1
|
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
|
2
|
Liu L, Zhou Q, Xu T, Deng Q, Sun Y, Fu J, Chen M, Chen X, Ma Z, Dong Q, Ma B, Jiao Y, Zhou Y, Wu T, Zou H, Shi J, Wang Y, Sheng Y, Tang L, Zheng C, Wu W, Ma W, Sun W, Hu S, Zhou H, He Y, Kong X, Chen L. Non-differential gut microbes contribute to hypertension and its severity through co-abundances: A multi-regional prospective cohort study. IMETA 2025; 4:e268. [PMID: 40027484 PMCID: PMC11865328 DOI: 10.1002/imt2.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 03/05/2025]
Abstract
Microbial dysbiosis, characterized by an imbalanced microbial community structure and function, has been linked to hypertension. While prior research has primarily focused on differential abundances, our study highlights the role of non-differential microbes in hypertension. We propose that non-differential microbes contribute to hypertension through their ecological interactions, as defined by co-abundances (pairs of microbes exhibiting correlated abundance patterns). Using gut microbiome data from the Guangdong Gut Microbiome Project, which includes 2355 hypertensive and 4644 non-hypertensive participants across 14 regions, we identified replicable hypertension-related microbial interactions. Notably, most co-abundances involved non-differential microbes, which were found to correlate with both hypertension severity and hypertension-related microbial metabolic pathways. These findings emphasize the importance of microbial interactions in hypertension pathogenesis and propose a novel perspective for microbiome-based therapeutic strategies.
Collapse
Affiliation(s)
- Lu Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Qianyi Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Tianbao Xu
- Department of Cardiology, The Affiliated Kezhou People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityXinjiangChina
| | - Qiufeng Deng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Yuhao Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Jingxiang Fu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Muxuan Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaojiao Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhenchao Ma
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Quanbin Dong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Beining Ma
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Yuwen Jiao
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Yan Zhou
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Tingting Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Huayiyang Zou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Jing Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Yifeng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Yanhui Sheng
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouChina
| | - Liming Tang
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Chao Zheng
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
- Department of Endocrinology, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Wei Wu
- Guangdong Provincial Institute of Public HealthGuangdong Provincial Center for Disease Control and PreventionGuangzhouChina
| | - Wenjun Ma
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouChina
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Clinical Research Center for Laboratory MedicineGuangzhouChina
- State Key Laboratory of Organ Failure ResearchSouthern Medical UniversityGuangzhouChina
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Clinical Research Center for Laboratory MedicineGuangzhouChina
- State Key Laboratory of Organ Failure ResearchSouthern Medical UniversityGuangzhouChina
- Key Laboratory of Mental Health of the Ministry of EducationGuangzhouChina
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouChina
| | - Lianmin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| |
Collapse
|
3
|
Kindschuh WF, Austin GI, Meydan Y, Park H, Urban JA, Watters E, Pollak S, Saade GR, Chung J, Mercer BM, Grobman WA, Haas DM, Silver RM, Serrano M, Buck GA, McNeil R, Nandakumar R, Reddy U, Wapner RJ, Kav AB, Uhlemann AC, Korem T. Early prediction of preeclampsia using the first trimester vaginal microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626267. [PMID: 39677801 PMCID: PMC11642775 DOI: 10.1101/2024.12.01.626267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Preeclampsia is a severe obstetrical syndrome which contributes to 10-15% of all maternal deaths. Although the mechanisms underlying systemic damage in preeclampsia-such as impaired placentation, endothelial dysfunction, and immune dysregulation-are well studied, the initial triggers of the condition remain largely unknown. Furthermore, although the pathogenesis of preeclampsia begins early in pregnancy, there are no early diagnostics for this life-threatening syndrome, which is typically diagnosed much later, after systemic damage has already manifested. Here, we performed deep metagenomic sequencing and multiplex immunoassays of vaginal samples collected during the first trimester from 124 pregnant individuals, including 62 who developed preeclampsia with severe features. We identified multiple significant associations between vaginal immune factors, microbes, clinical factors, and the early pathogenesis of preeclampsia. These associations vary with BMI, and stratification revealed strong associations between preeclampsia and Bifidobacterium spp., Prevotella timonensis, and Sneathia vaginalis. Finally, we developed machine learning models that predict the development of preeclampsia using this first trimester data, collected ~5.7 months prior to clinical diagnosis, with an auROC of 0.78. We validated our models using data from an independent cohort (MOMS-PI), achieving an auROC of 0.80. Our findings highlight robust associations among the vaginal microbiome, local host immunity, and early pathogenic processes of preeclampsia, paving the way for early detection, prevention and intervention for this devastating condition.
