on ulcerative colitis (UC) mice based on the perspective of intestinal barrier, and this study provides a new consultation for the clinical application of SGD. METHODS The chemical composition of SGD was characterized by HPLC. The UC mouse model was constructed by 3 % dextran sodium sulfate (DSS), which were randomly divided into the model group (DSS), the positive drug group (5-ASA), the Shaoyao group (SYD), Gancao group (GCD), and the Shaoyao Gancao Decoction group (SGD) at low, medium, and high dosages, respectively. The effects of each drug treatment group on UC were evaluated by the rate of body weight loss, disease activity index (DAI), colon length, spleen index, histopathological evaluations, and the levels of serum inflammatory factors (IL-1β, IL-6, IL-10, IL-21, and TNF-α). The goblet cell was observed by Alcian blue/periodic acid-Schiff (AB/PAS) straining, ELISA was used to detect the content of LPS in serum, and Western blot was used to detect the changes in the expression of tight junction proteins ZO-1, occludin, and the pathway proteins TLR4 and NF-κBp65 in the colonic tissues, to explore the protective effect of SGD on the intestinal barrier of UC mice. The vivo absorption process of the main active ingredients in the SG, SY and GC groups was determined by LC-MS. RESULTS The contents of albiflorin, paeoniflorin, liquiritin apioside, liquiritin and glycyrrhetinic acid were 6.1227 mg/g, 20.8993 mg/g, 4.0054 mg/g, 3.6140 mg/g and 8.2515 mg/g, respectively. Compared with DSS group, SGD reduced weight loss(P<0.01) and DAI scores(P<0.05), prevented colon shortening(P<0.01), and ameliorated histopathological damage of the colon in UC mice(P<0.01). SGD also protected the intestinal barrier to alleviate UC by significantly reducing serum LPS and inflammatory factor levels, altering the number of goblet cells, promoting tight junction proteins (ZO-1 and occludin) and decreasing the expression of TLR4 and NF-κB in colonic tissues. Pharmacokinetic results showed that there was no significant difference in Cmax, AUC0-t (μg/L.h) and Tmax of albiflorin and paeoniflorin between the SY and SG groups, the Tmax was within 1 h; the AUC0-t (μg/L.h) of liquiritin and glycyrrhizic acid were about 1.6 and 1.9 times higher in the SG group compared to the GC group, respectively. The Cmax, Tmax and AUC0-t (μg/L.h) of glycyrrhizinic acid were significantly reduced to 0.73, 0.68 and 0.68 times of that of the GC group. CONCLUSION SGD may have a therapeutic effect on DSS-induced UC mice by repairing the damaged intestinal barrier through the TLR4/NF-κB pathway. The combination of Shaoyao and Gancao increased the absorption of liquiritin and glycyrrhizic acid in vivo. The combination of Shaoyao and Gancao could promote the absorption of Gancao, and that the pairing of the two herbs could have a synergistic effect.
Collapse
Affiliation(s)
- Nini Jia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yun Meng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jing Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Mengyao Cui
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yaqing Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | | | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230012, China; Engineering Technology Research Center of Modern Pharmaceutical Preparation, Hefei, Anhui Province 230012, China.
| |
Collapse
|
44
|
Lee MH, Nuccio SP, Mohanty I, Hagey LR, Dorrestein PC, Chu H, Raffatellu M. How bile acids and the microbiota interact to shape host immunity. Nat Rev Immunol 2024; 24:798-809. [PMID: 39009868 DOI: 10.1038/s41577-024-01057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/17/2024]
Abstract
Bile acids are increasingly appearing in the spotlight owing to their novel impacts on various host processes. Similarly, there is growing attention on members of the microbiota that are responsible for bile acid modifications. With recent advances in technology enabling the discovery and continued identification of microbially conjugated bile acids, the chemical complexity of the bile acid landscape in the body is increasing at a rapid pace. In this Review, we summarize our current understanding of how bile acids and the gut microbiota interact to modulate immune responses during homeostasis and disease, with a particular focus on the gut.
