1
|
Yang C, Camargo Tavares L, Lee HC, Steele JR, Ribeiro RV, Beale AL, Yiallourou S, Carrington MJ, Kaye DM, Head GA, Schittenhelm RB, Marques FZ. Faecal metaproteomics analysis reveals a high cardiovascular risk profile across healthy individuals and heart failure patients. Gut Microbes 2025; 17:2441356. [PMID: 39709554 DOI: 10.1080/19490976.2024.2441356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
The gut microbiota is a crucial link between diet and cardiovascular disease (CVD). Using fecal metaproteomics, a method that concurrently captures human gut and microbiome proteins, we determined the crosstalk between gut microbiome, diet, gut health, and CVD. Traditional CVD risk factors (age, BMI, sex, blood pressure) explained < 10% of the proteome variance. However, unsupervised human protein-based clustering analysis revealed two distinct CVD risk clusters (low-risk and high-risk) with different blood pressure (by 9 mmHg) and sex-dependent dietary potassium and fiber intake. In the human proteome, the low-risk group had lower angiotensin-converting enzymes, inflammatory proteins associated with neutrophil extracellular trap formation and auto-immune diseases. In the microbial proteome, the low-risk group had higher expression of phosphate acetyltransferase that produces SCFAs, particularly in fiber-fermenting bacteria. This model identified severity across phenotypes in heart failure patients and long-term risk of cardiovascular events in a large population-based cohort. These findings underscore multifactorial gut-to-host mechanisms that may underlie risk factors for CVD.
Collapse
Affiliation(s)
- Chaoran Yang
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Leticia Camargo Tavares
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Han-Chung Lee
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Joel R Steele
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | | | - Anna L Beale
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
| | - Stephanie Yiallourou
- Preclinical Disease and Prevention Unit, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Melinda J Carrington
- Preclinical Disease and Prevention Unit, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - David M Kaye
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- School of Translational Medicine, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Geoffrey A Head
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Pharmacology, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Victorian Heart Institute, Monash University, Clayton, Australia
| |
Collapse
|
2
|
Snelson M, Muralitharan RR, Liu CF, Markó L, Forslund SK, Marques FZ, Tang WHW. Gut-Heart Axis: The Role of Gut Microbiota and Metabolites in Heart Failure. Circ Res 2025; 136:1382-1406. [PMID: 40403109 PMCID: PMC12101525 DOI: 10.1161/circresaha.125.325516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 05/24/2025]
Abstract
Heart failure is a global health issue with significant mortality and morbidity. There is increasing evidence that alterations in the gastrointestinal microbiome, gut epithelial permeability, and gastrointestinal disorders contribute to heart failure progression through various pathways, including systemic inflammation, metabolic dysregulation, and modulation of cardiac function. Moreover, several medications used to treat heart failure directly impact the microbiome. The relationship between the gastrointestinal tract and the heart is bidirectional, termed the gut-heart axis. It is increasingly understood that diet-derived microbial metabolites are key mechanistic drivers of the gut-heart axis. This includes, for example, trimethylamine N-oxide and short-chain fatty acids. This review discusses current insights into the interplay between heart failure, its associated risk factors, and the gut microbiome, focusing on key metabolic pathways, the role of dietary interventions, and the potential for gut-targeted therapies. Understanding these complex interactions could pave the way for novel strategies to mitigate heart failure progression and improve patient outcomes.
Collapse
Affiliation(s)
- Matthew Snelson
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Rikeish R. Muralitharan
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Chia-Feng Liu
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland OH, USA
| | - Lajos Markó
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
| | - Sofia K. Forslund
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Francine Z. Marques
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - W. H. Wilson Tang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland OH, USA
| |
Collapse
|
3
|
Camargo Tavares L, Muralitharan RR, Snelson M, Marques FZ. Rare pathogenic variants in G-protein-coupled receptor genes involved in gut-to-host communication are associated with cardiovascular disease risk. Cardiovasc Res 2025:cvaf070. [PMID: 40402583 DOI: 10.1093/cvr/cvaf070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/02/2025] [Accepted: 02/23/2025] [Indexed: 05/24/2025] Open
Affiliation(s)
- Leticia Camargo Tavares
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 2, Victorian Heart Hospital 631 Blackburn Road Clayton, Melbourne, VIC 3168, Australia
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 2, Victorian Heart Hospital 631 Blackburn Road Clayton, Melbourne, VIC 3168, Australia
| | - Matthew Snelson
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 2, Victorian Heart Hospital 631 Blackburn Road Clayton, Melbourne, VIC 3168, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 2, Victorian Heart Hospital 631 Blackburn Road Clayton, Melbourne, VIC 3168, Australia
- Baker Heart and Diabetes Institute, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| |
Collapse
|
4
|
Barber TM, Kabisch S, Pfeiffer AFH, Weickert MO. The Gut Microbiome as a Key Determinant of the Heritability of Body Mass Index. Nutrients 2025; 17:1713. [PMID: 40431453 PMCID: PMC12114430 DOI: 10.3390/nu17101713] [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: 04/19/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
The pathogenesis of obesity is complex and incompletely understood, with an underlying interplay between our genetic architecture and obesogenic environment. The public understanding of the development of obesity is shrouded in myths with widespread societal misconceptions. Body Mass Index (BMI) is a highly heritable trait. However, despite reports from recent genome-wide association studies, only a small proportion of the overall heritability of BMI is known to be lurking within the human genome. Other non-genetic heritable traits may contribute to BMI. The gut microbiome is an excellent candidate, implicating complex interlinks with hypothalamic control of appetite and metabolism via entero-endocrine, autonomic, and neuro-humeral pathways. The neonatal gut microbiome derived from the mother via transgenerational transmission (vaginal delivery and breastfeeding) tends to have a permanence within the gut. Conversely, non-maternally derived gut microbiota manifest mutability that responds to changes in lifestyle and diet. We should all strive to optimize our lifestyles and ensure a diet that is replete with varied and unprocessed plant-based foods to establish and nurture a healthy gut microbiome. Women of reproductive age should optimize their gut microbiome, particularly pre-conception, ante- and postnatally to enable the establishment of a healthy neonatal gut microbiome in their offspring. Finally, we should redouble our efforts to educate the populace on the pathogenesis of obesity, and the role of heritable (but modifiable) factors such as the gut microbiome. Such renewed understanding and insights would help to promote the widespread adoption of healthy lifestyles and diets, and facilitate a transition from our current dispassionate and stigmatized societal approach towards people living with obesity towards one that is epitomized by understanding, support, and compassion.
Collapse
Affiliation(s)
- Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK;
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
| | - Stefan Kabisch
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany (A.F.H.P.)
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany (A.F.H.P.)
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK;
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| |
Collapse
|
5
|
Lambert GW, Patel M, Lambert EA. The Influence of the Sympathetic Nervous System on Cardiometabolic Health in Response to Weight Gain or Weight Loss. Metabolites 2025; 15:286. [PMID: 40422864 DOI: 10.3390/metabo15050286] [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: 03/19/2025] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Alterations in sympathetic nervous activity are evident in response to changes in body weight. Sympathetic nervous activity and sympathetic responses to weight change are regionalized, with alterations in end organ function dependent on the changes occurring in the brain regulatory pathways invoked and in the effector organs engaged. The obesity-induced activation of the sympathetic nervous system likely contributes to the initiation and worsening of cardiometabolic risk factors, including elevated blood pressure, cardiac dysfunction, dyslipidaemia, increased fasting blood glucose, insulin resistance, and non-alcoholic steatohepatitis. Unintended weight loss, as occurs in cachexia, is driven, at least in part, by the activation of sympathetic nervous-stimulated thermogenesis. The complexity of sympathetic nervous regulation renders the use of global measures of sympathetic activity problematic and the development of targeted therapies difficult, but these are not without promise or precedent. Knowledge of the central and peripheral pathways involved in sympathetic nervous regulation has opened up opportunities for pharmacological, surgical, and device-based approaches to mitigating the burden of disease development and progression. In this narrative review, we elaborate on sympathetic activity in response to changes in body weight, the brain pathways involved, and the cardiovascular and metabolic risks associated with perturbations in regional sympathetic activity.
Collapse
Affiliation(s)
- Gavin W Lambert
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Mariya Patel
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Elisabeth A Lambert
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
- Medical Technology Victoria (MedTechVic) Research Hub, Hawthorn, VIC 3122, Australia
| |
Collapse
|
6
|
Mahgoup EM. "Gut Microbiota as a Therapeutic Target for Hypertension: Challenges and Insights for Future Clinical Applications" "Gut Microbiota and Hypertension Therapy". Curr Hypertens Rep 2025; 27:14. [PMID: 40261509 DOI: 10.1007/s11906-025-01331-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW Systemic hypertension is a major risk factor for cardiovascular disease and remains challenging to manage despite the widespread use of antihypertensive medications and lifestyle modifications. This review explores the role of gut microbiota in hypertension development and regulation, highlighting key mechanisms such as inflammation, gut-brain axis modulation, and bioactive metabolite production. We also assess the potential of microbiota-targeted therapies for hypertension management. RECENT FINDINGS Emerging evidence indicates that microbial dysbiosis, high-salt diets, and gut-derived metabolites such as short-chain fatty acids (SCFAs) and bile acids significantly influence blood pressure regulation. Preclinical and early clinical studies suggest that interventions targeting gut microbiota, including probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), and dietary modifications, may help modulate hypertension. However, variability in gut microbiota composition among individuals and limited human trial data pose challenges to translating these findings into clinical practice. While microbiota-based therapies show promise for hypertension management, further research is needed to establish their efficacy and long-term effects. Large-scale, standardized clinical trials are crucial for understanding the therapeutic potential and limitations of gut microbiota interventions. A deeper understanding of the gut-hypertension axis could lead to novel, personalized treatment strategies for hypertension.
Collapse
Affiliation(s)
- Elsayed M Mahgoup
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt.
- Department of Internal Medicine, Division of Cardiovascular Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
7
|
González-Correa C, Moleón J, Miñano S, Robles-Vera I, de la Visitación N, Guerra-Hernández E, Toral M, Jiménez R, Duarte J, Romero M. Protective Effect of Dietary Fiber on Blood Pressure and Vascular Dysfunction Through Regulation of Sympathetic Tone and Immune Response in Genetic Hypertension. Phytother Res 2025; 39:1858-1875. [PMID: 40122676 DOI: 10.1002/ptr.8484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
The mechanisms underlying the antihypertensive effect of dietary fibers remain poorly understood. This study investigates whether dietary fiber supplementation can prevent cardiovascular damage and high blood pressure in a genetic model of neurogenic hypertension. Six-week-old male spontaneously hypertensive rats (SHR) and their respective normotensive control, Wistar Kyoto rats (WKY), were divided into four groups: Untreated WKY, untreated SHR, SHR treated with resistant starch (SHR + RS), and SHR treated with inulin-type fructans (SHR + ITF) for 12 weeks. Additionally, a faecal microbiota transplantation (FMT) experiment was conducted, transferring faecal content from treated SHR donors to recipient SHRs. A diet rich in RS fiber reduced vascular oxidative stress, inflammation, and high blood pressure. These protective effects were associated with a reshaped gut microbiota, leading to increased short-chain fatty acid production, reduced endotoxemia, decreased sympathetic activity, and a restored balance between Th17 and Treg lymphocytes in mesenteric lymph nodes and aorta. Elevated plasma levels of acetate and butyrate in the SHR + RS group correlated with increased expression of aortic GPR41, GRP43 and PPARδ. Conversely, ITF treatment failed to prevent hypertension or endothelial dysfunction in SHR. FMT from the SHR + RS group to recipient SHR partially replicated these beneficial effects. This study highlights the antihypertensive benefits of dietary insoluble RS fiber, which are attributed to enhanced short-chain fatty acids production in the gut. This leads to improved gut permeability, reduced sympathetic tone, and diminished vascular T-cell accumulation. Therefore, dietary interventions with RS fiber may offer promising therapeutic strategies for preventing hypertension.
Collapse
Affiliation(s)
- Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | | | - Marta Toral
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| |
Collapse
|
8
|
Tillett BJ, Dwiyanto J, Secombe KR, George T, Zhang V, Anderson D, Duggan E, Giri R, Loo D, Stoll T, Morrison M, Begun J, Hill MM, Gurzov EN, Bell KJ, Saad S, Barlow CK, Creek DJ, Chong CW, Mariño E, Hamilton-Williams EE. SCFA biotherapy delays diabetes in humanized gnotobiotic mice by remodeling mucosal homeostasis and metabolome. Nat Commun 2025; 16:2893. [PMID: 40133336 PMCID: PMC11937418 DOI: 10.1038/s41467-025-58319-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: 04/17/2024] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Type 1 diabetes (T1D) is linked to an altered gut microbiota characterized by reduced short-chain fatty acid (SCFA) production. Oral delivery of a SCFA-yielding biotherapy in adults with T1D was followed by increased SCFAs, altered gut microbiota and immunoregulation, as well as delaying diabetes in preclinical models. Here, we show that SCFA-biotherapy in humans is accompanied by remodeling of the gut proteome and mucosal immune homeostasis. Metabolomics showed arginine, glutamate, nucleotide and tryptophan metabolism were enriched following the SCFA-biotherapy, and found metabolites that correlated with glycemic control. Fecal microbiota transfer demonstrated that the microbiota of SCFA-responders delayed diabetes progression in humanized gnotobiotic mice. The protected mice increased similar metabolite pathways to the humans including producing aryl-hydrocarbon receptor ligands and reducing inflammatory mucosal immunity and increasing IgA production in the gut. These data demonstrate that a potent SCFA immunomodulator promotes multiple beneficial pathways and supports targeting the microbiota as an approach against T1D. Trial registration: Australia New Zealand Clinical Trials Registry ACTRN12618001391268.