Collapse
Affiliation(s)
- William F. Kindschuh
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - George I. Austin
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Yoli Meydan
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia A. Urban
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily Watters
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Susan Pollak
- Biomarkers Core, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - George R. Saade
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Judith Chung
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of California Irvine, CA, USA
| | - Brian M. Mercer
- Departments of Obstetrics and Gynecology, Case Western Reserve University, Cleveland, OH, USA
| | | | - David M. Haas
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, IN, USA
| | - Robert M. Silver
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA
| | - Myrna Serrano
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, VA, USA
| | - Gregory A. Buck
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, VA, USA
- Department of Computer Science, School of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Renu Nandakumar
- Biomarkers Core, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Uma Reddy
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ronald J. Wapner
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Aya Brown Kav
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
4
|
Wu Y, Li Z, Shi L, Zhu Y, Wang Y, Yan N, Yang Y, He S, Li J. Effects of leachate from disposable plastic takeout containers on the cardiovascular system after thermal contact. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117383. [PMID: 39591733 DOI: 10.1016/j.ecoenv.2024.117383] [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: 09/23/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
The study investigated the cardiovascular effects of daily exposure to plastic products by simulating the oral heat exposure mode of disposable plastic takeout containers (DPTC) commonly used in society. Questionnaires were used to randomly choose 3179 people in order to examine any possible correlation between the frequency of plastic exposure and the risk of cardiovascular diseases (CVD). Additionally, Sprague-Dawley(SD) rats consumed leachate from DPTC exposed to boiling water for 1 minute,5 minutes and 15 minutes respectively, over three months. After intervention, fecal samples were taken for microbiota and metabolomics analysis, and rat cardiac tissue was studied by staining and electron microscopy. Serum parameters were tested to analyze cardiovascular system changes. The population-based plastic exposure questionnaire data revealed that high-frequency exposure to plastics is significantly associated with an increased risk of congestive heart failure, with an odds ratio of 1.13 (95 % CI: 1.03-1.24). Rat fecal analysis revealed that β diversity and composition of gut microbiota in experimental groups were changed. Inflammatory cell infiltration, mitochondrial swelling, and serum indicators of oxidative stress and inflammation were significantly elevated in the myocardium, without temporal differences observed. The study shows plastic exposure as a significant CVD risk factor regardless of duration. It leads to changes in myocardial tissue, gut microbiota, and metabolites, all closely tied to CVD.
Collapse
Affiliation(s)
- Yueping Wu
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Zhuoyuan Li
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Liping Shi
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Yongbin Zhu
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Yanrong Wang
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Ning Yan
- Heart Centre & Department of Cardiovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Yue Yang
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Shulan He
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Jiangping Li
- Department of Epidemiology and Health Statistics, School of public health, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| |
Collapse
|
5
|
Młynarska E, Wasiak J, Gajewska A, Bilińska A, Steć G, Jasińska J, Rysz J, Franczyk B. Gut Microbiota and Gut-Brain Axis in Hypertension: Implications for Kidney and Cardiovascular Health-A Narrative Review. Nutrients 2024; 16:4079. [PMID: 39683474 DOI: 10.3390/nu16234079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
INTRODUCTION Arterial hypertension is a major contributor to a wide range of health complications, with cardiac hypertrophy and chronic kidney disease being among the most prevalent. Consequently, novel strategies for the treatment and prevention of hypertension are actively being explored. Recent research has highlighted a potential link between hypertension and the gut-brain axis. A bidirectional communication between the microbiota and the brain via the vagus nerve, enteric nervous system, hypothalamus-pituitary-adrenal axis, secreted short-chain fatty acids, and neurotransmitter metabolism. MATERIALS AND METHODS A comprehensive literature search was conducted using databases such as PubMed to identify studies exploring the relationship between gut microbiota and hypertension, along with the effects of dietary interventions and probiotics on blood pressure regulation. DISCUSSION Studies in both animal models and human subjects have demonstrated a strong correlation between alterations in gut microbiota composition and the development of hypertension. By influencing blood pressure, the gut microbiota can potentially affect the progression of cardiovascular and kidney disorders. Modulating gut microbiota through dietary interventions and probiotics has shown promise in regulating blood pressure and reducing systemic inflammation, offering a novel approach to managing hypertension. Diets such as the Mediterranean diet, which is rich in polyphenols and omega-3 fatty acids and low in sodium, promote the growth of beneficial gut bacteria that support cardiovascular health. Additionally, probiotics have been found to enhance gut barrier function, reduce inflammation, and modulate the Renin-Angiotensin System, all of which contribute to lowering blood pressure. CONCLUSIONS Further research is needed to determine the mechanisms of action of the microbiota in hypertension. The aim of this study was to evaluate the influence of gut microbiota on blood pressure regulation and the progression of hypertension-related complications, such as cardiovascular and kidney disorders.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Aleksandra Bilińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Greta Steć
