1
|
Thakkar B, Dadhaniya H, Dudhat K. Exploring hypertension-linked diseases: a comprehensive review of innovative drug combinations with enhanced therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6643-6673. [PMID: 39888363 DOI: 10.1007/s00210-025-03819-3] [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: 11/04/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025]
Abstract
Hypertension, a prevalent cardiovascular condition affecting a substantial portion of the global population, remains a formidable health challenge associated with a multitude of complications. This review article provides a comprehensive examination of hypertension, its various complications, and the emergence of a novel management technique that shows promising potential in transforming the therapeutic landscape. Over the years, conventional treatment approaches, encompassing lifestyle modifications, dietary interventions, and pharmacotherapy, have been the mainstay in managing hypertension. However, these strategies fall short in achieving optimal blood pressure control and preventing complications in a significant number of patients. Consequently, the medical community has ventured into exploring innovative management techniques to tackle this unmet medical need. The focal point of this review centers on the emergence of a new management technique for hypertension that exhibits promise in preclinical and clinical studies. The latest research findings shed light on the efficacy and safety of this innovative approach, which encompasses pharmaceutical agents, medical devices, and non-invasive interventions. Through critical analysis and discussion, we explore the potential impact of these novel strategies on hypertension management and patient outcomes. In conclusion, this review article provides a comprehensive overview of hypertension, its complications, and the promising emergence of innovative management techniques. By acknowledging the complexity of hypertension and the potential of new therapeutic avenues, we aspire to pave the way for improved patient care, enhanced quality of life, and ultimately, the mitigation of hypertension-related morbidity and mortality.
Collapse
Affiliation(s)
- Bhavesh Thakkar
- School of Pharmacy, RK University, Kasturbadham, Rajkot, Gujarat, 360020, India
| | - Hetvi Dadhaniya
- School of Pharmacy, RK University, Kasturbadham, Rajkot, Gujarat, 360020, India
| | - Kiran Dudhat
- School of Pharmacy, RK University, Kasturbadham, Rajkot, Gujarat, 360020, India.
| |
Collapse
|
2
|
Saeedi Saravi SS, Pugin B, Constancias F, Shabanian K, Spalinger M, Thomas A, Le Gludic S, Shabanian T, Karsai G, Colucci M, Menni C, Attaye I, Zhang X, Allemann MS, Lee P, Visconti A, Falchi M, Alimonti A, Ruschitzka F, Paneni F, Beer JH. Gut microbiota-dependent increase in phenylacetic acid induces endothelial cell senescence during aging. NATURE AGING 2025:10.1038/s43587-025-00864-8. [PMID: 40355758 DOI: 10.1038/s43587-025-00864-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/02/2025] [Indexed: 05/15/2025]
Abstract
Endothelial cell senescence is a key driver of cardiovascular aging, yet little is known about the mechanisms by which it is induced in vivo. Here we show that the gut bacterial metabolite phenylacetic acid (PAA) and its byproduct, phenylacetylglutamine (PAGln), are elevated in aged humans and mice. Metagenomic analyses reveal an age-related increase in PAA-producing microbial pathways, positively linked to the bacterium Clostridium sp. ASF356 (Clos). We demonstrate that colonization of young mice with Clos increases blood PAA levels and induces endothelial senescence and angiogenic incompetence. Mechanistically, we find that PAA triggers senescence through mitochondrial H2O2 production, exacerbating the senescence-associated secretory phenotype. By contrast, we demonstrate that fecal acetate levels are reduced with age, compromising its function as a Sirt1-dependent senomorphic, regulating proinflammatory secretion and redox homeostasis. These findings define PAA as a mediator of gut-vascular crosstalk in aging and identify sodium acetate as a potential microbiome-based senotherapy to promote healthy aging.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland.
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland.
| | - Benoit Pugin
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Florentin Constancias
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Khatereh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Marianne Spalinger
- Department for Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Aurélien Thomas
- Faculty Unit of Toxicology, University Center of Legal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, University Center of Legal Medicine, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne, Geneva, Switzerland
| | - Sylvain Le Gludic
- Faculty Unit of Toxicology, University Center of Legal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, University Center of Legal Medicine, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne, Geneva, Switzerland
| | - Taraneh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland
| | - Manuel Colucci
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera Italiana, Lugano, Switzerland
| | - Cristina Menni
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Ilias Attaye
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
- Amsterdam Cardiovascular Sciences, Diabetes & Metabolism, Amsterdam, Netherlands
| | - Xinyuan Zhang
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
| | - Meret Sarah Allemann
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Pratintip Lee
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Alessia Visconti
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinial and Biological Sciences, University of Turin, Turin, Italy
| | - Mario Falchi
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medicine, University of Padova, Padova, Italy
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Zurich, Switzerland
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.
- Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland.
| |
Collapse
|
3
|
Nageswaran V, Carreras A, Reinshagen L, Beck KR, Steinfeldt J, Henricsson M, Ramezani Rad P, Peters L, Strässler ET, Lim J, Verhaar BJ, Döring Y, Weber C, König M, Steinhagen-Thiessen E, Demuth I, Kränkel N, Leistner DM, Potente M, Nieuwdorp M, Knaus P, Kuebler WM, Ferrell M, Nemet I, Hazen SL, Landmesser U, Bäckhed F, Haghikia A. Gut Microbial Metabolite Imidazole Propionate Impairs Endothelial Cell Function and Promotes the Development of Atherosclerosis. Arterioscler Thromb Vasc Biol 2025; 45:823-839. [PMID: 40143816 PMCID: PMC12017598 DOI: 10.1161/atvbaha.124.322346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND The microbially produced amino acid-derived metabolite imidazole propionate (ImP) contributes to the pathogenesis of type 2 diabetes. However, the effects of ImP on endothelial cell (EC) physiology and its role in atherosclerotic coronary artery disease are unknown. Using both human and animal model studies, we investigated the potential contributory role of ImP in the development of atherosclerosis. METHODS Plasma levels of ImP were measured in patients undergoing elective cardiac angiography (n=831) by ultra-high performance liquid chromatography coupled to tandem mass spectrometry. Odds ratios and corresponding 95% confidence intervals for coronary artery disease were calculated based on the ImP quartiles using both univariable and multivariable logistic regression models. The effects of ImP on functional properties of ECs were assessed using HAECs (human aortic endothelial cells). In a mouse model of carotid artery injury, the impact of ImP on vascular regeneration was examined. Additionally, atheroprone Apoe-/- mice fed a high-fat diet were treated with and without ImP (800 µg), and aortic atherosclerotic lesion area was evaluated after 12 weeks. Next-generation sequencing, Western blot analysis, small interfering RNA-based gene knockdown, and tamoxifen-inducible Cre-loxP experiments were performed to investigate ImP-mediated molecular mechanisms. RESULTS Plasma ImP levels in subjects undergoing cardiac evaluation were associated with increased risk of prevalent coronary artery disease. We found that ImP dose dependently impaired migratory and angiogenic properties of human ECs and promoted an increased inflammatory response. Long-term exposure to ImP compromised the repair potential of the endothelium after an arterial insult. In atheroprone Apoe-/- (apolipoprotein E-/-) mice, ImP increased atherosclerotic lesion size. Mechanistically, ImP attenuated insulin receptor signaling by suppressing the PI3K (phosphoinositide 3-kinase)/AKT pathway leading to sustained activation of the FOXO1 (forkhead box protein O1) transcription factor. Genetic inactivation of endothelial FOXO1 signaling in ImP-treated mice enhanced the angiogenic activity and preserved the vascular repair capacity of ECs after carotid injury. CONCLUSIONS Our findings reveal a hitherto unknown role of the microbially produced histidine-derived metabolite ImP in endothelial dysfunction and atherosclerosis, suggesting that ImP metabolism is a potential therapeutic target in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Vanasa Nageswaran
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (V.N., P.K.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
| | - Alba Carreras
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Leander Reinshagen
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
| | - Katharina R. Beck
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Jakob Steinfeldt
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Pegah Ramezani Rad
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
| | - Lisa Peters
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (L.P., W.M.K.)
- Institute of Biology, Freie Universität Berlin, Germany (L.P.)
| | - Elisabeth T. Strässler
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
| | - Joseph Lim
- Angiogenesis and Metabolism Laboratory, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.L., M.P.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.L., M.P.)
| | - Barbara J.H. Verhaar
- Department of Internal Medicine-Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), the Netherlands (B.J.H.V., M.N.)
- Department of Vascular Medicine, Amsterdam UMC, the Netherlands (B.J.H.V., M.N.)
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Germany (Y.D., C.W.)
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Switzerland (Y.D.)
- Department for BioMedical Research (DBMR), University of Bern, Switzerland (Y.D.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany (Y.D., C.W.)
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Germany (Y.D., C.W.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany (Y.D., C.W.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands (C.W.)
- Munich Cluster for Systems Neurology, Germany (C.W.)
| | - Maximilian König
- Department of Internal Medicine D–Geriatrics, University Medicine Greifswald, Germany (M.K.)
| | - Elisabeth Steinhagen-Thiessen
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (E.S.-T., I.D.)
| | - Ilja Demuth
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (E.S.-T., I.D.)
- Charité–Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Germany (I.D.)
| | - Nicolle Kränkel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
| | - David M. Leistner
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhine-Main, Germany (D.M.L.)
- Department of Medicine, Cardiology and Angiology, Goethe University Hospital, Frankfurt, Germany (D.M.L.)
| | - Michael Potente
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Angiogenesis and Metabolism Laboratory, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.L., M.P.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.L., M.P.)
| | - Max Nieuwdorp
- Department of Internal Medicine-Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), the Netherlands (B.J.H.V., M.N.)
- Department of Vascular Medicine, Amsterdam UMC, the Netherlands (B.J.H.V., M.N.)
| | - Petra Knaus
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (V.N., P.K.)
- Berlin-Brandenburg School for Regenerative Therapies, Germany (P.K.)
- International Max-Planck Research School for Biology and Computation, Berlin, Germany (P.K.)
| | - Wolfgang M. Kuebler
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (L.P., W.M.K.)
- German Center for Lung Research (DZL), Berlin, Germany (W.M.K.)
- Keenan Research Centre for Biomedical Science at St. Michael’s, Toronto, Canada (W.M.K.)
- Departments of Surgery and Physiology, University of Toronto, Canada (W.M.K.)
| | - Marc Ferrell
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
| | - Ina Nemet
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
| | - Stanley L. Hazen
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, OH (S.L.H.)
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden (F.B.)
| | - Arash Haghikia
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| |
Collapse
|
4
|
Deng L, Tan KSW. Decoding Blastocystis-Driven Mechanisms in Gut Microbiota and Host Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416325. [PMID: 40184630 PMCID: PMC12061313 DOI: 10.1002/advs.202416325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/25/2025] [Indexed: 04/06/2025]
Abstract
Blastocystis, a prevalent eukaryotic microorganism in the gut microbiota, has emerged as a potential link between healthy diets and improved cardiometabolic health. Despite its genetic diversity and varied host interactions, Blastocystis is consistently associated with healthier dietary patterns and reduced risk of cardiometabolic diseases. Current evidence suggests that Blastocystis may influence host metabolism by modulating gut microbial composition, short-chain fatty acids (SCFAs) production, and immune cell differentiation. Moreover, its role in tryptophan metabolism provides intriguing insights into its potential impact on host signaling pathways. However, mechanistic evidence connecting Blastocystis to improved metabolic health remains limited. This perspective explores plausible pathways, including SCFAs-mediated signaling, tryptophan metabolism, and immune modulation, through which Blastocystis may exert its effects. A systematic research framework integrating axenic cultivation, in vitro co-culture systems, animal models, and multi-omics approaches is proposed to further elucidate these mechanisms and expand the understanding of Blastocystis in gut health and disease.
Collapse
Affiliation(s)
- Lei Deng
- Shanghai Veterinary Research InstituteChinese Academy of Agricultural SciencesShanghai200241China
- Broad Institute of MIT and HarvardCambridgeMA02142USA
- Center for Computational and Integrative BiologyMassachusetts General HospitalBostonMA02134USA
| | - Kevin SW Tan
- Laboratory of Molecular and Cellular ParasitologyHealthy Longevity Translational Research Programme and Department of Microbiology and ImmunologyYong Loo Lin School of MedicineNational University of SingaporeSingapore117545Singapore
| |
Collapse
|
5
|
Leonardo J, Hertanto R, Surya R, Syahputra RA, Humayrah W, Sabrina N, Taslim NA, Tallei TE, Tjandrawinata RR, Nurkolis F. Delites™ supplementation prevents metabolic syndrome onset and modulates gut microbiome in male Sprague Dawley rats fed on cholesterol- and fat-enriched diet: a randomized preclinical trial study. Front Nutr 2025; 12:1571473. [PMID: 40331097 PMCID: PMC12052560 DOI: 10.3389/fnut.2025.1571473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Background Metabolic syndrome (MetS) is a global health concern, characterized by a combination of dyslipidemia, insulin resistance, obesity, and hypertension, significantly increasing the risk of type 2 diabetes mellitus (T2DM) and cardiovascular diseases. Gut microbiota plays a pivotal role in MetS pathophysiology, with dysbiosis exacerbating metabolic impairments. Delites™, a supplement inspired by Traditional Chinese Medicine, has shown potential in modulating gut microbiota and mitigating MetS. Objectives This study aimed to evaluate the effects of Delites™ supplementation on metabolic health and gut microbiota composition in male Sprague Dawley rats fed a cholesterol- and fat-enriched diet (CFED). Methods A randomized preclinical trial was conducted on 32 rats divided into four groups: control-normal, CFED, CFED+low-dose Delites™ (54 mg/kg), and CFED+high-dose Delites™ (108 mg/kg). Parameters including lipid profiles, enzymatic activity, molecular biomarkers, and gut microbiota composition were analyzed. Results Delites™ significantly improved lipid profiles, reduced inflammation (TNF-α), enhanced anti-inflammatory markers (IL-10), and increased energy metabolism regulator PGC-1α. Gut microbiota modulation showed increased beneficial genera (Bifidobacterium, Lactobacillus) and reduced pathogenic Proteus, improving microbial diversity. Conclusion Delites™ supplementation effectively mitigates MetS through metabolic and microbiota modulation. These findings highlight its potential for precision medicine approaches to combat metabolic disorders. Further research is needed to explore its long-term effects and translational relevance in humans.
