1
|
Romeo S, Vidal-Puig A, Husain M, Ahima R, Arca M, Bhatt DL, Diehl AM, Fontana L, Foo R, Frühbeck G, Kozlitina J, Lonn E, Pattou F, Plat J, Quaggin SE, Ridker PM, Rydén M, Segata N, Tuttle KR, Verma S, Roeters van Lennep J, Benn M, Binder CJ, Jamialahmadi O, Perkins R, Catapano AL, Tokgözoğlu L, Ray KK. Clinical staging to guide management of metabolic disorders and their sequelae: a European Atherosclerosis Society consensus statement. Eur Heart J 2025:ehaf314. [PMID: 40331343 DOI: 10.1093/eurheartj/ehaf314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Obesity rates have surged since 1990 worldwide. This rise is paralleled by increases in pathological processes affecting organs such as the heart, liver, and kidneys, here termed systemic metabolic disorders (SMDs). For clinical management of SMD, the European Atherosclerosis Society proposes a pathophysiology-based system comprising three stages: Stage 1, where metabolic abnormalities such as dysfunctional adiposity and dyslipidaemia occur without detectable organ damage; Stage 2, which involves early organ damage manifested as Type 2 diabetes, asymptomatic diastolic dysfunction, metabolic-associated steatohepatitis (MASH), and chronic kidney disease (CKD); and Stage 3, characterized by more advanced organ damage affecting multiple organs. Various forms of high-risk obesity, driven by maintained positive energy balance, are the most common cause of SMD, leading to ectopic lipid accumulation and insulin resistance. This progression affects various organs, promoting comorbidities such as hypertension and atherogenic dyslipidaemia. Genetic factors influence SMD susceptibility, and ethnic disparities in SMD are attributable to genetic and socioeconomic factors. Key SMD features include insulin resistance, inflammation, pre-diabetes, Type 2 diabetes, MASH, hypertension, CKD, atherogenic dyslipidaemia, and heart failure. Management strategies involve lifestyle changes, pharmacotherapy, and metabolic surgery in severe cases, with emerging treatments focusing on genetic approaches. The staging system provides a structured approach to understanding and addressing the multi-faceted nature of SMD, which is crucial for improving health outcomes. Categorization of SMD abnormalities by presence and progression is aimed to improve awareness of a multi-system trait and encourage a tailored and global approach to treatment, ultimately aiming to reduce the burden of obesity-related comorbidities.
Collapse
Affiliation(s)
- Stefano Romeo
- Department of Medicine, H7 Medicin, Huddinge, H7 Endokrinologi och Diabetes Romeo, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital Huddinge, 141 57 Huddinge, Stockholm, Sweden
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Viale Europa, 88100 Catanzaro, Italy
| | - Antonio Vidal-Puig
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- Centro de Investigacion Principe Felipe, C/ d'Eduardo Primo Yufera, 3, 46012 Valencia, Spain
- Cambridge University Nanjing Centre of Technology and Innovation, No. 23, Rongyue Road, Jiangbei New Area, Nanjing, Jiangsu, China
| | - Mansoor Husain
- Ted Rogers Centre for Heart Research, Department of Medicine, University of Toronto, 661 University Avenue, Toronto, ON, Canada M5G 1M1
| | - Rexford Ahima
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
- Unit of Internal Medicine and Metabolic Diseases, Hospital Policlinico Umberto I, Rome, Italy
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, NC, USA
| | - Luigi Fontana
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Roger Foo
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, National University Health Systems, Singapore
- Cardiovascular Metabolic Disease Translational Research Programme, National University Health Systems, Singapore
| | - Gema Frühbeck
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Metabolic Research Laboratory, CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Julia Kozlitina
- The Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eva Lonn
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Francois Pattou
- Department of Endocrine and Metabolic Surgery, CHU Lille, University of Lille, Inserm, Institut Pasteur Lille, Lille, France
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, NUTRIM School of Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Susan E Quaggin
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Nephrology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Katherine R Tuttle
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
- Providence Medical Research Center, Providence Inland Northwest Health, Spokane, WA, USA
| | - Subodh Verma
- Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Jeanine Roeters van Lennep
- Department of Internal Medicine, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marianne Benn
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Centre of Diagnostic Investigation, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Alberico L Catapano
- Center for the Study of Atherosclerosis, IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK
| |
Collapse
|
2
|
Guasch-Ferré M, Wittenbecher C, Palmnäs M, Ben-Yacov O, Blaak EE, Dahm CC, Fall T, Heitmann BL, Licht TR, Löf M, Loos R, Patel CJ, Quarta C, Redman LM, Segal E, Segata N, Snyder M, Sun Q, Tobias DK, Hu FB, Franks PW, Landberg R, Sargent JL, Merino J. Precision nutrition for cardiometabolic diseases. Nat Med 2025:10.1038/s41591-025-03669-9. [PMID: 40307513 DOI: 10.1038/s41591-025-03669-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/21/2025] [Indexed: 05/02/2025]
Abstract
Precision nutrition is a vibrant and rapidly evolving field of scientific research and innovation with the potential to deliver health, societal and economic benefits by improving healthcare delivery and policies. Advances in deep phenotyping technologies, digital tools and artificial intelligence have made possible early proof-of-concept research that expands the understanding of within- and between-person variability in responses to diet. These studies illustrate the promise of precision nutrition to complement the traditional 'one size fits all' dietary guidelines, which, while considering broad life-stage and disease-specific nutritional requirements, often lack the granularity to account fully for individual variations in nutritional needs and dietary responses. Despite these developments, however, considerable challenges remain before precision nutrition can be implemented on a broader scale. This Review examines the current state of precision nutrition research, with a focus on its application to reducing the incidence and burden of cardiometabolic diseases. We critically examine the evidence base, explore the potential benefits and discuss the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Marta Guasch-Ferré
- Section of Epidemiology, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Marie Palmnäs
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Orly Ben-Yacov
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ellen E Blaak
- Department of Human Biology, Maastricht University, Maastricht, the Netherlands
- NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Christina C Dahm
- Research Unit for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Berit L Heitmann
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, The Capital Region, Denmark
- Section for General Medicine, The Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- The Boden Initiative, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Marie Löf
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Ruth Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | | | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Deirdre K Tobias
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paul W Franks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Rikard Landberg
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Jennifer L Sargent
- School of Public Health, Imperial College, London, UK
- BabelFisk, Helsingborg, Sweden
| | - Jordi Merino
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Diabetes Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA, USA.
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Liu S, Lin Z, Huang Z, Yu M, Lin Z, Hu Z. Unique Microbial Characterisation of Oesophageal Squamous Cell Carcinoma Patients with Different Dietary Habits Based on Light Gradient Boosting Machine Learning Classifier. Nutrients 2025; 17:1340. [PMID: 40284204 PMCID: PMC12030675 DOI: 10.3390/nu17081340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Objectives: The microbiome plays an important role in cancer, but the relationship between dietary habits and the microbiota in oesophageal squamous cell carcinoma (ESCC) is not clear. The aim of this study is to explore the complex relationship between the microbiota in oesophagal tissue and dietary habits in ESCC patients. Methods: 173 ESCC patients were included. The method of 16S rRNA sequencing was used to analyze microbial composition and diversity. The LEfSe and Boruta methods were used to screen important microbes, and the LightGBM algorithm distinguished microbes associated with different dietary habits. PICRUST2 and DESeq2 predicted microbial function and screened differential functions. The Pearson test was used to analyze correlations between microbes and functions, and SPARCC microbial symbiotic networks and Cytoscape were used to determine microbial interactions. Results: Significant differences in microbial composition were observed among ESCC patients with different dietary habits. LEfSe and Boruta identified three, six, and two significantly different bacteria in the FF/FP, FF/PF, and FF/PP groups, respectively, with AUC values of 0.683, 0.830, and 0.715. PICRUST2 and DESeq2 analysis revealed 3, 11, and 5 significantly different metabolic pathways in each group. Eubacterium_B sulci was positively correlated with PWY-6285, PWY-3801, and PWY-5823. PWY-6397 was positively correlated with undefinded (Fusobacterium_C). Microbial network analysis confirmed unique microbial characteristics in different diet groups. Conclusions: Different dietary habits lead to alterations in Eubacterium_B sulci and undefinded (Fusobacterium_C) and related functional pathways.
Collapse
Affiliation(s)
- Shun Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Zhifeng Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Zhimin Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Menglin Yu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Zheng Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
4
|
Wu H, Lv B, Zhi L, Shao Y, Liu X, Mitteregger M, Chakaroun R, Tremaroli V, Hazen SL, Wang R, Bergström G, Bäckhed F. Microbiome-metabolome dynamics associated with impaired glucose control and responses to lifestyle changes. Nat Med 2025:10.1038/s41591-025-03642-6. [PMID: 40200054 DOI: 10.1038/s41591-025-03642-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025]
Abstract
Type 2 diabetes (T2D) is a complex disease shaped by genetic and environmental factors, including the gut microbiome. Recent research revealed pathophysiological heterogeneity and distinct subgroups in both T2D and prediabetes, prompting exploration of personalized risk factors. Using metabolomics in two Swedish cohorts (n = 1,167), we identified over 500 blood metabolites associated with impaired glucose control, with approximately one-third linked to an altered gut microbiome. Our findings identified metabolic disruptions in microbiome-metabolome dynamics as potential mediators of compromised glucose homeostasis, as illustrated by the potential interactions between Hominifimenecus microfluidus and Blautia wexlerae via hippurate. Short-term lifestyle changes, for example, diet and exercise, modulated microbiome-associated metabolites in a lifestyle-specific manner. This study suggests that the microbiome-metabolome axis is a modifiable target for T2D management, with optimal health benefits achievable through a combination of lifestyle modifications.
