1
|
Steinert RE, Rehman A, Sadabad MS, Milanese A, Wittwer-Schegg J, Burton JP, Spooren A. Microbial micronutrient sharing, gut redox balance and keystone taxa as a basis for a new perspective to solutions targeting health from the gut. Gut Microbes 2025; 17:2477816. [PMID: 40090884 PMCID: PMC11913388 DOI: 10.1080/19490976.2025.2477816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/05/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025] Open
Abstract
In health, the gut microbiome functions as a stable ecosystem maintaining overall balance and ensuring its own survival against environmental stressors through complex microbial interaction. This balance and protection from stressors is maintained through interactions both within the bacterial ecosystem as well as with its host. As a consequence, the gut microbiome plays a critical role in various physiological processes including maintaining the structure and function of the gut barrier, educating the gut immune system, and modulating the gut motor, digestive/absorptive, as well as neuroendocrine system all of which are crucial for human health and disease pathogenesis. Pre- and probiotics, widely available and clinically established, offer various health benefits primarily by beneficially modulating the gut microbiome. However, their clinical outcomes can vary significantly due to differences in host physiology, diets, individual microbiome compositions, and other environmental factors. This perspective paper highlights emerging scientific insights into the importance of microbial micronutrient sharing, gut redox balance, keystone species, and the gut barrier in maintaining a diverse and functional microbial ecosystem, and their relevance to human health. We propose a novel approach that targets microbial ecosystems and keystone taxa performance by supplying microbial micronutrients in the form of colon-delivered vitamins, and precision prebiotics [e.g. human milk oligosaccharides (HMOs) or synthetic glycans] as components of precisely tailored ingredient combinations to optimize human health. Such a strategy may effectively support and stabilize microbial ecosystems, providing a more robust and consistent approach across various individuals and environmental conditions, thus, overcoming the limitations of current single biotic solutions.
Collapse
Affiliation(s)
- Robert E. Steinert
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
- Department of Surgery and Transplantation, University Hospital Zurich (USZ) and University of Zurich (UZH), Zürich, Switzerland
| | - Ateequr Rehman
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| | | | - Alessio Milanese
- Data Science, Science & Research, Dsm-Firmenich, Delft, Netherlands
| | | | - Jeremy P. Burton
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada
| | - Anneleen Spooren
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| |
Collapse
|
2
|
Yang SJ, Yu XK, Zuo Q. Branched- Chain Fatty Acids and Obesity: A Narrative Review. Nutr Rev 2025; 83:1314-1326. [PMID: 40207993 DOI: 10.1093/nutrit/nuaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Branched- chain fatty acids (BCFAs) are a category of saturated fatty acids that are commonly present in various organisms and play a crucial role in a variety of metabolic reactions, including anticancer, lipid-lowering, anti-inflammatory, and neuroprotective actions. Currently, there is growing interest in the relationship between BCFAs and obesity. Branched- chain fatty acids regulate the gene expression of related enzymes by activating PPARα and sterol regulatory element-binding protein-1c, thereby reducing triglyceride synthesis in the body. Additionally, BCFAs reduce inflammation by decreasing the expression of pro-inflammatory factors in obesity such as cyclooxygenase-2, interleukin-6, and lipoxygenase-15 genes. Branched- chain fatty acids can also expedite the conversion of branched chain amino acids to BCFAs to regulate obesity-induced insulin resistance. In this article we provide a comprehensive review of research progress on how BCFAs affect obesity from the perspectives of lipid metabolism, inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Shi-Jiao Yang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xin-Kai Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Qun Zuo
- School of Sports Performance, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
3
|
Liu YH, Lee YL, Han CL, Lo YC, Liao ZA, Shih YS, Lin YW, Peng SW, Lee KY, Ho SC, Wu SM, Lin CW, Chung KF, Chang JH, Chuang HC. ITIH4 alleviates OVA-induced asthma by regulating lung-gut microbiota. Mol Med 2025; 31:204. [PMID: 40410722 PMCID: PMC12102971 DOI: 10.1186/s10020-025-01270-x] [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: 02/21/2025] [Accepted: 05/16/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4), a Type 2 acute phase protein, is critical for resolving inflammation and promoting tissue repair. While its role in chronic respiratory diseases is recognized, its effects on asthma remain unclear. This study investigated the effects of ITIH4 on the modulation of lung and gut microbiota, the attenuation of allergic inflammation, and the improvement of respiratory outcomes in an asthma mouse model. METHODS Six-week-old male Balb/c mice were divided into five groups: control, ITIH4, ovalbumin (OVA), and two OVA + ITIH4 treatment groups at different doses. Lung function and oxygen saturation were measured, and bronchoalveolar lavage fluid (BALF) was analyzed for white blood cell counts and cytokines. Lung and gut microbiota were profiled using 16 S rRNA gene sequencing, and short-chain fatty acids (SCFAs) were measured using gas chromatography-mass spectrometry (GC-MS). Proteomic profiling of intestinal tissues was conducted to identify ITIH4-associated signaling pathways. RESULTS ITIH4 administration significantly mitigated OVA-induced asthma symptoms by reducing weight loss, airway resistance, and tissue damping (p < 0.05). Histological analysis showed decreased airway wall thickening and lung injury scores (p < 0.05). ITIH4 also lowered BALF eosinophils and lymphocytes, IgE, and Th2 cytokines (IL-4, IL-5, and IL-13) (p < 0.05). ITIH4 treatment modulated microbiome composition, enriching Gram-positive taxa (Nocardioidaceae and Acholeplasmataceae) and depleting Gram-negative Helicobacteraceae (p < 0.05). SCFAs correlated with microbiome alterations, notably reduced 4-methylpentanoic acid levels (p < 0.05). Proteomic analysis revealed a dose-dependent activation of granzyme A signaling and suppression of metabolic and solute transport pathways. CONCLUSIONS ITIH4 ameliorates asthma symptoms by modulating lung and gut microbiota, dampening Th2-driven inflammation, and restoring mucosal immune balance. These findings support ITIH4 as a potential candidate for microbiome-targeted asthma therapy.
Collapse
Affiliation(s)
- Yi-Hsuan Liu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Li Han
- Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chun Lo
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Zih-An Liao
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yu-Syuan Shih
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yi-Wen Lin
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Syue-Wei Peng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Cheng-Wei Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, Taipei Medical University, Taipei, Taiwan
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- National Heart and Lung Institute, Imperial College London, London, UK.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
4
|
Guo H, Bi Y, Zhang G, Luo S, Jia X, Yang R, Peng Y, Zhang L. Alcohol-induced bone loss driven by dysregulated spatial distribution of gut microbiota and PGD2-IL17 pathway-mediated osteoclast activation. Front Microbiol 2025; 16:1551028. [PMID: 40443995 PMCID: PMC12121995 DOI: 10.3389/fmicb.2025.1551028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/25/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Alcohol-induced damage to bone microstructure leads to alcoholic osteoporosis (AOP). While prior studies have demonstrated alcohol's negative impact on bone density, the mechanisms by which alcohol induces osteoporosis through immune pathways, gut microbiota dysbiosis, and metabolic alterations remain insufficiently characterized. Given that alcohol is primarily absorbed in the upper gastrointestinal tract, in this research, we aimed to elucidate the role of spatial distribution disorders in gut microbiota and metabolites in the pathogenesis of alcohol-induced osteoporosis. We further sought to evaluate the potential of microbiota supplementation and targeted immunosuppressants as therapeutic strategies for related bone diseases. Methods An osteoporosis model using mice was established using alcohol drinking bottles, and bone loss was validated using micro-computed tomography. Segmented intestinal samples and fecal samples were analyzed using 16S rRNA sequencing and metabolomics. Mechanistic studies were conducted by supplementing R. intestinalis, prostaglandin D2 (PGD2), and its specific immune inhibitor, ramatroban. Analytical methods included tartrate-resistant acid phosphatase staining, flow cytometry, and enzyme-linked immunosorbent assay. Results Alcohol disrupted the spatial complexity of intestinal segments and fecal microbiota in mice, causing metabolic dysregulation and ultimately leading to elevated PGD2 levels. This, in turn, triggered Th17/Treg immune imbalance and osteoclast activation, resulting in bone loss. Supplementation with the probiotic R. intestinalis or inhibition of PGD2 significantly improved bone density and alleviate inflammation. Conclusion This study demonstrates that alcohol-induced elevation of PGD2 is a key pathogenic factor in AOP. PGD2 accelerates bone loss by promoting osteoclast formation through the activation of Th17 cells. Furthermore, this study highlights the importance of investigating the spatial distribution of gut microbiota and metabolites, providing potential targets and novel strategies for the precise treatment of AOP and other diseases associated with external stimuli.
Collapse
Affiliation(s)
- Haoyu Guo
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Gongzi Zhang
- Department of Rehabilitation, Chinese PLA General Hospital, Beijing, China
| | - Shicheng Luo
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xiaopeng Jia
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Jinzhou Medical University, Jinzhou, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Ye Peng
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Lihai Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Zhang T, Wang Y, Gu Y, Wu J, Zhan X, Gong P. Gellan gum-sialoglycan conjugates: a mucin mimic for alleviating inflammation in Caco-2 cells and modulating gut microbiota in the elderly. Int J Biol Macromol 2025; 310:143478. [PMID: 40286969 DOI: 10.1016/j.ijbiomac.2025.143478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/13/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
The sialylation of intestinal mucins plays a crucial role in maintaining intestinal homeostasis and shaping the gut microbiota. This study aims to develop a mucin-like polymer to modulate the intestinal microbiota in the elderly individuals. We synthesized functionalized conjugates using sialic acid monomer, 3'-sialyllactose, and Gellan gum, resulting in a series of GG-sialoglycan conjugates with diverse glycan chain lengths and terminal structures. The sialic acid contents of GG-EDA-Sia, GG-EDA-SL, GG-HAD-Sia, and GG-HAD-SL were 28.48 %, 29.78 %, 23.45 and 41.58 %, respectively. These conjugates exhibited anti-digestion effects. In addition, they demonstrated favorable biocompatibility and exhibited notable anti-inflammatory properties. In vitro fermentation experiments using fecal bacteria from elderly individuals revealed that GG-sialoglycan conjugates enhanced the proliferation of beneficial bacteria such as Lactobacillus, Bifidobacterium, and Blautia, while simultaneously suppressing pathogens like Escherichia-Shigella. It is worth noting that GG-sialoglycan conjugates containing 3'-sialyllactose exhibited superior prebiotic activity compared to those with sialic acid monomer. In summary, our findings strongly support the promising potential of functional sialic acid-based macromolecular glycan conjugates as a novel strategy to improve aging-related intestinal health.
Collapse
Affiliation(s)
- Tiantian Zhang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yuying Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yiqun Gu
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Jianrong Wu
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Xiaobei Zhan
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Ping Gong
- Department of Obstetrical, Affiliated Hospital of Jiangnan University, Wuxi 214123, China
| |
Collapse
|
6
|
Abavisani M, Tafti P, Khoshroo N, Ebadpour N, Khoshrou A, Kesharwani P, Sahebkar A. The heart of the matter: How gut microbiota-targeted interventions influence cardiovascular diseases. Pathol Res Pract 2025; 269:155931. [PMID: 40174272 DOI: 10.1016/j.prp.2025.155931] [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: 09/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
The human body is habitat to a wide spectrum of microbial populations known as microbiota, which play an important role in overall health. The considerable research has mostly focused on the gut microbiota due to its potential to impact numerous physiological functions and its correlation with a variety of disorders, such as cardiovascular diseases (CVDs). Imbalances in the gut microbiota, known as dysbiosis, have been linked to the development and progression of CVDs through various processes, including the generation of metabolites like trimethylamine-N-oxide and short-chain fatty acids. Studies have also looked at the idea of using therapeutic interventions, like changing your diet, taking probiotics or prebiotics, or even fecal microbiota transplantation (FMT), to change the gut microbiota's make-up and how it works in order to prevent or treat CVDs. Exploring the cause-and-effect connection between the gut microbiota and CVDs offers a hopeful path for creating innovative microbiome-centered strategies to prevent and cure CVDs. This review presents an in-depth review of the correlation between the gut microbiota and CVDs, as well as potential therapeutic approaches for manipulating the gut microbiota to enhance cardiovascular health.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pourya Tafti
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khoshroo
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pardesh, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Yang H, Zhao Y, Zhang R, Zhao L, Yang H, Liao X. CiLi (Rosa roxburghii Tratt.) polyphenols improve colitis via gut microbiota-lipid mediator-immunity axis. Food Res Int 2025; 209:116257. [PMID: 40253185 DOI: 10.1016/j.foodres.2025.116257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Dysbiosis of gut microbiome is one of the most important factors leading to inflammatory bowel disease (IBD). Intake of phytochemicals from fruits and vegetables is an effective way to improve IBD, but how these bioactivators regulate gut microbiota to exert healthy effects remains unclear. Here, we found that pretreatment with CiLi juice, particularly its polyphenol component, alleviated dextran sulfate sodium (DSS)-induced colitis while preserving intestinal barrier integrity. CiLi polyphenols (CL_PP) reduced inflammation and oxidative stress in colon tissue and enriched fecal short-chain fatty acids. Importantly, CL_PP significantly regulated the gut microbiome diversity, increasing beneficial bacteria (e.g., Clostridia_UCG-014, f_Muribaculaceae and Ileibacterium_valens) while decreasing harmful bacteria (Escherichia-Shigella and Romboutsia). Multiomics analysis revealed that CL_PP upregulated bioactive lipid metabolites, particularly those derived from polyunsaturated fatty acids (e.g., resolvin D2, prostaglandin A1, and glycerophosphocholine) related gene expressions (Pltp, Alox15 and Pld4). Additionally, CL-PP downregulated the oxidative stress markers (oxidized glutathione and glutathione peroxidase 3), and immune cell markers (CD8 and CD68). Fecal microbiota transplantation confirmed that the fecal microbiota from CL_PP-treated mice exhibited anti-colitis effects. These effects were diminished in antibiotic-treated mice, underscoring the importance of the gut microbiota in mediating the CL_PP's anti-inflammatory benefits. This study suggests that CL_PP is a potential modulator of gut microbiome dysbiosis for the prevention and treatment of colitis.
