1
|
Xie Z, Chen Z, Chai Y, Yao W, Ma G. Unveiling the placental bacterial microbiota: implications for maternal and infant health. Front Physiol 2025; 16:1544216. [PMID: 40161970 PMCID: PMC11949977 DOI: 10.3389/fphys.2025.1544216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
The human placenta is a unique organ that forms under specific physiological conditions and plays a crucial role in nutrient and metabolite exchange between the mother and fetus. Research on the placenta is important for understanding maternal-fetal diseases. Traditionally, the placenta was considered "sterile," but advancements in detection techniques have revealed the presence of a low level of microorganisms. This discovery challenges the traditional notion that the uterine placenta is sterile. The revelation of this truth marks a significant breakthrough in medical research, prompting more researchers to focus on this vital organ, the placenta. Placental microbial communities may originate from the oral, vaginal, and intestinal microbiota of expectant mothers. These microorganisms may reach the maternal-fetal interface, collectively shaping the placental microbiota and contributing to the composition of normal placental microbial communities. Abnormal placental microbial communities may be associated with some pregnancy complications and fetal developmental issues such as preterm birth, gestational hypertension, fetal growth restriction, and gestational diabetes mellitus. Intervention strategies targeting microbial communities, which include modulation of placental microbiota composition or function, such as probiotics, may help prevent or treat complications related to abnormal placental microbiota during pregnancy.
Collapse
Affiliation(s)
- Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Chai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wang Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Okunlola FO, Okunlola AR, Adetuyi BO, Soliman MES, Alexiou A, Papadakis M, Fawzy MN, El-Saber Batiha G. Beyond the gut: Unraveling the multifaceted influence of microbiome on cardiovascular health. Clin Nutr ESPEN 2025; 67:71-89. [PMID: 40064239 DOI: 10.1016/j.clnesp.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Cardiovascular disease is one of the leading causes of death worldwide. Even while receiving adequate pharmacological treatment for their hypertension, people are nonetheless at greater risk for cardiovascular disease. There is growing evidence that the gut microbiota may have major positive and negative effects on blood pressure and illnesses related with it as more study into this topic is conducted. Trimethylamine n-oxide (TMAO) and short-chain fatty acids (SCFA) are two major by-products of the gut microbiota. TMAO is involved in the formation of other coronary artery diseases, including atherosclerosis and hypertension, while SCFAs play an important role in controlling blood pressure. Numerous investigations have confirmed the established link between dietary salt intake and hypertension. Reducing sodium in the diet is linked to lower rates of cardiovascular disease morbidity and mortality as well as lower rates of blood pressure and hypertension. In both human and animal research, high salt diets increase local and systemic tissue inflammation and compromise gut architecture. Given that the gut microbiota constantly interacts with the immune system and is required for the correct maturation of immune cells, it is scientifically conceivable that it mediates the inflammatory response. This review highlights the therapeutic possibilities for focusing on intestinal microbiomes as well as the potential functions of the gut microbiota and its metabolites in the development of hypertension.
Collapse
Affiliation(s)
- Felix Oladele Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Abimbola Rafiat Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Babatunde Oluwafemi Adetuyi
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
3
|
Liu TH, Chen MH, Zhang CY, Xie T, Zhang S, Hao H, Bai ZY, Xue YZ, Wang JW, Xiao Y, Wei H, Chen LG. Hypertension inhibition by Dubosiella newyorkensis via reducing pentosidine synthesis. NPJ Biofilms Microbiomes 2025; 11:34. [PMID: 39987250 PMCID: PMC11846869 DOI: 10.1038/s41522-025-00645-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 12/31/2024] [Indexed: 02/24/2025] Open
Abstract
Gut dysbiosis has been associated with hypertension. Herein, we aimed to discover the potential association between gut microbiota and high-salt diet (HSD) induced endothelial dysfunction in conventional hypertensive mice. Dubosiella newyorkensis was found highly sensitive to salt in HSD-induced hypertension. The salt-sensitive nature of Dubosiella newyorkensis was confirmed by bacteria culture in vitro. Oral Dubosiella newyorkensis in HSD-induced hypertensive mice decreased blood pressure, inhibited activation of vascular endothelium, attenuated inflammation and alleviated intestinal vascular barrier injury. Similar effects of Dubosiella newyorkensis were observed in germ-free mice. Interestingly, serum pentosidine was found to function as a biomarker for Dubosiella newyorkensis in response to HSD in both metabolic modes. Supplement of pentosidine, deteriorated hypertension and vascular endothelial damage. Differential genes enriched in the glycerophospholipid metabolism were markedly altered in cultured bacteria. Our study has identified Dubosiella newyorkensis as a new salt-sensitive gut microbe that inhibits pentosidine production thereby alleviating hypertension.
Collapse
Affiliation(s)
- Tian-Hao Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, China
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Ming-Hao Chen
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Chen-Yang Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ting Xie
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Sitong Zhang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Haining Hao
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Zhen-Yu Bai
- College of Chinese Medicine, Jinan University, Guangzhou, China
| | - Yu-Zheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Ya Xiao
- College of Chinese Medicine, Jinan University, Guangzhou, China.
| | - Hong Wei
- Yu-Yue Pathology Scientific Research Center, Jinfeng Laboratory, Chongqing, China.
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Li-Guo Chen
- College of Chinese Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
4
|
Zhou T, Wang Z, Lv X, Guo M, Zhang N, Liu L, Geng L, Shao J, Zhang K, Gao M, Mao A, Zhu Y, Yu F, Feng L, Wang X, Zhai Q, Chen W, Ma X. Targeting gut S. aureofaciens Tü117 serves as a new potential therapeutic intervention for the prevention and treatment of hypertension. Cell Metab 2025; 37:496-513.e11. [PMID: 39908987 DOI: 10.1016/j.cmet.2025.01.004] [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: 01/03/2024] [Revised: 09/13/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025]
Abstract
Currently, the regulation of specific gut microbial metabolism for the development and/or treatment of hypertension remains largely unexplored. Here, we show that α-lipomycin, produced by Streptomyces aureofaciens (S. aureofaciens) Tü117, is upregulated in the serum of high-salt diet (HSD) mice and patients with essential hypertension. α-lipomycin causes vasodilation impairment involving transient receptor potential vanilloid 4 (TRPV4)-mediated nitric oxide and endothelium-derived hyperpolarizing factor pathways in mice. We also find that Lactobacillus plantarum (L. plantarum) CCFM639 attenuates the increase in blood pressure (BP) potentially through inhibiting the proliferation of S. aureofaciens Tü117 in mice. An exploratory intervention trial indicates that L. plantarum CCFM639 supplementation reduces BPs in subjects newly diagnosed with pre-hypertension or stage 1 hypertension without antihypertensive medication. Our findings provide evidence for a role of S. aureofaciens Tü117-associated α-lipomycin elevation in the pathogenesis of HSD-induced hypertension, highlighting that targeting gut bacteria serves as a new therapeutic intervention for hypertension.
Collapse
Affiliation(s)
- Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Zhiwei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Xiaowang Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Mengting Guo
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ning Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Liangju Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Jing Shao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Mengru Gao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Yifei Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Xiaoyan Wang
- Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Qixiao Zhai
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Affiliated Hospital of Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
5
|
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
|
6
|
Li X, Cui J, Ding Z, Tian Z, Kong Y, Li L, Liu Y, Zhao W, Chen X, Guo H, Cui Z, Li X, Yuan J, Zhang H. Klebsiella pneumoniae-derived extracellular vesicles impair endothelial function by inhibiting SIRT1. Cell Commun Signal 2025; 23:21. [PMID: 39800699 PMCID: PMC11726972 DOI: 10.1186/s12964-024-02002-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The potential role of Klebsiella pneumoniae (K.pn) in hypertension development has been emphasized, although the specific mechanisms have not been well understood. Bacterial extracellular vesicles (BEVs) released by Gram-negative bacteria modulate host cell functions by delivering bacterial components to host cells. Endothelial dysfunction is an important early event in the pathogenesis of hypertension, yet the impact of K.pn-secreted EVs (K.pn EVs) on endothelial function remains unclear. This study aimed to investigate the effects of K.pn EVs on endothelial function and to elucidate the underlying mechanisms. METHODS K.pn EVs were purified from the bacterial suspension using ultracentrifugation and characterized by transmission electron microscopy nanoparticle tracking analysis, and EV marker expression. Endothelium-dependent relaxation was measured using a wire myograph after in vivo or ex vivo treatment with K.pn EVs. Superoxide anion production was measured by confocal microscopy and HUVEC senescence was assessed by SA-β-gal activity. SIRT1 overexpression or activator was utilized to investigate the underlying mechanisms. RESULTS Our data showed that K.pn significantly impaired acetylcholine-induced endothelium-dependent relaxation and increased superoxide anion production in endothelial cells in vivo. Similarly, in vivo and ex vivo studies showed that K.pn EVs caused significant endothelial dysfunction, endothelial provocation, and increased blood pressure. Further examination revealed that K.pn EVs reduced the levels of SIRT1 and p-eNOS and increased the levels of NOX2, COX-2, ET-1, and p53 in endothelial cells. Notably, overexpression or activation of SIRT1 attenuated the adverse effects and protein changes induced by K.pn EVs on endothelial cells. CONCLUSION This study reveals a novel role of K.pn EVs in endothelial dysfunction and dissects the relevant mechanism involved in this process, which will help to establish a comprehensive understanding of K.pn EVs in endothelial dysfunction and hypertension from a new scope.
Collapse
Affiliation(s)
- Xinxin Li
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Jinghua Cui
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China
| | - Zanbo Ding
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China
| | - Ziyan Tian
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China
| | - Yiming Kong
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China
| | - Linghai Li
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Yang Liu
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Wen Zhao
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Xueying Chen
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Han Guo
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Zhengshuo Cui
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Xinwei Li
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Jing Yuan
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China.
| | - Huina Zhang
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China.
| |
Collapse
|
7
|
Brosolo G, Da Porto A, Marcante S, Capilupi F, Bertin N, Vivarelli C, Bulfone L, Vacca A, Catena C, Sechi LA. The role for ω-3 polyunsaturated and short chain fatty acids in hypertension: An updated view on the interaction with gut microbiota. Eur J Pharmacol 2024; 985:177107. [PMID: 39515560 DOI: 10.1016/j.ejphar.2024.177107] [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: 06/01/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
As of 2024, arterial hypertension is still considered the leading modifiable cardiovascular risk factor and, due to high rates of undertreatment and poor blood pressure control, the major contributor to human morbidity and mortality. Development of new treatment options and better interventions in lifestyle correction have become a priority of experimental and clinical research. In the last decades, dietary supplementation of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) and generation of gut microbiota-derived short chain fatty acids (SCFAs) have surged as potential and promising interventions for hypertension and cardiovascular prevention. ω-3 PUFAs are considered "essential" fatty acids that can be obtained only from dietary sources. Although previous intervention trials were not consistent in reporting a significant benefit of ω-3 PUFAs, the recent REDUCE-IT trial has provided robust evidence in support of their role in cardiovascular prevention. Recent studies have also identified the intestinal microbiota as a potential player in the pathophysiology and progression of hypertension. Although this might occur through many pathways, generation of SCFAs that is highly dependent on dietary fiber intake is primarily involved, providing an additional target for the development of novel therapeutic strategies. For these reasons, some scientific societies currently recommend dietary supplementation of ω-3 PUFAs and fiber-containing foods in patients with hypertension. In this narrative review, we summarize the results of studies that examined the effects of ω-3 PUFAs and SCFAs on blood pressure, highlighting the mechanisms of action on the vascular system and their possible impact on hypertension, hypertension-related organ damage and, ultimately, cardiovascular outcomes.