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Jasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
6
|
Nagasawa H, Suzuki S, Kobayashi T, Otsuka T, Okuma T, Matsushita S, Amano A, Shimizu Y, Suzuki Y, Ueda S. Effect of fruits granola (Frugra®) consumption on blood pressure reduction and intestinal microbiome in patients undergoing hemodialysis. Hypertens Res 2024; 47:3214-3224. [PMID: 39300301 PMCID: PMC11534689 DOI: 10.1038/s41440-024-01895-1] [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: 03/13/2024] [Revised: 08/06/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
Cardiovascular diseases (CVDs) are a major cause of death in patients undergoing hemodialysis (HD). Blood pressure (BP) and uremic toxins are well-known risk factors for CVDs, which are influenced by diet. Dietary fiber supplementation in patients undergoing HD may reduce the risk of CVDs by improving lipid profiles and inflammatory status and lowering the levels of the uremic toxin indoxyl sulfate (IS). In this study, we investigated the relationship between the intestinal microbiota and risk factors for CVDs, such as BP and serum IS, in patients undergoing HD who consumed fruits granola (FGR). The study participants were selected from patients undergoing HD at the Izu Nagaoka Daiichi Clinic and consumed FGR for 2 months. Body composition and blood samples were tested at months 0, 1, 2 and fecal samples were collected at months 0 and 2 for intestinal microbiota analysis. FGR consumption decreased systolic and diastolic BP, estimated salt intake, and serum IS levels and improved the stool characteristics according to the Bristol Stool Form Scale (N = 24). Gut microbiota analysis showed an increase in the alpha diversity and abundance of Blautia and Neglecta. The abundance of lactic acid- and ethanol-producing bacteria also significantly increased, whereas the abundance of indole-producing bacteria significantly decreased. FGR consumption could be a useful tool for salt reduction, fiber supplementation, and improvement of the intestinal environment, thus contributing to improvement of BP and the reduction of other risk factors for CVDs in patients undergoing HD.
Collapse
Affiliation(s)
- Hajime Nagasawa
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Division of Nephrology, Department of Internal Medicine, Juntendo University Shizuoka Hospital, Shizuoka, Japan
- Department of Granola Health Care and Preventive Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Division of Kidney Health and Aging, the Center for Integrated Kidney Research and Advance, Shimane University Faculty of Medicine, Shimane, Japan
| | - Shogo Suzuki
- Department of Radiological Technology, Juntendo University Faculty of Health Science, Tokyo, Japan
| | - Takashi Kobayashi
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Granola Health Care and Preventive Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Tomoyuki Otsuka
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Granola Health Care and Preventive Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Teruyuki Okuma
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Granola Health Care and Preventive Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Division of Kidney Health and Aging, the Center for Integrated Kidney Research and Advance, Shimane University Faculty of Medicine, Shimane, Japan
| | - Satoshi Matsushita
- Department of Granola Health Care and Preventive Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Cardiovascular Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Atsushi Amano
- Department of Granola Health Care and Preventive Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Cardiovascular Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yoshio Shimizu
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Division of Nephrology, Department of Internal Medicine, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Seiji Ueda
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan.
- Department of Granola Health Care and Preventive Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan.
- Division of Kidney Health and Aging, the Center for Integrated Kidney Research and Advance, Shimane University Faculty of Medicine, Shimane, Japan.
| |
Collapse
|
7
|
Li J, Wang S, Yan K, Wang P, Jiao J, Wang Y, Chen M, Dong Y, Zhong J. Intestinal microbiota by angiotensin receptor blocker therapy exerts protective effects against hypertensive damages. IMETA 2024; 3:e222. [PMID: 39135690 PMCID: PMC11316932 DOI: 10.1002/imt2.222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 08/15/2024]
Abstract
Dysbiosis of the gut microbiota has been implicated in hypertension, and drug-host-microbiome interactions have drawn considerable attention. However, the influence of angiotensin receptor blocker (ARB)-shaped gut microbiota on the host is not fully understood. In this work, we assessed the alterations of blood pressure (BP), vasculatures, and intestines following ARB-modified gut microbiome treatment and evaluated the changes in the intestinal transcriptome and serum metabolome in hypertensive rats. Hypertensive patients with well-controlled BP under ARB therapy were recruited as human donors, spontaneously hypertensive rats (SHRs) receiving normal saline or valsartan were considered animal donors, and SHRs were regarded as recipients. Histological and immunofluorescence staining was used to assess the aorta and small intestine, and 16S rRNA amplicon sequencing was performed to examine gut bacteria. Transcriptome and metabonomic analyses were conducted to determine the intestinal transcriptome and serum metabolome, respectively. Notably, ARB-modified fecal microbiota transplantation (FMT), results in marked decreases in systolic BP levels, collagen deposition and reactive oxygen species accumulation in the vasculature, and alleviated intestinal structure impairments in SHRs. These changes were linked with the reconstruction of the gut microbiota in SHR recipients post-FMT, especially with a decreased abundance of Lactobacillus, Aggregatibacter, and Desulfovibrio. Moreover, ARB-treated microbes contributed to increased intestinal Ciart, Per1, Per2, Per3, and Cipc gene levels and decreased Nfil3 and Arntl expression were detected in response to ARB-treated microbes. More importantly, circulating metabolites were dramatically reduced in ARB-FMT rats, including 6beta-Hydroxytestosterone and Thromboxane B2. In conclusion, ARB-modified gut microbiota exerts protective roles in vascular remodeling and injury, metabolic abnormality and intestinal dysfunctions, suggesting a pivotal role in mitigating hypertension and providing insights into the cross-talk between antihypertensive medicines and the gut microbiome.