Collapse
Affiliation(s)
- Huanzhi Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yang Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ruiqi Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Liang Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Haixia Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Xiaojun Liao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
8
|
Manhães LDM, Santana L, Alves MR, Leal PM, de Velasco PC, Santana I, de Brito JS, Mafra D, Borges NA. Trimethylamine N-Oxide Plasma Levels Following Red Meat and Cod Fish Intake: A Pilot Crossover Trial in Hemodialysis Patients. Mol Nutr Food Res 2025; 69:e70031. [PMID: 40079058 DOI: 10.1002/mnfr.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 03/14/2025]
Abstract
SCOPE The uremic toxin trimethylamine N-oxide (TMAO) accumulates in patients with chronic kidney disease (CKD) and is associated with its progression, cardiovascular disease, and other complications. The gut microbiota produces TMAO from substrates mainly found in red meat, eggs, and dairy. However, some saltwater fish also contain high levels of TMAO. Although fish consumption is generally linked to beneficial effects, its effects on CKD patients require further research. METHODS AND RESULTS This study compares the effect of red meat and cod fish intake on TMAO plasma levels in CKD patients undergoing hemodialysis (HD). Participants received a single animal protein source (red meat vs. cod fish) for lunch and dinner for four consecutive days (each intervention), with a 2-week washout period in between. TMAO plasma levels were analyzed using LC-MS/MS. All 14 patients concluded the red meat intervention, while one refused to participate in the fish intervention. No significant difference in TMAO plasma levels was found post-red meat (p = 0.21) or fish intervention (p = 0.91), as well as between groups (p = 0.43). CONCLUSION In this study, 4 days of red meat and cod fish intake did not significantly impact TMAO levels in HD patients, while other factors may be associated with their circulating levels.
Collapse
Affiliation(s)
- Larissa de Mattos Manhães
- Graduate Program in Food, Nutrition and Health - Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rio de Janeiro (RJ), Brazil
| | - Ludmilla Santana
- Graduate Program in Food, Nutrition and Health - Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rio de Janeiro (RJ), Brazil
| | - Marcelo Ribeiro Alves
- HIV/AIDS Clinical Research Center, National Institute of Infectology (INI/Fiocruz), Rio de Janeiro (RJ), Brazil
| | - Priscila Mansur Leal
- Pedro Ernesto University Hospital, State University of Rio de Janeiro (UERJ), Rio de Janeiro (RJ), Brazil
| | | | - Isabelle Santana
- Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rio de Janeiro (RJ), Brazil
| | - Jessyca Sousa de Brito
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
| | - Denise Mafra
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro (RJ), Brazil
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói (RJ), Brazil
| | - Natália Alvarenga Borges
- Graduate Program in Food, Nutrition and Health - Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rio de Janeiro (RJ), Brazil
| |
Collapse
|
9
|
Chauhan G, Rieder F. The Pathogenesis of Inflammatory Bowel Diseases. Surg Clin North Am 2025; 105:201-215. [PMID: 40015812 PMCID: PMC11868724 DOI: 10.1016/j.suc.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Inflammatory bowel diseases (IBDs) are relapsing, remitting inflammatory diseases of the intestinal tract. Familial aggregation and genome-wide association studies revealed susceptibility variants that point toward a combination of innate immune and adaptive immune dysregulation that in concert with environmental factors, such as our microbiome, can initiate and perpetuate inflammation. Innate immune perturbations include functional abnormalities in the intestinal barrier, endoplasmic reticulum stress, and abnormal recognition of microbes. Adaptive immune changes include dysregulation of cytokines, regulatory T cells, and leukocyte migration. IBD is linked with an abnormal wound-healing response leading to fibrosis. This article summarizes key pathogenic mechanisms in the pathogenesis of IBDs.
Collapse
Affiliation(s)
- Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA; Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases Institute; Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|
10
|
Wang Y, Guo J, Mao Z, Chen Y. Symphony of the gut microbiota and endocannabinoidome: a molecular and functional perspective. Front Cell Infect Microbiol 2025; 15:1566290. [PMID: 40207053 PMCID: PMC11979265 DOI: 10.3389/fcimb.2025.1566290] [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: 01/24/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
This review examines the impact of interactions between the gut microbiota and the endocannabinoidome (eCBome) on health and disease, highlighting their significance for physiological and pathological processes. We identify key research gaps and challenges to advance the field. The review discusses the role of dietary patterns and physical activity in regulating these interactions. It also explores the complex nature of these interactions in conditions such as inflammatory bowel disease (IBD), depression, anxiety, Alzheimer's disease (AD), and metabolic disorders. This analysis evaluates their contributions to disease onset and progression, and examines the molecular mechanisms and signaling pathways involved. From this, we provide forward-looking perspectives on future research directions, advocating for a more nuanced understanding of the gut microbiota-eCBome axis. We anticipate that future research will integrate gut microbiota-endocannabinoidome interactions into therapeutic strategies for a broad range of diseases.
Collapse
Affiliation(s)
| | | | | | - Ying Chen
- Department of Pediatric Gastroenterology, Shengjing Hospital of China Medical
University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Wagner CA, Frey-Wagner I, Ortiz A, Unwin R, Liabeuf S, Suzumoto Y, Iervolino A, Stasi A, Di Marzo V, Gesualdo L, Massy ZA. The role of the intestinal microbiome in cognitive decline in patients with kidney disease. Nephrol Dial Transplant 2025; 40:ii4-ii17. [PMID: 40080091 PMCID: PMC11905753 DOI: 10.1093/ndt/gfae253] [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: 08/15/2024] [Indexed: 03/15/2025] Open
Abstract
Cognitive decline is frequently seen in patients with chronic kidney disease (CKD). The causes of cognitive decline in these patients are likely to be multifactorial, including vascular disease, uraemic toxins, blood-brain barrier leakage, and metabolic and endocrine changes. Gut dysbiosis is common in patients with CKD and contributes to the increase in uraemic toxins. However, the gut microbiome modulates local and systemic levels of several metabolites such as short-chain fatty acids or derivatives of tryptophan metabolism, neurotransmitters, endocannabinoid-like mediators, bile acids, hormones such as glucagon-like peptide 1 (GLP1) or cholecystokinin (CCK). These factors can affect gut function, immunity, autonomic nervous system activity and various aspects of brain function. Key areas include blood-brain barrier integrity, nerve myelination and survival/proliferation, appetite, metabolism and thermoregulation, mood, anxiety and depression, stress and local inflammation. Alterations in the composition of the gut microbiota and the production of biologically active metabolites in patients with CKD are well documented and are favoured by low-fiber diets, elevated urea levels, sedentary lifestyles, slow stool transit times and polypharmacy. In turn, dysbiosis can modulate brain function and cognitive processes, as discussed in this review. Thus, the gut microbiome may contribute to alterations in cognition in patients with CKD and may be a target for therapeutic interventions using diet, prebiotics and probiotics.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology and Zurich Kidney Center, University of Zurich, Switzerland
| | | | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, RICORS2040, Madrid, Spain
| | - Robert Unwin
- Department of Renal Medicine, University College London, London, UK
| | - Sophie Liabeuf
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens-Picardie University Medical Center, Amiens, France
- MP3CV Laboratory, Jules Verne University of Picardie, Amiens, France
| | - Yoko Suzumoto
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| | - Anna Iervolino
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- University of Campania “L. Vanvitelli”, Naples, Italy
| | - Alessandra Stasi
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Di Marzo
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, CRIUCPQ and INAF, Centre NUTRISS, Faculties of Medicine and Agriculture and Food Sciences, Université Laval, Québec City, Canada
- Joint International Research Unit for Chemical and Biomolecular Research on the Microbiome and its impact on Metabolic Health and Nutrition (JIRU-MicroMeNu) between Université Laval Québec, Canada and Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Loreto Gesualdo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Ziad A Massy
- INSERM Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Saclay University and Versailles Saint-Quentin-en-Yvelines University (UVSQ), Villejuif, France
- Association pour l'Utilisation du Rein Artificiel dans la région parisienne (AURA) Paris, France and Ambroise Paré University Hospital, APHP, Department of Nephrology Boulogne-Billancourt, Paris, France
| |
Collapse
|
12
|
Yu Y, Wang H, Jin X, Huang W, Zhao Y, Wang N, Lu D, Wei B, Wang H. Structural Characterization of Dendrobium officinale Polysaccharides and Their Regulation Effect on Intestinal Microbiota During In Vitro Fermentation. Polymers (Basel) 2025; 17:727. [PMID: 40292547 PMCID: PMC11944827 DOI: 10.3390/polym17060727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 04/30/2025] Open
Abstract
Polysaccharides derived from Dendrobium officinale have been demonstrated to exhibit metabolic regulatory properties. However, the correlation between their structure and function, particularly their mechanism of action through gut microbiota, remains underexplored. This study systematically elucidates the structural characteristics of Dendrobium officinale polysaccharide (DOP) from the Guizhou (GZ) and Zhejiang (ZJ) provinces of China using nuclear magnetic resonance (NMR) and a series of chromatographic analyses, revealing their unique molecular features. Additionally, the metabolic regulatory activities were assessed through α-glucosidase inhibitory assay and in vitro intestinal flora activity assay. The findings include the following: (1) both DOP-GZ and DOP-ZJ predominantly consist of glycosidic linkages of β-1,4-Manp and β-1,4-Glcp; (2) zhe monosaccharide composition ratios of mannose to glucose are 2.51:1 for DOP-GZ and 2.66:1 for DOP-ZJ, with molecular weights of 356 kDa and 544 kDa, respectively, indicating significant structural differences between DOPs from different sources; (3) treatment with DOP-GZ and DOP-ZJ led to alterations in the α-diversity indices and Firmicutes-to-Bacteroidota ratios; (4) more importantly, DOP-GZ and DOP-ZJ significantly increase the abundance of beneficial bacteria (e.g., g_Proteobacteria_unclassified) while suppressing the growth of pathogenic bacteria (e.g., f_Enterobacteriaceae_unclassified), with statistically significant results. These findings not only uncover a novel mechanism by which DOPs regulate metabolism through gut microbiota but also provide a crucial theoretical basis for the application of DOPs in functional foods and pharmaceutical development.
Collapse
Affiliation(s)
- Yanlei Yu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Honggang Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Xiaoshu Jin
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Wenjing Huang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Yunjie Zhao
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Ningning Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Dongze Lu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
- Binjiang Cyberspace Security Institute of ZJUT, Hangzhou 310056, China
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Y.Y.); (B.W.)
- Binjiang Cyberspace Security Institute of ZJUT, Hangzhou 310056, China
| |
Collapse
|
13
|
Li F, Armet AM, Korpela K, Liu J, Quevedo RM, Asnicar F, Seethaler B, Rusnak TBS, Cole JL, Zhang Z, Zhao S, Wang X, Gagnon A, Deehan EC, Mota JF, Bakal JA, Greiner R, Knights D, Segata N, Bischoff SC, Mereu L, Haqq AM, Field CJ, Li L, Prado CM, Walter J. Cardiometabolic benefits of a non-industrialized-type diet are linked to gut microbiome modulation. Cell 2025; 188:1226-1247.e18. [PMID: 39855197 DOI: 10.1016/j.cell.2024.12.034] [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: 05/29/2024] [Revised: 10/24/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Industrialization adversely affects the gut microbiome and predisposes individuals to chronic non-communicable diseases. We tested a microbiome restoration strategy comprising a diet that recapitulated key characteristics of non-industrialized dietary patterns (restore diet) and a bacterium rarely found in industrialized microbiomes (Limosilactobacillus reuteri) in a randomized controlled feeding trial in healthy Canadian adults. The restore diet, despite reducing gut microbiome diversity, enhanced the persistence of L. reuteri strain from rural Papua New Guinea (PB-W1) and redressed several microbiome features altered by industrialization. The diet also beneficially altered microbiota-derived plasma metabolites implicated in the etiology of chronic non-communicable diseases. Considerable cardiometabolic benefits were observed independently of L. reuteri administration, several of which could be accurately predicted by baseline and diet-responsive microbiome features. The findings suggest that a dietary intervention targeted toward restoring the gut microbiome can improve host-microbiome interactions that likely underpin chronic pathologies, which can guide dietary recommendations and the development of therapeutic and nutritional strategies.
Collapse
Affiliation(s)
- Fuyong Li
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Animal Science and Technology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Anissa M Armet
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Katri Korpela
- Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Uusimaa, Finland
| | - Junhong Liu
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Rodrigo Margain Quevedo
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Francesco Asnicar
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento 38123, Trentino, Italy
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Baden-Württemberg, Germany
| | - Tianna B S Rusnak
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Janis L Cole
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Zhihong Zhang
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Shuang Zhao
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada
| | - Xiaohang Wang
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada
| | - Adele Gagnon
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Edward C Deehan
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588, USA
| | - João F Mota
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Munster, Ireland; Faculty of Nutrition, Federal University of Goiás, Goiânia, Goiás 74605-080, Brazil
| | - Jeffrey A Bakal
- Division of General Internal Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Russell Greiner
- Department of Computing Science, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Machine Intelligence Institute, Edmonton, AB T5J 3B1, Canada
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento 38123, Trentino, Italy
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Baden-Württemberg, Germany
| | - Laurie Mereu
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Andrea M Haqq
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Catherine J Field
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Liang Li
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada; Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Carla M Prado
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jens Walter
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; APC Microbiome Ireland, University College Cork, Cork T12 YT20, Munster, Ireland; School of Microbiology, University College Cork, Cork T12 YT20, Munster, Ireland; Department of Medicine, University College Cork, Cork T12 YT20, Munster, Ireland; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
14
|
Safarchi A, Al-Qadami G, Tran CD, Conlon M. Understanding dysbiosis and resilience in the human gut microbiome: biomarkers, interventions, and challenges. Front Microbiol 2025; 16:1559521. [PMID: 40104586 PMCID: PMC11913848 DOI: 10.3389/fmicb.2025.1559521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
The healthy gut microbiome is important in maintaining health and preventing various chronic and metabolic diseases through interactions with the host via different gut-organ axes, such as the gut-brain, gut-liver, gut-immune, and gut-lung axes. The human gut microbiome is relatively stable, yet can be influenced by numerous factors, such as diet, infections, chronic diseases, and medications which may disrupt its composition and function. Therefore, microbial resilience is suggested as one of the key characteristics of a healthy gut microbiome in humans. However, our understanding of its definition and indicators remains unclear due to insufficient experimental data. Here, we review the impact of key drivers including intrinsic and extrinsic factors such as diet and antibiotics on the human gut microbiome. Additionally, we discuss the concept of a resilient gut microbiome and highlight potential biomarkers including diversity indices and some bacterial taxa as recovery-associated bacteria, resistance genes, antimicrobial peptides, and functional flexibility. These biomarkers can facilitate the identification and prediction of healthy and resilient microbiomes, particularly in precision medicine, through diagnostic tools or machine learning approaches especially after antimicrobial medications that may cause stable dysbiosis. Furthermore, we review current nutrition intervention strategies to maximize microbial resilience, the challenges in investigating microbiome resilience, and future directions in this field of research.