Collapse
Affiliation(s)
| | - Robby Hertanto
- Increase Laboratorium Indonesia, Biomedical Campus BSD City, Tangerang, Banten, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, University of North Sumatra, Medan, Indonesia
| | - Wardina Humayrah
- Nutrition Study Program, Faculty of Food Technology and Health, Sahid University, Jakarta, Indonesia
| | - Nindy Sabrina
- Nutrition Study Program, Faculty of Food Technology and Health, Sahid University, Jakarta, Indonesia
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, Indonesia
| | - Raymond Rubianto Tjandrawinata
- Faculty of Biotechnology, Center for Pharmaceutical and Nutraceutical Research and Policy, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Fahrul Nurkolis
- State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, Indonesia
- Master of Basic Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Medical Research Center of Indonesia, Surabaya, East Java, Indonesia
| |
Collapse
|
6
|
Liu H, Xiang D, Zhou J, Xie J. Protective Effect of Dictyophora rubrovolvata Extract on Intestinal and Liver Tissue Toxicity Induced by Metformin Disinfection Byproducts. TOXICS 2025; 13:310. [PMID: 40278626 PMCID: PMC12030868 DOI: 10.3390/toxics13040310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Metformin disinfection byproducts Y and C have emerged as pollutants of concern in drinking water systems and are suspected to possess significant toxicity to mammals. However, effective strategies to mitigate the effects of Y and C exposure in mammals have not been thoroughly formulated. This study aimed to investigate the toxicity and characteristic phenotypes of short-term, high-dose exposure to Y and C in the intestine and liver of mice and to evaluate the protective effects of Dictyophora rubrovolvata extract (DRE) on Y- and C-induced intestinal and liver damage. The results showed that exposure-induced intestinal toxicity manifested mainly as intestinal barrier dysfunction, induction of immune response and oxidative stress, and disruption of intestinal flora homeostasis. Hepatotoxicity was mainly characterized by histopathological changes such as vacuolar degeneration, abnormal liver function, and oxidative stress. Additionally, marked changes in gut microbiota and biochemical indicators were closely related to hepatic and intestinal injuries after exposure. DRE effectively alleviated Y- and C-induced intestinal and liver damage, reshaped the gut microbiota, and maintained gut-liver axis homeostasis. These findings provide new insights into the toxic effects of disinfection byproduct exposure through the gut-liver axis and suggest that functional food extracts may serve to protect against these adverse health outcomes.
Collapse
Affiliation(s)
- Huijuan Liu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 561113, China;
| | - Dong Xiang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 561113, China;
| | - Jie Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China;
| | - Jiao Xie
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Guizhou Medical University, Guiyang 561113, China;
| |
Collapse
|
7
|
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
|
8
|
Zhou S, Zhou X, Zhang P, Zhang W, Huang J, Jia X, He X, Sun X, Su H. The gut microbiota-inflammation-HFpEF axis: deciphering the role of gut microbiota dysregulation in the pathogenesis and management of HFpEF. Front Cell Infect Microbiol 2025; 15:1537576. [PMID: 40182777 PMCID: PMC11965942 DOI: 10.3389/fcimb.2025.1537576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Heart failure with preserved left ventricular ejection fraction (HFpEF) is a disease that affects multiple organs throughout the body, accounting for over 50% of heart failure cases. HFpEF has a significant impact on individuals' life expectancy and quality of life, but the exact pathogenesis remains unclear. Emerging evidence implicates low-grade systemic inflammation as a crucial role in the onset and progression of HFpEF. Gut microbiota dysregulation and associated metabolites alteration, including short-chain fatty acids, trimethylamine N-oxides, amino acids, and bile acids can exacerbate chronic systemic inflammatory responses and potentially contribute to HFpEF. In light of these findings, we propose the hypothesis of a "gut microbiota-inflammation-HFpEF axis", positing that the interplay within this axis could be a crucial factor in the development and progression of HFpEF. This review focuses on the role of gut microbiota dysregulation-induced inflammation in HFpEF's etiology. It explores the potential mechanisms linking dysregulation of the gut microbiota to cardiac dysfunction and evaluates the therapeutic potential of restoring gut microbiota balance in mitigating HFpEF severity. The objective is to offer novel insights and strategies for the management of HFpEF.
Collapse
Affiliation(s)
- Shenghua Zhou
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xuan Zhou
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Wei Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Jinli Huang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xuzhao Jia
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xiaole He
- Department of General Practice, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| |
Collapse
|
9
|
Jena PK, Arditi M, Rivas MN. Gut Microbiota Alterations in Patients With Kawasaki Disease. Arterioscler Thromb Vasc Biol 2025; 45:345-358. [PMID: 39846163 PMCID: PMC11998981 DOI: 10.1161/atvbaha.124.321201] [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: 10/14/2024] [Revised: 12/09/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025]
Abstract
The intestinal microbiota influences many host biological processes, including metabolism, intestinal barrier functions, and immune responses in the gut and distant organs. Alterations in its composition have been associated with the development of inflammatory disorders and cardiovascular diseases, including Kawasaki disease (KD). KD is an acute pediatric vasculitis of unknown etiology and the leading cause of acquired heart disease in children in the United States. The presence of gastrointestinal symptoms in the acute phase of KD has been associated with an increased risk of treatment resistance and the development of coronary artery aneurysms. Studies report alterations in fecal bacterial communities of patients with KD, characterized by the blooming of pathogenic bacteria and decreased relative abundance of short-chain fatty acid-producing bacteria. However, causality and functionality cannot be established from these observational patient cohorts of KD. This highlights the need for more advanced and rigorous studies to establish causality and functionality in both experimental models of KD vasculitis and patient cohorts. Here, we review the evidence linking an altered gut microbiota composition to the development of KD, assess the potential mechanisms involved in this process, and discuss the potential therapeutic value of these observations.
Collapse
Affiliation(s)
- Prasant K. Jena
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
10
|
Zheng T, Meng C, Lv Z, Wu C, Zhou X, Mao W. The Critical Role of Faecalibacterium prausnitzii in Cardiovascular Diseases. Rev Cardiovasc Med 2025; 26:26740. [PMID: 40160596 PMCID: PMC11951488 DOI: 10.31083/rcm26740] [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: 09/26/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
Due to the continued aging of the global population, cardiovascular diseases (CVDs) remain the main cause of death worldwide, with millions of fatalities from diseases, including stroke and coronary artery disease, reported annually. Thus, novel therapeutic approaches and targets are urgently required for diagnosing and treating CVDs. Recent studies emphasize the vital part of gut microbiota in both CVD prevention and management. Among these, Faecalibacterium prausnitzii (F. prausnitzii) has emerged as a promising probiotic capable of improving intestinal health. Although preliminary investigations demonstrate that F. prausnitzii positively enhances cardiovascular health, research specifically connecting this strain to CVD outcomes remains limited. Based on current research and assessment of possible clinical applications, this paper aimed to investigate the positive effects on cardiovascular health using F. prausnitzii and its metabolites. Targeting gut flora is expected to become a mainstay in CVD treatment as research develops.
Collapse
Affiliation(s)
- Tiantian Zheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Chenchen Meng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Zhengtian Lv
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Chenxia Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, Zhejiang, China
| | - Xinbin Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006 Hangzhou, Zhejiang, China
| | - Wei Mao
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Integrative Chinese and Western Medicine for Diagnosis and Treatment of Circulatory Diseases, 310030 Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Li R, Kurilshikov A, Yang S, van Oortmerssen JA, van Hilten A, Ahmadizar F, Roshchupkin G, Kraaij R, Duijts L, Fu J, Ikram MK, Jaddoe VW, Uitterlinden AG, Rivadeneira F, Kavousi M, Zhernakova A, Medina-Gomez C. Association between gut microbiome profiles and host metabolic health across the life course: a population-based study. THE LANCET REGIONAL HEALTH. EUROPE 2025; 50:101195. [PMID: 39834614 PMCID: PMC11743806 DOI: 10.1016/j.lanepe.2024.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Background The human gut microbiome changes considerably over time. Previous studies have shown that gut microbiome profiles correlate with multiple metabolic traits. As disease development is likely a lifelong process, evidence gathered at different life stages would help gain a better understanding of this correlation. Therefore, we aim to investigate how the association of the gut microbiome and metabolic traits change over the lifespan. Methods We identified microbiome patterns (clusters) within two population-based cohorts at different life stages, i.e., pre-adolescents of the Generation R Study (mean age 9.8 years; n = 1488) and older adults of the Rotterdam Study (RS, mean age 62.7 years; n = 1265) using K-Means clustering, and surveyed for host metabolic phenotypes, lifestyles and other factors driving these patterns. Analyses were replicated in the Lifelines-DEEP Study (mean age 45.0 years; n = 1117). The association between microbiome clusters and host metabolic health was evaluated as well as the link between microbiome clusters and incident atherosclerotic cardiovascular disease (ASCVD) in RS during follow-up (median 6.5 years). Findings We identified two distinct microbiome clusters (U and H) within each study population presenting contrasting metabolic statuses. Cluster U was characterized by lower microbiome diversity, increased Streptococcus, Fusicatenibacter, and decreased Prevotella_9 and Christensenellaceae_R-7_group; wherein individuals showed higher fat percentage, triglycerides, use of medications, and lower socioeconomic status. Individuals in cluster U had increased odds (between 1.10 and 1.65) of being relatively metabolically unhealthy and presented a higher 5-year ASCVD risk (mean risk 0.059 ± 0.071 vs 0.047 ± 0.042, p < 0.001). Interpretation We provide evidence of a life-course relationship between gut microbiome profiles and metabolic health. Funding R.L is supported by European UnionHorizon 2020 research and innovation program under Marie Skłodowska-Curie grant agreement No 860898 [FIDELIO].
Collapse
Affiliation(s)
- Ruolin Li
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Shuyue Yang
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Arno van Hilten
- Department of Medical Informatics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Fariba Ahmadizar
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Gennady Roshchupkin
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Robert Kraaij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Liesbeth Duijts
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - M. Kamran Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Vincent W.V. Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
12
|
Gu P, Wei R, Liu R, Yang Q, He Y, Guan J, He W, Li J, Zhao Y, Xie L, He J, Guo Q, Hu J, Bao J, Wang W, Guo J, Zeng Z, Chen Z, Jiang Y, Liu Z, Chen P. Aging-induced Alternation in the Gut Microbiota Impairs Host Antibacterial Defense. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411008. [PMID: 39792643 PMCID: PMC11948050 DOI: 10.1002/advs.202411008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/10/2024] [Indexed: 01/12/2025]
Abstract
Older individuals experience increased susceptibility and mortality to bacterial infections, but the underlying etiology remains unclear. Herein, it is shown that aging-associated reduction of commensal Parabacteroides goldsteinii (P. goldsteinii) in both aged mice and humans critically contributes to worse outcomes of bacterial infection. The colonization of live P. goldsteinii conferred protection against aging-associated bacterial infections. Metabolomic profiling reveals a protective compound, apigenin, generated by P. goldsteinii, antagonizes bacterial clearance defects in aged mice. AMP-binding protein (ampB) is identified as a key gene involved in apigenin synthesis in P. goldsteinii using homologous recombination in bacteria. Mechanistically, apigenin binds directly to the potential sites on Fgr (M341 and D404), preventing its inhibitory role on Vav1 phosphorylation, and therefore promoting the activation of Cdc42/Rac1, Arp2/3 expression and subsequent actin reorganization, which contributes to the enhanced phagocytosis of macrophages to bacteria. Collectively, the findings suggest that dysbiosis of the gut microbiota may impair host defense mechanisms and increase susceptibility to bacterial infections in older adults and highlight the microbiota-apigenin-Fgr axis as a possible route to ameliorate aging-associated antibacterial defects.
