1
|
Kulkarni H, Gaikwad AB. The mitochondria-gut microbiota crosstalk - A novel frontier in cardiovascular diseases. Eur J Pharmacol 2025; 998:177562. [PMID: 40157703 DOI: 10.1016/j.ejphar.2025.177562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Cardiovascular diseases (CVDs), including hypertension, atherosclerosis, and cardiomyopathy among others, remain the leading cause of global morbidity and mortality. Despite advances in treatment, the complex pathophysiology of CVDs necessitates innovative approaches to improve patient outcomes. Recent research has uncovered a dynamic interplay between mitochondria and gut microbiota, fundamentally altering our understanding of cardiovascular health. However, while existing studies have primarily focused on individual components of this axis, this review examines the bidirectional communication between these biological systems and their collective impact on cardiovascular health. Mitochondria, serving as cellular powerhouses, are crucial for maintaining cardiovascular homeostasis through oxidative phosphorylation (OXPHOS), calcium regulation, and redox balance. Simultaneously, the gut microbiota influences cardiovascular function through metabolite production, barrier integrity maintenance, and immune system modulation. The mitochondria-gut microbiota axis operates through various molecular mechanisms, including microbial metabolites such as trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFA), and secondary bile acids, which directly influence mitochondrial function. Conversely, mitochondrial stress signals and damage-associated molecular patterns (DAMPs) affect gut microbial communities and barrier function. Key signalling pathways, including AMP-activated protein kinase (AMPK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and the silent information regulator 1-peroxisome proliferator-activated receptor gamma coactivator 1-alpha (SIRT1-PGC-1α) axis, integrate these interactions, highlighting their role in CVD pathogenesis. Understanding these interactions has revealed promising therapeutic targets, suggesting new therapies aimed at both mitochondrial function and gut microbiota composition. Thus, this review provides a comprehensive framework for leveraging the mitochondria-gut microbiota axis in providing newer therapeutics for CVDs by targeting the AMPK/SIRT-1/PGC-1α/NF-κB signalling.
Collapse
Affiliation(s)
- Hrushikesh Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani, Rajasthan 333031, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani, Rajasthan 333031, India.
| |
Collapse
|
2
|
Okunlola FO, Okunlola AR, Adetuyi BO, Soliman MES, Alexiou A, Papadakis M, Fawzy MN, El-Saber Batiha G. Beyond the gut: Unraveling the multifaceted influence of microbiome on cardiovascular health. Clin Nutr ESPEN 2025; 67:71-89. [PMID: 40064239 DOI: 10.1016/j.clnesp.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Cardiovascular disease is one of the leading causes of death worldwide. Even while receiving adequate pharmacological treatment for their hypertension, people are nonetheless at greater risk for cardiovascular disease. There is growing evidence that the gut microbiota may have major positive and negative effects on blood pressure and illnesses related with it as more study into this topic is conducted. Trimethylamine n-oxide (TMAO) and short-chain fatty acids (SCFA) are two major by-products of the gut microbiota. TMAO is involved in the formation of other coronary artery diseases, including atherosclerosis and hypertension, while SCFAs play an important role in controlling blood pressure. Numerous investigations have confirmed the established link between dietary salt intake and hypertension. Reducing sodium in the diet is linked to lower rates of cardiovascular disease morbidity and mortality as well as lower rates of blood pressure and hypertension. In both human and animal research, high salt diets increase local and systemic tissue inflammation and compromise gut architecture. Given that the gut microbiota constantly interacts with the immune system and is required for the correct maturation of immune cells, it is scientifically conceivable that it mediates the inflammatory response. This review highlights the therapeutic possibilities for focusing on intestinal microbiomes as well as the potential functions of the gut microbiota and its metabolites in the development of hypertension.
Collapse
Affiliation(s)
- Felix Oladele Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Abimbola Rafiat Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Babatunde Oluwafemi Adetuyi
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
3
|
Lv M, Shafagh G, Yu S. Effect of synbiotics on the cardiovascular risk factors in patients with non-alcoholic fatty liver: a GRADE assessed systematic review and meta-analysis. BMC Gastroenterol 2025; 25:407. [PMID: 40419987 DOI: 10.1186/s12876-025-03789-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/17/2025] [Indexed: 05/28/2025] Open
Abstract
INTRODUCTION Several studies revealed that synbiotics have been beneficial in managing non-alcoholic fatty liver disease (NAFLD), but the findings are conflicting. We aimed to assess the effect of synbiotic supplementation on cardiovascular risk factors in patients with NAFLD. METHODS A thorough literature search was conducted to identify relevant studies up to July 2024, including publications from Pubmed, Embase, Cochrane, Scopus, and the Web of Science. Standardized mean difference (SMD) was used, and data with a 95% confidence interval (CI) were presented. RESULTS Sixteen RCTs were included, involving NAFLD patients and synbiotic supplementation. No significant effects were found on blood pressure (SBP: SMD - 6.68, p = 0.234; DBP: SMD - 4.31, p = 0.391), anthropometric measures (weight: SMD - 0.78, p = 0.126; BMI: SMD - 0.21, p = 0.182; waist circumference: SMD - 0.53, p = 0.095), or lipid profile (triglycerides: SMD - 3.67, p = 0.266; total cholesterol: SMD - 0.61, p = 0.059; low-density lipoprotein: SMD - 3.09, p = 0.215; high-density lipoprotein: SMD 0.49, p = 0.219). Synbiotics significantly reduced CRP levels (SMD - 1.83, p = 0.019) but had no significant effect on TNF-α (SMD - 1.95, p = 0.087). Synbiotic led to a significant reduction in weight following adjustment of publication bias (SMD - 1.16, p < 0.05). CONCLUSION Overall, synbiotic supplementation does not significantly impact blood pressure, anthropometric indices, or lipid profile parameters in patients with NAFLD, except for a significant reduction in weight after adjusting for publication bias. Synbiotic supplementation significantly reduces CRP levels but has no effect on TNF-α.