Collapse
Affiliation(s)
- Azadeh Safarchi
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Ghanyah Al-Qadami
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Cuong D Tran
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Michael Conlon
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| |
Collapse
|
15
|
Katare PB, Tingstad RH, Beajani ST, Indseth JP, Telle-Hansen VH, Myhrstad MCW, Rustan AC, Eide L, Witczak O, Aas V. Divergent effects of monomethyl branched-chain fatty acids on energy metabolism and insulin signaling in human myotubes. J Lipid Res 2025; 66:100764. [PMID: 40010610 PMCID: PMC11982973 DOI: 10.1016/j.jlr.2025.100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025] Open
Abstract
Branched-chain fatty acids (BCFAs) are predominantly saturated fatty acids with one or more methyl branches on the carbon chain, typically found in dairy products and measured in micromolar concentrations in human plasma. The biological function of BCFAs in humans remains ill-defined, but a relationship between circulating BCFAs and cardiometabolic health has been suggested. The objective of this study was to evaluate the impact of BCFAs on energy metabolism in human myotubes. The results revealed distinct effects of BCFAs. 12-Methyltetradecanoic acid (12-MTD) increased glucose uptake and glycogen synthesis, while 13-methyltetradecanoic acid (13-MTD), 14-methylhexadecanoic acid (14-MHD), and 15-methylhexadecanoic acid (15-MHD) increased oleic acid uptake and 13-MTD and 15-MHD oleic acid oxidation, indicating a more general stimulatory effect on fatty acid than glucose metabolism. Interestingly, the same BCFAs, 13-MTD, 14-MHD, and 15-MHD, appeared to reduce insulin-stimulated glycogen synthesis. Insulin-stimulated phosphorylation of IRS1 was not apparent after exposure to 12-MTD, 13-MTD, and 15-MHD, whereas insulin-stimulated phosphorylation of Akt was unchanged by BCFAs. Incorporation of [14C]leucine into lipids was affected, as 13-MTD increased the total lipid content, and 12-MTD altered the distribution of lipid classes. Metabolic flux analysis indicated that 14-MHD stimulated extracellular acidification. The effects of BCFAs might involve increased mRNA expression of pyruvate dehydrogenase kinase 4. In conclusion, the study demonstrates that different BCFAs have distinct effects on energy metabolism in myotubes, 12-MTD mainly affect glucose metabolism, while 13-MTD, 14-MHD, and 15-MHD modulated oleic acid metabolism. These data suggest that some BCFAs might have therapeutic applications by improving energy metabolism.
Collapse
Affiliation(s)
- Parmeshwar Bajirao Katare
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Ragna H Tingstad
- Department of Life Sciences and Health, Oslo Metropolitan University - OsloMet, Oslo, Norway
| | - Sivar T Beajani
- Department of Life Sciences and Health, Oslo Metropolitan University - OsloMet, Oslo, Norway
| | | | - Vibeke H Telle-Hansen
- Department of Nursing and Health Promotion, Oslo Metropolitan University - OsloMet, Oslo, Norway
| | - Mari C W Myhrstad
- Department of Nursing and Health Promotion, Oslo Metropolitan University - OsloMet, Oslo, Norway
| | - Arild C Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Lars Eide
- Department of Medical Biochemistry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Oliwia Witczak
- Department of Life Sciences and Health, Oslo Metropolitan University - OsloMet, Oslo, Norway
| | - Vigdis Aas
- Department of Life Sciences and Health, Oslo Metropolitan University - OsloMet, Oslo, Norway.
| |
Collapse
|
16
|
JanssenDuijghuijsen L, Fransen K, Deng R, Perenboom C, de Wit N, Hooiveld G, van Trijp M. How to Study the Effects of Dietary Lipids on the Small Intestinal Microbiome? Methodological Design and Evaluation of the Human HealThy fAt, haPpy mIcRobiome (TAPIR) Proof-of-Concept Study. Curr Dev Nutr 2025; 9:104564. [PMID: 40092654 PMCID: PMC11908603 DOI: 10.1016/j.cdnut.2025.104564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 03/19/2025] Open
Abstract
Background Emerging evidence highlights the importance of the small intestinal microbiota in digestion and metabolism, underscoring the challenging need for human studies beyond fecal analyses. Objective The TAPIR (acronym of "healthy fat, happy microbiome") proof-of-concept study was primarily designed to confirm the interaction between the small intestinal microbiota and dietary lipids in healthy adults with a challenge test. We also aimed to assess the impact of a plant-based mild-ketogenic preconditioning diet on microbiome composition and function. Here, we comprehensively describe our extensive study protocol and evaluate the study execution. Methods Participants consumed an 8-day preconditioning diet, followed by a high-fat shake challenge test on day 9. During this test, fasting and postprandial small intestinal aspirates were collected every 20 min via a naso-intestinal catheter, and blood samples were collected hourly. Participants ingested small intestine aspiration capsules before (day 0), on day 6 of the preconditioning diet, and during the challenge test. Dietary compliance, capsule retrieval, sample collection, stool pattern, and gastrointestinal complaints were monitored to evaluate study execution. Results Twenty adults with a mean age of 48 y (19-88 y) and a mean body mass index (BMI) of 24.3 kg/m2 (19.5-30 kg/m2) consumed a preconditioning diet with a 96% compliance. There were no significant changes in gastrointestinal complaints and stool patterns during the study. Mean aspiration capsule retrieval rate was 94.7%, with mean sample weights per timepoint between 84.2 and 95.4 mg and median transit times between 32.8 and 49.3 h. The average success rate of aspirate collection by catheter was 49%, varying significantly between time points. Conclusion The dietary intervention was successful and well-tolerated. We sampled in the small intestine with capsules and catheters, each with its own (dis)advantages. The comprehensive description and evaluation of our study execution offer practical insights supporting future study designs in food-microbe interactions in the small intestine.The trial is registered at clinicaltrials.gov as NCT06064266.
Collapse
Affiliation(s)
| | - Karen Fransen
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| | - Ruolei Deng
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| | - Corine Perenboom
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| | - Nicole de Wit
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, the Netherlands
| | - Guido Hooiveld
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| | - Mara van Trijp
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| |
Collapse
|
17
|
Sievenpiper JL, Purkayastha S, Grotz VL, Mora M, Zhou J, Hennings K, Goody CM, Germana K. Dietary Guidance, Sensory, Health and Safety Considerations When Choosing Low and No-Calorie Sweeteners. Nutrients 2025; 17:793. [PMID: 40077663 PMCID: PMC11902030 DOI: 10.3390/nu17050793] [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: 12/17/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 03/14/2025] Open
Abstract
The growing global focus on the adverse health conditions associated with excessive sugar consumption has prompted health and policy organizations as well as the public to take a more mindful approach to health and wellness. In response, food and beverage companies have proactively innovated and reformulated their product portfolios to incorporate low and no-calorie sweeteners (LNCSs) as viable alternatives to sugar. LNCSs offer an effective and safe approach to delivering sweetness to foods and beverages and reducing calories and sugar intake while contributing to the enjoyment of eating. The objective of this paper is to enhance the understanding of LNCSs segmentation and definitions, dietary consumption and reduction guidance, front-of-package labeling, taste and sensory perception and physiology, metabolic efficacy and impact, as well as the overall safety of LNCSs and sugar.
Collapse
Affiliation(s)
- John L. Sievenpiper
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada;
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
| | - Sidd Purkayastha
- SP Advisors Inc., Chicago, IL 60605, USA;
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - V. Lee Grotz
- ToxInsight, LLC, Fort Washington, PA 19034, USA;
| | - Margaux Mora
- Ingredion Inc., Bridgewater, NJ 08807, USA; (M.M.); (K.G.)
| | - Jing Zhou
- Ingredion Inc., Bridgewater, NJ 08807, USA; (M.M.); (K.G.)
| | | | | | | |
Collapse
|
18
|
Höyhtyä M, Haaramo A, Nikkonen A, Ventin-Holmberg R, Agrawal N, Ritari J, Hickman B, Partanen J, Alapulli H, Tuokkola J, Salonen A, de Vos WM, Kolho KL. Fecal microbiota and genetics in pediatric-onset orofacial granulomatosis and Crohn´s disease. Sci Rep 2025; 15:6020. [PMID: 39972069 PMCID: PMC11839994 DOI: 10.1038/s41598-025-90243-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
Orofacial granulomatosis (OFG) is a rare chronic inflammatory condition. It is under debate, whether it is a condition of its own or merely a subtype of Crohn's disease (CD). We aimed to search for markers characteristic of patients with pediatric-onset OFG compared to patients with pediatric-onset CD. We recruited young patients with OFG (with or without CD, n = 29), CD (n = 24), and healthy controls (n = 20). All participants provided a fecal sample for microbiota and calprotectin analyses and saliva for DNA analysis of genes associated with OFG and kept a 3-day food diary. Oral disease activity was evaluated using The Oral Disease Activity Score by an otorhinolaryngologist and a dentist. We observed decreased relative abundance in class Clostridia and increased relative abundances of classes Actinobacteria and Bacilli in the feces of patients with OFG when compared to patients with CD and healthy controls. The relative abundances of Bifidobacterium adolescentis increased and Faecalibacterium prausnitzii decreased along with the increase in the Oral Disease Activity Score. We found the NOD2 gene rs8057341 allele A to be enriched in patients with OFG compared to patients with CD. These findings support the theory that OFG is a distinct disease phenotype.
Collapse
Affiliation(s)
- Miikka Höyhtyä
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu Haaramo
- Department of Otorhinolaryngology, Head and Neck Surgery, Helsinki University Hospital HUS and University of Helsinki, Helsinki, Finland
| | - Anne Nikkonen
- Children´S Hospital, University of Helsinki and HUS, Helsinki, Finland
| | - Rebecka Ventin-Holmberg
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Nitin Agrawal
- Fin-HIT Research Group, Folkhälsan Research Center, Department of Public Health, Helsinki, Finland
| | - Jarmo Ritari
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - Brandon Hickman
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Heikki Alapulli
- Department of Oral and Maxillofacial Diseases, Faculty of Medicine, University of Helsinki and Helsinki University Hospital HUS, Helsinki, Finland
| | - Jetta Tuokkola
- Clinical Nutrition Unit, Internal Medicine and Rehabilitation, Institute of Public Health, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Clinical Nutrition, Kuopio and Department of Medicine, Endocrinology and Clinical Nutrition, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands
| | - Kaija-Leena Kolho
- Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Children´S Hospital, University of Helsinki and HUS, Helsinki, Finland.
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
19
|
Castonguay-Paradis S, Parent L, St-Arnaud G, Perron J, Dumais É, Flamand N, Raymond F, Di Marzo V, Veilleux A. The Human Fecal Endocannabinoidome Mediator Profile Is Mainly Defined by the Fecal Microbiota and Diet. J Clin Endocrinol Metab 2025; 110:739-747. [PMID: 39171571 PMCID: PMC11834705 DOI: 10.1210/clinem/dgae586] [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: 05/07/2024] [Revised: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
CONTEXT The endocannabinoid system and its extension, the endocannabinoidome (eCBome), are involved in numerous biological processes, notably energy homeostasis, across virtually all tissues. While the circulating eCBome mediator profile is associated with dietary intakes and metabolic status, an important knowledge gap resides in the identification of the precise determinants of these mediators in the gut lumen. OBJECTIVE We aimed at establishing the profile of eCBome mediators in human feces and investigating their association with circulating eCBome mediators, dietary intakes, metabolic status, and gut microbiota composition. METHODS N-acyl-ethanolamines (NAEs) and 2-monoacyl-glycerols (2-MAGs) were profiled by liquid chromatography coupled to tandem mass spectrometry in plasma and feces of a cross-sectional cohort (n = 195) and a short-term dietary intervention trial (n = 21) with comprehensive dietary intakes and gut microbiota measures. RESULTS Six NAEs and 7 2-MAGs were identified in fecal samples, but some, especially omega-3-derived mediators, were undetectable in the majority of samples. Fecal NAEs, and to a lower extent 2-MAGs, were positively albeit weakly correlated with the circulating levels of eCBome mediators. Fecal 2-arachidonoyl-glycerol, N-palmitoyl-ethanolamine, and N-docosahexaenoyl-ethanolamine levels were positively associated with visceral adiposity and with some parameters of the metabolic profile. Dietary intakes of foods rich in fibers were associated with lower fecal levels of several eCBome mediators, while intakes of unsaturated fatty acids were associated with fecal 2-oleoyl-glycerol and 2-linoleoyl-glycerol. Interestingly, gut microbiota diversity and composition were a strong correlate of the fecal eCBome profile. CONCLUSION The fecal eCBome profile is associated with gut microbiota composition and dietary intakes, more than with the circulating profile. These results strengthen the hypothesis of an interrelation between the gut microbiome and eCBome signaling involved in the regulation of numerous host biological processes.