Collapse
Affiliation(s)
- Gabriele Brosolo
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Andrea Da Porto
- Department of Medicine, University of Udine, 33100, Udine, Italy; Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Stefano Marcante
- Department of Medicine, University of Udine, 33100, Udine, Italy.
| | - Filippo Capilupi
- Department of Medicine, University of Udine, 33100, Udine, Italy.
| | - Nicole Bertin
- Department of Medicine, University of Udine, 33100, Udine, Italy; Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Cinzia Vivarelli
- Department of Medicine, University of Udine, 33100, Udine, Italy.
| | - Luca Bulfone
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Antonio Vacca
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Cristiana Catena
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy.
| | - Leonardo A Sechi
- Department of Medicine, University of Udine, 33100, Udine, Italy; European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100, Udine, Italy; Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100, Udine, Italy; Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100, Udine, Italy.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Eguchi S, Torimoto K, Adebiyi A, Kanthakumar P, Bomfim GF, Wenceslau CF, Dahlen SA, Osei-Owusu P. Milestone Papers on Signal Transduction Mechanisms of Hypertension and Its Complications. Hypertension 2024; 81:977-990. [PMID: 38372140 PMCID: PMC11023792 DOI: 10.1161/hypertensionaha.123.21365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
To celebrate 100 years of American Heart Association-supported cardiovascular disease research, this review article highlights milestone papers that have significantly contributed to the current understanding of the signaling mechanisms driving hypertension and associated cardiovascular disorders. This article also includes a few of the future research directions arising from these critical findings. To accomplish this important mission, 4 principal investigators gathered their efforts to cover distinct yet intricately related areas of signaling mechanisms pertaining to the pathogenesis of hypertension. The renin-angiotensin system, canonical and novel contractile and vasodilatory pathways in the resistance vasculature, vascular smooth muscle regulation by membrane channels, and noncanonical regulation of blood pressure and vascular function will be described and discussed as major subjects.
Collapse
Affiliation(s)
- Satoru Eguchi
- Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University
| | - Keiichi Torimoto
- Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University
| | - Adebowale Adebiyi
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, Missouri
- NextGen Precision Health, University of Missouri, Columbia, Missouri
| | - Praghalathan Kanthakumar
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, Missouri
- NextGen Precision Health, University of Missouri, Columbia, Missouri
| | - Gisele F. Bomfim
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine
| | - Camilla Ferreira Wenceslau
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine
| | - Shelby A. Dahlen
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University
| | - Patrick Osei-Owusu
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University
| |
Collapse
|
11
|
Longtine AG, Greenberg NT, Bernaldo de Quirós Y, Brunt VE. The gut microbiome as a modulator of arterial function and age-related arterial dysfunction. Am J Physiol Heart Circ Physiol 2024; 326:H986-H1005. [PMID: 38363212 PMCID: PMC11279790 DOI: 10.1152/ajpheart.00764.2023] [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] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 02/17/2024]
Abstract
The arterial system is integral to the proper function of all other organs and tissues. Arterial function is impaired with aging, and arterial dysfunction contributes to the development of numerous age-related diseases, including cardiovascular diseases. The gut microbiome has emerged as an important regulator of both normal host physiological function and impairments in function with aging. The purpose of this review is to summarize more recently published literature demonstrating the role of the gut microbiome in supporting normal arterial development and function and in modulating arterial dysfunction with aging in the absence of overt disease. The gut microbiome can be altered due to a variety of exposures, including physiological aging processes. We explore mechanisms by which the gut microbiome may contribute to age-related arterial dysfunction, with a focus on changes in various gut microbiome-related compounds in circulation. In addition, we discuss how modulating circulating levels of these compounds may be a viable therapeutic approach for improving artery function with aging. Finally, we identify and discuss various experimental considerations and research gaps/areas of future research.
Collapse
Affiliation(s)
- Abigail G Longtine
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nathan T Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Yara Bernaldo de Quirós
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
- Instituto Universitario de Sanidad Animal y Seguridad Alimentaria, Universidad de las Palmas de Gran Canaria, Las Palmas, Spain
| | - Vienna E Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
12
|
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
|
13
|
Gao C, Wei J, Lu C, Wang L, Dong D, Sun M. A new perspective in intestinal microecology: lifting the veil of exercise regulation of cardiometabolic diseases. Gut Microbes 2024; 16:2404141. [PMID: 39305272 PMCID: PMC11418258 DOI: 10.1080/19490976.2024.2404141] [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: 03/21/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Cardiometabolic diseases (CMDs), encompassing cardiovascular and metabolic dysfunctions, characterized by insulin resistance, dyslipidemia, hepatic steatosis, and inflammation, have been identified with boosting morbidity and mortality due to the dearth of efficacious therapeutic interventions. In recent years, studies have shown that variations in gut microbiota and its own metabolites can influence the occurrence of CMDs. Intriguingly, the composition and function of the gut microbiota are susceptible to exercise patterns, thus affecting inflammatory, immune, and metabolic responses within the host. In this review, we introduce the key mechanisms of intestinal microecology involved in the onset and development of CMDs, discuss the relationship between exercise and intestinal microecology, and then analyze the role of intestinal microecology in the beneficial effects of exercise on CMDs, aiming at elucidating the gut-heart axis mechanisms of exercise mediated protective effect on CMDs, building avenues for the application of exercise in the management of CMDs.
Collapse
Affiliation(s)
- Can Gao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, P. R. China
| | - Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, P. R. China
| | - Changxu Lu
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, P. R. China
| | - Lijie Wang
- Department of Cardiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, P. R. China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, P. R. China
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, P. R. China
| |
Collapse
|
14
|
Liu X, Fang H, Pan L, Zhang P, Lin H, Gao H, Ye C, Mao D, Luo Y. S-amlodipine induces liver inflammation and dysfunction through the alteration of intestinal microbiome in a rat model. Gut Microbes 2024; 16:2316923. [PMID: 38400721 PMCID: PMC10896145 DOI: 10.1080/19490976.2024.2316923] [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: 06/25/2023] [Accepted: 02/06/2024] [Indexed: 02/26/2024] Open
Abstract
S-amlodipine, a commonly prescribed antihypertensive agent, is widely used in clinical settings to treat hypertension. However, the potential adverse effects of long-term S-amlodipine treatment on the liver remain uncertain, given the cautionary recommendations from clinicians regarding its administration in individuals with impaired liver function. To address this, we conducted a study using an eight-week-old male rat model and administered a daily dose of 0.6 ~ 5 mg/kg of S-amlodipine for 7 weeks. Our findings demonstrated that 1.2 ~ 5 mg/kg of S-amlodipine treatment induced liver inflammation and associated dysfunction in rats, further in vitro experiments revealed that the observed liver inflammation and dysfunction were not attributable to direct effects of S-amlodipine on the liver. Metagenome sequencing analysis revealed that S-amlodipine treatment led to alterations in the gut microbiome of rats, with the bloom of E. coli (4.5 ~ 6.6-fold increase) and a decrease in A. muciniphila (1,613.4 ~ 2,000-fold decrease) and B. uniformis (20.6 ~ 202.7-fold decrease), subsequently causing an increase in the gut bacterial lipopolysaccharide (LPS) content (1.4 ~ 1.5-fold increase in feces). S-amlodipine treatment also induced damage to the intestinal barrier and increased intestinal permeability, as confirmed by elevated levels of fecal albumin; furthermore, the flux of gut bacterial LPS into the bloodstream through the portal vein resulted in an increase in serum LPS content (3.3 ~ 4-fold increase). LPS induces liver inflammation and subsequent dysfunction in rats by activating the TLR4 pathway. This study is the first to show that S-amlodipine induces liver inflammation and dysfunction by perturbing the rat gut microbiome. These results indicate the adverse effects of S-amlodipine on the liver and provide a rich understanding of the safety of long-term S-amlodipine administration.
Collapse
Affiliation(s)
- Xinxin Liu
- College of Environmental Sciences and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Hui Fang
- School of Medicine, Nankai University, Tianjin, China
| | - Liuzhu Pan
- School of Medicine, Nankai University, Tianjin, China
| | - Peng Zhang
- College of Environmental Sciences and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Huai Lin
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Huihui Gao
- School of Medicine, Nankai University, Tianjin, China
| | - Chaolin Ye
- School of Medicine, Nankai University, Tianjin, China
| | - Daqing Mao
- School of Medicine, Nankai University, Tianjin, China
| | - Yi Luo
- College of Environmental Sciences and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| |
Collapse
|
15
|
Li D, Chen F. Effects of Gut Microbiota on Hypertension and the Cardiovascular System. Nutrients 2023; 15:4633. [PMID: 37960287 PMCID: PMC10650547 DOI: 10.3390/nu15214633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiovascular diseases, which include hypertension and atherosclerosis, are a group of disorders that affect the heart and blood vessels [...].
Collapse
Affiliation(s)
| | - Fang Chen
- National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, College of Food Science and Nutritional Engineering, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China;
| |
Collapse
|
16
|
Yang Y, Karampoor S, Mirzaei R, Borozdkin L, Zhu P. The interplay between microbial metabolites and macrophages in cardiovascular diseases: A comprehensive review. Int Immunopharmacol 2023; 121:110546. [PMID: 37364331 DOI: 10.1016/j.intimp.2023.110546] [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: 05/10/2023] [Revised: 06/11/2023] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
The gut microbiome has emerged as a crucial player in developing and progressing cardiovascular diseases (CVDs). Recent studies have highlighted the role of microbial metabolites in modulating immune cell function and their impact on CVD. Macrophages, which have a significant function in the pathogenesis of CVD, are very vulnerable to the effects of microbial metabolites. Microbial metabolites, such as short-chain fatty acids (SCFAs) and trimethylamine-N-oxide (TMAO), have been linked to atherosclerosis and the regulation of immune functions. Butyrate has been demonstrated to reduce monocyte migration and inhibit monocyte attachment to injured endothelial cells, potentially contributing to the attenuation of the inflammatory response and the progression of atherosclerosis. On the other hand, TMAO, another compound generated by gut bacteria, has been linked to atherosclerosis due to its impact on lipid metabolism and the accumulation of cholesterol in macrophages. Indole-3-propionic acid, a tryptophan metabolite produced solely by microbes, has been found to promote the development of atherosclerosis by stimulating macrophage reverse cholesterol transport (RCT) and raising the expression of ABCA1. This review comprehensively discusses how various microbiota-produced metabolites affect macrophage polarization, inflammation, and foam cell formation in CVD. We also highlight the mechanisms underlying these effects and the potential therapeutic applications of targeting microbial metabolites in treating CVD.
Collapse
Affiliation(s)
- Yongzheng Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Leonid Borozdkin
- Department of Maxillofacial Surgery, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510100, China.
| |
Collapse
|
17
|
Chakraborty S, Lulla A, Cheng X, Yeo JY, Mandal J, Yang T, Mei X, Saha P, Golonka RM, Yeoh BS, Mell B, Jia W, Putluri V, Piyarathna DWB, Putluri N, Sreekumar A, Meyer K, Vijay-Kumar M, Joe B. Conjugated bile acids are nutritionally re-programmable antihypertensive metabolites. J Hypertens 2023; 41:979-994. [PMID: 37071431 PMCID: PMC10158603 DOI: 10.1097/hjh.0000000000003423] [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: 11/13/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Hypertension is the largest risk factor affecting global mortality. Despite available medications, uncontrolled hypertension is on the rise, whereby there is an urgent need to develop novel and sustainable therapeutics. Because gut microbiota is now recognized as an important entity in blood pressure regulation, one such new avenue is to target the gut-liver axis wherein metabolites are transacted via host-microbiota interactions. Knowledge on which metabolites within the gut-liver axis regulate blood pressure is largely unknown. METHOD To address this, we analyzed bile acid profiles of human, hypertensive and germ-free rat models and report that conjugated bile acids are inversely correlated with blood pressure in humans and rats. RESULTS Notably intervening with taurine or tauro-cholic acid rescued bile acid conjugation and reduced blood pressure in hypertensive rats. Subsequently, untargeted metabolomics uncovered altered energy metabolism following conjugation of bile acids as a mechanism alleviating high blood pressure. CONCLUSION Together this work reveals conjugated bile acids as nutritionally re-programmable anti-hypertensive metabolites.