Collapse
Affiliation(s)
- Hao Wu
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China.
| | - Bomin Lv
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Luqian Zhi
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yikai Shao
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Xinyan Liu
- Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, and State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Matthias Mitteregger
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rima Chakaroun
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland, OH, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ru Wang
- School of Kinesiology, Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Göran Bergström
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
5
|
Deng K, Wang L, Nguyen SM, Shrubsole MJ, Cai Q, Lipworth L, Gupta DK, Zheng W, Shu XO, Yu D. A dietary pattern promoting gut sulfur metabolism is associated with increased mortality and altered circulating metabolites in low-income American adults. EBioMedicine 2025; 115:105690. [PMID: 40188743 PMCID: PMC12001102 DOI: 10.1016/j.ebiom.2025.105690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/12/2025] [Accepted: 03/26/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Excessive hydrogen sulfide in the gut, generated by sulfur-metabolising bacteria from foods, has been linked to intestinal inflammation and human diseases. We aim to investigate the interplay between diet and sulphur-metabolising bacteria in relation to mortality and circulating metabolites in understudied populations. METHODS In the Southern Community Cohort Study (SCCS), a prospective cohort of primarily low-income American adults, habitual diets were assessed using a food frequency questionnaire at baseline (2002-2009). A sulfur microbial diet score (SMDS) was developed among 514 Black/African American participants by linking habitual dietary intakes with the abundance of sulfur-metabolising bacteria profiled by faecal shotgun metagenomics. The SMDS was then constructed among all eligible SCCS participants (50,114 Black/African American and 23,923 non-Hispanic White adults), and its associations with mortality outcomes were examined by Cox proportional hazards model and Fine-Grey subdistribution hazard model. The association between SMDS and 1110 circulating metabolites was examined by linear regression among 1688 SCCS participants with untargeted metabolomic profiling of baseline plasma samples. FINDINGS Over an average 13.9-year follow-up, SMDS was associated with increased all-cause mortality (HR [95% CI] for the highest vs. lowest quartiles: 1.21 [1.15-1.27]) and cardiovascular disease (1.18 [1.08-1.29]), cancer (1.13 [1.02-1.25]), and gastrointestinal cancer-specific (1.22 [1.00-1.49]) mortality among Black/African American participants (all P-trend<0.05). The associations were largely consistent across participant subgroups. Similar results were observed among non-Hispanic White participants. The SMDS was associated with 112 circulating metabolites, which mediated 36.15% of the SMDS-mortality association (P = 0.002). INTERPRETATION A dietary pattern promoting sulfur-metabolising gut bacteria may contribute to increased total and disease mortality in low-income American adults. FUNDING This study was funded by the National Institutes of Health, United States, to Vanderbilt University Medical Center, United States, and Anne Potter Wilson Chair endowment to Vanderbilt University, United States.
Collapse
Affiliation(s)
- Kui Deng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Lei Wang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Sang Minh Nguyen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Martha J Shrubsole
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA; International Epidemiology Field Station, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Loren Lipworth
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Deepak K Gupta
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt Translational and Clinical Cardiovascular Research Centre, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA
| | - Danxia Yu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Centre, Vanderbilt University Medical Centre, Nashville, TN, USA.
| |
Collapse
|
6
|
Lin Y, Hou J, Li B, Shu W, Wan J. Advancements in Nanomaterials and Molecular Probes for Spatial Omics. ACS NANO 2025; 19:11604-11624. [PMID: 40125910 DOI: 10.1021/acsnano.4c18470] [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: 03/25/2025]
Abstract
Spatial omics is emerging as a focus of life sciences because of its applications in investigating the molecular mechanisms of cancer, mapping cellular distributions, and revealing specific cellular ecological niches. Notably, the in-depth acquisition of spatial omics information relies on highly sensitive, high-resolution, and high-throughput biological analysis tools and techniques. However, conventional methods of omics data acquisition still suffer from some drawbacks such as limited-resolution and low-throughput and are difficult to adapt directly to the collection of high-quality spatial omics data. Recently, an increasing number of advanced nanomaterials and molecular probes are employed in spatial omics due to their excellent optoelectronic properties, biocompatibility, and multifunction. These well-designed innovative nanoscaffolds successfully enhance the key parameters of spatial omics and, thus, increase the spatial resolution, detection sensitivity, and detection throughput. This review summarizes the design and application of functional nanoscaffolds for spatial omics in recent years, with a particular emphasis on nanomaterials and molecular probes. We believe that the present review can inspire and motivate researchers in designing and selecting appropriate materials and probes for high-quality spatial omics, thus promoting the development of spatial omics and life sciences.
Collapse
Affiliation(s)
- Yingying Lin
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Jiaxin Hou
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Bin Li
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Weikang Shu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| |
Collapse
|
7
|
Sun J, Geng L, Zhou D, Teng X, Chen M. Gut microbiota participates in polystyrene microplastics-induced defective implantation through impairing uterine receptivity. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 380:124997. [PMID: 40101486 DOI: 10.1016/j.jenvman.2025.124997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/22/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025]
Abstract
Microplastics (MPs) are widespread in global ecosystems and could pose risks to human health. However, crucial information on the impact of MP exposure on female reproductive health remains insufficient. In this study, we constructed an MP-exposure mice model through oral administration of polystyrene microplastics (PS-MPs) and found that it resulted in impaired uterine receptivity and defective implantation. An accumulation of plastic particles was detected in MP mice intestines. Metagenomic sequencing of feces samples indicated a structural and functional alteration of gut microbiota. Alistipes played a prominent role in MP biodegradation, while among the biodegradable functional genes, ACSL made the greatest contribution. Both had a significant increase in MP group, suggesting a potential occurrence of ferroptosis. Ferroptosis, a form of programmed cell death, is closely associated with uterine receptivity impairment and defective implantation. We detected MDA contents and ferroptosis-related proteins, and the results indicated the activation of ferroptosis in the process. Our research is the first to elucidate that exposure to MPs impairs uterine receptivity and results in deficient implantation, while also providing initial evidence that gut microbiota plays a critical role in this process.
Collapse
Affiliation(s)
- Jiani Sun
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Lulu Geng
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Dan Zhou
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoming Teng
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Miaoxin Chen
- Centre for Assisted Reproduction, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
8
|
Li Y, Lyu L, Ding H. The potential roles of gut microbiome in porto-sinusoidal vascular disease: an under-researched crossroad. Front Microbiol 2025; 16:1556667. [PMID: 40099185 PMCID: PMC11911366 DOI: 10.3389/fmicb.2025.1556667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Accumulating evidence indicates that patients with liver diseases exhibit distinct microbiological profiles, which can be attributed to the bidirectional relationship of the gut-liver axis. Porto-sinusoidal vascular disease (PSVD) has recently been introduced to describe a group of vascular diseases of the liver, involving the portal venules and sinusoids. Although the pathophysiology of PSVD is not yet fully understood, several predisposing conditions, including immunodeficiency, inflammatory bowel disease, abdominal bacterial infections are associated with the increasing in intestinal permeability and microbial translocation, supporting the role of altered gut microbiota and gut-derived endotoxins in PSVD etiopathogenesis. Recent studies have proposed that the gut microbiome may play a crucial role in the pathophysiology of intrahepatic vascular lesions, potentially influencing the onset and progression of PSVD in this context. This review aims to summarize the current understanding of the gut microbiome's potential role in the pathogenesis of hepatic microvascular abnormalities and thrombosis, and to briefly describe their interactions with PSVD. The insights into gut microbiota and their potential influence on the onset and progression of PSVD may pave the way for new diagnostic, prognostic, and therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing Youan Hospital Affiliated with Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Kirk D, Louca P, Attaye I, Zhang X, Wong KE, Michelotti GA, Falchi M, Valdes AM, Williams FMK, Menni C. Multifluid Metabolomics Identifies Novel Biomarkers for Irritable Bowel Syndrome. Metabolites 2025; 15:121. [PMID: 39997746 PMCID: PMC11857683 DOI: 10.3390/metabo15020121] [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: 11/15/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Background/Objectives: Irritable bowel syndrome (IBS) is a complex disorder affecting 10% of the global population, but the underlying mechanisms remain poorly understood. By integrating multifluid metabolomics, we aimed to identify metabolite markers of IBS in a large population-based cohort. Methods: We included individuals from TwinsUK with and without IBS, ascertained using the Rome III criteria, and analysed serum (232 cases, 1707 controls), urine (185 cases, 1341 controls), and stool (186 cases, 1284 controls) metabolites (Metabolon Inc.). Results: After adjusting for covariates, and multiple testing, 44 unique metabolites (25 novel) were associated with IBS, including lipids, amino acids, and xenobiotics. Androsterone sulphate, a sulfated steroid hormone precursor, was associated with lower odds of IBS in both urine (0.69 [95% confidence interval = 0.56-0.85], p = 2.34 × 10-4) and serum (0.75 [0.63-0.90], p = 1.54 × 10-3. Moreover, suberate (C8-DC) was associated with higher odds of IBS in serum (1.36 [1.15-1.61]; p = 1.84 × 10-4) and lower odds of IBS in stool (0.76 [0.63-0.91]; p = 2.30 × 10-3). On the contrary, 32 metabolites appeared to be fluid-specific, including indole, 13-HODE + 9-HODE, pterin, bilirubin (E,Z or Z,Z), and urolithin. The remaining 10 metabolites were associated with IBS in one fluid with suggestive evidence (p < 0.05) in another fluid. Finally, we identified androgenic signalling, dicarboxylates, haemoglobin, and porphyrin metabolism to be significantly over-represented in individuals with IBS compared to controls. Conclusions: Our results highlight the utility of a multi-fluid approach in IBS research, revealing distinct metabolic signatures across biofluids.
Collapse
Affiliation(s)
- Daniel Kirk
- Department of Twin Research & Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (D.K.); (P.L.); (I.A.); (X.Z.); (M.F.); (A.M.V.); (F.M.K.W.)
| | - Panayiotis Louca
- Department of Twin Research & Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (D.K.); (P.L.); (I.A.); (X.Z.); (M.F.); (A.M.V.); (F.M.K.W.)
| | - Ilias Attaye
- Department of Twin Research & Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (D.K.); (P.L.); (I.A.); (X.Z.); (M.F.); (A.M.V.); (F.M.K.W.)
- Amsterdam Cardiovascular Sciences, Diabetes & Metabolism, 1105 AZ Amsterdam, The Netherlands
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Xinyuan Zhang
- Department of Twin Research & Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (D.K.); (P.L.); (I.A.); (X.Z.); (M.F.); (A.M.V.); (F.M.K.W.)
| | - Kari E. Wong
- Metabolon Inc., Research Triangle Park, Morrisville, NC 27560, USA; (K.E.W.); (G.A.M.)
| | - Gregory A. Michelotti
- Metabolon Inc., Research Triangle Park, Morrisville, NC 27560, USA; (K.E.W.); (G.A.M.)
| | - Mario Falchi
- Department of Twin Research & Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (D.K.); (P.L.); (I.A.); (X.Z.); (M.F.); (A.M.V.); (F.M.K.W.)
| | - Ana M. Valdes
- Department of Twin Research & Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (D.K.); (P.L.); (I.A.); (X.Z.); (M.F.); (A.M.V.); (F.M.K.W.)
- Nottingham NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
- Inflammation, Recovery and Injury Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Frances M. K. Williams
- Department of Twin Research & Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (D.K.); (P.L.); (I.A.); (X.Z.); (M.F.); (A.M.V.); (F.M.K.W.)
| | - Cristina Menni
- Department of Twin Research & Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (D.K.); (P.L.); (I.A.); (X.Z.); (M.F.); (A.M.V.); (F.M.K.W.)