Collapse
Affiliation(s)
- Jing Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Si‐Yuan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Kai‐Xin Yan
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Pan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Jie Jiao
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Yi‐Dan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Mu‐Lei Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Ying Dong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| | - Jiu‐Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
- Department of Cardiology, Beijing Chaoyang HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
8
|
Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, Asleh R. Human Gut Microbiota in Cardiovascular Disease. Compr Physiol 2024; 14:5449-5490. [PMID: 39109979 DOI: 10.1002/cphy.c230012] [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: 01/03/2025]
Abstract
The gut ecosystem, termed microbiota, is composed of bacteria, archaea, viruses, protozoa, and fungi and is estimated to outnumber human cells. Microbiota can affect the host by multiple mechanisms, including the synthesis of metabolites and toxins, modulating inflammation and interaction with other organisms. Advances in understanding commensal organisms' effect on human conditions have also elucidated the importance of this community for cardiovascular disease (CVD). This effect is driven by both direct CV effects and conditions known to increase CV risk, such as obesity, diabetes mellitus (DM), hypertension, and renal and liver diseases. Cardioactive metabolites, such as trimethylamine N -oxide (TMAO), short-chain fatty acids (SCFA), lipopolysaccharides, bile acids, and uremic toxins, can affect atherosclerosis, platelet activation, and inflammation, resulting in increased CV incidence. Interestingly, this interaction is bidirectional with microbiota affected by multiple host conditions including diet, bile acid secretion, and multiple diseases affecting the gut barrier. This interdependence makes manipulating microbiota an attractive option to reduce CV risk. Indeed, evolving data suggest that the benefits observed from low red meat and Mediterranean diet consumption can be explained, at least partially, by the changes that these diets may have on the gut microbiota. In this article, we depict the current epidemiological and mechanistic understanding of the role of microbiota and CVD. Finally, we discuss the potential therapeutic approaches aimed at manipulating gut microbiota to improve CV outcomes. © 2024 American Physiological Society. Compr Physiol 14:5449-5490, 2024.
Collapse
Affiliation(s)
- Daniel Ronen
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Rokach
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abed Qadan
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samar Daana
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
9
|
Cui X, Zhang T, Xie T, Guo FX, Zhang YY, Deng YJ, Wang Q, Guo YX, Dong MH, Luo XT. Research Progress on the Correlation Between Hypertension and Gut Microbiota. J Multidiscip Healthc 2024; 17:2371-2387. [PMID: 38770171 PMCID: PMC11104380 DOI: 10.2147/jmdh.s463880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
Among cardiovascular diseases, hypertension is the most important risk factor for morbidity and mortality worldwide, and its pathogenesis is complex, involving genetic, dietary and environmental factors. The characteristics of the gut microbiota can vary in response to increased blood pressure (BP) and influence the development and progression of hypertension. This paper describes five aspects of the relationship between hypertension and the gut microbiota, namely, the different types of gut microbiota, metabolites of the gut microbiota, sympathetic activation, gut-brain interactions, the effects of exercise and dietary patterns and the treatment of the gut microbiota through probiotics, faecal microbiota transplantation (FMT) and herbal remedies, providing new clues for the future prevention of hypertension. Diet, exercise and traditional Chinese medicine may contribute to long-term improvements in hypertension, although the effects of probiotics and FMT still need to be validated in large populations.