Collapse
Affiliation(s)
- Saroj Chakraborty
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Anju Lulla
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - Xi Cheng
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Ji-Youn Yeo
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Juthika Mandal
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Tao Yang
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Xue Mei
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Piu Saha
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Rachel M. Golonka
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Beng San Yeoh
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Blair Mell
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Wei Jia
- University of Hawaii Cancer Center, Honolulu, Hawaii
| | | | | | - Nagireddy Putluri
- Dan L. Duncan Cancer Center, Advanced Technology Core
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Arun Sreekumar
- Dan L. Duncan Cancer Center, Advanced Technology Core
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Katie Meyer
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina,USA
| | - Matam Vijay-Kumar
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Bina Joe
- Program in Physiological Genomics, Microbiome Consortium and Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
18
|
Aryal S, Manandhar I, Mei X, Yeoh BS, Tummala R, Saha P, Osman I, Zubcevic J, Durgan DJ, Vijay-Kumar M, Joe B. Combating hypertension beyond genome-wide association studies: Microbiome and artificial intelligence as opportunities for precision medicine. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e26. [PMID: 38550938 PMCID: PMC10953772 DOI: 10.1017/pcm.2023.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 11/03/2024]
Abstract
The single largest contributor to human mortality is cardiovascular disease, the top risk factor for which is hypertension (HTN). The last two decades have placed much emphasis on the identification of genetic factors contributing to HTN. As a result, over 1,500 genetic alleles have been associated with human HTN. Mapping studies using genetic models of HTN have yielded hundreds of blood pressure (BP) loci but their individual effects on BP are minor, which limits opportunities to target them in the clinic. The value of collecting genome-wide association data is evident in ongoing research, which is beginning to utilize these data at individual-level genetic disparities combined with artificial intelligence (AI) strategies to develop a polygenic risk score (PRS) for the prediction of HTN. However, PRS alone may or may not be sufficient to account for the incidence and progression of HTN because genetics is responsible for <30% of the risk factors influencing the etiology of HTN pathogenesis. Therefore, integrating data from other nongenetic factors influencing BP regulation will be important to enhance the power of PRS. One such factor is the composition of gut microbiota, which constitute a more recently discovered important contributor to HTN. Studies to-date have clearly demonstrated that the transition from normal BP homeostasis to a state of elevated BP is linked to compositional changes in gut microbiota and its interaction with the host. Here, we first document evidence from studies on gut dysbiosis in animal models and patients with HTN followed by a discussion on the prospects of using microbiota data to develop a metagenomic risk score (MRS) for HTN to be combined with PRS and a clinical risk score (CRS). Finally, we propose that integrating AI to learn from the combined PRS, MRS and CRS may further enhance predictive power for the susceptibility and progression of HTN.
Collapse
Affiliation(s)
- Sachin Aryal
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ishan Manandhar
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Xue Mei
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Beng S. Yeoh
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ramakumar Tummala
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Piu Saha
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Islam Osman
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jasenka Zubcevic
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - David J. Durgan
- Integrative Physiology & Anesthesiology, Baylor College of Medicine, Houston, TX, USA
| | - Matam Vijay-Kumar
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
19
|
Grüner N, Ortlepp AL, Mattner J. Pivotal Role of Intestinal Microbiota and Intraluminal Metabolites for the Maintenance of Gut-Bone Physiology. Int J Mol Sci 2023; 24:5161. [PMID: 36982235 PMCID: PMC10048911 DOI: 10.3390/ijms24065161] [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: 01/29/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Intestinal microbiota, and their mutual interactions with host tissues, are pivotal for the maintenance of organ physiology. Indeed, intraluminal signals influence adjacent and even distal tissues. Consequently, disruptions in the composition or functions of microbiota and subsequent altered host-microbiota interactions disturb the homeostasis of multiple organ systems, including the bone. Thus, gut microbiota can influence bone mass and physiology, as well as postnatal skeletal evolution. Alterations in nutrient or electrolyte absorption, metabolism, or immune functions, due to the translocation of microbial antigens or metabolites across intestinal barriers, affect bone tissues, as well. Intestinal microbiota can directly and indirectly alter bone density and bone remodeling. Intestinal dysbiosis and a subsequently disturbed gut-bone axis are characteristic for patients with inflammatory bowel disease (IBD) who suffer from various intestinal symptoms and multiple bone-related complications, such as arthritis or osteoporosis. Immune cells affecting the joints are presumably even primed in the gut. Furthermore, intestinal dysbiosis impairs hormone metabolism and electrolyte balance. On the other hand, less is known about the impact of bone metabolism on gut physiology. In this review, we summarized current knowledge of gut microbiota, metabolites and microbiota-primed immune cells in IBD and bone-related complications.
Collapse
Affiliation(s)
- Niklas Grüner
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Lisa Ortlepp
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
20
|
Gumz ML, Shimbo D, Abdalla M, Balijepalli RC, Benedict C, Chen Y, Earnest DJ, Gamble KL, Garrison SR, Gong MC, Hogenesch JB, Hong Y, Ivy JR, Joe B, Laposky AD, Liang M, MacLaughlin EJ, Martino TA, Pollock DM, Redline S, Rogers A, Dan Rudic R, Schernhammer ES, Stergiou GS, St-Onge MP, Wang X, Wright J, Oh YS. Toward Precision Medicine: Circadian Rhythm of Blood Pressure and Chronotherapy for Hypertension - 2021 NHLBI Workshop Report. Hypertension 2023; 80:503-522. [PMID: 36448463 PMCID: PMC9931676 DOI: 10.1161/hypertensionaha.122.19372] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Healthy individuals exhibit blood pressure variation over a 24-hour period with higher blood pressure during wakefulness and lower blood pressure during sleep. Loss or disruption of the blood pressure circadian rhythm has been linked to adverse health outcomes, for example, cardiovascular disease, dementia, and chronic kidney disease. However, the current diagnostic and therapeutic approaches lack sufficient attention to the circadian rhythmicity of blood pressure. Sleep patterns, hormone release, eating habits, digestion, body temperature, renal and cardiovascular function, and other important host functions as well as gut microbiota exhibit circadian rhythms, and influence circadian rhythms of blood pressure. Potential benefits of nonpharmacologic interventions such as meal timing, and pharmacologic chronotherapeutic interventions, such as the bedtime administration of antihypertensive medications, have recently been suggested in some studies. However, the mechanisms underlying circadian rhythm-mediated blood pressure regulation and the efficacy of chronotherapy in hypertension remain unclear. This review summarizes the results of the National Heart, Lung, and Blood Institute workshop convened on October 27 to 29, 2021 to assess knowledge gaps and research opportunities in the study of circadian rhythm of blood pressure and chronotherapy for hypertension.
Collapse
Affiliation(s)
- Michelle L Gumz
- Department of Physiology and Aging; Center for Integrative Cardiovascular and Metabolic Disease, Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, FL (M.L.G.)
| | - Daichi Shimbo
- Department of Medicine, The Columbia Hypertension Center, Columbia University Irving Medical Center, New York, NY (D.S.)
| | - Marwah Abdalla
- Department of Medicine, Center for Behavioral Cardiovascular Health, Columbia University Irving Medical Center, New York, NY (M.A.)
| | - Ravi C Balijepalli
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (R.C.B., Y.H., J.W., Y.S.O.)
| | - Christian Benedict
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Sweden (C.B.)
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, and Research Department, Birmingham VA Medical Center, AL (Y.C.)
| | - David J Earnest
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University, Bryan, TX (D.J.E.)
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, AL (K.L.G.)
| | - Scott R Garrison
- Department of Family Medicine, University of Alberta, Canada (S.R.G.)
| | - Ming C Gong
- Department of Physiology, University of Kentucky, Lexington, KY (M.C.G.)
| | | | - Yuling Hong
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (R.C.B., Y.H., J.W., Y.S.O.)
| | - Jessica R Ivy
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, United Kingdom (J.R.I.)
| | - Bina Joe
- Department of Physiology and Pharmacology and Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (B.J.)
| | - Aaron D Laposky
- National Center on Sleep Disorders Research, Division of Lung Diseases, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (A.D.L.)
| | - Mingyu Liang
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI (M.L.)
| | - Eric J MacLaughlin
- Department of Pharmacy Practice, Texas Tech University Health Sciences Center, Amarillo, TX (E.J.M.)
| | - Tami A Martino
- Center for Cardiovascular Investigations, Department of Biomedical Sciences, University of Guelph, Ontario, Canada (T.A.M.)
| | - David M Pollock
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, AL (D.M.P.)
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.R.)
| | - Amy Rogers
- Division of Molecular and Clinical Medicine, University of Dundee, United Kingdom (A.R.)
| | - R Dan Rudic
- Department of Pharmacology and Toxicology, Augusta University, GA (R.D.R.)
| | - Eva S Schernhammer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (E.S.S.)
| | - George S Stergiou
- Hypertension Center, STRIDE-7, National and Kapodistrian University of Athens, School of Medicine, Third Department of Medicine, Sotiria Hospital, Athens, Greece (G.S.S.)
| | - Marie-Pierre St-Onge
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center' New York, NY (M.-P.S.-O.)
| | - Xiaoling Wang
- Georgia Prevention Institute, Department of Medicine, Augusta University, GA (X.W.)
| | - Jacqueline Wright
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (R.C.B., Y.H., J.W., Y.S.O.)
| | - Young S Oh
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD (R.C.B., Y.H., J.W., Y.S.O.)
| |
Collapse
|
21
|
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
|
22
|
Campbell C, Kandalgaonkar MR, Golonka RM, Yeoh BS, Vijay-Kumar M, Saha P. Crosstalk between Gut Microbiota and Host Immunity: Impact on Inflammation and Immunotherapy. Biomedicines 2023; 11:294. [PMID: 36830830 PMCID: PMC9953403 DOI: 10.3390/biomedicines11020294] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Gut microbes and their metabolites are actively involved in the development and regulation of host immunity, which can influence disease susceptibility. Herein, we review the most recent research advancements in the gut microbiota-immune axis. We discuss in detail how the gut microbiota is a tipping point for neonatal immune development as indicated by newly uncovered phenomenon, such as maternal imprinting, in utero intestinal metabolome, and weaning reaction. We describe how the gut microbiota shapes both innate and adaptive immunity with emphasis on the metabolites short-chain fatty acids and secondary bile acids. We also comprehensively delineate how disruption in the microbiota-immune axis results in immune-mediated diseases, such as gastrointestinal infections, inflammatory bowel diseases, cardiometabolic disorders (e.g., cardiovascular diseases, diabetes, and hypertension), autoimmunity (e.g., rheumatoid arthritis), hypersensitivity (e.g., asthma and allergies), psychological disorders (e.g., anxiety), and cancer (e.g., colorectal and hepatic). We further encompass the role of fecal microbiota transplantation, probiotics, prebiotics, and dietary polyphenols in reshaping the gut microbiota and their therapeutic potential. Continuing, we examine how the gut microbiota modulates immune therapies, including immune checkpoint inhibitors, JAK inhibitors, and anti-TNF therapies. We lastly mention the current challenges in metagenomics, germ-free models, and microbiota recapitulation to a achieve fundamental understanding for how gut microbiota regulates immunity. Altogether, this review proposes improving immunotherapy efficacy from the perspective of microbiome-targeted interventions.
