1
|
Mahmod AI, Govindaraju K, Lokanathan Y, Said NABM, Ibrahim B. Exploring the Potential of Stem Cells in Modulating Gut Microbiota and Managing Hypertension. Stem Cells Dev 2025; 34:99-116. [PMID: 39836384 DOI: 10.1089/scd.2024.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Hypertension, commonly known as high blood pressure, is a significant health issue that increases the risk of cardiovascular diseases, stroke, and renal failure. This condition broadly encompasses both primary and secondary forms. Despite extensive research, the underlying mechanisms of systemic arterial hypertension-particularly primary hypertension, which has no identifiable cause and is affected by genetic and lifestyle agents-remain complex and not fully understood. Recent studies indicate that an imbalance in gut microbiota, referred to as dysbiosis, may promote hypertension, affecting blood pressure regulation through metabolites such as short-chain fatty acids and trimethylamine N-oxide. Current antihypertensive medications face limitations, including resistance and adherence issues, highlighting the need for novel therapeutic approaches. Stem cell therapy, an emerging field in regenerative medicine, shows promise in addressing these challenges. Stem cells, with mesenchymal stem cells being a prime example, have regenerative, anti-inflammatory, and immunomodulatory properties. Emerging research indicates that stem cells can modulate gut microbiota, reduce inflammation, and improve vascular health, potentially aiding in blood pressure management. Research has shown the positive impact of stem cells on gut microbiota in various disorders, suggesting their potential therapeutic role in treating hypertension. This review synthesizes the recent studies on the complex interactions between gut microbiota, stem cells, and systemic arterial hypertension. By offering a thorough analysis of the current literature, it highlights key insights, uncovers critical gaps, and identifies emerging trends that will inform and guide future investigations in this rapidly advancing field.
Collapse
Affiliation(s)
- Asma Ismail Mahmod
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Kayatri Govindaraju
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Nur Akmarina B M Said
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Baharudin Ibrahim
- Department of Clinical Pharmacy and Pharmacy Practices, Faculty of Pharmacy, University Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
2
|
Tang J, Aheyeerke H, Ni X, Cao T, Wang X, Chen S, Han Y, Kong L, Yang X. Directional Temporal Relationship Between Hypertension and Non-Alcoholic Fatty Liver Disease: A Cross-Lagged Cohort Study. J Clin Hypertens (Greenwich) 2025; 27:e70012. [PMID: 39994951 PMCID: PMC11850434 DOI: 10.1111/jch.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/26/2025]
Abstract
The temporal relationship between non-alcoholic fatty liver disease (NAFLD) and hypertension (HTN) remains unclear despite their known association. Using data from the Beijing Health Management Cohort (BHMC) with a 5-year follow-up, we investigated these bidirectional links through Cox proportional hazards regression and a cross-lagged panel model (CLPM), adjusting for confounders. Systolic/diastolic blood pressure (SBP/DBP) and hepatic steatosis index (HSI) were treated as continuous variables to enhance biological interpretability. Cox regression revealed that HTN increased the risk of NAFLD (hazard ratio [HR]: 1.15, 95% confidence interval [CI]: 1.01-1.30, p < 0.05) among participants without NAFLD at baseline, while NAFLD elevated the risk of HTN (HR: 1.11, 95% CI: 1.02-1.21, p < 0.05) among those without HTN at baseline. However, CLPM involving 7349 participants identified a unidirectional temporal relationship from HTN to NAFLD, regression coefficients βSBP2017→HSI2022: 0.036 (95% CI: 0.012, 0.059), βDBP2017→HSI2022: -0.044 (95% CI: -0.068, -0.020), both p < 0.05; but not from NAFLD to HTN, regression coefficients βHSI2017→SBP2022: 0.017 (95% CI: -0.003,0.037), βHSI2017→DBP2022:0.006 (95% CI: -0.016,0.028), both p > 0.05. Overall, our study demonstrates a unidirectional temporal association from HTN to NAFLD. However, a bidirectional relationship was also observed in individuals under 60 years and in those without central obesity. These findings highlight the importance of considering age and central obesity to manage HTN to reduce the risk of future NAFLD and to manage NAFLD to reduce the risk of future HTN.
Collapse
Affiliation(s)
- Jianmin Tang
- School of Public HealthCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Clinical EpidemiologyBeijingChina
| | - Halengbieke Aheyeerke
- School of Public HealthCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Clinical EpidemiologyBeijingChina
| | - Xuetong Ni
- School of Public HealthCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Clinical EpidemiologyBeijingChina
| | - Tengrui Cao
- School of Public HealthCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Clinical EpidemiologyBeijingChina
| | - Xuan Wang
- School of Public HealthCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Clinical EpidemiologyBeijingChina
| | - Shuo Chen
- Beijing Medical Examination CenterBeijingChina
| | - Yumei Han
- Beijing Medical Examination CenterBeijingChina
| | - Linrun Kong
- Beijing Medical Examination CenterBeijingChina
| | - Xinghua Yang
- School of Public HealthCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Clinical EpidemiologyBeijingChina
| |
Collapse
|
3
|
Khalil M, Di Ciaula A, Mahdi L, Jaber N, Di Palo DM, Graziani A, Baffy G, Portincasa P. Unraveling the Role of the Human Gut Microbiome in Health and Diseases. Microorganisms 2024; 12:2333. [PMID: 39597722 PMCID: PMC11596745 DOI: 10.3390/microorganisms12112333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
The human gut is a complex ecosystem that supports billions of living species, including bacteria, viruses, archaea, phages, fungi, and unicellular eukaryotes. Bacteria give genes and enzymes for microbial and host-produced compounds, establishing a symbiotic link between the external environment and the host at both the gut and systemic levels. The gut microbiome, which is primarily made up of commensal bacteria, is critical for maintaining the healthy host's immune system, aiding digestion, synthesizing essential nutrients, and protecting against pathogenic bacteria, as well as influencing endocrine, neural, humoral, and immunological functions and metabolic pathways. Qualitative, quantitative, and/or topographic shifts can alter the gut microbiome, resulting in dysbiosis and microbial dysfunction, which can contribute to a variety of noncommunicable illnesses, including hypertension, cardiovascular disease, obesity, diabetes, inflammatory bowel disease, cancer, and irritable bowel syndrome. While most evidence to date is observational and does not establish direct causation, ongoing clinical trials and advanced genomic techniques are steadily enhancing our understanding of these intricate interactions. This review will explore key aspects of the relationship between gut microbiota, eubiosis, and dysbiosis in human health and disease, highlighting emerging strategies for microbiome engineering as potential therapeutic approaches for various conditions.
Collapse
Affiliation(s)
- Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Laura Mahdi
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Nour Jaber
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Domenica Maria Di Palo
- Division of Hygiene, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, 8055 Graz, Austria;
| | - Gyorgy Baffy
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02130, USA;
- Section of Gastroenterology, Department of Medicine, VA Boston Healthcare System, Boston, MA 02130, USA
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| |
Collapse
|
4
|
Hu M, Du Y, Li W, Zong X, Du W, Sun H, Liu H, Zhao K, Li J, Farooq MZ, Wu J, Xu Q. Interplay of Food-Derived Bioactive Peptides with Gut Microbiota: Implications for Health and Disease Management. Mol Nutr Food Res 2024; 68:e2400251. [PMID: 39097954 DOI: 10.1002/mnfr.202400251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/19/2024] [Indexed: 08/06/2024]
Abstract
Bioactive peptides (BPs) are protein fragments with beneficial effects on metabolism, physiology, and diseases. This review focuses on proteolytic BPs, which are produced by the action of gut microbiota on proteins in food and have demonstrated to influence the composition of gut microbes. And gut microbiota are candidate targets of BPs to alleviate oxidative stress, enhance immunity, and control diseases, including diabetes, hypertension, obesity, cancer, and immune and neurodegenerative diseases. Despite promising results, further research is needed to understand the mechanisms underlying the interactions between BPs and gut microbes, and to identify and screen more BPs for industrial applications. Overall, BPs offer potential as therapeutic agents for various diseases through their interactions with gut microbes, highlighting the importance of continued research in this area.
Collapse
Affiliation(s)
- Mingyang Hu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yufeng Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenyue Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaomei Zong
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjuan Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huizeng Sun
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hongyun Liu
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ke Zhao
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310058, China
| | - Jianxiong Li
- Wuhan Jason Biotech Co., Ltd., Wuhan, 430070, China
| | - Muhammad Zahid Farooq
- Department of Animal Science, University of Veterinary and Animal Science, Lahore, 54000, Pakistan
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-10 Ag/For Building, Edmonton, Alberta, T6G 2P5, Canada
| | - Qingbiao Xu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
5
|
Majumder S, Pushpakumar SB, Almarshood H, Ouseph R, Gondim DD, Jala VR, Sen U. Toll-like receptor 4 mutation mitigates gut microbiota-mediated hypertensive kidney injury. Pharmacol Res 2024; 206:107303. [PMID: 39002869 PMCID: PMC11287947 DOI: 10.1016/j.phrs.2024.107303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Hypertension-associated dysbiosis is linked to several clinical complications, including inflammation and possible kidney dysfunction. Inflammation and TLR4 activation during hypertension result from gut dysbiosis-related impairment of intestinal integrity. However, the contribution of TLR4 in kidney dysfunction during hypertension-induced gut dysbiosis is unclear. We designed this study to address this knowledge gap by utilizing TLR4 normal (TLR4N) and TLR4 mutant (TLR4M) mice. These mice were infused with high doses of Angiotensin-II for four weeks to induce hypertension. Results suggest that Ang-II significantly increased renal arterial resistive index (RI), decreased renal vascularity, and renal function (GFR) in TLR4N mice compared to TLR4M. 16 S rRNA sequencing analysis of gut microbiome revealed that Ang-II-induced hypertension resulted in alteration of Firmicutes: Bacteroidetes ratio in the gut of both TLR4N and TLR4M mice; however, it was not comparably rather differentially. Additionally, Ang-II-hypertension decreased the expression of tight junction proteins and increased gut permeability, which were more prominent in TLR4N mice than in TLR4M mice. Concomitant with gut hyperpermeability, an increased bacterial component translocation to the kidney was observed in TLR4N mice treated with Ang-II compared to TLR4N plus saline. Interestingly, microbiota translocation was mitigated in Ang-II-hypertensive TLR4M mice. Furthermore, Ang-II altered the expression of inflammatory (IL-1β, IL-6) and anti-inflammatory IL-10) markers, and extracellular matrix proteins, including MMP-2, -9, -14, and TIMP-2 in the kidney of TLR4N mice, which were blunted in TLR4M mice. Our data demonstrate that ablation of TLR4 attenuates hypertension-induced gut dysbiosis resulting in preventing gut hyperpermeability, bacterial translocation, mitigation of renal inflammation and alleviation of kidney dysfunction.