Collapse
Affiliation(s)
- Peng Gu
- Department of Critical Care MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Rongjuan Wei
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Ruofan Liu
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Qin Yang
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
- Department of GastroenterologyThe Seventh Affiliated Hospital of Southern Medical UniversityFoshan528244China
| | - Yuxuan He
- Department of Critical Care MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Jianbin Guan
- Department of Critical Care MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Wenhao He
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Jiaxin Li
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Yunfei Zhao
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Li Xie
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Jie He
- Department of Critical Care MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Qingling Guo
- Department of Critical Care MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Jiajia Hu
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Jingna Bao
- Department of Critical Care MedicineNanfang HospitalSouthern Medical UniversityGuangzhou510510China
| | - Wandang Wang
- Department of Clinical Medicine LaboratoryAffiliated Xiaolan HospitalSouthern Medical UniversityZhongshan528415China
| | - Jiayin Guo
- NMPA Key Laboratory for Research and Evaluation of Drug MetabolismGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Zhenhua Zeng
- Department of Critical Care MedicineNanfang HospitalSouthern Medical UniversityGuangzhou510510China
| | - Zhongqing Chen
- Department of Critical Care MedicineNanfang HospitalSouthern Medical UniversityGuangzhou510510China
| | - Yong Jiang
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
- Department of Respiratory and Critical Care MedicineThe Tenth Affiliated HospitalSouthern Medical UniversityDongguan523059China
| | - Zhanguo Liu
- Department of Critical Care MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Peng Chen
- Department of Critical Care MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
- Department of PathophysiologyGuangdong Provincial Key Laboratory of ProteomicsSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
13
|
Zhang H, Li Y, Li L. Blood metabolites reflect the effect of gut microbiota on differentiated thyroid cancer: a Mendelian randomization analysis. BMC Cancer 2025; 25:368. [PMID: 40022019 PMCID: PMC11869591 DOI: 10.1186/s12885-025-13598-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 01/27/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Studies have linked gut microbiome and differentiated thyroid cancer (DTC). However, their causal relationships and potential mediating factors have not been well defined. Our study investigated the causal relationships between the gut microbiome, papillary thyroid cancer (PTC) and follicular thyroid cancer (FTC), as well as the mediating effect of potential blood metabolites, using genetic approaches. METHODS Leveraging the summary statistics of gut microbial taxa, blood metabolites, PTC and FTC from the largest genome-wide association studies (GWAS) to date, we applied the bidirectional and mediation Mendelian randomization (MR) design. The multivariable MR approach based on Bayesian model averaging (MR-BMA) was used to prioritize the most likely causal taxa. Furthermore, metabolic pathway analysis was performed via the web-based Metaconflict 4.0. RESULTS After sensitivity analyses, we identified 4 taxa, 19 blood metabolites, and 5 gut bacterial pathways were causally associated with PTC. Similarly, 3 taxa, 31 blood metabolites, and 3 gut bacterial pathways were found to be causally associated with FTC, with 2 blood metabolites exhibiting bidirectional causal relationships. Metabolic pathway analysis revealed 8 significant pathways in PTC and FTC. MR-BMA analysis pinpointed species Bifidobacterium longum as the primary causal taxon for PTC and genus Bacteroides for FTC. The mediation MR analysis showed that sphingomyelin (d18:2/23:0, d18:1/23:1, d17:1/24:1) and 2-hydroxysebacate mediated the causal effects of specific gut microbiota on PTC and FTC, respectively. CONCLUSION The study suggested a causal relationship between several gut microbial taxa and DTC, and that specific blood metabolites might mediate this relationship.
Collapse
Affiliation(s)
- Hanfei Zhang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China
| | - Yuhao Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China
| | - Lin Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, No. 37, Guo Xue Xiang, Chengdu, 610041, China.
| |
Collapse
|
14
|
Zeng Y, Wu Q, Guo M, Teng F, Jiang C, Chen J, Tan X, Zeng C, Long Y, Law BYK, Xu Y. Gut microbiota-derived imidazole propionate: an emerging target for the prevention and treatment of cardiometabolic diseases. Front Endocrinol (Lausanne) 2025; 16:1409119. [PMID: 40034229 PMCID: PMC11872695 DOI: 10.3389/fendo.2025.1409119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Despite significant advancements in prevention and treatment, cardiometabolic diseases continue to pose a high burden of incidence and mortality. The chronic progression of these diseases necessitates the identification of early and complementary therapeutic targets to elucidate and mitigate residual risks in patient care. The gut microbiota acts as a sentinel between internal and external environments, transmitting modified risks associated with these factors to the host. Imidazole propionate (ImP), a histidine metabolite originating from the gut microbiota, gained attention after being found to impair glucose tolerance and insulin signaling several years ago. Epidemiological studies over the past five years have demonstrated a robust correlation between ImP and an increased risk of onset of type 2 diabetes (T2D) and obesity, exacerbation of kidney traits in chronic kidney disease (CKD), progression of atherosclerotic plaques, and elevated mortality rates in heart failure (HF). These findings suggest that ImP may serve as a pivotal target for the prevention and treatment of cardiometabolic diseases. Mechanistic insights have uncovered associations between ImP and insulin resistance, impaired glucose metabolism, chronic inflammation, and intestinal barrier damage. This review provides a comprehensive summary of the current evidence regarding the association between ImP and cardiometabolic impairment, highlighting its potential in advancing personalized approaches to disease prevention and management, and exploring the intricate interplay of diet, gut microbiota, and ImP in cardiovascular metabolic impairment. Overall, this review offers valuable insights into the multifaceted roles of ImP in cardiometabolic diseases, identifies current knowledge gaps, and discusses future research directions.
Collapse
Affiliation(s)
- Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, The Afiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Man Guo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Fangyuan Teng
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxia Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jiao Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaozhen Tan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Long
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
15
|
Sun T, Song B, Li B. Gut microbiota and atrial cardiomyopathy. Front Cardiovasc Med 2025; 12:1541278. [PMID: 39968343 PMCID: PMC11832500 DOI: 10.3389/fcvm.2025.1541278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Atrial cardiomyopathy is a multifaceted heart disease characterized by structural and functional abnormalities of the atria and is closely associated with atrial fibrillation and its complications. Its etiology involves a number of factors, including genetic, infectious, immunologic, and metabolic factors. Recent research has highlighted the critical role of the gut microbiota in the pathogenesis of atrial cardiomyopathy, and this is consistent with the gut-heart axis having major implications for cardiac health. The aim of this work is to bridge the knowledge gap regarding the interactions between the gut microbiota and atrial cardiomyopathy, with a particular focus on elucidating the mechanisms by which gut dysbiosis may induce atrial remodeling and dysfunction. This article provides an overview of the role of the gut microbiota in the pathogenesis of atrial cardiomyopathy, including changes in the composition of the gut microbiota and the effects of its metabolites. We also discuss how diet and exercise affect atrial cardiomyopathy by influencing the gut microbiota, as well as possible future therapeutic approaches targeting the gut-heart axis. A healthy gut microbiota can prevent disease, but ecological dysbiosis can lead to a variety of symptoms, including the induction of heart disease. We focus on the pathophysiological aspects of atrial cardiomyopathy, the impact of gut microbiota dysbiosis on atrial structure and function, and therapeutic strategies exploring modulation of the microbiota for the treatment of atrial cardiomyopathy. Finally, we discuss the role of gut microbiota in the treatment of atrial cardiomyopathy, including fecal microbiota transplantation and oral probiotics or prebiotics. Our study highlights the importance of gut microbiota homeostasis for cardiovascular health and suggests that targeted interventions on the gut microbiota may pave the way for innovative preventive and therapeutic strategies targeting atrial cardiomyopathy.
Collapse
Affiliation(s)
- Tingting Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Beibei Song
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Bo Li
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| |
Collapse
|
16
|
López-Yerena A, de Santisteban Villaplana V, Badimon L, Vilahur G, Padro T. Probiotics: A Potential Strategy for Preventing and Managing Cardiovascular Disease. Nutrients 2024; 17:52. [PMID: 39796486 PMCID: PMC11722674 DOI: 10.3390/nu17010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Probiotics are gaining recognition as a viable strategy for mitigating cardiovascular risk factors. Specifically, recent studies highlight their potential benefits in managing cholesterol levels, blood pressure, and inflammation, which are critical components in the prevention of cardiovascular diseases (CVD). This comprehensive review aims to elucidate the impact of probiotic consumption on major cardiovascular risk factors, including individuals with hypertension, type II diabetes mellitus, metabolic syndrome, hypercholesterolemia, and in secondary prevention in coronary artery disease. Scientific evidence based on human studies suggests that probiotic consumption is associated with positive effects on anthropometric measures, inflammation markers, blood pressure, glucose metabolism markers, lipid profiles, and endothelial function. However, these findings should be interpreted pragmatically and acknowledge the significant variability in results. This variability may be attributed to factors such as probiotic composition (single strain or multiple strains), the characteristics of the delivery matrix (food, capsules, and sachets), the duration of the intervention, the dosage regimen, and baseline health profiles of the participants. Incorporating probiotics as part of a comprehensive and healthy lifestyle approach can be considered a feasible strategy for both the prevention and management of CVD. However, further research is needed on factors influencing the effect of probiotics, such as: (i) optimal probiotic strain(s), (ii) appropriate dosage, (iii) duration of treatment, (iv) optimal delivery vehicle, and (v) sex-specific differences.
Collapse
Affiliation(s)
- Anallely López-Yerena
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
| | - Victoria de Santisteban Villaplana
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
- School of Pharmacy and Food Sciences, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Lina Badimon
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cardiovascular Research Foundation for Health Prevention and Innovation (FICSI), 08017 Barcelona, Spain
| | - Gemma Vilahur
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Teresa Padro
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
17
|
Li D, Chen R, Xu X, Hou Y, Li Z, Huang C, Zhang G, Wang B, Li B, Chu X. Integrated metabolomics and network pharmacology to reveal the mechanisms of Shexiang Baoxin pill against atherosclerosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156138. [PMID: 39423481 DOI: 10.1016/j.phymed.2024.156138] [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: 01/19/2024] [Revised: 07/07/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Atherosclerosis is a disease marked by the development of lipid lesions within the endothelium and continues to be a prominent contributor to global mortality. Shexiang Baoxin pill (SBP) has been employed in the management of numerous cardiovascular diseases, but the complex mechanisms by which it operates remain obscure. This research was conducted to determine the potential impact of SBP on atherosclerosis and the underlying regulatory mechanism involved. METHOD Network pharmacology was utilized to predict the key drug-disease targets, and a nontargeted metabolomic assay was applied to identify the key metabolites and metabolic pathways. A mouse atherosclerosis model was constructed to clarify the protective effect of SBP on atherosclerosis, and in vivo and in vitro tests were performed to verify the analysis results and clarify the mechanism through which SBP affects atherosclerosis. RESULTS The results show that SBP can exert a protective effect in vivo by decreasing lipid levels, plaque formation and endothelial damage. Network pharmacology and metabolomics revealed that MAPK3, AKT1 and STAT3 were the hub targets and that trimethylamine n-oxide (TMAO) was the pivotal metabolite. Due to the atherogenic effect of TMAO, the corresponding protective effect of SBP was investigated in vitro. SBP inhibited TMAO-induced endothelial cell apoptosis and oxidative stress and counteracted the upregulation of MAPK3, AKT1, and STAT3 expression. Molecular docking and enzymatic inhibition suggested that the active components of SBP could bind stably to key target proteins. CONCLUSION Taken together, based on the integrated metabolomics and network pharmacology, our findings suggest that SBP may be implicated in TMAO-induced atherosclerosis by affecting endothelial function and bile acid synthesis. We observed that SBP may ameliorate atherosclerosis by regulating TMAO levels through multiple pathways, which may provide a novel direction and insight for SBP involved in cardiovascular protection by mediating the gut-heart axis.
Collapse
Affiliation(s)
- Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China; Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruolan Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaojian Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanyuan Hou
- Department of Dermatology, The Affiliated Haici Hospital of Qingdao University, Qingdao, China
| | - Zhaoqing Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Huang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guoliang Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Banghui Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China; Department of Dermatology, The Affiliated Haici Hospital of Qingdao University, Qingdao, China
| | - Xianming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China; Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
18
|
Dharmarathne G, Kazi S, King S, Jayasinghe TN. The Bidirectional Relationship Between Cardiovascular Medications and Oral and Gut Microbiome Health: A Comprehensive Review. Microorganisms 2024; 12:2246. [PMID: 39597635 PMCID: PMC11596509 DOI: 10.3390/microorganisms12112246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of widespread morbidity and mortality. It has been found that the gut and oral microbiomes differ in individuals with CVDs compared to healthy individuals. Patients with CVDs often require long-term pharmacological interventions. While these medications have been extensively studied for their cardiovascular benefits, emerging research indicates that they may also impact the diversity and composition of the oral and gut microbiomes. However, our understanding of how these factors influence the compositions of the oral and gut microbiomes in individuals remains limited. Studies have shown that statins and beta-blockers, in particular, cause gut and oral microbial dysbiosis, impacting the metabolism and absorption of these medications. These alterations can lead to variations in drug responses, highlighting the need for personalized treatment approaches. The microbiome's role in drug metabolism and the impact of CVD medications on the microbiome are crucial in understanding these variations. However, there are very few studies in this area, and not all medications have been studied, emphasizing the necessity for further research to conclusively establish cause-and-effect relationships and determine the clinical significance of these interactions. This review will provide evidence of how the oral and gut microbiomes in patients with cardiovascular diseases (CVDs) interact with specific drugs used in CVD treatment.