Collapse
Affiliation(s)
- Connor Campbell
- Department of Physiology & Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Mrunmayee R. Kandalgaonkar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Rachel M. Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Piu Saha
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
23
|
Badran M, Khalyfa A, Ericsson AC, Puech C, McAdams Z, Bender SB, Gozal D. Gut microbiota mediate vascular dysfunction in a murine model of sleep apnoea: effect of probiotics. Eur Respir J 2023; 61:2200002. [PMID: 36028255 PMCID: PMC11556237 DOI: 10.1183/13993003.00002-2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 08/10/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Obstructive sleep apnoea (OSA) is a chronic prevalent condition characterised by intermittent hypoxia (IH), and is associated with endothelial dysfunction and coronary artery disease (CAD). OSA can induce major changes in gut microbiome diversity and composition, which in turn may induce the emergence of OSA-associated morbidities. However, the causal effects of IH-induced gut microbiome changes on the vasculature remain unexplored. Our objective was to assess if vascular dysfunction induced by IH is mediated through gut microbiome changes. METHODS Faecal microbiota transplantation (FMT) was conducted on C57BL/6J naïve mice for 6 weeks to receive either IH or room air (RA) faecal slurry with or without probiotics (VSL#3). In addition to 16S rRNA amplicon sequencing of their gut microbiome, FMT recipients underwent arterial blood pressure and coronary artery and aorta function testing, and their trimethylamine N-oxide (TMAO) and plasma acetate levels were determined. Finally, C57BL/6J mice were exposed to IH, IH treated with VSL#3 or RA for 6 weeks, and arterial blood pressure and coronary artery function assessed. RESULTS Gut microbiome taxonomic profiles correctly segregated IH from RA in FMT mice and the normalising effect of probiotics emerged. Furthermore, IH-FMT mice exhibited increased arterial blood pressure and TMAO levels, and impairments in aortic and coronary artery function (p<0.05) that were abrogated by probiotic administration. Lastly, treatment with VSL#3 under IH conditions did not attenuate elevations in arterial blood pressure or CAD. CONCLUSIONS Gut microbiome alterations induced by chronic IH underlie, at least partially, the typical cardiovascular disturbances of sleep apnoea and can be mitigated by concurrent administration of probiotics.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Abdelnaby Khalyfa
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Aaron C Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- University of Missouri Metagenomics Center, University of Missouri, Columbia, MO, USA
| | - Clementine Puech
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Zachary McAdams
- Department of Molecular Microbiology and Immunology, Molecular Pathogenesis and Therapeutics Program, University of Missouri, Columbia, MO, USA
| | - Shawn B Bender
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans Hospital, University of Missouri, Columbia, MO, USA
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
24
|
Gokula V, Terrero D, Joe B. Six Decades of History of Hypertension Research at the University of Toledo: Highlighting Pioneering Contributions in Biochemistry, Genetics, and Host-Microbiota Interactions. Curr Hypertens Rep 2022; 24:669-685. [PMID: 36301488 PMCID: PMC9708772 DOI: 10.1007/s11906-022-01226-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW The study aims to capture the history and lineage of hypertension researchers from the University of Toledo in Ohio and showcase their collective scientific contributions dating from their initial discoveries of the physiology of adrenal and renal systems and genetics regulating blood pressure (BP) to its more contemporary contributions including microbiota and metabolomic links to BP regulation. RECENT FINDINGS The University of Toledo College of Medicine and Life Sciences (UTCOMLS), previously known as the Medical College of Ohio, has contributed significantly to our understanding of the etiology of hypertension. Two of the scientists, Patrick Mulrow and John Rapp from UTCOMLS, have been recognized with the highest honor, the Excellence in Hypertension award from the American Heart Association for their pioneering work on the physiology and genetics of hypertension, respectively. More recently, Bina Joe has continued their legacy in the basic sciences by uncovering previously unknown novel links between microbiota and metabolites to the etiology of hypertension, work that has been recognized by the American Heart Association with multiple awards. On the clinical research front, Christopher Cooper and colleagues lead the CORAL trials and contributed importantly to the investigations on renal artery stenosis treatment paradigms. Hypertension research at this institution has not only provided these pioneering insights, but also grown careers of scientists as leaders in academia as University Presidents and Deans of Medical Schools. Through the last decade, the university has expanded its commitment to Hypertension research as evident through the development of the Center for Hypertension and Precision Medicine led by Bina Joe as its founding Director. Hypertension being the top risk factor for cardiovascular diseases, which is the leading cause of human mortality, is an important area of research in multiple international universities. The UTCOMLS is one such university which, for the last 6 decades, has made significant contributions to our current understanding of hypertension. This review is a synthesis of this rich history. Additionally, it also serves as a collection of audio archives by more recent faculty who are also prominent leaders in the field of hypertension research, including John Rapp, Bina Joe, and Christopher Cooper, which are cataloged at Interviews .
Collapse
Affiliation(s)
- Veda Gokula
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo College of Medicine and Life Sciences, Block Health Science Building, 3000 Arlington Ave, Toledo, OH, 43614-2598, USA
| | - David Terrero
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy, University of Toledo, Toledo, OH, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo College of Medicine and Life Sciences, Block Health Science Building, 3000 Arlington Ave, Toledo, OH, 43614-2598, USA.
| |
Collapse
|
25
|
Saha P, Mell B, Golonka RM, Bovilla VR, Abokor AA, Mei X, Yeoh BS, Doris PA, Gewirtz AT, Joe B, Vijay-Kumar M. Selective IgA Deficiency in Spontaneously Hypertensive Rats With Gut Dysbiosis. Hypertension 2022; 79:2239-2249. [PMID: 35950503 PMCID: PMC9458624 DOI: 10.1161/hypertensionaha.122.19307] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/07/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND The spontaneously hypertensive rat (SHR) is extensively used to study hypertension. Gut microbiota dysbiosis is a notable feature in SHR for reasons unknown. Immunoglobulin A (IgA) is a major host factor required for gut microbiota homeostasis. We hypothesized that inadequate IgA contributes to gut microbiota dysbiosis in SHR. METHODS IgA was measured in feces, cecum, serum, liver, gut-associated lymphoid tissue, and milk from SHR and Wistar Kyoto rats. IgA regulatory factors like IgM, IgG, and pIgR (polymeric immunoglobulin receptor) were analyzed. IgA and IgG antibodies and blood pressure (BP) were measured before and after administrating a bacterial antigen (ie, flagellin). RESULTS Compared with Wistar Kyoto rats, SHR displayed remarkably near-deficient IgA levels accompanied by compensatory increases in serum IgM and IgG and gut-liver pIgR expression. Inadequate milk IgA in SHR emphasized this immune defect stemmed from the neonatal stage. Reduced IgA+ B cells in circulation and Peyer patches indicated a possible reason for the lower IgA in SHR. Noteworthy, a genetic insufficiency was unlikely because administering flagellin to SHR induced anti-flagellin IgA antibodies. This immune response surprisingly accelerated hypertension development in SHR, suggesting IgA quiescence may help maintain lower BP. CONCLUSIONS This study is the first to reveal IgA deficiency in SHR as one host factor associated with gut microbiota dysbiosis and invigorates future research to determine the pathophysiological role of IgA in hypertension.
Collapse
Affiliation(s)
- Piu Saha
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Blair Mell
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Rachel M. Golonka
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Venugopal R. Bovilla
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Ahmed A. Abokor
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Xue Mei
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Peter A. Doris
- Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Andrew T. Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Bina Joe
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Matam Vijay-Kumar
- UT Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
26
|
Martín Giménez VM, Rukavina Mikusic NL, Lee HJ, García Menéndez S, Choi MR, Manucha W. Physiopathological mechanisms involved in the development of hypertension associated with gut dysbiosis and the effect of nutritional/pharmacological interventions. Biochem Pharmacol 2022; 204:115213. [PMID: 35985404 DOI: 10.1016/j.bcp.2022.115213] [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: 07/04/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/27/2022]
Abstract
The gut microbiota dysbiosis represents a triggering factor for cardiovascular diseases, including hypertension. In addition to the harmful impact caused by hypertension on different target organs, gut dysbiosis is capable of causing direct damage to critical organs such as the brain, heart, blood vessels, and kidneys. In this sense, it should be noted that pharmacological and nutritional interventions may influence gut microbiota composition, either inducing or preventing the development of hypertension. Some of the most important nutritional interventions at this level are represented by pro-, pre-, post- and/or syn-biotics, as well as polysaccharides, polyunsaturated fatty acids ω-3, polyphenols and fiber contained in different foods. Meanwhile, certain natural and synthetic active pharmaceutical ingredients, including antibiotics, antihypertensive and immunosuppressive drugs, vegetable extracts and vitamins, may also have a key role in the modulation of both gut microbiota and cardiovascular health. Additionally, gut microbiota may influence drugs and food-derived bioactive compounds metabolism, positively or negatively affecting their biological behavior facing established hypertension. The understanding of the complex interactions between gut microbiome and drug/food response results of great importance to developing improved pharmacological therapies for hypertension prevention and treatment. The purpose of this review is to critically outline the most relevant and recent findings on cardiovascular, renal and brain physiopathological mechanisms involved in the development of hypertension associated with changes in gut microbiota, besides the nutritional and pharmacological interventions potentially valuable for the prevention and treatment of this prevalent pathology. Finally, harmful food/drug interventions on gut microbiota are also described.
Collapse
Affiliation(s)
- Virna Margarita Martín Giménez
- Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, Universidad Católica de Cuyo, Sede San Juan, Argentina
| | - Natalia Lucía Rukavina Mikusic
- Universidad de Buenos Aires. CONICET. Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), Buenos Aires, Argentina; Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Ciencias Biológicas. Cátedra de Anatomía e Histología, Buenos Aires, Argentina
| | - Hyun Jin Lee
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Ciencias Biológicas. Cátedra de Anatomía e Histología, Buenos Aires, Argentina
| | - Sebastián García Menéndez
- Laboratorio de Farmacología Experimental Básica y Traslacional. Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina; Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Argentina
| | - Marcelo Roberto Choi
- Universidad de Buenos Aires. CONICET. Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), Buenos Aires, Argentina; Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Ciencias Biológicas. Cátedra de Anatomía e Histología, Buenos Aires, Argentina
| | - Walter Manucha
- Laboratorio de Farmacología Experimental Básica y Traslacional. Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina; Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Argentina.
| |
Collapse
|
27
|
Xu J, Moore BN, Pluznick JL. Short-Chain Fatty Acid Receptors and Blood Pressure Regulation: Council on Hypertension Mid-Career Award for Research Excellence 2021. Hypertension 2022; 79:2127-2137. [PMID: 35912645 PMCID: PMC9458621 DOI: 10.1161/hypertensionaha.122.18558] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The gut microbiome influences host physiology and pathophysiology through several pathways, one of which is microbial production of chemical metabolites which interact with host signaling pathways. Short-chain fatty acids (SCFAs) are a class of gut microbial metabolites known to activate multiple signaling pathways in the host. Growing evidence indicates that the gut microbiome is linked to blood pressure, that SCFAs modulate blood pressure regulation, and that delivery of exogenous SCFAs lowers blood pressure. Given that hypertension is a key risk factor for cardiovascular disease, the examination of novel contributors to blood pressure regulation has the potential to lead to novel approaches or treatments. Thus, this review will discuss SCFAs with a focus on their host G protein-coupled receptors including GPR41 (G protein-coupled receptor 41), GPR43, and GPR109A, as well as OLFR78 (olfactory receptor 78) and OLFR558. This includes a discussion of the ligand profiles, G protein coupling, and tissue distribution of each receptor. We will also review phenotypes relevant to blood pressure regulation which have been reported to date for Gpr41, Gpr43, Gpr109a, and Olfr78 knockout mice. In addition, we will consider how SCFA signaling influences physiology at baseline, and, how SCFA signaling may contribute to blood pressure regulation in settings of hypertension. In sum, this review will integrate current knowledge regarding how SCFAs and their receptors regulate blood pressure.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Brittni N. Moore
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jennifer L. Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
28
|
Huang YH, Tain YL, Hsu CN. Maternal Supplementation of Probiotics, Prebiotics or Postbiotics to Prevent Offspring Metabolic Syndrome: The Gap between Preclinical Results and Clinical Translation. Int J Mol Sci 2022; 23:10173. [PMID: 36077575 PMCID: PMC9456151 DOI: 10.3390/ijms231710173] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/21/2022] Open
Abstract
Metabolic syndrome (MetS) is an extremely prevalent complex trait and it can originate in early life. This concept is now being termed the developmental origins of health and disease (DOHaD). Increasing evidence supports that disturbance of gut microbiota influences various risk factors of MetS. The DOHaD theory provides an innovative strategy to prevent MetS through early intervention (i.e., reprogramming). In this review, we summarize the existing literature that supports how environmental cues induced MetS of developmental origins and the interplay between gut microbiota and other fundamental underlying mechanisms. We also present an overview of experimental animal models addressing implementation of gut microbiota-targeted reprogramming interventions to avert the programming of MetS. Even with growing evidence from animal studies supporting the uses of gut microbiota-targeted therapies start before birth to protect against MetS of developmental origins, their effects on pregnant women are still unknown and these results require further clinical translation.
