1
|
Chen X, Lin WY, Zhang FW, Guo LQ, Ge H, Ge DZ, Tan JJ, Liu BC, Wang RR, Zhang L. Investigation of oral microbiome composition in elderly Chinese patients with hypertension: a cross-sectional study. J Oral Microbiol 2025; 17:2489603. [PMID: 40270620 PMCID: PMC12016255 DOI: 10.1080/20002297.2025.2489603] [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: 10/15/2024] [Revised: 02/10/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Background Hypertension is a prevalent metabolic disorder in the elderly, with its pathogenesis linked to gut microbiota dysbiosis. Recent studies suggested that oral microbiota may also play a role in hypertension development, yet its relationship with hypertension in the elderly remains underexplored. Objective This cross-sectional study aimed to examine the structure of the oral microbiota and its association with hypertension in elderly patients, providing insights into hypertension prevention and treatment. Methods A total of 206 subjects (60-89 years) were categorized into normal (CON) and hypertensive (HTN) groups, based on the Chinese Hypertension Guidelines. Saliva samples were analyzed using 16S rRNA gene sequencing. Results Oral microbiota composition was significantly influenced by blood pressure. At the phylum level, Synergistetes and Spirochaetes were more significantly abundant in the HTN group, while at the genus level Treponema and Leptothrix was higher, Actinomyces and Capnocytophaga were lower in HTN. Random Forest analysis identified 15 key microbiota as strong discriminators of HTN (AUC 0.74). Blood pressure was negatively correlated with Actinomycetes and positively correlated with Leptothrix. PICRUST2 analysis revealed elevated chlorinated compound degradation in HTN patients. Conclusions This study identified distinct oral microbiota in elderly hypertensive patients, highlighting the role of the oral microbiome in hypertension pathogenesis.
Collapse
Affiliation(s)
- Xin Chen
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Yong Lin
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng-Wei Zhang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Qiang Guo
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Han Ge
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ding-Zuo Ge
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Juan-Juan Tan
- Institute of Integrative Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
- Institute of Integrative Medicine, Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Bao-Cheng Liu
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui-Rui Wang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Puel EM, Taruhn LF, Damé-Teixeira N, Stefani CM, Lataro RM. Is there a link between the abundance of nitrate-reducing bacteria and arterial hypertension? A systematic review. Nitric Oxide 2025:S1089-8603(25)00030-8. [PMID: 40220988 DOI: 10.1016/j.niox.2025.04.001] [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: 02/05/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
CONTEXT Nitric oxide is a vasodilator molecule that acts on blood pressure (BP) control, and its production can occur through the reduction of nitrates by oral or intestinal nitrate-reducing bacteria. However, the relationship between nitrate-reducing bacteria and arterial hypertension (HTN) remains under debate. OBJECTIVE Systematically review if there is an association between the abundance of oral and intestinal nitrate-reducing bacteria and the occurrence of HTN in humans. DATABASES AND ELIGIBILITY CRITERIA MEDLINE, Scopus, Cochrane Library, EMBASE, LILACS, Web of Science, Livivo, ProQuest Dissertations, and Google Scholar were searched for eligible articles until February 10th, 2024. Studies were included if they: (1) were observational studies or clinical trials; (2) included adults (≥ 18 years old) with HTN (systolic BP ≥130 mmHg and/or diastolic BP >80 mmHg and/or use of BP lowering medication); (3) compared (or not) to no-HTN adults; and (4) used next-generation sequencing microbiome analysis to identify bacterial taxa in the oral and/or gut nitrate-reducing bacteria. RESULTS The search identified 9365 articles, and 28 were included in the study after applying the inclusion and exclusion criteria; 23 articles assessed the gut microbiota, 4 assessed the oral microbiota, and 1 that assessed both. Depletion of nitrate-reducing bacteria was not consistently showed in the studies. The included studies reported reduction, increase, and no change in the nitrate-reducing bacteria genera or species in oral or gut microbiota. CONCLUSION We found no association between the abundance of oral and gut nitrate-reducing bacteria and the occurrence of HTN in humans. REGISTRATION PROSPERO identification number CRD42022315891.
Collapse
Affiliation(s)
- Esthela M Puel
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Lillian F Taruhn
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Nailê Damé-Teixeira
- Department of Dentistry, School of Health Sciences, University of Brasília, Brasília, Brazil
| | - Cristine M Stefani
- Department of Dentistry, School of Health Sciences, University of Brasília, Brasília, Brazil
| | - Renata M Lataro
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
3
|
González-Correa C, Moleón J, Miñano S, Robles-Vera I, de la Visitación N, Guerra-Hernández E, Toral M, Jiménez R, Duarte J, Romero M. Protective Effect of Dietary Fiber on Blood Pressure and Vascular Dysfunction Through Regulation of Sympathetic Tone and Immune Response in Genetic Hypertension. Phytother Res 2025; 39:1858-1875. [PMID: 40122676 DOI: 10.1002/ptr.8484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
The mechanisms underlying the antihypertensive effect of dietary fibers remain poorly understood. This study investigates whether dietary fiber supplementation can prevent cardiovascular damage and high blood pressure in a genetic model of neurogenic hypertension. Six-week-old male spontaneously hypertensive rats (SHR) and their respective normotensive control, Wistar Kyoto rats (WKY), were divided into four groups: Untreated WKY, untreated SHR, SHR treated with resistant starch (SHR + RS), and SHR treated with inulin-type fructans (SHR + ITF) for 12 weeks. Additionally, a faecal microbiota transplantation (FMT) experiment was conducted, transferring faecal content from treated SHR donors to recipient SHRs. A diet rich in RS fiber reduced vascular oxidative stress, inflammation, and high blood pressure. These protective effects were associated with a reshaped gut microbiota, leading to increased short-chain fatty acid production, reduced endotoxemia, decreased sympathetic activity, and a restored balance between Th17 and Treg lymphocytes in mesenteric lymph nodes and aorta. Elevated plasma levels of acetate and butyrate in the SHR + RS group correlated with increased expression of aortic GPR41, GRP43 and PPARδ. Conversely, ITF treatment failed to prevent hypertension or endothelial dysfunction in SHR. FMT from the SHR + RS group to recipient SHR partially replicated these beneficial effects. This study highlights the antihypertensive benefits of dietary insoluble RS fiber, which are attributed to enhanced short-chain fatty acids production in the gut. This leads to improved gut permeability, reduced sympathetic tone, and diminished vascular T-cell accumulation. Therefore, dietary interventions with RS fiber may offer promising therapeutic strategies for preventing hypertension.
Collapse
Affiliation(s)
- Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | | | - Marta Toral
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| |
Collapse
|
4
|
Dardi P, Coutinho CP, de Oliveira S, Teixeira SA, Gacek RRF, Purgatto E, Vinolo MAR, Muscará MN, Rossoni LV. Changes in the intestinal microbiota induced by the postnatal environment and their association with hypertension. Pharmacol Res 2025; 212:107621. [PMID: 39848350 DOI: 10.1016/j.phrs.2025.107621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
It has been established that cross-fostering impacts the development of hypertension in spontaneously hypertensive rats (SHR). However, the ability of the cross-fostering protocol to shape gut microbiota profile in SHR and impact hypertension is not known. In this sense, the current study explored the influence of normotensive and hypertensive postnatal environments on the intestinal microbiota structure, composition, and functional capacity of SHR and Wistar rats. Our findings revealed significant differences in the microbiota's composition and its metabolic activity in young non-fostered SHR (SS) vs. Wistar (WW) rats, even before hypertension onset, characterized by a reduction of the "low-abundance" bacterial genera, a diminished availability of fecal butyrate and elevated hydrogen sulfide (H2S) production by the SS gut microbiota. Despite influencing the microbiota of both strains, cross-fostering did not fully replicate the microbiota composition of the naturally reared groups in the SHR nursed by Wistar mothers (SW), or in the Wistar rats breastfed by SHR mothers (WS). The SW group had fewer significant genera identified at the Partial Least Squares Discriminant Analysis (PLS-DA), despite resembling the genera profile identified in the normotensive group. While sharing bacterial genera with both SS and WW groups, the WS group is distinguished by its unique microbial composition, particularly by a greater diversity of the 'low-abundance' bacterial genera. Moreover, decreased systolic blood pressure was observed in the SW group compared to the SS group in adulthood. Thus, we could establish a link between microbiota composition and hypertension development, associating it with the loss of the "low-abundance" bacterial taxa. Our data suggest that the postnatal environment is pivotal to promoting gut microbiota compositional changes and contributes to hypertension development.