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
10
|
Marette A, Pilon G. Host-microbiome determinants of insulin resistance in obesity: alone we go faster, together we go further! Gut 2025:gutjnl-2024-333855. [PMID: 39933914 DOI: 10.1136/gutjnl-2024-333855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Affiliation(s)
- Andre Marette
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
- Department of Medicine and IUCPQ, Laval University, Quebec, Quebec, Canada
| | - Genevieve Pilon
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
| |
Collapse
|
11
|
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: 0] [Impact Index Per Article: 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
|
12
|
Battistelli A, Proietti S, Paglialunga G, Mattioni M, Nazzaro F, Fratianni F, Colla G, Cardarelli M, Del Bianco M, Moscatello S. Establishing baselines for prebiotic production in controlled environments for applications in space and vertical farming. Heliyon 2025; 11:e42112. [PMID: 39925353 PMCID: PMC11804542 DOI: 10.1016/j.heliyon.2025.e42112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/11/2025] Open
Abstract
Prebiotics might contribute to astronauts' health in space, yet their production in Bioregenerative Life Support Systems (BLSS) remains largely unexplored. Here, chicory (Cichorium intybus L.) exhibited the most appropriate characteristics among seven candidate species as the ideal crop for space prebiotic production systems. Furthermore, we evaluated chicory's performance under different light intensities (250 μmol m-2 s-1 and 500 μmol m-2 s-1 of photosynthetically active radiation) and growing cycle lengths (76 and 91 days after sowing) to optimize prebiotic production. Our findings reveal chicory's remarkable adaptability to fully controlled growing conditions and its ability to accumulate high levels of inulin in the young taproot, despite the short growing cycle. The length of the growing period influenced the productivity of biomass and inulin, the latter depending more on taproot biomass than on inulin content. A growing area of 6.3 m2 might be sufficient for a daily production of 12 g of inulin, the amount of "native chicory inulin" recognized by EFSA to promote health benefits in humans. These results establish important baselines for prebiotic production in BLSS or Controlled Environment Agriculture (CEA) and may be used to design fully controlled cropping systems for space and Earth based vertical farming facilities.
Collapse
Affiliation(s)
- Alberto Battistelli
- National Research Council of Italy (CNR), Research Institute on Terrestrial Ecosystems (IRET), Porano, Terni, Italy
| | - Simona Proietti
- National Research Council of Italy (CNR), Research Institute on Terrestrial Ecosystems (IRET), Porano, Terni, Italy
| | - Gabriele Paglialunga
- National Research Council of Italy (CNR), Research Institute on Terrestrial Ecosystems (IRET), Porano, Terni, Italy
- Department of Agriculture and Forest Sciences, University of Tuscia, 01100, Viterbo, Italy
| | - Michele Mattioni
- National Research Council of Italy (CNR), Research Institute on Terrestrial Ecosystems (IRET), Porano, Terni, Italy
| | - Filomena Nazzaro
- National Research Council of Italy (CNR), Institute of Food Sciences (ISA) Avellino, Italy
| | - Florinda Fratianni
- National Research Council of Italy (CNR), Institute of Food Sciences (ISA) Avellino, Italy
| | - Giuseppe Colla
- Department of Agriculture and Forest Sciences, University of Tuscia, 01100, Viterbo, Italy
| | - Mariateresa Cardarelli
- Department of Agriculture and Forest Sciences, University of Tuscia, 01100, Viterbo, Italy
| | | | - Stefano Moscatello
- National Research Council of Italy (CNR), Research Institute on Terrestrial Ecosystems (IRET), Porano, Terni, Italy
| |
Collapse
|
13
|
Lin Z, Zeng M, Sui Z, Wu Y, Tang X, Liu T. Moderate full-fat and low-fat yoghurt consumption correlates with reduced mortality risk: a large-scale prospective analysis. J Glob Health 2025; 15:04014. [PMID: 39820103 PMCID: PMC11737816 DOI: 10.7189/jogh.15.04014] [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] [Indexed: 01/19/2025] Open
Abstract
Background Yoghurt is a commonly consumed fermented food recommended by many guidelines. Yoghurt consumption can contribute to the intake of multiple nutrients and reduce the risk of several diseases. However, prospective evidence is limited on the associations between full/low-fat yoghurt consumption and mortality risk. In this prospective cohort study, we aimed to assess the dose-dependent associations between full/low-fat yoghurt intake and all-cause or cause-specific mortality. Methods We enrolled 186 168 participants from the UK Biobank who had joined the study between 2006 and 2010 and were followed up until 2022. We obtained data on self-reported intake of full/low-fat yoghurt and mortality from all causes and specific causes of death, including cancers and cardiovascular diseases (CVDs). We then used Cox proportional hazard models to calculate the hazard ratio (HR) and 95% confidence interval (CI) to evaluate the associations between full-fat and low-fat yoghurt intake and mortality. Lastly, we conducted subgroup and sensitivity analyses to examine the robustness of our findings. Results A total of 9402 deaths occurred during a mean follow-up of 13.4 years, including 1687 CVD-related and 5073 cancer-related deaths. Relative to non-consumers, the HRs (95% CIs) for all-cause mortality risk in participants consuming >0-50, 50-100, and >100 g of full-fat yoghurt a day were 0.82 (95% CI = 0.72, 0.93), 0.97 (95% CI = 0.86, 1.09), and 0.96 (95% CI = 0.84, 1.1) respectively. The corresponding HR estimates relative to non-consumers for participants consuming low-fat yoghurt were 0.88 (95% CI = 0.81, 0.95), 0.91 (95% CI = 0.85, 0.98), and 0.95 (95% CI = 0.89, 1.01), respectively. Subgroup analysis indicated women who had moderate consumption of full-fat yoghurt had lower all-cause mortality risk, while men consuming low-fat yoghurt had lower all-cause mortality risk. Conclusions Moderate consumption of full-fat and low-fat yoghurt was correlated with decreased all-cause mortality. Future cohort studies are warranted to verify the potential of adopting yoghurt consumption as part of a healthy diet to reduce mortality.
Collapse
Affiliation(s)
- Zhengjun Lin
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Zeng
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, China
| | - Zijian Sui
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, China
| | - Yanlin Wu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Metabolic Disease, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xianzhe Tang
- Department of Orthopedics, Chenzhou No.1 People's Hospital, Chenzhou, Hunan, China
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
Fackelmann G, Manghi P, Carlino N, Heidrich V, Piccinno G, Ricci L, Piperni E, Arrè A, Bakker E, Creedon AC, Francis L, Capdevila Pujol J, Davies R, Wolf J, Bermingham KM, Berry SE, Spector TD, Asnicar F, Segata N. Gut microbiome signatures of vegan, vegetarian and omnivore diets and associated health outcomes across 21,561 individuals. Nat Microbiol 2025; 10:41-52. [PMID: 39762435 PMCID: PMC11726441 DOI: 10.1038/s41564-024-01870-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 10/25/2024] [Indexed: 01/12/2025]
Abstract
As plant-based diets gain traction, interest in their impacts on the gut microbiome is growing. However, little is known about diet-pattern-specific metagenomic profiles across populations. Here we considered 21,561 individuals spanning 5 independent, multinational, human cohorts to map how differences in diet pattern (omnivore, vegetarian and vegan) are reflected in gut microbiomes. Microbial profiles distinguished these common diet patterns well (mean AUC = 0.85). Red meat was a strong driver of omnivore microbiomes, with corresponding signature microbes (for example, Ruminococcus torques, Bilophila wadsworthia and Alistipes putredinis) negatively correlated with host cardiometabolic health. Conversely, vegan signature microbes were correlated with favourable cardiometabolic markers and were enriched in omnivores consuming more plant-based foods. Diet-specific gut microbes partially overlapped with food microbiomes, especially with dairy microbes, for example, Streptococcus thermophilus, and typical soil microbes in vegans. The signatures of common western diet patterns can support future nutritional interventions and epidemiology.
Collapse
Affiliation(s)
- Gloria Fackelmann
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Paolo Manghi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Niccolò Carlino
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Vitor Heidrich
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Gianmarco Piccinno
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Liviana Ricci
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Elisa Piperni
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | | | | | | | | | | | | | | | | | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, London, UK
| | - Tim D Spector
- ZOE Ltd., London, UK
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
| | - Francesco Asnicar
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
- European Institute of Oncology, Scientific Institute for Research, Hospitalization and Healthcare, Milan, Italy.
| |
Collapse
|
15
|
Simbirtseva KY, O'Toole PW. Healthy and Unhealthy Aging and the Human Microbiome. Annu Rev Med 2025; 76:115-127. [PMID: 39531852 DOI: 10.1146/annurev-med-042423-042542] [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: 11/16/2024]
Abstract
An altered gut microbiome is a feature of many multifactorial diseases, and microbiome effects on host metabolism, immune function, and possibly neurological function are implicated. Increased biological age is accompanied by a change in the gut microbiome. However, age-related health loss does not occur uniformly across all subjects but rather depends on differential loss of gut commensals and gain of pathobionts. In this article, we summarize the known and possible effects of the gut microbiome on the hallmarks of aging and describe the most plausible mechanisms. Understanding and targeting these factors could lead to prolonging health span by rationally maintaining the gut microbiome.
Collapse
Affiliation(s)
- Kseniya Y Simbirtseva
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland;
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland;
| |
Collapse
|
16
|
Wang S, Jin Z, Wu B, Morris AJ, Deng P. Role of dietary and nutritional interventions in ceramide-associated diseases. J Lipid Res 2025; 66:100726. [PMID: 39667580 PMCID: PMC11754522 DOI: 10.1016/j.jlr.2024.100726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
Ceramides are important intermediates in sphingolipid metabolism and serve as signaling molecules with independent biological significance. Elevated cellular and circulating ceramide levels are consistently associated with pathological conditions including cardiometabolic diseases, neurological diseases, autoimmune diseases, and cancers. Although pharmacological inhibition of ceramide formation often protects against these diseases in animal models, pharmacological modulation of ceramides in humans remains impractical. Dietary interventions including the Mediterranean diet, lacto-ovo-vegetarian diet, calorie-restricted diet, restriction of dairy product consumption, and dietary supplementation with polyunsaturated fatty acids, dietary fibers, and polyphenols, all have beneficial effects on modulating ceramide levels. Mechanistic insights into these interventions are discussed. This article reviews the relationships between ceramides and disease pathogenesis, with a focus on dietary intervention as a viable strategy for lowering the concentration of circulating ceramides.