Collapse
Affiliation(s)
- Gangani Dharmarathne
- Australian Laboratory Services Global, Water and Hydrographic, Hume, ACT 2620, Australia
| | - Samia Kazi
- Westmead Applied Research Centre, The University of Sydney, Sydney, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Shalinie King
- Westmead Applied Research Centre, The University of Sydney, Sydney, NSW 2145, Australia
- The Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Thilini N. Jayasinghe
- The Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- The Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
19
|
Moonsamy G, Roets-Dlamini Y, Langa CN, Ramchuran SO. Advances in Yeast Probiotic Production and Formulation for Preventative Health. Microorganisms 2024; 12:2233. [PMID: 39597622 PMCID: PMC11596959 DOI: 10.3390/microorganisms12112233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
The use of probiotics has been gaining popularity in terms of inclusion into human diets over recent years. Based on properties exerted by these organisms, several benefits have been elucidated and conferred to the host. Bacteria have been more commonly used in probiotic preparations compared to yeast candidates; however, yeast exhibit several beneficial properties, such as the prevention and treatment of diarrhea, the production of antimicrobial agents, the prevention of pathogen adherence to intestinal sites, the maintenance of microbial balance, the modulation of the immune system, antibiotic resistance, amongst others. Saccharomyces boulardii is by far the most studied strain; however, the potential for the use of other yeast candidates, such as Kluyveromyces lactis and Debaryomyces hansenii, amongst others, have also been evaluated in this review. Furthermore, a special focus has been made regarding the production considerations for yeast-based probiotics and their formulation into different delivery formats. When drafting this review, evidence suggests that the use of yeasts, both wild-type and genetically modified candidates, can extend beyond gut health to support skin, the respiratory system, and overall immune health. Hence, this review explores the potential of yeast probiotics as a safe, effective strategy for preventative health in humans, highlighting their mechanisms of action, clinical applications, and production considerations.
Collapse
Affiliation(s)
- Ghaneshree Moonsamy
- Council for Scientific and Industrial Research (CSIR) Future Production Chemicals, Meiring Naude Drive, Pretoria 0081, South Africa; (Y.R.-D.); (C.N.L.); (S.O.R.)
| | | | | | | |
Collapse
|
20
|
Warmbrunn MV, Attaye I, Horak A, Banerjee R, Massey WJ, Varadharajan V, Rampanelli E, Hao Y, Dutta S, Nemet I, Aron-Wisnewsky J, Clément K, Koopen A, Wortelboer K, Bergh PO, Davids M, Mohamed N, Kemper EM, Hazen S, Groen AK, van Raalte DH, Herrema H, Backhed F, Brown JM, Nieuwdorp M. Kinetics of imidazole propionate from orally delivered histidine in mice and humans. NPJ Biofilms Microbiomes 2024; 10:118. [PMID: 39496629 PMCID: PMC11535228 DOI: 10.1038/s41522-024-00592-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
Imidazole Propionate (ImP), a gut-derived metabolite from histidine, affects insulin signaling in mice and is elevated in type 2 diabetes (T2D). However, the source of histidine and the role of the gut microbiota remain unclear. We conducted an intervention study in mice and humans, comparing ImP kinetics in mice on a high-fat diet with varying histidine levels and antibiotics, and assessed ImP levels in healthy and T2D subjects with histidine supplementation. Results show that dietary histidine is metabolized to ImP, with antibiotic-induced gut microbiota suppression reducing ImP levels in mice. In contrast, oral histidine supplementation resulted in increases in circulating ImP levels in humans, whereas antibiotic treatment increased ImP levels, which was associated with a bloom of several bacterial genera that have been associated with ImP production, such as Lactobacilli. Our findings highlight the gut microbiota's crucial role in regulating ImP and the complexity of translating mouse models to humans.
Collapse
Affiliation(s)
- Moritz V Warmbrunn
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands.
- Amsterdam UMC, University of Amsterdam, Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, Netherlands.
| | - Ilias Attaye
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Anthony Horak
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rakhee Banerjee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - William J Massey
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Elena Rampanelli
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Youling Hao
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Sumita Dutta
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ina Nemet
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Judith Aron-Wisnewsky
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France
- Assistance Publique Hôpitaux de Paris,Pitie-Salpêtrière Hospital, Nutrition department, CRNH Ile de France, Paris, France
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France
- Assistance Publique Hôpitaux de Paris,Pitie-Salpêtrière Hospital, Nutrition department, CRNH Ile de France, Paris, France
| | - Annefleur Koopen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Koen Wortelboer
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam UMC, University of Amsterdam, Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, Netherlands
| | - Per-Olof Bergh
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mark Davids
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Nadia Mohamed
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - E Marleen Kemper
- Department of Clinical Pharmacology, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Stanley Hazen
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Albert K Groen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Daniel H van Raalte
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- VU University, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Hilde Herrema
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Fredrik Backhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden
| | - J Mark Brown
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Wang A, Guan B, Yu L, Liu Q, Hou Y, Li Z, Sun D, Xu H. Palmatine protects against atherosclerosis by gut microbiota and phenylalanine metabolism. Pharmacol Res 2024; 209:107413. [PMID: 39293583 DOI: 10.1016/j.phrs.2024.107413] [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: 05/01/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024]
Abstract
Accumulating evidence illuminated that gut microbiota directly modulates the development of atherosclerosis (AS) through interactions with metaflammation. The natural bioactive isoquinoline alkaloid palmatine (PAL), which is extracted from one of the herbs (Coptis chinensis) of the anti-AS formular, is of particular interest due to its pharmacological properties. ApoE-/- mice were administered PAL or vehicle; plaque areas, and stability were assessed by histopathological and immunohistochemistry analysis, serum glycolysis and lipid levels, and inflammation levels were also evaluated. 16S rRNA sequencing and metabolomics analysis were employed to evaluate microbial composition and serum metabolites. Microbial culture experiments were designed to reveal the target microbiota and associated metabolites. Cell culture and transcriptome were performed to elucidate the function of microbial metabolites on THP-1. PAL reduced the area of plaque and necrotic core, improving inflammatory infiltration within plaques, improving glycolipid metabolism, and reducing the levels of serum inflammatory cytokines in a dose-dependent manner. PAL treatment reshaped the composition of the gut microbiota, especially, reducing the relative abundance of Desulfovibrio piger (D. piger) in a dose-dependent manner and serum level of hippuric acid (HA). D. piger was able to convert phenylalanine into 3-phenylpropionic acid (precursor of HA). Finally, we verified HA accelerated the progression of AS and increased the secretions of inflammatory cytokines in vivo and in vitro. In conclusion, PAL exhibited anti-AS effects by regulating the gut microbiota-phenylalanine metabolism axis.
Collapse
Affiliation(s)
- Anlu Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Baoyi Guan
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Linghua Yu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qiyu Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuanlong Hou
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ziguang Li
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Daming Sun
- Laboratory of Metabolism and Drug Target Discovery, State Key Laboratory of Natural Medicines, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
22
|
Bao W, Zhang Y, Huang XJ, Gu N. The role of gut microbiome in mediating the effect of inflammatory bowel disease on hypertension: a two-step, two-sample Mendelian randomization study. Front Cardiovasc Med 2024; 11:1396973. [PMID: 39479396 PMCID: PMC11521848 DOI: 10.3389/fcvm.2024.1396973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/02/2024] [Indexed: 11/02/2024] Open
Abstract
OBJECTIVE Investigating the causal connection that exists between inflammatory bowel disease (IBD) and hypertension (HT). To gain a deeper insight into the correlation among IBD, gut microbiota, and HT, we conducted a two-step, two-sample Mendelian randomization study. METHODS An investigation of genome-wide association study (GWAS) summary-level data was utilized to conduct a two-sample Mendelian randomization (MR) analysis of genetically predicted inflammatory bowel disease: (12,882cases, 21,770controls) on Systolic/Diastolic blood pressure (N = 2,564). Subsequently, two-step MR analyses revealed that the relationship between IBD and SBP was partly mediated by Faecalicatena glycyrrhizinilyticum. The robustness of the findings was confirmed through several sensitivity assessments. RESULTS This MR study showed that increase in genetically predicted IBD was associated with higher risk of genetically predicted SBP (OR: 1.08, 95% CI: 1.01-1.16, P < 0.05) and DBP (OR: 1.09, 95% CI: 1.02-1.17, P < 0.05), respectively. Inverse variance weighted (IVW) MR analysis also showed that increase in genetically predicted IBD was associated with higher abundance Faecalicatena glycyrrhizinilyticum (OR: 1.03, 95% CI: 1.01-1.04, P < 0.05), which subsequently associated with increased SBP risk (OR: 1.42, 95% CI: 1.06-1.9, P < 0.05). Faecalicatena glycyrrhizinilyticum abundance in stool was responsible for mediating 11% of the genetically predicted IBD on SBP. CONCLUSION The research proposed a causal link between Inflammatory Bowel Disease (IBD) and Hypertension (HT), with a little percentage of the impact being influenced by Faecalicatena glycyrrhizinilyticum in stool. Mitigating gut microbiome may decrease the heightened risk of hypertension in people with inflammatory bowel disease.
Collapse
Affiliation(s)
| | | | | | - Ning Gu
- Department of Cardiovascular Disease, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
23
|
Santos AA, Duarte R, Duarte M, Arella F, Marques V, Roos S, Rodrigues CMP. Impact of Lactobacillaceae supplementation on the multi-organ axis during MASLD. Life Sci 2024; 354:122948. [PMID: 39117140 DOI: 10.1016/j.lfs.2024.122948] [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: 05/20/2024] [Revised: 07/15/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
The gut-liver axis plays a pivotal role in maintaining body homeostasis. Disruption of the gut-liver axis is linked to a multitude of diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD). Probiotic strains from the Lactobacillaceae family are commonly used to mitigate experimental MASLD. Over the years, numerous studies have demonstrated the efficacy of these probiotics, often focusing on the outcome of liver disease. This review aims to further understand MASLD as a systemic metabolic dysfunction and to highlight the effects of probiotics on multi-organ axis, including organs such as the gastrointestinal tract, pancreas, muscle, adipose tissue, and the immune system. We specifically discuss evidence on how supplementation with Lactobacillaceae strains may alleviate MASLD by not only restoring liver health but also by modulating the physiology of other organ systems.
Collapse
Affiliation(s)
- André A Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal.
| | - Raquel Duarte
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Madalena Duarte
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Fabiola Arella
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Vanda Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Sweden
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| |
Collapse
|
24
|
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: 18] [Impact Index Per Article: 18.0] [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
|
25
|
Wilcox NS, Amit U, Reibel JB, Berlin E, Howell K, Ky B. Cardiovascular disease and cancer: shared risk factors and mechanisms. Nat Rev Cardiol 2024; 21:617-631. [PMID: 38600368 PMCID: PMC11324377 DOI: 10.1038/s41569-024-01017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Cardiovascular disease (CVD) and cancer are among the leading causes of morbidity and mortality globally, and these conditions are increasingly recognized to be fundamentally interconnected. In this Review, we present the current epidemiological data for each of the modifiable risk factors shared by the two diseases, including hypertension, hyperlipidaemia, diabetes mellitus, obesity, smoking, diet, physical activity and the social determinants of health. We then review the epidemiological data demonstrating the increased risk of CVD in patients with cancer, as well as the increased risk of cancer in patients with CVD. We also discuss the shared mechanisms implicated in the development of these conditions, highlighting their inherent bidirectional relationship. We conclude with a perspective on future research directions for the field of cardio-oncology to advance the care of patients with CVD and cancer.
Collapse
Affiliation(s)
- Nicholas S Wilcox
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Uri Amit
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob B Reibel
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eva Berlin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kendyl Howell
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Yang F, Song H, Tang W, Liu L, Zhu Z, Ouyang B, Zhang L, He G, Qin W. Causal relationship between the gut microbiota, immune cells, and coronary heart disease: a mediated Mendelian randomization analysis. Front Microbiol 2024; 15:1449935. [PMID: 39161605 PMCID: PMC11332803 DOI: 10.3389/fmicb.2024.1449935] [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: 06/16/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024] Open
Abstract
Background Recent studies have shown that the gut microbiota (GM), immune cells, and coronary heart disease (CHD) are closely related, but the causal nature of these relationships is largely unknown. This study aimed to investigate this causal relationship and reveal the effect of GM and immune cells on the risk of developing CHD using mediated Mendelian randomization (MR) analysis. Methods First, we searched for data related to GM, immune cells, and CHD through published genome-wide association studies (GWAS). We filtered the single nucleotide polymorphisms (SNPs) associated with GM and immune cells and then performed the first MR analysis to identify disease-associated intestinal bacteria and disease-associated immune cells. Subsequently, three MR analyses were conducted: from disease-associated GM to disease-associated immune cells, from disease-associated immune cells to CHD, and from disease-associated GM to CHD. Each MR analysis was conducted using inverse variance weighting (IVW), MR-Egger regression, weighted median, weighted models, and simple models. Results A total of six GM and 25 immune cells were found to be associated with CHD. In the MR analysis using the inverse variance weighting (IVW) method, g__Desulfovibrio.s__Desulfovibrio_piger was associated with EM DN (CD4-CD8-) %T cells (P < 0.05 and OR > 1), EM DN (CD4-CD8-) %T cells was associated with CHD (P < 0.05 and OR < 1), and g__Desulfovibrio.s__Desulfovibrio_piger was associated with CHD (P < 0.05 and OR < 1). Conclusion An increase in the abundance of g__Desulfovibrio.s__Desulfovibrio_piger leads to an increase in the amount of EM DN (CD4-CD8-) %T cells, and an increase in the amount of EM DN (CD4-CD8-) %T cells reduces the risk of developing CHD. Our study provides some references for reducing the incidence of CHD by regulating GM and immune cells.