Collapse
Affiliation(s)
- Ying-Hua Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
29
|
Li Z, Liu K, Zhao J, Yang L, Chen G, Liu A, Wang Q, Wang S, Li X, Cao H, Tao F, Zhang D. Antibiotics in elderly Chinese population and their relations with hypertension and pulse pressure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:67026-67045. [PMID: 35513617 DOI: 10.1007/s11356-022-20613-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 04/30/2022] [Indexed: 06/14/2023]
Abstract
Although antibiotic exposure in the general population has been well documented by a biomonitoring approach, epidemiologic data on the relationships between urinary antibiotic burden in the elderly with blood pressure (BP) are still lacking. The current study revealed thirty-four antibiotics in urine specimens from 990 elderly patients in Lu'an City, China, with detection frequencies ranging from 0.2 to 35.5%. Among the elderly, the prevalence of hypertension was 72.0%, and 12 antibiotics were detected in more than 10% of individuals with hypertension. The elderly with hypertension had the maximum daily exposure (5450.45 μg/kg/day) to fluoroquinolones (FQs). Multiple linear regression analyses revealed significant associations of BP and pulse pressure (PP) with exposure to specific antibiotics. The estimated β values (95% confidence interval) of associations with systolic blood pressure (SBP) in the right arm were 4.42 (1.15, 7.69) for FQs, 4.26 (0.52, 8.01) for the preferred as human antibiotics (PHAs), and 3.48 (0.20, 6.77) for the mixtures (FQs + tetracyclines [TCs] (tertile 3 vs. tertile 1)), respectively. Increased concentrations of TCs were associated with decreased diastolic BP (DBP; tertile 3: -1.75 [-3.39, -0.12]) for the right arm. Higher levels of FQs (tertile 3: 4.28 [1.02, 7.54]), PHAs (tertile 3: 4.25 [0.49, 8.01]), and FQs + TCs (tertile 3: 3.99 [0.71, 7.26]) were associated with increased SBP, and an increase in DBP for FQs (tertile 3: 1.82 [0.22, 3.42]) was shown in the left arm. Also, higher urinary concentrations of FQs (tertile 3: 3.18 [0.53, 5.82]), PHAs (tertile 3: 3.42 [0.40, 6.45]), and FQs + TCs (tertile 3: 3.06 [0.40, 5.72]) were related to increased PP, whereas a decline in PP for TCs (tertile 2: -2.93 [-5.60, -0.25]) in the right arm. And increased concentrations of penicillin V (tertile 3: 5.31 [1.53, 9.10]) and FQs + TCs (tertile 3: 2.84 [0.19, 5.49]) were related to higher PP in the left arm. By utilizing restricted cubic splines, our current study revealed a potential nonlinear dose-response association between FQ exposure and hypertension risk. In conclusion, this investigation is the first to present antibiotic exposure using a biomonitoring approach, and informs understanding of impacts of antibiotic residues, as emerging hazardous pollutants, on the hypertension risk in the elderly.
Collapse
Affiliation(s)
- Zhenkun Li
- School of Health Management, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kaiyong Liu
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jianing Zhao
- The Fourth Affiliated Hospital of Anhui Medical University, Huaihai Road, Hefei, 230012, Anhui, China
| | - Linsheng Yang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Guimei Chen
- School of Health Management, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Annuo Liu
- School of Nursing, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Qunan Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Sufang Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xiude Li
- Lu'an Center of Disease Control and Prevention, Lu'an, 237000, Anhui, China
| | - Hongjuan Cao
- Lu'an Center of Disease Control and Prevention, Lu'an, 237000, Anhui, China
| | - Fangbiao Tao
- School of Health Management, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Dongmei Zhang
- School of Health Management, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
30
|
Yang T, Mei X, Tackie-Yarboi E, Akere MT, Kyoung J, Mell B, Yeo JY, Cheng X, Zubcevic J, Richards EM, Pepine CJ, Raizada MK, Schiefer IT, Joe B. Identification of a Gut Commensal That Compromises the Blood Pressure-Lowering Effect of Ester Angiotensin-Converting Enzyme Inhibitors. Hypertension 2022; 79:1591-1601. [PMID: 35538603 PMCID: PMC9278702 DOI: 10.1161/hypertensionaha.121.18711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background: Despite the availability of various classes of antihypertensive medications, a large proportion of hypertensive individuals remain resistant to treatments. The reason for what contributes to low efficacy of antihypertensive medications in these individuals is elusive. The knowledge that gut microbiota is involved in pathophysiology of hypertension and drug metabolism led us to hypothesize that gut microbiota catabolize antihypertensive medications and compromised their blood pressure (BP)-lowering effects. Methods and Results: To test this hypothesis, we examined the BP responses to a representative ACE (angiotensin-converting enzyme) inhibitor quinapril in spontaneously hypertensive rats (SHR) with or without antibiotics. BP-lowering effect of quinapril was more pronounced in the SHR+antibiotics, indicating that gut microbiota of SHR lowered the antihypertensive effect of quinapril. Depletion of gut microbiota in the SHR+antibiotics was associated with decreased gut microbial catabolism of quinapril as well as significant reduction in the bacterial genus Coprococcus. C. comes, an anaerobic species of Coprococcus, harbored esterase activity and catabolized the ester quinapril in vitro. Co-administration of quinapril with C. comes reduced the antihypertensive effect of quinapril in the SHR. Importantly, C. comes selectively reduced the antihypertensive effects of ester ramipril but not nonester lisinopril. Conclusions: Our study revealed a previously unrecognized mechanism by which human commensal C. comes catabolizes ester ACE inhibitors in the gut and lowers its antihypertensive effect.
Collapse
Affiliation(s)
- Tao Yang
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Xue Mei
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Ethel Tackie-Yarboi
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH.,Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH
| | - Millicent Tambari Akere
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH.,Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH
| | - Jun Kyoung
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Blair Mell
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Ji-Youn Yeo
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Xi Cheng
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| | - Elaine M Richards
- Department of Physiology and Functional Genomics (E.M.R., M.K.R.), University of Florida College of Medicine, Gainesville
| | - Carl J Pepine
- Division of Cardiovascular Medicine (C.J.P.), University of Florida College of Medicine, Gainesville
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics (E.M.R., M.K.R.), University of Florida College of Medicine, Gainesville
| | - Isaac T Schiefer
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH.,Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences (E.T.-Y., M.T.A., I.T.S.), University of Toledo, OH
| | - Bina Joe
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences (T.Y., X.M., J.K., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH.,UT Microbiome Consortium, Center for Hypertension and Precision Medicine (T.Y., X.M., B.M., J.-Y.Y., X.C., J.Z., B.J.), University of Toledo, OH
| |
Collapse
|
31
|
Avery EG, Bartolomaeus H, Rauch A, Chen CY, N'Diaye G, Löber U, Bartolomaeus TUP, Fritsche-Guenther R, Rodrigues AF, Yarritu A, Zhong C, Fei L, Tsvetkov D, Todiras M, Park JK, Markó L, Maifeld A, Patzak A, Bader M, Kempa S, Kirwan JA, Forslund SK, Müller DN, Wilck N. Quantifying the impact of gut microbiota on inflammation and hypertensive organ damage. Cardiovasc Res 2022:6651675. [PMID: 35904261 DOI: 10.1093/cvr/cvac121] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
AIMS Hypertension (HTN) can lead to heart and kidney damage. The gut microbiota has been linked to HTN, although it is difficult to estimate its significance due to the variety of other features known to influence HTN. In the present study, we used germ-free (GF) and colonized (COL) littermate mice to quantify the impact of microbial colonization on organ damage in HTN. METHODS AND RESULTS Four-week-old male GF C57BL/6J littermates were randomized to remain GF or receive microbial colonization. HTN was induced by subcutaneous infusion with angiotensin (Ang) II (1.44 mg/kg/d) and 1% NaCl in the drinking water; sham-treated mice served as control. Renal damage was exacerbated in GF mice, whereas cardiac damage was more comparable between COL and GF, suggesting that the kidney is more sensitive to microbial influence. Multivariate analysis revealed a larger effect of HTN in GF mice. Serum metabolomics demonstrated that the colonization status influences circulating metabolites relevant to HTN. Importantly, GF mice were deficient in anti-inflammatory fecal short-chain fatty acids (SCFA). Flow cytometry showed that the microbiome has an impact on the induction of anti-hypertensive myeloid-derived suppressor cells and pro-inflammatory Th17 cells in HTN. In vitro inducibility of Th17 cells was significantly higher for cells isolated from GF than conventionally raised mice. CONCLUSIONS Microbial colonization status of mice had potent effects on their phenotypic response to a hypertensive stimulus, and the kidney is a highly microbiota-susceptible target organ in HTN. The magnitude of the pathogenic response in GF mice underscores the role of the microbiome in mediating inflammation in HTN. TRANSLATION PERSPECTIVE To assess the potential of microbiota-targeted interventions to prevent organ damage in hypertension, an accurate quantification of microbial influence is necessary. We provide evidence that the development of hypertensive organ damage is dependent on colonization status and suggest that a healthy microbiota provides anti-hypertensive immune and metabolic signals to the host. In the absence of normal symbiotic host-microbiome interactions, hypertensive damage to the kidney in particular is exacerbated. We suggest that hypertensive patients experiencing perturbations to the microbiota, which are common in CVD, may be at a greater risk for target-organ damage than those with a healthy microbiome.
Collapse
Affiliation(s)
- Ellen G Avery
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Internal Intensive Care Medicine, Berlin, Germany
| | - Ariana Rauch
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Internal Intensive Care Medicine, Berlin, Germany
| | - Chia-Yu Chen
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gabriele N'Diaye
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Löber
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Theda U P Bartolomaeus
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Raphaela Fritsche-Guenther
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Metabolomics Platform, Berlin, Germany
| | - André F Rodrigues
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Alex Yarritu
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Internal Intensive Care Medicine, Berlin, Germany
| | - Cheng Zhong
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Lingyan Fei
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Dmitry Tsvetkov
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Geriatrics, University of Greifswald, University District Hospital Wolgast, Greifswald, Germany
| | - Mihail Todiras
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Nicolae Testemianu State University of Medicine and Pharmacy, Chisinau, Moldova
| | | | - Lajos Markó
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - András Maifeld
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Patzak
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Kempa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Integrative Proteomics and Metabolomics Platform, Berlin Institute for Medical Systems Biology BIMSB, Berlin, Germany
| | - Jennifer A Kirwan
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Metabolomics Platform, Berlin, Germany
| | - Sofia K Forslund
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Internal Intensive Care Medicine, Berlin, Germany
| |
Collapse
|
32
|
Yan D, Sun Y, Zhou X, Si W, Liu J, Li M, Wu M. Regulatory effect of gut microbes on blood pressure. Animal Model Exp Med 2022; 5:513-531. [PMID: 35880388 PMCID: PMC9773315 DOI: 10.1002/ame2.12233] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/25/2022] [Indexed: 12/30/2022] Open
Abstract
Hypertension is an important global public health issue because of its high morbidity as well as the increased risk of other diseases. Recent studies have indicated that the development of hypertension is related to the dysbiosis of the gut microbiota in both animals and humans. In this review, we outline the interaction between gut microbiota and hypertension, including gut microbial changes in hypertension, the effect of microbial dysbiosis on blood pressure (BP), indicators of gut microbial dysbiosis in hypertension, and the microbial genera that affect BP at the taxonomic level. For example, increases in Lactobacillus, Roseburia, Coprococcus, Akkermansia, and Bifidobacterium are associated with reduced BP, while increases in Streptococcus, Blautia, and Prevotella are associated with elevated BP. Furthermore, we describe the potential mechanisms involved in the regulation between gut microbiota and hypertension. Finally, we summarize the commonly used treatments of hypertension that are based on gut microbes, including fecal microbiota transfer, probiotics and prebiotics, antibiotics, and dietary supplements. This review aims to find novel potential genera for improving hypertension and give a direction for future studies on gut microbiota in hypertension.