Collapse
Affiliation(s)
- Minfang Lv
- Department of Nursing, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Ghavam Shafagh
- Faculty of Medicine, Lorestan Medical University, Khorramabad, Iran.
| | - Saiting Yu
- Department of Nursing, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
López-Moreno M, Fresán U, Marchena-Giráldez C, Bertotti G, Roldán-Ruiz A. Industry study sponsorship and conflicts of interest on the effect of unprocessed red meat on cardiovascular disease risk: a systematic review of clinical trials. Am J Clin Nutr 2025:S0002-9165(25)00126-1. [PMID: 40379522 DOI: 10.1016/j.ajcnut.2025.02.030] [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: 04/04/2024] [Revised: 02/12/2025] [Accepted: 02/26/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Experimental research on the link between unprocessed red meat and cardiovascular disease risk is inconsistent and may differ according to the financial interests of red meat industry sponsors. OBJECTIVES This study aims to assess whether studies sponsorship or conflicts of interest with the red meat industry are associated with reported outcomes of unprocessed red meat consumption effect on risk factors for cardiovascular disease. METHODS PubMed, Cochrane Library, and Scopus were searched from the inception of the databases to 3 March 2024. Studies were classified as "Red meat industry-related" if any of the authors declared affiliation or financial disclosure indicating a link to the red meat industry; or "Red Meat industry-independent." Reported outcomes were independently graded as favorable, neutral, or unfavorable. Studies were also categorized by type of control group. The quality of evidence for each outcome was evaluated using the Grading of Recommendations Assessment, Development and Evaluation approach. RESULTS A total of 44 studies were included, of which 66% had a link to the red meat industry. All independent studies reported either unfavorable (73.3%) or neutral (26.7%) cardiovascular outcomes when consuming unprocessed red meat. Conversely, all studies related to the red meat industry reported either favorable (20.7%) or neutral (79.3%) cardiovascular outcomes for red meat intake. A total of 69.6% of trials (16 of 23) showed a neutral effect of unprocessed red meat compared with other animal proteins, whereas 70% (7 of 10) reported an unfavorable effect compared with plant proteins. Studies with conflicts of interest were nearly 4 times more likely to report "Favorable/Neutral" outcomes compared with independent studies (odds ratio 3.75, 95% confidence interval: 1.62, 8.67). CONCLUSIONS Our findings highlight that using animal protein as a comparator in industry-funded studies, with quality of evidence rated as very low to low, may underestimate the cardiovascular benefits of reducing red meat intake. Most studies without conflicts of interest with the red meat industry suggested an unfavorable effect of unprocessed red meat consumption on risk factors for cardiovascular disease. TRIAL REGISTRATION NUMBER This trial for systematic reviews or meta-analyses was registered at PROSPERO as CRD42024525197 (https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=525197).
Collapse
Affiliation(s)
- Miguel López-Moreno
- Diet, Planetary Health and Performance, Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo, Spain.
| | | | | | - Gabriele Bertotti
- Diet, Planetary Health and Performance, Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo, Spain; School of Physiotherapy, Faculty of Health Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Alberto Roldán-Ruiz
- Diet, Planetary Health and Performance, Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo, Spain; School of Physiotherapy, Faculty of Health Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| |
Collapse
|
5
|
Abavisani M, Tafti P, Khoshroo N, Ebadpour N, Khoshrou A, Kesharwani P, Sahebkar A. The heart of the matter: How gut microbiota-targeted interventions influence cardiovascular diseases. Pathol Res Pract 2025; 269:155931. [PMID: 40174272 DOI: 10.1016/j.prp.2025.155931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
The human body is habitat to a wide spectrum of microbial populations known as microbiota, which play an important role in overall health. The considerable research has mostly focused on the gut microbiota due to its potential to impact numerous physiological functions and its correlation with a variety of disorders, such as cardiovascular diseases (CVDs). Imbalances in the gut microbiota, known as dysbiosis, have been linked to the development and progression of CVDs through various processes, including the generation of metabolites like trimethylamine-N-oxide and short-chain fatty acids. Studies have also looked at the idea of using therapeutic interventions, like changing your diet, taking probiotics or prebiotics, or even fecal microbiota transplantation (FMT), to change the gut microbiota's make-up and how it works in order to prevent or treat CVDs. Exploring the cause-and-effect connection between the gut microbiota and CVDs offers a hopeful path for creating innovative microbiome-centered strategies to prevent and cure CVDs. This review presents an in-depth review of the correlation between the gut microbiota and CVDs, as well as potential therapeutic approaches for manipulating the gut microbiota to enhance cardiovascular health.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pourya Tafti
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khoshroo
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pardesh, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Przewodowska D, Alster P, Madetko-Alster N. Role of the Intestinal Microbiota in the Molecular Pathogenesis of Atypical Parkinsonian Syndromes. Int J Mol Sci 2025; 26:3928. [PMID: 40362171 PMCID: PMC12071724 DOI: 10.3390/ijms26093928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 05/15/2025] Open
Abstract
The role of the intestinal microbiota and its influence on neurodegenerative disorders has recently been extensively explored, especially in the context of Parkinson's disease (PD). In particular, its role in immunomodulation, impact on inflammation, and participation in the gut-brain axis are under ongoing investigations. Recent studies have revealed new data that could be important for exploring the neurodegeneration mechanisms connected with the gut microbiota, potentially leading to the development of new methods of treatment. In this review, the potential roles of the gut microbiota in future disease-modifying therapies were discussed and the properties of the intestinal microbiota-including its impacts on metabolism and short-chain fatty acids and vitamins-were summarized, with a particular focus on atypical Parkinsonian syndromes. This review focused on a detailed description of the numerous mechanisms through which the microbiota influences neurodegenerative processes. This review explored potentially important connections between the gut microbiota and the evolution and progression of atypical Parkinsonian syndromes. Finally, a description of recently derived results regarding the microbiota alterations in atypical Parkinsonian syndromes in comparison with results previously described in PD was also included.
Collapse
Affiliation(s)
- Dominika Przewodowska
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242 Warsaw, Poland;
| | | | - Natalia Madetko-Alster
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242 Warsaw, Poland;
| |
Collapse
|
7
|
Tonon CR, Pereira AG, Ferreira NF, Monte MG, Vieira NM, Fujimori ASS, Ballin PDS, de Paiva SAR, Zornoff LAM, Minicucci MF, Polegato BF. The Gut-Heart Axis and Its Role in Doxorubicin-Induced Cardiotoxicity: A Narrative Review. Microorganisms 2025; 13:855. [PMID: 40284691 PMCID: PMC12029146 DOI: 10.3390/microorganisms13040855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Doxorubicin is a widely used chemotherapy for the treatment of several types of cancer. However, its application is restricted due to adverse effects, particularly cardiotoxicity, which can progress to heart failure-a chronic and debilitating condition. Several mechanisms have been identified in the pathophysiology of doxorubicin-induced cardiotoxicity, including oxidative stress, mitochondrial dysfunction, inflammation, and disruption of collagen homeostasis. More recently, dysbiosis of the gut microbiota has been implicated in the development and perpetuation of cardiac injury. Studies have reported alterations in the composition and abundance of the microbiota during doxorubicin treatment. Therefore, as of recent, there is a new field of research in order to develop strategies involving the gut microbiota to prevent or attenuate cardiotoxicity since there is no effective therapy at the moment. This narrative review aims to provide an update on the role of gut microbiota and intestinal permeability in the pathophysiology of cardiovascular diseases, and more specifically doxorubicin-induced cardiotoxicity. Additionally, it seeks to establish a foundation for future research targeting gut microbiota to alleviate cardiotoxicity.