Collapse
Affiliation(s)
- Bree J Tillett
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jacky Dwiyanto
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kate R Secombe
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Thomas George
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Vivian Zhang
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Dovile Anderson
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, MelbourneVIC, Australia
| | - Emily Duggan
- Translational Research Institute, Brisbane, QLD, Australia
| | - Rabina Giri
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | - Dorothy Loo
- Translational Research Institute, Brisbane, QLD, Australia
| | - Thomas Stoll
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Mark Morrison
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Jakob Begun
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | - Michelle M Hill
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Esteban N Gurzov
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | - Kirstine J Bell
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Sonia Saad
- Department of Medicine, Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Monash University, MelbourneVIC, Australia
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Darren J Creek
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, MelbourneVIC, Australia
| | - Chun Wie Chong
- Monash University Microbiome Research Centre, School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
| | - Eliana Mariño
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
- ImmunoBiota Therapeutics Pty Ltd, Melbourne, VIC, Australia.
| | | |
Collapse
|
9
|
Li L, Zhang Y, Huang Z, Hong Y, Cheng L, Gu Z. Steric Inhibition by Butyryl Groups on Gut Microbial Amylases Significantly Impacts In Vitro Fecal Fermentation of Butyrylated Starch. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6943-6954. [PMID: 40048501 DOI: 10.1021/acs.jafc.4c10445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Colonic starch fermentation requires the assistance of gut microbial amylases (GMAs). However, it remains unknown whether chemically substituted butyryl groups induce a steric inhibitory effect on GMAs analogous to that observed with pancreatic amylase during the intestinal digestion of butyrylated starch. In this study, we investigated the in vitro fermentation and enzymatic hydrolysis performance of three types of butyrylated starch. The results showed that the esterolysis of butyryl groups was a rate-limiting process, and the fermentation of butyrylated starch was partially inhibited by steric inhibition of the butyryl groups on GMAs. X-ray photoelectron spectroscopy (XPS) results further confirmed the negative correlation between the fermentation rate of starch and the relative content of butyryl groups accessible to the gut microbiota. Moreover, analyses of the enzymatic characteristics and the resulting hydrolysate composition demonstrated that a relatively high multiple attack degree (MAD) made GMAs more susceptible to steric inhibition by butyryl groups, thus producing more malto-oligosaccharides, which are preferred by butyrate-producing bacteria. These findings provide important insights into the fermentation behavior of butyrylated starch from the perspective of interactions between microbial amylases and starch.
Collapse
Affiliation(s)
- Lingjin Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yi Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zehao Huang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, China
| | - Li Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, China
| |
Collapse
|
10
|
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
|
11
|
Wang S, Yan K, Dong Y, Chen Y, Song J, Chen Y, Liu X, Qi R, Zhou X, Zhong J, Li J. The influence of microplastics on hypertension-associated cardiovascular injury via the modulation of gut microbiota. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125760. [PMID: 39880356 DOI: 10.1016/j.envpol.2025.125760] [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: 09/11/2024] [Revised: 11/15/2024] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Microplastics (MPs) have been found to interfere with the gut microbiota and compromise the integrity of the gut barrier. Excessive exposure to MPs markedly elevates the risk of cardiovascular disease, yet their influence on hypertension remains elusive, calling for investigation into their potential impacts on blood pressure (BP) regulation. In the present study, an increase in the concentration of MPs was observed in the fecal samples of individuals suffering from hypertension, as compared to the controls. Oral administration of MPs led to obvious increases in systolic, diastolic and mean BP levels in mice. MPs were associated with promoting myocardial hypertrophy, fibrosis, and cardiac remodeling through alterations in gut microbial composition, such as Prevotella and Coprobacillus, or fecal metabolites Betaine and Glycyrrhetinic acid. The hypertensive damage mediated by MPs was significantly mitigated by the high-fiber diet or antibiotics that targeted the gut microbiota. Notablely, fecal microbiota transplantation from mice treated with MPs led to an increase in systolic BP levels and the development of cardiac dysfunction. Our findings offer valuable insights into the complex interplay between MPs and the gut microbiome in the context of hypertension, and suggest potential strategies for reducing the vascular and cardiac injury caused by MPs.
Collapse
Affiliation(s)
- Siyuan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Kaixin Yan
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ying Dong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yihang Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jiawei Song
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yufei Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing ChaoYang Hospital, Capital Medical University, Beijing, China
| | - Ruiqiang Qi
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xinyu Zhou
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - Jing Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Shremo Msdi A, Haghparast A, Garey KW, Wang EM. Microbiome-Based Therapeutics for Salt-Sensitive Hypertension: A Scoping Review. Nutrients 2025; 17:825. [PMID: 40077695 PMCID: PMC11901721 DOI: 10.3390/nu17050825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 01/29/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The purpose of this scoping review was to provide a comprehensive understanding of the current knowledge concerning the gut microbiome and SCFAs as emerging treatments for salt-sensitive hypertension. Relevant animal and human studies were identified via PubMed through August 2024. Twenty-four human (n = 9) and animal (n = 15) trials were included. Most human studies were observational (n = 6), aiming to compare gut microbiota differences between hypertensive and normotensive individuals. Three human studies evaluated microbiome-based interventions either via a sodium-restricted diet (n = 2) or prebiotic supplementation (n = 1). Fifteen animal trials involving either mice or rats were identified, all of which were interventional. These included dietary changes (n = 9), probiotic treatments (n = 1), postbiotic primarily bacterial metabolites (n = 4), and live biotherapeutic products (n = 4). All interventions were effective in decreasing blood pressure. Microbiome-based therapies as biologic modifiers for salt-sensitive hypertension are emerging. Substantial knowledge gaps remain, warranting further research to fully explore this promising therapeutic avenue.
Collapse
Affiliation(s)
- Abdulwhab Shremo Msdi
- Department of Pharmacy Practice and Translational Research, College of Pharmacy, University of Houston, 4349 Martin Luther King Boulevard, Houston, TX 77204, USA
| | | | | | | |
Collapse
|
13
|
Christensen LJ, Larsen AM, Homilius C, Gopalasingam N, Moeslund N, Berg-Hansen K, Boedtkjer E, Jensen RV, Johannsen M, Hansen J, Seefeldt J, Bøtker HE, Schütz J, Hørsdal OK, Nielsen BRR. Butyrate increases cardiac output and causes vasorelaxation in a healthy porcine model. Life Sci 2025; 363:123407. [PMID: 39828226 DOI: 10.1016/j.lfs.2025.123407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Butyrate, a short-chain fatty acid, has shown potential to improve left ventricular (LV) function and induce vasorelaxation in rodents. Butyrate may either be produced by the microbiome in the colon, be ingested or administered intravenously. This study aimed to evaluate effects of butyrate on cardiac output (CO) and associated hemodynamic variables in a porcine model. METHODS In a randomized, blinded crossover study, ten healthy 60-kg pigs were given three hour infusions of 600 mM butyrate and equimolar sodium chloride (control). CO was measured by thermodilution via a pulmonary artery catheter. LV contractility was assessed using pressure-volume admittance catheterization. Additionally, isolated porcine coronary arteries were exposed to butyrate in a wire myograph to evaluate vasorelaxation. RESULTS Butyrate infusion increased plasma butyrate concentration to 0.53 mM (95 % confidence interval (CI): 0.49 to 0.58 mM, P < 0.58 mM, P < 0.001) and CO by 1.6 L/min (95 % CI: 1.0 to 2.1 L/min, P < 0.001) compared with the control. Heart rate, LV ejection fraction, cardiac efficiency and dP/dtmax rose, while systemic vascular resistance, arterial elastance, mean arterial pressure and LV end-systolic volume decreased. Load-independent LV contractility and stroke volume did not significantly differ. In the myograph, porcine coronary arteries relaxed in response to butyrate in a concentration-dependent manner. CONCLUSION Butyrate increases cardiac output and lowers vascular resistance in a large animal model, through increased HR and systemic vasorelaxation. Load-independent LV contractility was not significantly altered. We observed indices of increased end-organ perfusion. These potentially beneficial cardiovascular properties of butyrate should be further studied.
Collapse
Affiliation(s)
- Lasse Juul Christensen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aaarhus N, Denmark.
| | - Alexander Møller Larsen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aaarhus N, Denmark
| | - Casper Homilius
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus, Denmark
| | - Nigopan Gopalasingam
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aaarhus N, Denmark; Department of Cardiology, Gødstrup Hospital, Hospitalsparken 15, 7400 Herning, Denmark
| | - Niels Moeslund
- Department of Cardiothoracic Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark
| | - Kristoffer Berg-Hansen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aaarhus N, Denmark
| | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus, Denmark
| | - Rebekka Vibjerg Jensen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark
| | - Mogens Johannsen
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aaarhus N, Denmark
| | - Jakob Hansen
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aaarhus N, Denmark
| | - Jacob Seefeldt
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aaarhus N, Denmark
| | - Julian Schütz
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aaarhus N, Denmark
| | - Oskar Kjærgaard Hørsdal
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aaarhus N, Denmark
| | - Bent Roni Ranghøj Nielsen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 69, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aaarhus N, Denmark
| |
Collapse
|
14
|
Khan MM, Kirabo A. Fiber, Fatty Acids, and Blood Pressure: A Gut-Level Solution. Circ Res 2025; 136:358-360. [PMID: 39946444 PMCID: PMC11839179 DOI: 10.1161/circresaha.125.326065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Affiliation(s)
- Mohd Mabood Khan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 (M.M.K., A.K.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 (M.M.K., A.K.)
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology, and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| |
Collapse
|
15
|
R Muralitharan R, Zheng T, Dinakis E, Xie L, Barbaro-Wahl A, Jama HA, Nakai M, Paterson M, Leung KC, McArdle Z, Mirabito Colafella K, Johnson C, Qin W, Salimova E, Bitto NJ, Kaparakis-Liaskos M, Kaye DM, O'Donnell JA, Mackay CR, Marques FZ. Gut Microbiota Metabolites Sensed by Host GPR41/43 Protect Against Hypertension. Circ Res 2025; 136:e20-e33. [PMID: 39840468 DOI: 10.1161/circresaha.124.325770] [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: 10/25/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Fermentation of dietary fiber by the gut microbiota leads to the production of metabolites called short-chain fatty acids, which lower blood pressure and exert cardioprotective effects. Short-chain fatty acids activate host signaling responses via the functionally redundant receptors GPR41 (G-protein-coupled receptor 41) and GPR43 (G-protein-coupled receptor 43), which are highly expressed by immune cells. Whether and how these receptors protect against hypertension or mediate the cardioprotective effects of dietary fiber remains unknown. METHODS Cardiovascular phenotype was assessed in untreated and Ang II (angiotensin II) treated hypertensive wild-type and GPR41/43 knockout (KO) double knockout male mice fed diets with different levels of fiber content. Some mice received TLR4 (toll-like receptor 4)-antagonist treatment and bone marrow chimeras. SNPs (single-nucleotide polymorphisms) associated with GPR41 and GPR43 expression were assessed in UK Biobank participants. RESULTS Untreated GPR41/43KO mice had unaltered blood pressure but had greater cardiac and renal collagen deposition with higher macrophage numbers in the kidney compared with wild-type mice. Ang II-treated GPR41/43KO mice showed higher systolic blood pressure, cardiorenal weights and collagen deposition, and increased gut permeability, which allows the translocation of gastrointestinal bacterial components such as lipopolysaccharides into the circulation. The use of an antagonist to the lipopolysaccharide receptor, TLR4, a potent proinflammatory signaling molecule, restored the cardiovascular phenotype in GPR41/43KO mice. The lack of GPR41/43 expression in the immune compartment was sufficient to lead to a worsened hypertensive phenotype. We also demonstrate that GPR41/43 is, at least partially, responsible for the blood pressure-lowering and cardioprotective effects of a high-fiber diet. Finally, using the UK Biobank, we provide translational evidence that variants associated with lower expression of both GPR41 and GPR43 are more prevalent in participants with hypertension. CONCLUSIONS Our findings highlight that lack of short-chain fatty acid-receptor signaling via both GPR41 and GPR43 increases risk of high blood pressure, suggesting treatments that target these receptors could be a novel strategy to prevent or treat hypertension.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Tenghao Zheng
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Evany Dinakis
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Liang Xie
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia (L.X., C.R.M.)