Collapse
Affiliation(s)
- Shengnan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Zihui Jin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Biyu Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Andrew J Morris
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Affairs Healthcare System, Little Rock, Arkansas, USA
| | - Pan Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
17
|
Lotankar M, Houttu N, Mokkala K, Laitinen K. Diet-Gut Microbiota Relations: Critical Appraisal of Evidence From Studies Using Metagenomics. Nutr Rev 2024:nuae192. [PMID: 39718602 DOI: 10.1093/nutrit/nuae192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
Diet may influence the gut microbiota and subsequently affect the host's health. Recent developments in methods analyzing the composition and function of the gut microbiota allow a deeper understanding of diet-gut microbiota relationships. A state-of-the-art methodology, shotgun metagenomics sequencing, offers a higher taxonomic resolution of the gut microbiota at the bacterial species and strain levels, and more accurate information regarding the functional potential of gut microbiota. Here, the available evidence on the relationship between diet and gut microbiota was critically reviewed, focusing on results emerging from recent metagenomics sequencing studies applied in randomized controlled trials and observational studies. The PubMed and Embase databases were used to search publications between January 2011 and September 2023. Thus far, the number of studies is limited, and the study designs and methods utilized have been variable. Nevertheless, the cumulative evidence from interventions relates to dietary fiber as a modifier of bacterial species, such as Anaerostipes hadrus and Faecalibacterium prausnitzii. Furthermore, observational studies have detected associations between different dietary patterns and food groups with certain microbial species. Utilization of metagenomics sequencing is becoming more common and will undoubtedly provide further insights into diet-gut microbiota relationships at the species level as well as their functional pathways in the near future. For reproducible results and to draw reliable conclusions across various studies on diet-gut microbiota relationships, there is a need for harmonization of the study designs and standardized ways of reporting.
Collapse
Affiliation(s)
- Mrunalini Lotankar
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Noora Houttu
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Kati Mokkala
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
- Nutrition and Food Research Center, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Kirsi Laitinen
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, 20520 Turku, Finland
- Nutrition and Food Research Center, Faculty of Medicine, University of Turku, 20520 Turku, Finland
- Department of Obstetrics and Gynecology, Turku University Hospital, Wellbeing Services County of Southwest Finland, 20520 Turku, Finland
| |
Collapse
|
18
|
Sankaran K, Kodikara S, Li JJ, Cao KAL. Semisynthetic simulation for microbiome data analysis. Brief Bioinform 2024; 26:bbaf051. [PMID: 39927858 PMCID: PMC11808806 DOI: 10.1093/bib/bbaf051] [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: 12/19/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
High-throughput sequencing data lie at the heart of modern microbiome research. Effective analysis of these data requires careful preprocessing, modeling, and interpretation to detect subtle signals and avoid spurious associations. In this review, we discuss how simulation can serve as a sandbox to test candidate approaches, creating a setting that mimics real data while providing ground truth. This is particularly valuable for power analysis, methods benchmarking, and reliability analysis. We explain the probability, multivariate analysis, and regression concepts behind modern simulators and how different implementations make trade-offs between generality, faithfulness, and controllability. Recognizing that all simulators only approximate reality, we review methods to evaluate how accurately they reflect key properties. We also present case studies demonstrating the value of simulation in differential abundance testing, dimensionality reduction, network analysis, and data integration. Code for these examples is available in an online tutorial (https://go.wisc.edu/8994yz) that can be easily adapted to new problem settings.
Collapse
Affiliation(s)
- Kris Sankaran
- Department of Statistics, University of Wisconsin-Madison, 1300 University Ave, Madison,WI 53703, United States
| | - Saritha Kodikara
- Melbourne Integrative Genomics, School of Mathematics and Statistics, University of Melbourne, Building 184/30 Royal Parade, Melbourne, VIC 3052, Australia
| | - Jingyi Jessica Li
- Department of Statistics and Data Science, University of California, Los Angeles, 520 Portola Plaza, Los Angeles, CA 90095, United States
- Department of Human Genetics, University of California, Los Angeles, 695 Charles E Young Dr S, Los Angeles, CA 90095, United States
- Department of Biostatistics, University of California, Los Angeles, 650 Charles E. Young Dr S, Los Angeles, CA 90095, United States
| | - Kim-Anh Lê Cao
- Melbourne Integrative Genomics, School of Mathematics and Statistics, University of Melbourne, Building 184/30 Royal Parade, Melbourne, VIC 3052, Australia
| |
Collapse
|
19
|
Chandra M. Developmental Origins of Non-Communicable Chronic Diseases: Role of Fetal Undernutrition and Gut Dysbiosis in Infancy. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1387. [PMID: 39594962 PMCID: PMC11592819 DOI: 10.3390/children11111387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024]
Abstract
There is an increasing prevalence of non-communicable chronic diseases (NCCDs) like obesity, metabolic syndrome, type 2 diabetes mellitus (T2DM), hypertension, allergic asthma, and neuro-developmental/psychiatric problems in many parts of the world. A suboptimal lifestyle as an adult is often blamed for the occurrence of NCCDs. This review discusses the developmental origin of health and disease theory and how suboptimal nutrition in intrauterine life and the establishment of a suboptimal gut microbiome during infancy can influence the predisposition to NCCDs.
Collapse
Affiliation(s)
- Manju Chandra
- Division of Pediatric Nephrology, Department of Pediatrics, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| |
Collapse
|
20
|
Mancin L, Paoli A, Berry S, Gonzalez JT, Collins AJ, Lizarraga MA, Mota JF, Nicola S, Rollo I. Standardization of gut microbiome analysis in sports. Cell Rep Med 2024; 5:101759. [PMID: 39368478 PMCID: PMC11514603 DOI: 10.1016/j.xcrm.2024.101759] [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: 05/14/2024] [Revised: 08/09/2024] [Accepted: 09/10/2024] [Indexed: 10/07/2024]
Abstract
The gut microbiome plays a significant role in physiological functions such as nutrient processing, vitamin production, inflammatory response, and immune modulation, which, in turn, are important contributors to athlete health and performance. To date, the interpretation, discussion, and visualization of microbiome results of athletes are challenging, due to a lack of standard parameters and reference data for collection and comparison. The purpose of this perspective piece is to provide researchers with an easy-to-understand framework for the collection, analysis, and data management related to the gut microbiome with a specific focus on athletic populations. In the absence of a consensus on microbiome research in the sports field, we hope that these considerations serve as foundational "best practice." Adherence to these standard operating procedures will accelerate the path toward improving the quality of data and ultimately our understanding of the influence of the gut microbiome in sport settings.
Collapse
Affiliation(s)
- Laura Mancin
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Human Inspired Technology Research Center HIT, University of Padua, Padua, Italy.
| | - Antonio Paoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Human Inspired Technology Research Center HIT, University of Padua, Padua, Italy
| | - Sara Berry
- Department of Nutritional Sciences, King's College London, London, UK
| | | | - Adam J Collins
- Department for Health, University of Bath, BA2 7AY Bath, UK
| | | | - Joao Felipe Mota
- APC Microbiome Ireland, Department of Medicine, School of Microbiology, University College Cork, T12 YT20 Cork, Ireland
| | - Segata Nicola
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Ian Rollo
- Gatorade Sports Science Institute, PepsiCo Life Sciences, Global R&D, Leicestershire, UK; School of Sports Exercise and Health Sciences, Loughborough University, Leicestershire, UK
| |
Collapse
|
21
|
Carlino N, Blanco-Míguez A, Punčochář M, Mengoni C, Pinto F, Tatti A, Manghi P, Armanini F, Avagliano M, Barcenilla C, Breselge S, Cabrera-Rubio R, Calvete-Torre I, Coakley M, Cobo-Díaz JF, De Filippis F, Dey H, Leech J, Klaassens ES, Knobloch S, O'Neil D, Quijada NM, Sabater C, Skírnisdóttir S, Valentino V, Walsh L, Alvarez-Ordóñez A, Asnicar F, Fackelmann G, Heidrich V, Margolles A, Marteinsson VT, Rota Stabelli O, Wagner M, Ercolini D, Cotter PD, Segata N, Pasolli E. Unexplored microbial diversity from 2,500 food metagenomes and links with the human microbiome. Cell 2024; 187:5775-5795.e15. [PMID: 39214080 DOI: 10.1016/j.cell.2024.07.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/17/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Complex microbiomes are part of the food we eat and influence our own microbiome, but their diversity remains largely unexplored. Here, we generated the open access curatedFoodMetagenomicData (cFMD) resource by integrating 1,950 newly sequenced and 583 public food metagenomes. We produced 10,899 metagenome-assembled genomes spanning 1,036 prokaryotic and 108 eukaryotic species-level genome bins (SGBs), including 320 previously undescribed taxa. Food SGBs displayed significant microbial diversity within and between food categories. Extension to >20,000 human metagenomes revealed that food SGBs accounted on average for 3% of the adult gut microbiome. Strain-level analysis highlighted potential instances of food-to-gut transmission and intestinal colonization (e.g., Lacticaseibacillus paracasei) as well as SGBs with divergent genomic structures in food and humans (e.g., Streptococcus gallolyticus and Limosilactobabillus mucosae). The cFMD expands our knowledge on food microbiomes, their role in shaping the human microbiome, and supports future uses of metagenomics for food quality, safety, and authentication.
Collapse
Affiliation(s)
- Niccolò Carlino
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Aitor Blanco-Míguez
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Michal Punčochář
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Claudia Mengoni
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Pinto
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Alessia Tatti
- Scuola Universitaria Superiore IUSS Pavia, Pavia, Italy; Centre for Agriculture Food Environment, University of Trento, Trento, Italy; Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Italy
| | - Paolo Manghi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Armanini
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Michele Avagliano
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy
| | - Coral Barcenilla
- Department of Food Hygiene and Technology, Universidad de León, León, Spain
| | - Samuel Breselge
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Raul Cabrera-Rubio
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; Department of Biotechnology, Institute of Agrochemistry and Food Technology - National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Inés Calvete-Torre
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias - Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain; Microhealth Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Mairéad Coakley
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - José F Cobo-Díaz
- Department of Food Hygiene and Technology, Universidad de León, León, Spain
| | - Francesca De Filippis
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Portici, Italy
| | - Hrituraj Dey
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - John Leech
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | | | | | | | - Narciso M Quijada
- Austrian Competence Centre for Feed and Food Quality, Safety, and Innovation, FFoQSI GmbH, Tulln an der Donau, Austria; Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria; Institute for Agribiotechnology Research (CIALE), Department of Microbiology and Genetics, University of Salamanca, Salamanca, Spain
| | - Carlos Sabater
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias - Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain; Microhealth Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | - Vincenzo Valentino
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy
| | - Liam Walsh
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; School of Microbiology, University College Cork, Cork, Ireland
| | | | - Francesco Asnicar
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Gloria Fackelmann
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Vitor Heidrich
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias - Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Spain; Microhealth Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Viggó Thór Marteinsson
- Microbiology Research Group, Matís, Reykjavík, Iceland; University of Iceland, Faculty of Food Science and Nutrition, Reykjavík, Iceland
| | - Omar Rota Stabelli
- Centre for Agriculture Food Environment, University of Trento, Trento, Italy; Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Italy
| | - Martin Wagner
- Austrian Competence Centre for Feed and Food Quality, Safety, and Innovation, FFoQSI GmbH, Tulln an der Donau, Austria; Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Danilo Ercolini
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Portici, Italy
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; VistaMilk SFI Research Centre, Teagasc, Moorepark, Fermoy, Co. Cork, Ireland
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy; Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| | - Edoardo Pasolli
- Department of Agricultural Sciences, Division of Microbiology, University of Naples Federico II, Portici, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Portici, Italy
| |
Collapse
|
22
|
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
|
23
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [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: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
24
|
Dinakis E, O'Donnell JA, Marques FZ. The gut-immune axis during hypertension and cardiovascular diseases. Acta Physiol (Oxf) 2024; 240:e14193. [PMID: 38899764 DOI: 10.1111/apha.14193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/04/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
The gut-immune axis is a relatively novel phenomenon that provides mechanistic links between the gut microbiome and the immune system. A growing body of evidence supports it is key in how the gut microbiome contributes to several diseases, including hypertension and cardiovascular diseases (CVDs). Evidence over the past decade supports a causal link of the gut microbiome in hypertension and its complications, including myocardial infarction, atherosclerosis, heart failure, and stroke. Perturbations in gut homeostasis such as dysbiosis (i.e., alterations in gut microbial composition) may trigger immune responses that lead to chronic low-grade inflammation and, ultimately, the development and progression of these conditions. This is unsurprising, as the gut harbors one of the largest numbers of immune cells in the body, yet is a phenomenon not entirely understood in the context of cardiometabolic disorders. In this review, we discuss the role of the gut microbiome, the immune system, and inflammation in the context of hypertension and CVD, and consolidate current evidence of this complex interplay, whilst highlighting gaps in the literature. We focus on diet as one of the major modulators of the gut microbiota, and explain key microbial-derived metabolites (e.g., short-chain fatty acids, trimethylamine N-oxide) as potential mediators of the communication between the gut and peripheral organs such as the heart, arteries, kidneys, and the brain via the immune system. Finally, we explore the dual role of both the gut microbiome and the immune system, and how they work together to not only contribute, but also mitigate hypertension and CVD.