Collapse
Affiliation(s)
- Carolina Rodrigues Tonon
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, Brazil (B.F.P.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Kalkan AE, BinMowyna MN, Raposo A, Ahmad MF, Ahmed F, Otayf AY, Carrascosa C, Saraiva A, Karav S. Beyond the Gut: Unveiling Butyrate's Global Health Impact Through Gut Health and Dysbiosis-Related Conditions: A Narrative Review. Nutrients 2025; 17:1305. [PMID: 40284169 PMCID: PMC12029953 DOI: 10.3390/nu17081305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Short-chain fatty acids (SCFAs), mainly produced by gut microbiota through the fermentation process of dietary fibers and proteins, are crucial to human health, with butyrate, a famous four-carbon SCFA, standing out for its inevitably regulatory impact on both gut and immune functions. Within this narrative review, the vital physiological functions of SCFAs were examined, with emphasis on butyrate's role as an energy source for colonocytes and its ability to enhance the gut barrier while exhibiting anti-inflammatory effects. Knowledge of butyrate synthesis, primarily generated by Firmicutes bacteria, can be influenced by diets with specifically high contents of resistant starches and fiber. Butyrate can inhibit histone deacetylase, modulate gene expression, influence immune functionality, and regulate tight junction integrity, supporting the idea of its role in gut barrier preservation. Butyrate possesses systemic anti-inflammatory properties, particularly, its capacity to reduce pro-inflammatory cytokines and maintain immune homeostasis, highlighting its therapeutic potential in managing dysbiosis and inflammatory diseases. Although butyrate absorption into circulation is typically minimal, its broader health implications are substantial, especially regarding obesity and type 2 diabetes through its influence on metabolic regulation and inflammation. Furthermore, this narrative review thoroughly examines butyrate's growing recognition as a modulator of neurological health via its interaction with the gut-brain axis. Additionally, butyrate's neuroprotective effects are mediated through activation of specific G-protein-coupled receptors, such as FFAR3 and GPR109a, and inhibition of histone deacetylases (HDACs). Research indicates that butyrate can alleviate neurological disorders, including Alzheimer's, Parkinson's, autism spectrum disorder, and Huntington's disease, by reducing neuroinflammation, enhancing neurotransmitter modulation, and improving histone acetylation. This focus will help unlock its full therapeutic potential for metabolic and neurological health, rather than exclusively on its well-known benefits for gut health, as these are often interconnected.
Collapse
Affiliation(s)
- Arda Erkan Kalkan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| | - Mona N. BinMowyna
- College of Education, Shaqra University, Shaqra 11911, Saudi Arabia;
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Faiyaz Ahmed
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia;
| | - Abdullah Y. Otayf
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Conrado Carrascosa
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain;
| | - Ariana Saraiva
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal;
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| |
Collapse
|
9
|
Ponce Alencastro JA, Salinas Lucero DA, Solis RP, Herrera Giron CG, Estrella López AS, Anda Suárez PX. Molecular Mechanisms and Emerging Precision Therapeutics in the Gut Microbiota-Cardiovascular Axis. Cureus 2025; 17:e83022. [PMID: 40421334 PMCID: PMC12104768 DOI: 10.7759/cureus.83022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2025] [Indexed: 05/28/2025] Open
Abstract
A microbiome in the gut plays a significant role in cardiovascular health and disease. Dysbiosis is an imbalance in the gut microbiome, leading to multiple cardiovascular diseases (CVD) such as atherosclerosis, hypertension, and heart failure. Gut microbe-derived metabolites such as trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs) are important mediators of the gut-heart axis. Evaluation of the relationship between the gut microbiome and host biomarkers with CVD requires the integration of metagenomics and metabolomics with meta-omics approaches. The literature review found that microbes and metabolic signatures are associated with the risk and progression of CVD. The development of precision therapeutic approaches for targeting gut microbiota includes preventing adverse microbial effects using probiotics, prebiotics, and the drug-as-bug approach to inhibit harmful metabolites of microbiomes, and fecal microbiota transplantation (FMT). However, the implication and practice of these findings in clinical settings face challenges due to the heterogeneity of study designs, difficulty in the determination of causality, and the impact of confounding factors such as diet, medication, and potential inter-individual gut microbiome variability. Future researchers are recommended to conduct longitudinal studies to further establish both gut microbiome associations with CVD and develop successful precision therapeutics approaches based on the microbiome for the treatment of CVD.
Collapse
Affiliation(s)
| | | | - Ricardo Perez Solis
- Material Sciences, Instituto Tecnológico Superior de Atlixco, Tecnológico Nacional de México (TecNM), Atlixco, MEX
| | | | | | | |
Collapse
|
10
|
Fadhillah FS, Habibah K, Juniarto AZ, Sobirin MA, Maharani N, Pramono A. Diet and the gut microbiota profiles in individuals at risk of chronic heart failure - A review on the Asian population. Asia Pac J Clin Nutr 2025; 34:141-152. [PMID: 40134053 PMCID: PMC11937497 DOI: 10.6133/apjcn.202504_34(2).0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/22/2024] [Accepted: 07/09/2024] [Indexed: 03/27/2025]
Abstract
BACKGROUND AND OBJECTIVES Chronic Heart Failure (CHF) is one of the leading cardiovascular diseases (CVDs), particularly in the Asian population. Individuals with specific health risks, such as obesity, type 2 diabetes, hypertension, dyslipidemia, and coronary artery disease (CAD), are more susceptible to developing CHF. Current evidence is limited to understanding the link between gut microbiota dysbiosis and CHF. Therefore, this review aims to explore the potential connection between dietary patterns, gut microbiota, and its metabolites in individuals at risk of CHF in the Asian population. METHODS AND STUDY DESIGN A literature review of cross-sectional studies was conducted using primary keywords such as "Asian", "obesity", "type 2 diabetes", "hypertension", "dyslipidemia", "coronary artery disease", and "chronic heart failure". There was no restriction on sample size. RESULTS Several gut microbiotas were found to correlate with CHF risk factors. There were increased levels of Prevotella, Klebsiella, Romboutsia, Catenibacterium, Clostridium, Holdemanella, Ruminococcus, Coprococcus, Parabacteroides, Bacteroides, Lachnoclostridium, Streptococcus, and Megamonas, while decreased levels of Oscillibacter, Bifidobacterium, Lactobacillus, Akkermansia, Roseburia, Faecalibacterium, Pseudobutyrivibrio, and Eubacterium were reported. These microbiota shifts were linked to increased TMAO production and impaired short-chain fatty acids (SCFAs) production. Dietary intake and microbial metabolites were also identified as contributors to the gut microbiota associated with CHF. CONCLUSIONS A potential link exists between the gut microbiota profile and CHF risk factors, possibly mediated by microbial metabolites. Dietary patterns may influence CHF-associated gut microbiota and me-tabolites. Future research is needed to investigate how dietary modifications can modulate gut microbiota and its metabolites in CHF patients.