Collapse
Affiliation(s)
- Sophie Castonguay-Paradis
- Centre Nutrition, Santé et Société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, QC, Canada, G1V 0A6
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada, G1V 0A6
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC, Canada, G1V 0A6
| | - Lydiane Parent
- Centre Nutrition, Santé et Société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, QC, Canada, G1V 0A6
| | - Gabrielle St-Arnaud
- Centre Nutrition, Santé et Société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, QC, Canada, G1V 0A6
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada, G1V 0A6
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC, Canada, G1V 0A6
| | - Julie Perron
- Centre Nutrition, Santé et Société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, QC, Canada, G1V 0A6
| | - Élizabeth Dumais
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC, Canada, G1V 0A6
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Université Laval, Québec, QC, Canada, G1V 4G5
| | - Nicolas Flamand
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC, Canada, G1V 0A6
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Université Laval, Québec, QC, Canada, G1V 4G5
- Département de médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada, G1V 0A6
| | - Frédéric Raymond
- Centre Nutrition, Santé et Société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, QC, Canada, G1V 0A6
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada, G1V 0A6
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC, Canada, G1V 0A6
| | - Vincenzo Di Marzo
- Centre Nutrition, Santé et Société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, QC, Canada, G1V 0A6
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada, G1V 0A6
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC, Canada, G1V 0A6
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Université Laval, Québec, QC, Canada, G1V 4G5
- Département de médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada, G1V 0A6
- Joint International Unit on Chemical and Biomolecular Research on the Microbiome and its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Québec, QC, Canada, G1V 0A6
| | - Alain Veilleux
- Centre Nutrition, Santé et Société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec, QC, Canada, G1V 0A6
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada, G1V 0A6
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC, Canada, G1V 0A6
| |
Collapse
|
20
|
Liu J, Huang S. Dietary index for gut microbiota is associated with stroke among US adults. Food Funct 2025; 16:1458-1468. [PMID: 39898733 DOI: 10.1039/d4fo04649h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Aims: Emerging evidence underscores the diet-microbiota-gut-brain axis as vital to brain health. The dietary index for gut microbiota (DI-GM), quantifying diet quality linked to gut microbiota diversity, reflects healthier gut microbiota with higher scores. Therefore, this study was designed to explore the unclear association between DI-GM and stroke. Methods: A cross-sectional analysis was conducted using data from 48 677 participants aged ≥20 years in the National Health and Nutrition Examination Survey (NHANES). Demographic and dietary data were collected, and multivariable weighted logistic regression analysis was performed to evaluate the association between the DI-GM and stroke. Additionally, restricted cubic spline (RCS), subgroup analyses, and receiver operating characteristic (ROC) curve were conducted. Results: In participants aged ≥20 years, the odds ratio (OR) was 0.96 (95% CI: 0.92-1.00, P = 0.075) in the crude model, but after adjustment, the OR was 0.93 (95% CI: 0.89-0.98, P = 0.003), while higher beneficial to gut microbiota scores were consistently associated with lower stroke prevalence with ORs of 0.87 (95% CI: 0.83-0.90, P < 0.001) in the crude model and 0.88 (95% CI: 0.83-0.93, P < 0.001) after adjustment. Among participants aged 20-29 years, no significant association was observed. For those aged ≥30 years, higher DI-GM and beneficial to gut microbiota scores were associated with lower stroke prevalence, with DI-GM showing ORs of 0.93 (95% CI: 0.89-0.97, P < 0.001) in the crude model and 0.93 (95% CI: 0.89-0.98, P = 0.003) after adjustment, and beneficial to gut microbiota scores showing ORs of 0.82 (95% CI: 0.79-0.86, P < 0.001) in the crude model and 0.88 (95% CI: 0.83-0.93, P < 0.001) after adjustment. RCS indicated a linear relationship between DI-GM and stroke. Conclusion: The DI-GM was inversely and linearly associated with stroke prevalence, particularly in adults aged 30 years and above.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China.
| | - Shaoqiang Huang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China.
| |
Collapse
|
21
|
Senthakumaran T, Tannæs TM, Moen AEF, Brackmann SA, Jahanlu D, Rounge TB, Bemanian V, Tunsjø HS. Detection of colorectal-cancer-associated bacterial taxa in fecal samples using next-generation sequencing and 19 newly established qPCR assays. Mol Oncol 2025; 19:412-429. [PMID: 38970464 PMCID: PMC11793011 DOI: 10.1002/1878-0261.13700] [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: 12/12/2023] [Revised: 05/15/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024] Open
Abstract
We have previously identified increased levels of distinct bacterial taxa within mucosal biopsies from colorectal cancer (CRC) patients. Following prior research, the aim of this study was to investigate the detection of the same CRC-associated bacteria in fecal samples and to evaluate the suitability of fecal samples as a non-invasive material for the detection of CRC-associated bacteria. Next-generation sequencing (NGS) of the 16S ribosomal RNA (rRNA) V4 region was performed to evaluate the detection of the CRC-associated bacteria in the fecal microbiota of cancer patients, patients with adenomatous polyp and healthy controls. Furthermore, 19 novel species-specific quantitative PCR (qPCR) assays were established to detect the CRC-associated bacteria. Approximately, 75% of the bacterial taxa identified in biopsies were reflected in fecal samples. NGS failed to detect low-abundance CRC-associated taxa in fecal samples, whereas qPCR exhibited high sensitivity and specificity in identifying all targeted taxa. Comparison of fecal microbial composition between the different patient groups showed enrichment of Fusobacterium nucleatum, Parvimonas micra, and Gemella morbillorum in cancer patients. Our findings suggest that low-abundance mucosa-associated bacteria can be detected in fecal samples using sensitive qPCR assays.
Collapse
Affiliation(s)
| | - Tone M. Tannæs
- Section for Clinical Molecular Biology (EpiGen)Akershus University HospitalLørenskogNorway
- Department of Clinical Molecular Biology, Institute of Clinical MedicineUniversity of OsloNorway
| | - Aina E. F. Moen
- Section for Clinical Molecular Biology (EpiGen)Akershus University HospitalLørenskogNorway
- Department of Clinical Molecular Biology, Institute of Clinical MedicineUniversity of OsloNorway
- Department of Methods Development and AnalyticsNorwegian Institute of Public HealthOsloNorway
| | - Stephan A. Brackmann
- Department of Gastroenterology, Division of MedicineAkershus University HospitalLørenskogNorway
- Institute for Clinical MedicineUniversity of OsloNorway
| | - David Jahanlu
- Department of Life Sciences and HealthOslo Metropolitan UniversityNorway
| | - Trine B. Rounge
- Department of Pharmacy, Centre for BioinformaticsUniversity of OsloNorway
- Department of ResearchCancer Registry of NorwayOsloNorway
| | - Vahid Bemanian
- Department of PathologyAkershus University HospitalLørenskogNorway
| | - Hege S. Tunsjø
- Department of Life Sciences and HealthOslo Metropolitan UniversityNorway
| |
Collapse
|
22
|
Lee DB, Hwang IS. Macronutrient balance determines the human gut microbiome eubiosis: insights from in vitro gastrointestinal digestion and fermentation of eight pulse species. Front Microbiol 2025; 15:1512217. [PMID: 39949350 PMCID: PMC11823474 DOI: 10.3389/fmicb.2024.1512217] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/26/2024] [Indexed: 02/16/2025] Open
Abstract
The interactions between macronutrients, the human gut microbiome, and their metabolites (short-chain fatty acids) were comprehensively investigated via an in vitro digestion and fermentation model subjected to eight pulse species. 16S rRNA sequencing and taxonomic analysis of pulse digesta fermented for up to 24 h revealed an increase in the relative abundance of gut health-detrimental genera represented by Escherichia-Shigella in kidney bean, soybean, cowpea, chickpea, and black bean samples. In contrast, the relative abundance of health-positive genera, including Bacteroides, Eubacterium, and Akkermansia, was elevated in red bean, mung bean, and Heunguseul. At the same time, the proportion of the pathogenic Escherichia-Shigella decreased. Concurrently, these three species exhibited an increase in microbial diversity as evidenced by the calculation of α-diversity (Shannon index) and β-diversity (Bray-Curtis distance). Despite the lower nutrient contents in the three pulses, represented by carbohydrates, amino acids, and fatty acids, network analysis revealed that the nutrient contents in the pulse digesta possess complex positive or negative correlations with a variety of bacteria, as well as their metabolites. These correlations were more pronounced in red bean, mung bean, and Heunguseul than in the other pulses. It was postulated that the overall potential to nourish gut environments in these species was due to the balance of their nutritional components. The linear regression analysis demonstrated that there was a negative association between carbohydrate and amino acid contents and the increase in Shannon indices. Furthermore, the ratio of carbohydrates to fatty acids and amino acids to fatty acids displayed negative correlations with the diversity increase. The ratio of carbohydrates to amino acids showed a weak positive correlation. It is noteworthy that a diet comprising foods with a balanced nutritional profile supports the growth of beneficial gut microbes, thereby promoting microbial eubiosis. Consistent work on different ingredients is essential for precise insight into the interplay between food and the human microbiome in complex dietary patterns.
Collapse
Affiliation(s)
| | - In Seon Hwang
- Food and Nutrition Division, Department of Agri-food Resources, National Institute of Agricultural Sciences, Wanju, Republic of Korea
| |
Collapse
|
23
|
Bizzozero-Peroni B, Martínez-Vizcaíno V, Fernández-Rodríguez R, Jiménez-López E, Núñez de Arenas-Arroyo S, Saz-Lara A, Díaz-Goñi V, Mesas AE. The impact of the Mediterranean diet on alleviating depressive symptoms in adults: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 2025; 83:29-39. [PMID: 38219230 DOI: 10.1093/nutrit/nuad176] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
CONTEXT High adherence to the Mediterranean diet (MD) has been associated with a reduced risk of depression in prospective cohort studies, but whether MD interventions are effective among adults with depression is uncertain. OBJECTIVE This study aimed to synthesize findings on the effects of MD interventions on the severity of depressive symptoms in adults with depression. DATA SOURCES PubMed, Cochrane CENTRAL, PsycINFO, Scopus, and Web of Science were systematically searched from database inception to March 2023. The Preferred Reporting Items for Systematic Review and Meta-Analyses guidelines and the Cochrane recommendations were followed. We included randomized controlled trials (RCTs) comparing outcomes after MD interventions with outcomes for control conditions in adults with depressive disorders or depressive symptoms. DATA EXTRACTION Two authors extracted the data independently. The Sidik-Jonkman estimator, the I2 metric, and the prediction interval were used to estimate between-study heterogeneity. To determine the risk of bias and the certainty of evidence from RCTs, we used the Cochrane Collaboration's Risk of Bias 2 and Grades of Recommendation, Assessment, Development, and Evaluation tools, respectively. DATA ANALYSIS In total, 1507 participants (mean age range: 22.0 years-53.3 years) with depression were initially included in the 5 RCTs of this review. Compared with control conditions, MD interventions significantly reduced depressive symptoms among young and middle-aged adults with major depression or mild to moderate depressive symptoms (standardized mean difference: -0.53; 95% confidence interval: -0.90 to -0.16; I2 = 87.1%). The prediction interval ranged from -1.86 to 0.81. The overall risk of bias was within the range of "some concerns" to "high," while the certainty of evidence was low. CONCLUSION MD interventions appear to have substantial potential for alleviating depressive symptoms in people experiencing major or mild depression. However, to establish robust recommendations, there remains a need for high-quality, large-scale, and long-term RCTs. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42022341895.
Collapse
Affiliation(s)
- Bruno Bizzozero-Peroni
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Department of Physical Education and Health, Higher Institute of Physical Education, Universidad de la República, Rivera, Uruguay
| | - Vicente Martínez-Vizcaíno
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Grupo de Investigación en Educación Física, Salud y Calidad de Vida, Facultad de Educación, Universidad Autónoma de Chile, Temuco, Chile
| | | | - Estela Jiménez-López
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Department of Psychiatry, Hospital Virgen de La Luz, Cuenca, Spain
- Network Centre for Biomedical Research in Mental Health (CIBERSAM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Alicia Saz-Lara
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| | - Valentina Díaz-Goñi
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| | - Arthur Eumann Mesas
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| |
Collapse
|
24
|
Reis LG, Teeple K, Schoonmaker JL, Davis C, Scinto S, Schinckel A, Casey T. Constant light and high fat diet alter daily patterns of activity, feed intake and fecal corticosterone levels in pregnant and lactating female ICR mice. PLoS One 2024; 19:e0312419. [PMID: 39565751 PMCID: PMC11578523 DOI: 10.1371/journal.pone.0312419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/05/2024] [Indexed: 11/22/2024] Open
Abstract
The prevalence of constant light exposure and high-fat diet in modern society raises concerns regarding their impact on maternal and offspring health outcomes. In rodents, exposure to maternal high-fat diet or continuous light negatively program metabolic and stress response outcomes of offspring. A 2x3 factorial study was conducted to investigate the impact of diet (control-CON, 10% fat, or high fat-HF, 60% fat) and exposure to different lighting conditions: regular 12-hour light-dark cycles (LD), continuous dim light (L5), or continuous bright light (L100) on female ICR mice daily patterns of time in and out of the nest, feed intake, and fecal corticosterone levels during gestation and lactation. Our previous analysis of these mice found HF diet decreased number of pups born, but increased litter growth rate to postnatal (PN) d12. Whereas continuous light increased gestation length and tended to increase PN litter growth. Here we report that patterns of grams of feed intake, an indicator of feeding activity, were affected by light, diet, period of the day (day versus night) and physiological state (gestation and lactation), with significant interactions among all these variables (P<0.05). HF diet and light treatment increased fecal corticosterone output (P<0.05) during lactation. Dams exhibited significant 12 h and 24 h rhythms of activity out of the nest in the first 48 h postnatal, with time outside of the nest greater in the second 24 h period. L100 treatment and HF diet attenuated rhythms and shifted phase of rhythms relative to LD and CON, respectively (P<0.05). Alterations in behavior affect maternal physiology, including level and timing of release of corticosteroids. Elevated fecal corticosterone levels due to high-fat diet and continuous light may have potential implications on maternal-offspring health, and potentially underlie some of the adverse effects of modern lifestyle factors on maternal and offspring health.