- Now with Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore (L.X.)
| | - Anastasia Barbaro-Wahl
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Hamdi A Jama
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Michael Nakai
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Madeleine Paterson
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Kwan Charmaine Leung
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Zoe McArdle
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia (Z.M., K.M.C.)
| | - Katrina Mirabito Colafella
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia (Z.M., K.M.C.)
| | - Chad Johnson
- Bioimaging Platform, La Trobe University, Melbourne, Australia (C.J.)
| | - Wendy Qin
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Ekaterina Salimova
- Monash Bioimaging Facility, Monash University, Melbourne, Australia (E.S.)
| | - Natalie J Bitto
- Department of Microbiology, Anatomy, Physiology and Pharmacology (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- La Trobe Research Centre for Extracellular Vesicles (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
| | - Maria Kaparakis-Liaskos
- Department of Microbiology, Anatomy, Physiology and Pharmacology (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- La Trobe Research Centre for Extracellular Vesicles (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- Now with Department of Microbiology & Immunology, University of Melbourne, Australia (M. K-L.)
| | - David M Kaye
- Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia (D.M.K.)
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (D.M.K., F.Z.M.)
- Department of Cardiology, Alfred Hospital, Melbourne, Australia (D.M.K.)
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Charles R Mackay
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia (L.X., C.R.M.)
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China (C.R.M.)
| | - Francine Z Marques
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (D.M.K., F.Z.M.)
| |
Collapse
|
16
|
Chen S, Huan P, Ma T, Zhong Y, Ning D, Zhuang Y. Walnut peptide relieves hypertension and associated kidney and heart injury by regulating the renin-angiotensin-aldosterone system and intestinal microbiota. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:1170-1184. [PMID: 39291451 DOI: 10.1002/jsfa.13907] [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: 06/22/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Hypertension is a chronic disease with high morbidity and mortality. Previously, we screened a walnut meal peptide FDWLR (PEP) with significant angiotensin-converting enzyme inhibitory activity. The present study further investigated the anti-hypertensive effects of PEP in vivo using spontaneously hypertensive rats. RESULTS The results indicated that PEP reduced blood pressure and the indices in the renin-angiotensin-aldosterone system (RAAS) including angiotensin-converting enzyme (ACE) (decreased by 15.36%), angiotensin II (Ang II) (decreased by 31.56%), angiotensinogen (AGT) (decreased by 58.84%) and aldosterone (ALD) (decreased by 18.27%), whereas NO levels increased by 54.96%. The pathological analysis showed that PEP relieved cardiac and renal damage. PEP also alleviated oxidative stress, inflammation and fibrosis in the heart and kidney. Mechanistically, PEP mitigated cardiac and renal damage by simultaneously regulating ACE-Ang II-AT1R and the ACE2-Ang (1-7)-MAS axis. Additionally, PEP increased the levels of short chain fatty acids by 224.16% and improved gut microbiota by increasing the abundance of Prevotella, Phascolarctobacterium, Clostridium_sensu_stricto and Bifidobacterium, at the same time as decreasing Bacteroides and Alistipes abundances. CONCLUSION This study indicated that PEP prevented hypertension and associated heart and kidney damage by modulating the RAAS system and gut microbiota, which is valuable in guiding future development and optimal utilization of walnut meal. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shupeng Chen
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Pengtao Huan
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Ting Ma
- Yunnan Academy of Forestry and Grassland, Kunming, China
- Yunnan Technology Innovation Center of Woody Oil, Kunming, China
| | - Yujie Zhong
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
| | - Delu Ning
- Yunnan Academy of Forestry and Grassland, Kunming, China
- Yunnan Technology Innovation Center of Woody Oil, Kunming, China
| | - Yongliang Zhuang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, China
- Yunnan Technology Innovation Center of Woody Oil, Kunming, China
| |
Collapse
|
17
|
Sharma A, Kapur S, Kancharla P, Yang T. Sex differences in gut microbiota, hypertension, and cardiovascular risk. Eur J Pharmacol 2025; 987:177183. [PMID: 39647571 PMCID: PMC11714433 DOI: 10.1016/j.ejphar.2024.177183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
The intricate ecosystem of the gut microbiome exhibits sex-specific differences, influencing the susceptibility to cardiovascular diseases (CVD). Imbalance within the gut microbiome compromises the gut barrier, activates inflammatory pathways, and alters the production of metabolites, all of which initiate chronic diseases including CVD. In particular, the interplay between lifestyle choices, hormonal changes, and metabolic byproducts uniquely affects sex-specific gut microbiomes, potentially shaping the risk profiles for hypertension and CVD differently in men and women. Understanding the gut microbiome's role in CVD risk offers informative reasoning behind the importance of developing tailored preventative strategies based on sex-specific differences in CVD risk. Furthermore, insight into the differential impact of social determinants and biological factors on CVD susceptibility emphasizes the necessity for more nuanced approaches. This review also outlines specific dietary interventions that may enhance gut microbiome health, offering a glimpse into potential therapeutic avenues for reducing CVD risk that require greater awareness. Imbalance in natural gut microbiomes may explain etiologies of chronic diseases; we advocate for future application to alter the gut microbiome as possible treatment of the aforementioned diseases. This review mentions the idea of altering the gut microbiome through interventions such as fecal microbiota transplantation (FMT), a major application of microbiome-based therapy that is first-line for Clostridium difficile infections and patient-specific probiotics highlights more innovative approaches to hypertension and CVD prevention. Through increased analysis of gut microbiota compositions along with patient-centric probiotics and microbiome transfers, this review advocates for future preventative strategies for hypertension.
Collapse
Affiliation(s)
- Anish Sharma
- Center for Hypertension and Precision Medicine, Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH, USA
| | - Sahil Kapur
- Center for Hypertension and Precision Medicine, Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH, USA
| | - Priyal Kancharla
- Center for Hypertension and Precision Medicine, Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH, USA
| | - Tao Yang
- Center for Hypertension and Precision Medicine, Microbiome Consortium, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH, USA.
| |
Collapse
|
18
|
Kamath S, Sokolenko E, Collins K, Chan NSL, Mills N, Clark SR, Marques FZ, Joyce P. IUPHAR themed review: The gut microbiome in schizophrenia. Pharmacol Res 2025; 211:107561. [PMID: 39732352 DOI: 10.1016/j.phrs.2024.107561] [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: 11/25/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Gut microbial dysbiosis or altered gut microbial consortium, in schizophrenia suggests a pathogenic role through the gut-brain axis, influencing neuroinflammatory and neurotransmitter pathways critical to psychotic, affective, and cognitive symptoms. Paradoxically, conventional psychotropic interventions may exacerbate this dysbiosis, with antipsychotics, particularly olanzapine, demonstrating profound effects on microbial architecture through disruption of bacterial phyla ratios, diminished taxonomic diversity, and attenuated short-chain fatty acid synthesis. To address these challenges, novel therapeutic strategies targeting the gut microbiome, encompassing probiotic supplementation, prebiotic compounds, faecal microbiota transplantation, and rationalised co-pharmacotherapy, show promise in attenuating antipsychotic-induced metabolic disruptions while enhancing therapeutic efficacy. Harnessing such insights, precision medicine approaches promise to transform antipsychotic prescribing practices by identifying patients at risk of metabolic side effects based on their microbial profiles. This IUPHAR review collates the current literature landscape of the gut-brain axis and its intricate relationship with schizophrenia while advocating for integrating microbiome assessments and therapeutic management. Such a fundamental shift in proposing microbiome-informed psychotropic prescriptions to optimise therapeutic efficacy and reduce adverse metabolic impacts would align antipsychotic treatments with microbiome safety, prioritising 'gut-neutral' or gut-favourable drugs to safeguard long-term patient outcomes in schizophrenia therapy.
Collapse
Affiliation(s)
- Srinivas Kamath
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Elysia Sokolenko
- Discipline of Anatomy and Pathology, School of Biomedicine, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kate Collins
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Nicole S L Chan
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Natalie Mills
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Francine Z Marques
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Hypertension Research Laboratory, School of Biological Sciences and Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | - Paul Joyce
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia.
| |
Collapse
|
19
|
Muralitharan RR, Buikema JW, Marques FZ. Minimizing gut microbiome confounding factors in cardiovascular research. Cardiovasc Res 2024; 120:e60-e62. [PMID: 39495203 DOI: 10.1093/cvr/cvae228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/23/2024] [Indexed: 11/05/2024] Open
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, 25 Rainforest Walk, Melbourne, Clayton, 3800 VIC, Australia
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, 3168 VIC, Australia
| | - Jan W Buikema
- Amsterdam Heart Center, Department of Cardiology, Amsterdam Cardiovascular Sciences, Department of Physiology, Amsterdam University Medical Center, VU University, Amsterdam, The Netherlands
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, 25 Rainforest Walk, Melbourne, Clayton, 3800 VIC, Australia
- Victorian Heart Institute, Monash University, 631 Blackburn Rd, Clayton, 3168 VIC, Australia
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, 3004 VIC, Australia
| |
Collapse
|
20
|
Ng YH, Koay YC, Marques FZ, Kaye DM, O’Sullivan JF. Leveraging metabolism for better outcomes in heart failure. Cardiovasc Res 2024; 120:1835-1850. [PMID: 39351766 PMCID: PMC11630082 DOI: 10.1093/cvr/cvae216] [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: 03/09/2024] [Revised: 06/26/2024] [Accepted: 08/07/2024] [Indexed: 12/11/2024] Open
Abstract
Whilst metabolic inflexibility and substrate constraint have been observed in heart failure for many years, their exact causal role remains controversial. In parallel, many of our fundamental assumptions about cardiac fuel use are now being challenged like never before. For example, the emergence of sodium-glucose cotransporter 2 inhibitor therapy as one of the four 'pillars' of heart failure therapy is causing a revisit of metabolism as a key mechanism and therapeutic target in heart failure. Improvements in the field of cardiac metabolomics will lead to a far more granular understanding of the mechanisms underpinning normal and abnormal human cardiac fuel use, an appreciation of drug action, and novel therapeutic strategies. Technological advances and expanding biorepositories offer exciting opportunities to elucidate the novel aspects of these metabolic mechanisms. Methodologic advances include comprehensive and accurate substrate quantitation such as metabolomics and stable-isotope fluxomics, improved access to arterio-venous blood samples across the heart to determine fuel consumption and energy conversion, high quality cardiac tissue biopsies, biochemical analytics, and informatics. Pairing these technologies with recent discoveries in epigenetic regulation, mitochondrial dynamics, and organ-microbiome metabolic crosstalk will garner critical mechanistic insights in heart failure. In this state-of-the-art review, we focus on new metabolic insights, with an eye on emerging metabolic strategies for heart failure. Our synthesis of the field will be valuable for a diverse audience with an interest in cardiac metabolism.
Collapse
Affiliation(s)
- Yann Huey Ng
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
| | - Yen Chin Koay
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, VIC 3800, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3800, Australia
- Victorian Heart Institute, Monash University, Melbourne, VIC 3800, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3800, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Monash-Alfred-Baker Centre for Cardiovascular Research, Monash University, Melbourne, VIC 3800, Australia
| | - John F O’Sullivan
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Department of Medicine, Technische Univeristat Dresden, 01062 Dresden, Germany
| |
Collapse
|
21
|
Deehan EC, Al Antwan S, Witwer RS, Guerra P, John T, Monheit L. Revisiting the Concepts of Prebiotic and Prebiotic Effect in Light of Scientific and Regulatory Progress-A Consensus Paper From the Global Prebiotic Association. Adv Nutr 2024; 15:100329. [PMID: 39481540 PMCID: PMC11616045 DOI: 10.1016/j.advnut.2024.100329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
The term prebiotic has been used for almost 3 decades and has undergone numerous updates over the years. The scientific literature reveals that despite continuous efforts to establish a globally unified definition to guide jurisdictional regulations and product innovations, ambiguity continues to surround the terms prebiotic and prebiotic effect, leading to products that lack in full regulatory adherence being marketed worldwide. Thus, to reflect the current state of scientific research and knowledge and for the continuous advancement of the category, an update to the current prebiotic definition is warranted. This update includes removing the term selectivity, considering additional locations of action besides the gut, highlighting prebiotic performance benefits such as cognitive and athletic, and providing a clear standalone definition for prebiotic effect. The Global Prebiotic Association (GPA) is a leading information and industry hub committed to raising awareness about prebiotics, their emerging and well-established health benefits, and prebiotic product integrity and efficacy. In this position paper, GPA builds on previous prebiotic definitions to propose the following expanded definition for prebiotic: "a compound or ingredient that is utilized by the microbiota producing a health or performance benefit." In addition to prebiotic, GPA also defines prebiotic effect as "a health or performance benefit that arises from alteration of the composition and/or activity of the microbiota, as a direct or indirect result of the utilization of a specific and well-defined compound or ingredient by microorganisms." With these 2 definitions, GPA aims to paint a clearer picture for the term prebiotic, and by incorporating an industry point of view, these updated definitions may be used alongside current scientific and regulatory perspectives to move the category forward.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States; Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, United States; Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States.
| | | | - Rhonda S Witwer
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; ADM, Decatur, IL, United States
| | - Paula Guerra
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; SGS Nutrasource, Guelph, Ontario, Canada.