Collapse
Affiliation(s)
- Evany Dinakis
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Kallapura G, Prakash AS, Sankaran K, Manjappa P, Chaudhary P, Ambhore S, Dhar D. Microbiota based personalized nutrition improves hyperglycaemia and hypertension parameters and reduces inflammation: a prospective, open label, controlled, randomized, comparative, proof of concept study. PeerJ 2024; 12:e17583. [PMID: 38948211 PMCID: PMC11214429 DOI: 10.7717/peerj.17583] [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: 01/19/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Background Recent studies suggest that gut microbiota composition, abundance and diversity can influence many chronic diseases such as type 2 diabetes. Modulating gut microbiota through targeted nutrition can provide beneficial effects leading to the concept of personalized nutrition for health improvement. In this prospective clinical trial, we evaluated the impact of a microbiome-based targeted personalized diet on hyperglycaemic and hyperlipidaemic individuals. Specifically, BugSpeaks®-a microbiome profile test that profiles microbiota using next generation sequencing and provides personalized nutritional recommendation based on the individual microbiota profile was evaluated. Methods A total of 30 participants with type 2 diabetes and hyperlipidaemia were recruited for this study. The microbiome profile of the 15 participants (test arm) was evaluated using whole genome shotgun metagenomics and personalized nutritional recommendations based on their microbiota profile were provided. The remaining 15 participants (control arm) were provided with diabetic nutritional guidance for 3 months. Clinical and anthropometric parameters such as HbA1c, systolic/diastolic pressure, c-reactive protein levels and microbiota composition were measured and compared during the study. Results The test arm (microbiome-based nutrition) showed a statistically significant decrease in HbA1c level from 8.30 (95% confidence interval (CI), [7.74-8.85]) to 6.67 (95% CI [6.2-7.05]), p < 0.001 after 90 days. The test arm also showed a 5% decline in the systolic pressure whereas the control arm showed a 7% increase. Incidentally, a sub-cohort of the test arm of patients with >130 mm Hg systolic pressure showed a statistically significant decrease of systolic pressure by 14%. Interestingly, CRP level was also found to drop by 19.5%. Alpha diversity measures showed a significant increase in Shannon diversity measure (p < 0.05), after the microbiome-based personalized dietary intervention. The intervention led to a minimum two-fold (Log2 fold change increase in species like Phascolarctobacterium succinatutens, Bifidobacterium angulatum, and Levilactobacillus brevis which might have a beneficial role in the current context and a similar decrease in species like Alistipes finegoldii, and Sutterella faecalis which have been earlier shown to have some negative effects in the host. Overall, the study indicated a net positive impact of the microbiota based personalized dietary regime on the gut microbiome and correlated clinical parameters.
Collapse
|
26
|
Bernatova I, Bartekova M. Molecular Aspects of Cardiometabolic Diseases: From Etiopathogenesis to Potential Therapeutic Targets. Int J Mol Sci 2024; 25:5841. [PMID: 38892029 PMCID: PMC11172306 DOI: 10.3390/ijms25115841] [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: 05/06/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiometabolic diseases (CMDs) encompass a range of prevalent, often preventable, non-communicable illnesses, including myocardial infarction, stroke, cardiac insufficiency, arterial hypertension, obesity, type 2 diabetes mellitus, insulin resistance, chronic renal dysfunction, non-alcoholic fatty liver disease, and rare metabolic disorders [...].
Collapse
Affiliation(s)
- Iveta Bernatova
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Sienkiewiczova 1, 81371 Bratislava, Slovakia
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 84104 Bratislava, Slovakia
| |
Collapse
|
27
|
Bosch TCG, Wigley M, Colomina B, Bohannan B, Meggers F, Amato KR, Azad MB, Blaser MJ, Brown K, Dominguez-Bello MG, Ehrlich SD, Elinav E, Finlay BB, Geddie K, Geva-Zatorsky N, Giles-Vernick T, Gros P, Guillemin K, Haraoui LP, Johnson E, Keck F, Lorimer J, McFall-Ngai MJ, Nichter M, Pettersson S, Poinar H, Rees T, Tropini C, Undurraga EA, Zhao L, Melby MK. The potential importance of the built-environment microbiome and its impact on human health. Proc Natl Acad Sci U S A 2024; 121:e2313971121. [PMID: 38662573 PMCID: PMC11098107 DOI: 10.1073/pnas.2313971121] [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: 05/18/2024] Open
Abstract
There is increasing evidence that interactions between microbes and their hosts not only play a role in determining health and disease but also in emotions, thought, and behavior. Built environments greatly influence microbiome exposures because of their built-in highly specific microbiomes coproduced with myriad metaorganisms including humans, pets, plants, rodents, and insects. Seemingly static built structures host complex ecologies of microorganisms that are only starting to be mapped. These microbial ecologies of built environments are directly and interdependently affected by social, spatial, and technological norms. Advances in technology have made these organisms visible and forced the scientific community and architects to rethink gene-environment and microbe interactions respectively. Thus, built environment design must consider the microbiome, and research involving host-microbiome interaction must consider the built-environment. This paradigm shift becomes increasingly important as evidence grows that contemporary built environments are steadily reducing the microbial diversity essential for human health, well-being, and resilience while accelerating the symptoms of human chronic diseases including environmental allergies, and other more life-altering diseases. New models of design are required to balance maximizing exposure to microbial diversity while minimizing exposure to human-associated diseases. Sustained trans-disciplinary research across time (evolutionary, historical, and generational) and space (cultural and geographical) is needed to develop experimental design protocols that address multigenerational multispecies health and health equity in built environments.
Collapse
Affiliation(s)
- Thomas C. G. Bosch
- Zoological Institute, University of Kiel, Kiel24118, Germany
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
| | - Mark Wigley
- Graduate School of Architecture, Planning and Preservation, Columbia University, New York, NY10027
| | - Beatriz Colomina
- School of Architecture, Princeton University, Princeton, NJ08544
| | - Brendan Bohannan
- The Institute of Ecology and Evolution, University of Oregon, Eugene, OR97403-5289
| | - Forrest Meggers
- Princeton University School of Architecture & Andlinger Center for Energy and the Environment, Princeton, NJ08540
| | - Katherine R. Amato
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Anthropology, Northwestern University, Evanston, IL60208
| | - Meghan B. Azad
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 0Z3, Canada
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
| | - Martin J. Blaser
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MBR3E 3P4, Canada
- Center for Advanced Biotechnology and Medicine at Rutgers Biomedical and Health Sciences, Rutgers University, Piscataway, NJ08854-8021
| | - Kate Brown
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Program in Science, Technology and Society, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Maria Gloria Dominguez-Bello
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ08901
- Department of Anthropology, Rutgers University, New Brunswick, NJ08901
| | - Stanislav Dusko Ehrlich
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Institute of Neurology, University College London, LondonWC1N 3RX, United Kingdom
| | - Eran Elinav
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Systems Immunology Department, Weizmann Institute of Science, Rehovot761000, Israel
- Division of Microbiome & Cancer, Deutsches Krebsforschungszentrum, 69120Heidelberg, Germany
| | - B. Brett Finlay
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BCV6T 1Z4, Canada
| | - Kate Geddie
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Medical and Related Sciences Centre, The Canadian Institute for Advanced Research, Toronto, ONM5G 1L7, Canada
| | - Naama Geva-Zatorsky
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Technion Integrated Cancer Center, Technion-Israel Institute of Technology, Haifa3525433, Israel
- Department of Cell Biology and Cancer Science, Technion-Israel Institute of Technology, Haifa3525433, Israel
| | - Tamara Giles-Vernick
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Anthropology & Ecology of Disease Emergence, Institut Pasteur, Université Paris Cité, Paris75015, France
| | - Philippe Gros
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3G 1Y6, Canada
| | - Karen Guillemin
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Institute of Molecular Biology, University of Oregon, Eugene, OR97403
| | - Louis-Patrick Haraoui
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, CanadaJ1E 4K8
| | - Elizabeth Johnson
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- College of Human Ecology, Cornell University, IthakaNY14853
| | - Frédéric Keck
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Laboratoire d’Anthropologie Sociale, Collège de France, Paris75005, France
| | - Jamie Lorimer
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- School of Geography and the Environment, University of Oxford, OX1 3QY, United Kingdom
| | - Margaret J. McFall-Ngai
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA91125
| | - Mark Nichter
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- School of Anthropology, University of Arizona, Tucson, AZ85721
| | - Sven Pettersson
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Nanyang Technological University, Singapore637715, Singapore
| | - Hendrik Poinar
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Anthropology, McMaster University, Hamilton, ONL8S 4M4, Canada
| | - Tobias Rees
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- LIMN, Berkeley, CA94708
| | - Carolina Tropini
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Microbiology and Immunology and School of Biomedical Engineering, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| | - Eduardo A. Undurraga
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Escuela de Gobierno, Pontificia Universidad Católica de Chile, Santiago7820436, Chile
| | - Liping Zhao
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ08901
| | - Melissa K. Melby
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ONM5G 1M1, Canada
- Department of Anthropology, University of Delaware, Newark, DE19716
| |
Collapse
|
28
|
Bijla M, Saini SK, Pathak AK, Bharadwaj KP, Sukhavasi K, Patil A, Saini D, Yadav R, Singh S, Leeuwenburgh C, Kumar P. Microbiome interactions with different risk factors in development of myocardial infarction. Exp Gerontol 2024; 189:112409. [PMID: 38522483 DOI: 10.1016/j.exger.2024.112409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/10/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Among all non-communicable diseases, Cardiovascular Diseases (CVDs) stand as the leading global cause of mortality. Within this spectrum, Myocardial Infarction (MI) strikingly accounts for over 15 % of all deaths. The intricate web of risk factors for MI, comprising family history, tobacco use, oral health, hypertension, nutritional pattern, and microbial infections, is firmly influenced by the human gut and oral microbiota, their diversity, richness, and dysbiosis, along with their respective metabolites. Host genetic factors, especially allelic variations in signaling and inflammatory markers, greatly affect the progression or severity of the disease. Despite the established significance of the human microbiome-nutrient-metabolite interplay in associations with CVDs, the unexplored terrain of the gut-heart-oral axis has risen as a critical knowledge gap. Moreover, the pivotal role of the microbiome and the complex interplay with host genetics, compounded by age-related changes, emerges as an area of vital importance in the development of MI. In addition, a distinctive disease susceptibility and severity influenced by gender-based or ancestral differences, adds a crucial insights to the association with increased mortality. Here, we aimed to provide an overview on interactions of microbiome (oral and gut) with major risk factors (tobacco use, alcohol consumption, diet, hypertension host genetics, gender, and aging) in the development of MI and therapeutic regulation.