Collapse
Affiliation(s)
- Leriana Garcia Reis
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Kelsey Teeple
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Jenna Lynn Schoonmaker
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Corrin Davis
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Sara Scinto
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Allan Schinckel
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Theresa Casey
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| |
Collapse
|
25
|
Pearce CS, Bukovsky D, Douchant K, Katoch A, Greenlaw J, Gale DJ, Nashed JY, Brien D, Kuhlmeier VA, Sabbagh MA, Blohm G, De Felice FG, Pare M, Cook DJ, Scott SH, Munoz DP, Sjaarda CP, Tusche A, Sheth PM, Winterborn A, Boehnke S, Gallivan JP. Changes in social environment impact primate gut microbiota composition. Anim Microbiome 2024; 6:66. [PMID: 39538341 PMCID: PMC11562706 DOI: 10.1186/s42523-024-00355-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The gut microbiota (GM) has proven to be essential for both physical health and mental wellbeing, yet the forces that ultimately shape its composition remain opaque. One critical force known to affect the GM is the social environment. Prior work in humans and free-ranging non-human primates has shown that cohabitation and frequent social interaction can lead to changes in GM composition. However, it is difficult to assess the direction of causation in these studies, and interpretations are complicated by the influence of uncontrolled but correlated factors, such as shared diet. RESULTS We performed a 15-month longitudinal investigation wherein we disentangled the impacts of diet and social living conditions on GM composition in a captive cohort of 13 male cynomolgus macaques. The animals were in single housing for the first 3 months of the study initially with a variable diet. After baseline data collection they were placed on a controlled diet for the remainder of the study. Following this diet shift the animals were moved to paired housing for 6 months, enabling enhanced social interaction, and then subsequently returned to single housing at the end of our study. This structured sequencing of diet and housing changes allowed us to assess their distinct impacts on GM composition. We found that the early dietary adjustments led to GM changes in both alpha and beta diversity, whereas changes in social living conditions only altered beta diversity. With respect to the latter, we found that two particular bacterial families - Lactobacillaceae and Clostridiaceae - demonstrated significant shifts in abundance during the transition from single housing to paired housing, which was distinct from the shifts we observed based on a change in diet. Conversely, we found that other bacteria previously associated with sociality were not altered based on changes in social living conditions but rather only by changes in diet. CONCLUSIONS Together, these findings decouple the influences that diet and social living have on GM composition and reconcile previous observations in the human and animal literatures. Moreover, the results indicate biological alterations of the gut that may, in part, mediate the relationship between sociality and wellbeing.
Collapse
Affiliation(s)
- Colleen S Pearce
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | | | - Katya Douchant
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Abhay Katoch
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Biology, Queen's University, Kingston, ON, Canada
| | - Jill Greenlaw
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Daniel J Gale
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Joseph Y Nashed
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Don Brien
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Valerie A Kuhlmeier
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Mark A Sabbagh
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Gunnar Blohm
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Fernanda G De Felice
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Martin Pare
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Douglas J Cook
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Neurosurgery, Queen's University, Kingston, ON, Canada
| | - Stephen H Scott
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Calvin P Sjaarda
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Anita Tusche
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Prameet M Sheth
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Andrew Winterborn
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Susan Boehnke
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Jason P Gallivan
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
- Department of Psychology, Queen's University, Kingston, ON, Canada.
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
26
|
Ross FC, Patangia D, Grimaud G, Lavelle A, Dempsey EM, Ross RP, Stanton C. The interplay between diet and the gut microbiome: implications for health and disease. Nat Rev Microbiol 2024; 22:671-686. [PMID: 39009882 DOI: 10.1038/s41579-024-01068-4] [Citation(s) in RCA: 103] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 07/17/2024]
Abstract
Diet has a pivotal role in shaping the composition, function and diversity of the gut microbiome, with various diets having a profound impact on the stability, functionality and diversity of the microbial community within our gut. Understanding the profound impact of varied diets on the microbiome is crucial, as it will enable us not only to make well-informed dietary decisions for better metabolic and intestinal health, but also to prevent and slow the onset of specific diet-related diseases that stem from suboptimal diets. In this Review, we explore how geographical location affects the gut microbiome and how different diets shape its composition and function. We examine the mechanisms by which whole dietary regimes, such as the Mediterranean diet, high-fibre diet, plant-based diet, high-protein diet, ketogenic diet and Western diet, influence the gut microbiome. Furthermore, we underscore the need for exhaustive studies to better understand the causal relationship between diet, host and microorganisms for the development of precision nutrition and microbiome-based therapies.
Collapse
Affiliation(s)
- Fiona C Ross
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Dhrati Patangia
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Cork, Ireland
| | - Ghjuvan Grimaud
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Moorepark Food Research Centre, Cork, Ireland
| | - Aonghus Lavelle
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- INFANT Centre, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| |
Collapse
|
27
|
Li Y, Sun M, Tian X, Bao T, Yu Q, Ma NL, Gan R, Cheang WS, Wu X. Gymnemic acid alleviates gut barrier disruption and lipid dysmetabolism via regulating gut microbiota in HFD hamsters. J Nutr Biochem 2024; 133:109709. [PMID: 39053860 DOI: 10.1016/j.jnutbio.2024.109709] [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: 01/23/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Gut microbiota dysbiosis and gut barrier disruption are key events associated with high-fat diet (HFD)-induced systemic metabolic disorders. Gymnemic acid (GA) has been reported to have an important role in alleviating HFD-induced disorders of glycolipid metabolism, but its regulatory role in HFD-induced disorders of the gut microbiota and gut barrier function has not been elucidated. Here we showed that GA intervention in HFD-induced hamsters increased the relative abundance of short-chain fatty acid (SCFA)-producing microbes including Lactobacillus (P<.05) and Lachnoclostridium (P<.01) in the gut, and reduced the relative abundance of lipopolysaccharide (LPS)-producing microbes including Enterococcus (P<.05) and Bacteroides (P<.05), subsequently improving HFD-induced intestinal barrier dysfunction and systemic inflammation. Specifically, GA intervention reduced mRNA expression of inflammatory cytokines, including IL-1β, IL-6, and TNF-α (P<.01), increased mRNA expression of antioxidant-related genes, including Nfe2l2, Ho-1, and Nqo1 (P<.01), and increased mRNA expression of intestinal tight junction proteins, including Occludin and Claudin-1 (P<.01), thereby improving gut barrier function of HFD hamsters. This ameliorative effect of GA on the gut of HFD hamsters may further promote lipid metabolic balance in liver and adipose tissue by regulating the Toll-like receptor 4 (TLR4)-nuclear factor-κB (NF-κB) signaling pathway. Taken together, these results systematically revealed the important role of GA in regulating HFD-induced gut microbiota disturbance and gut barrier function impairment, providing a potential clinical theoretical basis for targeted treatment of HFD-induced microbiota dysbiosis.
Collapse
Affiliation(s)
- Yumeng Li
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China; TIB-UM Joint Laboratory of Synthetic Biology for Traditional Chinese Medicine, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, China
| | - Mingzhe Sun
- Air Force Medical Center of People's Liberation Army, Beijing, China; College of food science & nutritional engineering, China Agricultural University, Beijing, China
| | - Xutong Tian
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China
| | - Tongtong Bao
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China
| | - Qian Yu
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China
| | - Nyuk Ling Ma
- BIOSES Research Interest Group, Faculty of Science & Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia; Center for Global Health Research (CGHR), Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Renyou Gan
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China; TIB-UM Joint Laboratory of Synthetic Biology for Traditional Chinese Medicine, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, China
| | - Xin Wu
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China; TIB-UM Joint Laboratory of Synthetic Biology for Traditional Chinese Medicine, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, China.
| |
Collapse
|
28
|
Matsuki T, Nakamura S, Nishiyama M, Narimatsu H. Holistic Evaluation of the Gut Microbiota through Data Envelopment Analysis: A Cross-Sectional Study. Curr Dev Nutr 2024; 8:104469. [PMID: 39524216 PMCID: PMC11550754 DOI: 10.1016/j.cdnut.2024.104469] [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: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
Background The gut microbiome plays a crucial role in human health, but maintaining a healthy gut microbiome remains challenging. Current approaches often focus on individual components rather than providing a holistic assessment. Objectives To introduce and evaluate a novel approach using data envelopment analysis (DEA) for assessing gut microbiota efficiency and identifying potential targets for personalized interventions. Methods We conducted a cross-sectional analysis of 577 participants from the Kanagawa "ME-BYO" Prospective Cohort Study. Lifestyle factors and gut microbiota composition were assessed. DEA was employed to calculate an efficiency score for each participant, incorporating multiple inputs (lifestyle factors) and outputs (gut microbiotas). This score represents how efficiently an individual's lifestyle factors contribute to their gut microbiota composition. Tobit regression analysis was used to assess associations between efficiency scores and demographic and health-related factors. Results The mean efficiency score was 0.86, with 14.2% of participants classified as efficient. Efficiency scores showed positive correlations with alcohol intake and Faith's phylogenetic diversity. Tobit regression analysis revealed significant associations between efficiency scores and sex, fat intake, and yogurt consumption. DEA identified specific targets for improving gut microbiota composition in inefficient individuals. Conclusions This study demonstrates the potential of DEA as a tool for evaluating gut microbiota efficiency and providing personalized recommendations for microbiota optimization. This approach could lead to more effective strategies for optimizing gut health across diverse populations.
Collapse
Affiliation(s)
- Taizo Matsuki
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Sho Nakamura
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
- Department of Medical Genetics, Kanagawa Cancer Center, Nakao, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Minami Nishiyama
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Hiroto Narimatsu
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
- Department of Medical Genetics, Kanagawa Cancer Center, Nakao, Asahi-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
29
|
Ma X, Zhu N, Yu X, Wang W, Wu W. Research on preventive effect of Akkermansia muciniphilaAKK PROBIO on acute gouty arthritis in mice. Food Sci Nutr 2024; 12:7644-7656. [PMID: 39479687 PMCID: PMC11521663 DOI: 10.1002/fsn3.4367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 11/02/2024] Open
Abstract
In mice with acute gouty arthritis, this study intends to examine the mechanism of action of Akkermansia muciniphila AKK PROBIO. We developed a mouse model of acute gouty arthritis using sodium urate. The efficiency and mechanism of AKK PROBIO in preventing acute gouty arthritis in mice were then determined by examining the degree of foot swelling, pain threshold, blood biochemical indicators, histological alterations, and messenger RNA (mRNA) expression changes. The results of the animal experiment showed that AKK PROBIO can lessen mouse foot edema severity and increase pain threshold. AKK PROBIO can enhance the enzyme activity of superoxide dismutase (SOD) and the level of glutathione (GSH) in the ankle joint tissues of mice with acute arthritis while decreasing the enzyme activity of myeloperoxidase (MPO) and the level of malondialdehyde (MDA). Interleukin 6 (IL-6), interleukin 10 (IL-10), interleukin 1 beta (IL-1β), and tumor necrosis factor-alpha (TNF-α) levels are all reduced by AKK PROBIO in the blood of mice with acute arthritis. Results from histopathology showed that AKK PROBIO reduced tissue damage in the mouse ankle and foot joints. In the tissues of the ankle joints of mice with acute arthritis, the results of the quantitative polymerase chain reaction (qPCR) and Western blot experiments suggested that AKK PROBIO may inhibit the mRNA and protein expression of extracellular signal-regulated kinase 1/2 (ERK1/2), cyclooxygenase-2 (COX-2), and prostaglandin E2 (PGE2) in the tissues. AKK PROBIO can also regulate gut microbiota, inhibit harmful bacteria, and enhance valeric acid in the intestine, isobutyric acid, and isovaleric acid. Therefore, it is evident that AKK PROBIO prevents acute gouty arthritis better than glucosamine sulfate. It is a strain that has probiotic potential.
Collapse
Affiliation(s)
- Xin Ma
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Na Zhu
- Department of AnesthesiaFirst Affiliated Hospital of Chengdu Medical CollegeChengduChina
| | - Xueping Yu
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Wei Wang
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Wenzhong Wu
- Heilongjiang Red Cross Sengong General HospitalHarbinHeilongjiangChina
| |
Collapse
|
30
|
Ferreira H, Duarte D, Carneiro TJ, Costa C, Barbosa JC, Rodrigues JE, Alves P, Vasconcelos M, Pinto E, Gomes A, Gil AM. Impact of a legumes diet on the human gut microbiome articulated with fecal and plasma metabolomes: A pilot study. Clin Nutr ESPEN 2024; 63:332-345. [PMID: 38964655 DOI: 10.1016/j.clnesp.2024.06.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/12/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND & AIMS Legumes intake is known to be associated with several health benefits the origins of which is still a matter of debate. This paper addresses a pilot small cohort to probe for metabolic aspects of the interplay between legumes intake, human metabolism and gut microbiota. METHODS Untargeted nuclear magnetic resonance (NMR) metabolomics of blood plasma and fecal extracts was carried out, in tandem with qPCR analysis of feces, to assess the impact of an 8-week pilot legumes diet intervention on the fecal and plasma metabolomes and gut microbiota of 19 subjects. RESULTS While the high inter-individual variability hindered the detection of statistically significant changes in the gut microbiome, increased fecal glucose and decreased threonine levels were noted. Correlation analysis between the microbiome and fecal metabolome lead to putative hypotheses regarding the metabolic activities of prevalent bacteria groups (Clostridium leptum subgroup, Roseburia spp., and Faecalibacterium prausnitzii). These included elevated fecal glucose as a preferential energy source, the involvement of valerate/isovalerate and reduced protein degradation in gut microbiota. Plasma metabolomics advanced mannose and betaine as potential markers of legume intake and unveiled a decrease in formate and ketone bodies, the latter suggesting improved energy utilization through legume carbohydrates. Amino acid metabolism was also apparently affected, as suggested by lowered urea, histidine and threonine levels. CONCLUSIONS Despite the high inter-individual gut microbiome variability characterizing the small cohort addressed, combination of microbiological measurements and untargeted metabolomics unveiled several metabolic effects putatively related to legumes intake. If confirmed in larger cohorts, our findings will support the inclusion of legumes in diets and contribute valuable new insight into the origins of associated health benefits.
Collapse
Affiliation(s)
- Helena Ferreira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal; Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Daniela Duarte
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Tatiana J Carneiro
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Célia Costa
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Joana C Barbosa
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - João E Rodrigues
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Paulo Alves
- Universidade Católica Portuguesa, CIIS - Centro de Investigação Interdisciplinar em Saúde, Escola Enfermagem (Porto), Portugal
| | - Marta Vasconcelos
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Elisabete Pinto
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal; EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
| | - Ana Gomes
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Ana M Gil
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
31
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
32
|
He CYY, Zhou Z, Kan MMP, Chan DHY, Wong ACT, Mok KHY, Lam FMH, Chan SCC, Cheung CKC, Yeung MKC, Wong AYL. Modifiable risk factors for mild cognitive impairment among cognitively normal community-dwelling older adults: A systematic review and meta-analysis. Ageing Res Rev 2024; 99:102350. [PMID: 38942197 DOI: 10.1016/j.arr.2024.102350] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 06/30/2024]
Abstract
Although numerous studies have investigated modifiable risk factors for mild cognitive impairment (MCI) among community-dwelling seniors, no meta-analysis has summarized these findings. Five databases were searched from January 1, 2000, to December 30, 2023. The protocol was registered with PROSPERO. Data were extracted and reported following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Relevant meta-analyses of modifiable risk factors were performed. The evidence of each factor was assessed by the GRADE for cohort studies. Of 16,651 citations, 87 studies involving 225,584 community-dwelling seniors were included. Fourteen meta-analyses involving 20 studies with 44,199 participants were performed. The analyses revealed low-to-moderate-quality evidence supporting that diabetes, 2 or more comorbidities, anxiety, apathy, depressive symptoms, and physical frailty were risk factors for incident MCI in older adults. Conversely, hypertension, agitation, and irritability might not be risk factors. Additionally, moderate-quality evidence supports the protective effect of engaging in cognitive-demanding activities on the onset of MCI. Collectively, this study constitutes the first extensive compilation of evidence regarding the various risk factors for the development of MCI in older adults. Our findings hold significant potential to guide the formulation of prevention and management strategies to either prevent or potentially reverse the onset of MCI.