| | - Tania John
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; SGS Nutrasource, Guelph, Ontario, Canada
| | - Len Monheit
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; Global Prebiotic Association/Industry Transparency Center, Chicago, IL, United States
| |
Collapse
|
22
|
R Muralitharan R, Marques FZ, O'Donnell JA. Recent advancements in targeting the immune system to treat hypertension. Eur J Pharmacol 2024; 983:177008. [PMID: 39304109 DOI: 10.1016/j.ejphar.2024.177008] [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: 05/15/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Hypertension is the key leading risk factor for death globally, affecting ∼1.3 billion adults, particularly in low- and middle-income countries. Most people living with hypertension have uncontrolled high blood pressure, increasing their likelihood of cardiovascular events. Significant issues preventing blood pressure control include lack of diagnosis, treatment, and response to existing therapy. For example, monotherapy and combination therapy are often unable to lower blood pressure to target levels. New therapies are urgently required to tackle this issue, particularly those that target the mechanisms behind hypertension instead of treating its symptoms. Acting via an increase in systemic and tissue-specific inflammation, the immune system is a critical contributor to blood pressure regulation and is considered an early mechanism leading to hypertension development. Here, we review the immune system's role in hypertension, evaluate clinical trials that target inflammation, and discuss knowledge gaps in pre-clinical and clinical data. We examine the effects of anti-inflammatory drugs colchicine and methotrexate on hypertension and evaluate the blockade of pro-inflammatory cytokines IL-1β and TNF-α on blood pressure in clinical trials. Lastly, we highlight how we can move forward to target specific components of the immune system to lower blood pressure. This includes targeting isolevuglandins, which accumulate in dendritic cells to promote T cell activation and cytokine production in salt-induced hypertension. We discuss the potential of the dietary fibre-derived metabolites short-chain fatty acids, which have anti-inflammatory and blood pressure-lowering effects via the gut microbiome. This would limit adverse events, leading to improved medication adherence and better blood pressure control.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
23
|
Wimmer MI, Bartolomaeus H, Anandakumar H, Chen CY, Vecera V, Kedziora S, Kamboj S, Schumacher F, Pals S, Rauch A, Meisel J, Potapenko O, Yarritu A, Bartolomaeus TUP, Samaan M, Thiele A, Stürzbecher L, Geisberger SY, Kleuser B, Oefner PJ, Haase N, Löber U, Gronwald W, Forslund-Startceva SK, Müller DN, Wilck N. Metformin modulates microbiota and improves blood pressure and cardiac remodeling in a rat model of hypertension. Acta Physiol (Oxf) 2024; 240:e14226. [PMID: 39253815 DOI: 10.1111/apha.14226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/29/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024]
Abstract
AIMS Metformin has been attributed to cardiovascular protection even in the absence of diabetes. Recent observations suggest that metformin influences the gut microbiome. We aimed to investigate the influence of metformin on the gut microbiota and hypertensive target organ damage in hypertensive rats. METHODS Male double transgenic rats overexpressing the human renin and angiotensinogen genes (dTGR), a model of angiotensin II-dependent hypertension, were treated with metformin (300 mg/kg/day) or vehicle from 4 to 7 weeks of age. We assessed gut microbiome composition and function using shotgun metagenomic sequencing and measured blood pressure via radiotelemetry. Cardiac and renal organ damage and inflammation were evaluated by echocardiography, histology, and flow cytometry. RESULTS Metformin treatment increased the production of short-chain fatty acids (SCFA) acetate and propionate in feces without altering microbial composition and diversity. It significantly reduced systolic and diastolic blood pressure and improved cardiac function, as measured by end-diastolic volume, E/A, and stroke volume despite increased cardiac hypertrophy. Metformin reduced cardiac inflammation by lowering macrophage infiltration and shifting macrophage subpopulations towards a less inflammatory phenotype. The observed improvements in blood pressure, cardiac function, and inflammation correlated with fecal SCFA levels in dTGR. In vitro, acetate and propionate altered M1-like gene expression in macrophages, reinforcing anti-inflammatory effects. Metformin did not affect hypertensive renal damage or microvascular structure. CONCLUSION Metformin modulated the gut microbiome, increased SCFA production, and ameliorated blood pressure and cardiac remodeling in dTGR. Our findings confirm the protective effects of metformin in the absence of diabetes, highlighting SCFA as a potential mediators.
Collapse
Affiliation(s)
- Moritz I Wimmer
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Hendrik Bartolomaeus
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Harithaa Anandakumar
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Chia-Yu Chen
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Valentin Vecera
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Sarah Kedziora
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sakshi Kamboj
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | | | - Sidney Pals
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Ariana Rauch
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Jutta Meisel
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olena Potapenko
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Alex Yarritu
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Theda U P Bartolomaeus
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mariam Samaan
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Arne Thiele
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Lucas Stürzbecher
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabrina Y Geisberger
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Löber
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wolfram Gronwald
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nicola Wilck
- Department of Nephrology and Medical Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbruck Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
24
|
Camargo Tavares L, Marques FZ. Clinical Trial Evidence of the Gut Microbial Metabolite Butyrate in Hypertension. Hypertension 2024; 81:2137-2139. [PMID: 39292798 DOI: 10.1161/hypertensionaha.124.23736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Affiliation(s)
- Leticia Camargo Tavares
- Hypertension Research Laboratory, Faculty of Science, School of Biological Sciences (L.C.T., F.Z.M.), Monash University, Melbourne, VIC, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, Faculty of Science, School of Biological Sciences (L.C.T., F.Z.M.), Monash University, Melbourne, VIC, Australia
- Victorian Heart Institute (F.Z.M.), Monash University, Melbourne, VIC, Australia
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (F.Z.M.)
| |
Collapse
|
25
|
Zheng T, Camargo Tavares L, D'Amato M, Marques FZ. Constipation is associated with an increased risk of major adverse cardiac events in a UK population. Am J Physiol Heart Circ Physiol 2024; 327:H956-H964. [PMID: 39150392 DOI: 10.1152/ajpheart.00519.2024] [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: 07/30/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
Traditional cardiovascular risk factors, including hypertension, only explain part of major adverse cardiac events (MACEs). Understanding what other risk factors contribute to MACE is essential for prevention. Constipation shares common risk factors with hypertension and is associated with an increased risk of several cardiovascular diseases. We hypothesized that constipation is an underappreciated risk factor for MACE. We used the population healthcare and genomic data in the UK Biobank (n = 408,354) to study the contribution of constipation (ICD10 K59.0) to the risk of MACE, defined by any episode of acute coronary syndrome (ACS), ischemic stroke, and heart failure (HF). Analyses were controlled for traditional cardiovascular risk factors. We also assessed genetic correlations (rg) between constipation and MACE. Constipation cases (n = 23,814) exhibited a significantly higher risk of MACE compared with those with normal bowel habits [odds ratio (OR) = 2.15, P < 1.00 × 10-300]. Constipation was also significantly associated with individual MACE subgroups, in order: HF (OR = 2.72, P < 1.00 × 10-300), ischemic stroke (OR = 2.36, P = 2.02 × 10-230), and ACS (OR = 1.62, P = 5.82 × 10-113). In comparison with patients with constipation-free hypertension, patients with hypertension with constipation showed significantly higher odds of MACE (OR = 1.68, P = 1.05 × 10-136) and a 34% increased risk of MACE occurrence (P = 2.3 × 10-50) after adjustment for medications that affect gut motility and other traditional cardiovascular risk factors. Finally, we detected positive genetic correlations between constipation and MACE subgroups ACS (rg = 0.27, P = 2.12 × 10-6), ischemic stroke (rg = 0.23, P = 0.011), and HF (rg = 0.21, P = 0.0062). We identified constipation as a potential risk factor independently associated with higher MACE prevalence. These findings warrant further studies on their causal relationship and identification of pathophysiological mechanisms.NEW & NOTEWORTHY Analyzing 408,354 participants of the UK Biobank, we show that constipation cases exhibited a significantly higher risk of major adverse cardiac events (MACEs) than those with regular bowel habits. In comparison with patients with constipation-free hypertension, patients with hypertension with constipation showed significantly higher odds of MACE and a 34% increased risk of subsequent MACE occurrence. Finally, we detected positive genetic correlations between constipation and MACE. This association holds potential for therapeutic exploitation and prevention based on individuals' risk assessment.
Collapse
Affiliation(s)
- Tenghao Zheng
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
| | - Leticia Camargo Tavares
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
| | - Mauro D'Amato
- Gastrointestinal Genetics Laboratory, CIC bioGUNE-BRTA, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Department of Medicine and Surgery, LUM University, Bari, Italy
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
R Muralitharan R, Nakai ME, Snelson M, Zheng T, Dinakis E, Xie L, Jama H, Paterson M, Shihata W, Wassef F, Vinh A, Drummond GR, Kaye DM, Mackay CR, Marques FZ. Influence of angiotensin II on the gut microbiome: modest effects in comparison to experimental factors. Cardiovasc Res 2024; 120:1155-1163. [PMID: 38518247 PMCID: PMC11368123 DOI: 10.1093/cvr/cvae062] [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: 10/18/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 03/24/2024] Open
Abstract
AIMS Animal models are regularly used to test the role of the gut microbiome in hypertension. Small-scale pre-clinical studies have investigated changes to the gut microbiome in the angiotensin II hypertensive model. However, the gut microbiome is influenced by internal and external experimental factors, which are not regularly considered in the study design. Once these factors are accounted for, it is unclear if microbiome signatures are reproduceable. We aimed to determine the influence of angiotensin II treatment on the gut microbiome using a large and diverse cohort of mice and to quantify the magnitude by which other factors contribute to microbiome variations. METHODS AND RESULTS We conducted a retrospective study to establish a diverse mouse cohort resembling large human studies. We sequenced the V4 region of the 16S rRNA gene from 538 samples across the gastrointestinal tract of 303 male and female C57BL/6J mice randomized into sham or angiotensin II treatment from different genotypes, diets, animal facilities, and age groups. Analysing over 17 million sequencing reads, we observed that angiotensin II treatment influenced α-diversity (P = 0.0137) and β-diversity (i.e. composition of the microbiome, P < 0.001). Bacterial abundance analysis revealed patterns consistent with a reduction in short-chain fatty acid producers, microbial metabolites that lower blood pressure. Furthermore, animal facility, genotype, diet, age, sex, intestinal sampling site, and sequencing batch had significant effects on both α- and β-diversity (all P < 0.001). Sampling site (6.8%) and diet (6%) had the largest impact on the microbiome, while angiotensin II and sex had the smallest effect (each 0.4%). CONCLUSION Our large-scale data confirmed findings from small-scale studies that angiotensin II impacted the gut microbiome. However, this effect was modest relative to most of the other factors studied. Accounting for these factors in future pre-clinical hypertensive studies will increase the likelihood that microbiome findings are replicable and translatable.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
- Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Clayton, 3800 Melbourne, Australia
| | - Michael E Nakai
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
| | - Matthew Snelson
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Clayton, 3800 Melbourne, Australia
| | - Tenghao Zheng
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
| | - Evany Dinakis
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
| | - Liang Xie
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
| | - Hamdi Jama
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
| | - Madeleine Paterson
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
| | - Waled Shihata
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Road, 3004 Melbourne, Australia
| | - Flavia Wassef
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), Bundoora, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), Bundoora, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), Bundoora, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Road, 3004 Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Charles R Mackay
- Infection and Immunity Program, Monash Biodiscovery Institute, Monash University, Melbourne, Australia
- Department of Biochemistry, Monash University, Melbourne, Australia
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, 18 Innovation Walk, Clayton, 3800 Melbourne, Australia
- Victorian Heart Institute, Monash University, 631 Blackburn Road, Clayton, 3800 Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Road, 3004 Melbourne, Australia
| |
Collapse
|
27
|
Jama HA, Dona MSI, Dinakis E, Nakai M, Paterson MR, Shihata WA, Krstevski C, Cohen CD, Weeks KL, Farrugia GE, Johnson C, Salimova E, Donner DG, Kiriazis H, Kaipananickal H, Okabe J, Anderson D, Creek DJ, Mackay CR, El-Osta A, Pinto AR, Kaye DM, Marques FZ. Maternal Diet and Gut Microbiota Influence Predisposition to Cardiovascular Disease in Offspring. Circ Res 2024; 135:537-539. [PMID: 39016011 DOI: 10.1161/circresaha.124.324614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Affiliation(s)
- Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., E.D., M.N., M.R.P., F.Z.M.), Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group (H.A.J., W.A.S., D.M.K., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Malathi S I Dona
- Cardiac Cellular Systems (M.S.I.D., C.K., C.D.C., G.E.F., A.R.P.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Evany Dinakis
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., E.D., M.N., M.R.P., F.Z.M.), Monash University, Melbourne, Victoria, Australia
| | - Michael Nakai
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., E.D., M.N., M.R.P., F.Z.M.), Monash University, Melbourne, Victoria, Australia
| | - Madeleine R Paterson
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., E.D., M.N., M.R.P., F.Z.M.), Monash University, Melbourne, Victoria, Australia
| | - Waled A Shihata
- Heart Failure Research Group (H.A.J., W.A.S., D.M.K., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Crisdion Krstevski
- Cardiac Cellular Systems (M.S.I.D., C.K., C.D.C., G.E.F., A.R.P.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research (C.K., C.D.C., A.R.P.), La Trobe University, Melbourne, Victoria, Australia
| | - Charles D Cohen
- Cardiac Cellular Systems (M.S.I.D., C.K., C.D.C., G.E.F., A.R.P.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research (C.K., C.D.C., A.R.P.), La Trobe University, Melbourne, Victoria, Australia
| | - Kate L Weeks
- Department of Anatomy and Physiology (K.L.W.), The University of Melbourne, Parkville, Victoria, Australia
- Baker Department of Cardiometabolic Health (K.L.W., D.G.D., H. Kiriazis, H. Kaipananickal, J.O., A.E.-O.), The University of Melbourne, Parkville, Victoria, Australia
| | - Gabriella E Farrugia
- Cardiac Cellular Systems (M.S.I.D., C.K., C.D.C., G.E.F., A.R.P.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Chad Johnson
- Monash Micro Imaging (C.J.), Monash University, Melbourne, Victoria, Australia
- La Trobe Institute of Molecular Science (C.J.), La Trobe University, Melbourne, Victoria, Australia
- Burnet Institute, Melbourne, Victoria, Australia (C.J.)