Collapse
Affiliation(s)
- Manisha Bijla
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, India
| | - Sunil Kumar Saini
- Department of Zoology, Swami Shraddhanand College, Delhi University, India
| | - Ajai Kumar Pathak
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia; Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | | | - Katyayani Sukhavasi
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital & Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
| | - Ayurshi Patil
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | - Diksha Saini
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | - Rakesh Yadav
- Department of Cardiology, AIIMS, New Delhi, India
| | - Shalini Singh
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | | | - Pramod Kumar
- ICMR-National Institute of Cancer Prevention and Research, Noida, India.
| |
Collapse
|
29
|
Tang H, Huang Y, Yuan D, Liu J. Atherosclerosis, gut microbiome, and exercise in a meta-omics perspective: a literature review. PeerJ 2024; 12:e17185. [PMID: 38584937 PMCID: PMC10999153 DOI: 10.7717/peerj.17185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Background Cardiovascular diseases are the leading cause of death worldwide, significantly impacting public health. Atherosclerotic cardiovascular diseases account for the majority of these deaths, with atherosclerosis marking the initial and most critical phase of their pathophysiological progression. There is a complex relationship between atherosclerosis, the gut microbiome's composition and function, and the potential mediating role of exercise. The adaptability of the gut microbiome and the feasibility of exercise interventions present novel opportunities for therapeutic and preventative approaches. Methodology We conducted a comprehensive literature review using professional databases such as PubMed and Web of Science. This review focuses on the application of meta-omics techniques, particularly metagenomics and metabolomics, in studying the effects of exercise interventions on the gut microbiome and atherosclerosis. Results Meta-omics technologies offer unparalleled capabilities to explore the intricate connections between exercise, the microbiome, the metabolome, and cardiometabolic health. This review highlights the advancements in metagenomics and metabolomics, their applications in research, and examines how exercise influences the gut microbiome. We delve into the mechanisms connecting these elements from a metabolic perspective. Metagenomics provides insight into changes in microbial strains post-exercise, while metabolomics sheds light on the shifts in metabolites. Together, these approaches offer a comprehensive understanding of how exercise impacts atherosclerosis through specific mechanisms. Conclusions Exercise significantly influences atherosclerosis, with the gut microbiome serving as a critical intermediary. Meta-omics technology holds substantial promise for investigating the gut microbiome; however, its methodologies require further refinement. Additionally, there is a pressing need for more extensive cohort studies to enhance our comprehension of the connection among these element.
Collapse
Affiliation(s)
- Haotian Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yanqing Huang
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Didi Yuan
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Junwen Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
30
|
Muse ED, Topol EJ. Transforming the cardiometabolic disease landscape: Multimodal AI-powered approaches in prevention and management. Cell Metab 2024; 36:670-683. [PMID: 38428435 PMCID: PMC10990799 DOI: 10.1016/j.cmet.2024.02.002] [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: 10/26/2023] [Revised: 01/25/2024] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
The rise of artificial intelligence (AI) has revolutionized various scientific fields, particularly in medicine, where it has enabled the modeling of complex relationships from massive datasets. Initially, AI algorithms focused on improved interpretation of diagnostic studies such as chest X-rays and electrocardiograms in addition to predicting patient outcomes and future disease onset. However, AI has evolved with the introduction of transformer models, allowing analysis of the diverse, multimodal data sources existing in medicine today. Multimodal AI holds great promise in more accurate disease risk assessment and stratification as well as optimizing the key driving factors in cardiometabolic disease: blood pressure, sleep, stress, glucose control, weight, nutrition, and physical activity. In this article we outline the current state of medical AI in cardiometabolic disease, highlighting the potential of multimodal AI to augment personalized prevention and treatment strategies in cardiometabolic disease.
Collapse
Affiliation(s)
- Evan D Muse
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA 92037, USA; Division of Cardiovascular Diseases, Scripps Clinic, La Jolla, CA 92037, USA
| | - Eric J Topol
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA 92037, USA; Division of Cardiovascular Diseases, Scripps Clinic, La Jolla, CA 92037, USA.
| |
Collapse
|
31
|
Kirk D, Costeira R, Visconti A, Khan Mirzaei M, Deng L, Valdes AM, Menni C. Bacteriophages, gut bacteria, and microbial pathways interplay in cardiometabolic health. Cell Rep 2024; 43:113728. [PMID: 38300802 PMCID: PMC11554570 DOI: 10.1016/j.celrep.2024.113728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/18/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
Cardiometabolic diseases are leading causes of mortality in Western countries. Well-established risk factors include host genetics, lifestyle, diet, and the gut microbiome. Moreover, gut bacterial communities and their activities can be altered by bacteriophages (also known simply as phages), bacteria-infecting viruses, making these biological entities key regulators of human cardiometabolic health. The manipulation of bacterial populations by phages enables the possibility of using phages in the treatment of cardiometabolic diseases through phage therapy and fecal viral transplants. First, however, a deeper understanding of the role of the phageome in cardiometabolic diseases is required. In this review, we first introduce the phageome as a component of the gut microbiome and discuss fecal viral transplants and phage therapy in relation to cardiometabolic diseases. We then summarize the current state of phageome research in cardiometabolic diseases and propose how the phageome might indirectly influence cardiometabolic health through gut bacteria and their metabolites.
Collapse
Affiliation(s)
- Daniel Kirk
- Department of Twin Research & Genetic Epidemiology, King's College London, St Thomas Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Ricardo Costeira
- Department of Twin Research & Genetic Epidemiology, King's College London, St Thomas Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Alessia Visconti
- Department of Twin Research & Genetic Epidemiology, King's College London, St Thomas Hospital, Westminster Bridge Road, London SE1 7EH, UK; Center for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Centre Munich - German Research Centre for Environmental Health, 85764 Neuherberg, Germany; School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Li Deng
- Institute of Virology, Helmholtz Centre Munich - German Research Centre for Environmental Health, 85764 Neuherberg, Germany; School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Ana M Valdes
- Academic Rheumatology, Clinical Sciences Building, Nottingham City Hospital, University of Nottingham, Nottingham, UK
| | - Cristina Menni
- Department of Twin Research & Genetic Epidemiology, King's College London, St Thomas Hospital, Westminster Bridge Road, London SE1 7EH, UK.
| |
Collapse
|
32
|
Raju SC, Molinaro A, Awoyemi A, Jørgensen SF, Braadland PR, Nendl A, Seljeflot I, Ueland PM, McCann A, Aukrust P, Vestad B, Mayerhofer C, Broch K, Gullestad L, Lappegård KT, Halvorsen B, Kristiansen K, Hov JR, Trøseid M. Microbial-derived imidazole propionate links the heart failure-associated microbiome alterations to disease severity. Genome Med 2024; 16:27. [PMID: 38331891 PMCID: PMC10854170 DOI: 10.1186/s13073-024-01296-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Interactions between the gut microbiota, diet, and host metabolism contribute to the development of cardiovascular disease, but a firm link between disease-specific gut microbiota alterations and circulating metabolites is lacking. METHODS We performed shot-gun sequencing on 235 samples from 166 HF patients and 69 healthy control samples. Separate plasma samples from healthy controls (n = 53) were used for the comparison of imidazole propionate (ImP) levels. Taxonomy and functional pathways for shotgun sequencing data was assigned using MetaPhlAn3 and HUMAnN3 pipelines. RESULTS Here, we show that heart failure (HF) is associated with a specific compositional and functional shift of the gut microbiota that is linked to circulating levels of the microbial histidine-derived metabolite ImP. Circulating ImP levels are elevated in chronic HF patients compared to controls and associated with HF-related gut microbiota alterations. Contrary to the microbiota composition, ImP levels provide insight into etiology and severity of HF and also associate with markers of intestinal permeability and systemic inflammation. CONCLUSIONS Our findings establish a connection between changes in the gut microbiota, the presence, etiology, and severity of HF, and the gut-microbially produced metabolite ImP. While ImP appears promising as a circulating biomarker reflecting gut dysbiosis related to HF, further studies are essential to demonstrate its causal or contributing role in HF pathogenesis. TRIAL REGISTRATION NCT02637167, registered December 22, 2015.
Collapse
Affiliation(s)
- Sajan C Raju
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Antonio Molinaro
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ayodeji Awoyemi
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Silje F Jørgensen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Peder R Braadland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andraz Nendl
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Ingebjørg Seljeflot
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | | | | | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Beate Vestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Cristiane Mayerhofer
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Knut T Lappegård
- Division of Internal Medicine, Nordlandssykehuset, 8005, Bodø, Norway
- Institute of Clinical Medicine, University of Tromsø, 9037, Tromsø, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen, 518083, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Johannes R Hov
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| |
Collapse
|
33
|
Siddiqui R, Qaisar R, Al-Dahash K, Altelly AH, Elmoselhi AB, Khan NA. Cardiovascular changes under the microgravity environment and the gut microbiome. LIFE SCIENCES IN SPACE RESEARCH 2024; 40:89-96. [PMID: 38245353 DOI: 10.1016/j.lssr.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 01/22/2024]
Abstract
In view of the critical role the gut microbiome plays in human health, it has become clear that astronauts' gut microbiota composition changes after spending time in space. Astronauts are exposed to several risks in space, including a protracted period of microgravity, radiation, and mechanical unloading of the body. Several deleterious effects of such an environment are reported, including orthostatic intolerance, cardiovascular endothelial dysfunction, cellular and molecular changes, and changes in the composition of the gut microbiome. Herein, the correlation between the gut microbiome and cardiovascular disease in a microgravity environment is evaluated. Additionally, the relationship between orthostatic hypotension, cardiac shrinkage and arrhythmias during spaceflight, and cellular alterations during spaceflight is reviewed. Given its impact on human health in general, modifying the gut microbiota may significantly promote astronaut health and performance. This is merited, given the prospect of augmented human activities in future space missions.