Collapse
Affiliation(s)
- Christo Y Y He
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Zhixing Zhou
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Mandy M P Kan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Dorothy H Y Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Athena C T Wong
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Kenny H Y Mok
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Freddy M H Lam
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Sam C C Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Chelsia K C Cheung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Michael K C Yeung
- Department of Psychology, The Education University of Hong Kong, 999077, Hong Kong Special Administrative Region of China.
| | - Arnold Y L Wong
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China; Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China.
| |
Collapse
|
33
|
Zang X, Du Y, Jiang M, Zhou S, Wang L, Han X. A thorough investigation into the correlation between migraines and the gut microbiome: an in-depth analysis using Mendelian randomization studies. Front Neurol 2024; 15:1356974. [PMID: 39015315 PMCID: PMC11250663 DOI: 10.3389/fneur.2024.1356974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/29/2024] [Indexed: 07/18/2024] Open
Abstract
Objective A growing body of evidence underscores a significant association between neurological disorders, particularly migraines, and the gut microbiota. However, a research gap persists in understanding the cause-and-effect dynamics between these elements. Therefore, we employed robust methodologies aimed at thoroughly exploring the causal relationship between the gut microbiome and migraines. Methods Employing bidirectional Two Sample Mendelian Randomization (TSMR) analysis, we investigated the causal association between the composition of the gut microbiota and migraines. Data summarizing the relationship between gut microbiota and migraines were extracted from one or more genome-wide association studies. The TSMR analysis employed five methods to assess the correlation between the gut microbiota and migraines, with the inverse variance-weighted method serving as the primary approach for analyzing causal links. Sensitivity analyses were applied to address horizontal pleiotropy and heterogeneity. Simultaneously, a meta-analysis was performed to strengthen the robustness of the findings. Additionally, a reverse TSMR was carried out to explore potential occurrences of reverse causal relationships. Results The ongoing TSMR analysis identified a collection of 14 bacterial taxa connected to migraines. Among these, 8 taxa exhibited a protective effect, while 5 taxa had a detrimental impact, and 1 taxon maintained a neutral relationship. The reverse Mendelian randomization analysis highlighted stable outcomes for only one bacterial taxonomic group. Conclusion The study confirms a causal relationship between the gut microbiota and migraines, offering a new perspective for migraine research. Strategically targeting specific bacterial taxa with dysregulation may be effective in both preventing and treating migraines, thus opening new avenues for therapeutic strategies.
Collapse
Affiliation(s)
- Xuege Zang
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchuan, Jilin, China
| | - Yongkun Du
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchuan, Jilin, China
| | - Mengshu Jiang
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchuan, Jilin, China
| | | | - Libo Wang
- Unity Health Toronto, Toronto, ON, Canada
| | - Xuemei Han
- Unity Health Toronto, Toronto, ON, Canada
| |
Collapse
|
34
|
Masi D, Le Roy T, Adriouch S, Clément K. Nourishing the gut: the impact of diet on host-gut microbiota interaction. Curr Opin Clin Nutr Metab Care 2024; 27:361-371. [PMID: 38260940 DOI: 10.1097/mco.0000000000001009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
PURPOSE OF REVIEW Understanding the spectrum of drivers that influence the gut microbiome (GM) remains a crucial field of investigation. Among these factors, diet has received particular attention, as it could explain up to 20% of the variability in GM composition between individuals. This review focuses on the complex relationships between different dietary patterns and GM in humans, based on recent findings. RECENT FINDINGS Current evidence underscores the multifaceted impact of diet on GM richness, diversity, and overall composition. Key contributing factors encompass dietary habits, nutritional interventions, food quality and variety, macronutrient distribution, timing of feeding, and selective exclusion of certain foods. SUMMARY The intricate interplay between diet and GM is of fundamental importance in shaping the interaction between the host and the environment. Further understanding the causal impact of diet on GM has promising potential for the advancement of strategies to promote health and mitigate cardio-metabolic disease risks through dietary interventions. GRAPHICAL ABSTRACT http://links.lww.com/COCN/A21.
Collapse
Affiliation(s)
- Davide Masi
- Sorbonne University, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, Paris
- Sapienza University of Rome, Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, Rome
| | - Tiphaine Le Roy
- Sorbonne University, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, Paris
| | - Solia Adriouch
- Sorbonne University, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, Paris
| | - Karine Clément
- Sorbonne University, Inserm, Nutrition and Obesities: Systemic Approaches (NutriOmics) Research Unit, Paris
- Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
35
|
Florkowski M, Abiona E, Frank KM, Brichacek AL. Obesity-associated inflammation countered by a Mediterranean diet: the role of gut-derived metabolites. Front Nutr 2024; 11:1392666. [PMID: 38978699 PMCID: PMC11229823 DOI: 10.3389/fnut.2024.1392666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/03/2024] [Indexed: 07/10/2024] Open
Abstract
The prevalence of obesity has increased dramatically worldwide and has become a critical public health priority. Obesity is associated with many co-morbid conditions, including hypertension, diabetes, and cardiovascular disease. Although the physiology of obesity is complex, a healthy diet and sufficient exercise are two elements known to be critical to combating this condition. Years of research on the Mediterranean diet, which is high in fresh fruits and vegetables, nuts, fish, and olive oil, have demonstrated a reduction in numerous non-communicable chronic diseases associated with this diet. There is strong evidence to support an anti-inflammatory effect of the diet, and inflammation is a key driver of obesity. Changes in diet alter the gut microbiota which are intricately intertwined with human physiology, as gut microbiota-derived metabolites play a key role in biological pathways throughout the body. This review will summarize recent published studies that examine the potential role of gut metabolites, including short-chain fatty acids, bile acids, trimethylamine-N-oxide, and lipopolysaccharide, in modulating inflammation after consumption of a Mediterranean-like diet. These metabolites modulate pathways of inflammation through the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, toll-like receptor 4 signaling, and macrophage driven effects in adipocytes, among other mechanisms.
Collapse
Affiliation(s)
- Melanie Florkowski
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Esther Abiona
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Karen M Frank
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Allison L Brichacek
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| |
Collapse
|
36
|
Brichacek AL, Florkowski M, Abiona E, Frank KM. Ultra-Processed Foods: A Narrative Review of the Impact on the Human Gut Microbiome and Variations in Classification Methods. Nutrients 2024; 16:1738. [PMID: 38892671 PMCID: PMC11174918 DOI: 10.3390/nu16111738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Ultra-processed foods (UPFs) are foods that are industrially processed and are often pre-packaged, convenient, energy-dense, and nutrient-poor. UPFs are widespread in the current Western diet and their proposed contribution to non-communicable diseases such as obesity and cardiovascular disease is supported by numerous studies. UPFs are hypothesized to affect the body in multiple ways, including by inducing changes in the gut microbiome. This review summarizes the available research on the effect of UPFs on the gut microbiome. We also review current usage of the NOVA food classification system in randomized controlled trials and observational studies and how its implementation effects UPF research. Despite some differences in methodology between studies, results often associate UPF consumption with a number of negative health consequences. There are attempts to standardize a UPF classification system; however, reaching and implementing a consensus is difficult. Future studies focusing on the mechanisms by which UPFs effect the body, including through the microbiome and metabolome, will be essential to refine our understanding of the effects of UPFs on human health.
Collapse
Affiliation(s)
| | | | | | - Karen M. Frank
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Building 10, 10 Center Drive MSC 1508, Bethesda, MD 20892, USA; (A.L.B.); (M.F.); (E.A.)
| |
Collapse
|
37
|
García-Gavilán JF, Atzeni A, Babio N, Liang L, Belzer C, Vioque J, Corella D, Fitó M, Vidal J, Moreno-Indias I, Torres-Collado L, Coltell O, Toledo E, Clish C, Hernando J, Yun H, Hernández-Cacho A, Jeanfavre S, Dennis C, Gómez-Pérez AM, Martínez MA, Ruiz-Canela M, Tinahones FJ, Hu FB, Salas-Salvadó J. Effect of 1-year lifestyle intervention with energy-reduced Mediterranean diet and physical activity promotion on the gut metabolome and microbiota: a randomized clinical trial. Am J Clin Nutr 2024; 119:1143-1154. [PMID: 38428742 DOI: 10.1016/j.ajcnut.2024.02.021] [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: 11/30/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND The health benefits of the Mediterranean diet (MedDiet) have been linked to the presence of beneficial gut microbes and related metabolites. However, its impact on the fecal metabolome remains poorly understood. OBJECTIVES Our goal was to investigate the weight-loss effects of a 1-y lifestyle intervention based on an energy-reduced MedDiet coupled with physical activity (intervention group), compared with an ad libitum MedDiet (control group), on fecal metabolites, fecal microbiota, and their potential association with cardiovascular disease risk factors. METHODS A total of 400 participants (200 from each study group), aged 55-75 y, and at high cardiovascular disease risk, were included. Dietary and lifestyle information, anthropometric measurements, blood biochemical parameters, and stool samples were collected at baseline and after 1 y of follow-up. Liquid chromatography-tandem mass spectrometry was used to profile endogenous fecal metabolites, and 16S amplicon sequencing was employed to profile the fecal microbiota. RESULTS Compared with the control group, the intervention group exhibited greater weight loss and improvement in various cardiovascular disease risk factors. We identified intervention effects on 4 stool metabolites and subnetworks primarily composed of bile acids, ceramides, and sphingosines, fatty acids, carnitines, nucleotides, and metabolites of purine and the Krebs cycle. Some of these were associated with changes in several cardiovascular disease risk factors. In addition, we observed a reduction in the abundance of the genera Eubacterium hallii group and Dorea, and an increase in alpha diversity in the intervention group after 1 y of follow-up. Changes in the intervention-related microbiota profiles were also associated with alterations in different fecal metabolite subnetworks and some cardiovascular disease risk factors. CONCLUSIONS An intervention based on an energy-reduced MedDiet and physical activity promotion, compared with an ad libitum MedDiet, was associated with improvements in cardiometabolic risk factors, potentially through modulation of the fecal microbiota and metabolome. This trial was registered at https://www.isrctn.com/ as ISRCTN89898870 (https://doi.org/10.1186/ISRCTN89898870).
Collapse
Affiliation(s)
- Jesús F García-Gavilán
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Departament de Bioquímica i Biotecnologia, Alimentaciò, Nutrició, Desenvolupament i Salut Mental (ANUT-DSM), Universitat Rovira i Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Alessandro Atzeni
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Departament de Bioquímica i Biotecnologia, Alimentaciò, Nutrició, Desenvolupament i Salut Mental (ANUT-DSM), Universitat Rovira i Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| | - Nancy Babio
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Departament de Bioquímica i Biotecnologia, Alimentaciò, Nutrició, Desenvolupament i Salut Mental (ANUT-DSM), Universitat Rovira i Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Jesús Vioque
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernández (ISABIAL-UMH), Alicante, Spain
| | - Dolores Corella
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Montserrat Fitó
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas Municipal d'Investigació Médica (IMIM), Barcelona, Spain
| | - Josep Vidal
- CIBER Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Department of Endocrinology, Institut d'Investigacions Biomédiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Isabel Moreno-Indias
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Laura Torres-Collado
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernández (ISABIAL-UMH), Alicante, Spain
| | - Oscar Coltell
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Computer Languages and Systems, Jaume I University, Castellón, Spain
| | - Estefanía Toledo
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain; Epidemiología y Salud Pública, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Clary Clish
- Metabolomics Platform, The Broad Institute of MIT and Harvard, Boston, MA, United States
| | - Javier Hernando
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas Municipal d'Investigació Médica (IMIM), Barcelona, Spain
| | - Huan Yun
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Adrián Hernández-Cacho
- Departament de Bioquímica i Biotecnologia, Alimentaciò, Nutrició, Desenvolupament i Salut Mental (ANUT-DSM), Universitat Rovira i Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Sarah Jeanfavre
- Metabolomics Platform, The Broad Institute of MIT and Harvard, Boston, MA, United States
| | - Courtney Dennis
- Metabolomics Platform, The Broad Institute of MIT and Harvard, Boston, MA, United States
| | - Ana M Gómez-Pérez
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Maria Angeles Martínez
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Departament de Bioquímica i Biotecnologia, Alimentaciò, Nutrició, Desenvolupament i Salut Mental (ANUT-DSM), Universitat Rovira i Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Miguel Ruiz-Canela
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain; Epidemiología y Salud Pública, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Francisco J Tinahones
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, United States; Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jordi Salas-Salvadó
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Departament de Bioquímica i Biotecnologia, Alimentaciò, Nutrició, Desenvolupament i Salut Mental (ANUT-DSM), Universitat Rovira i Virgili, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.