Collapse
Affiliation(s)
- Michael H Lee
- Division of Host-Microbe Systems and Therapeutics, Department of Paediatrics, University of California San Diego, La Jolla, CA, USA
| | - Sean-Paul Nuccio
- Division of Host-Microbe Systems and Therapeutics, Department of Paediatrics, University of California San Diego, La Jolla, CA, USA
| | - Ipsita Mohanty
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lee R Hagey
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Hiutung Chu
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), La Jolla, CA, USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems and Therapeutics, Department of Paediatrics, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), La Jolla, CA, USA.
| |
Collapse
|
45
|
Lin SKK, Kuo PH, Hsu CY, Chiu YH, Chen CH. The effects of Lactobacillus plantarum PS128 in patients with major depressive disorder: an eight-week double-blind, placebo-controlled study. Asian J Psychiatr 2024; 101:104210. [PMID: 39232392 DOI: 10.1016/j.ajp.2024.104210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/23/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Major depressive disorder (MDD) is a complex mental disorder, potentially linked to the gut-microbiota-brain axis. Probiotics like Lactobacillus plantarum PS128 (PS128) may improve depressive symptoms by modulating the gut microbiota based on our previous open trial. We conducted an 8-week double-blind, placebo-controlled trial to investigate the impact of PS128 on depression severity, markers of inflammation and gut permeability, and the gut microbiota composition in 32 patients with MDD with stable antidepressant treatment but moderate symptom severity. Following the 8-week intervention, both the Hamilton Depression Rating Scale-17 score (HAMD), and Depression and Somatic Symptoms Scale (DSSS) showed a significant decrease in both groups (p<0.001). However, there was no significant difference in the change of depression severity between groups (p=0.203). Moreover, alterations in serum levels of high sensitivity C-reactive protein, interleukin-6, tumor necrosis factor-α, and intestinal fatty acid binding protein, as well as changes in the gut microbiota composition, did not exhibit significant differences before and after intervention or between the groups. In comparison to the placebo group, our study did not find significant effects of PS128 on depressive symptoms, biomarkers of inflammation and gut permeability, and the overall gut microbiota composition. Nonetheless, we observed a potential impact of PS128 on the symbiosis of specific taxa. To comprehensively understand the psychophysiological effects of PS128 in patients with MDD, further research with a larger sample size is imperative.
Collapse
Affiliation(s)
- Shih-Kai Kevin Lin
- Department of Public Health & Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Department of Public Health & Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan; Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Chia-Yueh Hsu
- Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Hung Chiu
- Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chun-Hsin Chen
- Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
46
|
Budau R, Okamura T, Hasegawa Y, Nakanishi N, Hamaguchi M, Fukui M. Supplementation of Miso to a Western-Type Diet Stimulates ILC3s and Decreases Inflammation in the Small Intestine. Nutrients 2024; 16:3743. [PMID: 39519576 PMCID: PMC11547460 DOI: 10.3390/nu16213743] [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: 09/30/2024] [Revised: 10/16/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Western-type diets (WDs) damage the intestinal barrier by disrupting the gut microbiota composition and causing inflammation, leading to the development of obesity, type 2 diabetes, and non-alcoholic fatty liver disease. Short-chain fatty acids (SCFAs) are produced by the gut microbiota and found in fermented foods and can stimulate the anti-inflammatory action of type 3 innate lymphoid cells (ILCS3s) in the intestine. This study hypothesised that supplementing miso, a Japanese fermented food, to a WD could increase the levels of SCFAs and thus stimulate ILC3s, decreasing inflammation in the intestine and protecting intestinal barrier integrity. METHODS Mice with RORγt total (KI/KI) or partial (KI/w) knockout were fed a high-fat high-sugar diet (HFHSD) for eight weeks as a model of WD. Half of the mice received miso supplementation in addition to the HFHSD. Weight gain, glucose tolerance and insulin resistance, intestinal barrier integrity, intestinal immunity, and liver condition were assessed. RESULTS Miso supplementation increased SCFA levels in the small intestine, which stimulated ILC3 function in KI/w mice. Glucose tolerance was improved, intestinal barrier integrity was ameliorated, and mucus production was increased. The level of IL-22 was increased, while pro-inflammatory ILC1s, M1 macrophages, TNF-α, and IL-1β were decreased. Liver condition was not affected. CONCLUSIONS This study demonstrated that miso supplementation influenced several factors involved in inflammation and intestinal barrier integrity by stimulating ILC3s in RORγt heterozygous mice. Moreover, it showed that the number of ILC3s is not the key factor in immune regulation, but rather the ability of ILC3 to produce IL-22 and employ it to control the immune response in the small intestine.