Collapse
Affiliation(s)
- Suravi Majumder
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States; Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, United States
| | - Sathnur B Pushpakumar
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, United States
| | - Hebah Almarshood
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, United States
| | - Rosemary Ouseph
- Division of Nephrology and Hypertension, University of Louisville, School of Medicine, Louisville, KY, United States
| | - Dibson D Gondim
- Department of Pathology and Laboratory Medicine, and University of Louisville, School of Medicine, Louisville, KY, United States
| | - Venkatakrishna R Jala
- Department of Microbiology and Immunology, University of Louisville, School of Medicine, Louisville, KY, United States
| | - Utpal Sen
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, United States.
| |
Collapse
|
6
|
Shariff S, Kwan Su Huey A, Parag Soni N, Yahia A, Hammoud D, Nazir A, Uwishema O, Wojtara M. Unlocking the gut-heart axis: exploring the role of gut microbiota in cardiovascular health and disease. Ann Med Surg (Lond) 2024; 86:2752-2758. [PMID: 38694298 PMCID: PMC11060260 DOI: 10.1097/ms9.0000000000001744] [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: 09/23/2023] [Accepted: 12/24/2023] [Indexed: 05/04/2024] Open
Abstract
Introduction Gut microbiota has emerged as a pivotal player in cardiovascular health and disease, extending its influence beyond the gut through intricate metabolic processes and interactions with the immune system. Accumulating evidence supports a significant association between gut microbiota and cardiovascular diseases such as atherosclerosis, hypertension, and heart failure. Dietary patterns have been identified as key factors shaping the composition of the gut microbiota and exerting notable impacts on cardiovascular health. Probiotics and prebiotics have shown promise in mitigating the risks of cardiovascular disease by modulating key cardiovascular parameters. Faecal microbiota transplantation (FMT) has recently emerged as a novel and intriguing therapeutic strategy. Aim This review paper aims to explore and elucidate the multifaceted role of gut microbiota in cardiovascular health. It will also address the prevailing challenges and limitations in gut microbiota studies, emphasizing the importance of future research in overcoming these obstacles to expand our understanding of the gut-heart axis. Materials and methods A comprehensive literature search was conducted using various databases including ClinicalTrials, Google Scholar, PubMed, ScienceDirect, MEDLINE, and Ovid Resources. The search strategy included utilizing keywords such as "Gut microbiota," "Randomized controlled trials (RCTs)," "Gut-heart axis," "Dysbiosis," "Diet," "Probiotics," "Prebiotics," "Faecal Microbiota transplantation," "cardiovascular disease," "Meta-analyses," and other compatible terms thereof. Only articles written in English were considered, and selection criteria included relevance to the research objectives, reasonable sample sizes, and robust methodology. In addition to the identified articles, meta-analyses, animal models and studies, and references from the selected articles were also examined to ensure a comprehensive review of the literature. Results Dietary patterns exert a significant influence on the composition of the gut microbiota, and certain diets, such as the Mediterranean diet, have been associated with a favourable gut microbiota profile and a reduced risk of cardiovascular disease (CVD). Probiotics and prebiotics have emerged as potential interventions to mitigate CVD risks by modulating blood pressure, glycemic control, lipid profiles, and gut dysbiosis. Another innovative therapeutic approach is FMT, which involves transferring faecal material from a healthy donor to restore a balanced gut microbiota. FMT holds promise for improving cardiometabolic parameters in individuals with CVD, although further research is needed to elucidate its precise mechanisms and assess its effectiveness. Conclusion The gut microbiota is emerging as a potential therapeutic target for CVD prevention and management. However, current research has limitations, including the need for larger and more diverse studies, the challenges of establishing causality, and concerns regarding the long-term consequences and safety of gut microbiota modulation. Despite these limitations, understanding the gut-heart axis holds promise for the development of personalized therapies and interventions for cardiovascular health. Further research is needed to expand our knowledge and address the ethical and safety issues associated with gut microbiota modification.
Collapse
Affiliation(s)
- Sanobar Shariff
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Yerevan State Medical University, Yerevan, Armenia
| | - Alicia Kwan Su Huey
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Medical School, University of Glasgow, Glasgow, UK
| | - Nishant Parag Soni
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- GMERS Medical College, Ahmedabad, India
| | - Amer Yahia
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Mohamed 6 of Health Sciences, Casablanca, Morocco
| | - Doha Hammoud
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- National University of Pharmacy, Kharkiv, Ukraine
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Clinton Global Initiative University, New York, NY
- Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Magda Wojtara
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- University of Michigan Medical School; Ann Arbor, MI
| |
Collapse
|
7
|
Liu X, Zeng X, Li X, Xin S, Zhang F, Liu F, Zeng Y, Wu J, Zou Y, Xiong X. Landscapes of gut bacterial and fecal metabolic signatures and their relationship in severe preeclampsia. J Transl Med 2024; 22:360. [PMID: 38632606 PMCID: PMC11022388 DOI: 10.1186/s12967-024-05143-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Preeclampsia is a pregnancy-specific disease leading to maternal and perinatal morbidity. Hypertension and inflammation are the main characteristics of preeclampsia. Many factors can lead to hypertension and inflammation, including gut microbiota which plays an important role in hypertension and inflammation in humans. However, alterations to the gut microbiome and fecal metabolome, and their relationships in severe preeclampsia are not well known. This study aims to identify biomarkers significantly associated with severe preeclampsia and provide a knowledge base for treatments regulating the gut microbiome. METHODS In this study, fecal samples were collected from individuals with severe preeclampsia and healthy controls for shotgun metagenomic sequencing to evaluate changes in gut microbiota composition. Quantitative polymerase chain reaction analysis was used to validate the reliability of our shotgun metagenomic sequencing results. Additionally, untargeted metabolomics analysis was performed to measure fecal metabolome concentrations. RESULTS We identified several Lactobacillaceae that were significantly enriched in the gut of healthy controls, including Limosilactobacillus fermentum, the key biomarker distinguishing severe preeclampsia from healthy controls. Limosilactobacillus fermentum was significantly associated with shifts in KEGG Orthology (KO) genes and KEGG pathways of the gut microbiome in severe preeclampsia, such as flagellar assembly. Untargeted fecal metabolome analysis found that severe preeclampsia had higher concentrations of Phenylpropanoate and Agmatine. Increased concentrations of Phenylpropanoate and Agmatine were associated with the abundance of Limosilactobacillus fermentum. Furthermore, all metabolites with higher abundances in healthy controls were enriched in the arginine and proline metabolism pathway. CONCLUSION Our research indicates that changes in metabolites, possibly due to the gut microbe Limosilactobacillus fermentum, can contribute to the development of severe preeclampsia. This study provides insights into the interaction between gut microbiome and fecal metabolites and offers a basis for improving severe preeclampsia by modulating the gut microbiome.
Collapse
Affiliation(s)
- Xianxian Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Xiaoming Zeng
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Xing Li
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Siming Xin
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Feng Zhang
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Faying Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Yang Zeng
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Jilin Wu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China
| | - Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, 330006, China.
| | - Xinwei Xiong
- Institute of Biological Technology, Nanchang Normal University, Nanchang, Jiangxi, 330032, China.
| |
Collapse
|
8
|
de Luna Freire MO, Cruz Neto JPR, de Albuquerque Lemos DE, de Albuquerque TMR, Garcia EF, de Souza EL, de Brito Alves JL. Limosilactobacillus fermentum Strains as Novel Probiotic Candidates to Promote Host Health Benefits and Development of Biotherapeutics: A Comprehensive Review. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10235-1. [PMID: 38393628 DOI: 10.1007/s12602-024-10235-1] [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: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Fruits and their processing by-products are sources of potentially probiotic strains. Limosilactobacillus (L.) fermentum strains isolated from fruit processing by-products have shown probiotic-related properties. This review presents and discusses the results of the available studies that evaluated the probiotic properties of L. fermentum in promoting host health benefits, their application by the food industry, and the development of biotherapeutics. The results showed that administration of L. fermentum for 4 to 8 weeks promoted host health benefits in rats, including the modulation of gut microbiota, improvement of metabolic parameters, and antihypertensive, antioxidant, and anti-inflammatory effects. The results also showed the relevance of L. fermentum strains for application in the food industry and for the formulation of novel biotherapeutics, especially nutraceuticals. This review provides evidence that L. fermentum strains isolated from fruit processing by-products have great potential for promoting host health and indicate the need for a translational approach to confirm their effects in humans using randomized, double-blind, placebo-controlled trials.
Collapse
Affiliation(s)
- Micaelle Oliveira de Luna Freire
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 58051-900, Brazil
| | - José Patrocínio Ribeiro Cruz Neto
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 58051-900, Brazil
| | | | | | - Estefânia Fernandes Garcia
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 58051-900, Brazil
| | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 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, PB, 58051-900, Brazil.
| |
Collapse
|
9
|
Huang X, Cai H, Zhao Y, Kang Y. The Gut Microbiome and Acute Leukemia: Implications for Early Diagnostic and New Therapies. Mol Nutr Food Res 2024; 68:e2300551. [PMID: 38059888 DOI: 10.1002/mnfr.202300551] [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: 08/02/2023] [Revised: 10/04/2023] [Indexed: 12/08/2023]
Abstract
Acute leukemia (AL), one of the hematological malignancies, shows high heterogeneity. Tremendous progresses are achieved in treating AL with novel targeted drugs and allogeneic hematopoietic stem cell transplantation, there are numerous issues including pathogenesis, early diagnosis, and therapeutic efficacy of AL to be solved. In recent years, an increasing number of studies regarding microbiome have shed more lights on the role of gut microbiota in promoting AL progression. Mechanisms related to the role of gut microbiota in enhancing AL genesis are summarized in the present work, especially on critical pathways like leaky gut, bacterial dysbiosis, microorganism-related molecular patterns, and bacterial metabolites, resulting in AL development. Additionally, the potential of gut microbiota as the biomarker for early AL diagnosis is discussed. It also outlooks therapies targeting gut microbiota for preventing AL development.
Collapse
Affiliation(s)
- Xinwei Huang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Haibo Cai
- Department of Oncology, Yunfeng Hospital, Xuanwei City, Yunnan Province, 655400, China
| | - Yanqin Zhao
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| | - Yongbo Kang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030000, China
| |
Collapse
|
10
|
Almeida C, Gonçalves-Nobre JG, Alpuim Costa D, Barata P. The potential links between human gut microbiota and cardiovascular health and disease - is there a gut-cardiovascular axis? FRONTIERS IN GASTROENTEROLOGY 2023; 2. [DOI: 10.3389/fgstr.2023.1235126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut-heart axis is an emerging concept highlighting the crucial link between gut microbiota and cardiovascular diseases (CVDs). Recent studies have demonstrated that gut microbiota is pivotal in regulating host metabolism, inflammation, and immune function, critical drivers of CVD pathophysiology. Despite a strong link between gut microbiota and CVDs, this ecosystem’s complexity still needs to be fully understood. The short-chain fatty acids, trimethylamine N-oxide, bile acids, and polyamines are directly or indirectly involved in the development and prognosis of CVDs. This review explores the relationship between gut microbiota metabolites and CVDs, focusing on atherosclerosis and hypertension, and analyzes personalized microbiota-based modulation interventions, such as physical activity, diet, probiotics, prebiotics, and fecal microbiota transplantation, as a promising strategy for CVD prevention and treatment.