Collapse
Affiliation(s)
- Patrizia Dardi
- Laboratory of Vascular Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Camille Perella Coutinho
- Department of Food and Experimental Nutrition /FoRC - Food Research Center, University of Sao Paulo, Sao Paulo, Brazil
| | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Simone Aparecida Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Eduardo Purgatto
- Department of Food and Experimental Nutrition /FoRC - Food Research Center, University of Sao Paulo, Sao Paulo, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Marcelo Nicolás Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Luciana Venturini Rossoni
- Laboratory of Vascular Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
5
|
Cheng CK, Ye L, Wang Y, Wang YL, Xia Y, Wong SHS, Chen S, Huang Y. Exercised gut microbiota improves vascular and metabolic abnormalities in sedentary diabetic mice through gut‒vascular connection. JOURNAL OF SPORT AND HEALTH SCIENCE 2025; 14:101026. [PMID: 39827989 PMCID: PMC11937666 DOI: 10.1016/j.jshs.2025.101026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Exercise elicits cardiometabolic benefits, reducing the risks of cardiovascular diseases and type 2 diabetes. This study aimed to investigate the vascular and metabolic effects of gut microbiota from exercise-trained donors on sedentary mice with type 2 diabetes and the potential mechanism. METHODS Leptin receptor-deficient diabetic (db/db) and nondiabetic (db/m+) mice underwent running treadmill exercise for 8 weeks, during which fecal microbiota transplantation (FMT) was parallelly performed from exercise-trained to sedentary diabetic (db/db) mice. Endothelial function, glucose homeostasis, physical performance, and vascular signaling of recipient mice were assessed. Vascular and intestinal stresses, including inflammation, oxidative stress, and endoplasmic reticulum (ER) stress, were investigated. RNA sequencing analysis on mouse aortic and intestinal tissues was performed. Gut microbiota profiles of recipient mice were evaluated by metagenomic sequencing. RESULTS Chronic exercise improved vascular and metabolic abnormalities in donor mice. Likewise, FMT from exercised donors retarded body weight gain and slightly improved grip strength and rotarod performance in recipient mice. Exercise-associated FMT enhanced endothelial function in different arteries, suppressed vascular and intestinal stresses, and improved glucose homeostasis in recipient mice, with noted microRNA-181b upregulation in aortas and intestines. Altered gut microbiota profiles and gut-derived factors (e.g., short-chain fatty acids and glucagon-like peptide-1) as well as improved intestinal integrity shall contribute to the cardiometabolic benefits, implying a gut‒vascular connection. CONCLUSION This proof-of-concept study indicates that exercised microbiota confers cardiometabolic benefits on sedentary db/db mice, extending the beneficial mechanism of exercise through gut‒vascular communication. The findings open up new therapeutic opportunities for cardiometabolic diseases and shed light on the development of exercise mimetics by targeting the gut microbiota.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR 999077, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Lianwei Ye
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR 999077, China
| | - Yu Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen 518071, China
| | - Ya-Ling Wang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR 999077, China
| | - Yin Xia
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Stephen Heung-Sang Wong
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Sheng Chen
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR 999077, China.
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR 999077, China; Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR 999077, China.
| |
Collapse
|
6
|
Avery EG, Haag LM, McParland V, Kedziora SM, Zigra GJ, Valdes DS, Kirchner M, Popp O, Geisberger S, Nonn O, Karlsen TV, N'Diaye G, Yarritu A, Bartolomaeus H, Bartolomaeus TUP, Tagiyeva NA, Wimmer MI, Haase N, Zhang YD, Wilhelm A, Grütz G, Tenstad O, Wilck N, Forslund SK, Klopfleisch R, Kühl AA, Atreya R, Kempa S, Mertins P, Siegmund B, Wiig H, Müller DN. Intestinal interstitial fluid isolation provides novel insight into the human host-microbiome interface. Cardiovasc Res 2025:cvae267. [PMID: 39804196 DOI: 10.1093/cvr/cvae267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/13/2024] [Accepted: 11/12/2024] [Indexed: 03/28/2025] Open
Abstract
AIMS The gastrointestinal (GI) tract is composed of distinct sub-regions, which exhibit segment-specific differences in microbial colonization and (patho)physiological characteristics. Gut microbes can be collectively considered as an active endocrine organ. Microbes produce metabolites, which can be taken up by the host and can actively communicate with the immune cells in the gut lamina propria with consequences for cardiovascular health. Variation in bacterial load and composition along the GI tract may influence the mucosal microenvironment and thus be reflected its interstitial fluid (IF). Characterization of the segment-specific microenvironment is challenging and largely unexplored because of lack of available tools. METHODS AND RESULTS Here, we developed methods, namely tissue centrifugation and elution, to collect IF from the mucosa of different intestinal segments. These methods were first validated in rats and mice, and the tissue elution method was subsequently translated for use in humans. These new methods allowed us to quantify microbiota-derived metabolites, mucosa-derived cytokines, and proteins at their site-of-action. Quantification of short-chain fatty acids showed enrichment in the colonic IF. Metabolite and cytokine analyses revealed differential abundances within segments, often significantly increased compared to plasma, and proteomics revealed that proteins annotated to the extracellular phase were site-specifically identifiable in IF. Lipopolysaccharide injections in rats showed significantly higher ileal IL-1β levels in IF compared to the systemic circulation, suggesting the potential of local as well as systemic effect. CONCLUSION Collection of IF from defined segments and the direct measurement of mediators at the site-of-action in rodents and humans bypasses the limitations of indirect analysis of faecal samples or serum, providing direct insight into this understudied compartment.
Collapse
Affiliation(s)
- Ellen G Avery
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Lea-Maxie Haag
- Department for Medicine (Gastroenterology, Infectious Diseases, Rheumatology) Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Charitéplatz 1, Berlin 10117, Germany
| | - Victoria McParland
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sarah M Kedziora
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Gabriel J Zigra
- Department for Medicine (Gastroenterology, Infectious Diseases, Rheumatology) Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Daniela S Valdes
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charite-Universitätsmedizin Berlin, Berlin, Germany
- Proteomics Platform, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Oliver Popp
- Proteomics Platform, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sabrina Geisberger
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Integrative Proteomics and Metabolomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Olivia Nonn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Tine V Karlsen
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| | - Gabriele N'Diaye
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alex Yarritu
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Hendrik Bartolomaeus
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Theda U P Bartolomaeus
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nurana A Tagiyeva
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Moritz I Wimmer
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Faculty of Medicine, Universität Tübingen, Tübingen, Germany
| | - Nadine Haase
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Yiming D Zhang
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Integrative Proteomics and Metabolomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Andreas Wilhelm
- CheckImmune GmbH, BerlinBioCube, Robert-Rössle Str. 10, Berlin 13125, Germany
| | - Gerald Grütz
- CheckImmune GmbH, BerlinBioCube, Robert-Rössle Str. 10, Berlin 13125, Germany
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| | - Nicola Wilck
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin, Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Sofia K Forslund
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja A Kühl
- Department for Medicine (Gastroenterology, Infectious Diseases, Rheumatology) Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Univeristät Berlin and Humboldt Universität zu Berlin, iPATH, Berlin, Berlin, Germany
| | - Raja Atreya
- Department of Medicine 1, Friedrich-Alexander University, Erlangen, Germany
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Integrative Proteomics and Metabolomics Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charite-Universitätsmedizin Berlin, Berlin, Germany
- Proteomics Platform, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Britta Siegmund
- Department for Medicine (Gastroenterology, Infectious Diseases, Rheumatology) Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| | - Dominik N Müller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
7
|
Escobar C, Aldeguer X, Vivas D, Manzano Fernández S, Gonzalez Caballero E, Garcia Martín A, Barrios V, Freixa-Pamias R. The gut microbiota and its role in the development of cardiovascular disease. Expert Rev Cardiovasc Ther 2025; 23:23-34. [PMID: 39915986 DOI: 10.1080/14779072.2025.2463366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION The pathophysiology of cardiovascular diseases encompasses a complex interplay of genetic and environmental risk factors. Even if traditional risk factors are treated to target, there remains a residual risk. AREAS COVERED This manuscript reviews the potential role of gut microbiota in the development of cardiovascular disease, and as potential target. A systematic search was conducted until 30 October 2024 on PubMed (MEDLINE), using the MeSH terms [Gut microbiota] + [Dysbiosis] + [Cardiovascular] + [TMAO] + [bile acids] + [short-chain fatty acids]. EXPERT OPINION The term dysbiosis implies changes in equilibrium, with modifications in the composition and functionality of microbiota and a series of additional factors: reduced diversity and uniformity of microorganisms; reduced short-chain fatty acid-producing bacteria; increased gut permeability; release of metabolites, such as trimethylamine N-oxide, betaine, phenylalanine, tryptophan-kynurenine, phenylacetylglutamine, and lipopolysaccharides; and reduced secondary bile acid excretion, leading to inflammation, oxidative stress, and endothelial dysfunction and facilitating the onset of pathological conditions, including obesity, hypertension, diabetes, atherosclerosis, and heart failure. Attempts to restore gut microbiota balance through different interventions, mainly changes in diet, have been shown to positively affect individual components and metabolites and reduce the risk of cardiovascular disease. In addition, probiotics and prebiotics are potentially useful. Fecal microbiota transplantation is a promising therapy.