| | - Ekaterina Salimova
- Monash Biomedical Imaging (E.S.), Monash University, Melbourne, Victoria, Australia
| | - Daniel G Donner
- School of Translational Medicine, Faculty of Medicine Nursing and Health Sciences (D.G.D., D.M.K.), Monash University, Melbourne, Victoria, Australia
- Preclinical Cardiology, Microsurgery and Imaging Platform (D.D., H. Kiriazis), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health (K.L.W., D.G.D., H. Kiriazis, H. Kaipananickal, J.O., A.E.-O.), The University of Melbourne, Parkville, Victoria, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (D.G.D., D.M.K.)
| | - Helen Kiriazis
- Preclinical Cardiology, Microsurgery and Imaging Platform (D.D., H. Kiriazis), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health (K.L.W., D.G.D., H. Kiriazis, H. Kaipananickal, J.O., A.E.-O.), The University of Melbourne, Parkville, Victoria, Australia
| | - Harikrishnan Kaipananickal
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Alfred Centre (H. Kaipananickal, J.O., A.E.-O.), Monash University, Melbourne, Victoria, Australia
- Epigenetics in Human Health and Disease (H. Kaipananickal, J.O., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health (K.L.W., D.G.D., H. Kiriazis, H. Kaipananickal, J.O., A.E.-O.), The University of Melbourne, Parkville, Victoria, Australia
| | - Jun Okabe
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Alfred Centre (H. Kaipananickal, J.O., A.E.-O.), Monash University, Melbourne, Victoria, Australia
- Epigenetics in Human Health and Disease (H. Kaipananickal, J.O., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health (K.L.W., D.G.D., H. Kiriazis, H. Kaipananickal, J.O., A.E.-O.), The University of Melbourne, Parkville, Victoria, Australia
| | - Dovile Anderson
- Monash Institute of Pharmaceutical Sciences (D.A., D.J.C.), Monash University, Melbourne, Victoria, Australia
| | - Darren J Creek
- Monash Institute of Pharmaceutical Sciences (D.A., D.J.C.), Monash University, Melbourne, Victoria, Australia
| | - Charles R Mackay
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China (C.R.M.)
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Alfred Centre (H. Kaipananickal, J.O., A.E.-O.), Monash University, Melbourne, Victoria, Australia
- Epigenetics in Human Health and Disease (H. Kaipananickal, J.O., A.E.-O.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health (K.L.W., D.G.D., H. Kiriazis, H. Kaipananickal, J.O., A.E.-O.), The University of Melbourne, Parkville, Victoria, Australia
| | - Alexander R Pinto
- Cardiac Cellular Systems (M.S.I.D., C.K., C.D.C., G.E.F., A.R.P.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research (C.K., C.D.C., A.R.P.), La Trobe University, Melbourne, Victoria, Australia
| | - David M Kaye
- School of Translational Medicine, Faculty of Medicine Nursing and Health Sciences (D.G.D., D.M.K.), Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group (H.A.J., W.A.S., D.M.K., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Victoria, Australia (D.G.D., D.M.K.)
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., E.D., M.N., M.R.P., F.Z.M.), Monash University, Melbourne, Victoria, Australia
- Victorian Heart Hospital (F.Z.M.), Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group (H.A.J., W.A.S., D.M.K., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| |
Collapse
|
28
|
Dinakis E, O'Donnell JA, Marques FZ. The gut-immune axis during hypertension and cardiovascular diseases. Acta Physiol (Oxf) 2024; 240:e14193. [PMID: 38899764 DOI: 10.1111/apha.14193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/04/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
The gut-immune axis is a relatively novel phenomenon that provides mechanistic links between the gut microbiome and the immune system. A growing body of evidence supports it is key in how the gut microbiome contributes to several diseases, including hypertension and cardiovascular diseases (CVDs). Evidence over the past decade supports a causal link of the gut microbiome in hypertension and its complications, including myocardial infarction, atherosclerosis, heart failure, and stroke. Perturbations in gut homeostasis such as dysbiosis (i.e., alterations in gut microbial composition) may trigger immune responses that lead to chronic low-grade inflammation and, ultimately, the development and progression of these conditions. This is unsurprising, as the gut harbors one of the largest numbers of immune cells in the body, yet is a phenomenon not entirely understood in the context of cardiometabolic disorders. In this review, we discuss the role of the gut microbiome, the immune system, and inflammation in the context of hypertension and CVD, and consolidate current evidence of this complex interplay, whilst highlighting gaps in the literature. We focus on diet as one of the major modulators of the gut microbiota, and explain key microbial-derived metabolites (e.g., short-chain fatty acids, trimethylamine N-oxide) as potential mediators of the communication between the gut and peripheral organs such as the heart, arteries, kidneys, and the brain via the immune system. Finally, we explore the dual role of both the gut microbiome and the immune system, and how they work together to not only contribute, but also mitigate hypertension and CVD.
Collapse
Affiliation(s)
- Evany Dinakis
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Jama HA, Snelson M, Schutte AE, Muir J, Marques FZ. Recommendations for the Use of Dietary Fiber to Improve Blood Pressure Control. Hypertension 2024; 81:1450-1459. [PMID: 38586958 DOI: 10.1161/hypertensionaha.123.22575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
According to several international, regional, and national guidelines on hypertension, lifestyle interventions are the first-line treatment to lower blood pressure (BP). Although diet is one of the major lifestyle modifications described in hypertension guidelines, dietary fiber is not specified. Suboptimal intake of foods high in fiber, such as in Westernized diets, is a major contributing factor to mortality and morbidity of noncommunicable diseases due to higher BP and cardiovascular disease. In this review, we address this deficiency by examining and advocating for the incorporation of dietary fiber as a key lifestyle modification to manage elevated BP. We explain what dietary fiber is, review the existing literature that supports its use to lower BP and prevent cardiovascular disease, describe the mechanisms involved, propose evidence-based target levels of fiber intake, provide examples of how patients can achieve the recommended targets, and discuss outstanding questions in the field. According to the evidence reviewed here, the minimum daily dietary fiber for adults with hypertension should be >28 g/day for women and >38 g/day for men, with each extra 5 g/day estimated to reduce systolic BP by 2.8 mm Hg and diastolic BP by 2.1 mm Hg. This would support a healthy gut microbiota and the production of gut microbiota-derived metabolites called short-chain fatty acids that lower BP. Awareness about dietary fiber targets and how to achieve them will guide medical teams on better educating patients and empowering them to increase their fiber intake and, as a result, lower their BP and cardiovascular disease risk.
Collapse
Affiliation(s)
- Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
| | - Matthew Snelson
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
- Victorian Heart Institute (M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
| | - Aletta E Schutte
- School of Population Health, University of New South Wales, Sydney, Australia (A.E.S.)
- George Institute for Global Health, Sydney, NSW, Australia (A.E.S.)
- Hypertension in Africa Research Team, MRC Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa (A.E.S.)
| | - Jane Muir
- Department of Gastroenterology, School of Translational Medicine (J.M.), Monash University, Melbourne, VIC, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences (H.A.J., M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
- Victorian Heart Institute (M.S., F.Z.M.), Monash University, Melbourne, VIC, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (F.Z.M.)
| |
Collapse
|
30
|
Chrysant SG. The role of gut microbiota in the development of salt-sensitive hypertension and the possible preventive effect of exercise. Expert Rev Cardiovasc Ther 2024; 22:265-271. [PMID: 38823009 DOI: 10.1080/14779072.2024.2364031] [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: 10/31/2023] [Accepted: 05/31/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION The aim of the present study is to analyze the data indicating an association between high salt intake and the gastrointestinal microbiota in the development of salt-sensitive hypertension in animals and men. It is also, to discuss the preventive effects of exercise on gut-induced hypertension by favorably modifying the composition of gut microbiota. AREAS COVERED Salt sensitivity is quite common, accounting for 30%-60% in hypertensive subjects. Recently, a novel cause for salt-sensitive hypertension has been discovered through the action of gut microbiota by the secretion of several hormones and the action of short chain fatty acids (SCFAs). In addition, recent studies indicate that exercise might favorably modify the adverse effects of gut microbiota regarding their effects on BP. To identify the role of gut microbiota on the incidence of hypertension and CVD and the beneficial effect of exercise, a Medline search of the English literature was conducted between 2018 and 2023 and 42 pertinent papers were selected. EXPERT OPINION The analysis of data from the selected papers disclosed that the gut microbiota contribute significantly to the development of salt-sensitive hypertension and that exercise modifies their gut composition and ameliorates their adverse effects on BP.
Collapse
Affiliation(s)
- Steven G Chrysant
- Cardiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
31
|
Guzik TJ, Nosalski R, Maffia P, Drummond GR. Immune and inflammatory mechanisms in hypertension. Nat Rev Cardiol 2024; 21:396-416. [PMID: 38172242 DOI: 10.1038/s41569-023-00964-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Hypertension is a global health problem, with >1.3 billion individuals with high blood pressure worldwide. In this Review, we present an inflammatory paradigm for hypertension, emphasizing the crucial roles of immune cells, cytokines and chemokines in disease initiation and progression. T cells, monocytes, macrophages, dendritic cells, B cells and natural killer cells are all implicated in hypertension. Neoantigens, the NLRP3 inflammasome and increased sympathetic outflow, as well as cytokines (including IL-6, IL-7, IL-15, IL-18 and IL-21) and a high-salt environment, can contribute to immune activation in hypertension. The activated immune cells migrate to target organs such as arteries (especially the perivascular fat and adventitia), kidneys, the heart and the brain, where they release effector cytokines that elevate blood pressure and cause vascular remodelling, renal damage, cardiac hypertrophy, cognitive impairment and dementia. IL-17 secreted by CD4+ T helper 17 cells and γδ T cells, and interferon-γ and tumour necrosis factor secreted by immunosenescent CD8+ T cells, exert crucial effector roles in hypertension, whereas IL-10 and regulatory T cells are protective. Effector mediators impair nitric oxide bioavailability, leading to endothelial dysfunction and increased vascular contractility. Inflammatory effector mediators also alter renal sodium and water balance and promote renal fibrosis. These mechanisms link hypertension with obesity, autoimmunity, periodontitis and COVID-19. A comprehensive understanding of the immune and inflammatory mechanisms of hypertension is crucial for safely and effectively translating the findings to clinical practice.
Collapse
Affiliation(s)
- Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK.
- Department of Medicine and Omicron Medical Genomics Laboratory, Jagiellonian University, Collegium Medicum, Kraków, Poland.
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK.
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Pasquale Maffia
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grant R Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Seefeldt JM, Homilius C, Hansen J, Lassen TR, Jespersen NR, Jensen RV, Boedtkjer E, Bøtker HE, Nielsen R. Short-Chain Fatty Acid Butyrate Is an Inotropic Agent With Vasorelaxant and Cardioprotective Properties. J Am Heart Assoc 2024; 13:e033744. [PMID: 38686853 PMCID: PMC11179878 DOI: 10.1161/jaha.123.033744] [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: 11/29/2023] [Accepted: 03/21/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND The heart can metabolize the microbiota-derived short-chain fatty acid butyrate. Butyrate may have beneficial effects in heart failure, but the underlying mechanisms are unknown. We tested the hypothesis that butyrate elevates cardiac output by mechanisms involving direct stimulation of cardiac contractility and vasorelaxation in rats. METHODS AND RESULTS We examined the effects of butyrate on (1) in vivo hemodynamics using parallel echocardiographic and invasive blood pressure measurements, (2) isolated perfused hearts in Langendorff systems under physiological conditions and after ischemia and reperfusion, and (3) isolated coronary arteries mounted in isometric wire myographs. We tested Na-butyrate added to injection solutions or physiological buffers and compared its effects with equimolar doses of NaCl. Butyrate at plasma concentrations of 0.56 mM increased cardiac output by 48.8±14.9%, stroke volume by 38.5±12.1%, and left ventricular ejection fraction by 39.6±6.2%, and lowered systemic vascular resistance by 33.5±6.4% without affecting blood pressure or heart rate in vivo. In the range between 0.1 and 5 mM, butyrate increased left ventricular systolic pressure by up to 23.7±3.4% in isolated perfused hearts and by 9.4±2.9% following ischemia and reperfusion, while reducing myocardial infarct size by 81.7±16.9%. Butyrate relaxed isolated coronary septal arteries concentration dependently with an EC50=0.57 mM (95% CI, 0.23-1.44). CONCLUSIONS We conclude that butyrate elevates cardiac output through mechanisms involving increased cardiac contractility and vasorelaxation. This effect of butyrate was not associated with adverse myocardial injury in damaged hearts exposed to ischemia and reperfusion.