Collapse
Affiliation(s)
- Xiaomei Cui
- Key Laboratory of Cardio Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, People’s Republic of China
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Ting Zhang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Tao Xie
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Fang-xi Guo
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Yu-ying Zhang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Yuan-jia Deng
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Qi Wang
- Key Laboratory of Cardio Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, People’s Republic of China
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Yi-xing Guo
- Key Laboratory of Cardio Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, People’s Republic of China
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Ming-hua Dong
- Key Laboratory of Cardio Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, People’s Republic of China
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, People’s Republic of China
| | - Xiao-ting Luo
- Key Laboratory of Cardio Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, People’s Republic of China
- School of General Medicine, Gannan Medical University, Ganzhou, People’s Republic of China
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Gao K, Wang PX, Mei X, Yang T, Yu K. Untapped potential of gut microbiome for hypertension management. Gut Microbes 2024; 16:2356278. [PMID: 38825779 PMCID: PMC11152106 DOI: 10.1080/19490976.2024.2356278] [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: 12/08/2023] [Accepted: 05/13/2024] [Indexed: 06/04/2024] Open
Abstract
The gut microbiota has been shown to be associated with a range of illnesses and disorders, including hypertension, which is recognized as the primary factor contributing to the development of serious cardiovascular diseases. In this review, we conducted a comprehensive analysis of the progression of the research domain pertaining to gut microbiota and hypertension. Our primary emphasis was on the interplay between gut microbiota and blood pressure that are mediated by host and gut microbiota-derived metabolites. Additionally, we elaborate the reciprocal communication between gut microbiota and antihypertensive drugs, and its influence on the blood pressure of the host. The field of computer science has seen rapid progress with its great potential in the application in biomedical sciences, we prompt an exploration of the use of microbiome databases and artificial intelligence in the realm of high blood pressure prediction and prevention. We propose the use of gut microbiota as potential biomarkers in the context of hypertension prevention and therapy.
Collapse
Affiliation(s)
- Kan Gao
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Pu Xiu Wang
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xue Mei
- School of Pharmacy, Institute of Materia Medica, North Sichuan Medical College, Nanchang, Sichuan, China
| | - Tao Yang
- Department of Physiology and Pharmacology, Center for Hypertension and Precision Medicine, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, USA
| | - Kai Yu
- Department of General Practice, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
13
|
Perrotta S, Carnevale D. Brain-Splenic Immune System Interactions in Hypertension: Cellular and Molecular Mechanisms. Arterioscler Thromb Vasc Biol 2024; 44:65-75. [PMID: 37942610 DOI: 10.1161/atvbaha.123.318230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 10/20/2023] [Indexed: 11/10/2023]
Abstract
Hypertension represents a major worldwide cause of death and disability, and it is becoming increasingly clear that available therapies are not sufficient to reduce the risk of major cardiovascular events. Various mechanisms contribute to blood pressure increase: neurohormonal activation, autonomic nervous system imbalance, and immune activation. Of note, the brain is an important regulator of blood pressure levels; it recognizes the peripheral perturbation and organizes a reflex response by modulating immune system and hormonal release to attempt at restoring the homeostasis. The connection between the brain and peripheral organs is mediated by the autonomic nervous system, which also modulates immune and inflammatory responses. Interestingly, an increased autonomic nervous system activity has been correlated with an altered immune response in cardiovascular diseases. The spleen is the largest immune organ exerting a potent influence on the cardiovascular system during disease and is characterized by a dense noradrenergic innervation. Taken together, these aspects led to hypothesize a key role of neuroimmune mechanisms in the onset and progression of hypertension. This review discusses how the nervous and splenic immune systems interact and how the mechanisms underlying the neuroimmune cross talk influence the disease progression.
Collapse
Affiliation(s)
- Sara Perrotta
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
- Department of Molecular Medicine, "Sapienza" University of Rome, Italy (D.C.)
| |
Collapse
|
14
|
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
|
15
|
Pedroza Matute S, Iyavoo S. Exploring the gut microbiota: lifestyle choices, disease associations, and personal genomics. Front Nutr 2023; 10:1225120. [PMID: 37867494 PMCID: PMC10585655 DOI: 10.3389/fnut.2023.1225120] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
The gut microbiota is a rich and dynamic ecosystem that actively interacts with the human body, playing a significant role in the state of health and disease of the host. Diet, exercise, mental health, and other factors have exhibited the ability to influence the gut bacterial composition, leading to changes that can prevent and improve, or favor and worsen, both intestinal and extra-intestinal conditions. Altered gut microbial states, or 'dysbiosis', associated with conditions and diseases are often characterized by shifts in bacterial abundance and diversity, including an impaired Firmicutes to Bacteroidetes ratio. By understanding the effect of lifestyle on the gut microbiota, personalized advice can be generated to suit each individual profile and foster the adoption of lifestyle changes that can both prevent and ameliorate dysbiosis. The delivery of effective and reliable advice, however, depends not only on the available research and current understanding of the topic, but also on the methods used to assess individuals and to discover the associations, which can introduce bias at multiple stages. The aim of this review is to summarize how human gut microbial variability is defined and what lifestyle choices and diseases have shown association with gut bacterial composition. Furthermore, popular methods to investigate the human gut microbiota are outlined, with a focus on the possible bias caused by the lack of use of standardized methods. Finally, an overview of the current state of personalized advice based on gut microbiota testing is presented, underlining its power and limitations.