Collapse
Affiliation(s)
- Carlos Escobar
- Cardiology Department, University Hospital La Paz, Madrid, Spain
| | - Xavier Aldeguer
- Gastroenterology Department, Hospital Doctor Josep Trueta i Santa Caterina, Institut d'investigació Biomèdica de Girona IDIBGI, Girona/Salt, Spain
| | - David Vivas
- Cardiovascular Institute, San Carlos University Hospital, Madrid, Spain
- Cardiology Department, Cardiovascular Institute Vithas Milagrosa and Aravaca, Madrid, Spain
| | | | | | - Ana Garcia Martín
- Cardiology Department, University Hospital Ramón y Cajal, Alcalá University, Madrid, Spain
| | - Vivencio Barrios
- Cardiology Department, University Hospital Ramón y Cajal, Alcalá University, Madrid, Spain
| | - Román Freixa-Pamias
- Cardiology Department, Complex Hospitalari Moisès Broggi, Sant Joan Despí, Barcelona, Spain
| |
Collapse
|
8
|
Ribeiro FPB, de Luna Freire MO, de Oliveira Coutinho D, de Santana Cirilo MA, de Brito Alves JL. Gut Dysbiosis and Probiotic Therapy in Chronic Kidney Disease: A Comprehensive Review. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10427-9. [PMID: 39668321 DOI: 10.1007/s12602-024-10427-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
Chronic kidney disease (CKD) is a multifactorial disease affecting more than 13.4% of the world's population and is a growing public health problem. It is silent in its early stages and leads to irreversible kidney damage as the disease progresses. A key factor in this progression is the bidirectional relationship between CKD and gut dysbiosis, which creates an imbalance that promotes the accumulation of uremic toxins (UTs), contributing to renal fibrosis, endothelial dysfunction, and decreased glomerular filtration rate. In addition, CKD itself exacerbates gut dysbiosis by altering the composition of the gut microbiota (GM) and promoting the growth of pathogenic microorganisms. Therefore, it is crucial to explore new therapeutic strategies, and the use of probiotics and synbiotics has shown promise in modulating the GM. Numerous preclinical studies have shown that the use of probiotics in CKD has a beneficial effect on the kidney by reducing UTs, apoptosis, inflammation, and oxidative stress. Probiotic treatment has also been associated with restoration of intestinal integrity, modulation of microbial composition and diversity, and increased production of short-chain fatty acids (SCFAs). These positive results have also been observed in patients at different stages of CKD, where the use of probiotics and/or synbiotics was able to improve creatinine levels and uremic parameters and alleviate abdominal discomfort, in addition to modulating GM and reducing serum endotoxin levels. Although recent studies have explored the benefits of probiotics in the treatment of CKD, further research is needed to determine their long-term efficacy and clinical relevance. This review focuses on the factors driving gut dysbiosis in CKD, its role in disease progression, and the potential of probiotics as a therapeutic strategy.
Collapse
Affiliation(s)
- Fernanda Priscila Barbosa Ribeiro
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I - Jd, Cidade Universitária, João Pessoa, 58051-900, Brazil
| | - Micaelle Oliveira de Luna Freire
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I - Jd, Cidade Universitária, João Pessoa, 58051-900, Brazil
| | - Daniella de Oliveira Coutinho
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I - Jd, Cidade Universitária, João Pessoa, 58051-900, Brazil
| | | | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I - Jd, Cidade Universitária, João Pessoa, 58051-900, Brazil.
| |
Collapse
|
9
|
Barron M, Fernando DG, Atkinson SN, Kirby J, Kindel TL. Sleeve Gastrectomy Protects Against Hypertension in Rats due to Changes in the Gut Microbiome. J Surg Res 2024; 301:118-126. [PMID: 38925098 DOI: 10.1016/j.jss.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/26/2024] [Accepted: 05/11/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Sleeve gastrectomy (SG), results in improvement in hypertension. We have previously published that rodent SG improves hypertension independent of weight loss associated with unique shifts in the gut microbiome. We tested if the gut microbiome directly improves blood pressure by performing fecal material transfer (FMT) from post-SG rats to surgery-naïve animals. METHODS We performed SG or Sham surgery in male, Zucker rats (n = 6-7) with obesity. Stool was collected postop from surgical donors for treatment of recipient rats. Three nonsurgical groups received daily, oral consumption of SG stool, sham stool, or vehicle alone (Nutella) for 10 wk (n = 7-8). FMT treatment was assessed for effects on body weight, food intake, oral glucose tolerance, and blood pressure. Genomic deoxyribonucleic acid of stool from donor and recipient groups were sequenced by 16S ribosomal ribonucleic acid and analyzed for diversity, abundance, and importance. RESULTS Ten weeks of SG-FMT treatment significantly lowered systolic blood pressures in surgery-naïve, recipient rats compared to vehicle treatment alone (126.8 ± 13.3 mmHg versus 151.8 ± 12.2 mmHg, P = 0.001). SG-FMT treatment also significantly altered beta diversity metrics compared to Sham-FMT and vehicle treatment. In random forest analysis, amplicon sequence variant level significantly predicted FMT group, P = 0.01. CONCLUSIONS We have found a direct link between gut microbial changes after SG and regulation of blood pressure. Future mechanistic studies are required to learn what specific gut microbial changes are required to induce improvements in obesity-associated hypertension and translation to clinical, metabolic surgery.
Collapse
Affiliation(s)
| | | | - Samantha N Atkinson
- Department of Microbiology and Immunology, Milwaukee, Wisconsin; Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John Kirby
- Department of Microbiology and Immunology, Milwaukee, Wisconsin
| | | |
Collapse
|
10
|
González-Correa C, Moleón J, Miñano S, Robles-Vera I, Toral M, Barranco AM, Martín-Morales N, O'Valle F, Guerra-Hernández E, Sánchez M, Gómez-Guzmán M, Jiménez R, Romero M, Duarte J. Differing contributions of the gut microbiota to the blood pressure lowering effects induced by first-line antihypertensive drugs. Br J Pharmacol 2024; 181:3420-3444. [PMID: 38770714 DOI: 10.1111/bph.16410] [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: 05/23/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND AND PURPOSE This study analyses whether first-line antihypertensive drugs ameliorate the dysbiosis state in hypertension, and to test if this modification contributes to their blood pressure (BP) lowering properties in a genetic model of neurogenic hypertension. EXPERIMENTAL APPROACH Twenty-week-old male Wistar Kyoto rats (WKY) and spontaneously hypertensive rats (SHR) were untreated or treated with captopril, amlodipine or hydrochlorothiazide. A faecal microbiota transplantation (FMT) experiment was also performed by gavage of faecal content from donor SHR-treated groups to SHR recipients for 3 weeks. KEY RESULTS Faeces from SHR showed gut dysbiosis, characterized by lower acetate- and higher lactate-producing bacteria and lower strict anaerobic bacteria. All three drugs increased the anaerobic bacteria proportion, captopril and amlodipine restored the proportion of acetate-producing bacterial populations to WKY levels, whereas hydrochlorothiazide decreased butyrate-producing bacteria. Captopril and amlodipine decreased gut pathology and permeability and attenuated sympathetic drive in the gut. Both drugs decreased neuroinflammation and oxidative stress in the hypothalamic paraventricular nuclei. Hydrochlorothiazide was unable to reduce neuroinflammation, gut sympathetic tone and gut integrity. FMT from SHR-amlodipine to SHR decreased BP, ameliorated aortic endothelium-dependent relaxation to acetylcholine, lowered NADPH oxidase activity, aortic Th17 infiltration and reduced neuroinflammation, whereas FMT from SHR-hydrochlorothiazide did not have these effects. CONCLUSIONS AND IMPLICATIONS First-line antihypertensive drugs induced different modifications of gut integrity and gut dysbiosis in SHR, which result in no contribution of microbiota in the BP lowering effects of hydrochlorothiazide, whereas the vasculo-protective effect induced by amlodipine involves gut microbiota reshaping and gut-immune system communication.