Collapse
Affiliation(s)
- Dong Yan
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Ye Sun
- Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| | - Xiaoyue Zhou
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Wenhao Si
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina,Department of Dermatologythe First Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| | - Jieyu Liu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Min Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Minna Wu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
33
|
Varela-Trinidad GU, Domínguez-Díaz C, Solórzano-Castanedo K, Íñiguez-Gutiérrez L, Hernández-Flores TDJ, Fafutis-Morris M. Probiotics: Protecting Our Health from the Gut. Microorganisms 2022; 10:1428. [PMID: 35889147 PMCID: PMC9316266 DOI: 10.3390/microorganisms10071428] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota (GM) comprises billions of microorganisms in the human gastrointestinal tract. This microbial community exerts numerous physiological functions. Prominent among these functions is the effect on host immunity through the uptake of nutrients that strengthen intestinal cells and cells involved in the immune response. The physiological functions of the GM are not limited to the gut, but bidirectional interactions between the gut microbiota and various extraintestinal organs have been identified. These interactions have been termed interorganic axes by several authors, among which the gut-brain, gut-skin, gut-lung, gut-heart, and gut-metabolism axes stand out. It has been shown that an organism is healthy or in homeostasis when the GM is in balance. However, altered GM or dysbiosis represents a critical factor in the pathogenesis of many local and systemic diseases. Therefore, probiotics intervene in this context, which, according to various published studies, allows balance to be maintained in the GM, leading to an individual's good health.
Collapse
Affiliation(s)
- Gael Urait Varela-Trinidad
- Doctorado en Ciencias Biomédicas, Con Orientaciones en Inmunología y Neurociencias, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico; (G.U.V.-T.); (C.D.-D.)
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
| | - Carolina Domínguez-Díaz
- Doctorado en Ciencias Biomédicas, Con Orientaciones en Inmunología y Neurociencias, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico; (G.U.V.-T.); (C.D.-D.)
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
| | - Karla Solórzano-Castanedo
- Doctorado en Ciencias de la Nutrición Traslacional, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico;
| | - Liliana Íñiguez-Gutiérrez
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara, Coronel Calderón 777, Guadalajara 44280, Mexico; (L.Í.-G.); (T.d.J.H.-F.)
| | - Teresita de Jesús Hernández-Flores
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara, Coronel Calderón 777, Guadalajara 44280, Mexico; (L.Í.-G.); (T.d.J.H.-F.)
- Departamento de Disciplinas Filosóficas Metodológicas e Intrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico
| | - Mary Fafutis-Morris
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico
| |
Collapse
|
34
|
Dwaib HS, AlZaim I, Ajouz G, Eid AH, El-Yazbi A. Sex Differences in Cardiovascular Impact of Early Metabolic Impairment: Interplay between Dysbiosis and Adipose Inflammation. Mol Pharmacol 2022; 102:481-500. [PMID: 34732528 DOI: 10.1124/molpharm.121.000338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/23/2021] [Indexed: 11/22/2022] Open
Abstract
The evolving view of gut microbiota has shifted toward describing the colonic flora as a dynamic organ in continuous interaction with systemic physiologic processes. Alterations of the normal gut bacterial profile, known as dysbiosis, has been linked to a wide array of pathologies. Of particular interest is the cardiovascular-metabolic disease continuum originating from positive energy intake and high-fat diets. Accumulating evidence suggests a role for sex hormones in modulating the gut microbiome community. Such a role provides an additional layer of modulation of the early inflammatory changes culminating in negative metabolic and cardiovascular outcomes. In this review, we will shed the light on the role of sex hormones in cardiovascular dysfunction mediated by high-fat diet-induced dysbiosis, together with the possible involvement of insulin resistance and adipose tissue inflammation. Insights into novel therapeutic interventions will be discussed as well. SIGNIFICANCE STATEMENT: Increasing evidence implicates a role for dysbiosis in the cardiovascular complications of metabolic dysfunction. This minireview summarizes the available data on the sex-based differences in gut microbiota alterations associated with dietary patterns leading to metabolic impairment. A role for a differential impact of adipose tissue inflammation across sexes in mediating the cardiovascular detrimental phenotype following diet-induced dysbiosis is proposed. Better understanding of this pathway will help introduce early approaches to mitigate cardiovascular deterioration in metabolic disease.
Collapse
Affiliation(s)
- Haneen S Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ghina Ajouz
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| |
Collapse
|
35
|
Depletion of the gut microbiota enhances the blood pressure-lowering effect of captopril: implication of the gut microbiota in resistant hypertension. Hypertens Res 2022; 45:1505-1510. [PMID: 35513487 DOI: 10.1038/s41440-022-00921-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/08/2022] [Accepted: 03/22/2022] [Indexed: 11/08/2022]
Abstract
The role of the gut microbiota in the initiation and progression of hypertension has been newly identified, suggesting that targeting the gut microbiota may provide a new treatment strategy. This entails a complicated interaction between the gut microbiota and different host systems (e.g., immune system) or organs (e.g., gut, spleen) that contribute to blood pressure control. The significance of the gut microbiota in treatment-resistant hypertension is still unknown, owing to a lack of appropriate animal models. Given that the gut microbiota has a variety of enzymatic activities, we hypothesized that the gut microbiota may be involved in the metabolism of antihypertensive medications, causing treatment-resistant hypertension. We investigated this hypothesis in a simple, new hypertension paradigm and found that hypertensive rats pretreated with antibiotics to reduce the gut microbiota had a better response to the angiotensin-converting enzyme inhibitor captopril. This is a simple rodent model for testing the effectiveness of antihypertensive medications. Further mechanistic research may shed light on the pathogenic function of the gut microbiota in resistant hypertension. Our method presents a novel model that has the potential to be employed in the research of resistant hypertension.
Collapse
|
36
|
Zhang X, Gérard P. Diet-gut microbiota interactions on cardiovascular disease. Comput Struct Biotechnol J 2022; 20:1528-1540. [PMID: 35422966 PMCID: PMC8983311 DOI: 10.1016/j.csbj.2022.03.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/23/2022] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) are a group of disorders of the heart and blood vessels and remain the leading cause of morbidity and mortality worldwide. Over the past decades, accumulating studies indicated that the gut microbiota, an indispensable "invisible organ", plays a vital role in human metabolism and disease states including CVD. Among many endogenous and exogenous factors that can impact gut microbial communities, the dietary nutrients emerge as an essential component of host-microbiota relationships that can be involved in CVD susceptibility. In this review, we summarize the major concepts of dietary modulation of the gut microbiota and the chief principles of the involvement of this microbiota in CVD development. We also discuss the mechanisms of diet-microbiota crosstalk that regulate CVD progression, including endotoxemia, inflammation, gut barrier dysfunction and lipid metabolism dysfunction. In addition, we describe how metabolites produced by the microbiota, including trimethylamine-N-oxide (TMAO), secondary bile acids (BAs), short chain fatty acids (SCFAs) as well as aromatic amino acids (AAAs) derived metabolites play a role in CVD pathogenesis. Finally, we present the potential dietary interventions which interacted with gut microbiota as novel preventive and therapeutic strategies for CVD management.
Collapse
Affiliation(s)
- Xufei Zhang
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Philippe Gérard
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| |
Collapse
|
37
|
Tain YL, Hsu CN. Hypertension of Developmental Origins: Consideration of Gut Microbiome in Animal Models. Biomedicines 2022; 10:biomedicines10040875. [PMID: 35453625 PMCID: PMC9030804 DOI: 10.3390/biomedicines10040875] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/29/2022] [Accepted: 04/08/2022] [Indexed: 11/29/2022] Open
Abstract
Hypertension is the leading cause of global disease burden. Hypertension can arise from early life. Animal models are valuable for giving cogent evidence of a causal relationship between various environmental insults in early life and the hypertension of developmental origins in later life. These insults consist of maternal malnutrition, maternal medical conditions, medication use, and exposure to environmental chemicals/toxins. There is a burgeoning body of evidence on maternal insults can shift gut microbiota, resulting in adverse offspring outcomes later in life. Emerging evidence suggests that gut microbiota dysbiosis is involved in hypertension of developmental origins, while gut microbiota-targeted therapy, if applied early, is able to help prevent hypertension in later life. This review discusses the innovative use of animal models in addressing the mechanisms behind hypertension of developmental origins. We will also highlight the application of animal models to elucidate how the gut microbiota connects with other core mechanisms, and the potential of gut microbiota-targeted therapy as a novel preventive strategy to prevent hypertension of developmental origins. These animal models have certainly enhanced our understanding of hypertension of developmental origins, closing the knowledge gap between animal models and future clinical translation.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-975-368-975; Fax: +886-7733-8009
| |
Collapse
|
38
|
Wang Z, Wu F, Zhou Q, Qiu Y, Zhang J, Tu Q, Zhou Z, Shao Y, Xu S, Wang Y, Tao J. Berberine Improves Vascular Dysfunction by Inhibiting Trimethylamine-N-oxide via Regulating the Gut Microbiota in Angiotensin II-Induced Hypertensive Mice. Front Microbiol 2022; 13:814855. [PMID: 35350612 PMCID: PMC8957906 DOI: 10.3389/fmicb.2022.814855] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/15/2022] [Indexed: 01/14/2023] Open
Abstract
Berberine (BBR) has been demonstrated to exert cardiovascular protective effects by regulating gut microbiota. However, few studies examine the effect of BBR on the gut microbiota in hypertension. This study aims to investigate the role of BBR in regulating microbial alterations and vascular function in hypertension. C57BL/6 J mice were infused with Ang II (0.8 mg/kg/day) via osmotic minipumps and treated with BBR (150 mg/kg/day) or choline (1%) for 4 weeks. Blood pressure was detected by tail-cuff measurement once a week. Abdominal aorta pulse wave velocity (PWV) and endothelium dependent vasodilatation were measured to evaluate vascular function. Vascular remodeling was assessed by histological staining of aortic tissue. The fecal microbiota was profiled using 16S ribosomal DNA (rDNA) sequencing. Plasma trimethylamine (TMA)/trimethylamine-N-oxide (TMAO) and hepatic FMO3 expression were measured. We found that BBR treatment significantly alleviated the elevated blood pressure, vascular dysfunction, and pathological remodeling in Ang II-induced hypertensive mice, while choline treatment aggravated hypertension-related vascular dysfunction. 16S rDNA gene sequencing results showed that BBR treatment altered gut microbiota composition (reduced the Firmicutes/Bacteroidetes (F/B) ratio and increased the abundances of Lactobacillus). Moreover, BBR inhibited FMO3 expression and plasma TMA/TMAO production in hypertensive mice. TMAO treatment increased the apoptosis and oxidative stress of human aortic endothelial cells (HAECs) and aggravated Ang II-induced HAECs dysfunction in vitro. These results indicate that the protective effect of BBR in hypertension might be attributed (at least partially) to the inhibition of TMAO production via regulating the gut microbiota.
Collapse
Affiliation(s)
- Zhichao Wang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fang Wu
- Institute of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qianbing Zhou
- Department of Stomatology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yumin Qiu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianning Zhang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiang Tu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhe Zhou
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yijia Shao
- Institute of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shiyue Xu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Shiyue Xu,
| | - Yan Wang
- Institute of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Yan Wang,
| | - Jun Tao
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jun Tao,
| |
Collapse
|
39
|
Trikha SRJ, Lee DM, Ecton KE, Wrigley SD, Vazquez AR, Litwin NS, Thomas KN, Wei Y, Battson ML, Johnson SA, Kuhn KA, Colgan SP, Gentile CL, Weir TL. Transplantation of an obesity-associated human gut microbiota to mice induces vascular dysfunction and glucose intolerance. Gut Microbes 2021; 13:1940791. [PMID: 34313540 PMCID: PMC8317959 DOI: 10.1080/19490976.2021.1940791] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent preclinical data suggest that alterations in the gut microbiota may be an important factor linking obesity to vascular dysfunction, an early sign of cardiovascular disease. The purpose of this study was to begin translation of these preclinical data by examining whether vascular phenotypes in humans are transmissible through the gut microbiota. We hypothesized that germ-free mice colonized with gut microbiota from obese individuals would display diminished vascular function compared to germ-free mice receiving microbiota from lean individuals.We transplanted fecal material from obese and lean age-and sex-matched participants with disparate vascular function to germ-free mice. Using Principle Component Analysis, the microbiota of colonized mice separated by donor group along the first principle component, accounting for between 70-93% of the total variability in the dataset. The microbiota of mice receiving transplants from lean individuals was also characterized by increased alpha diversity, as well as increased relative abundance of potentially beneficial bacteria, including Bifidobacterium, Lactobacillus, and Bacteroides ovatis. Endothelium-dependent dilation, aortic pulse wave velocity and glucose tolerance were significantly altered in mice receiving microbiota from the obese donor relative to those receiving microbiota from the lean donor or those remaining germ-free.These data indicate that the obesity-associated human gut microbiota is sufficient to alter the vascular phenotype in germ-free mice in the absence of differences in body weight or dietary manipulation, and provide justification for future clinical trials to test the efficacy of microbiota-targeted therapies in the prevention or treatment of cardiovascular disease.