Collapse
|
11
|
Modulatory effects of Lactiplantibacillus plantarum on chronic metabolic diseases. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
12
|
Mutengo KH, Masenga SK, Mweemba A, Mutale W, Kirabo A. Gut microbiota dependant trimethylamine N-oxide and hypertension. Front Physiol 2023; 14:1075641. [PMID: 37089429 PMCID: PMC10118022 DOI: 10.3389/fphys.2023.1075641] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/31/2023] [Indexed: 04/25/2023] Open
Abstract
The human gut microbiota environment is constantly changing and some specific changes influence the host's metabolic, immune, and neuroendocrine functions. Emerging evidence of the gut microbiota's role in the development of cardiovascular disease (CVD) including hypertension is remarkable. There is evidence showing that alterations in the gut microbiota and especially the gut-dependant metabolite trimethylamine N-oxide is associated with hypertension. However, there is a scarcity of literature addressing the role of trimethylamine N-oxide in hypertension pathogenesis. In this review, we discuss the impact of the gut microbiota and gut microbiota dependant trimethylamine N-oxide in the pathogenesis of hypertension. We present evidence from both human and animal studies and further discuss new insights relating to potential therapies for managing hypertension by altering the gut microbiota.
Collapse
Affiliation(s)
- Katongo H. Mutengo
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Schools of Public Health and Medicine, University of Zambia, Lusaka, Zambia
| | - Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Schools of Public Health and Medicine, University of Zambia, Lusaka, Zambia
| | - Aggrey Mweemba
- Department of Medicine, Levy Mwanawasa Medical University, Lusaka, Zambia
| | - Wilbroad Mutale
- School of Public Health, University of Zambia, Lusaka, Zambia
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
13
|
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
|
14
|
Fan Y, Liang L, Tang X, Zhu J, Mu L, Wang M, Huang X, Gong S, Xu J, Liu T, Zhang T. Changes in the gut microbiota structure and function in rats with doxorubicin-induced heart failure. Front Cell Infect Microbiol 2023; 13:1135428. [PMID: 37180435 PMCID: PMC10173310 DOI: 10.3389/fcimb.2023.1135428] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Objectives The rat model of heart failure (HF) induced by doxorubicin (DOX), a broad spectrum and highly effective chemotherapeutic anthracycline with high-affinity to myocardial tissue that causes severe dose-dependent irreversible cardiotoxicity has been widely recognized and applied in HF pathogenesis and drug therapy studies. The gut microbiota (GM) has attracted significant attention due to its potential role in HF, and research in this area may provide beneficial therapeutic strategies for HF. Considering the differences in the route, mode, and total cumulative dose of DOX administration used to establish HF models, the optimal scheme for studying the correlation between GM and HF pathogenesis remains to be determined. Therefore, focusing on establishing the optimal scheme, we evaluated the correlation between GM composition/function and DOX-induced cardiotoxicity (DIC). Methods Three schemes were investigated: DOX (at total cumulative doses of 12, 15 or 18 mg/kg using a fixed or alternating dose via a tail vein or intraperitoneal injection) was administered to Sprague Dawley (SD) for six consecutive weeks. The M-mode echocardiograms performed cardiac function evaluation. Pathological changes in the intestine were observed by H&E staining and in the heart by Masson staining. The serum levels of N-terminal pre-B-type natriuretic peptide (NT-proBNP) and cardiac troponin I (cTnI) were measured by ELISA. The GM was analysed by 16S rRNA gene sequencing. Key findings Strikingly, based on the severity of cardiac dysfunction, there were marked differences in the abundance and grouping of GM under different schemes. The HF model established by tail vein injection of DOX (18 mg/kg, alternating doses) was more stable; moreover, the degree of myocardial injury and microbial composition were more consistent with the clinical manifestations of HF. Conclusions The model of HF established by tail vein injection of doxorubicin, administered at 4mg/kg body weight (2mL/kg) at weeks 1, 3 and 5, and at 2mg/kg body weight (1mL/kg) at weeks 2, 4 and 6, with a cumulative total dose of 18mg/kg, is a better protocol to study the correlation between HF and GM.
Collapse
Affiliation(s)
- Yawen Fan
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Lichang Liang
- Department of Preventive Treatment, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xinzheng Tang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Jinxian Zhu
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Lei Mu
- Department of Encephalopathy Diseases, Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen, China
| | - Mengni Wang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xuecheng Huang
- Department of Spinal Surgery, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Shenglan Gong
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Jinghan Xu
- Department of Endocrinology, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Tianjiao Liu
- Department of Endocrinology, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Tianfeng Zhang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
- *Correspondence: Tianfeng Zhang,
| |
Collapse
|
15
|
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
|
16
|
Tan YQ, Lin F, Ding YK, Dai S, Liang YX, Zhang YS, Li J, Chen HW. Pharmacological properties of total flavonoids in Scutellaria baicalensis for the treatment of cardiovascular diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154458. [PMID: 36152591 DOI: 10.1016/j.phymed.2022.154458] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/21/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Scutellaria baicalensis, a medicinal herb belonging to the Lamiaceae family, has been recorded in the Chinese, European, and British Pharmacopoeias. The medicinal properties of this plant are attributed to the total flavonoids of Scutellaria baicalensis (TFSB), particularly the main component, baicalin. This study provides a systematic and comprehensive list of the identified TFSB components and their chemical structures. The quality control process, pharmacokinetics, clinical application, and safety of Scutellaria baicalensis are discussed, and its pharmacological effect on cardiovascular diseases (CVDs) is detailed. Finally, the future research trends and prospects of this medicinal plant are provided. METHODS The Chinese and English papers related to TFSB were collected from the PubMed and CNKI databases using the relevant keywords. To highlight the pharmacological mechanism, clinical application, and safety of TFSB, the collected articles were screened and classified based on their research content. RESULTS TFSB contains at least 100 different kinds of flavonoids, of which baicalin, baicalein, wogonin, wogonoside, scutellarin, and scutellarein are the main active ingredients. The preparation process of TFSB is relatively well established, and the extraction rate can be significantly increased by enzymatic pretreatment and ultrasonication. The low oral availability of TFSB may be effectively enhanced using nanoformulations. The available pharmacokinetic data show that flavonoid glycosides and aglycones with the same parent nucleus may be converted to structures that are conducive to absorption in vivo. Moreover, TFSB can protect against CVDs by inhibiting apoptosis, regulating oxidative stress response, participating in inflammatory response, protecting against myocardial fibrosis, inhibiting myocardial hypertrophy, and regulating blood vessels. In terms of clinical application and animal safety, the available studies show that TFSB can be applied in a wide range of clinical treatments and is safe to use is animals. CONCLUSION This article systematically reviews the therapeutic effect and underlying pharmacological mechanism of TFSB against CVDs. The available studies clearly suggest that TFSB has great potential for the treatment of CVDs and is worthy of in-depth research and development.
Collapse
Affiliation(s)
- Yu-Qing Tan
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fei Lin
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100 Henan, China
| | - Yu-Kun Ding
- Beijing University of Chinese Medicine, Beijing 100029, China; Department of Cardiology, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100700, China
| | - Shuang Dai
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ying-Xin Liang
- Traditional Chinese Medicine Orthopedics, Liuzhou Worker's Hospital, Liuzhou 545007, China
| | - Yun-Shu Zhang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Heng-Wen Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
17
|
Bhat MA, Mishra AK, Tantray JA, Alatawi HA, Saeed M, Rahman S, Jan AT. Gut Microbiota and Cardiovascular System: An Intricate Balance of Health and the Diseased State. Life (Basel) 2022; 12:1986. [PMID: 36556351 PMCID: PMC9780831 DOI: 10.3390/life12121986] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
Gut microbiota encompasses the resident microflora of the gut. Having an intricate relationship with the host, it plays an important role in regulating physiology and in the maintenance of balance between health and disease. Though dietary habits and the environment play a critical role in shaping the gut, an imbalance (referred to as dysbiosis) serves as a driving factor in the occurrence of different diseases, including cardiovascular disease (CVD). With risk factors of hypertension, diabetes, dyslipidemia, etc., CVD accounts for a large number of deaths among men (32%) and women (35%) worldwide. As gut microbiota is reported to have a direct influence on the risk factors associated with CVDs, this opens up new avenues in exploring the possible role of gut microbiota in regulating the gross physiological aspects along the gut-heart axis. The present study elaborates on different aspects of the gut microbiota and possible interaction with the host towards maintaining a balance between health and the occurrence of CVDs. As the gut microbiota makes regulatory checks for these risk factors, it has a possible role in shaping the gut and, as such, in decreasing the chances of the occurrence of CVDs. With special emphasis on the risk factors for CVDs, this paper includes information on the prominent bacterial species (Firmicutes, Bacteriodetes and others) towards an advance in our understanding of the etiology of CVDs and an exploration of the best possible therapeutic modules for implementation in the treatment of different CVDs along the gut-heart axis.
Collapse
Affiliation(s)
- Mujtaba Aamir Bhat
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Javeed Ahmad Tantray
- Department of Zoology, Central University of Kashmir, Ganderbal 191131, Jammu and Kashmir, India
| | - Hanan Ali Alatawi
- Department of Biological Sciences, University College of Haqel, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail 55476, Saudi Arabia
| | - Safikur Rahman
- Department of Botany, MS College, BR Ambedkar Bihar University, Muzaffarpur 842001, Bihar, India
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India
| |
Collapse
|
18
|
Xu J, Moore BN, Pluznick JL. Short-Chain Fatty Acid Receptors and Blood Pressure Regulation: Council on Hypertension Mid-Career Award for Research Excellence 2021. Hypertension 2022; 79:2127-2137. [PMID: 35912645 PMCID: PMC9458621 DOI: 10.1161/hypertensionaha.122.18558] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The gut microbiome influences host physiology and pathophysiology through several pathways, one of which is microbial production of chemical metabolites which interact with host signaling pathways. Short-chain fatty acids (SCFAs) are a class of gut microbial metabolites known to activate multiple signaling pathways in the host. Growing evidence indicates that the gut microbiome is linked to blood pressure, that SCFAs modulate blood pressure regulation, and that delivery of exogenous SCFAs lowers blood pressure. Given that hypertension is a key risk factor for cardiovascular disease, the examination of novel contributors to blood pressure regulation has the potential to lead to novel approaches or treatments. Thus, this review will discuss SCFAs with a focus on their host G protein-coupled receptors including GPR41 (G protein-coupled receptor 41), GPR43, and GPR109A, as well as OLFR78 (olfactory receptor 78) and OLFR558. This includes a discussion of the ligand profiles, G protein coupling, and tissue distribution of each receptor. We will also review phenotypes relevant to blood pressure regulation which have been reported to date for Gpr41, Gpr43, Gpr109a, and Olfr78 knockout mice. In addition, we will consider how SCFA signaling influences physiology at baseline, and, how SCFA signaling may contribute to blood pressure regulation in settings of hypertension. In sum, this review will integrate current knowledge regarding how SCFAs and their receptors regulate blood pressure.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Brittni N. Moore
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jennifer L. Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
19
|
Afzaal M, Saeed F, Shah YA, Hussain M, Rabail R, Socol CT, Hassoun A, Pateiro M, Lorenzo JM, Rusu AV, Aadil RM. Human gut microbiota in health and disease: Unveiling the relationship. Front Microbiol 2022; 13:999001. [PMID: 36225386 PMCID: PMC9549250 DOI: 10.3389/fmicb.2022.999001] [Citation(s) in RCA: 194] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/31/2022] [Indexed: 12/04/2022] Open
Abstract
The human gut possesses millions of microbes that define a complex microbial community. The gut microbiota has been characterized as a vital organ forming its multidirectional connecting axis with other organs. This gut microbiota axis is responsible for host-microbe interactions and works by communicating with the neural, endocrinal, humoral, immunological, and metabolic pathways. The human gut microorganisms (mostly non-pathogenic) have symbiotic host relationships and are usually associated with the host’s immunity to defend against pathogenic invasion. The dysbiosis of the gut microbiota is therefore linked to various human diseases, such as anxiety, depression, hypertension, cardiovascular diseases, obesity, diabetes, inflammatory bowel disease, and cancer. The mechanism leading to the disease development has a crucial correlation with gut microbiota, metabolic products, and host immune response in humans. The understanding of mechanisms over gut microbiota exerts its positive or harmful impacts remains largely undefined. However, many recent clinical studies conducted worldwide are demonstrating the relation of specific microbial species and eubiosis in health and disease. A comprehensive understanding of gut microbiota interactions, its role in health and disease, and recent updates on the subject are the striking topics of the current review. We have also addressed the daunting challenges that must be brought under control to maintain health and treat diseases.