Collapse
Affiliation(s)
- Feifei Yang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Hui Song
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Weizhi Tang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Lingyun Liu
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Ziyi Zhu
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Bin Ouyang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Liwen Zhang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Guixin He
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Weibin Qin
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
27
|
Merenstein DJ, Tancredi DJ, Karl JP, Krist AH, Lenoir-Wijnkoop I, Reid G, Roos S, Szajewska H, Sanders ME. Is There Evidence to Support Probiotic Use for Healthy People? Adv Nutr 2024; 15:100265. [PMID: 38977065 PMCID: PMC11342770 DOI: 10.1016/j.advnut.2024.100265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
Probiotics are typically marketed as foods and dietary supplements, categories for products intended to maintain health in generally healthy populations and which, unlike drugs, cannot claim to treat or cure disease. This review addresses the existing evidence that probiotics are beneficial to healthy people. Our approach was to perform a descriptive review of efficacy evidence that probiotics can prevent urinary, vaginal, gastrointestinal, and respiratory infections, and improve risk factors associated with cardiovascular health or reduce antibiotic use. Other endpoints such as mental, dental, or immune health were not specifically addressed. We concluded that there is sufficient evidence of efficacy and safety for clinicians and consumers to consider using specific probiotics for some indications - such as the use of probiotics to support gut function during antibiotic use or to reduce the risk of respiratory tract infections - for certain people. However, we did not find a sufficiently high level of evidence to support unconditional, population-wide recommendations for other preventive endpoints we reviewed for healthy people. Although evidence for some indications is suggestive of the preventive benefits of probiotics, additional research is needed.
Collapse
Affiliation(s)
- Daniel J Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Daniel J Tancredi
- Department of Pediatrics, University of California, Davis, CA, United States
| | - J Philip Karl
- Military Nutrition Division, United States Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Alex H Krist
- Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Gregor Reid
- Departments of Microbiology and Immunology and Surgery, Western University, London, ON, Canada
| | - Stefan Roos
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Uppsala BioCenter, Uppsala, Sweden; Research & Development, BioGaia AB, Stockholm, Sweden
| | - Hania Szajewska
- Department of Paediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, CO, United States.
| |
Collapse
|
28
|
Li Y, Yao M, Xie F, Qiu Y, Zhao X, Li R. Gut microbiota as a residual risk factor causally influencing cardiac structure and function: Mendelian randomization analysis and biological annotation. Front Microbiol 2024; 15:1410272. [PMID: 39132134 PMCID: PMC11316272 DOI: 10.3389/fmicb.2024.1410272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 07/04/2024] [Indexed: 08/13/2024] Open
Abstract
Background The gut microbiota (GM) is widely acknowledged to have a significant impact on cardiovascular health and may act as a residual risk factor affecting cardiac structure and function. However, the causal relationship between GM and cardiac structure and function remains unclear. Objective This study aims to employ a two-sample Mendelian randomization (MR) approach to investigate the causal association between GM and cardiac structure and function. Methods Data on 119 GM genera were sourced from a genome-wide association study (GWAS) meta-analysis (13,266 European participants) conducted by the MiBioGen consortium, while data on 16 parameters of cardiac structure and function were obtained from the UK Biobank's GWAS of cardiac magnetic resonance imaging (up to 41,135 European participants). Inverse variance weighted (IVW), MR-Egger, and weighted median (WM) methods were utilized for causal association assessments, with sensitivity analyses conducted to reinforce the findings. Finally, biological annotation was performed on the GWAS data of GM and cardiac phenotypes with causal associations to explore potential mechanisms. Results The MR analysis, predominantly based on the IVW model, revealed 93 causal associations between the genetically predicted abundance of 44 GM genera and 16 cardiac structure and function parameters. These associations maintained consistent directions in MR-Egger and WM models, with no evidence of pleiotropy detected. Biological annotations suggest that GM may influence cardiac structure and function through pathways involved in myocardial cell development, cardiac contractility, and apoptosis. Conclusion The MR analysis supports a causal association between certain abundances of genetically predicted GM and cardiac structure and function, suggesting that GM could be a residual risk factor impacting cardiac phenotypes.
Collapse
Affiliation(s)
- Yihua Li
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meidan Yao
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- National Key Laboratory of Chinese Medicine Evidence, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fei Xie
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yijun Qiu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinjun Zhao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
29
|
Massier L, Musat N, Stumvoll M, Tremaroli V, Chakaroun R, Kovacs P. Tissue-resident bacteria in metabolic diseases: emerging evidence and challenges. Nat Metab 2024; 6:1209-1224. [PMID: 38898236 DOI: 10.1038/s42255-024-01065-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/13/2024] [Indexed: 06/21/2024]
Abstract
Although the impact of the gut microbiome on health and disease is well established, there is controversy regarding the presence of microorganisms such as bacteria and their products in organs and tissues. However, recent contamination-aware findings of tissue-resident microbial signatures provide accumulating evidence in support of bacterial translocation in cardiometabolic disease. The latter provides a distinct paradigm for the link between microbial colonizers of mucosal surfaces and host metabolism. In this Perspective, we re-evaluate the concept of tissue-resident bacteria including their role in metabolic low-grade tissue and systemic inflammation. We examine the limitations and challenges associated with studying low bacterial biomass samples and propose experimental and analytical strategies to overcome these issues. Our Perspective aims to encourage further investigation of the mechanisms linking tissue-resident bacteria to host metabolism and their potentially actionable health implications for prevention and treatment.
Collapse
Affiliation(s)
- Lucas Massier
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Niculina Musat
- Aarhus University, Department of Biology, Section for Microbiology, Århus, Denmark
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rima Chakaroun
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
30
|
Huang P, Dong Q, Wang Y, Tian Y, Wang S, Zhang C, Yu L, Tian F, Gao X, Guo H, Yi S, Li M, Liu Y, Zhang Q, Lu W, Wang G, Yang B, Cui S, Hua D, Wang X, Jiao Y, Liu L, Deng Q, Ma B, Wu T, Zou H, Shi J, Zhang H, Fan D, Sheng Y, Zhao J, Tang L, Zhang H, Sun W, Chen W, Kong X, Chen L, Zhai Q. Gut microbial genomes with paired isolates from China illustrate probiotic and cardiometabolic effects. CELL GENOMICS 2024; 4:100559. [PMID: 38740021 PMCID: PMC11228888 DOI: 10.1016/j.xgen.2024.100559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/04/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
The gut microbiome displays genetic differences among populations, and characterization of the genomic landscape of the gut microbiome in China remains limited. Here, we present the Chinese Gut Microbial Reference (CGMR) set, comprising 101,060 high-quality metagenomic assembled genomes (MAGs) of 3,707 nonredundant species from 3,234 fecal samples across primarily rural Chinese locations, 1,376 live isolates mainly from lactic acid bacteria, and 987 novel species relative to worldwide databases. We observed region-specific coexisting MAGs and MAGs with probiotic and cardiometabolic functionalities. Preliminary mouse experiments suggest a probiotic effect of two Faecalibacillus intestinalis isolates in alleviating constipation, cardiometabolic influences of three Bacteroides fragilis_A isolates in obesity, and isolates from the genera Parabacteroides and Lactobacillus in host lipid metabolism. Our study expands the current microbial genomes with paired isolates and demonstrates potential host effects, contributing to the mechanistic understanding of host-microbe interactions.
Collapse
Affiliation(s)
- Pan Huang
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Quanbin Dong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China; Department of Gastroenterology, Changzhou Medical Center, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Yifeng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China; Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yunfan Tian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Shunhe Wang
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaoxiang Gao
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hang Guo
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Shanrong Yi
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Mingyang Li
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yang Liu
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qingsong Zhang
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Dongxu Hua
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Xiuchao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yuwen Jiao
- Department of Gastroenterology, Changzhou Medical Center, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Lu Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Qiufeng Deng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Beining Ma
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Tingting Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Huayiyang Zou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jing Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Haifeng Zhang
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Daming Fan
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yanhui Sheng
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Liming Tang
- Department of Gastroenterology, Changzhou Medical Center, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China; Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.
| | - Lianmin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China; Department of Gastroenterology, Changzhou Medical Center, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, China.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources & School of Food Science and Technology, Jiangnan University, Wuxi, China.
| |
Collapse
|
31
|
Fernández-Ruiz I. Gut bacteria can break down cholesterol. Nat Rev Cardiol 2024; 21:357. [PMID: 38627565 DOI: 10.1038/s41569-024-01026-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
|
32
|
Heavey MK, Hazelton A, Wang Y, Garner M, Anselmo AC, Arthur JC, Nguyen J. Targeted delivery of the probiotic Saccharomyces boulardii to the extracellular matrix enhances gut residence time and recovery in murine colitis. Nat Commun 2024; 15:3784. [PMID: 38710716 PMCID: PMC11074276 DOI: 10.1038/s41467-024-48128-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
Probiotic and engineered microbe-based therapeutics are an emerging class of pharmaceutical agents. They represent a promising strategy for treating various chronic and inflammatory conditions by interacting with the host immune system and/or delivering therapeutic molecules. Here, we engineered a targeted probiotic yeast platform wherein Saccharomyces boulardii is designed to bind to abundant extracellular matrix proteins found within inflammatory lesions of the gastrointestinal tract through tunable antibody surface display. This approach enabled an additional 24-48 h of probiotic gut residence time compared to controls and 100-fold increased probiotic concentrations within the colon in preclinical models of ulcerative colitis in female mice. As a result, pharmacodynamic parameters including colon length, colonic cytokine expression profiles, and histological inflammation scores were robustly improved and restored back to healthy levels. Overall, these studies highlight the potential for targeted microbial therapeutics as a potential oral dosage form for the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Mairead K Heavey
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Anthony Hazelton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yuyan Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mitzy Garner
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Aaron C Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- VitaKey Incorporation, Durham, NC, 27701, USA
| | - Janelle C Arthur
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Center for Gastrointestinal Biology and Disease, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
33
|
Chen B, Zeng G, Sun L, Jiang C. When smoke meets gut: deciphering the interactions between tobacco smoking and gut microbiota in disease development. SCIENCE CHINA. LIFE SCIENCES 2024; 67:854-864. [PMID: 38265598 DOI: 10.1007/s11427-023-2446-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/09/2023] [Indexed: 01/25/2024]
Abstract
Tobacco smoking is a prevalent and detrimental habit practiced worldwide, increasing the risk of various diseases, including chronic obstructive pulmonary disease (COPD), cardiovascular disease, liver disease, and cancer. Although previous research has explored the detrimental health effects of tobacco smoking, recent studies suggest that gut microbiota dysbiosis may play a critical role in these outcomes. Numerous tobacco smoke components, such as nicotine, are found in the gastrointestinal tract and interact with gut microbiota, leading to lasting impacts on host health and diseases. This review delves into the ways tobacco smoking and its various constituents influence gut microbiota composition and functionality. We also summarize recent advancements in understanding how tobacco smoking-induced gut microbiota dysbiosis affects host health. Furthermore, this review introduces a novel perspective on how changes in gut microbiota following smoking cessation may contribute to withdrawal syndrome and the degree of health improvements in smokers.
Collapse
Affiliation(s)
- Bo Chen
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Guangyi Zeng
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Lulu Sun
- State Key Laboratory of Women's Reproductive Health and Fertility Promotion, Peking University, Beijing, 100191, China.
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, 100191, China.
| | - Changtao Jiang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Women's Reproductive Health and Fertility Promotion, Peking University, Beijing, 100191, China.
| |
Collapse
|
34
|
Spasova N, Somleva D, Krastev B, Ilieva R, Borizanova A, Svinarov D, Kinova E, Goudev A. Association of the trimethylamine N-oxide with cardiovascular risk and vascular alterations in middle-aged patients with risk factors for cardiovascular diseases. Biosci Rep 2024; 44:BSR20232090. [PMID: 38669041 PMCID: PMC12046062 DOI: 10.1042/bsr20232090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/15/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO) is synthesized by the intestinal microbiota and is an independent predictor of cardiovascular disease (CVD). However, its underlying mechanisms remain unclear. We investigated TMAO levels across different CVD-risk patient groups, and evaluated associations between TMAO and vascular alterations (e.g., arterial stiffness, intima-media thickness [IMT], and the presence and grade of carotid artery plaques [CAPs]). METHODS We examined 95 patients (58.5 ± 7.3 years): 40 with clinical atherosclerotic cardiovascular disease (ASCVD), 40 with atherosclerosis risk factors (RF), and 15 controls. Arterial stiffness was measured by Carotid-Femoral Pulse Wave Velocity (C-F PWV). B-mode ultrasound was used to evaluate the presence and grade of CAPs and carotid IMT (CIMT). TMAO was measured by high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) and results were presented as the median (interquartile range). RESULTS TMAO levels were higher in patients with ASCVD (251.5 [164.5] µg/l) when compared with patients with RFs (194.0 [174] µg/l, P=0.04) and controls (122.0 (77) µg/l, P<0.001). A significant correlation was observed between TMAO and PWV (r = 0.31, P=0.003), which was not confirmed after adjustment for RFs. TMAO levels were significantly correlated with plaque score (r = 0.46, P<0.001) and plaque height (r=0.41, P=0.003), and were independent predictors for grade III plaques (odds ratio [OR] = 1.002, confidence interval (CI) 95%: 1.000047-1.003, P=0.044). CONCLUSIONS TMAO levels are increased with expanded CVD risk. Across different types of vascular damage, TMAO is associated with atherosclerotic changes.