Collapse
Affiliation(s)
- Jacob Marthinsen Seefeldt
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
| | | | - Jakob Hansen
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Forensic Medicine Aarhus University Hospital Aarhus Denmark
| | | | | | | | - Ebbe Boedtkjer
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Hans Erik Bøtker
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
| | - Roni Nielsen
- Department of Clinical Medicine Aarhus University Aarhus Denmark
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
| |
Collapse
|
33
|
Durgan DJ, Zubcevic J, Vijay-Kumar M, Yang T, Manandhar I, Aryal S, Muralitharan RR, Li HB, Li Y, Abais-Battad JM, Pluznick JL, Muller DN, Marques FZ, Joe B. Prospects for Leveraging the Microbiota as Medicine for Hypertension. Hypertension 2024; 81:951-963. [PMID: 38630799 DOI: 10.1161/hypertensionaha.124.21721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Affiliation(s)
- David J Durgan
- Department of Integrative Physiology and Anesthesiology, Baylor College of Medicine, Houston, TX (D.J.D.)
| | - Jasenka Zubcevic
- Center for Hypertension and Precision Medicine, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Microbiome Consortium, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
| | - Matam Vijay-Kumar
- Center for Hypertension and Precision Medicine, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Microbiome Consortium, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
| | - Tao Yang
- Center for Hypertension and Precision Medicine, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Microbiome Consortium, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
| | - Ishan Manandhar
- Center for Hypertension and Precision Medicine, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Microbiome Consortium, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
| | - Sachin Aryal
- Center for Hypertension and Precision Medicine, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Microbiome Consortium, Toledo, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, OH (J.Z., M.V.-K., T.Y., I.M., S.A., B.J.)
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Australia (R.R.M., F.Z.M.)
- Victorian Heart Institute, Monash University, Melbourne, Australia (R.R.M., F.Z.M.)
- Baker Heart and Diabetes Institute, Melbourne, Australia (R.R.M., F.Z.M.)
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, PR China (H.-B.L., Y.L.)
| | - Ying Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, PR China (H.-B.L., Y.L.)
| | | | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD (J.L.P.)
| | - Dominik N Muller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (D.N.M.)
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Germany (D.N.M.)
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (D.N.M.)
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany (D.N.M.)
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Australia (R.R.M., F.Z.M.)
- Victorian Heart Institute, Monash University, Melbourne, Australia (R.R.M., F.Z.M.)
- Baker Heart and Diabetes Institute, Melbourne, Australia (R.R.M., F.Z.M.)
| | - Bina Joe
- Department of Integrative Physiology and Anesthesiology, Baylor College of Medicine, Houston, TX (D.J.D.)
| |
Collapse
|
34
|
Tan DSY, Akelew Y, Snelson M, Nguyen J, O’Sullivan KM. Unravelling the Link between the Gut Microbiome and Autoimmune Kidney Diseases: A Potential New Therapeutic Approach. Int J Mol Sci 2024; 25:4817. [PMID: 38732038 PMCID: PMC11084259 DOI: 10.3390/ijms25094817] [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: 03/22/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
The gut microbiota and short chain fatty acids (SCFA) have been associated with immune regulation and autoimmune diseases. Autoimmune kidney diseases arise from a loss of tolerance to antigens, often with unclear triggers. In this review, we explore the role of the gut microbiome and how disease, diet, and therapy can alter the gut microbiota consortium. Perturbations in the gut microbiota may systemically induce the translocation of microbiota-derived inflammatory molecules such as liposaccharide (LPS) and other toxins by penetrating the gut epithelial barrier. Once in the blood stream, these pro-inflammatory mediators activate immune cells, which release pro-inflammatory molecules, many of which are antigens in autoimmune diseases. The ratio of gut bacteria Bacteroidetes/Firmicutes is associated with worse outcomes in multiple autoimmune kidney diseases including lupus nephritis, MPO-ANCA vasculitis, and Goodpasture's syndrome. Therapies that enhance SCFA-producing bacteria in the gut have powerful therapeutic potential. Dietary fiber is fermented by gut bacteria which in turn release SCFAs that protect the gut barrier, as well as modulating immune responses towards a tolerogenic anti-inflammatory state. Herein, we describe where the current field of research is and the strategies to harness the gut microbiome as potential therapy.
Collapse
Affiliation(s)
- Diana Shu Yee Tan
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, VIC 3168, Australia; (D.S.Y.T.); (Y.A.)
| | - Yibeltal Akelew
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, VIC 3168, Australia; (D.S.Y.T.); (Y.A.)
| | - Matthew Snelson
- School of Biological Science, Monash University, Clayton, VIC 3168, Australia;
| | - Jenny Nguyen
- The Alfred Centre, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Kim Maree O’Sullivan
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, VIC 3168, Australia; (D.S.Y.T.); (Y.A.)
| |
Collapse
|
35
|
Wang Q, Mackay CR. High metabolite concentrations in portal venous blood as a possible mechanism for microbiota effects on the immune system and Western diseases. J Allergy Clin Immunol 2024; 153:980-982. [PMID: 37992816 DOI: 10.1016/j.jaci.2023.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/02/2023] [Accepted: 10/25/2023] [Indexed: 11/24/2023]
Affiliation(s)
- Quanbo Wang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in the Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Charles R Mackay
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in the Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China; Department of Microbiology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
| |
Collapse
|
36
|
Cardoso AM. Microbial influence on blood pressure: unraveling the complex relationship for health insights. MICROBIOME RESEARCH REPORTS 2024; 3:22. [PMID: 38841410 PMCID: PMC11149090 DOI: 10.20517/mrr.2023.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/29/2024] [Accepted: 03/13/2024] [Indexed: 06/07/2024]
Abstract
Hypertension, a critical global health concern, is characterized by persistent high blood pressure and is a major cause of cardiovascular events. This perspective explores the multifaceted implications of hypertension, its association with cardiovascular diseases, and the emerging role of the gut microbiota. The gut microbiota, a dynamic community in the gastrointestinal tract, plays a pivotal role in hypertension by influencing blood pressure through the generation of antioxidant, anti-inflammatory, and short-chain fatty acids metabolites, and the conversion of nitrates into nitric oxide. Antihypertensive medications interact with the gut microbiota, impacting drug pharmacokinetics and efficacy. Prebiotics and probiotics present promising avenues for hypertension management, with prebiotics modulating blood pressure through lipid and cholesterol modulation, and probiotics exhibiting a general beneficial effect. Personalized choices based on individual factors are crucial for optimizing prebiotic and probiotic interventions. In conclusion, the gut microbiota's intricate influence on blood pressure regulation offers innovative perspectives in hypertension therapeutics, with targeted strategies proving valuable for holistic blood pressure management and health promotion.
Collapse
|
37
|
Xiong Y, He Y, Chen Z, Wu T, Xiong Y, Peng Y, Yang X, Liu Y, Zhou J, Zhou H, Zhang W, Shu Y, Li X, Li Q. Lactobacillus induced by irbesartan on spontaneously hypertensive rat contribute to its antihypertensive effect. J Hypertens 2024; 42:460-470. [PMID: 38009301 DOI: 10.1097/hjh.0000000000003613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
OBJECTIVE Hypertension is linked to gut dysbiosis. Here, the impact of the angiotensin receptor antagonist irbesartan on the gut microbiota of spontaneously hypertensive rats (SHR) were investigated. In addition, we assessed their contribution to its antihypertensive effect. METHODS Eight-week-old Wistar-Kyoto (WKY) rats and SHR were administered irbesartan for 8 weeks. Fecal microbiota transplantation (FMT) was performed from SHR treated with irbesartan or untreated SHR to recipient untreated SHR. The preventive effect of Lactobacillus on hypertension in SHR was evaluated. Blood pressure (BP) was calculated using a tail-sleeve sphygmomanometer. To better assess the composition of the gut microbiota, the V3-V4 region of the 16S rRNA gene was amplified while short-chain fatty acids (SCFAs) in feces were tested by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). RESULTS Irbesartan restored gut dysbiosis, increased the abundance of Lactobacillus , and improved anti-inflammatory ability, antioxidative ability, intestinal integrity, and intestinal inflammation in SHR. The microbiota in SHR-treated irbesartan could reduce BP and improve antioxidative ability and gut integrity in SHR. Lactobacillus johnsonii ( L. johnsonii ) and Lactobacillus reuteri ( L. reuteri ) reduced BP, restored gut dysbiosis and improved anti-inflammatory ability, antioxidative ability, intestinal integrity in SHR. Most notably, irbesartan, L. johnsonii , and L. reuteri can significantly increase SCFA content in SHR feces. CONCLUSION The current study demonstrated that irbesartan treatment ameliorated gut dysbiosis in SHR. Irbesartan induced alterations in gut microbiota, with increased prevalence of Lactobacillus .
Collapse
Affiliation(s)
- Yanling Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Department of Pharmacy, First hospital of Nanchang, Nanchang
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Yanping He
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou
| | - Zhi Chen
- Department of Hypertension, Xingsha Hospital, Changsha, China
| | - Tianyuan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Yalan Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Yilei Peng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Xuechun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Jian Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland, USA
| | - Xiong Li
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education
- National Clinical Research Center for Geriatric Disorders, Changsha
| |
Collapse
|
38
|
Cheng J, Zhou J. Unraveling the gut health puzzle: exploring the mechanisms of butyrate and the potential of High-Amylose Maize Starch Butyrate (HAMSB) in alleviating colorectal disturbances. Front Nutr 2024; 11:1285169. [PMID: 38304546 PMCID: PMC10830644 DOI: 10.3389/fnut.2024.1285169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Colorectal disturbances encompass a variety of disorders that impact the colon and rectum, such as colitis and colon cancer. Butyrate, a short-chain fatty acid, plays a pivotal role in supporting gut health by nourishing colonocytes, promoting barrier function, modulating inflammation, and fostering a balanced microbiome. Increasing colorectal butyrate concentration may serve as a critical strategy to improve colon function and reduce the risk of colorectal disturbances. Butyrylated high-amylose maize starch (HAMSB) is an edible ingredient that efficiently delivers butyrate to the colon. HAMSB is developed by esterifying a high-amylose starch backbone with butyric anhydride. With a degree of substitution of 0.25, each hydroxy group of HAMSB is substituted by a butyryl group in every four D-glucopyranosyl units. In humans, the digestibility of HAMSB is 68% (w/w), and 60% butyrate molecules attached to the starch backbone is absorbed by the colon. One clinical trial yielded two publications, which showed that HAMSB significantly reduced rectal O6-methyl-guanine adducts and epithelial proliferation induced by the high protein diet. Fecal microbial profiles were assessed in three clinical trials, showing that HAMSB supplementation was consistently linked to increased abundance of Parabacteroides distasonis. In animal studies, HAMSB was effective in reducing the risk of diet- or AOM-induced colon cancer by reducing genetic damage, but the mechanisms differed. HAMSB functioned through affecting cecal ammonia levels by modulating colon pH in diet-induced cancer, while it ameliorated chemical-induced colon cancer through downregulating miR19b and miR92a expressions and subsequently activating the caspase-dependent apoptosis. Furthermore, animal studies showed that HAMSB improved colitis via regulating the gut immune modulation by inhibiting histone deacetylase and activating G protein-coupled receptors, but its role in bacteria-induced colon colitis requires further investigation. In conclusion, HAMSB is a food ingredient that may deliver butyrate to the colon to support colon health. Further clinical trials are warranted to validate earlier findings and determine the minimum effective dose of HAMSB.