Collapse
Affiliation(s)
- S. Raj J. Trikha
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Dustin M. Lee
- Department of Nutritional Medicine, Brooke Army Medical Center, San Antonio, TX, USA
| | - Kayl E. Ecton
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Scott D. Wrigley
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Allegra R. Vazquez
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Nicole S. Litwin
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Keely N. Thomas
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Yuren Wei
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Micah L. Battson
- Department of Nutrition, Metropolitan State University, Denver, CO, USA
| | - Sarah A. Johnson
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Kristine A. Kuhn
- School of Medicine in the Division of Rheumatology and Gnotobiotic Core Director, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sean P. Colgan
- School of Medicine in the Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Christopher L. Gentile
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA,CONTACT Christopher L. Gentile 208 Gifford Bldg, 1571 Campus Delivery, Colorado State University, Fort Collins, CO 80523-1571, USA
| | - Tiffany L. Weir
- Department of Food Science & Human Nutrition, Colorado State University, Fort Collins, CO, USA,Tiffany L. Weir 210 Gifford Bldg, 1571 Campus Delivery, Colorado State University, Fort Collins, CO 80523-1571, USA
| |
Collapse
|
40
|
Role of the microbiota in hypertension and antihypertensive drug metabolism. Hypertens Res 2021; 45:246-253. [PMID: 34887530 DOI: 10.1038/s41440-021-00804-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/17/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022]
Abstract
Recent evidence suggests that the gut microbiota plays an important role in the development and pathogenesis of hypertension. Dysbiosis, an imbalance in the composition and function of the gut microbiota, was shown to be associated with hypertension in both animal models and humans. In this review, we provide insights into host-microbiota interactions and summarize the evidence supporting the importance of the microbiota in blood pressure (BP) regulation. Metabolites produced by the gut microbiota, especially short-chain fatty acids (SCFAs), modulate BP and vascular responses. Harmful gut-derived metabolites, such as trimethylamine N-oxide and several uremic toxins, exert proatherosclerotic, prothrombotic, and proinflammatory effects. High-salt intake alters the composition of the microbiota, and this microbial alteration contributes to the pathogenesis of salt-sensitive hypertension. In addition, the microbiota may impact the metabolism of drugs and steroid hormones in the host. The drug-metabolizing activities of the microbiota affect the pharmacokinetic parameters of antihypertensive drugs and contribute to the pathogenesis of licorice-induced pseudohyperaldosteronism. Furthermore, the oral microbiota plays a role in BP regulation by producing nitric oxide, which lowers BP via its vasodilatory effects. Thus, antihypertensive intervention strategies targeting the microbiota, such as the use of prebiotics, probiotics, and postbiotics (e.g., SCFAs), are considered new therapeutic options for the treatment of hypertension.
Collapse
|
41
|
Czernik PJ, Golonka RM, Chakraborty S, Yeoh BS, Abokor AA, Saha P, Yeo JY, Mell B, Cheng X, Baroi S, Tian Y, Patterson AD, Joe B, Vijay-Kumar M, Lecka-Czernik B. Reconstitution of the host holobiont in germ-free born male rats acutely increases bone growth and affects marrow cellular content. Physiol Genomics 2021; 53:518-533. [PMID: 34714176 PMCID: PMC8714805 DOI: 10.1152/physiolgenomics.00017.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 11/22/2022] Open
Abstract
Integration of microbiota in a host begins at birth and progresses during adolescence, forming a multidirectional system of physiological interactions. Here, we present an instantaneous effect of natural, bacterial gut colonization on the acceleration of longitudinal and radial bone growth in germ-free born, 7-wk-old male rats. Changes in bone mass and structure were analyzed after 10 days following the onset of colonization through cohousing with conventional rats and revealed unprecedented acceleration of bone accrual in cortical and trabecular compartments, increased bone tissue mineral density, improved proliferation and hypertrophy of growth plate chondrocytes, bone lengthening, and preferential deposition of periosteal bone in the tibia diaphysis. In addition, the number of small in size adipocytes increased, whereas the number of megakaryocytes decreased, in the bone marrow of conventionalized germ-free rats indicating that not only bone mass but also bone marrow environment is under control of gut microbiota signaling. The changes in bone status paralleled with a positive shift in microbiota composition toward short-chain fatty acids (SCFA)-producing microbes and a considerable increase in cecal SCFA concentrations, specifically butyrate. Furthermore, reconstitution of the host holobiont increased hepatic expression of IGF-1 and its circulating levels. Elevated serum levels of 25-hydroxy vitamin D and alkaline phosphatase pointed toward an active process of bone formation. The acute stimulatory effect on bone growth occurred independently of body mass increase. Overall, the presented model of conventionalized germ-free rats could be used to study microbiota-based therapeutics for combatting dysbiosis-related bone disorders.
Collapse
Affiliation(s)
- Piotr J Czernik
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Rachel M Golonka
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Saroj Chakraborty
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Beng San Yeoh
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Ahmed A Abokor
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Piu Saha
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Ji-Youn Yeo
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Blair Mell
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Xi Cheng
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Sudipta Baroi
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Bina Joe
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Matam Vijay-Kumar
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Beata Lecka-Czernik
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
42
|
Morris DJ, Brem AS, Odermatt A. Modulation of 11β-hydroxysteroid dehydrogenase functions by the cloud of endogenous metabolites in a local microenvironment: The glycyrrhetinic acid-like factor (GALF) hypothesis. J Steroid Biochem Mol Biol 2021; 214:105988. [PMID: 34464733 DOI: 10.1016/j.jsbmb.2021.105988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/08/2021] [Accepted: 08/25/2021] [Indexed: 01/09/2023]
Abstract
11β-Hydroxysteroid dehydrogenase (11β-HSD)-dependent conversion of cortisol to cortisone and corticosterone to 11-dehydrocorticosterone are essential in regulating transcriptional activities of mineralocorticoid receptors (MR) and glucocorticoid receptors (GR). Inhibition of 11β-HSD by glycyrrhetinic acid metabolites, bioactive components of licorice, causes sodium retention and potassium loss, with hypertension characterized by low renin and aldosterone. Essential hypertension is a major disease, mostly with unknown underlying mechanisms. Here, we discuss a putative mechanism for essential hypertension, the concept that endogenous steroidal compounds acting as glycyrrhetinic acid-like factors (GALFs) inhibit 11β-HSD dehydrogenase, and allow for glucocorticoid-induced MR and GR activation with resulting hypertension. Initially, several metabolites of adrenally produced glucocorticoids and mineralocorticoids were shown to be potent 11β-HSD inhibitors. Such GALFs include modifications in the A-ring and/or at positions 3, 7 and 21 of the steroid backbone. These metabolites may be formed in peripheral tissues or by gut microbiota. More recently, metabolites of 11β-hydroxy-Δ4androstene-3,17-dione and 7-oxygenated oxysterols have been identified as potent 11β-HSD inhibitors. In a living system, 11β-HSD isoforms are not exposed to a single substrate but to several substrates, cofactors, and various inhibitors simultaneously, all at different concentrations depending on physical state, tissue and cell type. We propose that this "cloud" of steroids and steroid-like substances in the microenvironment determines the 11β-HSD-dependent control of MR and GR activity. A dysregulated composition of this cloud of metabolites in the respective microenvironment needs to be taken into account when investigating disease mechanisms, for forms of low renin, low aldosterone hypertension.
Collapse
Affiliation(s)
- David J Morris
- Department of Pathology and Laboratory Medicine, The Miriam Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Andrew S Brem
- Division of Kidney Diseases and Hypertension, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
43
|
Kong CY, Li ZM, Mao YQ, Chen HL, Hu W, Han B, Wang LS. Probiotic yogurt blunts the increase of blood pressure in spontaneously hypertensive rats via remodeling of the gut microbiota. Food Funct 2021; 12:9773-9783. [PMID: 34494630 DOI: 10.1039/d1fo01836a] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dietary intake of probiotic yogurt, which has beneficial effects on intestinal microecology, is associated with a lower incidence of hypertension. Recent studies have shown that the gut microbiota plays a vital role in the development of hypertension. However, the impact of the gut microbiota in the antihypertensive effect of probiotic yogurt remains unclear. Here, we evaluated the impact of the gut microbiota in the antihypertensive effect of probiotic yogurt in spontaneously hypertensive rats (SHR). SHR were treated with probiotic yogurt (0.2 mL per 100 g body weight) (SHR-Y group) for seven weeks and compared with whole milk-treated (0.2 mL per 100 g body weight) SHR (SHR group) and with normotensive Wistar-Kyoto rats (WKY group). The blood pressure and heart function of the rats in the WKY, SHR, and SHR-Y groups were measured. Fecal microbiota was assessed by 16S ribosomal RNA (16S rRNA) gene sequencing. To investigate whether probiotic yogurt prevents hypertension in spontaneously hypertensive rats through the gut microbiota, we co-housed SHR rats (SHRCOH) with SHR-Y rats (SHRCOH-Y), thus allowing the transfer of microbiota via coprophagy. Compared with whole milk, supplementation of probiotic yogurt significantly reduced the blood pressure, heart rate (HR), and cardiac function. We found that the probiotic yogurt modified the gut microbiota populations and increased the alpha diversity. Gut microbiota remodeling by co-housing partly rescued the increase of blood pressure and impaired the cardiac function of SHR rats. Moreover, probiotic yogurt modulated the gut microbiota in mice by increasing the abundance of short-chain fatty acid (SCFA)-producing bacteria and SCFA levels (acetic acid, propionic acid, butyric acid, and valeic acid) in the feces. Together, the presented data revealed that probiotic yogurt exhibited antihypertensive effects in SHR rats via remodeling of the gut microbiota.