Collapse
Affiliation(s)
- Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
| | - Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta Toral
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Antonio Manuel Barranco
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
| | | | - Francisco O'Valle
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, Granada, Spain
| | | | - Manuel Sánchez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
| | - Manuel Gómez-Guzmán
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, ibs. GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
11
|
Maia LA, de Souza JR, da Silva LDFR, Magnani M, de Souza EL, de Brito Alves JL. Effects of Probiotics on Inflammatory Biomarkers and Its Associations With Cardiac Autonomic Function in Women With Arterial Hypertension: A Secondary Analysis of a Randomized Clinical Trial. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10303-6. [PMID: 38842655 DOI: 10.1007/s12602-024-10303-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Preclinical evidence suggests that probiotic administration may exert an anti-inflammatory effect and reduce autonomic dysfunction and blood pressure. This study evaluated the effects of probiotic therapy on inflammatory biomarkers and characterized the correlations between inflammation and cardiac autonomic function in women with arterial hypertension. Women were randomized into probiotics (n = 20) or placebo (n = 20). The probiotic group received 109 CFU/day of Lactobacillus (L.) paracasei LPC-37, L. rhamnosus HN001, L. acidophilus NCFM, and Bifidobacterium lactis HN019, and the placebo group received polydextrose. Clinical, electrocardiogram, heart rate variability (HRV) analysis, and cytokine levels were assessed at baseline and after 8 weeks. Women who received probiotics for 8 weeks had increased serum levels of IL-17A (p = 0.02) and decreased INF-γ (p = 0.02) compared to baseline. Probiotic supplementation increased serum levels of IL-10 compared to the placebo group (p = 0.03). Probiotic or placebo administration did not change serum levels of TNFα and IL-6. Serum levels of IL-2 (p = 0.001, and p = 0.001) and IL-4 (p = 0.001, and p = 0.001) were reduced in women receiving placebo or probiotics, respectively. Correlations between HRV indices and inflammatory variables showed that INF-γ was positively correlated with heart rate (HR) and sympathetic HRV indices and negatively correlated with vagal HRV indices. IL-10 was negatively correlated with HR and sympathetic HRV indices. IL-6 was negatively correlated with parasympathetic HRV indices and positively correlated with SD2/SD1 ratio. Probiotic therapy has a discreet anti-inflammatory effect in hypertensive women, and pro-inflammatory cytokines were negatively correlated with vagal modulation and positively correlated with sympathetic modulation of HRV. The clinical trial was registered in the Brazilian Registry of Clinical Trials (ReBEC) with the identification RBR-9mj2dt.
Collapse
Affiliation(s)
- Larissa Araújo Maia
- Department of Nutrition, Health Sciences Center, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | | | | | - Marciane Magnani
- Department of Food Engineering, Technology Center, Federal University of Paraiba, João Pessoa, PB, Brazil
| | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraiba, Joao Pessoa, PB, Brazil.
| |
Collapse
|
12
|
Dosh L, Ghazi M, Haddad K, El Masri J, Hawi J, Leone A, Basset C, Geagea AG, Jurjus R, Jurjus A. Probiotics, gut microbiome, and cardiovascular diseases: An update. Transpl Immunol 2024; 83:102000. [PMID: 38262540 DOI: 10.1016/j.trim.2024.102000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Cardiovascular diseases (CVD) are one of the most challenging diseases and many factors have been demonstrated to affect their pathogenesis. One of the major factors that affect CVDs, especially atherosclerosis, is the gut microbiota (GM). Genetics play a key role in linking CVDs with GM, in addition to some environmental factors which can be either beneficial or harmful. The interplay between GM and CVDs is complex due to the numerous mechanisms through which microbial components and their metabolites can influence CVDs. Within this interplay, the immune system plays a major role, mainly based on the immunomodulatory effects of microbial dysbiosis and its resulting metabolites. The resulting modulation of chronic inflammatory processes was found to reduce the severity of CVDs and to maintain cardiovascular health. To better understand the specific roles of GM-related metabolites in this interplay, this review presents an updated perspective on gut metabolites related effects on the cardiovascular system, highlighting the possible benefits of probiotics in therapeutic strategies.
Collapse
Affiliation(s)
- Laura Dosh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Maya Ghazi
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Karim Haddad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Jad El Masri
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| | - Jihad Hawi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Faculty of Medicine and Medical Sciences, University of Balamand, Al Kurah, Lebanon.
| | - Angelo Leone
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, Palermo, Italy.
| | - Charbel Basset
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, Palermo, Italy.
| | - Alice Gerges Geagea
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rosalyn Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
13
|
Dai Y, Shen Z, Khachatryan LG, Vadiyan DE, Karampoor S, Mirzaei R. Unraveling mechanistic insights into the role of microbiome in neurogenic hypertension: A comprehensive review. Pathol Res Pract 2023; 249:154740. [PMID: 37567034 DOI: 10.1016/j.prp.2023.154740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
Neurogenic hypertension, a complex and multifactorial cardiovascular disorder, is known to be influenced by various genetic, environmental, and lifestyle factors. In recent years, there has been growing interest in the role of the gut microbiome in hypertension pathogenesis. The bidirectional communication between the gut microbiota and the central nervous system, known as the microbiota-gut-brain axis, has emerged as a crucial mechanism through which the gut microbiota exerts its influence on neuroinflammation, immune responses, and blood pressure regulation. Recent studies have shown how the microbiome has a substantial impact on a variety of physiological functions, such as cardiovascular health. The increased sympathetic activity to the gut may cause microbial dysbiosis, increased permeability of the gut, and increased inflammatory reactions by altering a number of intestinal bacteria producing short-chain fatty acids (SCFAs) and the concentrations of lipopolysaccharide (LPS) in the plasma. Collectively, these microbial metabolic and structural compounds stimulate sympathetic stimulation, which may be an important stage in the onset of hypertension. The result is an upsurge in peripheral and central inflammatory response. In addition, it has recently been shown that a link between the immune system and the gut microbiota might play a significant role in hypertension. The therapeutic implications of the gut microbiome including probiotic usage, prebiotics, dietary modifications, and fecal microbiota transplantation in neurogenic hypertension have also been found. A large body of research suggests that probiotic supplementation might help reduce chronic inflammation and hypertension that have an association with dysbiosis in the gut microbiota. Overall, this review sheds light on the intricate interplay between the gut microbiome and neurogenic hypertension, providing valuable insights for both researchers and clinicians. As our knowledge of the microbiome's role in hypertension expands, novel therapeutic strategies and diagnostic biomarkers may pave the way for more effective management and prevention of this prevalent cardiovascular disorder. Exploring the potential of the microbiome in hypertension offers an exciting avenue for future research and offers opportunities for precision medicine and improved patient care.