Collapse
MESH Headings
- Animals
- Intestine, Small/metabolism
- Intestine, Small/immunology
- Lymphocytes/immunology
- Lymphocytes/metabolism
- Mice
- Inflammation
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Diet, Western/adverse effects
- Gastrointestinal Microbiome
- Dietary Supplements
- Mice, Knockout
- Diet, High-Fat/adverse effects
- Male
- Fatty Acids, Volatile/metabolism
- Insulin Resistance
- Interleukin-22
- Immunity, Innate
- Mice, Inbred C57BL
- Interleukins/metabolism
Collapse
Affiliation(s)
| | - Takuro Okamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (R.B.)
| | | | | | | | | |
Collapse
|
47
|
Iqbal ZS, Halkjær SI, Ghathian KSA, Heintz JE, Petersen AM. The Role of the Gut Microbiome in Urinary Tract Infections: A Narrative Review. Nutrients 2024; 16:3615. [PMID: 39519448 PMCID: PMC11547363 DOI: 10.3390/nu16213615] [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: 10/03/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Urinary tract infections (UTIs) represent a substantial health concern worldwide. Although it is known that the gut can act as a reservoir for UTI-causing pathogens, the exact role of the gut microbiome in developing UTIs remains unclear. This review aims to investigate the link between the gut microbiome and UTIs and whether gut dysbiosis increases the risk of getting a UTI. METHODS To find relevant studies, a search was conducted across three databases, PubMed, EMBASE and Cochrane Library. Only records that directly described the association between the gut microbiome and UTIs were included in this review. RESULTS Of the numerous studies retrieved, eight studies met the pre-set criteria and were selected for the review. The findings suggest several potential ways in which gut dysbiosis might enhance UTI susceptibility. A low gut microbiome diversity, a reduced level of bacteria involved in short-chain fatty acid (SCFA) production and a high abundance of Escherichia coli (E. coli) among UTI patients all offer a reasonable explanation for the existence of a link between an altered gut microbiome and UTIs. However, contradictory study results make it difficult to verify this. CONCLUSIONS Research on the link between the gut microbiome and UTIs is limited, and further studies need to be carried out to substantiate this relationship, as this can bring attention to finding improved and more relevant treatment for UTIs.
Collapse
Affiliation(s)
- Zaryan Safdar Iqbal
- Gastrounit, Medical Section, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre, Denmark; (Z.S.I.); (S.I.H.)
| | - Sofie Ingdam Halkjær
- Gastrounit, Medical Section, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre, Denmark; (Z.S.I.); (S.I.H.)
| | - Khaled Saoud Ali Ghathian
- Department of Clinical Microbiology, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre, Denmark; (K.S.A.G.); (J.E.H.)
| | - Julie Elm Heintz
- Department of Clinical Microbiology, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre, Denmark; (K.S.A.G.); (J.E.H.)
| | - Andreas Munk Petersen
- Gastrounit, Medical Section, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre, Denmark; (Z.S.I.); (S.I.H.)
- Department of Clinical Microbiology, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre, Denmark; (K.S.A.G.); (J.E.H.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
48
|
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
|
49
|
Cao Y, Chen J, Xiao J, Hong Y, Xu K, Zhu Y. Butyrate: a bridge between intestinal flora and rheumatoid arthritis. Front Immunol 2024; 15:1475529. [PMID: 39478858 PMCID: PMC11521846 DOI: 10.3389/fimmu.2024.1475529] [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: 08/04/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
In patients with rheumatoid arthritis (RA), intestinal flora imbalance and butyrate metabolism disorders precede clinical arthritis and are associated with the pathogenesis of RA. This imbalance can alter the immunology and intestinal permeability of the intestinal mucosa, leading to damage to the intestinal barrier. In this context, bacteria and their metabolites can enter the bloodstream and reach the distant target tissues of the host, resulting in local inflammation and aggravating arthritis. Additionally, arthritis is also exacerbated by bone destruction and immune tolerance due to disturbed differentiation of osteoclasts and adaptive immune cells. Of note, butyrate is a metabolite of intestinal flora, which not only locally inhibits intestinal immunity and targets zonulin and tight junction proteins to alleviate intestinal barrier-mediated arthritis but also inhibits osteoclasts and autoantibodies and balances the immune responses of T and B lymphocytes throughout the body to repress bone erosion and inflammation. Therefore, butyrate is a key intermediate linking intestinal flora to the host. As a result, restoring the butyrate-producing capacity of intestinal flora and using exogenous butyrate are potential therapeutic strategies for RA in the future.
Collapse
Affiliation(s)
- Yang Cao
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jingjing Chen
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jing Xiao
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yujie Hong
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Yan Zhu
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
50
|
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
|