Collapse
Affiliation(s)
- Chao-Yue Kong
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, 201100 Shanghai, China. .,Institute of Fudan-Minhang academic health system, Minhang Hospital, Fudan University, 201100 Shanghai, China
| | - Zhan-Ming Li
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, 201100 Shanghai, China. .,Institute of Fudan-Minhang academic health system, Minhang Hospital, Fudan University, 201100 Shanghai, China
| | - Yu-Qin Mao
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, 201100 Shanghai, China. .,Institute of Fudan-Minhang academic health system, Minhang Hospital, Fudan University, 201100 Shanghai, China
| | - Hui-Ling Chen
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, 201100 Shanghai, China. .,Institute of Fudan-Minhang academic health system, Minhang Hospital, Fudan University, 201100 Shanghai, China
| | - Wei Hu
- Department of Cardiology, Minhang Hospital, Fudan University, 201100 Shanghai, China
| | - Bing Han
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, 201100 Shanghai, China. .,Institute of Fudan-Minhang academic health system, Minhang Hospital, Fudan University, 201100 Shanghai, China
| | - Li-Shun Wang
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital, Fudan University, 201100 Shanghai, China. .,Institute of Fudan-Minhang academic health system, Minhang Hospital, Fudan University, 201100 Shanghai, China
| |
Collapse
|
44
|
Xiong Y, Xiong Y, Zhu P, Wang Y, Yang H, Zhou R, Shu Y, Zhou H, Li Q. The Role of Gut Microbiota in Hypertension Pathogenesis and the Efficacy of Antihypertensive Drugs. Curr Hypertens Rep 2021; 23:40. [PMID: 34487269 DOI: 10.1007/s11906-021-01157-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 02/05/2023]
Abstract
PURPOSE OF HEADING To review the relationship between intestinal microbes and hypertension and its impact on the efficacy of antihypertensive drugs, and help to address some of these knowledge gaps. RECENT FINDINGS Hypertension is associated with cardiovascular diseases and is the most important modifiable risk factor for all-cause morbidity and mortality worldwide. The pathogenesis of hypertension is complex, including factors such as dietary, environmental and genetics. Recently, the studies have shown that the gut microbiota influences the occurrence and development of hypertension through a variety of ways, including affecting the production of short-chain fatty acids, dysfunction of the brain-gut axis, and changes in serotonin content that cause the imbalance of vagus and sympathetic nerve output associated with hypertension. However, patients with hypertension typically take antihypertensive drugs orally on a long-term basis, and most antihypertensive drugs are absorbed by the gastrointestinal tract. Studies have shown that the pharmacokinetics and metabolism of antihypertensive drugs may be influenced by microbiota, or antihypertensive drugs act directly on the intestinal flora to exert efficacy, including regulation of intestinal microbial metabolism, intestinal inflammation, and intestinal sympathetic nervous system disorders. The intestinal flora can affect the pharmacokinetics and metabolism of antihypertensive drugs in the rats, and intestinal microbiota also can be the target "organ" by antihypertensive drugs.
Collapse
Affiliation(s)
- Yanling Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China
| | - Yalan Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China
| | - Peng Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China
| | - Yusheng Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, ShanTou, Guangdong, China
| | - Haijun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China
| | - Rong Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China
| | - Yan Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, China.
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, China.
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, China.
| |
Collapse
|
45
|
Gut microbiota dependent trimethylamine N-oxide aggravates angiotensin II-induced hypertension. Redox Biol 2021; 46:102115. [PMID: 34474396 PMCID: PMC8408632 DOI: 10.1016/j.redox.2021.102115] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota produce Trimethylamine N-oxide (TMAO) by metabolizing dietary phosphatidylcholine, choline, l-carnitine and betaine. TMAO is implicated in the pathogenesis of chronic kidney disease (CKD), diabetes, obesity and atherosclerosis. We test, whether TMAO augments angiotensin II (Ang II)-induced vasoconstriction and hence promotes Ang II-induced hypertension. Plasma TMAO levels were indeed elevated in hypertensive patients, thus the potential pathways by which TMAO mediates these effects were explored. Ang II (400 ng/kg−1min−1) was chronically infused for 14 days via osmotic minipumps in C57Bl/6 mice. TMAO (1%) or antibiotics were given via drinking water. Vasoconstriction of renal afferent arterioles and mesenteric arteries were assessed by microperfusion and wire myograph, respectively. In Ang II-induced hypertensive mice, TMAO elevated systolic blood pressure and caused vasoconstriction, which was alleviated by antibiotics. TMAO enhanced the Ang II-induced acute pressor responses (12.2 ± 1.9 versus 20.6 ± 1.4 mmHg; P < 0.05) and vasoconstriction (32.3 ± 2.6 versus 55.9 ± 7.0%, P < 0.001). Ang II-induced intracellular Ca2+ release in afferent arterioles (147 ± 7 versus 234 ± 26%; P < 0.001) and mouse vascular smooth muscle cells (VSMC, 123 ± 3 versus 157 ± 9%; P < 0.001) increased by TMAO treatment. Preincubation of VSMC with TMAO activated the PERK/ROS/CaMKII/PLCβ3 pathway. Pharmacological inhibition of PERK, ROS, CaMKII and PLCβ3 impaired the effect of TMAO on Ca2+ release. Thus, TMAO facilitates Ang II-induced vasoconstriction, thereby promoting Ang II-induced hypertension, which involves the PERK/ROS/CaMKII/PLCβ3 axis. Orally administered TMAO aggravates Ang II-induced hypertension. Antibiotics alleviate Ang II-induced hypertension by reducing TMAO generation. High concentrations of TMAO constrict afferent arterioles and mesenteric arteries and increase blood pressure. Low concentrations of TMAO enhance Ang II-induced vasoconstriction and acute pressor response via activating PERK/ROS/CaMKII/PLCβ3/Ca2+ pathway.
Collapse
|
46
|
Tomasova L, Grman M, Ondrias K, Ufnal M. The impact of gut microbiota metabolites on cellular bioenergetics and cardiometabolic health. Nutr Metab (Lond) 2021; 18:72. [PMID: 34266472 PMCID: PMC8281717 DOI: 10.1186/s12986-021-00598-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022] Open
Abstract
Recent research demonstrates a reciprocal relationship between gut microbiota-derived metabolites and the host in controlling the energy homeostasis in mammals. On the one hand, to thrive, gut bacteria exploit nutrients digested by the host. On the other hand, the host utilizes numerous products of gut bacteria metabolism as a substrate for ATP production in the colon. Finally, bacterial metabolites seep from the gut into the bloodstream and interfere with the host’s cellular bioenergetics machinery. Notably, there is an association between alterations in microbiota composition and the development of metabolic diseases and their cardiovascular complications. Some metabolites, like short-chain fatty acids and trimethylamine, are considered markers of cardiometabolic health. Others, like hydrogen sulfide and nitrite, demonstrate antihypertensive properties. Scientific databases were searched for pre-clinical and clinical studies to summarize current knowledge on the role of gut microbiota metabolites in the regulation of mammalian bioenergetics and discuss their potential involvement in the development of cardiometabolic disorders. Overall, the available data demonstrates that gut bacteria products affect physiological and pathological processes controlling energy and vascular homeostasis. Thus, the modulation of microbiota-derived metabolites may represent a new approach for treating obesity, hypertension and type 2 diabetes.
Collapse
Affiliation(s)
- Lenka Tomasova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05, Bratislava, Slovak Republic.
| | - Marian Grman
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05, Bratislava, Slovak Republic
| | - Karol Ondrias
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05, Bratislava, Slovak Republic
| | - Marcin Ufnal
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-091, Warsaw, Poland.
| |
Collapse
|
47
|
Silva CBP, Elias-Oliveira J, McCarthy CG, Wenceslau CF, Carlos D, Tostes RC. Ethanol: striking the cardiovascular system by harming the gut microbiota. Am J Physiol Heart Circ Physiol 2021; 321:H275-H291. [PMID: 34142885 DOI: 10.1152/ajpheart.00225.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ethanol consumption represents a significant public health problem, and excessive ethanol intake is a risk factor for cardiovascular disease (CVD), one of the leading causes of death and disability worldwide. The mechanisms underlying the effects of ethanol on the cardiovascular system are complex and not fully comprehended. The gut microbiota and their metabolites are indispensable symbionts essential for health and homeostasis and therefore, have emerged as potential contributors to ethanol-induced cardiovascular system dysfunction. By mechanisms that are not completely understood, the gut microbiota modulates the immune system and activates several signaling pathways that stimulate inflammatory responses, which in turn, contribute to the development and progression of CVD. This review summarizes preclinical and clinical evidence on the effects of ethanol in the gut microbiota and discusses the mechanisms by which ethanol-induced gut dysbiosis leads to the activation of the immune system and cardiovascular dysfunction. The cross talk between ethanol consumption and the gut microbiota and its implications are detailed. In summary, an imbalance in the symbiotic relationship between the host and the commensal microbiota in a holobiont, as seen with ethanol consumption, may contribute to CVD. Therefore, manipulating the gut microbiota, by using antibiotics, probiotics, prebiotics, and fecal microbiota transplantation might prove a valuable opportunity to prevent/mitigate the deleterious effects of ethanol and improve cardiovascular health and risk prevention.
Collapse
Affiliation(s)
- Carla B P Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jefferson Elias-Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Camilla F Wenceslau
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
48
|
Li J, Richards EM, Handberg EM, Pepine CJ, Raizada MK. Distinct Gene Expression Profiles in Colonic Organoids from Normotensive and the Spontaneously Hypertensive Rats. Cells 2021; 10:cells10061523. [PMID: 34204247 PMCID: PMC8234507 DOI: 10.3390/cells10061523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Hypertension is associated with gut bacterial dysbiosis and gut pathology in animal models and people. Butyrate-producing gut bacteria are decreased in hypertension. RNA-seq analysis of gut colonic organoids prepared from spontaneously hypertensive rats (SHR) and normotensive Wistar Kyoto (WKY) rats was used to test the hypothesis that impaired interactions between the gut microbiome and gut epithelium are involved and that these would be remediated with butyrate supplementation. Gene expressions in immune responses including antigen presentation and antiviral pathways were decreased in the gut epithelium of the SHR in organoids and confirmed in vivo; these deficits were corrected by butyrate supplementation. Deficits in gene expression driving epithelial proliferation and differentiation were also observed in SHR. These findings highlight the importance of aligned interactions of the gut microbiome and gut immune responses to blood pressure homeostasis.
Collapse
Affiliation(s)
- Jing Li
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, USA; (J.L.); (E.M.R.)
| | - Elaine M. Richards
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, USA; (J.L.); (E.M.R.)
| | - Eileen M. Handberg
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA; (E.M.H.); (C.J.P.)
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA; (E.M.H.); (C.J.P.)
| | - Mohan K. Raizada
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL 32610, USA; (J.L.); (E.M.R.)
- Correspondence: ; Tel.: +1-352-392-9299
| |
Collapse
|
49
|
Yang T, Chakraborty S, Mandal J, Mei X, Joe B. Microbiota and Metabolites as Factors Influencing Blood Pressure Regulation. Compr Physiol 2021; 11:1731-1757. [PMID: 33792901 DOI: 10.1002/cphy.c200009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The study of microbes has rapidly expanded in recent years due to a surge in our understanding that humans host a plethora of commensal microbes, which reside in their bodies and depending upon their composition, contribute to either normal physiology or pathophysiology. This article provides a general foundation for learning about host-commensal microbial interactions as an emerging area of research. The article is divided into two sections. The first section is dedicated to introducing commensal microbiota and its known effects on the host. The second section is on metabolites, which are biochemicals that the host and the microbes use for bi-directional communication with each other. Together, the sections review what is known about how microbes interact with the host to impact cardiovascular physiology, especially blood pressure regulation. © 2021 American Physiological Society. Compr Physiol 11:1731-1757, 2021.
Collapse
Affiliation(s)
- Tao Yang
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Saroj Chakraborty
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Juthika Mandal
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Xue Mei
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Rapidly emerging evidence implicates an important role of gut-brain-bone marrow (BM) axis involving gut microbiota (GM), gut epithelial wall permeability, increased production of pro-inflammatory BM cells and neuroinflammation in hypertension (HTN). However, the precise sequence of events involving these organs remains to be established. Furthermore, whether an impaired gut-brain-BM axis is a cause or consequence of HTN is actively under investigation. This will be extremely important for translation of this fundamental knowledge to novel, innovative approaches for the control and management of HTN. Therefore, our objectives are to summarize the latest hypothesis, provide evidence for and against the impaired gut, BM and brain interactions in HTN and discuss perspectives and future directions. RECENT FINDINGS Hypertensive stimuli activate autonomic neural pathways resulting in increased sympathetic and decreased parasympathetic cardiovascular modulation. This directly affects the functions of cardiovascular-relevant organs to increase blood pressure. Increases in sympathetic drive to the gut and BM also trigger sequences of signaling events that ultimately contribute to altered GM, increased gut permeability, enhanced gut- and brain-targeted pro-inflammatory cells from the BM in perpetuation and establishment of HTN. SUMMARY In this review, we present the mechanisms involving the brain, gut, and BM, whose dysfunctional interactions may be critical in persistent neuroinflammation and key in the development and establishment of HTN.
Collapse
Affiliation(s)
- Jing Li
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, USA
| | | | | |
Collapse
|