Collapse
Affiliation(s)
- Muhammad Afzaal
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
- *Correspondence: Muhammad Afzaal,
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Yasir Abbas Shah
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muzzamal Hussain
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Roshina Rabail
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | | | - Abdo Hassoun
- Sustainable AgriFoodtech Innovation & Research (SAFIR), Arras, France
- Syrian Academic Expertise (SAE), Gaziantep, Turkey
| | - Mirian Pateiro
- Centro Tecnológico de la Carne de Galicia, Ourense, Spain
| | - José M. Lorenzo
- Centro Tecnológico de la Carne de Galicia, Ourense, Spain
- Área de Tecnoloxía dos Alimentos, Faculdade de Ciências de Ourense, Universidade de Vigo, Ourense, Spain
| | - Alexandru Vasile Rusu
- Life Science Institute, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
- Faculty of Animal Science and Biotechnology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Rana Muhammad Aadil
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
- Rana Muhammad Aadil,
| |
Collapse
|
20
|
Jin J, Gao L, Zou X, Zhang Y, Zheng Z, Zhang X, Li J, Tian Z, Wang X, Gu J, Zhang C, Wu T, Wang Z, Zhang Q. Gut Dysbiosis Promotes Preeclampsia by Regulating Macrophages and Trophoblasts. Circ Res 2022; 131:492-506. [PMID: 35950704 DOI: 10.1161/circresaha.122.320771] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Preeclampsia is one of the leading causes of maternal and perinatal morbidity and is characterized by hypertension, inflammation, and placental dysfunction. Gut microbiota plays key roles in inflammation and hypertension. However, its roles and mechanisms in preeclampsia have not been fully elucidated. METHODS 16S rRNA gene sequencing and targeted metabolomics were conducted on stool samples from 92 preeclamptic patients and 86 normal late-pregnant women. Then, fecal microbiota transplantation and in vitro and in vivo functional experiments were performed to explore the roles and mechanisms of gut microbiota in preeclampsia development. RESULTS We revealed the gut microbiota dysbiosis in preeclamptic patients, including significant reductions in short-chain fatty acid-producing bacteria and short-chain fatty acids. The gut microbiota of preeclamptic patients significantly exacerbated pathologies and symptoms of preeclamptic rats, whereas the gut microbiota of healthy pregnant women had significant protective effects. Akkermansia muciniphila, propionate, or butyrate significantly alleviated the symptoms of preeclamptic rats. Mechanistically, they significantly promoted autophagy and M2 polarization of macrophages in placental bed, thereby suppressing inflammation. Propionate also significantly promoted trophoblast invasion, thereby improved spiral arterial remodeling. Additionally, we identified a marker set consisting of Akkermansia, Oscillibacter, and short-chain fatty acids that could accurately diagnose preeclampsia. CONCLUSIONS Our study revealed that gut microbiota dysbiosis is an important etiology of preeclampsia. Gut microbiota and their active metabolites have great potential for the treatment and diagnosis of preeclampsia. Our findings enrich the gut-placenta axis theory and contribute to the development of microecological products for preeclampsia.
Collapse
Affiliation(s)
- Jiajia Jin
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.)
| | - Liaomei Gao
- Division of Geriatrics, Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China (L.G., J.L., J.G., Z.W.).,Maternal and child health care hospital of Shandong province, Shandong University, Jinan, China (L.G.)
| | - Xiuli Zou
- Intensive Care Unit, Liaocheng People's Hospital, China (X.Z., T.W.)
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.)
| | - Zhijian Zheng
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.)
| | - Xinjie Zhang
- Department of Biology, University College London, United Kingdom (X.Z.)
| | - Jiaxuan Li
- Division of Geriatrics, Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China (L.G., J.L., J.G., Z.W.)
| | - Zhenyu Tian
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.)
| | - Xiaowei Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.)
| | - Junfei Gu
- Division of Geriatrics, Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China (L.G., J.L., J.G., Z.W.)
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.)
| | - Tiejun Wu
- Intensive Care Unit, Liaocheng People's Hospital, China (X.Z., T.W.)
| | - Zhe Wang
- Division of Geriatrics, Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, China (L.G., J.L., J.G., Z.W.)
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China (J.J., Y.Z., Z.Z., Z.T., X.W., C.Z., Q.Z.).,Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China (Q.Z.)
| |
Collapse
|
21
|
Ghorbani Z, Kazemi A, Bartolomaeus TUP, Martami F, Noormohammadi M, Salari A, Löber U, Balou HA, Forslund SK, Mahdavi-Roshan M. The effect of probiotic and synbiotic supplementation on lipid parameters among patients with cardiometabolic risk factors: a systematic review and meta-analysis of clinical trials. Cardiovasc Res 2022; 119:933-956. [PMID: 35934838 DOI: 10.1093/cvr/cvac128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
Although the available evidence emphasizes the beneficial effects of probiotics in normalizing various cardiometabolic markers, there is still substantial uncertainty in this regard. Thus, we set out to determine the effect sizes of probiotics on blood lipid parameters more coherently. A systematic literature search of the Medline (PubMed) and Scopus databases was conducted from inception to February 12, 2021, applying both MeSH terms and free text terms to find the relevant randomized controlled trials (RCTs). The meta-analysis was conducted based on a random-effect model to calculate the mean effect sizes demonstrated as weighted mean differences (WMD) and the 95% confidence intervals (95%CI). To explore the heterogeneity, the Cochrane Chi-squared test, and analysis of Galbraith plots were performed. Meta-analysis of data from 40 RCTs (n = 2795) indicated a significant decrease in serum/plasma triglyceride (WMD (95%CI) -12.26 (-17.11- -7.41) mg/dL; P-value <0.001; I2 (%)= 29.9; P heterogeneity = 0.034)), total cholesterol (with high heterogeneity) (WMD (95%CI) -8.43 (-11.90- -4.95) mg/dL; P-value <0.001; I2 (%) =56.8; P heterogeneity < 0.001), LDL-C (WMD (95%CI) -5.08 (-7.61, -2.56) mg/dL; P-value <0.001; I2 (%) =42.7; P heterogeneity =0.002), and HDL-C (with high heterogeneity) (WMD (95%CI) 1.14 (0.23, 2.05) mg/dL; P-value =0.014; I2 (%) = 59.8; P heterogeneity < 0.001) following receiving probiotic/synbiotic supplements. Collectively, the current preliminary evidence supports the effectiveness of probiotics/synbiotics in improving dyslipidemia and various lipid parameters more prominently among subjects with hyperlipidemia, diabetes, and metabolic syndrome. However, large and well conducted RCTs are required to provide further convincing support for these results.
Collapse
Affiliation(s)
- Zeinab Ghorbani
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Asma Kazemi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Theda U P Bartolomaeus
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany
| | - Fahimeh Martami
- School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Morvarid Noormohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ulrike Löber
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany
| | - Heydar Ali Balou
- Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sofia K Forslund
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany.,Structural and Computational Biology Unit, European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany
| | - Marjan Mahdavi-Roshan
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
22
|
Yan D, Sun Y, Zhou X, Si W, Liu J, Li M, Wu M. Regulatory effect of gut microbes on blood pressure. Animal Model Exp Med 2022; 5:513-531. [PMID: 35880388 PMCID: PMC9773315 DOI: 10.1002/ame2.12233] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/25/2022] [Indexed: 12/30/2022] Open
Abstract
Hypertension is an important global public health issue because of its high morbidity as well as the increased risk of other diseases. Recent studies have indicated that the development of hypertension is related to the dysbiosis of the gut microbiota in both animals and humans. In this review, we outline the interaction between gut microbiota and hypertension, including gut microbial changes in hypertension, the effect of microbial dysbiosis on blood pressure (BP), indicators of gut microbial dysbiosis in hypertension, and the microbial genera that affect BP at the taxonomic level. For example, increases in Lactobacillus, Roseburia, Coprococcus, Akkermansia, and Bifidobacterium are associated with reduced BP, while increases in Streptococcus, Blautia, and Prevotella are associated with elevated BP. Furthermore, we describe the potential mechanisms involved in the regulation between gut microbiota and hypertension. Finally, we summarize the commonly used treatments of hypertension that are based on gut microbes, including fecal microbiota transfer, probiotics and prebiotics, antibiotics, and dietary supplements. This review aims to find novel potential genera for improving hypertension and give a direction for future studies on gut microbiota in hypertension.
Collapse
Affiliation(s)
- Dong Yan
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Ye Sun
- Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical CenterPeking Union Medical CollegeBeijingChina
| | - Xiaoyue Zhou
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Wenhao Si
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina,Department of Dermatologythe First Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| | - Jieyu Liu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Min Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| | - Minna Wu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical SciencesXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
23
|
Tegegne BA, Kebede B. Probiotics, their prophylactic and therapeutic applications in human health development: A review of the literature. Heliyon 2022; 8:e09725. [PMID: 35785237 PMCID: PMC9240980 DOI: 10.1016/j.heliyon.2022.e09725] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/03/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023] Open
Abstract
Antibiotics do not differentiate between good and bad germs, disrupting normal microflora and causing vitamin deficiency in the human body. They also kill healthy bacteria in the gut and genital tract on a large scale, weakening the host's defense mechanism. Probiotics are a colony of bacteria that live in our intestines and are regarded as a metabolic 'organ' due to their beneficial effects on human health, including metabolism and immunological function. They are used in clinical settings to prevent and treat conditions such as diarrhoea, colon cancer, hypertension, diabetes, acute pancreatitis, Helicobacter pylori infection, ventilator-associated pneumonia, migraine and autism. Probiotics may modify immunological activity by increasing innate and adaptive immune responses, altering microbial habitat in the intestine, improving gut barrier function, competitive adherence to the mucosa and epithelium, and producing antimicrobial compounds. The aim of this study is to index that further in depth researches to be conducted on probiotics pivotal role in the prophylaxis and therapeutic usage for a variety of disease that may or may not have treatment alternatives. Key words such as probiotics, microbiota, prophylactics, and therapeutic applications were searched extensively in research databases such as PubMed, PubMed Central (PMC), Scopus, Web of Science, Research Gate, Google Scholar, and Cochrane Library. This concise narrative review article summarized primarily the history, selection, mechanism/mode of action, recent advances in prophylactic and therapeutic applications, and future directions in the use of probiotics for prophylactic and therapeutic applications.