Collapse
Affiliation(s)
- Yusang Dai
- Physical Examination Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Zheng Shen
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Lusine G Khachatryan
- Department of Pediatric Diseases, N.F. Filatov Clinical Institute of Children's Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia
| | - Diana E Vadiyan
- Institute of Dentistry, Department of Pediatric, Preventive Dentistry and Orthodontics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia
| | - 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.
| |
Collapse
|
14
|
Abstract
Hypertension is a major healthcare issue that afflicts one in every three adults worldwide and contributes to cardiovascular diseases, morbidity and mortality. Bioactive lipids contribute importantly to blood pressure regulation via actions on the vasculature, kidney, and inflammation. Vascular actions of bioactive lipids include blood pressure lowering vasodilation and blood pressure elevating vasoconstriction. Increased renin release by bioactive lipids in the kidney is pro-hypertensive whereas anti-hypertensive bioactive lipid actions result in increased sodium excretion. Bioactive lipids have pro-inflammatory and anti-inflammatory actions that increase or decrease reactive oxygen species and impact vascular and kidney function in hypertension. Human studies provide evidence that fatty acid metabolism and bioactive lipids contribute to sodium and blood pressure regulation in hypertension. Genetic changes identified in humans that impact arachidonic acid metabolism have been associated with hypertension. Arachidonic acid cyclooxygenase, lipoxygenase and cytochrome P450 metabolites have pro-hypertensive and anti-hypertensive actions. Omega-3 fish oil fatty acids eicosapentaenoic acid and docosahexaenoic acid are known to be anti-hypertensive and cardiovascular protective. Lastly, emerging fatty acid research areas include blood pressure regulation by isolevuglandins, nitrated fatty acids, and short chain fatty acids. Taken together, bioactive lipids are key contributors to blood pressure regulation and hypertension and their manipulation could decrease cardiovascular disease and associated morbidity and mortality.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
15
|
Calabrò S, Kankowski S, Cescon M, Gambarotta G, Raimondo S, Haastert-Talini K, Ronchi G. Impact of Gut Microbiota on the Peripheral Nervous System in Physiological, Regenerative and Pathological Conditions. Int J Mol Sci 2023; 24:ijms24098061. [PMID: 37175764 PMCID: PMC10179357 DOI: 10.3390/ijms24098061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
It has been widely demonstrated that the gut microbiota is responsible for essential functions in human health and that its perturbation is implicated in the development and progression of a growing list of diseases. The number of studies evaluating how the gut microbiota interacts with and influences other organs and systems in the body and vice versa is constantly increasing and several 'gut-organ axes' have already been defined. Recently, the view on the link between the gut microbiota (GM) and the peripheral nervous system (PNS) has become broader by exceeding the fact that the PNS can serve as a systemic carrier of GM-derived metabolites and products to other organs. The PNS as the communication network between the central nervous system and the periphery of the body and internal organs can rather be affected itself by GM perturbation. In this review, we summarize the current knowledge about the impact of gut microbiota on the PNS, with regard to its somatic and autonomic divisions, in physiological, regenerative and pathological conditions.
Collapse
Affiliation(s)
- Sonia Calabrò
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Svenja Kankowski
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Kirsten Haastert-Talini
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Center for Systems Neuroscience Hannover (ZSN), Buenteweg 2, 30559 Hannover, Germany
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| |
Collapse
|
16
|
Gómez-Contreras A, Franco-Ávila T, Miró L, Juan ME, Moretó M, Planas JM. Dietary intake of table olives exerts antihypertensive effects in association with changes in gut microbiota in spontaneously hypertensive rats. Food Funct 2023; 14:2793-2806. [PMID: 36861461 DOI: 10.1039/d2fo02928f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Arbequina table olive (AO) consumption lowers blood pressure (BP) in spontaneously hypertensive rats (SHR). This study evaluates whether dietary supplementation with AO induced changes in the gut microbiota that are consistent with the purported antihypertensive effects. Wistar-Kyoto rats (WKY-c) and SHR-c received water, while SHR-o were supplemented by gavage with AO (3.85 g kg-1) for 7 weeks. Faecal microbiota was analysed by 16S rRNA gene sequencing. SHR-c showed increased Firmicutes and decreased Bacteroidetes compared to WKY-c. AO supplementation in SHR-o decreased BP by approximately 19 mmHg, and reduced plasmatic concentrations of malondialdehyde and angiotensin II. Moreover, reshaped faecal microbiota associated with antihypertensive activity by lowering Peptoniphilus and increasing Akkermansia, Sutterella, Allobaculum, Ruminococcus, and Oscillospira. Also promoted the growth of probiotic strains of Lactobacillus and Bifidobacterium and modified the relationship of Lactobacillus with other microorganisms, from competitive to symbiotic. In SHR, AO promotes a microbiota profile compatible with the antihypertensive effects of this food.
Collapse
Affiliation(s)
- Aldo Gómez-Contreras
- Grup de Fisiologia i Nutrició Experimental, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB, Maria de Maeztu Unit of Excellence), Universitat de Barcelona (UB), and Food Innovation Network (XIA), Av. Joan XXIII 27-31, 08028-Barcelona, Spain.
| | - Talia Franco-Ávila
- Grup de Fisiologia i Nutrició Experimental, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB, Maria de Maeztu Unit of Excellence), Universitat de Barcelona (UB), and Food Innovation Network (XIA), Av. Joan XXIII 27-31, 08028-Barcelona, Spain.
| | - Lluïsa Miró
- Grup de Fisiologia i Nutrició Experimental, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB, Maria de Maeztu Unit of Excellence), Universitat de Barcelona (UB), and Food Innovation Network (XIA), Av. Joan XXIII 27-31, 08028-Barcelona, Spain.
| | - M Emília Juan
- Grup de Fisiologia i Nutrició Experimental, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB, Maria de Maeztu Unit of Excellence), Universitat de Barcelona (UB), and Food Innovation Network (XIA), Av. Joan XXIII 27-31, 08028-Barcelona, Spain.
| | - Miquel Moretó
- Grup de Fisiologia i Nutrició Experimental, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB, Maria de Maeztu Unit of Excellence), Universitat de Barcelona (UB), and Food Innovation Network (XIA), Av. Joan XXIII 27-31, 08028-Barcelona, Spain.
| | - Joana M Planas
- Grup de Fisiologia i Nutrició Experimental, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB, Maria de Maeztu Unit of Excellence), Universitat de Barcelona (UB), and Food Innovation Network (XIA), Av. Joan XXIII 27-31, 08028-Barcelona, Spain.
| |
Collapse
|
17
|
Does the Composition of Gut Microbiota Affect Hypertension? Molecular Mechanisms Involved in Increasing Blood Pressure. Int J Mol Sci 2023; 24:ijms24021377. [PMID: 36674891 PMCID: PMC9863380 DOI: 10.3390/ijms24021377] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Arterial hypertension is a chronic disease which is very prevalent contemporarily. The aim of this review was to investigate the impact of gut microbiota on the development and potential treatment of hypertension, taking into consideration underlying molecular mechanisms. The bacteria present in the intestines have the ability to secrete different metabolites, which might play a significant role in the regulation of blood pressure. The most important include short-chain fatty acids (SCFAs), vasoactive hormones, trimethylamine (TMA) and trimethylamine N-oxide (TMAO) and uremic toxins, such as indoxyl sulfate (IS) and p-cresyl sulfate (PCS). Their action in regulating blood pressure is mainly based on their pro- or anti-inflammatory function. The use of specifically formulated probiotics to modify the composition of gut microbiota might be a beneficial way of supportive treatment of hypertension; however, further research on this topic is needed to choose the species of bacteria that could induce the hypotensive pattern.
Collapse
|
18
|
Food Peptides, Gut Microbiota Modulation, and Antihypertensive Effects. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248806. [PMID: 36557936 PMCID: PMC9788432 DOI: 10.3390/molecules27248806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
The gut microbiota is increasingly important in the overall human health and as such, it is a target in the search of novel strategies for the management of metabolic disorders including blood pressure, and cardiovascular diseases. The link between microbiota and hypertension is complex and this review is intended to provide an overview of the mechanism including the production of postbiotics, mitigation of inflammation, and the integration of food biological molecules within this complex system. The focus is on hydrolyzed food proteins and peptides which are less commonly investigated for prebiotic properties. The analysis of available data showed that food peptides are multifunctional and can prevent gut dysbiosis by positively affecting the production of postbiotics or gut metabolites (short-chain fatty acids, polysaccharides, biogenic amines, bile acids). Peptides and the postbiotics then displayed antihypertensive effects via the renin-angiotensin system, the gut barrier, the endothelium, and reduction in inflammation and oxidative stress. Despite the promising antihypertensive effect of the food peptides via the modulation of the gut, there is a lack of human studies as most of the works have been conducted in animal models.
Collapse
|
19
|
Sun D, Xiang H, Yan J, He L. Intestinal microbiota: A promising therapeutic target for hypertension. Front Cardiovasc Med 2022; 9:970036. [PMID: 36457803 PMCID: PMC9705378 DOI: 10.3389/fcvm.2022.970036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/28/2022] [Indexed: 10/29/2023] Open
Abstract
Hypertension has developed into an escalating serious global public health problem with multiple and unclear pathophysiological mechanisms. Recent studies have identified intestinal microbiota as a key perpetrator of hypertension through a variety of mechanisms. In this review, we highlight the potential roles of the intestinal microbiota and its metabolites in the development of hypertension, as well as the therapeutic potential for targeting intestinal microbiomes. We also shed light on the main limitations and challenges of the current research and suggest directions for future investigations. Finally, we discuss the development of accurate and personalized preventive and therapeutic strategies for hypotension by the modulation of intestinal microbes and metabolites.