Collapse
Affiliation(s)
- Natalia Spasova
- Department of Cardiology, University Hospital, UMHAT “Tsaritsa Yoanna - ISUL”, Sofia, Bulgaria
| | - Desislava Somleva
- Department of Cardiology, University Hospital, UMHAT “Tsaritsa Yoanna - ISUL”, Sofia, Bulgaria
| | - Bozhidar Krastev
- Department of Cardiology, University Hospital, UMHAT “Tsaritsa Yoanna - ISUL”, Sofia, Bulgaria
| | - Radostina Ilieva
- Department of Cardiology, University Hospital, UMHAT “Tsaritsa Yoanna - ISUL”, Sofia, Bulgaria
| | - Angelina Borizanova
- Department of Cardiology, University Hospital, UMHAT “Tsaritsa Yoanna - ISUL”, Sofia, Bulgaria
| | - Dobrin Svinarov
- University Hospital Alexandrovska, Faculty of Medicine, Medical University, Sofia
| | - Elena Kinova
- Department of Cardiology, University Hospital, UMHAT “Tsaritsa Yoanna - ISUL”, Sofia, Bulgaria
| | - Assen Goudev
- Department of Cardiology, University Hospital, UMHAT “Tsaritsa Yoanna - ISUL”, Sofia, Bulgaria
| |
Collapse
|
35
|
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
|
36
|
Zhang S, Liu R, Ma Y, Ma Y, Feng H, Ding X, Zhang Q, Li Y, Shan J, Bian H, Zhu R, Meng Q. Lactiplantibacillus plantarum ATCC8014 Alleviates Postmenopausal Hypercholesterolemia in Mice by Remodeling Intestinal Microbiota to Increase Secondary Bile Acid Excretion. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6236-6249. [PMID: 38484389 DOI: 10.1021/acs.jafc.3c08232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Hypercholesterolemia poses a significant cardiovascular risk, particularly in postmenopausal women. The anti-hypercholesterolemic properties of Lactiplantibacillus plantarum ATCC8014 (LP) are well recognized; however, its improving symptoms on postmenopausal hypercholesterolemia and the possible mechanisms have yet to be elucidated. Here, we utilized female ApoE-deficient (ApoE-/-) mice undergoing bilateral ovariectomy, fed a high-fat diet, and administered 109 colony-forming units (CFU) of LP for 13 consecutive weeks. LP intervention reduces total cholesterol (TC) and triglyceride (TG) accumulation in the serum and liver and accelerates their fecal excretion, which is mainly accomplished by increasing the excretion of fecal secondary bile acids (BAs), thereby facilitating cholesterol conversion. Correlation analysis revealed that lithocholic acid (LCA) is an important regulator of postmenopausal lipid abnormalities. LP can reduce LCA accumulation in the liver and serum while enhancing its fecal excretion, accomplished by elevating the relative abundances of Allobaculum and Olsenella in the ileum. Our findings demonstrate that postmenopausal lipid dysfunction is accompanied by abnormalities in BA metabolism and dysbiosis of the intestinal microbiota. LP holds therapeutic potential for postmenopausal hypercholesterolemia. Its effectiveness in ameliorating lipid dysregulation is primarily achieved through reshaping the diversity and abundance of the intestinal microbiota to correct BA abnormalities.
Collapse
Affiliation(s)
- Shurui Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ronghui Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuxin Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuting Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Han Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xue Ding
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qichun Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinjun Shan
- First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Huimin Bian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruigong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qinghai Meng
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
37
|
Zhang XY, Khakisahneh S, Han SY, Song EJ, Nam YD, Kim H. Ginseng extracts improve circadian clock gene expression and reduce inflammation directly and indirectly through gut microbiota and PI3K signaling pathway. NPJ Biofilms Microbiomes 2024; 10:24. [PMID: 38503759 PMCID: PMC10950852 DOI: 10.1038/s41522-024-00498-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024] Open
Abstract
Despite the potential benefits of herbal medicines for therapeutic application in preventing and treating various metabolic disorders, the mechanisms of action were understood incompletely. Ginseng (Panax ginseng), a commonly employed plant as a dietary supplement, has been reported to play its hot property in increasing body temperature and improving gut health. However, a comprehensive understanding of the mechanisms by which ginseng regulates body temperature and gut health is still incomplete. This paper illustrates that intermittent supplementation with ginseng extracts improved body temperature rhythm and suppressed inflammatory responses in peripheral metabolic organs of propylthiouracil (PTU)-induced hypothermic rats. These effects were associated with changes in gut hormone secretion and the microbiota profile. The in-vitro studies in ICE-6 cells indicate that ginseng extracts can not only act directly on the cell to regulate the genes related to circadian clock and inflammation, but also may function through the gut microbiota and their byproducts such as lipopolysaccharide. Furthermore, administration of PI3K inhibitor blocked ginseng or microbiota-induced gene expression related with circadian clock and inflammation in vitro. These findings demonstrate that the hot property of ginseng may be mediated by improving circadian clock and suppressing inflammation directly or indirectly through the gut microbiota and PI3K-AKT signaling pathways.
Collapse
Affiliation(s)
- Xue-Ying Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Saeid Khakisahneh
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, 814 Siksa-dong, Ilsandong-gu, Goyang-si, 10326, Republic of Korea
| | - Song-Yi Han
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, 814 Siksa-dong, Ilsandong-gu, Goyang-si, 10326, Republic of Korea
| | - Eun-Ji Song
- Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, 245, Republic of Korea
- Department of Food Biotechnology, Korea University of Science and Technology, Wanju, Republic of Korea
| | - Young-Do Nam
- Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, 245, Republic of Korea.
- Department of Food Biotechnology, Korea University of Science and Technology, Wanju, Republic of Korea.
| | - Hojun Kim
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, 814 Siksa-dong, Ilsandong-gu, Goyang-si, 10326, Republic of Korea.
| |
Collapse
|
38
|
Angelini G, Galvao Neto M, Boskoski I, Caristo ME, Russo S, Proto L, Previti E, Olsson L, Aggarwal H, Pezzica S, Ferrari E, Bove V, Genco A, Bornstein S, Tremaroli V, Gastaldelli A, Mingrone G. ForePass endoscopic bypass device for obesity and insulin resistance-metabolic treatment in a swine model. Gut 2024; 73:568-572. [PMID: 38129101 PMCID: PMC10958257 DOI: 10.1136/gutjnl-2023-331335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Affiliation(s)
- Giulia Angelini
- Department of Translational Medicine and Surgery, Universtà Cattlica del Sacro Cuore, Rome, Italy
| | - Manoel Galvao Neto
- Bariatric Endoscopy department of Mohak Bariatric and Robotic Center, Sri Aurobindo Medical College, Indore, India, India, India
| | - Ivo Boskoski
- Digestive Endoscopy Unit, Digestive Endoscopy Unit. Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia, Rome, Italy
| | | | - Sara Russo
- Department of Translational Medicine and Surgery, Universtà Cattlica del Sacro Cuore, Rome, Italy
| | - Luca Proto
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Elena Previti
- Department of Translational Medicine and Surgery, Universtà Cattlica del Sacro Cuore, Rome, Italy
| | - Lisa Olsson
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hobby Aggarwal
- Department of Molecular and Clinical Medicine, University of Gothenburg, Goteborg, Sweden
| | | | | | - Vincenzo Bove
- Digestive Endoscopy Unit, Digestive Endoscopy Unit. Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia, Rome, Italy
| | - Alfredo Genco
- Surgery, University of Rome La Sapienza, Rome, Italy
| | - Stefan Bornstein
- Universitatsklinikum Carl Gustav Carus, Dresden, Sachsen, Germany
| | - Valentina Tremaroli
- Department of Molecular and Clinical Medicine Goteborg, University of Gothenburg, Goteborg, Västra Götaland, Sweden
| | | | - Geltrude Mingrone
- Department of Translational Medicine and Surgery, Universtà Cattlica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
- Diabetes, King's College London School of Medical Education, London, UK
| |
Collapse
|
39
|
Huang K, Li Z, He X, Dai J, Huang B, Shi Y, Fan D, Zhang Z, Liu Y, Li N, Zhang Z, Peng J, Liu C, Zeng R, Cen Z, Wang T, Yang W, Cen M, Li J, Yuan S, Zhang L, Hu D, Huang S, Chen P, Lai P, Lin L, Wen J, Zhao Z, Huang X, Yuan L, Zhou L, Wu H, Huang L, Feng K, Wang J, Liao B, Cai W, Deng X, Li Y, Li J, Hu Z, Yang L, Li J, Zhuo Y, Zhang F, Lin L, Luo Y, Zhang W, Ni Q, Hong X, Chang G, Zhang Y, Guan D, Cai W, Lu Y, Li F, Yan L, Ren M, Li L, Chen S. Gut microbial co-metabolite 2-methylbutyrylcarnitine exacerbates thrombosis via binding to and activating integrin α2β1. Cell Metab 2024; 36:598-616.e9. [PMID: 38401546 DOI: 10.1016/j.cmet.2024.01.014] [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: 05/24/2023] [Revised: 11/08/2023] [Accepted: 01/25/2024] [Indexed: 02/26/2024]
Abstract
Thrombosis represents the leading cause of death and disability upon major adverse cardiovascular events (MACEs). Numerous pathological conditions such as COVID-19 and metabolic disorders can lead to a heightened thrombotic risk; however, the underlying mechanisms remain poorly understood. Our study illustrates that 2-methylbutyrylcarnitine (2MBC), a branched-chain acylcarnitine, is accumulated in patients with COVID-19 and in patients with MACEs. 2MBC enhances platelet hyperreactivity and thrombus formation in mice. Mechanistically, 2MBC binds to integrin α2β1 in platelets, potentiating cytosolic phospholipase A2 (cPLA2) activation and platelet hyperresponsiveness. Genetic depletion or pharmacological inhibition of integrin α2β1 largely reverses the pro-thrombotic effects of 2MBC. Notably, 2MBC can be generated in a gut-microbiota-dependent manner, whereas the accumulation of plasma 2MBC and its thrombosis-aggravating effect are largely ameliorated following antibiotic-induced microbial depletion. Our study implicates 2MBC as a metabolite that links gut microbiota dysbiosis to elevated thrombotic risk, providing mechanistic insight and a potential therapeutic strategy for thrombosis.
Collapse
Affiliation(s)
- Kan Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China; Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xi He
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Jun Dai
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Bingding Huang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, Guangdong 518118, China
| | - Yongxia Shi
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Dongxiao Fan
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zefeng Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Yunchong Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Na Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zhongyu Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Jiangyun Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Chenshu Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Renli Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Zhipeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Wenchao Yang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Meifeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Jingyu Li
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, Guangdong 518118, China
| | - Shuai Yuan
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Lu Zhang
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Dandan Hu
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Shuxiang Huang
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Pin Chen
- National Supercomputer Center in Guangzhou, School of Computer Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Peilong Lai
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Liyan Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Jielu Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Zhengde Zhao
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xiuyi Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lining Yuan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Lifang Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Haoliang Wu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lihua Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Kai Feng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Jian Wang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Baolin Liao
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Weiping Cai
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Xilong Deng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Yueping Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Jianping Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Zhongwei Hu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Li Yang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Jiaojiao Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Youguang Zhuo
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Fuchun Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Lin Lin
- Department of Respiratory Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Yifeng Luo
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Sun Yat-sen University, Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Wei Zhang
- Wuhan Metware Biotechnology Co., Ltd., Wuhan, Hubei 430070, China
| | - Qianlin Ni
- Wuhan Metware Biotechnology Co., Ltd., Wuhan, Hubei 430070, China
| | - Xiqiang Hong
- Wuhan Metware Biotechnology Co., Ltd., Wuhan, Hubei 430070, China
| | - Guangqi Chang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yang Zhang
- School of Public Health, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Dongxian Guan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Weikang Cai
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568, USA
| | - Yutong Lu
- National Supercomputer Center in Guangzhou, School of Computer Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Fang Li
- Department of Obstetrics and Gynecology, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510620, China
| | - Li Yan
- Guangdong Clinical Research Center for Metabolic Diseases (Diabetes), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Meng Ren
- Guangdong Clinical Research Center for Metabolic Diseases (Diabetes), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Linghua Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China.
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China.
| |
Collapse
|
40
|
Sun Y, Wang X, Li L, Zhong C, Zhang Y, Yang X, Li M, Yang C. The role of gut microbiota in intestinal disease: from an oxidative stress perspective. Front Microbiol 2024; 15:1328324. [PMID: 38419631 PMCID: PMC10899708 DOI: 10.3389/fmicb.2024.1328324] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Recent studies have indicated that gut microbiota-mediated oxidative stress is significantly associated with intestinal diseases such as colorectal cancer, ulcerative colitis, and Crohn's disease. The level of reactive oxygen species (ROS) has been reported to increase when the gut microbiota is dysregulated, especially when several gut bacterial metabolites are present. Although healthy gut microbiota plays a vital role in defending against excessive oxidative stress, intestinal disease is significantly influenced by excessive ROS, and this process is controlled by gut microbiota-mediated immunological responses, DNA damage, and intestinal inflammation. In this review, we discuss the relationship between gut microbiota and intestinal disease from an oxidative stress perspective. In addition, we also provide a summary of the most recent therapeutic approaches for preventing or treating intestinal diseases by modifying gut microbiota.