Collapse
Affiliation(s)
| | - Sasitaran Iyavoo
- Nkaarco Diagnostics Limited, Norwich, United Kingdom
- School of Chemistry, College of Health and Science, University of Lincoln, Lincoln, United Kingdom
| |
Collapse
|
16
|
Murugesan S, Al Khodor S. Salivary microbiome and hypertension in the Qatari population. J Transl Med 2023; 21:454. [PMID: 37422685 PMCID: PMC10329805 DOI: 10.1186/s12967-023-04247-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/06/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND The prevalence of hypertension in Qatar is 33 percent of the adult population. It is postulated that the salivary microbiome can regulate blood pressure (BP). However, limited investigations exist to prove this hypothesis. Therefore, we examined the difference in the salivary microbiome composition between hypertensive and normotensive Qatari subjects. METHODS A total of 1190 Qatar Genome Project (QGP) participants (Mean age = 43 years) were included in this study. BP for all participants was classified into Normal (n = 357), Stage1 (n = 336), and Stage2: (n = 161) according to the American Heart Association guidelines. 16S-rRNA libraries were sequenced and analyzed using QIIME-pipeline, and PICRUST was used to predict functional metabolic routes. Machine Learning (ML) strategies were applied to identify salivary microbiome-based predictors of hypertension. RESULTS Differential abundant analysis (DAA) revealed that Bacteroides and Atopobium were the significant members of the hypertensive groups. Alpha and beta diversity indices indicated dysbiosis between the normotensive and hypertensive groups. ML-based prediction models revealed that these markers could predict hypertension with an AUC (Area under the curve) of 0.89. Functional predictive analysis disclosed that Cysteine and Methionine metabolism and the sulphur metabolic pathways involving the renin-angiotensin system were significantly higher in the normotensive group. Therefore, members of Bacteroides and Atopobium can serve as predictors of hypertension. Likewise, Prevotella, Neisseria, and Haemophilus can be the protectors that regulate BP via nitric acid synthesis and regulation of the renin-angiotensin system. CONCLUSION It is one of the first studies to assess salivary microbiome and hypertension as disease models in a large cohort of the Qatari population. Further research is needed to confirm these findings and validate the mechanisms involved.
Collapse
Affiliation(s)
- Selvasankar Murugesan
- Maternal and Child Health Division, Research Department, Sidra Medicine, 26999, Doha, Qatar
| | - Souhaila Al Khodor
- Maternal and Child Health Division, Research Department, Sidra Medicine, 26999, Doha, Qatar.
| |
Collapse
|
17
|
Wang X, Chen W, Jin R, Xu X, Wei J, Huang H, Tang Y, Zou C, Chen T. Engineered probiotics Clostridium butyricum-pMTL007-GLP-1 improves blood pressure via producing GLP-1 and modulating gut microbiota in spontaneous hypertension rat models. Microb Biotechnol 2023; 16:799-812. [PMID: 36528874 PMCID: PMC10034621 DOI: 10.1111/1751-7915.14196] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Hypertension is a significant risk factor of cardiovascular diseases (CVDs) with high prevalence worldwide, the current treatment has multiple adverse effects and requires continuous administration. The glucagon-like peptide-1 receptor (GLP-1R) agonists have shown great potential in treating diabetes mellitus, neurodegenerative diseases, obesity and hypertension. Butyric acid is a potential target in treating hypertension. Yet, the application of GLP-1 analogue and butyric acid in reducing blood pressure and reversing ventricular hypertrophy remains untapped. In this study, we combined the therapeutic capability of GLP-1 and butyric acid by transforming Clostridium butyricum (CB) with recombinant plasmid pMTL007 encoded with hGLP gene to construct the engineered probiotics Clostridium butyricum-pMTL007-GLP-1 (CB-GLP-1). We used spontaneous hypertensive rat (SHR) models to evaluate the positive effect of this strain in treating hypertension. The results revealed that the intragastric administration of CB-GLP-1 had markedly reduced blood pressure and improved cardiac marker ACE2, AT2R, AT1R, ANP, BNP, β-MHC, α-SMA and activating AMPK/mTOR/p70S6K/4EBP1 signalling pathway. The high-throughput sequencing further demonstrated that CB-GLP-1 treatments significantly improved the dysbiosis in the SHR rats via downregulating the relative abundance of Porphyromonadaceae at the family level and upregulating Lactobacillus at the genus level. Hence, we concluded that the CB-GLP-1 greatly improves blood pressure and cardiomegaly by restoring the gut microbiome and reducing ventricular hypertrophy in rat models. This is the first time using engineered CB in treating hypertension, which provides a new idea for the clinical treatment of hypertension.