Collapse
|
24
|
The Effects of Probiotics on Inflammation, Endothelial Dysfunction, and Atherosclerosis Progression: A Mechanistic Overview. Heart Lung Circ 2022; 31:e45-e71. [PMID: 35153150 DOI: 10.1016/j.hlc.2021.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/07/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The relationship between the intestinal microbiota dysbiosis, inflammation, and cardiovascular disorders (CVDs) has become evident, based on a growing body of literature from animal models and human studies. On the other hand, probiotics are believed to have promising effects on modifying dysbiosis and protecting against CVDs. OBJECTIVE This narrative review provides an overview of the link between gut microbiota, inflammation, endothelial dysfunction, and atherosclerosis. The influences of probiotic supplementation on biomarkers contributing to these conditions as the primary underlying risk factors for developing CVDs are also discussed. METHODS An up-to-date review was performed of the available evidence from experimental studies, clinical trials, and meta-analyses, considering their challenges and limitations. It also aimed to provide mechanistic insight into the likely mechanisms of probiotics that could prevent atherosclerosis initiation and progression. RESULTS Probiotic supplementation seems to be associated with reduced levels of inflammation and oxidative stress biomarkers (C-reactive protein, tumour necrosis factor-α, interleukin (IL)-6, IL-12, and malondialdehyde). Further, these agents might enhance antioxidant factors (IL-10, total antioxidant status, total antioxidant capacity, glutathione, and nitric oxide). Probiotics also appear to improve intestinal barrier integrity, reduce leakage of harmful metabolites (e.g., lipopolysaccharides), inhibit pro-inflammatory signalling pathways, and possibly suppress the formation of trimethylamine/trimethylamine oxide. Probiotics have also been found to enhance endothelial function and halter thrombosis. CONCLUSION The current clinical evidence underlines belief that probiotics might be associated with reduced levels of inflammation biomarkers. Experimental evidence reports that the beneficial effects of probiotics seem to be mainly imposed by triggering the secretion of short-chain fatty acids and bile acids, in addition to suppressing the NF-κB signalling pathway. However, the current studies are still in their infancy and it is of high priority to design further research on the topic.
Collapse
|
25
|
Kang Y, Cai Y, Yang Y. The Gut Microbiome and Hepatocellular Carcinoma: Implications for Early Diagnostic Biomarkers and Novel Therapies. Liver Cancer 2022; 11:113-125. [PMID: 35634424 PMCID: PMC9109080 DOI: 10.1159/000521358] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/04/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranks the third place among all causes inducing cancer-associated mortality, worldwide. HCC nearly exclusively occurs in cases suffering from chronic liver disease (CLD), which results from the vicious cycle of liver damage, inflammation, and regeneration possibly lasting for dozens of years. Recently, more and more investigation on microbiome-gut-liver axis enhances our understanding toward how gut microbiota promotes liver disease and even HCC development. In this review, we summarize the mechanisms underlying the effect of gut microbiota on promoting HCC occurrence, with the focus on key pathways such as bacterial dysbiosis, leaky gut, bacterial metabolites, and microorganism-related molecular patterns, which promote liver inflammation, genotoxicity, and fibrosis that finally lead to cancer occurrence. Furthermore, we discuss gut microbiota's important potential to be the early diagnostic biomarker for HCC. Gut microbiota may be the candidate targets to simultaneously prevent CLD and HCC occurrence among advanced liver disease cases. We outlook the gut microbiota-targeting treatments in detail to prevent CLD and HCC progression.
Collapse
Affiliation(s)
- Yongbo Kang
- Department of Endocrinology, Affiliated Hospital of Yunnan University, Kunming, China
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yue Cai
- Department of Endocrinology, Affiliated Hospital of Yunnan University, Kunming, China
| | - Ying Yang
- Department of Endocrinology, Affiliated Hospital of Yunnan University, Kunming, China
| |
Collapse
|
26
|
Georgescu D, Caraba A, Ionita I, Lascu A, Hut EF, Dragan S, Ancusa OE, Suceava I, Lighezan D. Dyspepsia and Gut Microbiota in Female Patients with Postcholecystectomy Syndrome. Int J Womens Health 2022; 14:41-56. [PMID: 35136356 PMCID: PMC8816732 DOI: 10.2147/ijwh.s342882] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/06/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gallstone disease (GSD) represents one of the most frequent digestive disorders, highly reported in female gender. The purpose of the study was to explore the clinical and gut microbiota particularities of female patients with postcholecystectomy syndrome (PCS) and the possible relationship between gut dysbiosis (DB) and abdominal complaints. PATIENTS AND METHODS In total, 129 female participants: 104 outpatients divided into two equal groups, 52 PCS (+), 52 PCS (-) and 25 healthy controls were consecutively enrolled in this observational study. Patients underwent clinical examination with assessment of pain, bloating, transit disturbances, abdominal ultrasound/computer tomography/magnetic resonance imaging/endoscopic retrograde cholangiopancreatography, upper and lower digestive endoscopies. Laboratory work-ups and stool microbiology assessments were performed for all study participants (patients and controls). Stool microorganisms were identified by matrix-assisted laser desorption ionization - time-of-flight- mass spectrometry and in patients with DB also by next-generation sequencing. RESULTS Older age, complicated gallstones disease, associated conditions like diabetes mellitus/impaired glucose tolerance and irritable bowel syndrome were significantly present in PCS (+) group, as well as sedentary lifestyle and diets characterized by a low fiber intake (p<0.0001). PCS (+) patients displayed significant differences related to the incidence and severity of overall gut microbiota DB, decreased H index of biodiversity and the unbalanced Firmicutes/Bacteroidetes (F/B) ratios by comparison to the PCS (-) group (p<0.0001). Strong positive correlations of the severity of overall DB with bloating and the intestinal habit disorders, as well as of F/B ratios to all abdominal symptoms were noted. CONCLUSION PCS in female patients was associated with older age, sedentary lifestyle, specific dietary habits, history of complicated gallstone disease, diabetes mellitus/impaired glucose tolerance and irritable bowel syndrome, as well as gut microbiota particularities. Overall DB and unbalanced F/B ratios were strongly correlated to abdominal complaints.
Collapse
Affiliation(s)
- Doina Georgescu
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Alexandru Caraba
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ioana Ionita
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ana Lascu
- Department of Functional Sciences, Discipline of Pathophysiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Emil Florin Hut
- Department IX of Surgery I/Compartment of Hepato-Bilio-Pancreatic Surgery, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Simona Dragan
- Department of Cardiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Oana Elena Ancusa
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ioana Suceava
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Daniel Lighezan
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
27
|
Kang Y, Kang X, Cai Y. The gut microbiome as a target for adjuvant therapy in insomnia disorder. Clin Res Hepatol Gastroenterol 2022; 46:101834. [PMID: 34800683 DOI: 10.1016/j.clinre.2021.101834] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023]
Abstract
Insomnia is a type of sleep disorder which has negative impacts on the quality of life, mood, cognitive function and health of humans. The etiology of insomnia may be related to many factors such as genetics, biochemistry, neuroendocrine, immune, and psychosocial factors. However, the detailed pathological aspects of insomnia remain unclear. Recent investigation of the microbiome-gut-brain axis enhances our understanding of the role of the gut microbiota in brain-related diseases. Gut microbiome has been shown to be associated with insomnia. However, the available data in this field remain limited and the relevant scientific work has only recently begun. This review aims to summarize the recent literature as an aid to better understanding how the alteration of gut microbiota composition contributes to insomnia while evaluating and prospecting the therapeutic effect of modulating gut microbiota in the treatment of insomnia based on previous publications.
Collapse
Affiliation(s)
- Yongbo Kang
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Xing Kang
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yue Cai
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
28
|
Probiotics supplementation and cardiometabolic risk factors: A new insight into recent advances, potential mechanisms, and clinical implications. PHARMANUTRITION 2021. [DOI: 10.1016/j.phanu.2021.100261] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
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
|
30
|
Gutiérrez-Cuevas J, Sandoval-Rodriguez A, Meza-Rios A, Monroy-Ramírez HC, Galicia-Moreno M, García-Bañuelos J, Santos A, Armendariz-Borunda J. Molecular Mechanisms of Obesity-Linked Cardiac Dysfunction: An Up-Date on Current Knowledge. Cells 2021; 10:629. [PMID: 33809061 PMCID: PMC8000147 DOI: 10.3390/cells10030629] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is defined as excessive body fat accumulation, and worldwide obesity has nearly tripled since 1975. Excess of free fatty acids (FFAs) and triglycerides in obese individuals promote ectopic lipid accumulation in the liver, skeletal muscle tissue, and heart, among others, inducing insulin resistance, hypertension, metabolic syndrome, type 2 diabetes (T2D), atherosclerosis, and cardiovascular disease (CVD). These diseases are promoted by visceral white adipocyte tissue (WAT) dysfunction through an increase in pro-inflammatory adipokines, oxidative stress, activation of the renin-angiotensin-aldosterone system (RAAS), and adverse changes in the gut microbiome. In the heart, obesity and T2D induce changes in substrate utilization, tissue metabolism, oxidative stress, and inflammation, leading to myocardial fibrosis and ultimately cardiac dysfunction. Peroxisome proliferator-activated receptors (PPARs) are involved in the regulation of carbohydrate and lipid metabolism, also improve insulin sensitivity, triglyceride levels, inflammation, and oxidative stress. The purpose of this review is to provide an update on the molecular mechanisms involved in obesity-linked CVD pathophysiology, considering pro-inflammatory cytokines, adipokines, and hormones, as well as the role of oxidative stress, inflammation, and PPARs. In addition, cell lines and animal models, biomarkers, gut microbiota dysbiosis, epigenetic modifications, and current therapeutic treatments in CVD associated with obesity are outlined in this paper.
Collapse
Affiliation(s)
- Jorge Gutiérrez-Cuevas
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Ana Sandoval-Rodriguez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Alejandra Meza-Rios
- Tecnologico de Monterrey, Campus Guadalajara, Zapopan, School of Medicine and Health Sciences, Jalisco 45201, Mexico; (A.M.-R.); (A.S.)
| | - Hugo Christian Monroy-Ramírez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Marina Galicia-Moreno
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Jesús García-Bañuelos
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
| | - Arturo Santos
- Tecnologico de Monterrey, Campus Guadalajara, Zapopan, School of Medicine and Health Sciences, Jalisco 45201, Mexico; (A.M.-R.); (A.S.)
| | - Juan Armendariz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Jalisco 44340, Mexico; (J.G.-C.); (A.S.-R.); (H.C.M.-R.); (M.G.-M.); (J.G.-B.)