Collapse
Affiliation(s)
- Farhan S Fadhillah
- Department of Nutrition Science, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Kona'atul Habibah
- Department of Nutrition Science, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Achmad Z Juniarto
- Department of Medical Study, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
- Center of Biomedical Research, Diponegoro University, Semarang, Indonesia
| | - Mochamad A Sobirin
- Department of Medical Study, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Nani Maharani
- Department of Medical Study, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Adriyan Pramono
- Department of Nutrition Science, Faculty of Medicine, Diponegoro University, Semarang, Indonesia. ;
- Center of Nutrition Research, Diponegoro University, Semarang, Indonesia
| |
Collapse
|
11
|
Wu Y, Chen X, Wu Q, Wang Q. Research progress on fecal microbiota transplantation in tumor prevention and treatment. Open Life Sci 2025; 20:20220954. [PMID: 40177417 PMCID: PMC11964189 DOI: 10.1515/biol-2022-0954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/27/2024] [Accepted: 08/07/2024] [Indexed: 04/05/2025] Open
Abstract
The application of fecal microbiota transplantation (FMT) as a therapeutic strategy to directly modify the makeup of the gut microbiota has made significant progress in the last few decades. The gut microbiota, a sizable microbial community present in the human gut, is essential for digestion, immunomodulation, and nutrition absorption. Alternatively, a growing body of research indicates that gut microbiota is a key contributor to cancer, and intratumoral bacteria are considered to be crucial "accomplices" in the development and metastasis of malignancies. The exceptional clinical effectiveness of FMT in treating melanoma patients has been adequately established in earlier research, which has created new avenues for the diagnosis and treatment of cancer and sparked an increasing interest in the treatment and prevention of other cancers. However, further research on the function and mechanisms of the gut microbiota is required to properly comprehend the impact and role of these organisms in tumor regulation. In this article, we present a detailed account of the influence of FMT on the entire course of cancer patients' illness and treatment, from tumor development, metastasis, and invasion, to the impact and application of treatment and prognosis, as well as address the associated mechanisms.
Collapse
Affiliation(s)
- Yijia Wu
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Xi Chen
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Qingming Wu
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Qiang Wang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
| |
Collapse
|
12
|
Quagliariello V, Berretta M, Bisceglia I, Giacobbe I, Iovine M, Barbato M, Maurea C, Canale ML, Paccone A, Inno A, Scherillo M, Oliva S, Cadeddu Dessalvi C, Mauriello A, Fonderico C, Maratea AC, Gabrielli D, Maurea N. In the Era of Cardiovascular-Kidney-Metabolic Syndrome in Cardio-Oncology: From Pathogenesis to Prevention and Therapy. Cancers (Basel) 2025; 17:1169. [PMID: 40227756 PMCID: PMC11988012 DOI: 10.3390/cancers17071169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Cardiovascular-kidney-metabolic (CKM) syndrome represents a complex interplay between cardiovascular disease (CVD), chronic kidney disease (CKD), and metabolic disorders, significantly impacting cancer patients. The presence of CKM syndrome in cancer patients not only worsens their prognosis but also increases the risk of major adverse cardiovascular events (MACE), reduces quality of life (QoL), and affects overall survival (OS). Furthermore, several anticancer therapies, including anthracyclines, tyrosine kinase inhibitors, immune checkpoint inhibitors, and hormonal treatments, can exacerbate CKM syndrome by inducing cardiotoxicity, nephrotoxicity, and metabolic dysregulation. This review explores the pathophysiology of CKM syndrome in cancer patients and highlights emerging therapeutic strategies to mitigate its impact. We discuss the role of novel pharmacological interventions, including sodium-glucose cotransporter-2 inhibitors (SGLT2i), proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), and soluble guanylate cyclase (sGC) activators, as well as dietary and lifestyle interventions. Optimizing the management of CKM syndrome in cancer patients is crucial to improving OS, enhancing QoL, and reducing MACE. By integrating cardiometabolic therapies into oncologic care, we can create a more comprehensive treatment approach that reduces the burden of cardiovascular and renal complications in this vulnerable population. Further research is needed to establish personalized strategies for CKM syndrome prevention and treatment in cancer patients.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Carlo Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, 23807 Naples, Italy
| | | | - Andrea Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy
| | - Marino Scherillo
- Cardiologia Interventistica e UTIC, A.O. San Pio, Presidio Ospedaliero Gaetano Rummo, 82100 Benevento, Italy
| | - Stefano Oliva
- Cardio-Oncology Unit, IRCCS Istituto Tumori, “Giovanni Paolo II”, 70124 Bari, Italy
| | | | - Alfredo Mauriello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Celeste Fonderico
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Anna Chiara Maratea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| |
Collapse
|
13
|
Khanmohammadi S, Fallahtafti P, Habibzadeh A, Ezzatollahi Tanha A, Alamdari AA, Fallahtafti P, Shafi Kuchay M. Effectiveness of body roundness index for the prediction of nonalcoholic fatty liver disease: a systematic review and meta-analysis. Lipids Health Dis 2025; 24:117. [PMID: 40148946 PMCID: PMC11948846 DOI: 10.1186/s12944-025-02544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Several anthropometric indices, such as body mass index and waist circumference, have been used as clinical screening tools for the prediction of nonalcoholic fatty liver disease (NAFLD). To further refine these clinical tools for NAFLD, the body roundness index (BRI) has recently been evaluated. In this systematic review and meta-analysis, the objective was to evaluate the relationship and predictive capability of the BRI in identifying NAFLD. METHODS A comprehensive search was conducted in PubMed, Embase, Web of Science, and Scopus up to December 31, 2024. Eligibility criteria included observational studies on adults (≥ 18 years old) with measured BRI and its association with NAFLD. The Joanna Briggs Institute tool was used for risk of bias assessment. Meta-analyses used random-effects models to pool data on mean difference, odds ratio, sensitivity, specificity, and the area under the curve (AUC), with heterogeneity and publication bias assessed. RESULTS Ten studies involving 59,466 participants were included. The pooled mean difference in BRI between the NAFLD and non-NAFLD groups was 1.73 (95% confidence interval [CI]: 1.31-2.15). The pooled sensitivity and specificity of BRI for diagnosing NAFLD were 0.806 and 0.692, respectively. The pooled AUC for BRI was 0.803 (95% CI: 0.775-0.830), indicating good diagnostic accuracy. Unlike subgroup analysis by country, subgroup analysis by sex showed no significant differences. Higher BRI values were associated with increased odds of NAFLD (pooled OR = 2.87, 95% CI: 1.39; 5.96). Studies provided mixed results on the predictive ability of BRI compared to other indices like body mass index, mostly favoring BRI over conventional indices. CONCLUSION BRI demonstrates a good diagnostic performance for NAFLD, suggesting it may be a valuable clinical tool for NAFLD assessment. Further research is necessary to validate these findings and strengthen the evidence base.