Collapse
Affiliation(s)
- Xin‐liang Wang
- Key Laboratory of Poyang Lake Environment and Resource Utilization, School of Resources Environmental and Chemical EngineeringMinistry of Education, Nanchang UniversityNanchangChina
- National Engineering Research Center for Bioengineering Drugs and TechnologiesInstitute of Translational Medicine, Nanchang UniversityNanchangChina
| | - Wen‐jie Chen
- National Engineering Research Center for Bioengineering Drugs and TechnologiesInstitute of Translational Medicine, Nanchang UniversityNanchangChina
| | - Rui Jin
- National Engineering Research Center for Bioengineering Drugs and TechnologiesInstitute of Translational Medicine, Nanchang UniversityNanchangChina
| | - Xuan Xu
- National Engineering Research Center for Bioengineering Drugs and TechnologiesInstitute of Translational Medicine, Nanchang UniversityNanchangChina
| | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and TechnologiesInstitute of Translational Medicine, Nanchang UniversityNanchangChina
| | - Hong Huang
- Key Laboratory of Poyang Lake Environment and Resource Utilization, School of Resources Environmental and Chemical EngineeringMinistry of Education, Nanchang UniversityNanchangChina
| | - Yan‐hua Tang
- Department of Cardiovascular SurgeryThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Chang‐wei Zou
- Key Laboratory of Poyang Lake Environment and Resource Utilization, School of Resources Environmental and Chemical EngineeringMinistry of Education, Nanchang UniversityNanchangChina
| | - Ting‐tao Chen
- National Engineering Research Center for Bioengineering Drugs and TechnologiesInstitute of Translational Medicine, Nanchang UniversityNanchangChina
| |
Collapse
|
18
|
Kang G, He H, Miao H, Zhang T, Meng Z, Li X. Predictive value of gut microbiota in long-term blood pressure control: a cross-sectional study. Eur J Med Res 2023; 28:115. [PMID: 36907902 PMCID: PMC10008596 DOI: 10.1186/s40001-022-00944-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/09/2022] [Indexed: 03/14/2023] Open
Abstract
OBJECTIVES To investigate the prediction of long-term blood pressure control using the intestinal flora of patients with hypertension. METHODS A total of 125 patients with primary grade-2 hypertension who attended the cardiovascular department of Hebei Province Hospital of Chinese Medicine between April 2021 and April 2022 were enrolled; these included 65 patients with substandard long-term blood pressure control (the uncontrolled group) and 60 patients with standard long-term blood pressure control (the controlled group). General clinical data and data on morning stools and diet were recorded for all the enrolled patients. The 16 s rDNA sequencing of faecal intestinal flora was also performed to analyse the differences in intestinal flora between the two groups of patients and to investigate the relationship between blood pressure compliance and the presence of flora. RESULTS The intestinal flora of the two groups of patients differed in terms of the Firmicutes-Bacteroidetes ratio (F/B), α-diversity analysis (Chao1, ACE and Shannon) results and β-diversity analysis results. At the genus level, the number of Streptococcus and Paraprevotella in patients in the uncontrolled group was greater than that of the controlled group, and the level of Akkermansia and Bifidobacterium was lower than that in the controlled group. A logistic regression analysis of the difference factors found differences in ACE, F/B, Streptococcus, Paraprevotella and Akkermansia in the two groups; these differences remained after correcting for age, gender and body mass index. The receiver operating characteristic curves revealed the following: ACE (area under the curve [AUC] = 85.282), Streptococcus (AUC = 82.705), Akkermansia (AUC = 77.333), Paraprevotella (AUC = 66.154) and F/B (AUC = 60.436). CONCLUSIONS There were significant differences in the intestinal flora of the patients in the controlled blood group compared with that of the uncontrolled group. Therefore, the ACE, genus levels of Streptococcus and Akkermansia could provide some prediction of late blood pressure compliance or non-compliance in patients with hypertension.