- Tecnologico de Monterrey, Campus Guadalajara, Zapopan, School of Medicine and Health Sciences, Jalisco 45201, Mexico; (A.M.-R.); (A.S.)
| |
Collapse
|
31
|
Grylls A, Seidler K, Neil J. Link between microbiota and hypertension: Focus on LPS/TLR4 pathway in endothelial dysfunction and vascular inflammation, and therapeutic implication of probiotics. Biomed Pharmacother 2021; 137:111334. [PMID: 33556874 DOI: 10.1016/j.biopha.2021.111334] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
High blood pressure (BP) presents a significant public health challenge. Recent findings suggest that altered microbiota can exert a hypertensive effect on the host. One of the possible mechanisms involved is the chronic translocation of its components, mainly lipopolysaccharides (LPS) into systemic circulation leading to metabolic endotoxemia. In animal models, LPS has been commonly used to induce endothelial dysfunction and vascular inflammation. In human studies, plasma LPS concentration has been positively correlated with hypertension, however, the mechanistic link has not been fully elucidated. It is hypothesised here that the LPS-induced direct alterations to the vascular endothelium and resulting hypertension are possible targets for probiotic intervention. The methodology of this review involved a systematic search of the literature with critical appraisal of papers. Three tranches of search were performed: 1) existing review papers; 2) primary mechanistic animal, in vitro and human studies; and 3) primary intervention studies. A total of 70 peer-reviewed papers were included across the three tranches and critically appraised using SIGN50 for human studies and the ARRIVE guidelines for animal studies. The extracted information was coded into key themes and summarized in a narrative analysis. Results highlight the role of LPS in the activation of endothelial toll-like receptor 4 (TLR4) initiating a cascade of interrelated signalling pathways including: 1) Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase/ Reactive oxygen species (ROS)/ Endothelial nitric oxide synthase (eNOS) pathway leading to endothelial dysfunction; and 2) Mitogen-Activated Protein Kinase (MAPK) and Nuclear factor kappa B (NF-κB) pathways leading to vascular inflammation. Findings from animal intervention studies suggest an improvement in vasorelaxation, vascular inflammation and hypertension following probiotic supplementation, which was mediated by downregulation of LPS-induced pathways. Randomised controlled trials (RCTs) and systematic reviews provided some evidence for the anti-inflammatory effect of probiotics with statistically significant antihypertensive effect in clinical samples and may offer a viable intervention for the management of hypertension.
Collapse
Affiliation(s)
- Alina Grylls
- Centre for Nutrition Education and Lifestyle Management, Chapel Gardens, 14 Rectory Road, Wokingham RG40 1DH, England, United Kingdom.
| | - Karin Seidler
- Centre for Nutrition Education and Lifestyle Management, Chapel Gardens, 14 Rectory Road, Wokingham RG40 1DH, England, United Kingdom
| | - James Neil
- Centre for Nutrition Education and Lifestyle Management, Chapel Gardens, 14 Rectory Road, Wokingham RG40 1DH, England, United Kingdom
| |
Collapse
|
32
|
Wang JM, Yang MX, Wu QF, Chen J, Deng SF, Chen L, Wei DN, Liang FR. Improvement of intestinal flora: accompany with the antihypertensive effect of electroacupuncture on stage 1 hypertension. Chin Med 2021; 16:7. [PMID: 33413552 PMCID: PMC7792359 DOI: 10.1186/s13020-020-00417-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 12/22/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increasing evidence have indicated the relationship between intestinal dysbiosis and hypertension. We aimed to evaluate the effect of the electroacupuncture (EA) on intestinal microbiota in patients with stage 1 hypertension. METHODS 93 hypertensive patients and 15 healthy subjects were enrolled in this study. Applying a highly accurate oscillometric device to evaluate the antihypertensive effect of EA. 16S rRNA sequencing was used to profile stool microbial communities from Healthy group, Before treatment (BT) group and After treatment (AT) group, and various multivariate analysis approaches were used to assess diversity, composition and abundance of intestinal microbiota. RESULTS In this study, EA significantly decreased the blood pressure (BP) of hypertensive patients. Higher abundance of Firmicutes and lower Bacteroidetes abundance were observed in the BT group compared to the Healthy group. And EA treatment significantly decreased the Firmicutes/Bacteroidetes ratio compared to the BT group. Moreover, at the genus level, there was an increased abundance of Escherichia-Shigella in patients with hypertension, while Blautia were decreased, and EA reversed these changes. CONCLUSIONS Our study indicates that EA can effectively lower BP and improve the structure of intestinal microbiota which are correlate with the alteration of blood pressure by electroacupuncture. TRIAL REGISTRATION Clinicaltrial.gov, NCT01701726. Registered 5 October 2012, https://clinicaltrials.gov/ct2/show/study/NCT01701726.
Collapse
Affiliation(s)
- Jun-Meng Wang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Ming-Xiao Yang
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, SAR, China.,Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Hai yuan Road, Futian District, Shenzhen, 518053, Guangdong, China
| | - Qiao-Feng Wu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China. .,Institute of Acupuncture and Homeostasis Regulation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| | - Ji Chen
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Shu-Fang Deng
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Lin Chen
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Da-Neng Wei
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Fan-Rong Liang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China.
| |
Collapse
|
33
|
Kim E, Cho S. CNS and peripheral immunity in cerebral ischemia: partition and interaction. Exp Neurol 2021; 335:113508. [PMID: 33065078 PMCID: PMC7750306 DOI: 10.1016/j.expneurol.2020.113508] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/28/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
Stroke elicits excessive immune activation in the injured brain tissue. This well-recognized neural inflammation in the brain is not just an intrinsic organ response but also a result of additional intricate interactions between infiltrating peripheral immune cells and the resident immune cells in the affected areas. Given that there is a finite number of immune cells in the organism at the time of stroke, the partitioned immune systems of the central nervous system (CNS) and periphery must appropriately distribute the limited pool of immune cells between the two domains, mounting a necessary post-stroke inflammatory response by supplying a sufficient number of immune cells into the brain while maintaining peripheral immunity. Stroke pathophysiology has mainly been neurocentric in focus, but understanding the distinct roles of the CNS and peripheral immunity in their concerted action against ischemic insults is crucial. This review will discuss stroke-induced influences of the peripheral immune system on CNS injury/repair and of neural inflammation on peripheral immunity, and how comorbidity influences each.
Collapse
Affiliation(s)
- Eunhee Kim
- Vivian L. Smith Department of Neurosurgery at University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Sunghee Cho
- Burke Neurological Institute, White Plains, NY, United States of America; Feil Brain Mind Research Institute, Weill Cornell Medicine, New York, NY, United States of America.
| |
Collapse
|
34
|
Kang Y, Kang X, Zhang H, Liu Q, Yang H, Fan W. Gut Microbiota and Parkinson's Disease: Implications for Faecal Microbiota Transplantation Therapy. ASN Neuro 2021; 13:17590914211016217. [PMID: 34053243 PMCID: PMC8165863 DOI: 10.1177/17590914211016217] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) ranks the second place among neurodegenerative diseases in terms of its morbidity, which affects 1-2% people aged over 65 years. In addition to genetics, some environmental factors may exert vital parts in PD occurrence as well. At present, more and more studies are conducted to elucidate the association between gut microbial dysbiosis and the incidence of PD. Gut microbial dysbiosis has a certain effect on both the central nervous system (CNS) and the enteric nervous system (ENS), which indicates that there is a gut-microbiota-brain axis that induces CNS disorders. Some gut microbial strains are suggested to suppress or weaken the neuroinflammation- and gut-inflammation-immune responses, which suggests the protective and pathogenic effects of certain gut microbial species on PD progression. Therefore, gut microbiome may contain plenty of targets for preventing and managing PD. Faecal microbiota transplantation (FMT) may serve as a direct and useful treatment for PD in the future. Nonetheless, there is little available scientific research in this field. The present work reviewed the latest research to examine the association of gut microbiota with PD, and the future prospects of FMT treatment.
Collapse
Affiliation(s)
- Yongbo Kang
- Department of Microbiology and Immunology,
School of Basic Medical Sciences, Shanxi Medical
University, Taiyuan, Shanxi, China
| | - Xing Kang
- Department of Microbiology and Immunology,
School of Basic Medical Sciences, Shanxi Medical
University, Taiyuan, Shanxi, China
| | - Hongfang Zhang
- Department of Microbiology and Immunology,
School of Basic Medical Sciences, Shanxi Medical
University, Taiyuan, Shanxi, China
| | - Qingqing Liu
- Department of Microbiology and Immunology,
School of Basic Medical Sciences, Shanxi Medical
University, Taiyuan, Shanxi, China
| | - Hao Yang
- Department of Microbiology and Immunology,
School of Basic Medical Sciences, Shanxi Medical
University, Taiyuan, Shanxi, China
| | - Weiping Fan
- Department of Microbiology and Immunology,
School of Basic Medical Sciences, Shanxi Medical
University, Taiyuan, Shanxi, China
| |
Collapse
|
35
|
Ghavami A, Ziaei R, Moradi S, Sharifi S, Reza Moravejolahkami A, Ghaffari S, Irandoost P, Khorvash F, Mokari_yamchi A, Nattagh-Eshtivani E, Roshanravan N. Potential of favorable effects of probiotics fermented milk supplementation on blood pressure: a systematic review and meta-analysis. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2020. [DOI: 10.1080/10942912.2020.1833030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Abed Ghavami
- Student Research Committee, Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Sajjad Moradi
- Halal Research Center of IRI, FDA, Tehran, Iran
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Sharifi
- Student Research Committee, Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Reza Moravejolahkami
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pardis Irandoost
- Student Research Committee, Department of Nutrition, School of Health, Iran University of Medical Sciences, Tehran, Iran
| | - Fariborz Khorvash
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amin Mokari_yamchi
- Student Research Committee, Department of Community Nutrition, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elyas Nattagh-Eshtivani
- Department of Nutrition, Faculty of Medicin, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Zhu Y, Shui X, Liang Z, Huang Z, Qi Y, He Y, Chen C, Luo H, Lei W. Gut microbiota metabolites as integral mediators in cardiovascular diseases (Review). Int J Mol Med 2020; 46:936-948. [PMID: 32705240 PMCID: PMC7388831 DOI: 10.3892/ijmm.2020.4674] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVDs), such as atherosclerosis, hypertension, myocardial infarction and diabetic heart disease, are associated with high morbidity and mortality rates worldwide, and may also induce multiple organ failure in their later stages, greatly reducing the long-term survival of the patients. There are several causes of CVDs, but after nearly a decade of investigation, researchers have found that CVDs are usually accompanied by an imbalance of gut microbiota and a decreased abundance of flora. More importantly, the metabolites produced by intestinal flora, such as trimethylamine and trimethylamine N-oxide, bile acids, short-chain fatty acids and aromatic amino acids, exert different effects on the occurrence and development of CVDs, as observed in the relevant pathways in the cells, which may either promote or protect against CVD occurrence. It is known that changes in the intestinal flora following antibiotic administration, diet supplementation with probiotics, or exercise, can interfere with the composition of the intestinal flora and may represent an effective approach to preventing or treating CVDs. The focus of this review was the analysis of gut microbiota metabolites to elucidate their effects on CVDs and to identify the most cost-effective and beneficial methods for treating CVDs with minimal side effects.