Collapse
Affiliation(s)
- Shaghayegh Khanmohammadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Parisa Fallahtafti
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cardiovascular Diseases Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Amir Ali Alamdari
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parsa Fallahtafti
- School of Pharmacy and Pharmaceutical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Shafi Kuchay
- Divison of Endocrinology and Diabetes, Medanta the Medicity Hospital, Gurugram, Haryana, 122001, India
| |
Collapse
|
14
|
Młynarska E, Barszcz E, Budny E, Gajewska A, Kopeć K, Wasiak J, Rysz J, Franczyk B. The Gut-Brain-Microbiota Connection and Its Role in Autism Spectrum Disorders. Nutrients 2025; 17:1135. [PMID: 40218893 PMCID: PMC11990867 DOI: 10.3390/nu17071135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Autism spectrum disorder (ASD) is a group of complex neurodevelopmental conditions with a heterogeneous and multifactorial etiology that is not yet fully understood. Among the various factors that may contribute to ASD development, alterations in the gut microbiota have been increasingly recognized. Microorganisms in the gastrointestinal tract play a crucial role in the gut-brain axis (GBA), affecting nervous system development and behavior. Dysbiosis, or an imbalance in the microbiota, has been linked to both behavioral and gastrointestinal (GI) symptoms in individuals with ASD. The microbiota interacts with the central nervous system through mechanisms such as the production of short-chain fatty acids (SCFAs), the regulation of neurotransmitters, and immune system modulation. Alterations in its composition, including reduced diversity or an overabundance of specific bacterial taxa, have been associated with the severity of ASD symptoms. Dietary modifications, such as gluten-free or antioxidant-rich diets, have shown potential for improving gut health and alleviating behavioral symptoms. Probiotics, with their anti-inflammatory properties, may support neural health and reduce neuroinflammation. Fecal microbiota transplantation (FMT) is being considered, particularly for individuals with persistent GI symptoms. It has shown promising outcomes in enhancing microbial diversity and mitigating GI and behavioral symptoms. However, its limitations should be considered, as discussed in this narrative review. Further research is essential to better understand the long-term effects and safety of these therapies. Emphasizing the importance of patient stratification and phenotype characterization is crucial for developing personalized treatment strategies that account for individual microbiota profiles, genetic predispositions, and coexisting conditions. This approach could lead to more effective interventions for individuals with ASD. Recent findings suggest that gut microbiota may play a key role in innovative therapeutic approaches to ASD management.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Barszcz
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Emilian Budny
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Kacper Kopeć
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
15
|
Jiang GH, Li HY, Xie LJ, Fan JY, Li SY, Yu WQ, Xu YT, He ML, Jiang Y, Bai X, Zhou J, Wang X. Intestinal flora was associated with occurrence risk of chronic non-communicable diseases. World J Gastroenterol 2025; 31:103507. [PMID: 40124279 PMCID: PMC11924013 DOI: 10.3748/wjg.v31.i11.103507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/26/2024] [Accepted: 02/24/2025] [Indexed: 03/13/2025] Open
Abstract
BACKGROUND The intestinal flora (IF) has been linked to risks of non-communicable diseases, especially various cancers, stroke, and Alzheimer's disease. However, many uncertainties of these associations during different stages of growth, development, and aging still exist. Therefore, further in-depth explorations are warranted. AIM To explore the associations of the human IF with disease risks during different stages of growth, development, and aging to achieve more accurate and convincing conclusions. METHODS Cohort, cross-sectional, case-control, and Mendelian randomization studies published in the PubMed and Web of Science databases until December 31, 2023 were systematically reviewed to clarify the associations of the IF at the genus level with the risks of various non-communicable diseases, which were grouped in accordance with the 10th revision of the International Classification of Diseases. RESULTS In total, 57 studies were included to quantitatively examine the influence of the IF on the risks of 30 non-communicable diseases during different stages of growth, development, and aging. Population studies and Mendelian randomization studies confirmed positive associations of the abundances of Bifidobacterium and Ruminococcus with multiple sclerosis. CONCLUSION These findings contribute to a deeper understanding of the roles of the IF and provide novel evidence for effective strategies for the prevention and treatment of non-communicable diseases. In the future, it will be necessary to explore a greater variety of research techniques to uncover the specific mechanisms by which gut microbiota trigger diseases and conduct in-depth studies on the temporal relationship between microbiota alterations and diseases, so as to clarify the causal relationship more accurately.
Collapse
Affiliation(s)
- Guo-Heng Jiang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hong-Yu Li
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lin-Jun Xie
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jing-Yuan Fan
- China Tobacco Sichuan Industry Co. Ltd., Technology Center, Chengdu 610101, Sichuan Province, China
| | - Shi-Yi Li
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Qian Yu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yi-Ting Xu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Meng-Lin He
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yi Jiang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xuan Bai
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jin Zhou
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Xin Wang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
16
|
Jha SS, Jeyaraman N, Jeyaraman M, Ramasubramanian S, Muthu S, Santos GS, da Fonseca LF, Lana JF. Cross-talks between osteoporosis and gut microbiome. World J Orthop 2025; 16:102274. [PMID: 40124724 PMCID: PMC11924030 DOI: 10.5312/wjo.v16.i3.102274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/06/2025] [Accepted: 02/06/2025] [Indexed: 03/12/2025] Open
Abstract
The gut microbiome comprises a vast community of microbes inhabiting the human alimentary canal, playing a crucial role in various physiological functions. These microbes generally live in harmony with the host; however, when dysbiosis occurs, it can contribute to the pathogenesis of diseases, including osteoporosis. Osteoporosis, a systemic skeletal disease characterized by reduced bone mass and increased fracture risk, has attracted significant research attention concerning the role of gut microbes in its development. Advances in molecular biology have highlighted the influence of gut microbiota on osteoporosis through mechanisms involving immunoregulation, modulation of the gut-brain axis, and regulation of the intestinal barrier and nutrient absorption. These microbes can enhance bone mass by inhibiting osteoclast differentiation, inducing apoptosis, reducing bone resorption, and promoting osteoblast proliferation and maturation. Despite these promising findings, the therapeutic effectiveness of targeting gut microbes in osteoporosis requires further investigation. Notably, gut microbiota has been increasingly studied for their potential in early diagnosis, intervention, and as an adjunct therapy for osteoporosis, suggesting a growing utility in improving bone health. Further research is essential to fully elucidate the therapeutic potential and clinical application of gut microbiome modulation in the management of osteoporosis.