| |
Collapse
|
38
|
Gong C, Jiao C, Liang H, Ma Y, Wu Q, Xie Y. Exome-Based Amino Acid Optimization: A Dietary Strategy to Satisfy Human Nutritional Demands and Enhance Muscle Strength in Breast Tumor Mice Undergoing Chemotherapy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7089-7099. [PMID: 38512774 DOI: 10.1021/acs.jafc.3c07256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Breast cancer patients undergoing chemotherapy often experience muscle wasting and weakness, which impact their quality of life. A potential solution lies in customizing amino acid compositions based on exome-derived formulations (ExAAs). The study hypothesized that tailoring dietary amino acids using ExAAs could enhance muscle health. Theoretical amino acid requirements were calculated from the genome's exome region, and a breast cancer mouse model undergoing paclitaxel treatment was established. The mice were supplemented with a cancer-specific nutritional formula (QJS), and the effects of QJS and amino acid-adjusted QJS (adjQJS) were compared. Both formulations improved the nutritional status without compromising tumor growth. Notably, adjQJS significantly enhanced muscle strength compared to QJS (1.51 ± 0.25 vs. 1.30 ± 0.08 fold change, p < 0.05). Transcriptome analysis revealed alterations in complement and coagulation cascades, with an observed upregulation of C3 gene expression in adjQJS. Immune regulation also changed, showing a decrease in B cells and an increase in monocytes in skeletal muscle with adjQJS. Importantly, adjQJS resulted in a notable increase in Alistipes abundance compared to QJS (10.19 ± 0.04% vs. 5.03 ± 1.75%). This study highlights the potential of ExAAs as valuable guide for optimizing amino acid composition in diets for breast cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Congcong Gong
- South China University of Technology, Guangzhou 510070, PrR. China
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou 510070, PR. China
| | - Chunwei Jiao
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou 510070, PR. China
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing 526000, PR. China
| | - Huijia Liang
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou 510070, PR. China
| | - Yuxin Ma
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing 526000, PR. China
| | - Qingping Wu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, PR. China
| | - Yizhen Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, PR. China
| |
Collapse
|
39
|
Manrique P, Montero I, Fernandez-Gosende M, Martinez N, Cantabrana CH, Rios-Covian D. Past, present, and future of microbiome-based therapies. MICROBIOME RESEARCH REPORTS 2024; 3:23. [PMID: 38841413 PMCID: PMC11149097 DOI: 10.20517/mrr.2023.80] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 06/07/2024]
Abstract
Technological advances in studying the human microbiome in depth have enabled the identification of microbial signatures associated with health and disease. This confirms the crucial role of microbiota in maintaining homeostasis and the host health status. Nowadays, there are several ways to modulate the microbiota composition to effectively improve host health; therefore, the development of therapeutic treatments based on the gut microbiota is experiencing rapid growth. In this review, we summarize the influence of the gut microbiota on the development of infectious disease and cancer, which are two of the main targets of microbiome-based therapies currently being developed. We analyze the two-way interaction between the gut microbiota and traditional drugs in order to emphasize the influence of gut microbial composition on drug effectivity and treatment response. We explore the different strategies currently available for modulating this ecosystem to our benefit, ranging from 1st generation intervention strategies to more complex 2nd generation microbiome-based therapies and their regulatory framework. Lastly, we finish with a quick overview of what we believe is the future of these strategies, that is 3rd generation microbiome-based therapies developed with the use of artificial intelligence (AI) algorithms.
Collapse
|
40
|
Zhang Q, Li X, Huang T, Zhang S, Teng K, Rousitemu N, Lan T, Wen Y. Alterations in the diversity, composition and function of the gut microbiota in Uyghur individuals with sarcopenia. Exp Gerontol 2024; 187:112376. [PMID: 38331300 DOI: 10.1016/j.exger.2024.112376] [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: 11/05/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Research on the gut microbiota has emerged as a new direction for understanding pathophysiologic changes in diseases associated with aging, such as sarcopenia. Several studies have shown that there are differences in the gut microbiota between individuals with sarcopenia and without sarcopenia. However, these differences are not consistent across regions and ethnic groups, and additional research is needed. METHODS In this study, we collected fresh fecal samples from 31 Uyghur individuals with sarcopenia and 31 healthy controls. We used 16S rRNA sequencing to obtain fecal base sequences and analyzed the diversity, composition and function of the gut microbiota. RESULTS There was no significant difference in alpha diversity between the sarcopenia group and the healthy control group (P > 0.05). There was a significant difference in beta diversity between the groups (P < 0.05). In the sarcopenia group, the abundances of Alloprevotella, un_f_Prevotellaceae, Anaerovibrio, Prevotellaceae_NK3B31_group, Mitsuokella, Prevotella and Allisonella were lower than those in the heathy control group, and the abundances of Flavobacteriales, Flavobacteriaceae, Catenibacterium, Romboutsia, Erysipelotrichaceae_UCG-003, GCA-900066575, Lachnospiraceae_FCS020_group, and un_f_Flavobacteriaceae were higher than those in the heathy control group. Linear discriminant analysis effect size (LEfSe) revealed that the microbial species in the control group that were significantly different from those in the sarcopenia group were concentrated in the genus Alloprevotella, while the species in the sarcopenia group were concentrated in the genus Catenibacterium. Functional prediction analysis revealed that D-alanine, glycine, serine, and threonine metabolism and transcription machinery, among others, were enriched in the sarcopenia group, which indicated that metabolic pathways related to amino acid metabolism and nutrient transport may be regulated to varying degrees in the pathophysiological context of sarcopenia. CONCLUSIONS There were significant differences in the composition and function of the gut microbiota between Xinjiang Uyghur sarcopenia individuals and healthy individuals. These findings might aid in the development of probiotics or microbial-based therapies for sarcopenia in Uyhur individuals.
Collapse
Affiliation(s)
- Qiuxi Zhang
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Xin Li
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Ting Huang
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Shuang Zhang
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Kunchen Teng
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Namuna Rousitemu
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Ting Lan
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Youfeng Wen
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
41
|
Gupta VK, Rajendraprasad S, Ozkan M, Ramachandran D, Ahmad S, Bakken JS, Laudanski K, Gajic O, Bauer B, Zec S, Freeman DW, Khanna S, Shah A, Skalski JH, Sung J, Karnatovskaia LV. Safety, feasibility, and impact on the gut microbiome of kefir administration in critically ill adults. BMC Med 2024; 22:80. [PMID: 38378568 PMCID: PMC10880344 DOI: 10.1186/s12916-024-03299-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/12/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Dysbiosis of the gut microbiome is frequent in the intensive care unit (ICU), potentially leading to a heightened risk of nosocomial infections. Enhancing the gut microbiome has been proposed as a strategic approach to mitigate potential adverse outcomes. While prior research on select probiotic supplements has not successfully shown to improve gut microbial diversity, fermented foods offer a promising alternative. In this open-label phase I safety and feasibility study, we examined the safety and feasibility of kefir as an initial step towards utilizing fermented foods to mitigate gut dysbiosis in critically ill patients. METHODS We administered kefir in escalating doses (60 mL, followed by 120 mL after 12 h, then 240 mL daily) to 54 critically ill patients with an intact gastrointestinal tract. To evaluate kefir's safety, we monitored for gastrointestinal symptoms. Feasibility was determined by whether patients received a minimum of 75% of their assigned kefir doses. To assess changes in the gut microbiome composition following kefir administration, we collected two stool samples from 13 patients: one within 72 h of admission to the ICU and another at least 72 h after the first stool sample. RESULTS After administering kefir, none of the 54 critically ill patients exhibited signs of kefir-related bacteremia. No side effects like bloating, vomiting, or aspiration were noted, except for diarrhea in two patients concurrently on laxatives. Out of the 393 kefir doses prescribed for all participants, 359 (91%) were successfully administered. We were able to collect an initial stool sample from 29 (54%) patients and a follow-up sample from 13 (24%) patients. Analysis of the 26 paired samples revealed no increase in gut microbial α-diversity between the two timepoints. However, there was a significant improvement in the Gut Microbiome Wellness Index (GMWI) by the second timepoint (P = 0.034, one-sided Wilcoxon signed-rank test); this finding supports our hypothesis that kefir administration can improve gut health in critically ill patients. Additionally, the known microbial species in kefir were found to exhibit varying levels of engraftment in patients' guts. CONCLUSIONS Providing kefir to critically ill individuals is safe and feasible. Our findings warrant a larger evaluation of kefir's safety, tolerability, and impact on gut microbiome dysbiosis in patients admitted to the ICU. TRIAL REGISTRATION NCT05416814; trial registered on June 13, 2022.
Collapse
Affiliation(s)
- Vinod K Gupta
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Surgery Research, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Sanu Rajendraprasad
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mahmut Ozkan
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Sumera Ahmad
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Johan S Bakken
- Section of Infectious Diseases, St Luke's Hospital, Duluth, MN, USA
| | - Krzysztof Laudanski
- Department of Anesthesiology and Perioperative Care, Mayo Clinic, Rochester, MN, USA
| | - Ognjen Gajic
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Brent Bauer
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Simon Zec
- Department of Anesthesiology and Perioperative Care, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - David W Freeman
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Sahil Khanna
- Division of Gastroenterology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Aditya Shah
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joseph H Skalski
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jaeyun Sung
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
- Division of Surgery Research, Department of Surgery, Mayo Clinic, Rochester, MN, USA.
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| | | |
Collapse
|
42
|
Suprunowicz M, Tomaszek N, Urbaniak A, Zackiewicz K, Modzelewski S, Waszkiewicz N. Between Dysbiosis, Maternal Immune Activation and Autism: Is There a Common Pathway? Nutrients 2024; 16:549. [PMID: 38398873 PMCID: PMC10891846 DOI: 10.3390/nu16040549] [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: 01/18/2024] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neuropsychiatric condition characterized by impaired social interactions and repetitive stereotyped behaviors. Growing evidence highlights an important role of the gut-brain-microbiome axis in the pathogenesis of ASD. Research indicates an abnormal composition of the gut microbiome and the potential involvement of bacterial molecules in neuroinflammation and brain development disruptions. Concurrently, attention is directed towards the role of short-chain fatty acids (SCFAs) and impaired intestinal tightness. This comprehensive review emphasizes the potential impact of maternal gut microbiota changes on the development of autism in children, especially considering maternal immune activation (MIA). The following paper evaluates the impact of the birth route on the colonization of the child with bacteria in the first weeks of life. Furthermore, it explores the role of pro-inflammatory cytokines, such as IL-6 and IL-17a and mother's obesity as potentially environmental factors of ASD. The purpose of this review is to advance our understanding of ASD pathogenesis, while also searching for the positive implications of the latest therapies, such as probiotics, prebiotics or fecal microbiota transplantation, targeting the gut microbiota and reducing inflammation. This review aims to provide valuable insights that could instruct future studies and treatments for individuals affected by ASD.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (M.S.); (N.T.); (A.U.); (K.Z.); (N.W.)
| | | |
Collapse
|
43
|
Tian Y, Fu M, Su J, Yan M, Yu J, Wang C, Niu Z, Du Y, Hu X, Zheng J, Tao B, Gao Z, Chen J, Chen S, Lv G. Gut microbiota dysbiosis and intestinal barrier impairment in diarrhea caused by cold drink and high-fat diet. Toxicology 2024; 502:153728. [PMID: 38216112 DOI: 10.1016/j.tox.2024.153728] [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: 11/15/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/14/2024]
Abstract
Cold drink and high-fat diet (CDHFD) are common diet patterns. However, the potential risks remain unclear. We investigated the effects of CDHFD in adult mice and explored the mechanisms of action. Twenty adult male mice were randomly divided into control and model groups, and the control group was fed a normal diet, whereas the model group was fed CDHFD for 28 days. We found that mice in the model group developed diarrhea symptoms accompanied by fatigue and weakness. Analysis of the intestinal flora revealed that the model group had a lower diversity and richness of microorganism species in the gut than the control group. Furthermore, the characteristic analysis indicated that CDHFD downregulated specific bacteria, such as norank_f_Muribaculaceae, Muribaculum, and Odoribacter, which are known to be associated with the systemic inflammatory response and mucosal barrier function. Blood tests showed that immune cells and inflammatory cytokines were significantly elevated in the model group, along with increased LPS induced by CDHFD. Pathological investigations demonstrated that CDHFD damages the intestinal mucosa while affecting the expression of tight junction proteins, including ZO-1, Claudin-1, Claudin-2, and Occludin, which may be attributed to the activation of the TRAF6/IκB/p65 signaling pathway. In conclusion, impaired gut microbial and mechanical barrier function is responsible for CDHFD-induced diarrhea. In this study, we constructed a model of diet-induced diarrhea by simulating human dietary patterns, evaluated the long-term effects of CDHFD on human intestinal barriers and immune systems, and revealed its mechanism of action based on chronic inflammation. This study validated the model's fit to provide an effective screening model for drug or functional food development.
Collapse
Affiliation(s)
- Yajuan Tian
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meng Fu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Su
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meiqiu Yan
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingjing Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxing Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuangwei Niu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuzhong Du
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xueling Hu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiayi Zheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bai Tao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zengguang Gao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianzhen Chen
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Suhong Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Hangzhou, China.
| | - Guiyuan Lv
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
44
|
Dridi C, Millette M, Salmieri S, Aguilar Uscanga BR, Lacroix S, Venneri T, Sarmast E, Allahdad Z, Di Marzo V, Silvestri C, Lacroix M. Effect of a Probiotic Beverage Enriched with Cricket Proteins on the Gut Microbiota: Composition of Gut and Correlation with Nutritional Parameters. Foods 2024; 13:204. [PMID: 38254505 PMCID: PMC10814958 DOI: 10.3390/foods13020204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
The health and balance of the gut microbiota are known to be linked to diet composition and source, with fermented products and dietary proteins potentially providing an exceptional advantage for the gut. The purpose of this study was to evaluate the effect of protein hydrolysis, using a probiotic beverage enriched with either cricket protein (CP) or cricket protein hydrolysates (CP.Hs), on the composition of the gut microbiota of rats. Taxonomic characterization of the gut microbiota in fecal samples was carried out after a 14-day nutritional study to identify modifications induced by a CP- and CP.H-enriched fermented probiotic product. The results showed no significant differences (p > 0.05) in the diversity and richness of the gut microbiota among the groups fed with casein (positive control), CP-enriched, and fermented CP.H-enriched probiotic beverages; however, the overall composition of the microbiota was altered, with significant modifications in the relative abundance of several bacterial families and genera. In addition, fermented CP.H-enriched probiotic beverages could be related to the decrease in the number of potential pathogens such as Enterococcaceae. The association of gut microbiota with the nutritional parameters was determined and the results showed that digestibility and the protein efficiency ratio (PER) were highly associated with the abundance of several taxa.
Collapse
Affiliation(s)
- Chaima Dridi
- Research Laboratories in Sciences, Applied to Food (RESALA), Canadian Irradiation Centre (CIC), INRS Armand-Frappier Health Biotechnology Research Centre, Laval, QC H7V 1B7, Canada (M.M.)