Collapse
Affiliation(s)
- Yiqi Sun
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xurui Wang
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Li
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Zhong
- Traditional Chinese Medicine Department of Orthopaedic and Traumatic, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Zhang
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, China
| | - Xiangdong Yang
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, China
| | - Mingyue Li
- Special Needs Outpatient Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Yang
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
41
|
Luqman A, Hassan A, Ullah M, Naseem S, Ullah M, Zhang L, Din AU, Ullah K, Ahmad W, Wang G. Role of the intestinal microbiome and its therapeutic intervention in cardiovascular disorder. Front Immunol 2024; 15:1321395. [PMID: 38343539 PMCID: PMC10853344 DOI: 10.3389/fimmu.2024.1321395] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
The gut microbiome is a heterogeneous population of microbes comprising viruses, bacteria, fungi, and protozoa. Such a microbiome is essential for sustaining host equilibrium, and its impact on human health can be altered by a variety of factors such as external variables, social behavior, age, nutrition, and genetics. Gut microbes' imbalances are related to a variety of chronic diseases including cancer, obesity, and digestive disorders. Globally, recent findings show that intestinal microbes have a significant role in the formation of cardiovascular disease (CVD), which is still the primary cause of fatalities. Atherosclerosis, hypertension, diabetes, inflammation, and some inherited variables are all cardiovascular risk variables. However, studies found correlations between metabolism, intestinal flora, and dietary intake. Variations in the diversity of gut microbes and changes in their activity are thought to influence CVD etiology. Furthermore, the gut microbiota acts as an endocrine organ, producing bioactive metabolites such as TMA (trimethylamine)/TMAO (trimethylamine N-oxide), SCFA (short-chain fatty acids), and bile acids, which have a substantial impact on host wellness and disease by multiple mechanisms. The purpose of this overview is to compile current evidence highlighting the intricate links between gut microbiota, metabolites, and the development of CVD. It focuses on how intestinal dysbiosis promotes CVD risk factors such as heart failure, hypertension, and atherosclerosis. This review explores the normal physiology of intestinal microbes and potential techniques for targeting gut bacteria for CVD treatment using various microbial metabolites. It also examines the significance of gut bacteria in disease treatment, including supplements, prebiotics, probiotics, antibiotic therapies, and fecal transplantation, which is an innovative approach to the management of CVD. As a result, gut bacteria and metabolic pathways become increasingly attractive as potential targets for CVD intervention.
Collapse
Affiliation(s)
- Ameer Luqman
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
| | - Adil Hassan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing, China
| | - Mehtab Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Sahar Naseem
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Mehraj Ullah
- School of Fermentation Engineering Tianjin University of Science and Technology, Tianjin, China
| | | | - Ahmad Ud Din
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, United States
| | - Kamran Ullah
- Department of Biology, The University of Haripur, Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Waqar Ahmad
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
| |
Collapse
|
42
|
Yuan L, Li Y, Chen M, Xue L, Wang J, Ding Y, Gu Q, Zhang J, Zhao H, Xie X, Wu Q. Therapeutic applications of gut microbes in cardiometabolic diseases: current state and perspectives. Appl Microbiol Biotechnol 2024; 108:156. [PMID: 38244075 PMCID: PMC10799778 DOI: 10.1007/s00253-024-13007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Cardiometabolic disease (CMD) encompasses a range of diseases such as hypertension, atherosclerosis, heart failure, obesity, and type 2 diabetes. Recent findings about CMD's interaction with gut microbiota have broadened our understanding of how diet and nutrition drive microbes to influence CMD. However, the translation of basic research into the clinic has not been smooth, and dietary nutrition and probiotic supplementation have yet to show significant evidence of the therapeutic benefits of CMD. In addition, the published reviews do not suggest the core microbiota or metabolite classes that influence CMD, and systematically elucidate the causal relationship between host disease phenotypes-microbiome. The aim of this review is to highlight the complex interaction of the gut microbiota and their metabolites with CMD progression and to further centralize and conceptualize the mechanisms of action between microbial and host disease phenotypes. We also discuss the potential of targeting modulations of gut microbes and metabolites as new targets for prevention and treatment of CMD, including the use of emerging technologies such as fecal microbiota transplantation and nanomedicine. KEY POINTS: • To highlight the complex interaction of the gut microbiota and their metabolites with CMD progression and to further centralize and conceptualize the mechanisms of action between microbial and host disease phenotypes. • We also discuss the potential of targeting modulations of gut microbes and metabolites as new targets for prevention and treatment of CMD, including the use of emerging technologies such as FMT and nanomedicine. • Our study provides insight into identification-specific microbiomes and metabolites involved in CMD, and microbial-host changes and physiological factors as disease phenotypes develop, which will help to map the microbiome individually and capture pathogenic mechanisms as a whole.
Collapse
Affiliation(s)
- Lin Yuan
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Moutong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Liang Xue
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Ding
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Science & Engineering, Jinan University, Guangzhou, 510632, China
| | - Qihui Gu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Hui Zhao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| |
Collapse
|
43
|
Yuan D, Xu N, Song Y, Zhang Z, Xu J, Liu Z, Tang X, Han Y, Chen Y, Zhang Y, Zhu P, Guo X, Wang Z, Liu R, Wang Q, Yao Y, Feng Y, Zhao X, Yuan J. Association Between Free Fatty Acids and Cardiometabolic Risk in Coronary Artery Disease: Results From the PROMISE Study. J Clin Endocrinol Metab 2023; 109:125-134. [PMID: 37540767 DOI: 10.1210/clinem/dgad416] [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: 01/14/2023] [Indexed: 08/06/2023]
Abstract
CONTEXT The association between free fatty acids (FFAs) and unfavorable clinical outcomes has been reported in the general population. However, evidence in the secondary prevention population is relatively scarce. OBJECTIVE We aimed to examine the relationship between FFA and cardiovascular risk in patients with coronary artery disease (CAD). METHODS This study was based on a multicenter cohort of patients with CAD enrolled from January 2015 to May 2019. The primary outcome was all-cause death. Secondary outcomes included cardiac death and major adverse cardiovascular events (MACE), a composite of death, myocardial infarction, and unplanned revascularization. RESULTS During a follow-up of 2 years, there were 468 (3.0%) all-cause deaths, 335 (2.1%) cardiac deaths, and 1279 (8.1%) MACE. Elevated FFA levels were independently associated with increased risks of all-cause death, cardiac death, and MACE (all P < .05). Moreover, When FFA were combined with an original model derived from the Cox regression, there were significant improvements in discrimination and reclassification for prediction of all-cause death (net reclassification improvement [NRI] 0.245, P < .001; integrated discrimination improvement [IDI] 0.004, P = .004), cardiac death (NRI 0.269, P < .001; IDI 0.003, P = .006), and MACE (NRI 0.268, P < .001; IDI 0.004, P < .001). Notably, when stratified by age, we found that the association between FFA with MACE risk appeared to be stronger in patients aged ≥60 years compared with those aged <60 years. CONCLUSION In patients with CAD, FFAs are associated with all-cause death, cardiac death, and MACE. Combined evaluation of FFAs with other traditional risk factors could help identify high-risk individuals who may require closer monitoring and aggressive treatment.
Collapse
Affiliation(s)
- Deshan Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Na Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Ying Song
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Zheng Zhang
- Department of Cardiology, the First Hospital of Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Jingjing Xu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Xiaofang Tang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Yaling Han
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110000, People's Republic of China
| | - Yan Chen
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Yongzhen Zhang
- Department of Cardiology, Peking University Third Hospital, Beijing 100037, People's Republic of China
| | - Pei Zhu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Xiaogang Guo
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine (FAHZU), Hangzhou 310000, People's Republic of China
| | - Zhifang Wang
- Department of Cardiology, Xinxiang Central Hospital, Xinxiang 453000, People's Republic of China
| | - Ru Liu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Qingsheng Wang
- Department of Cardiology, the First Hospital of Qinhuangdao, Qinhuangdao 066000, People's Republic of China
| | - Yi Yao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Yingqing Feng
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangzhou 510000, People's Republic of China
| | - Xueyan Zhao
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Jinqing Yuan
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| |
Collapse
|
44
|
Huang H, Kuang Z, Mo R, Meng M, Cai Y, Ni X. The preliminary evidence on the association of the gut microbiota with stroke risk stratification in South Chinese population. Front Cell Infect Microbiol 2023; 13:1227450. [PMID: 38222855 PMCID: PMC10785002 DOI: 10.3389/fcimb.2023.1227450] [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: 05/23/2023] [Accepted: 11/24/2023] [Indexed: 01/16/2024] Open
Abstract
Aims This study aimed to investigate the association between the gut microbiota and the risk of stroke. Methods Faecal samples from 60 participants in South China, including 45 individuals with risk factors for stroke and 15 healthy controls, were collected and subjected to 16S rRNA sequencing. A bioinformatics analysis was performed to characterise the gut microbial diversity and taxonomic compositions at different risk levels (low, moderate, and high) of stroke. Functional prediction and correlation analyses between the microbiota and laboratory markers were performed to explore the potential mechanisms. Results A significant difference in beta diversity was observed between the participants from the stroke risk and healthy control groups. Linear discriminant effect size analysis revealed a large number of vascular beneficial bacteria enriched in the participants from the healthy control and low-risk groups, but a few vascular harmful bacteria were more abundant in the participants from the high-risk group than in those from the other groups. In addition, Anaerostipes, Clostridium_XlVb, and Flavonifractor, all of which belonged to the Firmicutes phylum, were enriched in the participants from the low-risk group, and their relative abundances gradually decreased as the stroke risk increased. Spearman's analysis revealed that these outstanding microbiota correlated with the levels of triglycerides, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, white blood cells, neutrophils, and carotid intima-media thickness. Conclusion The preliminary evidence suggests that gut microbiota is associated with stroke risk. It potentially ameliorates atherosclerosis by targeting lipid metabolism and inflammation. This provides novel insights into the early screening of stroke risk and primary prevention.
Collapse
Affiliation(s)
- Haiyan Huang
- The Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuoran Kuang
- The Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruibi Mo
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Miaomiao Meng
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yefeng Cai
- The Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojia Ni
- The Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
45
|
Liu Y, Huang Y, He Q, Dou Z, Zeng M, Wang X, Li S. From heart to gut: Exploring the gut microbiome in congenital heart disease. IMETA 2023; 2:e144. [PMID: 38868221 PMCID: PMC10989834 DOI: 10.1002/imt2.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 06/14/2024]
Abstract
Congenital heart disease (CHD) is a prevalent birth defect and a significant contributor to childhood mortality. The major characteristics of CHD include cardiovascular malformations and hemodynamical disorders. However, the impact of CHD extends beyond the circulatory system. Evidence has identified dysbiosis of the gut microbiome in patients with CHD. Chronic hypoxia and inflammation associated with CHD affect the gut microbiome, leading to alterations in its number, abundance, and composition. The gut microbiome, aside from providing essential nutrients, engages in direct interactions with the host immune system and indirect interactions via metabolites. The abnormal gut microbiome or its products can translocate into the bloodstream through an impaired gut barrier, leading to an inflammatory state. Metabolites of the gut microbiome, such as short-chain fatty acids and trimethylamine N-oxide, also play important roles in the development, treatment, and prognosis of CHD. This review discusses the role of the gut microbiome in immunity, gut barrier, neurodevelopment, and perioperative period in CHD. By fostering a better understanding of the cross-talk between CHD and the gut microbiome, this review aims to contribute to improve clinical management and outcomes for CHD patients.
Collapse
Affiliation(s)
- Yuze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yuan Huang
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Qiyu He
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Zheng Dou
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Min Zeng
- Department of Pediatric Intensive Care Unit, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Xu Wang
- Department of Pediatric Intensive Care Unit, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Shoujun Li
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| |
Collapse
|
46
|
Zhao Q, Chen Y, Huang W, Zhou H, Zhang W. Drug-microbiota interactions: an emerging priority for precision medicine. Signal Transduct Target Ther 2023; 8:386. [PMID: 37806986 PMCID: PMC10560686 DOI: 10.1038/s41392-023-01619-w] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 07/20/2023] [Accepted: 08/24/2023] [Indexed: 10/10/2023] Open
Abstract
Individual variability in drug response (IVDR) can be a major cause of adverse drug reactions (ADRs) and prolonged therapy, resulting in a substantial health and economic burden. Despite extensive research in pharmacogenomics regarding the impact of individual genetic background on pharmacokinetics (PK) and pharmacodynamics (PD), genetic diversity explains only a limited proportion of IVDR. The role of gut microbiota, also known as the second genome, and its metabolites in modulating therapeutic outcomes in human diseases have been highlighted by recent studies. Consequently, the burgeoning field of pharmacomicrobiomics aims to explore the correlation between microbiota variation and IVDR or ADRs. This review presents an up-to-date overview of the intricate interactions between gut microbiota and classical therapeutic agents for human systemic diseases, including cancer, cardiovascular diseases (CVDs), endocrine diseases, and others. We summarise how microbiota, directly and indirectly, modify the absorption, distribution, metabolism, and excretion (ADME) of drugs. Conversely, drugs can also modulate the composition and function of gut microbiota, leading to changes in microbial metabolism and immune response. We also discuss the practical challenges, strategies, and opportunities in this field, emphasizing the critical need to develop an innovative approach to multi-omics, integrate various data types, including human and microbiota genomic data, as well as translate lab data into clinical practice. To sum up, pharmacomicrobiomics represents a promising avenue to address IVDR and improve patient outcomes, and further research in this field is imperative to unlock its full potential for precision medicine.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, PR China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, PR China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, PR China.