Collapse
Affiliation(s)
- Shiva Shankar Jha
- Department of Orthopaedics, Harishchandra Orthopaedic Research Institute, Patna 880023, Bihar, India
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, São Paulo, Brazil
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College and Hospital, Karur 639004, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Gabriel Silva Santos
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, São Paulo, Brazil
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, São Paulo, Brazil
| | - José Fábio Lana
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, São Paulo, Brazil
| |
Collapse
|
17
|
Schieferecke AJ, Kuxhausen Ralph N, Schaffer DV. The Application of DNA Viruses to Biotechnology. Viruses 2025; 17:414. [PMID: 40143341 PMCID: PMC11946468 DOI: 10.3390/v17030414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
The delivery of biomolecules to target cells has been a longstanding challenge in biotechnology. DNA viruses naturally evolved the ability to deliver genetic material to cells and modulate cellular processes. As such, they inherently possess requisite characteristics that have led to their extensive study, engineering, and development as biotechnological tools. Here, we overview the application of DNA viruses to biotechnology, with specific implications in basic research, health, biomanufacturing, and agriculture. For each application, we review how an increasing understanding of virology and technological methods to genetically manipulate DNA viruses has enabled advances in these fields. Additionally, we highlight the remaining challenges to unlocking the full biotechnological potential of DNA viral technologies. Finally, we discuss the importance of balancing continued technological progress with ethical and biosafety considerations.
Collapse
Affiliation(s)
- Adam J. Schieferecke
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nadia Kuxhausen Ralph
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
| | - David V. Schaffer
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
18
|
Williams EG, Alissa M, Alsugoor MH, Albakri GS, Altamimi AA, Alabdullateef AA, Almansour NM, Aldakheel FM, Alessa S, Marber M. Integrative approaches to atrial fibrillation prevention and management: Leveraging gut health for improved cardiovascular outcomes in the aging population. Curr Probl Cardiol 2025; 50:102952. [PMID: 39626858 DOI: 10.1016/j.cpcardiol.2024.102952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/14/2024]
Abstract
Atrial fibrillation (AF) is a prevalent clinical arrhythmia associated with a high incidence and severe complications such as cerebral embolism and heart failure. While the etiology and pathogenesis of AF involve numerous factors, recent research emphasizes the significant role of intestinal microbiota imbalance in the emergence and progression of AF, particularly among older adults. This review investigates the mechanisms by which intestinal flora and their metabolites contribute to the onset of AF in the elderly, highlighting novel interactions between gut health and cardiac function. Current literature often overlooks these critical connections, indicating a substantial research gap in understanding how dysbiosis may exacerbate AF and hinder recovery. Furthermore, exploring the bidirectional relationship between the gut microbiome and systemic inflammation in the context of AF provides a unique perspective that has yet to be thoroughly investigated. Future research should focus on longitudinal studies assessing gut microbiota composition and function in AF patients and consider probiotics or prebiotics as potential adjunctive therapies for mitigating AF. This comprehensive approach may pave the way for innovative treatments integrating cardiology with gastroenterology, enhancing patient outcomes through a holistic understanding of health.
Collapse
Affiliation(s)
- Emma Grace Williams
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112; 2 Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Mahdi H Alsugoor
- Department of Emergency Medical Services, Faculty of Health Sciences, AlQunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia
| | - Ghadah Shukri Albakri
- Department of Teaching and Learning, College of Education and Human Development, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Ali A Altamimi
- Department of Medical Laboratory, Prince Sultan Air Base Hospital, Al-Kharj, Saudi Arabia
| | | | - Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| | - Fahad M Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Salem Alessa
- Department of Medical Laboratory, Al Kharj Military Industries Corporation Hospital, Al-kharj, Saudi Arabia
| | - Michael Marber
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| |
Collapse
|
19
|
Rana JS, Chi FW, Acquah I, Sterling SA. Unhealthy alcohol use and risk of coronary heart disease among young and middle-aged adults. Am J Prev Cardiol 2025; 21:100947. [PMID: 40027094 PMCID: PMC11869880 DOI: 10.1016/j.ajpc.2025.100947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/21/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
Objective To examine the associations between unhealthy alcohol use and risk of coronary heart disease (CHD) among women and men aged 18-65 years. Methods An observational study in an integrated healthcare system with systematic alcohol screening. We identified 432,265 primary care patients aged 18-65 years who, in 2014-2015, reported weekly alcohol intake levels. Weekly alcohol intake, categorized into below (≤14/week men; ≤7/week women) and above limits (≥15/week men; ≥ 8/week women) per U.S. guidelines, and heavy episodic drinking (HED, ≥5/≥4 drinks any day in past 3 months for men/women, respectively). Main outcome was CHD during 4-year follow-up, based on inpatient ICD diagnoses of myocardial infarction and CHD. Cox proportional hazards models adjusted for age, sex, race/ethnicity, body mass index, physical activity, smoking, hypertension, diabetes, and hyperlipidemia. Results The cohort comprised 44 % women, mean age (standard deviation) of 43.5 years (±13.1). Weekly alcohol intake above limits was associated with higher prevalence of cardiovascular risk factors, and a 26 %, 19 % and 43 % higher risk on the overall, men- and women-specific risk of CHD after adjusting for these risk factors (hazard ratio [95 % confidence interval] = 1.26[1.13 -1.40], 1.19[1.04-1.35] and 1.43[1.20-1.71], respectively). Conclusions In a large, real-world, diverse population with a systematic alcohol screening program, having weekly alcohol intake above limits was associated with increased risk of CHD among young and middle-aged men and women. Increased CHD risk due to alcohol intake above limits warrants particular awareness and interventions.
Collapse
Affiliation(s)
- Jamal S. Rana
- Department of Cardiology, The Permanente Medical Group. Oakland CA, USA
- Division of Research, Kaiser Permanente Northern California, Oakland CA, USA
| | - Felicia W. Chi
- Division of Research, Kaiser Permanente Northern California, Oakland CA, USA
| | - Isaac Acquah
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore MD, USA
| | - Stacy A. Sterling
- Division of Research, Kaiser Permanente Northern California, Oakland CA, USA
| |
Collapse
|
20
|
Luo C, Liu L, Zhu D, Ge Z, Chen Y, Chen F. Risk of stroke in patients with inflammatory bowel disease: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:114. [PMID: 40000943 PMCID: PMC11853978 DOI: 10.1186/s12876-025-03702-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Current studies suggest a potential link between inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), and cardiovascular diseases, such as stroke. This study aimed to assess the risk of stroke in IBD patients compared to general population. METHODS Systematic search was done in PubMed, Embase, CENTRAL, Scopus, and CINAHL databases for studies published till September 2023. Using a random-effects model, the hazard ratios (HRs) with 95% confidence intervals (CIs) for stroke occurrence were calculated. Subgroup analyses were done to estimate pooled HR with 95%CI for CD, UC, and overall IBD cases separately. Publication bias assessment was done by Begg's and Egger's tests. RESULTS Thirteen studies with 2,802,955 participants were included. IBD patients in general had significantly higher risk of stroke, with HR of 1.30 [95% CI 1.21-1.39]. Subgroup analysis demonstrated an HR of 1.35 [95% CI 1.22-1.49] for CD and 1.15 [95% CI 1.09-1.22] for UC. Substantial heterogeneity was detected across studies, with no substantial publication bias. Sensitivity analyses affirmed the stability of findings. CONCLUSION IBD in general, and Crohn's disease in particular are associated with significantly higher risk of stroke. Our findings further emphasize the importance of cardiovascular risk assessment and management strategies in IBD care. PROTOCOL REGISTRATION PROSPERO, CRD42023470602.