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Mathieu Millette
- Research Laboratories in Sciences, Applied to Food (RESALA), Canadian Irradiation Centre (CIC), INRS Armand-Frappier Health Biotechnology Research Centre, Laval, QC H7V 1B7, Canada (M.M.)
- Bio-K+, a Kerry Company, Preclinical Research Division, Laval, QC H7V 4B3, Canada
| | - Stephane Salmieri
- Research Laboratories in Sciences, Applied to Food (RESALA), Canadian Irradiation Centre (CIC), INRS Armand-Frappier Health Biotechnology Research Centre, Laval, QC H7V 1B7, Canada (M.M.)
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Blanca R. Aguilar Uscanga
- Research Laboratory of Industrial Microbiology, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara 44430, Mexico;
| | - Sebastien Lacroix
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC G1V 0A6, Canada
- Faculty of Agriculture and Food Sciences (FSAA), Université Laval, Quebec, QC G1V 0A6, Canada;
| | - Tommaso Venneri
- Joint International Research Unit on Chemical and Biomolecular Research on the Microbiomeand Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Université Laval, Quebec, QC G1V 0A6, Canada
- Heart and Lung Institute Research Centre (IUCPQ), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Elham Sarmast
- Research Laboratories in Sciences, Applied to Food (RESALA), Canadian Irradiation Centre (CIC), INRS Armand-Frappier Health Biotechnology Research Centre, Laval, QC H7V 1B7, Canada (M.M.)
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Zahra Allahdad
- Research Laboratories in Sciences, Applied to Food (RESALA), Canadian Irradiation Centre (CIC), INRS Armand-Frappier Health Biotechnology Research Centre, Laval, QC H7V 1B7, Canada (M.M.)
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Vincenzo Di Marzo
- Joint International Research Unit on Chemical and Biomolecular Research on the Microbiomeand Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Université Laval, Quebec, QC G1V 0A6, Canada
- Heart and Lung Institute Research Centre (IUCPQ), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Cristoforo Silvestri
- Faculty of Agriculture and Food Sciences (FSAA), Université Laval, Quebec, QC G1V 0A6, Canada;
- Heart and Lung Institute Research Centre (IUCPQ), Université Laval, Quebec, QC G1V 0A6, Canada
- Nutrition, Health and Society (NUTRISS) Centre, Department of Medicine, Faculty of Medicine, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Monique Lacroix
- Research Laboratories in Sciences, Applied to Food (RESALA), Canadian Irradiation Centre (CIC), INRS Armand-Frappier Health Biotechnology Research Centre, Laval, QC H7V 1B7, Canada (M.M.)
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
45
|
Park G, Kadyan S, Hochuli N, Pollak J, Wang B, Salazar G, Chakrabarty P, Efron P, Sheffler J, Nagpal R. A modified Mediterranean-style diet enhances brain function via specific gut-microbiome-brain mechanisms. Gut Microbes 2024; 16:2323752. [PMID: 38444392 PMCID: PMC10936641 DOI: 10.1080/19490976.2024.2323752] [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: 09/08/2023] [Accepted: 02/22/2024] [Indexed: 03/07/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating brain disorder with rapidly mounting prevalence worldwide, yet no proven AD cure has been discovered. Using a multi-omics approach in a transgenic AD mouse model, the current study demonstrated the efficacy of a modified Mediterranean-ketogenic diet (MkD) on AD-related neurocognitive pathophysiology and underlying mechanisms related to the gut-microbiome-brain axis. The findings revealed that MkD induces profound shifts in the gut microbiome community and microbial metabolites. Most notably, MkD promoted growth of the Lactobacillus population, resulting in increased bacteria-derived lactate production. We discovered elevated levels of microbiome- and diet-derived metabolites in the serum as well, signaling their influence on the brain. Importantly, these changes in serum metabolites upregulated specific receptors that have neuroprotective effects and induced alternations in neuroinflammatory-associated pathway profiles in hippocampus. Additionally, these metabolites displayed strong favorable co-regulation relationship with gut-brain integrity and inflammatory markers, as well as neurobehavioral outcomes. The findings underscore the ameliorative effects of MkD on AD-related neurological function and the underlying gut-brain communication via modulation of the gut microbiome-metabolome arrays.
Collapse
Affiliation(s)
- Gwoncheol Park
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| | - Saurabh Kadyan
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| | - Nathaniel Hochuli
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| | - Julie Pollak
- Department of Chemistry and Chemical Engineering, Florida Institute of Technology, Melbourne, FL, USA
| | - Bo Wang
- Department of Chemistry and Chemical Engineering, Florida Institute of Technology, Melbourne, FL, USA
| | - Gloria Salazar
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Diseases, Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Philip Efron
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Julia Sheffler
- Center for Translational Behavioral Science, Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Ravinder Nagpal
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Science, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
46
|
Wang J, Zhang G, Lai H, Li Z, Shen M, Li C, Kwan P, O'Brien TJ, Wu T, Yang S, Zhang X, Zhang L. Characterizing Gut Microbiota in Older Chinese Adults with Cognitive Impairment: A Cross-Sectional Study. J Alzheimers Dis 2024; 101:761-771. [PMID: 39213074 DOI: 10.3233/jad-240597] [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: 09/04/2024]
Abstract
Background Cognitive impairment is a clinical manifestation that occurs in the course of dementia like Alzheimer's disease. The association between cognitive impairment and gut microbiota is unclear. Objective We aimed to identify gut microbiota characteristics and key gut microbiota biomarkers associated with cognitive impairment in a relatively large cohort of older adults in China. Methods A total of 229 adults aged ≥60 years from Shenzhen, China were recruited into this cross-sectional study. Participants were divided into cognitive impairment (CI) and no cognitive impairment (NCI) groups according to the results of the Mini-Mental State Examination. Diversity analysis and network analysis were used to characterize the gut microbiota between the two groups. The linear discriminant analysis effect size method and machine learning approaches were sequentially performed to identify gut microbiota biomarkers. The relationship between biomarkers and lifestyle factors was explored using Transformation-based redundancy analysis (tb-RDA). Results A total of 74 CI participants and 131 NCI participants were included in the analysis. The CI group demonstrated lower α-diversity compared to the NCI group (Shannon: 2.798 versus 3.152, p < 0.001). The density of the gut microbiota interaction network was lower in the CI group (0.074) compared to the NCI group (0.081). Megamonas, Blautia, Pseudomonas, Stenotrophomonas, and Veillonella were key biomarkers for CI. The tb-RDA revealed that increased fruit intake and exercise contribute to a higher abundance of Megamonas, Blautia, and Veillonella. Conclusions We identified a significantly reduced abundance of certain beneficial gut microbiota in older Chinese adults with cognitive impairment.
Collapse
Affiliation(s)
- Jing Wang
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, PR China
| | - Gong Zhang
- MOE Key Laboratory of Tumour Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Hao Lai
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, PR China
| | - Zengbin Li
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, PR China
| | - Mingwang Shen
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, PR China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China
| | - Chao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, China
| | - Patrick Kwan
- The Department of Neuroscience, The School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University & Alfred Health, Melbourne, Australia
| | - Terence J O'Brien
- The Department of Neuroscience, The School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University & Alfred Health, Melbourne, Australia
| | - Ting Wu
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Siyu Yang
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xueli Zhang
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lei Zhang
- China-Australia Joint Research Centre for Infectious Diseases, School of Public Health, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, PR China
- The Department of Neuroscience, The School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University & Alfred Health, Melbourne, Australia
- Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, Australia
| |
Collapse
|
47
|
Gauthier E, Milagro FI, Navas-Carretero S. Effect of low-and non-calorie sweeteners on the gut microbiota: A review of clinical trials and cross-sectional studies. Nutrition 2024; 117:112237. [PMID: 37897982 DOI: 10.1016/j.nut.2023.112237] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 10/30/2023]
Abstract
Use of non-nutritive sweeteners (NNSs) has increased worldwide in recent decades. However, evidence from preclinical studies shows that sweetener consumption may induce glucose intolerance through changes in the gut microbiota, which raises public health concerns. As studies conducted on humans are lacking, the aim of this review was to gather and summarize the current evidence on the effects of NNSs on human gut microbiota. Only clinical trials and cross-sectional studies were included in the review. Regarding NNSs (i.e, saccharin, sucralose, aspartame, and stevia), only two of five clinical trials showed significant changes in gut microbiota composition after the intervention protocol. These studies concluded that saccharin and sucralose impair glycemic tolerance. In three of the four cross-sectional studies an association between NNSs and the microbial composition was observed. All three clinical trials on polyols (i.e, xylitol) showed prebiotic effects on gut microbiota, but these studies had multiple limitations (publication date, dosage, duration) that jeopardize their validity. The microbial response to NNSs consumption could be strongly mediated by the gut microbial composition at baseline. Further studies in which the potential personalized microbial response to NNSs consumption is acknowledged, and that include longer intervention protocols, larger cohorts, and more realistic sweetener dosage are needed to broaden these findings.
Collapse
Affiliation(s)
- Ellie Gauthier
- School of Nutrition, Université Laval, Quebec City, Quebec, Canada; Centre Nutrition, santé et société (NUTRISS)-Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Quebec City, Quebec, Canada
| | - Fermin I Milagro
- Center for Nutrition Research; Department of Nutrition, Food Sciences and Physiology; School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Santiago Navas-Carretero
- Center for Nutrition Research; Department of Nutrition, Food Sciences and Physiology; School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
48
|
Prame Kumar K, Ooi JD, Goldberg R. The interplay between the microbiota, diet and T regulatory cells in the preservation of the gut barrier in inflammatory bowel disease. Front Microbiol 2023; 14:1291724. [PMID: 38107848 PMCID: PMC10722198 DOI: 10.3389/fmicb.2023.1291724] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
Inflammatory bowel disease (IBD) is becoming more common in the Western world due to changes in diet-related microbial dysbiosis, genetics and lifestyle. Incidences of gut permeability can predate IBD and continued gut barrier disruptions increase the exposure of bacterial antigens to the immune system thereby perpetuating chronic inflammation. Currently, most of the approved IBD therapies target individual pro-inflammatory cytokines and pathways. However, they fail in approximately 50% of patients due to their inability to overcome the redundant pro inflammatory immune responses. There is increasing interest in the therapeutic potential of T regulatory cells (Tregs) in inflammatory conditions due to their widespread capability to dampen inflammation, promote tolerance of intestinal bacteria, facilitate healing of the mucosal barrier and ability to be engineered for more targeted therapy. Intestinal Treg populations are inherently shaped by dietary molecules and gut microbiota-derived metabolites. Thus, understanding how these molecules influence Treg-mediated preservation of the intestinal barrier will provide insights into immune tolerance-mediated mucosal homeostasis. This review comprehensively explores the interplay between diet, gut microbiota, and immune system in influencing the intestinal barrier function to attenuate the progression of colitis.
Collapse
Affiliation(s)
- Kathryn Prame Kumar
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash Medical Centre, Monash University, Clayton, VIC, Australia
| | | | | |
Collapse
|
49
|
Kalnina I, Gudra D, Silamikelis I, Viksne K, Roga A, Skinderskis E, Fridmanis D, Klovins J. Variations in the Relative Abundance of Gut Bacteria Correlate with Lipid Profiles in Healthy Adults. Microorganisms 2023; 11:2656. [PMID: 38004667 PMCID: PMC10673050 DOI: 10.3390/microorganisms11112656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/04/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
The gut microbiome is a versatile system regulating numerous aspects of host metabolism. Among other traits, variations in the composition of gut microbial communities are related to blood lipid patterns and hyperlipidaemia, yet inconsistent association patterns exist. This study aims to assess the relationships between the composition of the gut microbiome and variations in lipid profiles among healthy adults. This study used data and samples from 23 adult participants of a previously conducted dietary intervention study. Circulating lipid measurements and whole-metagenome sequences of the gut microbiome were derived from 180 blood and faecal samples collected from eight visits distributed across an 11-week study. Lipid-related variables explained approximately 4.5% of the variation in gut microbiome compositions, with higher effects observed for total cholesterol and high-density lipoproteins. Species from the genera Odoribacter, Anaerostipes, and Parabacteroides correlated with increased serum lipid levels, whereas probiotic species like Akkermansia muciniphila were more abundant among participants with healthier blood lipid profiles. An inverse correlation with serum cholesterol was also observed for Massilistercora timonensis, a player in regulating lipid turnover. The observed correlation patterns add to the growing evidence supporting the role of the gut microbiome as an essential regulator of host lipid metabolism.
Collapse
Affiliation(s)
- Ineta Kalnina
- Latvian Biomedical Research and Study Centre 1, LV-1067 Riga, Latvia
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Balmant BD, Fonseca DC, Rocha IM, Callado L, Torrinhas RSMDM, Waitzberg DL. Dys-R Questionnaire: A Novel Screening Tool for Dysbiosis Linked to Impaired Gut Microbiota Richness. Nutrients 2023; 15:4261. [PMID: 37836545 PMCID: PMC10574031 DOI: 10.3390/nu15194261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Practical and affordable tools to screen intestinal dysbiosis are needed to support clinical decision making. Our study aimed to design a new subjective screening tool for the risk of intestinal dysbiosis from a previously described nonvalidated questionnaire (DYS/FQM) and based on subjective and objective data. A total of 219 individuals comprised the chronic diseases (CD; n = 167) and healthy control (HC; 52 subjects) groups. Sociodemographic, anthropometric, body composition, lifestyle, past history, intestinal health, and dietary data were collected. The gut microbiota (GM) profile was assessed from fecal samples using the 16S rRNA sequencing. Scores for the new tool (Dys-R Questionnaire) were assigned using discrete optimization techniques. The association between Dys-R scores and dysbiosis risk was assessed through correlation, simple linear models, sensitivity, specificity, as well as positive and negative predictive values. We found significant differences in the Chao1 Index between CD and HC groups (adjusted p-value = 0.029), highlighting lower GM richness as the primary marker for intestinal dysbiosis. DYS/FQM showed poor performance in identifying poor GM richness. Dys-R exhibited a 42% sensitivity, 82% specificity, 79% positive predictive value (PPV), and 55% negative predictive value (NPV) to identify poor GM richness. The new Dys-R questionnaire showed good performance in ruling out dysbiosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Dan Linetzky Waitzberg
- Laboratory of Nutrition and Metabolic Surgery of the Digestive System, LIM 35, Department of Gastroenterology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246 903, Brazil; (B.D.B.)
| |
Collapse
|