Collapse
Affiliation(s)
- Dating Sun
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Hui Xiang
- Infectious Disease Department, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liqun He
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| |
Collapse
|
20
|
Chao YM, Tain YL, Lee WC, Wu KLH, Yu HR, Chan JYH. Protection by -Biotics against Hypertension Programmed by Maternal High Fructose Diet: Rectification of Dysregulated Expression of Short-Chain Fatty Acid Receptors in the Hypothalamic Paraventricular Nucleus of Adult Offspring. Nutrients 2022; 14:nu14204306. [PMID: 36296991 PMCID: PMC9609147 DOI: 10.3390/nu14204306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
The role of short-chain fatty acids (SCFAs) in the brain on the developmental programming of hypertension is poorly understood. The present study explored dysregulated tissue levels of SCFAs and expression of SCFA-sensing receptors in the hypothalamic paraventricular nucleus (PVN), a key forebrain region engaged in neural regulation of blood pressure of offspring to maternal high fructose diet (HFD) exposure. We further investigated the engagement of SCFA-sensing receptors in PVN in the beneficial effects of -biotics (prebiotic, probiotic, synbiotic, and postbiotic) on programmed hypertension. Maternal HFD during gestation and lactation significantly reduced circulating butyrate, along with decreased tissue level of butyrate and increased expression of SCFA-sensing receptors, GPR41 and olfr78, and tissue oxidative stress and neuroinflammation in PVN of HFD offspring that were rectified by oral supplement with -biotics. Gene silencing of GPR41 or olfr78 mRNA in PVN also protected adult HFD offspring from programmed hypertension and alleviated the induced oxidative stress and inflammation in PVN. In addition, oral supplement with postbiotic butyrate restored tissue butyrate levels, rectified expressions of GPR41 and olfr78 in PVN, and protected against programmed hypertension in adult HFD offspring. These data suggest that alterations in tissue butyrate level, expression of GPR41 and olfr78, and activation of SCFA-sensing receptor-dependent tissue oxidative stress and neuroinflammation in PVN could be novel mechanisms that underlie hypertension programmed by maternal HFD exposure in adult offspring. Furthermore, oral -biotics supplementation may exert beneficial effects on hypertension of developmental origin by targeting dysfunctional SCFA-sensing receptors in PVN to exert antioxidant and anti-inflammatory actions in the brain.
Collapse
Affiliation(s)
- Yung-Mei Chao
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - You-Lin Tain
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Wei-Chia Lee
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Kay L. H. Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Julie Y. H. Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Correspondence: ; Tel./Fax: +886-7733-8415
| |
Collapse
|
21
|
Pattanakuhar S, Kaewchur T, Saiyasit N, Chattipakorn N, Chattipakorn SC. Level of injury is an independent determining factor of gut dysbiosis in people with chronic spinal cord injury: A cross-sectional study. Spinal Cord 2022; 60:1115-1122. [PMID: 35835855 DOI: 10.1038/s41393-022-00832-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN A cross-sectional study. OBJECTIVE To investigate the correlations between gut microbiota and metabolic parameters in people with different levels of chronic spinal cord injury (SCI). SETTING An SCI-specialized rehabilitation facility in a university hospital. METHODS Forty-three participants with chronic SCI were recruited. Blood samples of each participant were collected for analysis of metabolic parameters. Feces were collected after the bowel opening method the patient routinely uses to evaluate fecal bacterial microbiota using quantitative RT-PCR. Body composition was examined using dual-energy x-ray absorptiometry (DEXA). Data were analyzed to evaluate the correlations between gut microbiota and other parameters. RESULTS Of the 43 participants, 31 people (72.1%) were paraplegic and 12 people (27.9%) tetraplegic. Thirty-two people (74.4%) were diagnosed with obesity using the percentage of body fat (% body fat) criteria. The mean (SD) ratio of Firmicutes:Bacteroides (F/B), which represents the degree of gut dysbiosis, was 18.3 (2.45). Using stepwise multivariable linear regression analysis, both having tetraplegia and being diagnosed with obesity from % body fat evaluated by DEXA were independent positively-correlating factors of F/B (p < 0.001 and p = 0.001, respectively), indicating more severe gut dysbiosis in people with tetraplegia than paraplegia. CONCLUSION In people with chronic SCI, having tetraplegia and being diagnosed with obesity from % body fat evaluated by DEXA are independent positive-correlating factors of gut dysbiosis. These results indicate a significant association between gut microbiota and the characteristics of SCI as well as metabolic parameters.
Collapse
Affiliation(s)
- Sintip Pattanakuhar
- Department of Rehabilitation Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Tawika Kaewchur
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Napatsorn Saiyasit
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand. .,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
22
|
Méndez-Albiñana P, Martínez-González Á, Camacho-Rodríguez L, Ferreira-Lazarte Á, Villamiel M, Rodrigues-Díez R, Balfagón G, García-Redondo AB, Prieto-Nieto MI, Blanco-Rivero J. Supplementation with the Symbiotic Formulation Prodefen® Increases Neuronal Nitric Oxide Synthase and Decreases Oxidative Stress in Superior Mesenteric Artery from Spontaneously Hypertensive Rats. Antioxidants (Basel) 2022; 11:antiox11040680. [PMID: 35453365 PMCID: PMC9029967 DOI: 10.3390/antiox11040680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/23/2022] Open
Abstract
In recent years, gut dysbiosis has been related to some peripheral vascular alterations linked to hypertension. In this work, we explore whether gut dysbiosis is related to vascular innervation dysfunction and altered nitric oxide (NO) production in the superior mesenteric artery, one of the main vascular beds involved in peripheral vascular resistance. For this purpose, we used spontaneously hypertensive rats, either treated or not with the commercial synbiotic formulation Prodefen® (108 colony forming units/day, 4 weeks). Prodefen® diminished systolic blood pressure and serum endotoxin, as well as the vasoconstriction elicited by electrical field stimulation (EFS), and enhanced acetic and butyric acid in fecal samples, and the vasodilation induced by the exogenous NO donor DEA-NO. Unspecific nitric oxide synthase (NOS) inhibitor L-NAME increased EFS-induced vasoconstriction more markedly in rats supplemented with Prodefen®. Both neuronal NO release and neuronal NOS activity were enhanced by Prodefen®, through a hyperactivation of protein kinase (PK)A, PKC and phosphatidylinositol 3 kinase-AKT signaling pathways. The superoxide anion scavenger tempol increased both NO release and DEA-NO vasodilation only in control animals. Prodefen® caused an increase in both nuclear erythroid related factor 2 and superoxide dismutase activities, consequently reducing both superoxide anion and peroxynitrite releases. In summary, Prodefen® could be an interesting non-pharmacological approach to ameliorate hypertension.
Collapse
Affiliation(s)
- Pablo Méndez-Albiñana
- Department of Physiology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.-A.); (Á.M.-G.); (L.C.-R.); (G.B.); (A.B.G.-R.)
- Group of Chemistry and Functionality of Carbohydrates and Derivatives, Food Science Research Institute (CIAL) (CSIC-UAM), 28049 Madrid, Spain; (Á.F.-L.); (M.V.)
| | - Ángel Martínez-González
- Department of Physiology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.-A.); (Á.M.-G.); (L.C.-R.); (G.B.); (A.B.G.-R.)
| | - Laura Camacho-Rodríguez
- Department of Physiology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.-A.); (Á.M.-G.); (L.C.-R.); (G.B.); (A.B.G.-R.)
| | - Álvaro Ferreira-Lazarte
- Group of Chemistry and Functionality of Carbohydrates and Derivatives, Food Science Research Institute (CIAL) (CSIC-UAM), 28049 Madrid, Spain; (Á.F.-L.); (M.V.)
| | - Mar Villamiel
- Group of Chemistry and Functionality of Carbohydrates and Derivatives, Food Science Research Institute (CIAL) (CSIC-UAM), 28049 Madrid, Spain; (Á.F.-L.); (M.V.)
| | - Raquel Rodrigues-Díez
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
- Research Institute University Hospital la Paz (IdIPaz), 28029 Madrid, Spain
- Center for Biomedical Research Network (CIBER) in Cardiovascular Diseases, 28029 Madrid, Spain
| | - Gloria Balfagón
- Department of Physiology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.-A.); (Á.M.-G.); (L.C.-R.); (G.B.); (A.B.G.-R.)