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, PR China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, PR China.
- Central Laboratory of Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, Changsha, 410013, PR China.
| |
Collapse
|
47
|
Li Y, Zou C, Li J, Wang W, Wang F, Guo Y. Airway Microbiome Composition and Co-Occurrence Network Are Associated with Inflammatory Phenotypes of Asthma. Int Arch Allergy Immunol 2023; 184:1254-1263. [PMID: 37690443 PMCID: PMC10733928 DOI: 10.1159/000533315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/27/2023] [Indexed: 09/12/2023] Open
Abstract
INTRODUCTION The composition and co-occurrence network of the airway microbiome might influence the asthma inflammatory phenotype. Airway microbiota change with asthma phenotypes, and the structure of the bacterial community in the airway might differ between different asthma inflammatory phenotypes and may also influence therapy results. Identifying airway microbiota can help to investigate the role that microbiota play in the asthma inflammatory process. METHODS Induced sputum from 55 subjects and 12 healthy subjects from Beijing, China, was collected and analyzed for bacterial microbiota. Microbiome diversity, composition, co-occurrence networks, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were predicted and compared between the study groups. RESULTS Significant differences in the sputum microbiome composition, co-occurrence network, and predicted functional pathways were observed between the two inflammatory phenotypes. Asthmatics in the low FeNO group exhibited lower α-diversity in the sputum microbiota and had higher abundance of the phylum Proteobacteria compared with that of the high FeNO group. The network in the high FeNO group was more "closed" and "connected" compared with that of the low FeNO group, and an alteration in the abundance of keystone species T. socranskii was found. Significantly different predicted metabolic subfunctions including nucleotide metabolism, lipid metabolism, energy metabolism, replication and repair, and drug resistance antimicrobial and carbohydrate metabolism between the two studied phenotypes were also observed. CONCLUSION Our findings confirm that the airway microbiota is associated with the asthma inflammation process. The differences in the airway microbiome composition and co-occurrence network may affect distinct asthma inflammatory phenotypes, suggesting the possibility that more targeted therapies could be applied based on the airway bacterial genera.
Collapse
Affiliation(s)
- Yi Li
- State Key Laboratory of Severe Weather of CMA, Chinese Academy of Meteorological Sciences, Beijing, China
| | - Congying Zou
- Department of Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jieying Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Feiran Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yue Guo
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
48
|
Dai H, Hou T, Wang Q, Hou Y, Wang T, Zheng J, Lin H, Zhao Z, Li M, Wang S, Zhang D, Dai M, Zheng R, Lu J, Xu Y, Chen Y, Ning G, Wang W, Bi Y, Xu M. Causal relationships between the gut microbiome, blood lipids, and heart failure: a Mendelian randomization analysis. Eur J Prev Cardiol 2023; 30:1274-1282. [PMID: 37195998 DOI: 10.1093/eurjpc/zwad171] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 05/19/2023]
Abstract
AIMS Studies have linked gut microbiome and heart failure (HF). However, their causal relationships and potential mediating factors have not been well defined. To investigate the causal relationships between the gut microbiome and HF and the mediating effect of potential blood lipids by using genetics. METHODS AND RESULTS We performed a bidirectional and mediation Mendelian randomization (MR) study using summary statistics from the genome-wide association studies of gut microbial taxa (Dutch Microbiome Project, n = 7738), blood lipids (UK Biobank, n = 115 078), and a meta-analysis of HF (115 150 cases and 1550 331 controls). We applied the inverse-variance weighted estimation method as the primary method, with several other estimators as complementary methods. The multivariable MR approach based on Bayesian model averaging (MR-BMA) was used to prioritize the most likely causal lipids. Six microbial taxa are suggestively associated with HF causally. The most significant taxon was the species Bacteroides dorei [odds ratio = 1.059, 95% confidence interval (CI) = 1.022-1.097, P-value = 0.0017]. The MR-BMA analysis showed that apolipoprotein B (ApoB) was the most likely causal lipid for HF (the marginal inclusion probability = 0.717, P-value = 0.005). The mediation MR analysis showed that ApoB mediated the causal effects of species B. dorei on HF (proportion mediated = 10.1%, 95% CI = 0.2-21.6%, P-value = 0.031). CONCLUSION The study suggested a causal relationship between specific gut microbial taxa and HF and that ApoB might mediate this relationship as the primary lipid determinant of HF.
Collapse
Affiliation(s)
- Huajie Dai
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tianzhichao Hou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qi Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanan Hou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Di Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Meng Dai
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
49
|
Liang Y, Ju D, Liu W, Wu D, Zhao Y, Du Y, Li X, Zhao M. Natural Shikonin Potentially Alters Intestinal Flora to Alleviate Acute Inflammation. Microorganisms 2023; 11:2139. [PMID: 37763983 PMCID: PMC10534322 DOI: 10.3390/microorganisms11092139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 09/29/2023] Open
Abstract
Shikonin, derived from the herb Lithospermum erythrorhizon (Purple Cromwell), is extensively utilized in traditional Chinese medicine as an anti-inflammatory agent; however, its effect on the intestinal flora is not yet known. Herein, we demonstrate that, compared to a blank control group, the intragastric administration of shikonin suppressed the swelling rate of ears in a mouse model of acute inflammation in a dose-dependent manner via animal experiments; furthermore, the 20 mg/kg shikonin treatment exhibited the highest inhibitory effect. In formal animal experimentation, we discovered that the inhibitory effect of shikonin with 20 mg/kg on inflammation was closely linked to the intestinal flora, whereby the microbiota phylum was altered in feces through a 16S rDNA sequencing analysis, implying that shikonin improves gut microbiota structures and compositions to counteract inflammation. Notably, using a real-time quantitative polymerase chain reaction (RT-qPCR), a Western blotting assay, and an immunohistochemistry (IHC) assay, we found that inflammatory cytokines such as TNF-α, IL-6, and IL-1β reduced in both the shikonin-administration group and the positive control group than those in the blank control group, as expected. To the best of our knowledge, this is the first study to outline the underlying mechanism through which shikonin acts on gut microbes to alleviate acute inflammation, providing an alternative mechanism for shikonin to become a preventive agent in countering inflammation.
Collapse
Affiliation(s)
- Ying Liang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China; (Y.L.); (W.L.); (D.W.); (Y.D.); (X.L.)
| | - Dongen Ju
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China;
| | - Wenna Liu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China; (Y.L.); (W.L.); (D.W.); (Y.D.); (X.L.)
| | - Dan Wu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China; (Y.L.); (W.L.); (D.W.); (Y.D.); (X.L.)
| | - Yujia Zhao
- Department of Oncology, The First Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an 710086, China;
| | - Yaya Du
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China; (Y.L.); (W.L.); (D.W.); (Y.D.); (X.L.)
| | - Xi Li
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China; (Y.L.); (W.L.); (D.W.); (Y.D.); (X.L.)
| | - Minggao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China; (Y.L.); (W.L.); (D.W.); (Y.D.); (X.L.)
| |
Collapse
|
50
|
Sayols-Baixeras S, Dekkers KF, Baldanzi G, Jönsson D, Hammar U, Lin YT, Ahmad S, Nguyen D, Varotsis G, Pita S, Nielsen N, Eklund AC, Holm JB, Nielsen HB, Ericson U, Brunkwall L, Ottosson F, Larsson A, Ericson D, Klinge B, Nilsson PM, Malinovschi A, Lind L, Bergström G, Sundström J, Ärnlöv J, Engström G, Smith JG, Orho-Melander M, Fall T. Streptococcus Species Abundance in the Gut Is Linked to Subclinical Coronary Atherosclerosis in 8973 Participants From the SCAPIS Cohort. Circulation 2023; 148:459-472. [PMID: 37435755 PMCID: PMC10399955 DOI: 10.1161/circulationaha.123.063914] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND Gut microbiota have been implicated in atherosclerotic disease, but their relation with subclinical coronary atherosclerosis is unclear. This study aimed to identify associations between the gut microbiome and computed tomography-based measures of coronary atherosclerosis and to explore relevant clinical correlates. METHODS We conducted a cross-sectional study of 8973 participants (50 to 65 years of age) without overt atherosclerotic disease from the population-based SCAPIS (Swedish Cardiopulmonary Bioimage Study). Coronary atherosclerosis was measured using coronary artery calcium score and coronary computed tomography angiography. Gut microbiota species abundance and functional potential were assessed with shotgun metagenomics sequencing of fecal samples, and associations with coronary atherosclerosis were evaluated with multivariable regression models adjusted for cardiovascular risk factors. Associated species were evaluated for association with inflammatory markers, metabolites, and corresponding species in saliva. RESULTS The mean age of the study sample was 57.4 years, and 53.7% were female. Coronary artery calcification was detected in 40.3%, and 5.4% had at least 1 stenosis with >50% occlusion. Sixty-four species were associated with coronary artery calcium score independent of cardiovascular risk factors, with the strongest associations observed for Streptococcus anginosus and Streptococcus oralis subsp oralis (P<1×10-5). Associations were largely similar across coronary computed tomography angiography-based measurements. Out of the 64 species, 19 species, including streptococci and other species commonly found in the oral cavity, were associated with high-sensitivity C-reactive protein plasma concentrations, and 16 with neutrophil counts. Gut microbial species that are commonly found in the oral cavity were negatively associated with plasma indole propionate and positively associated with plasma secondary bile acids and imidazole propionate. Five species, including 3 streptococci, correlated with the same species in saliva and were associated with worse dental health in the Malmö Offspring Dental Study. Microbial functional potential of dissimilatory nitrate reduction, anaerobic fatty acid β-oxidation, and amino acid degradation were associated with coronary artery calcium score. CONCLUSIONS This study provides evidence of an association of a gut microbiota composition characterized by increased abundance of Streptococcus spp and other species commonly found in the oral cavity with coronary atherosclerosis and systemic inflammation markers. Further longitudinal and experimental studies are warranted to explore the potential implications of a bacterial component in atherogenesis.
Collapse
Affiliation(s)
- Sergi Sayols-Baixeras
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
- CIBER Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain (S.S.-B.)
| | - Koen F. Dekkers
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
| | - Gabriel Baldanzi
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
| | - Daniel Jönsson
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
- Public Dental Service of Skåne, Lund, Sweden (D.J.)
- Departments of Periodontology (D.J., B.K.), Faculty of Odontology, Malmö University, Sweden
| | - Ulf Hammar
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
| | - Yi-Ting Lin
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden (Y.-T.L., J.Ä.)
| | - Shafqat Ahmad
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
- Preventive Medicine Division, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA (S.A.)
| | - Diem Nguyen
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
| | - Georgios Varotsis
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
| | - Sara Pita
- Clinical Microbiomics A/S, Copenhagen, Denmark (S.P., N.N., A.C.E., J.B.H., H.B.N.)
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark (S.P.)
| | - Nynne Nielsen
- Clinical Microbiomics A/S, Copenhagen, Denmark (S.P., N.N., A.C.E., J.B.H., H.B.N.)
| | - Aron C. Eklund
- Clinical Microbiomics A/S, Copenhagen, Denmark (S.P., N.N., A.C.E., J.B.H., H.B.N.)
| | - Jacob B. Holm
- Clinical Microbiomics A/S, Copenhagen, Denmark (S.P., N.N., A.C.E., J.B.H., H.B.N.)
| | - H. Bjørn Nielsen
- Clinical Microbiomics A/S, Copenhagen, Denmark (S.P., N.N., A.C.E., J.B.H., H.B.N.)
| | - Ulrika Ericson
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
| | - Louise Brunkwall
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
- Clinical Studies Sweden, Forum Söder, Region Skåne, Lund, Sweden (L.B.)
| | - Filip Ottosson
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark (F.O.)
| | - Anna Larsson
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
| | - Dan Ericson
- Cariology (D.E.), Faculty of Odontology, Malmö University, Sweden
| | - Björn Klinge
- Departments of Periodontology (D.J., B.K.), Faculty of Odontology, Malmö University, Sweden
- Department of Dental Medicine, Karolinska Institutet, Solna, Sweden (B.K.)
| | - Peter M. Nilsson
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden (P.M.N.)
| | - Andrei Malinovschi
- Clinical Physiology (A.M.), Department of Medical Sciences, Uppsala University, Sweden
| | - Lars Lind
- Clinical Epidemiology (L.L., J.S.), Department of Medical Sciences, Uppsala University, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden (G. Bergström)
- Department of Clinical Physiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden (G. Bergström)
| | - Johan Sundström
- Clinical Epidemiology (L.L., J.S.), Department of Medical Sciences, Uppsala University, Sweden
- The George Institute for Global Health, University of New South Wales, Sydney, Australia (J.S.)
| | - Johan Ärnlöv
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden (Y.-T.L., J.Ä.)
- School of Health and Social Studies, Dalarna University, Falun, Sweden (J.Ä.)
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
| | - J. Gustav Smith
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Sweden (J.G.S.)
| | - Marju Orho-Melander
- Department of Clinical Sciences in Malmö, Lund University, Sweden (D.J., U.E., L.B., F.O., A.L., P.M.N., G.E., M.O.-M.)
| | - Tove Fall
- Molecular Epidemiology and Science for Life Laboratory (S.S.-B., K.F.D., G. Baldanzi, U.H., Y.-T.L., S.A., D.N., G.V., T.F.), Department of Medical Sciences, Uppsala University, Sweden
| |
Collapse
|