Collapse
Affiliation(s)
- Chao Luo
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Lingpei Liu
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Di Zhu
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Zuanmin Ge
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Yuehua Chen
- Department of General Practice, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China
| | - Feng Chen
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, 321000, China.
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua Municipal Central Hospital, No. 365, Renmin East Road, Jinhua City, Zhejiang Province, 321000, China.
| |
Collapse
|
21
|
Esposito S, McGuinness LR, Sharma P, Chadwick AE, Rainbow RD. Trimethylamine N-oxide (TMAO) acutely alters ionic currents but does not increase cardiac cell death. Front Physiol 2025; 16:1505813. [PMID: 40017801 PMCID: PMC11865236 DOI: 10.3389/fphys.2025.1505813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/20/2025] [Indexed: 03/01/2025] Open
Abstract
Background Trimethylamine N-oxide (TMAO) is a product of the action of gut microbiota on choline and other choline-containing compounds ingested in the diet. The presence of TMAO at high concentrations has been reported in the blood of patients with cardiovascular disease, suggesting the role for TMAO as either a marker or causative agent of the disease. These investigations examined whether TMAO had an effect on cardiomyocyte contractile function, calcium homoeostasis, and survival from metabolic insult. Results TMAO had no effect on metabolic function or the ability of cells to survive a metabolic insult; however, it did cause transient changes to contractile function. These changes included an increase in calcium current and an increase in Kir6.1 channel activity in the cell, causing a shortening of the action potential duration to 90% repolarised but lengthening the action potential to 30% repolarised. These effects occurred within minutes of TMAO application; however, they were not observed following 24 h culture. These data suggest that TMAO does modulate contractile function, albeit only in the short-term, but has no effect on metabolic behaviour or the ability to withstand a metabolic challenge. Conclusion These data suggest that high TMAO concentrations in the blood of patients may be a marker of potential cardiovascular disease rather than playing a causative role.
Collapse
Affiliation(s)
- Simona Esposito
- Department of Cardiovascular Sciences, Glenfield General Hospital, University of Leicester, Leicester, United Kingdom
- Department of Cardiovascular & Metabolic Medicine and Liverpool Centre for Cardiovascular Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Lauren R. McGuinness
- Department of Cardiovascular & Metabolic Medicine and Liverpool Centre for Cardiovascular Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Parveen Sharma
- Department of Cardiovascular & Metabolic Medicine and Liverpool Centre for Cardiovascular Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Amy E. Chadwick
- Department of Pharmacology and Therapeutics, Molecular and Integrative Biology, Institute of Systems, Liverpool, Merseyside, United Kingdom
| | - Richard D. Rainbow
- Department of Cardiovascular & Metabolic Medicine and Liverpool Centre for Cardiovascular Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, Merseyside, United Kingdom
| |
Collapse
|
22
|
Deslande M, Puig-Castellvi F, Castro-Dionicio I, Pacheco-Tapia R, Raverdy V, Caiazzo R, Lassailly G, Leloire A, Andrikopoulos P, Kahoul Y, Zaïbi N, Toussaint B, Oger F, Gambardella N, Lefebvre P, Derhourhi M, Amanzougarene S, Staels B, Pattou F, Froguel P, Bonnefond A, Dumas ME. Intrahepatic levels of microbiome-derived hippurate associates with improved metabolic dysfunction-associated steatotic liver disease. Mol Metab 2025; 92:102090. [PMID: 39746606 PMCID: PMC11772989 DOI: 10.1016/j.molmet.2024.102090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterised by lipid accumulation in the liver and is often associated with obesity and type 2 diabetes. The gut microbiome recently emerged as a significant player in liver metabolism and health. Hippurate, a host-microbial co-metabolite has been associated with human gut microbial gene richness and with metabolic health. However, its role on liver metabolism and homeostasis is poorly understood. METHODS We characterised liver biospies from 318 patients with obesity using RNAseq and metabolomics in liver and plasma to derive associations among hepatic hippurate, hepatic gene expression and MASLD and phenotypes. To test a potential beneficial role for hippurate in hepatic insulin resistance, we profile the metabolome of (IHH) using ultra-high-performance liquid chromatography coupled to high-resolution tandem mass spectrometry (UHPLC-MS/MS), and characterised intracellular triglyceride accumulation and glucose internalisation after a 24 h insulin exposure. RESULTS We first report significant associations among MASLD traits, plasma and hepatic hippurate. Further analysis of the hepatic transcriptome shows that liver and plasma hippurate are inversely associated with MASLD, implicating lipid metabolism and regulation of inflammatory responses pathways. Hippurate treatment inhibits lipid accumulation and rescues insulin resistance induced by 24-hour chronic insulin in IHH. Hippurate also improves hepatocyte metabolic profiles by increasing the abundance of metabolites involved in energy homeostasis that are depleted by chronic insulin treatment while decreasing those involved in inflammation. CONCLUSIONS Altogether, our results further highlight hippurate as a mechanistic marker of metabolic health, by its ability to improve metabolic homeostasis as a postbiotic candidate.
Collapse
Affiliation(s)
- Maxime Deslande
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Francesc Puig-Castellvi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Inés Castro-Dionicio
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Romina Pacheco-Tapia
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Violeta Raverdy
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Robert Caiazzo
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Guillaume Lassailly
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Audrey Leloire
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Petros Andrikopoulos
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Yasmina Kahoul
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Nawel Zaïbi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Bénédicte Toussaint
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Frédérik Oger
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Nicolas Gambardella
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Philippe Lefebvre
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Mehdi Derhourhi
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Souhila Amanzougarene
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France
| | - Bart Staels
- INSERM U1011 Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - François Pattou
- INSERM U1190, Institut Pasteur de Lille, University of Lille, Lille University Hospital, 59045, Lille, France
| | - Philippe Froguel
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Amélie Bonnefond
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom
| | - Marc-Emmanuel Dumas
- University of Lille, Lille University hospital, 59045, Lille, France; INSERM U1283, CNRS UMR 8199, Institut Pasteur de Lille, 59045, Lille, France; Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, United Kingdom; The Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, H3A 0G1, Canada.