- Research Institute University Hospital la Paz (IdIPaz), 28029 Madrid, Spain
- Center for Biomedical Research Network (CIBER) in Cardiovascular Diseases, 28029 Madrid, Spain
| | - Ana B. García-Redondo
- Department of Physiology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.-A.); (Á.M.-G.); (L.C.-R.); (G.B.); (A.B.G.-R.)
- Research Institute University Hospital la Paz (IdIPaz), 28029 Madrid, Spain
- Center for Biomedical Research Network (CIBER) in Cardiovascular Diseases, 28029 Madrid, Spain
| | - Mª Isabel Prieto-Nieto
- Research Institute University Hospital la Paz (IdIPaz), 28029 Madrid, Spain
- Department of General and Digestive Surgery, Hospital Universitario la Paz, 28046 Madrid, Spain
- Correspondence: (M.I.P.-N.); (J.B.-R.); Tel.: +34-91-497-5446 (J.B.-R.)
| | - Javier Blanco-Rivero
- Department of Physiology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.-A.); (Á.M.-G.); (L.C.-R.); (G.B.); (A.B.G.-R.)
- Research Institute University Hospital la Paz (IdIPaz), 28029 Madrid, Spain
- Center for Biomedical Research Network (CIBER) in Cardiovascular Diseases, 28029 Madrid, Spain
- Correspondence: (M.I.P.-N.); (J.B.-R.); Tel.: +34-91-497-5446 (J.B.-R.)
| |
Collapse
|
23
|
Trimethylamine N-Oxide Promotes Autoimmunity and a Loss of Vascular Function in Toll-like Receptor 7-Driven Lupus Mice. Antioxidants (Basel) 2021; 11:antiox11010084. [PMID: 35052589 PMCID: PMC8773414 DOI: 10.3390/antiox11010084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023] Open
Abstract
Plasma levels of trimethylamine N-oxide (TMAO) are elevated in lupus patients. We analyzed the implication of TMAO in autoimmunity and vascular dysfunction of the murine model of systemic lupus erythematosus (SLE) induced by the activation of the Toll-like receptor (TLR)7 with imiquimod (IMQ). Female BALB/c mice were randomly divided into four groups: untreated control mice, control mice treated with the trimethylamine lyase inhibitor 3,3-dimethyl-1-butanol (DMB), IMQ mice, and IMQ mice treated with DMB. The DMB-treated groups were administered the substance in their drinking water for 8 weeks. Treatment with DMB reduced plasma levels of TMAO in mice with IMQ-induced lupus. DMB prevents the development of hypertension, reduces disease progression (plasma levels of anti-dsDNA autoantibodies, splenomegaly, and proteinuria), reduces polarization of T lymphocytes towards Th17/Th1 in secondary lymph organs, and improves endothelial function in mice with IMQ-induced lupus. The deleterious vascular effects caused by TMAO appear to be associated with an increase in vascular oxidative stress generated by increased NADPH oxidase activity, derived in part from the vascular infiltration of Th17/Th1 lymphocytes, and reduced nrf2-driven antioxidant defense. In conclusion, our findings identified the bacterial-derived TMAO as a regulator of immune system, allowing for the development of autoimmunity and endothelial dysfunction in SLE mice.
Collapse
|
24
|
Gut Microbiota Has a Crucial Role in the Development of Hypertension and Vascular Dysfunction in Toll-like Receptor 7-Driven Lupus Autoimmunity. Antioxidants (Basel) 2021; 10:antiox10091426. [PMID: 34573058 PMCID: PMC8472682 DOI: 10.3390/antiox10091426] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Our group has investigated the involvement of gut microbiota in hypertension in a murine model of systemic lupus erythematosus induced by Toll-like receptor (TLR)-7 activation. Female BALB/c mice were randomly assigned to four experimental groups: an untreated control (CTR), a group treated with the TLR7 agonist imiquimod (IMQ), IMQ-treated with vancomycin, and IMQ-treated with a cocktail of broad-spectrum antibiotics. We carried out faecal microbiota transplant (FMT) from donor CTR or IMQ mice to recipient IMQ or CTR animals, respectively. Vancomycin inhibited the increase in blood pressure; improved kidney injury, endothelial function, and oxidative stress; and reduced T helper (Th)17 infiltration in aortas from IMQ-treated mice. The rise in blood pressure and vascular complications present in IMQ mice were also observed in the CTR mice recipients of IMQ microbiota. Reduced relative populations of Sutterella and Anaerovibrio were associated with high blood pressure in our animals, which were increased after stool transplantation of healthy microbiota to IMQ mice. The reduced endothelium-dependent vasodilator responses to acetylcholine induced by IMQ microbiota were normalized after interleukin-17 neutralization. In conclusion, gut microbiota plays a role in the TLR7-driven increase in Th17 cell, endothelial dysfunction, vascular inflammation, and hypertension. The vascular changes induced by IMQ microbiota were initiated by Th17 infiltrating the vasculature.
Collapse
|
25
|
Chen J, Hall S, Vitetta L. Altered gut microbial metabolites could mediate the effects of risk factors in Covid-19. Rev Med Virol 2021; 31:1-13. [PMID: 34546607 PMCID: PMC7995004 DOI: 10.1002/rmv.2211] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (Covid-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, is now pandemic. While most Covid-19 patients will experience mild symptoms, a small proportion will develop severe disease, which could be fatal. Clinically, Covid-19 patients manifest fever with dry cough, fatigue and dyspnoea, and in severe cases develop into acute respiratory distress syndrome (ARDS), sepsis and multi-organ failure. These severe patients are characterized by hyperinflammation with highly increased pro-inflammatory cytokines including IL-6, IL-17 and TNF-alpha as well as C-reactive protein, which are accompanied by decreased lymphocyte counts. Clinical evidence supports that gut microbiota dysregulation is common in Covid-19 and plays a key role in the pathogenesis of Covid-19. In this narrative review, we summarize the roles of intestinal dysbiosis in Covid-19 pathogenesis and posit that the associated mechanisms are being mediated by gut bacterial metabolites. Based on this premise, we propose possible clinical implications. Various risk factors could be causal for severe Covid-19, and these include advanced age, concomitant chronic disease, SARS-CoV-2 infection of enterocytes, use of antibiotics and psychological distress. Gut dysbiosis is associated with risk factors and severe Covid-19 due to decreased commensal microbial metabolites, which cause reduced anti-inflammatory mechanisms and chronic low-grade inflammation. The preconditioned immune dysregulation enables SARS-CoV-2 infection to progress to an uncontrolled hyperinflammatory response. Thus, a pre-existing gut microbiota that is diverse and abundant could be beneficial for the prevention of severe Covid-19, and supplementation with commensal microbial metabolites may facilitate and augment the treatment of severe Covid-19.
Collapse
Affiliation(s)
| | - Sean Hall
- Research DepartmentMedlab ClinicalSydneyAustralia
| | - Luis Vitetta
- Research DepartmentMedlab ClinicalSydneyAustralia
- Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| |
Collapse
|
26
|
Canale MP, Noce A, Di Lauro M, Marrone G, Cantelmo M, Cardillo C, Federici M, Di Daniele N, Tesauro M. Gut Dysbiosis and Western Diet in the Pathogenesis of Essential Arterial Hypertension: A Narrative Review. Nutrients 2021; 13:1162. [PMID: 33915885 PMCID: PMC8066853 DOI: 10.3390/nu13041162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndrome is a cluster of the most dangerous cardiovascular (CV) risk factors including visceral obesity, insulin resistance, hyperglycemia, alterations in lipid metabolism and arterial hypertension (AH). In particular, AH plays a key role in the complications associated with metabolic syndrome. High salt intake is a well-known risk factor for AH and CV diseases. Vasoconstriction, impaired vasodilation, extracellular volume expansion, inflammation, and an increased sympathetic nervous system (SNS) activity are the mechanisms involved in the pathogenesis of AH, induced by Western diet. Gut dysbiosis in AH is associated with reduction of short chain fatty acid-producing bacteria: acetate, butyrate and propionate, which activate different pathways, causing vasoconstriction, impaired vasodilation, salt and water retention and a consequent high blood pressure. Moreover, increased trimethylamine N-oxide and lipopolysaccharides trigger chronic inflammation, which contributes to endothelial dysfunction and target organs damage. Additionally, a high salt-intake diet impacts negatively on gut microbiota composition. A bidirectional neuronal pathway determines the "brain-gut" axis, which, in turn, influences blood pressure levels. Then, we discuss the possible adjuvant novel treatments related to gut microbiota modulation for AH control.