Collapse
Affiliation(s)
- Junrui Cheng
- Global Scientific and Regulatory Department, Ingredion Incorporated, Bridgewater, NJ, United States
| | - Jing Zhou
- Global Scientific and Regulatory Department, Ingredion Incorporated, Bridgewater, NJ, United States
| |
Collapse
|
39
|
Caparrós-Martín JA, Maher P, Ward NC, Saladié M, Agudelo-Romero P, Stick SM, Chan DC, Watts GF, O’Gara F. An Analysis of the Gut Microbiota and Related Metabolites following PCSK9 Inhibition in Statin-Treated Patients with Elevated Levels of Lipoprotein(a). Microorganisms 2024; 12:170. [PMID: 38257996 PMCID: PMC10818477 DOI: 10.3390/microorganisms12010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ASCVD) is a leading cause of global mortality, often associated with high blood levels of LDL cholesterol (LDL-c). Medications like statins and PCSK9 inhibitors, are used to manage LDL-c levels and reduce ASCVD risk. Recent findings connect the gut microbiota and its metabolites to ASCVD development. We showed that statins modulate the gut microbiota including the production of microbial metabolites involved in the regulation of cholesterol metabolism such as short chain fatty acids (SCFAs) and bile acids (BAs). Whether this pleiotropic effect of statins is associated with their antimicrobial properties or it is secondary to the modulation of cholesterol metabolism in the host is unknown. In this observational study, we evaluated whether alirocumab, a PCSK9 inhibitor administered subcutaneously, alters the stool-associated microbiota and the profiles of SCFAs and BAs. METHODS We used stool and plasma collected from patients enrolled in a single-sequence study using alirocumab. Microbial DNA was extracted from stool, and the bacterial component of the gut microbiota profiled following an amplicon sequencing strategy targeting the V3-V4 region of the 16S rRNA gene. Bile acids and SCFAs were profiled and quantified in stool and plasma using mass spectrometry. RESULTS Treatment with alirocumab did not alter bacterial alpha (Shannon index, p = 0.74) or beta diversity (PERMANOVA, p = 0.89) in feces. Similarly, circulating levels of SCFAs (mean difference (95% confidence interval (CI)), 8.12 [-7.15-23.36] µM, p = 0.25) and BAs (mean difference (95% CI), 0.04 [-0.11-0.19] log10(nmol mg-1 feces), p = 0.56) were equivalent regardless of PCSK9 inhibition. Alirocumab therapy was associated with increased concentration of BAs in feces (mean difference (95% CI), 0.20 [0.05-0.34] log10(nmol mg-1 feces), p = 0.01). CONCLUSION In statin-treated patients, the use of alirocumab to inhibit PCSK9 leads to elevated levels of fecal BAs without altering the bacterial population of the gut microbiota. The association of alirocumab with increased fecal BA concentration suggests an additional mechanism for the cholesterol-lowering effect of PCSK9 inhibition.
Collapse
Affiliation(s)
- Jose A. Caparrós-Martín
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Patrice Maher
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Natalie C. Ward
- Dobney Hypertension Centre, Medical School, The University of Western Australia, Perth, WA 6009, Australia
| | - Montserrat Saladié
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Patricia Agudelo-Romero
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- The University of Western Australia, Perth, WA 6009, Australia
| | - Stephen M. Stick
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- The University of Western Australia, Perth, WA 6009, Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA 6008, Australia
| | - Dick C. Chan
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Gerald F. Watts
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Cardiometabolic Service, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA 6000, Australia
| | - Fergal O’Gara
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
- BIOMERIT Research Centre, School of Microbiology, University College Cork, T12 XF62 Cork, Ireland
| |
Collapse
|
40
|
Avellaneda-Franco L, Xie L, Nakai M, Barr JJ, Marques FZ. Dietary fiber intake impacts gut bacterial and viral populations in a hypertensive mouse model. Gut Microbes 2024; 16:2407047. [PMID: 39340212 PMCID: PMC11567275 DOI: 10.1080/19490976.2024.2407047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/19/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
The gut microbiome is an emerging factor in preventing hypertension, yet the influence of gut bacteriophages, viruses infecting bacteria, on this condition remains unclear. Bacteriophage-bacteria interactions, which impact the gut microbiome, are influenced differentially by temperate and virulent bacteriophages. However, the standard technique for studying viral populations, viral-like particles (VLPs)-metagenomes, often overlook prophages, the intracellular stage of temperate bacteriophages, creating a knowledge gap. To address this, we investigated alterations in extracellular and intracellular bacteriophages, alongside bacterial populations, in the angiotensin II-hypertension model. We sequenced VLPs and bulk DNA from cecal-colonic samples collected from male C57BL/6J mice implanted with minipumps containing saline or angiotensin II. We assembled 106 bacterial and 816 viral genomes and found that gut viral and bacterial populations remained stable between hypertensive and normotensive mice. A higher number of temperate viruses were observed across all treatments. Although temperate viruses outnumbered virulent viruses, sequencing of both VLPs and bulk revealed that virions from virulent viruses were more abundant in the murine gut. We then evaluated the impact of low- and high-fiber intake on gut microbiome composition in the angiotensin II model. Fiber intake significantly influenced the gut microbiome composition and hypertension development. Mice receiving high-fiber had lower blood pressure, a higher bacterial-encoded carbohydrate-associated enzyme, and a higher total relative abundance of temperate viruses than those receiving low-fiber. Our findings suggest that phages are not associated with hypertension development in the angiotensin II model. However, they support a complex diet-bacteria/phage interaction that may be involved in blood pressure regulation.
Collapse
Affiliation(s)
| | - Liang Xie
- School of Biological Sciences, Monash University, Melbourne, Australia
- Precision Medicine Translational Research Programme, Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michael Nakai
- School of Biological Sciences, Monash University, Melbourne, Australia
| | - Jeremy J. Barr
- School of Biological Sciences, Monash University, Melbourne, Australia
| | - Francine Z. Marques
- School of Biological Sciences, Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| |
Collapse
|
41
|
Jeong S, Hunter SD, Cook MD, Grosicki GJ, Robinson AT. Salty Subjects: Unpacking Racial Differences in Salt-Sensitive Hypertension. Curr Hypertens Rep 2024; 26:43-58. [PMID: 37878224 PMCID: PMC11414742 DOI: 10.1007/s11906-023-01275-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 10/26/2023]
Abstract
PURPOSE OF REVIEW To review underlying mechanisms and environmental factors that may influence racial disparities in the development of salt-sensitive blood pressure. RECENT FINDINGS Our group and others have observed racial differences in diet and hydration, which may influence salt sensitivity. Dietary salt elicits negative alterations to the gut microbiota and immune system, which may increase hypertension risk, but little is known regarding potential racial differences in these physiological responses. Antioxidant supplementation and exercise offset vascular dysfunction following dietary salt, including in Black adults. Furthermore, recent work proposes the role of racial differences in exposure to social determinants of health, and differences in health behaviors that may influence risk of salt sensitivity. Physiological and environmental factors contribute to the mechanisms that manifest in racial differences in salt-sensitive blood pressure. Using this information, additional work is needed to develop strategies that can attenuate racial disparities in salt-sensitive blood pressure.
Collapse
Affiliation(s)
- Soolim Jeong
- Neurovascular Physiology Laboratory (NVPL), School of Kinesiology, Auburn University, Auburn, AL, 36849, USA
| | - Stacy D Hunter
- Department of Health & Human Performance, Texas State University, San Marcos, TX, 78666, USA
| | - Marc D Cook
- Department of Kinesiology, North Carolina Agriculture and Technology State University, Greensboro, NC, 27411, USA
| | - Gregory J Grosicki
- Biodynamics and Human Performance Center, Georgia Southern University (Armstrong Campus), Savannah, GA, 31419, USA
| | - Austin T Robinson
- Neurovascular Physiology Laboratory (NVPL), School of Kinesiology, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
42
|
Charchar FJ, Prestes PR, Mills C, Ching SM, Neupane D, Marques FZ, Sharman JE, Vogt L, Burrell LM, Korostovtseva L, Zec M, Patil M, Schultz MG, Wallen MP, Renna NF, Islam SMS, Hiremath S, Gyeltshen T, Chia YC, Gupta A, Schutte AE, Klein B, Borghi C, Browning CJ, Czesnikiewicz-Guzik M, Lee HY, Itoh H, Miura K, Brunström M, Campbell NR, Akinnibossun OA, Veerabhadrappa P, Wainford RD, Kruger R, Thomas SA, Komori T, Ralapanawa U, Cornelissen VA, Kapil V, Li Y, Zhang Y, Jafar TH, Khan N, Williams B, Stergiou G, Tomaszewski M. Lifestyle management of hypertension: International Society of Hypertension position paper endorsed by the World Hypertension League and European Society of Hypertension. J Hypertens 2024; 42:23-49. [PMID: 37712135 PMCID: PMC10713007 DOI: 10.1097/hjh.0000000000003563] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 09/16/2023]
Abstract
Hypertension, defined as persistently elevated systolic blood pressure (SBP) >140 mmHg and/or diastolic blood pressure (DBP) at least 90 mmHg (International Society of Hypertension guidelines), affects over 1.5 billion people worldwide. Hypertension is associated with increased risk of cardiovascular disease (CVD) events (e.g. coronary heart disease, heart failure and stroke) and death. An international panel of experts convened by the International Society of Hypertension College of Experts compiled lifestyle management recommendations as first-line strategy to prevent and control hypertension in adulthood. We also recommend that lifestyle changes be continued even when blood pressure-lowering medications are prescribed. Specific recommendations based on literature evidence are summarized with advice to start these measures early in life, including maintaining a healthy body weight, increased levels of different types of physical activity, healthy eating and drinking, avoidance and cessation of smoking and alcohol use, management of stress and sleep levels. We also discuss the relevance of specific approaches including consumption of sodium, potassium, sugar, fibre, coffee, tea, intermittent fasting as well as integrated strategies to implement these recommendations using, for example, behaviour change-related technologies and digital tools.
Collapse
Affiliation(s)
- Fadi J. Charchar
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
- Department of Physiology, University of Melbourne, Melbourne, Australia
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Priscilla R. Prestes
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Charlotte Mills
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | - Siew Mooi Ching
- Department of Family Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang
- Department of Medical Sciences, School of Medical and Live Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia
| | - Dinesh Neupane
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, USA
| | - Francine Z. Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne
| | - James E. Sharman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Liffert Vogt
- Department of Internal Medicine, Section Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Louise M. Burrell
- Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
| | - Lyudmila Korostovtseva
- Department of Hypertension, Almazov National Medical Research Centre, St Petersburg, Russia
| | - Manja Zec
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, USA
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Mansi Patil
- Department of Nutrition and Dietetics, Asha Kiran JHC Hospital, Chinchwad
- Hypertension and Nutrition, Core Group of IAPEN India, India
| | - Martin G. Schultz
- Department of Internal Medicine, Section Nephrology, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | | | - Nicolás F. Renna
- Unit of Hypertension, Hospital Español de Mendoza, School of Medicine, National University of Cuyo, IMBECU-CONICET, Mendoza, Argentina
| | | | - Swapnil Hiremath
- Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| | - Tshewang Gyeltshen
- Graduate School of Public Health, St. Luke's International University, Tokyo, Japan
| | - Yook-Chin Chia
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Selangor
- Department of Primary Care Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Abhinav Gupta
- Department of Medicine, Acharya Shri Chander College of Medical Sciences and Hospital, Jammu, India
| | - Aletta E. Schutte
- School of Population Health, University of New South Wales, The George Institute for Global Health, Sydney, New South Wales, Australia
- Hypertension in Africa Research Team, SAMRC Unit for Hypertension and Cardiovascular Disease, North-West University
- SAMRC Developmental Pathways for Health Research Unit, School of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Britt Klein
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Claudio Borghi
- Department of Medical and Surgical Sciences, Faculty of Medicine, University of Bologna, Bologna, Italy
| | - Colette J. Browning
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Marta Czesnikiewicz-Guzik
- School of Medicine, Dentistry and Nursing-Dental School, University of Glasgow, UK
- Department of Periodontology, Prophylaxis and Oral Medicine; Jagiellonian University, Krakow, Poland
| | - Hae-Young Lee
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hiroshi Itoh
- Department of Internal Medicine (Nephrology, Endocrinology and Metabolism), Keio University, Tokyo
| | - Katsuyuki Miura
- NCD Epidemiology Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Mattias Brunström
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Norm R.C. Campbell
- Libin Cardiovascular Institute, Department of Medicine, University of Calgary, Calgary, Canada
| | | | - Praveen Veerabhadrappa
- Kinesiology, Division of Science, The Pennsylvania State University, Reading, Pennsylvania
| | - Richard D. Wainford
- Department of Pharmacology and Experimental Therapeutics, The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston
- Division of Cardiology, Emory University, Atlanta, USA
| | - Ruan Kruger
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Shane A. Thomas
- Health Innovation and Transformation Centre, Federation University Australia, Ballarat, Australia
| | - Takahiro Komori
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Udaya Ralapanawa
- Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | | | - Vikas Kapil
- William Harvey Research Institute, Centre for Cardiovascular Medicine and Devices, NIHR Barts Biomedical Research Centre, BRC, Faculty of Medicine and Dentistry, Queen Mary University London
- Barts BP Centre of Excellence, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Yan Li
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai
| | - Yuqing Zhang
- Department of Cardiology, Fu Wai Hospital, Chinese Academy of Medical Sciences, Chinese Hypertension League, Beijing, China
| | - Tazeen H. Jafar
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore
- Duke Global Health Institute, Duke University, Durham, North Carolina, USA
| | - Nadia Khan
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Bryan Williams
- University College London (UCL), Institute of Cardiovascular Science, National Institute for Health Research (NIHR), UCL Hospitals Biomedical Research Centre, London, UK
| | - George Stergiou
- Hypertension Centre STRIDE-7, School of Medicine, Third Department of Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester
- Manchester Academic Health Science Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
43
|
Moleón J, González-Correa C, Miñano S, Robles-Vera I, de la Visitación N, Barranco AM, Gómez-Guzmán M, Sánchez M, Riesco P, Guerra-Hernández E, Toral M, Romero M, Duarte J. Protective effect of microbiota-derived short chain fatty acids on vascular dysfunction in mice with systemic lupus erythematosus induced by toll like receptor 7 activation. Pharmacol Res 2023; 198:106997. [PMID: 37972724 DOI: 10.1016/j.phrs.2023.106997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Our objective was to investigate whether short-chain fatty acids (SCFAs), specifically acetate and butyrate, could prevent vascular dysfunction and elevated blood pressure (BP) in mice with systemic lupus erythematosus (SLE) induced by TLR7 activation using imiquimod (IMQ). Treatment with both SCFAs and dietary fibers rich in resistant starch (RS) or inulin-type fructans (ITF) effectively prevented the development of hypertension and cardiac hypertrophy. Additionally, these treatments improved aortic relaxation induced by acetylcholine and mitigated vascular oxidative stress. Acetate and butyrate treatments also contributed to the maintenance of colonic integrity, reduced endotoxemia, and decreased the proportion of helper T (Th)17 cells in mesenteric lymph nodes (MLNs), blood, and aorta in TLR7-induced SLE mice. The observed changes in MLNs were correlated with increased levels of GPR43 mRNA in mice treated with acetate and increased GPR41 levels along with decreased histone deacetylase (HDAC)- 3 levels in mice treated with butyrate. Notably, the effects attributed to acetate, but not butyrate, were nullified when co-administered with the GPR43 antagonist GLPG-0974. T cell priming and differentiation into Th17 cells in MLNs, as well as increased Th17 cell infiltration, were linked to aortic endothelial dysfunction and hypertension subsequent to the transfer of faecal microbiota from IMQ-treated mice to germ-free (GF) mice. These effects were counteracted in GF mice through treatment with either acetate or butyrate. To conclude, these findings underscore the potential of SCFA consumption in averting hypertension by restoring balance to the interplay between the gut, immune system, and vascular wall in SLE induced by TLR7 activation.