Collapse
Affiliation(s)
- Ruqaiyyah Siddiqui
- Microbiota Research Center, Istinye University, Istanbul 34010, Turkey; College of Arts and Sciences, American University of Sharjah, University City, Sharjah 26666, United Arab Emirates
| | - Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Khulood Al-Dahash
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahmad Hashem Altelly
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Adel B Elmoselhi
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Naveed Ahmed Khan
- Microbiota Research Center, Istinye University, Istanbul 34010, Turkey.
| |
Collapse
|
34
|
You M, Tian M, Song Z, Liu Z, Yang B, Zhang S. Selection of GalNAc-Conjugated si Keap1 as Disease-Specific Delivery System for Chemotherapy-Induced Liver Injury and Chronic Liver Disease. NANO LETTERS 2024; 24:1096-1105. [PMID: 38251670 DOI: 10.1021/acs.nanolett.3c03609] [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: 01/23/2024]
Abstract
Chemotherapy-induced liver injury (CILI) is a pressing concern in cancer patients. One promising approach involves activating nuclear factor erythroid 2-related factor 2 (Nrf2) to mitigate CILI. However, selectively activating liver Nrf2 without compromising chemotherapy's efficacy has remained elusive. Herein, two RNAi delivery strategies were explored: lipid nanoparticle (LNP) and N-acetylgalactosamine (GalNAc) delivery systems loaded with siRNA designed to silence Kelch-like-ECH associated protein 1 (Keap1) by aiming for liver-specific Nrf2 activation. Remarkably, siKeap1-LNP exhibited unintended tumor targeting alongside liver effects, thereby potentially promoting tumor progression. Conversely, siKeap1-GalNAc did not compromise chemotherapy efficacy and outperformed the conventional Nrf2 activator, bardoxolone, in mitigating CILI. This study proposes siKeap1-GalNAc as a promising therapeutic avenue for liver injury. Importantly, our study bridges a crucial gap concerning the delivery system for liver targeting but not tumor targeting and underscores the importance of selecting nucleic acid delivery systems tailored to specific diseases, not just to specific organs.
Collapse
Affiliation(s)
- Mengmeng You
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Meng Tian
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhiling Song
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhen Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Bingxue Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shiyi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
35
|
Zhang Z, Mocanu V, Deehan EC, Hotte N, Zhu Y, Wei S, Kao DH, Karmali S, Birch DW, Walter J, Madsen KL. Recipient microbiome-related features predicting metabolic improvement following fecal microbiota transplantation in adults with severe obesity and metabolic syndrome: a secondary analysis of a phase 2 clinical trial. Gut Microbes 2024; 16:2345134. [PMID: 38685731 PMCID: PMC11062372 DOI: 10.1080/19490976.2024.2345134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 04/16/2024] [Indexed: 05/02/2024] Open
Abstract
Microbial-based therapeutics in clinical practice are of considerable interest, and a recent study demonstrated fecal microbial transplantation (FMT) followed by dietary fiber supplements improved glucose homeostasis. Previous evidence suggests that donor and recipient compatibility and FMT protocol are key determinants, but little is known about the involvement of specific recipient factors. Using data from our recent randomized placebo-control phase 2 clinical trial in adults with obesity and metabolic syndrome, we grouped participants that received FMT from one of 4 donors with either fiber supplement into HOMA-IR responders (n = 21) and HOMA-IR non-responders (n = 8). We further assessed plasma bile acids using targeted metabolomics and performed subgroup analyzes to evaluate the effects of recipient parameters and gastrointestinal factors on microbiota engraftment and homeostatic model assessment of insulin resistance (HOMA2-IR) response. The baseline fecal microbiota composition at genus level of recipients could predict the improvements in HOMA2-IR at week 6 (ROC-AUC = 0.70). Prevotella was identified as an important predictor, with responders having significantly lower relative abundance than non-responders (p = .02). In addition, recipients displayed a highly individualized degree of microbial engraftment from donors. Compared to the non-responders, the responders had significantly increased bacterial richness (Chao1) after FMT and a more consistent engraftment of donor-specific bacteria ASVs (amplicon sequence variants) such as Faecalibacillus intestinalis (ASV44), Roseburia spp. (ASV103), and Christensenellaceae spp. (ASV140) (p < .05). Microbiota engraftment was strongly associated with recipients' factors at baseline including initial gut microbial diversity, fiber and nutrient intakes, inflammatory markers, and bile acid derivative levels. This study identified that responders to FMT therapy had a higher engraftment rate in the transplantation of specific donor-specific microbes, which were strongly correlated with insulin sensitivity improvements. Further, the recipient baseline gut microbiota and related factors were identified as the determinants for responsiveness to FMT and fiber supplementation. The findings provide a basis for the development of precision microbial therapeutics for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Zhengxiao Zhang
- College of Ocean Food and Biological Engineering, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian, China
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Valentin Mocanu
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Edward C. Deehan
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
| | - Naomi Hotte
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Yuanyuan Zhu
- College of Ocean Food and Biological Engineering, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian, China
| | - Shanshan Wei
- College of Ocean Food and Biological Engineering, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian, China
| | - Dina H. Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Shahzeer Karmali
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Daniel W. Birch
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Jens Walter
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | - Karen L. Madsen
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
36
|
Bartsch M, Hahn A, Berkemeyer S. Bridging the Gap from Enterotypes to Personalized Dietary Recommendations: A Metabolomics Perspective on Microbiome Research. Metabolites 2023; 13:1182. [PMID: 38132864 PMCID: PMC10744656 DOI: 10.3390/metabo13121182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
Advances in high-throughput DNA sequencing have propelled research into the human microbiome and its link to metabolic health. We explore microbiome analysis methods, specifically emphasizing metabolomics, how dietary choices impact the production of microbial metabolites, providing an overview of studies examining the connection between enterotypes and diet, and thus, improvement of personalized dietary recommendations. Acetate, propionate, and butyrate constitute more than 95% of the collective pool of short-chain fatty acids. Conflicting data on acetate's effects may result from its dynamic signaling, which can vary depending on physiological conditions and metabolic phenotypes. Human studies suggest that propionate has overall anti-obesity effects due to its well-documented chemistry, cellular signaling mechanisms, and various clinical benefits. Butyrate, similar to propionate, has the ability to reduce obesity by stimulating the release of appetite-suppressing hormones and promoting the synthesis of leptin. Tryptophan affects systemic hormone secretion, with indole stimulating the release of GLP-1, which impacts insulin secretion, appetite suppression, and gastric emptying. Bile acids, synthesized from cholesterol in the liver and subsequently modified by gut bacteria, play an essential role in the digestion and absorption of dietary fats and fat-soluble vitamins, but they also interact directly with intestinal microbiota and their metabolites. One study using statistical methods identified primarily two groupings of enterotypes Bacteroides and Ruminococcus. The Prevotella-dominated enterotype, P-type, in humans correlates with vegetarians, high-fiber and carbohydrate-rich diets, and traditional diets. Conversely, individuals who consume diets rich in animal fats and proteins, typical in Western-style diets, often exhibit the Bacteroides-dominated, B-type, enterotype. The P-type showcases efficient hydrolytic enzymes for plant fiber degradation but has limited lipid and protein fermentation capacity. Conversely, the B-type features specialized enzymes tailored for the degradation of animal-derived carbohydrates and proteins, showcasing an enhanced saccharolytic and proteolytic potential. Generally, models excel at predictions but often struggle to fully elucidate why certain substances yield varied responses. These studies provide valuable insights into the potential for personalized dietary recommendations based on enterotypes.
Collapse
Affiliation(s)
- Madeline Bartsch
- NutritionLab, Faculty of Agricultural Sciences and Landscape Architecture, Osnabrueck University of Applied Sciences, Am Kruempel 31, 49090 Osnabrueck, Germany;
- Institute of Food Science and Human Nutrition, Leibniz University Hannover, 30167 Hannover, Germany;
| | - Andreas Hahn
- Institute of Food Science and Human Nutrition, Leibniz University Hannover, 30167 Hannover, Germany;
| | - Shoma Berkemeyer
- NutritionLab, Faculty of Agricultural Sciences and Landscape Architecture, Osnabrueck University of Applied Sciences, Am Kruempel 31, 49090 Osnabrueck, Germany;
| |
Collapse
|
37
|
Zhu Q, Li G, Ma L, Chen B, Zhang D, Gao J, Deng S, Chen Y. Virgin Camellia Seed Oil Improves Glycolipid Metabolism in the Kidney of High Fat-Fed Rats through AMPK-SREBP Pathway. Nutrients 2023; 15:4888. [PMID: 38068746 PMCID: PMC10708295 DOI: 10.3390/nu15234888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Camellia seed oil (CO) is used as edible oil in southern China because of its excellent fatty acid composition and abundant bioactive compounds. Chronic kidney disease (CKD) is one of the most common chronic degenerative diseases in China, and active compounds in vegetable oil, like virgin olive oil, have been demonstrated to be efficacious in the management of CKD. In this study, virgin CO was refined using a standard process. The refining had minimal impact on the fatty acid composition, but significantly reduced the presence of bioactive compounds like polyphenols in CO. Sprague-Dawley (SD) rats fed with high fat diet (Group G) were treated with either virgin (Group Z) or refined CO (Group R). The oral administration of CO alleviated lipid accumulation and decreased body and kidney weight gain. Furthermore, treatment with virgin CO increased the renal ATP content. The renal expression levels of AMPK and key enzymes involved in fatty acid oxidation (CPT-1 and ACOX1) and glycolysis (HK, PFK, PK and GAPDH) were up-regulated in Group Z, thereby enhancing the ATP production. Virgin CO treatment downregulated the expression level of SREBP2 and its downstream target genes, such as ACC, FAS, and HMGCR, which reduced lipid synthesis. These findings indicate that virgin CO improves glycolipid metabolism and restores energy homeostasis in the kidneys of rats fed with a high-fat diet by modulating the AMPK-SREBP-signaling pathway, suggesting the potential of active compounds in virgin CO for managing the renal failure associated with glycolipid dysmetabolism.