Collapse
Affiliation(s)
- Guobin Kang
- Department of Cardiology, Hebei Province Hospital of Chinese Medicine, No. 389 of Zhongshan East Street, Chang'an District, Shijiazhuang, 050000, Hebei, China
| | - Hongtao He
- Department of Cardiology, Hebei Province Hospital of Chinese Medicine, No. 389 of Zhongshan East Street, Chang'an District, Shijiazhuang, 050000, Hebei, China
| | - Huawei Miao
- Department of Cardiology, Hebei Province Hospital of Chinese Medicine, No. 389 of Zhongshan East Street, Chang'an District, Shijiazhuang, 050000, Hebei, China
| | - Tiejun Zhang
- Department of Cardiology, Hebei Province Hospital of Chinese Medicine, No. 389 of Zhongshan East Street, Chang'an District, Shijiazhuang, 050000, Hebei, China
| | - Zongde Meng
- Department of Internal Medicine, Hebei Province Hospital of Chinese Medicine, Shijiazhuang, 050000, Hebei, China
| | - Xia Li
- Department of Cardiology, Hebei Province Hospital of Chinese Medicine, No. 389 of Zhongshan East Street, Chang'an District, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
19
|
Abstract
A large body of evidence has emerged in the past decade supporting a role for the gut microbiome in the regulation of blood pressure. The field has moved from association to causation in the last 5 years, with studies that have used germ-free animals, antibiotic treatments and direct supplementation with microbial metabolites. The gut microbiome can regulate blood pressure through several mechanisms, including through gut dysbiosis-induced changes in microbiome-associated gene pathways in the host. Microbiota-derived metabolites are either beneficial (for example, short-chain fatty acids and indole-3-lactic acid) or detrimental (for example, trimethylamine N-oxide), and can activate several downstream signalling pathways via G protein-coupled receptors or through direct immune cell activation. Moreover, dysbiosis-associated breakdown of the gut epithelial barrier can elicit systemic inflammation and disrupt intestinal mechanotransduction. These alterations activate mechanisms that are traditionally associated with blood pressure regulation, such as the renin-angiotensin-aldosterone system, the autonomic nervous system, and the immune system. Several methodological and technological challenges remain in gut microbiome research, and the solutions involve minimizing confounding factors, establishing causality and acting globally to improve sample diversity. New clinical trials, precision microbiome medicine and computational methods such as Mendelian randomization have the potential to enable leveraging of the microbiome for translational applications to lower blood pressure.
Collapse
|
20
|
Zhang Q, Meng N, Liu Y, Zhao H, Zhao Z, Hao D, Li R, Han K, Li H, Ma J, Yu X, Qi Z, Li Q. Protection effect of gut microbiota composition and acetate absorption against hypertension-induced damages on the longevity population in Guangxi, China. Front Nutr 2023; 9:1070223. [PMID: 36726815 PMCID: PMC9884688 DOI: 10.3389/fnut.2022.1070223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/20/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Recent evidence supports a role for the gut microbe-metabolites in longevity. However, the phenomenon of hypertension is more common in the longevity area and whether hypertension is associated with longevity remains unclear. Here, we hypothesize that the levels of gut microbiota, SCFAs, and urine metabolites were different between hypertension elderly and hypertension longevity. Methods We recruited 46 elderly volunteers from Donglan County, Guangxi, and 32 were selected and included in the experiment. The subjects with hypertension were divided into two groups according to age, Hypertension Elderly (HTE, aged 70.5 ± 8.59, n = 19) and Hypertension Longevity (HTL, aged 100 ± 5.72, n = 13). The gut microbiota, SCFAs, and urine metabolites were determined by three-generation 16S rRNA full-length sequencing, GC-MS, and 1H-NMR, respectively. Results Compared with the HTL group, the HTE group had higher levels of hypertension-related genera Klebsiella and Streptococcus, while having lower levels of the SCFA-producing genera Bacteroides, Faecalibacterium, and Alistipes. Based on LEFse analysis, Klebsiella pneumoniae, Lactobacillus gasseri, Streptococcus salivarius, Ruminococcus, Actinomyces, Rikenellaceae, f_Saccharimonadaceae, Clostridium perfringens, and Bacteroids, Faecalibacterium prausnitzii, Parabacteroides, Alistipes were biomarkers that showed significant differences between the groups. In addition, the microbial pathways associated with K. pneumoniae and E. coli may promote hypertension, while A. muciniphila may play a role in reversing the development of hypertension in long-lived elderly. Metabolomics revealed that HTL contained a lower concentration of fecal acetate and propionate than HTE, while it contained a higher concentration of serum acetate and urine acetate. Furthermore, their immune cells exhibited no significant changes in SCFAs receptors. Conclusion Although long-lived elderly have extremely high systolic blood pressure, their unique gut microbiota composition and efficient acetate absorption in the colon may offset the damages caused by hypertension and maintain healthy homeostasis.
Collapse
Affiliation(s)
- Qinren Zhang
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Ning Meng
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Yu Liu
- Medical College, Guangxi University, Nanning, China
| | - Haiyan Zhao
- Medical College, Guangxi University, Nanning, China
| | - Zhengtao Zhao
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Dan Hao
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Ruiding Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Kunchen Han
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - He Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Jinke Ma
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Xiaohan Yu
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning, China,Zhongquan Qi,
| | - Quanyang Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning, China,*Correspondence: Quanyang Li,
| |
Collapse
|