Collapse
Affiliation(s)
- Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain
| | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Néstor de la Visitación
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Antonio Manuel Barranco
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Manuel Gómez-Guzmán
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Manuel Sánchez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Pedro Riesco
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain
| | | | - Marta Toral
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain; Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain; Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain.
| |
Collapse
|
44
|
Vallianou NG, Kounatidis D, Panagopoulos F, Evangelopoulos A, Stamatopoulos V, Papagiorgos A, Geladari E, Dalamaga M. Gut Microbiota and Its Role in the Brain-Gut-Kidney Axis in Hypertension. Curr Hypertens Rep 2023; 25:367-376. [PMID: 37632662 DOI: 10.1007/s11906-023-01263-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE OF REVIEW The role of the gut microbiota in modulating blood pressure is increasingly being recognized, currently. The purpose of this review is to summarize recent findings about the mechanisms involved in hypertension with regard to the phenomenon of "gut dysbiosis." RECENT FINDINGS Gut dysbiosis, i.e., the imbalance between the gut microbiota and the host, is characterized by a disruption of the tight junction proteins, such as occludins, claudins, and JAMs (junctional adhesion molecules), resulting in increased gut permeability or the so called "leaky gut." Due to the influence of genetic as well as environmental factors, various metabolites produced by the gut microbiota, such as indole and p-cresol, are increased. Thereby, uremic toxins, such as indoxyl sulfates and p-cresol sulfates, accumulate in the blood and the urine, causing damage in the podocytes and the tubular cells. In addition, immunological mechanisms are implicated as well. In particular, a switch from M2 macrophages to M1 macrophages, which produce pro-inflammatory cytokines, occurs. Moreover, a higher level of Th17 cells, releasing large amounts of interleukin-17 (IL-17), has been reported, when a diet rich in salt is consumed. Therefore, apart from the aggravation of uremic toxins, which may account for direct harmful effects on the kidney, there is inflammation not only in the gut, but in the kidneys as well. This crosstalk between the gut and the kidney is suggested to play a crucial role in hypertension. Notably, the brain is also implicated, with an increasing sympathetic output. The brain-gut-kidney axis seems to be deeply involved in the development of hypertension and chronic kidney disease (CKD). The notion that, by modulating the gut microbiota, we could regulate blood pressure is strongly supported by the current evidence. A healthy diet, low in animal protein and fat, and low in salt, together with the utilization of probiotics, prebiotics, synbiotics, or postbiotics, may contribute to our fight against hypertension.
Collapse
Affiliation(s)
| | | | - Fotis Panagopoulos
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | | | | | - Eleni Geladari
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece
| |
Collapse
|
45
|
Pacheco-Yanes J, Reynolds E, Li J, Mariño E. Microbiome-targeted interventions for the control of oral-gut dysbiosis and chronic systemic inflammation. Trends Mol Med 2023; 29:912-925. [PMID: 37730461 DOI: 10.1016/j.molmed.2023.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023]
Abstract
Recent research has confirmed the strong connection between imbalances in the oral and gut microbiome (oral-gut dysbiosis), periodontitis, and inflammatory conditions such as diabetes, Alzheimer's disease, and cardiovascular diseases. Microbiome modulation is crucial for preventing and treating several autoimmune and inflammatory diseases, including periodontitis. However, the causal relationships between the microbiome and its derived metabolites that mediate periodontitis and chronic inflammation constitute a notable knowledge gap. Here we review the mechanisms involved in the microbiome-host crosstalk, and describe novel precision medicine for the control of systemic inflammation. As microbiome-targeted therapies begin to enter clinical trials, the success of these approaches relies upon understanding these reciprocal microbiome-host interactions, and it may provide new therapeutic avenues to reduce the risk of periodontitis-associated diseases.
Collapse
Affiliation(s)
- Juan Pacheco-Yanes
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Eric Reynolds
- Oral Health Collaborative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Victoria, Australia
| | - Jian Li
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Eliana Mariño
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; ImmunoBiota Therapeutics Pty Ltd, Melbourne, Australia.
| |
Collapse
|
46
|
Mueller NT, Liu T, Debelius J, Zhao N. The Gut-Brain-Sleep Connection in Older Adults: What Do We Learn From the Microbiome? J Gerontol A Biol Sci Med Sci 2023; 78:1933-1934. [PMID: 37814931 DOI: 10.1093/gerona/glad049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023] Open
Affiliation(s)
- Noel T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Tiange Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justine Debelius
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ni Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
47
|
Saxami G, Kerezoudi EN, Eliopoulos C, Arapoglou D, Kyriacou A. The Gut-Organ Axis within the Human Body: Gut Dysbiosis and the Role of Prebiotics. Life (Basel) 2023; 13:2023. [PMID: 37895405 PMCID: PMC10608660 DOI: 10.3390/life13102023] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
The human gut microbiota (GM) is a complex microbial ecosystem that colonises the gastrointestinal tract (GIT) and is comprised of bacteria, viruses, fungi, and protozoa. The GM has a symbiotic relationship with its host that is fundamental for body homeostasis. The GM is not limited to the scope of the GIT, but there are bidirectional interactions between the GM and other organs, highlighting the concept of the "gut-organ axis". Any deviation from the normal composition of the GM, termed "microbial dysbiosis", is implicated in the pathogenesis of various diseases. Only a few studies have demonstrated a relationship between GM modifications and disease phenotypes, and it is still unknown whether an altered GM contributes to a disease or simply reflects its status. Restoration of the GM with probiotics and prebiotics has been postulated, but evidence for the effects of prebiotics is limited. Prebiotics are substrates that are "selectively utilized by host microorganisms, conferring a health benefit". This study highlights the bidirectional relationship between the gut and vital human organs and demonstrates the relationship between GM dysbiosis and the emergence of certain representative diseases. Finally, this article focuses on the potential of prebiotics as a target therapy to manipulate the GM and presents the gaps in the literature and research.
Collapse
Affiliation(s)
- Georgia Saxami
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.N.K.); (A.K.)
| | - Evangelia N. Kerezoudi
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.N.K.); (A.K.)
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden
| | - Christos Eliopoulos
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization—Demeter, L. Sof. Venizelou 1, 14123 Lykovryssi, Greece; (C.E.); (D.A.)
| | - Dimitrios Arapoglou
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization—Demeter, L. Sof. Venizelou 1, 14123 Lykovryssi, Greece; (C.E.); (D.A.)
| | - Adamantini Kyriacou
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.N.K.); (A.K.)
| |
Collapse
|
48
|
Ribeiro F, Teixeira M, Alves AJ, Sherwood A, Blumenthal JA. Lifestyle Medicine as a Treatment for Resistant Hypertension. Curr Hypertens Rep 2023; 25:313-328. [PMID: 37470944 DOI: 10.1007/s11906-023-01253-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/21/2023]
Abstract
PURPOSE OF REVIEW Approximately 10% of the adults with hypertension fail to achieve the recommended blood pressure treatment targets on 3 antihypertensive medications or require ≥ 4 medications to achieve goal. These patients with 'resistant hypertension' have an increased risk of target organ damage, adverse clinical events, and all-cause mortality. Although lifestyle modification is widely recommended as a first-line approach for the management of high blood pressure, the effects of lifestyle modifications in patients with resistant hypertension has not been widely studied. This review aims to provide an overview of the emerging evidence on the benefits of lifestyle modifications in patients with resistant hypertension, reviews potential mechanisms by which lifestyles may reduce blood pressure, and discusses the clinical implications of the recent findings in this field. RECENT FINDINGS Evidence from single-component randomized clinical trials demonstrated that aerobic exercise, weight loss and dietary modification can reduce clinic and ambulatory blood pressure in patients with resistant hypertension. Moreover, evidence from multi-component trials involving exercise and dietary modification and weight management can facilitate lifestyle change, reduce clinic and ambulatory blood pressure, and improve biomarkers of cardiovascular risk. This new evidence supports the efficacy of lifestyle modifications added to optimized medical therapy in reducing blood pressure and improving cardiovascular risk biomarkers in patients with resistant hypertension. These findings need to be confirmed in larger studies, and the persistence of benefit over extended follow-up needs further study.
Collapse
Affiliation(s)
- Fernando Ribeiro
- Institute of Biomedicine (iBiMED), School of Health Sciences, University of Aveiro, Aveiro, Portugal
| | - Manuel Teixeira
- Institute of Biomedicine (iBiMED), School of Health Sciences, University of Aveiro, Aveiro, Portugal
| | - Alberto J Alves
- University of Maia, Research Center in Sports Sciences, Health Sciences and Human Development, Castêlo da Maia, Portugal
| | - Andrew Sherwood
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, 27710, USA
| | - James A Blumenthal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
49
|
Vogt L, Marques FZ, Fujita T, Hoorn EJ, Danser AHJ. Novel mechanisms of salt-sensitive hypertension. Kidney Int 2023; 104:690-697. [PMID: 37454911 DOI: 10.1016/j.kint.2023.06.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
A high dietary sodium-consumption level is considered the most important lifestyle factor that can be modified to help prevent an increase in blood pressure and the development of hypertension. Despite numerous studies over the past decades, the pathophysiology explaining why some people show a salt-sensitive blood pressure response and others do not is incompletely understood. Here, a brief overview of the latest mechanistic insights is provided, focusing on the mononuclear phagocytic system and inflammation, the gut-kidney axis, and epigenetics. The article also discusses the effects of 3 types of novel drugs on salt-sensitive hypertension-sodium-glucose cotransporter 2 inhibitors, nonsteroidal mineralocorticoid receptor antagonists, and aldosterone synthase inhibitors. The conclusion is that besides kidney-centered mechanisms, vasoconstrictor mechanisms are also relevant for both the understanding and treatment of this blood pressure phenotype.
Collapse
Affiliation(s)
- Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, and Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Toshiro Fujita
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
50
|
Muralitharan RR, Snelson M, Meric G, Coughlan MT, Marques FZ. Guidelines for microbiome studies in renal physiology. Am J Physiol Renal Physiol 2023; 325:F345-F362. [PMID: 37440367 DOI: 10.1152/ajprenal.00072.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023] Open
Abstract
Gut microbiome research has increased dramatically in the last decade, including in renal health and disease. The field is moving from experiments showing mere association to causation using both forward and reverse microbiome approaches, leveraging tools such as germ-free animals, treatment with antibiotics, and fecal microbiota transplantations. However, we are still seeing a gap between discovery and translation that needs to be addressed, so that patients can benefit from microbiome-based therapies. In this guideline paper, we discuss the key considerations that affect the gut microbiome of animals and clinical studies assessing renal function, many of which are often overlooked, resulting in false-positive results. For animal studies, these include suppliers, acclimatization, baseline microbiota and its normalization, littermates and cohort/cage effects, diet, sex differences, age, circadian differences, antibiotics and sweeteners, and models used. Clinical studies have some unique considerations, which include sampling, gut transit time, dietary records, medication, and renal phenotypes. We provide best-practice guidance on sampling, storage, DNA extraction, and methods for microbial DNA sequencing (both 16S rRNA and shotgun metagenome). Finally, we discuss follow-up analyses, including tools available, metrics, and their interpretation, and the key challenges ahead in the microbiome field. By standardizing study designs, methods, and reporting, we will accelerate the findings from discovery to translation and result in new microbiome-based therapies that may improve renal health.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
- Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Guillaume Meric
- Cambridge-Baker Systems Genomics Initiative, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|