Collapse
Affiliation(s)
- Qinhe Zhu
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Guihui Li
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Li Ma
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
| | - Bolin Chen
- Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Dawei Zhang
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Jing Gao
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
| | - Senwen Deng
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
- Hunan Key Laboratory of Economic Crops Genetic Improvement and Integrated Utilization, School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Yongzhong Chen
- National Engineering Research Center of Oiltea Camellia, State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Shao Shan South Road, No. 658, Changsha 410004, China; (Q.Z.); (G.L.); (L.M.); (D.Z.)
| |
Collapse
|
38
|
Ruocco C, Malavazos AE, Ragni M, Carruba MO, Valerio A, Iacobellis G, Nisoli E. Amino acids contribute to adaptive thermogenesis. New insights into the mechanisms of action of recent drugs for metabolic disorders are emerging. Pharmacol Res 2023; 195:106892. [PMID: 37619907 DOI: 10.1016/j.phrs.2023.106892] [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: 05/19/2023] [Revised: 07/28/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Adaptive thermogenesis is the heat production by muscle contractions (shivering thermogenesis) or brown adipose tissue (BAT) and beige fat (non-shivering thermogenesis) in response to external stimuli, including cold exposure. BAT and beige fat communicate with peripheral organs and the brain through a variegate secretory and absorption processes - controlling adipokines, microRNAs, extracellular vesicles, and metabolites - and have received much attention as potential therapeutic targets for managing obesity-related disorders. The sympathetic nervous system and norepinephrine-releasing adipose tissue macrophages (ATM) activate uncoupling protein 1 (UCP1), expressed explicitly in brown and beige adipocytes, dissolving the electrochemical gradient and uncoupling tricarboxylic acid cycle and the electron transport chain from ATP production. Mounting evidence has attracted attention to the multiple effects of dietary and endogenously synthesised amino acids in BAT thermogenesis and metabolic phenotype in animals and humans. However, the mechanisms implicated in these processes have yet to be conclusively characterized. In the present review article, we aim to define the principal investigation areas in this context, including intestinal microbiota constitution, adipose autophagy modulation, and secretome and metabolic fluxes control, which lead to increased brown/beige thermogenesis. Finally, also based on our recent epicardial adipose tissue results, we summarise the evidence supporting the notion that the new dual and triple agonists of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon (GCG) receptor - with never before seen weight loss and insulin-sensitizing efficacy - promote thermogenic-like amino acid profiles in BAT with robust heat production and likely trigger sympathetic activation and adaptive thermogenesis by controlling amino acid metabolism and ATM expansion in BAT and beige fat.
Collapse
Affiliation(s)
- Chiara Ruocco
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Alexis Elias Malavazos
- Endocrinology Unit, Clinical Nutrition and Cardiovascular Prevention Service, IRCCS Policlinico San Donato, Piazza Edmondo Malan, 2, San Donato Milanese, 20097 Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, via della Commenda, 10, 20122 Milan, Italy
| | - Maurizio Ragni
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Michele O Carruba
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa, 11, 25123 Brescia, Italy
| | - Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami, 1400 NW 12th Ave, Miami, FL, USA
| | - Enzo Nisoli
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy.
| |
Collapse
|
39
|
Qi L, Heianza Y, Li X, Sacks FM, Bray GA. Toward Precision Weight-Loss Dietary Interventions: Findings from the POUNDS Lost Trial. Nutrients 2023; 15:3665. [PMID: 37630855 PMCID: PMC10458797 DOI: 10.3390/nu15163665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
The POUNDS Lost trial is a 2-year clinical trial testing the effects of dietary interventions on weight loss. This study included 811 adults with overweight or obesity who were randomized to one of four diets that contained either 15% or 25% protein and 20% or 40% fat in a 2 × 2 factorial design. By 2 years, participants on average lost from 2.9 to 3.6 kg in body weight in the four intervention arms, while no significant difference was observed across the intervention arms. In POUNDS Lost, we performed a series of ancillary studies to detect intrinsic factors particular to genomic, epigenomic, and metabolomic markers that may modulate changes in weight and other cardiometabolic traits in response to the weight-loss dietary interventions. Genomic variants identified from genome-wide association studies (GWASs) on obesity, type 2 diabetes, glucose and lipid metabolisms, gut microbiome, and dietary intakes have been found to interact with dietary macronutrients (fat, protein, and carbohydrates) in relation to weight loss and changes of body composition and cardiometabolic traits. In addition, we recently investigated epigenomic modifications, particularly blood DNA methylation and circulating microRNAs (miRNAs). We reported DNA methylation levels at NFATC2IP, CPT1A, TXNIP, and LINC00319 were related to weight loss or changes of glucose, lipids, and blood pressure; we also reported thrifty miRNA expression as a significant epigenomic marker related to changes in insulin sensitivity and adiposity. Our studies have also highlighted the importance of temporal changes in novel metabolomic signatures for gut microbiota, bile acids, and amino acids as predictors for achievement of successful weight loss outcomes. Moreover, our studies indicate that biochemical, behavioral, and psychosocial factors such as physical activity, sleep disturbance, and appetite may also modulate metabolic changes during dietary interventions. This review summarized our major findings in the POUNDS Lost trial, which provided preliminary evidence supporting the development of precision diet interventions for obesity management.
Collapse
Affiliation(s)
- Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
| | - Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - George A. Bray
- Department of Clinical Obesity, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| |
Collapse
|
40
|
Belli M, Barone L, Longo S, Prandi FR, Lecis D, Mollace R, Margonato D, Muscoli S, Sergi D, Federici M, Barillà F. Gut Microbiota Composition and Cardiovascular Disease: A Potential New Therapeutic Target? Int J Mol Sci 2023; 24:11971. [PMID: 37569352 PMCID: PMC10418329 DOI: 10.3390/ijms241511971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/13/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
A great deal of evidence has revealed an important link between gut microbiota and the heart. In particular, the gut microbiota plays a key role in the onset of cardiovascular (CV) disease, including heart failure (HF). In HF, splanchnic hypoperfusion causes intestinal ischemia resulting in the translocation of bacteria and their metabolites into the blood circulation. Among these metabolites, the most important is Trimethylamine N-Oxide (TMAO), which is responsible, through various mechanisms, for pathological processes in different organs and tissues. In this review, we summarise the complex interaction between gut microbiota and CV disease, particularly with respect to HF, and the possible strategies for influencing its composition and function. Finally, we highlight the potential role of TMAO as a novel prognostic marker and a new therapeutic target for HF.
Collapse
Affiliation(s)
- Martina Belli
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Lucy Barone
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| | - Susanna Longo
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy (R.M.)
| | - Francesca Romana Prandi
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
- Division of Cardiology, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Dalgisio Lecis
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| | - Rocco Mollace
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy (R.M.)
- Cardiovascular Department, Humanitas Gavazzeni, 24125 Bergamo, Italy
| | - Davide Margonato
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Saverio Muscoli
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| | - Domenico Sergi
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| | - Massimo Federici
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy (R.M.)
| | - Francesco Barillà
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| |
Collapse
|
41
|
Manghi P, Blanco-Míguez A, Manara S, NabiNejad A, Cumbo F, Beghini F, Armanini F, Golzato D, Huang KD, Thomas AM, Piccinno G, Punčochář M, Zolfo M, Lesker TR, Bredon M, Planchais J, Glodt J, Valles-Colomer M, Koren O, Pasolli E, Asnicar F, Strowig T, Sokol H, Segata N. MetaPhlAn 4 profiling of unknown species-level genome bins improves the characterization of diet-associated microbiome changes in mice. Cell Rep 2023; 42:112464. [PMID: 37141097 PMCID: PMC10242440 DOI: 10.1016/j.celrep.2023.112464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/10/2023] [Accepted: 04/17/2023] [Indexed: 05/05/2023] Open
Abstract
Mouse models are key tools for investigating host-microbiome interactions. However, shotgun metagenomics can only profile a limited fraction of the mouse gut microbiome. Here, we employ a metagenomic profiling method, MetaPhlAn 4, which exploits a large catalog of metagenome-assembled genomes (including 22,718 metagenome-assembled genomes from mice) to improve the profiling of the mouse gut microbiome. We combine 622 samples from eight public datasets and an additional cohort of 97 mouse microbiomes, and we assess the potential of MetaPhlAn 4 to better identify diet-related changes in the host microbiome using a meta-analysis approach. We find multiple, strong, and reproducible diet-related microbial biomarkers, largely increasing those identifiable by other available methods relying only on reference information. The strongest drivers of the diet-induced changes are uncharacterized and previously undetected taxa, confirming the importance of adopting metagenomic methods integrating metagenomic assemblies for comprehensive profiling.
Collapse
Affiliation(s)
- Paolo Manghi
- Department CIBIO, University of Trento, Trento, Italy
| | | | - Serena Manara
- Department CIBIO, University of Trento, Trento, Italy
| | - Amir NabiNejad
- Department CIBIO, University of Trento, Trento, Italy; IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Fabio Cumbo
- Department CIBIO, University of Trento, Trento, Italy
| | | | | | | | - Kun D Huang
- Department CIBIO, University of Trento, Trento, Italy
| | | | | | | | - Moreno Zolfo
- Department CIBIO, University of Trento, Trento, Italy
| | - Till R Lesker
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marius Bredon
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, CRSA, AP-HP, Saint Antoine Hospital, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Julien Planchais
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; INRAE, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France
| | - Jeremy Glodt
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; INRAE, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France
| | | | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples, Naples, Italy
| | | | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany; Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Harry Sokol
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, CRSA, AP-HP, Saint Antoine Hospital, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; INRAE, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy; IEO, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
42
|
Fusco W, Lorenzo MB, Cintoni M, Porcari S, Rinninella E, Kaitsas F, Lener E, Mele MC, Gasbarrini A, Collado MC, Cammarota G, Ianiro G. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023; 15:2211. [PMID: 37432351 DOI: 10.3390/nu15092211] [Citation(s) in RCA: 323] [Impact Index Per Article: 161.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 07/12/2023] Open
Abstract
Short-chain fatty acids (SCFAs) play a key role in health and disease, as they regulate gut homeostasis and their deficiency is involved in the pathogenesis of several disorders, including inflammatory bowel diseases, colorectal cancer, and cardiometabolic disorders. SCFAs are metabolites of specific bacterial taxa of the human gut microbiota, and their production is influenced by specific foods or food supplements, mainly prebiotics, by the direct fostering of these taxa. This Review provides an overview of SCFAs' roles and functions, and of SCFA-producing bacteria, from their microbiological characteristics and taxonomy to the biochemical process that lead to the release of SCFAs. Moreover, we will describe the potential therapeutic approaches to boost the levels of SCFAs in the human gut and treat different related diseases.
Collapse
Affiliation(s)
- William Fusco
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Manuel Bernabeu Lorenzo
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46022 Valencia, Spain
| | - Marco Cintoni
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
| | - Serena Porcari
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Emanuele Rinninella
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
| | - Francesco Kaitsas
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Elena Lener
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Maria Cristina Mele
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46022 Valencia, Spain
| | - Giovanni Cammarota
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, Digestive Disease Center, Universitary Policlinic Agostino Gemelli Foundation IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|