Collapse
Affiliation(s)
- Ying Zhu
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Zheng Liang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Zufeng Huang
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Can Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Lei
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
37
|
Marzullo P, Di Renzo L, Pugliese G, De Siena M, Barrea L, Muscogiuri G, Colao A, Savastano S. From obesity through gut microbiota to cardiovascular diseases: a dangerous journey. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2020; 10:35-49. [PMID: 32714511 PMCID: PMC7371682 DOI: 10.1038/s41367-020-0017-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The co-existence of humans and gut microbiota started millions of years ago. Until now, a balance gradually developed between gut bacteria and their hosts. It is now recognized that gut microbiota are key to form adequate immune and metabolic functions and, more in general, for the maintenance of good health. Gut microbiota are established before birth under the influence of maternal nutrition and metabolic status, which can impact the future metabolic risk of the offspring in terms of obesity, diabetes, and cardiometabolic disorders during the lifespan. Obesity and diabetes are prone to disrupt the gut microbiota and alter the gut barrier permeability, leading to metabolic endotoxaemia with its detrimental consequences on health. Specific bacterial sequences are now viewed as peculiar signatures of the metabolic syndrome across life stages in each individual, and are linked to pathogenesis of cardiovascular diseases (CVDs) via metabolic products (metabolites) and immune modulation. These mechanisms have been linked, in association with abnormalities in microbial richness and diversity, to an increased risk of developing arterial hypertension, systemic inflammation, nonalcoholic fatty liver disease, coronary artery disease, chronic kidney disease, and heart failure. Emerging strategies for the manipulation of intestinal microbiota represent a promising therapeutic option for the prevention and treatment of CVD especially in individuals prone to CV events.
Collapse
Affiliation(s)
- Paolo Marzullo
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, 28923 Piancavallo, Verbania Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
| | - Gabriella Pugliese
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Martina De Siena
- Division of Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
| | - Luigi Barrea
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Giovanna Muscogiuri
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Annamaria Colao
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Silvia Savastano
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - on behalf of Obesity Programs of nutrition, Education, Research and Assessment (OPERA) Group
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, 28923 Piancavallo, Verbania Italy
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
- Division of Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
38
|
Saroj C, Juthika M, Tao Y, Xi C, Ji-Youn Y, Cameron MG, Camilla WF, Lauren KG, Jennifer HW, Matam VK, Bina J. Metabolites and Hypertension: Insights into Hypertension as a Metabolic Disorder: 2019 Harriet Dustan Award. Hypertension 2020; 75:1386-1396. [PMID: 32336227 PMCID: PMC7225070 DOI: 10.1161/hypertensionaha.120.13896] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
For over 100 years, essential hypertension has been researched from different perspectives ranging from genetics, physiology, and immunology to more recent ones encompassing microbiology (microbiota) as a previously underappreciated field of study contributing to the cause of hypertension. Each field of study in isolation has uniquely contributed to a variety of underlying mechanisms of blood pressure regulation. Even so, clinical management of essential hypertension has remained somewhat static. We, therefore, asked if there are any converging lines of evidence from these individual fields that could be amenable for a better clinical prognosis. Accordingly, here we present converging evidence which support the view that metabolic dysfunction underlies essential hypertension.
Collapse
Affiliation(s)
- Chakraborty Saroj
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Mandal Juthika
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Yang Tao
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Cheng Xi
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Yeo Ji-Youn
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - McCarthy G. Cameron
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Wenceslau F. Camilla
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Koch G. Lauren
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Hill W. Jennifer
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Vijay-Kumar Matam
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Joe Bina
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
39
|
Zhu Y, Li Q, Jiang H. Gut microbiota in atherosclerosis: focus on trimethylamine N-oxide. APMIS 2020; 128:353-366. [PMID: 32108960 PMCID: PMC7318354 DOI: 10.1111/apm.13038] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 12/25/2022]
Abstract
The increasing prevalence of cardiovascular diseases cannot adequately be explained by traditional risk factors. Recently, accumulating evidence has suggested that gut microbiota‐derived numerous metabolites are contributors to atherosclerotic events. Among them, the role of trimethylamine N‐oxide (TMAO) in promoting atherosclerosis has gained attention. TMAO is reported to exert the proatherogenic effects by impacting on the traditional risk factors of atherosclerosis and is associated with high risk of cardiovascular events. Besides that, TMAO is involved in the complex pathological processes of atherosclerotic lesion formation, such as endothelial dysfunction, platelet activation and thrombus generation. In light of these promising findings, TMAO may serve as a potential target for atherosclerosis prevention and treatment, which is conceptually novel, when compared with existing traditional treatments. It is likely that regulating TMAO production and associated gut microbiota may become a promising strategy for the anti‐atherosclerosis therapy.
Collapse
Affiliation(s)
- Yingqian Zhu
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qingqing Li
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Jiang
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
40
|
de Almeida Silva M, Mowry FE, Peaden SC, Andrade TU, Biancardi VC. Kefir ameliorates hypertension via gut–brain mechanisms in spontaneously hypertensive rats. J Nutr Biochem 2020; 77:108318. [DOI: 10.1016/j.jnutbio.2019.108318] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 11/06/2019] [Accepted: 11/27/2019] [Indexed: 12/27/2022]
|
41
|
Hypertension Programmed by Perinatal High-Fat Diet: Effect of Maternal Gut Microbiota-Targeted Therapy. Nutrients 2019; 11:nu11122908. [PMID: 31810197 PMCID: PMC6950030 DOI: 10.3390/nu11122908] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 01/06/2023] Open
Abstract
Hypertension can originate in early life caused by perinatal high-fat (HF) consumption. Gut microbiota and their metabolites short chain fatty acids (SCFAs), trimethylamine (TMA), and trimethylamine N-oxide (TMAO) are involved in the development of hypertension. Despite the beneficial effects of prebiotic/probiotic on human health, little is known whether maternal use of prebiotics/probiotics could protect offspring against the development of hypertension in adulthood. We investigated whether perinatal HF diet-induced programmed hypertension in adult offspring can be prevented by therapeutic uses of prebiotic inulin or probiotic Lactobacillus casei during gestation and lactation. Pregnant Sprague–Dawley rats received regular chow or HF diet (D12331, Research Diets), with 5% w/w long chain inulin (PRE), or 2 × 108 CFU/day Lactobacillus casei via oral gavage (PRO) during pregnancy and lactation. Male offspring (n = 8/group) were assigned to four groups: control, HF, PRE, and PRO. Rats were sacrificed at 16 weeks of age. Maternal prebiotic or probiotic therapy prevents elevated blood pressure (BP) programmed by perinatal HF consumption. Both prebiotic and probiotic therapies decreased the Firmicutes to Bacteroidetes ratio and renal mRNA expression of Ace, but increased abundance of genus Lactobacillus and Akkermansia. Additionally, prebiotic treatment prevents HF-induced elevation of BP is associated with reduced fecal propionate and acetate levels, while probiotic therapy restored several Lactobacillus species. Maternal probiotic or prebiotic therapy caused a reduction in plasma TMAO level and TMAO-to-TMA ratio. The beneficial effects of prebiotic or probiotic therapy on elevated BP programmed by perinatal HF diet are relevant to alterations of microbial populations, modulation of microbial-derived metabolites, and mediation of the renin-angiotensin system. Our results cast a new light on the use of maternal prebiotic/probiotic therapy to prevent hypertension programmed by perinatal HF consumption. The possibility of applying gut microbiota-targeted therapies as a reprogramming strategy for hypertension warrants further clinical translation.
Collapse
|
42
|
Lin SR, Lin SY, Chen CC, Fu YS, Weng CF. Exploring a New Natural Treating Agent for Primary Hypertension: Recent Findings and Forthcoming Perspectives. J Clin Med 2019; 8:E2003. [PMID: 31744165 PMCID: PMC6912567 DOI: 10.3390/jcm8112003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022] Open
Abstract
Primary hypertension describes abnormally-high systolic/diastolic blood pressure in a resting condition caused by various genetic or environmental risk factors. Remarkably, severe complications, such as ischemic cardiovascular disease, stroke, and chronic renal disease have led to primary hypertension becoming a huge burden for almost one-third of the total population. Medication is the major regimen for treating primary hypertension; however, recent medications may have adverse effects that attenuate energy levels. Hence, the search for new hypotensive agents from folk or traditional medicine may be fruitful in the discovery and development of new drugs. This review assembles recent findings for natural antihypertensive agents, extracts, or decoctions published in PubMed, and provides insights into the search for new hypotensive compounds based on blood-pressure regulating mechanisms, including the renin-angiotensin-aldosterone system and the sympathetic/adrenergic receptor/calcium channel system.
Collapse
Affiliation(s)
- Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan; (S.-R.L.); (C.-C.C.)
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 110, Taiwan
| | - Shiuan-Yea Lin
- Department of Anatomy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ching-Cheng Chen
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan; (S.-R.L.); (C.-C.C.)
- Camillian Saint Mary’s Hospital Luodong,160 Zhongzheng S. Rd. Luodong, Yilan 26546, Taiwan
| | - Yaw-Syan Fu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Feng Weng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Basic Medical Science, Center for Transitional Medicine, Xiamen Medical College, Xiamen 361023, China
| |
Collapse
|
43
|
The Interplay between Maternal and Post-Weaning High-Fat Diet and Gut Microbiota in the Developmental Programming of Hypertension. Nutrients 2019; 11:nu11091982. [PMID: 31443482 PMCID: PMC6769506 DOI: 10.3390/nu11091982] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/21/2022] Open
Abstract
Excessive intake of saturated fat has been linked to hypertension. Gut microbiota and their metabolites, short-chain fatty acids (SCFAs), are known to be involved in the development of hypertension. We examined whether maternal and post-weaning high-fat (HF) diet-induced hypertension in adult male offspring is related to alterations of gut microbiota, mediation of SCFAs and their receptors, and downregulation of nutrient-sensing signals. Female Sprague–Dawley rats received either a normal diet (ND) or HF diet (D12331, Research Diets) during pregnancy and lactation. Male offspring were put on either the ND or HF diet from weaning to 16 weeks of age, and designated to four groups (maternal diet/post-weaning diet; n = 8/group): ND/ND, HF/ND, ND/HF, and HF/HF. Rats were sacrificed at 16 weeks of age. Combined HF/HF diets induced elevated blood pressure (BP) and increased body weight and kidney damage in male adult offspring. The rise in BP is related to a downregulated AMP-activated protein kinase (AMPK)–peroxisome proliferator-activated receptor co-activator 1α (PGC-1α) pathway. Additionally, HF/HF diets decreased fecal concentrations of propionate and butyrate and decreased G protein-coupled receptor 41 (GPR41), but increased olfactory receptor 78 (Oflr78) expression. Maternal HF diet has differential programming effects on the offspring’s microbiota at 3 and 16 weeks of age. Combined HF/HF diet induced BP elevation was associated with an increased Firmicutes to Bacteroidetes ratio, increased abundance of genus Akkermansia and phylum Verrucomicrobia, and reduced abundance in genus Lactobacillus. Maternal gut microbiota-targeted dietary interventions might be reprogramming strategies to protect against programmed hypertension in children and their mothers on consumption of a fat-rich diet.