Collapse
Affiliation(s)
- Maria Paola Canale
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.P.C.); (M.F.)
| | - Annalisa Noce
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.D.L.); (G.M.); (N.D.D.)
| | - Manuela Di Lauro
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.D.L.); (G.M.); (N.D.D.)
| | - Giulia Marrone
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.D.L.); (G.M.); (N.D.D.)
- PhD School of Applied Medical, Surgical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Maria Cantelmo
- School of Specialization in Geriatrics, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Carmine Cardillo
- Department of Internal Medicine and Geriatrics, Policlinico A. Gemelli IRCCS, 00168 Roma, Italy;
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.P.C.); (M.F.)
| | - Nicola Di Daniele
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.D.L.); (G.M.); (N.D.D.)
| | - Manfredi Tesauro
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.D.L.); (G.M.); (N.D.D.)
| |
Collapse
|
27
|
Lin YT, Lin TY, Hung SC, Liu PY, Hung WC, Tsai WC, Tsai YC, Delicano RA, Chuang YS, Kuo MC, Chiu YW, Wu PH. Differences in the Microbial Composition of Hemodialysis Patients Treated with and without β-Blockers. J Pers Med 2021; 11:jpm11030198. [PMID: 33809103 PMCID: PMC8002078 DOI: 10.3390/jpm11030198] [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] [Received: 02/06/2021] [Revised: 02/21/2021] [Accepted: 03/06/2021] [Indexed: 12/23/2022] Open
Abstract
β-blockers are commonly prescribed to treat cardiovascular disease in hemodialysis patients. Beyond the pharmacological effects, β-blockers have potential impacts on gut microbiota, but no study has investigated the effect in hemodialysis patients. Hence, we aim to investigate the gut microbiota composition difference between β-blocker users and nonusers in hemodialysis patients. Fecal samples collected from hemodialysis patients (83 β-blocker users and 110 nonusers) were determined by 16S ribosomal RNA amplification sequencing. Propensity score (PS) matching was performed to control confounders. The microbial composition differences were analyzed by the linear discriminant analysis effect size, random forest, and zero-inflated Gaussian fit model. The α-diversity (Simpson index) was greater in β-blocker users with a distinct β-diversity (Bray-Curtis Index) compared to nonusers in both full and PS-matched cohorts. There was a significant enrichment in the genus Flavonifractor in β-blocker users compared to nonusers in full and PS-matched cohorts. A similar finding was demonstrated in random forest analysis. In conclusion, hemodialysis patients using β-blockers had a different gut microbiota composition compared to nonusers. In particular, the Flavonifractor genus was increased with β-blocker treatment. Our findings highlight the impact of β-blockers on the gut microbiota in hemodialysis patients.
Collapse
Affiliation(s)
- Yi-Ting Lin
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (Y.-T.L.); (Y.-S.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-C.T.); (M.-C.K.); (Y.-W.C.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ting-Yun Lin
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; (T.-Y.L.); (S.-C.H.)
- School of Medicine, Tzu Chi University, Hualien 97071, Taiwan
| | - Szu-Chun Hung
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; (T.-Y.L.); (S.-C.H.)
- School of Medicine, Tzu Chi University, Hualien 97071, Taiwan
| | - Po-Yu Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100225, Taiwan;
| | - Wei-Chun Hung
- Department of Microbiology and Immunology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Wei-Chung Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yi-Chun Tsai
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-C.T.); (M.-C.K.); (Y.-W.C.)
- Division of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Faculty of Renal Care, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | | | - Yun-Shiuan Chuang
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (Y.-T.L.); (Y.-S.C.)
| | - Mei-Chuan Kuo
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-C.T.); (M.-C.K.); (Y.-W.C.)
- Faculty of Renal Care, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yi-Wen Chiu
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-C.T.); (M.-C.K.); (Y.-W.C.)
- Faculty of Renal Care, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ping-Hsun Wu
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-C.T.); (M.-C.K.); (Y.-W.C.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +886-7-3121101
| |
Collapse
|
28
|
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
|
29
|
Higaki A, Mahmoud AUM, Paradis P, Schiffrin EL. Role of interleukin-23/interleukin-17 axis in T-cell-mediated actions in hypertension. Cardiovasc Res 2020; 117:1274-1283. [PMID: 32870976 DOI: 10.1093/cvr/cvaa257] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/01/2020] [Accepted: 08/25/2020] [Indexed: 12/25/2022] Open
Abstract
Current knowledge suggests that hypertension is in part mediated by immune mechanisms. Both interleukin (IL)-23 and IL-17 are up-regulated in several experimental hypertensive rodent models, as well as in hypertensive humans in observational studies. Recent preclinical studies have shown that either IL-23 or IL-17A treatment induce blood pressure elevation. However, the IL-23/IL-17 axis has not been a major therapeutic target in hypertension, unlike in other autoimmune diseases. In this review, we summarize current knowledge on the role of these cytokines in immune mechanisms contributing to hypertension, and discuss the potential of IL-23/IL-17-targeted therapy for treatment of hypertension.
Collapse
Affiliation(s)
| | | | | | - Ernesto L Schiffrin
- Lady Davis Institute for Medical Research.,Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, 3755 Côte-Ste-Catherine Road, Montreal, Quebec H3T 1E2, Canada
| |
Collapse
|
30
|
Magno AL, Herat LY, Kiuchi MG, Schlaich MP, Ward NC, Matthews VB. The Influence of Hypertensive Therapies on Circulating Factors: Clinical Implications for SCFAs, FGF21, TNFSF14 and TNF-α. J Clin Med 2020; 9:jcm9092764. [PMID: 32858953 PMCID: PMC7576485 DOI: 10.3390/jcm9092764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/18/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023] Open
Abstract
Studying the role of circulatory factors in the pathogenesis of diseases has been key to the development of effective therapies. We sought to examine the effect of antihypertensive therapies on numerous circulatory factors including short chain fatty acids and growth factors in a human cohort. A subset of participants from an earlier study was characterized by their hypertensive and/or treatment status and separated into three groups: (i) normotensives; (ii) untreated hypertensive and (iii) treated hypertensive subjects. Circulating levels of short chain fatty acids, FGF21 and TNF superfamily members were measured as part of this study. Both F2-isoprostane and circulating lipid levels were reanalysed as part of this current study. We found that antihypertensive treatment increased butyrate levels and decreased acetate levels to levels similar to normotensives. We also found that antihypertensive treatments reduced levels of circulating FGF21, TNFSF14 and TNF-α. In conclusion, we identified several circulatory factors that are altered in hypertension.
Collapse
Affiliation(s)
- Aaron L. Magno
- Research Centre, Royal Perth Hospital, Perth, WA 6000, Australia;
| | - Lakshini Y. Herat
- Dobney Hypertension Centre, School of Biomedical Science—Royal Perth Hospital Unit, University of Western Australia, Crawley, WA 6009, Australia;
| | - Márcio G. Kiuchi
- Dobney Hypertension Centre, School of Medicine—Royal Perth Hospital Unit, University of Western Australia, Crawley, WA 6009, Australia; (M.G.K.); (M.P.S.)
| | - Markus P. Schlaich
- Dobney Hypertension Centre, School of Medicine—Royal Perth Hospital Unit, University of Western Australia, Crawley, WA 6009, Australia; (M.G.K.); (M.P.S.)
- Department of Cardiology and Department of Nephrology, Royal Perth Hospital, Perth, WA 6000, Australia
| | - Natalie C. Ward
- School of Public Health, Curtin University, Perth, WA 6102, Australia;
- School of Medicine, University of Western Australia, Perth, WA 6009, Australia
| | - Vance B. Matthews
- Dobney Hypertension Centre, School of Biomedical Science—Royal Perth Hospital Unit, University of Western Australia, Crawley, WA 6009, Australia;
- Correspondence: ; Tel.: +61-8-9224-0239
| |
Collapse
|