Collapse
|
44
|
Zhou YB, Li HT, Si KY, Zhang YL, Wang LL, Liu JM. Association of elective cesarean delivery with metabolic measures in childhood: A prospective cohort study in China. Nutr Metab Cardiovasc Dis 2019; 29:775-782. [PMID: 31151881 DOI: 10.1016/j.numecd.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/03/2019] [Accepted: 04/03/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND AIMS Cesarean delivery may increase the risk of childhood obesity, a precursor of metabolic syndrome (MetS). We aimed to investigate the association of elective cesarean delivery (ElCD) with MetS and its components in a Chinese birth cohort. METHODS AND RESULTS This cohort included 1467 children (737 delivered by ElCD and 730 by spontaneous vaginal delivery [SVD]) who were followed up at the age of 4-7 years in 2013. MetS was defined as the presence of ≥3 components: central obesity, hypertriglyceridemia, low high-density lipoprotein (HDL), high fasting glucose, and hypertension. Of the 1467 children, 93 (6.3%) were categorized as having MetS: 50 (6.8%) delivered by ElCD and 43 (5.9%) by SVD. After multivariable adjustment, ElCD was not associated with MetS (adjusted odds ratio [AOR] 1.15, 95% confidence interval [CI] 0.74, 1.78) or certain components including hypertriglyceridemia, low HDL, and high fasting glucose but was associated with central obesity (AOR 1.33, 95% CI 1.02, 1.72) and hypertension (AOR 1.50, 95% CI 1.15, 1.96), as well as higher levels of total cholesterol (3.43 vs. 3.04 mmol/L; P < 0.001), low-density lipoprotein-cholesterol (1.77 vs. 1.67 mmol/L, P = 0.002), fasting glucose (5.08 vs. 5.02 mmol/L, P = 0.022), systolic (97.57 vs. 94.69 mmHg, P < 0.001)/diastolic blood pressure (63.72 vs. 62.24 mmHg, P < 0.001), and BMI (15.46 vs. 14.83 kg/m2, P < 0.001) than SVD. CONCLUSIONS ElCD is not associated with MetS in early to middle childhood but is associated with its components including central obesity and hypertension, as well as various continuous indices.
Collapse
Affiliation(s)
- Y-B Zhou
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - H-T Li
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - K-Y Si
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Y-L Zhang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - L-L Wang
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - J-M Liu
- Institute of Reproductive and Child Health, Ministry of Health Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
45
|
Probiotics as Beneficial Dietary Supplements to Prevent and Treat Cardiovascular Diseases: Uncovering Their Impact on Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3086270. [PMID: 31205584 PMCID: PMC6530239 DOI: 10.1155/2019/3086270] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/01/2019] [Accepted: 04/14/2019] [Indexed: 02/07/2023]
Abstract
The gut microbiota, the ecosystem formed by a wide symbiotic community of nonpathogenic microorganisms that are present in the distal part of the human gut, plays a prominent role in the normal physiology of the organism. The gut microbiota's imbalance, gut dysbiosis, is directly related to the origin of various processes of acute or chronic dysfunction in the host. Therefore, the ability to intervene in the gut microbiota is now emerging as a possible tactic for therapeutic intervention in various diseases. From this perspective, evidence is growing that a functional dietary intervention with probiotics, which maintain or restore beneficial bacteria of the digestive tract, represents a promising therapeutic strategy for interventions in cardiovascular diseases and also reduces the risk of their occurrence. In the present work, we review the importance of maintaining the balance of the intestinal microbiota to prevent or combat such processes as arterial hypertension or endothelial dysfunction, which underlie many cardiovascular disorders. We also review how the consumption of probiotics can improve autonomic control of cardiovascular function and provide beneficial effects in patients with heart failure. Among the known effects of probiotics is their ability to decrease the generation of reactive oxygen species and, therefore, reduce oxidative stress. Therefore, in this review, we specifically focus on this antioxidant capacity and its relationship with the beneficial cardiovascular effects described for probiotics.
Collapse
|
46
|
Resveratrol prevents combined prenatal N G-nitro-L-arginine-methyl ester (L-NAME) treatment plus postnatal high-fat diet induced programmed hypertension in adult rat offspring: interplay between nutrient-sensing signals, oxidative stress and gut microbiota. J Nutr Biochem 2019; 70:28-37. [PMID: 31108332 DOI: 10.1016/j.jnutbio.2019.04.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/17/2019] [Accepted: 04/04/2019] [Indexed: 12/28/2022]
Abstract
Oxidative stress, nutrient-sensing signals, high-fat (HF) intake and dysbiosis of gut microbiota are involved in the development of hypertension, a disorder that can originate in early life. We examined whether postnatal HF diet can aggravate maternal NG-nitro-L-arginine-methyl ester (L-NAME) treatment-induced programmed hypertension and whether resveratrol therapy can prevent it. Pregnant Sprague-Dawley rats received L-NAME administration at 60 mg/kg/day subcutaneously during pregnancy alone, or with additional resveratrol (R) 50 mg/L in drinking water during the pregnancy and lactation. The offspring were onto either regular chow or HF diet (D12331) from weaning to 16 weeks of age. Male offspring rats were assigned to five groups (N=8/group): control, L-NAME, HF, L-NAME+HF and L-NAME+HF + R at weaning at 3 weeks of age. Rats were sacrificed at 16 weeks of age. We observed that postnatal HF diet exacerbates maternal L-NAME treatment-induced programmed hypertension in male adult offspring, which resveratrol attenuated. Combined L-LAME and HF diet-induced hypertension is related to increased oxidative stress, inhibiting AMP-activated protein kinase (AMPK)/ peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) pathway and altered gut microbiota compositions. L-NAME+HF caused an increase of the Firmicutes to Bacteroidetes ratio, which resveratrol therapy prevented. Additionally, the abundances of phylum Verrucomicrobia and genus Akkermansia were amplified by resveratrol therapy. Conclusively, our data highlighted the interactions between maternal NO deficiency, HF diet, AMPK/PGC-1α pathway and gut microbiota in which the blood pressure of adult offspring can be modified by resveratrol. Resveratrol might be a useful reprogramming strategy to prevent L-NAME and HF diet-induced hypertension of developmental origin.
Collapse
|
47
|
Kang Y, Cai Y. Altered Gut Microbiota in HIV Infection: Future Perspective of Fecal Microbiota Transplantation Therapy. AIDS Res Hum Retroviruses 2019; 35:229-235. [PMID: 29877092 DOI: 10.1089/aid.2017.0268] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
HIV infection progressively destroys CD4+ mononuclear cells, leading to profound cellular immune deficiency that manifests as life-threatening opportunistic infections and malignancies (i.e., AIDS). Gut microbiota plays key roles in the modulation of host metabolism and gene expression, maintenance of epithelial integrity, and mediation of inflammatory and immunity. Hence, the normal intestinal microbiota plays a major role in the maintenance of health and disease prevention. In fact, a large number of studies have shown that the alteration of the gut microbiota contributes to the pathogenesis of several diseases, such as inflammatory bowel diseases, irritable bowel syndrome, metabolic diseases, anorexia nervosa, autoimmune diseases, multiple sclerosis, cancer, neuropsychiatric disorders, and cardiovascular diseases. Recently, accumulating evidence has shed light on the association of dysbiosis of gut microbiota with HIV infection. Hence, the modification of gut microbiota may be a potential therapeutic tool. Fecal microbiota transplantation may improve the conditions of patients with HIV infection by manipulating the human intestinal bacteria. However, the relevant research is very limited, and a large amount of scientific research work needs to be done in the near future.
Collapse
Affiliation(s)
- Yongbo Kang
- 1 School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- 2 Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Yue Cai
- 1 School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- 3 Genetics and Pharmacogenomics Laboratory, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
48
|
|
49
|
Abstract
Proteomics is a widely used method for defining the protein composition of a complex sample. As this approach allows for identification and quantification of proteins across a broad dynamic range as well as detection of post-translational modifications, proteomics is an ideal platform to investigate the gut microbiome at a functional level. The gut microbiome is a dynamic environment which is crucial for overall health and fitness. Imbalances in the gut microbiome can influence nutrient absorption, pathogen resistance, inflammation, and various human diseases. Metaproteomic analysis of the gut microbiome is currently being performed on bacteria isolated from (1) fecal samples (2) colonic lavage, or (3) colon biopsies. Investigation of the gut microbiome has demonstrated that within the colon, there are distinct communities based on spatial location, and separable from the gut microbiomes isolated from stool. In addition to expanding our understanding of host-bacterial interactions for human health and disease, gut microbiome analysis is being utilized for biomarker development to discriminate normal individuals and diseased (i.e., inflammatory bowel disease or colon cancer) patients as well as to monitor disease activity and prognosis.
Collapse
Affiliation(s)
- Lisa A Lai
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Zachary Tong
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ru Chen
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Sheng Pan
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
50
|
Nikitina E, Larionova I, Choinzonov E, Kzhyshkowska J. Monocytes and Macrophages as Viral Targets and Reservoirs. Int J Mol Sci 2018; 19:E2821. [PMID: 30231586 PMCID: PMC6163364 DOI: 10.3390/ijms19092821] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 02/07/2023] Open
Abstract
Viruses manipulate cell biology to utilize monocytes/macrophages as vessels for dissemination, long-term persistence within tissues and virus replication. Viruses enter cells through endocytosis, phagocytosis, macropinocytosis or membrane fusion. These processes play important roles in the mechanisms contributing to the pathogenesis of these agents and in establishing viral genome persistence and latency. Upon viral infection, monocytes respond with an elevated expression of proinflammatory signalling molecules and antiviral responses, as is shown in the case of the influenza, Chikungunya, human herpes and Zika viruses. Human immunodeficiency virus initiates acute inflammation on site during the early stages of infection but there is a shift of M1 to M2 at the later stages of infection. Cytomegalovirus creates a balance between pro- and anti-inflammatory processes by inducing a specific phenotype within the M1/M2 continuum. Despite facilitating inflammation, infected macrophages generally display abolished apoptosis and restricted cytopathic effect, which sustains the virus production. The majority of viruses discussed in this review employ monocytes/macrophages as a repository but certain viruses use these cells for productive replication. This review focuses on viral adaptations to enter monocytes/macrophages, immune escape, reprogramming of infected cells and the response of the host cells.
Collapse
Affiliation(s)
- Ekaterina Nikitina
- Department of Episomal-Persistent DNA in Cancer- and Chronic Diseases, German Cancer Research Center, 69120 Heidelberg, Germany.
- Department of Oncovirology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russia.
- Department of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk 634050, Russia.
| | - Irina Larionova
- Department of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk 634050, Russia.
- Department of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russia.
| | - Evgeniy Choinzonov
- Head and Neck Department, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russia.
| | - Julia Kzhyshkowska
- Department of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk 634050, Russia.
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, 68167 Heidelberg, Germany.
| |
Collapse
|