| |
Collapse
|
23
|
Chen J, Zhu T, Yang J, Shen M, Wang D, Gu B, Xu J, Zhang M, Hao X, Tang Z, Tong J, Du Y, Zhang B, Li H, Xu M. Geniposide Protects Against Myocardial Infarction Injury via the Restoration in Gut Microbiota and Gut-Brain Axis. J Cell Mol Med 2025; 29:e70406. [PMID: 39910683 PMCID: PMC11798748 DOI: 10.1111/jcmm.70406] [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: 04/25/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
Improving gut dysbiosis and impaired gut-brain axis has been a potent therapeutic strategy for treating myocardial infarction (MI). Geniposide (GEN), a traditional Chinese medicine extract, has demonstrated substantial cardioprotective properties post-MI. Nevertheless, the effect of GEN on gut microbial, gut-brain communication, and its potential mechanism remains unclear. In this study, we initially found that GEN significantly alleviated MI-induced cardiac dysfunction from echocardiographic data and decreased myocardial fibrosis, inflammation, apoptosis and hypertrophy post-MI. Additionally, we investigated the effects of GEN on gut pathology, and observed that GEN led to a remarkable change in gut microbiota as evidenced by altering β-diversity and short-chain fatty acids (SCFAs) levels, and alleviated intestinal damage indicated by reduced inflammation and barrier permeability post-MI. Finally, our investigation into brain pathology revealed that GEN induced a remarkable inhibition in PVN inflammation and sympathetic activity following MI. Collectively, these findings imply that the cardioprotective effects of GEN against MI were mediated possibly via an improvement in the impaired gut-brain axis. Mechanically, GEN-induced increase of microbiota-derived SCFAs might be the critical factor linking gut microbiota and reduced neuroinflammation with PVN, which leads to the suppression of sympathetic activation, therefore protecting the myocardium against MI-induced damage.
Collapse
Affiliation(s)
- Jie Chen
- The Affiliated Xi'an International Medical Center HospitalNorthwest UniversityXi'anShaanxiChina
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Tong Zhu
- The Affiliated Xi'an International Medical Center HospitalNorthwest UniversityXi'anShaanxiChina
| | - Jinbao Yang
- The Affiliated Xi'an International Medical Center HospitalNorthwest UniversityXi'anShaanxiChina
| | - Mengqing Shen
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Danmei Wang
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Boyuan Gu
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Jin Xu
- The Affiliated Xi'an International Medical Center HospitalNorthwest UniversityXi'anShaanxiChina
| | - Mingxia Zhang
- The Affiliated Xi'an International Medical Center HospitalNorthwest UniversityXi'anShaanxiChina
| | - Xiuli Hao
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Zheng Tang
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Jie Tong
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Yan Du
- Department of NephrologyThe First Affiliated Hospital of Xi'an Medical UniversityXi'anShaanxiChina
| | - Bao Zhang
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Hongbao Li
- Department of Physiology and PathophysiologyXi'an Jiaotong University School of Basic Medical SciencesXi'anChina
| | - MengLu Xu
- Department of NephrologyThe First Affiliated Hospital of Xi'an Medical UniversityXi'anShaanxiChina
| |
Collapse
|
24
|
Yang C, Zhong H. Multiomics Analyses Demonstrate the Attenuation of Metabolic Cardiac Disorders Associated With Type 2 Diabetes by Stachydrine in Relation With the Transition of Gastrointestinal Microbiota. Chem Biol Drug Des 2025; 105:e70066. [PMID: 39994907 DOI: 10.1111/cbdd.70066] [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: 09/30/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/26/2025]
Abstract
Stachydrine (STA) has therapeutic effects on heart disorders. The current study assessed its effects on Type 2 diabetes (T2D) induced cardiac disorders by focusing on the heart-gut axis. Mice were subjected to high-fat diet (HFD) and streptozocin (STZ) to induce cardiac disorders such as inflammation and structural deteriorations, which were handled with STA. Changes regarding the composition and metabolism of gastrointestinal (GI) microbiota were then determined using a multiomics strategy, including amplicon sequencing and metabolomics. The data showed that STA improved heart function, reduced intestinal permeability, and suppressed inflammation in mice in a dose-dependent manner. However, the compound had little influence on the overall alpha diversity of gut microbiota, while it did influence the beta diversity. The analyses based on the multiomics strategy demonstrated that certain GI microbial groups, including Paramuribaculum, Allobaculum, Bifidobacterium, and Adlercreutzia, responded to the STA administration, which contributed to the alternatives of metabolites in the gut. Correlation analyses showed that Duncaniella and Ruminococcus negatively impacted health, while Muribaculum, Paramuribaculum, and Prevotella positively influenced intestinal permeability and heart health. Collectively, STA attenuated T2D-induced cardiac disorders by improving heart structure and function and suppressing inflammation, during which the GI homeostasis of the T2D mice changed to an alternative state that was different from that of healthy mice.
Collapse
Affiliation(s)
- Chaoxing Yang
- Department of Cardiovascular Medicine, Ganzhou People's Hospital, Ganzhou, China
| | - Huaping Zhong
- Heart Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
25
|
Li L, Zhong S, Ye J, Hu S, Xiong X, Chen G, Hu Z. Shenmai injection revives cardiac function in rats with hypertensive heart failure: involvement of microbial-host co-metabolism. BMC Complement Med Ther 2025; 25:24. [PMID: 39856640 PMCID: PMC11761217 DOI: 10.1186/s12906-024-04737-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
Heart failure (HF) is a complex syndrome marked by considerable expenditures and elevated mortality and morbidity rates globally. Shenmai injection (SMI), a form of Traditional Chinese Medicine-based therapy, has demonstrated effectiveness in treating HF. Recent research suggests that Traditional Chinese Medicine (TCM) may induce beneficial changes in microbial-host co-metabolism, potentially providing cardiovascular protection. This study used a rat model of hypertensive heart failure (H-HF) to explore the mechanism of SMI. The possible compounds and key targets of SMI against H-HF were investigated using network pharmacology. The pharmacodynamics of SMI were validated using the H-HF animal model, with analysis of fecal gut microbiota integrating metabolomics and 16S rRNA sequencing. Metorigin metabolite traceability analysis and the MetaboAnalyst platform were utilized to explore the action mechanism. To evaluate changes in serum TMAO levels, targeted metabolomics was performed. Finally, the study looked at the intrinsic relationships among modifications in the intestinal flora, metabolite profile changes, and the targets of SMI compounds to clarify how they might be used to treat H-HF. According to metabolomics and 16S rRNA sequencing, by reestablishing homeostasis in the gut microbiota, SMI affects vital metabolic pathways, such as energy metabolism, amino acid metabolism, and bile acid metabolism. Increased serum TMAO levels were identified to be a risk factor for H-HF, and SMI was able to downregulate the levels of TMAO-related metabolites. Network pharmacology analysis identified 13 active components of SMI targeting 46 proteins, resulting in differential expression changes in 8 metabolites and 24 gut microbes. In conclusion, this study highlights the effectiveness of SMI in alleviating H-HF and its potential to modulate microbial-host co-metabolism. Through a comprehensive discussion of the interconnected relationships among the components, targets, metabolites, and gut microbiota, it provided fresh light on the therapeutic mechanism of SMI on H-HF.
Collapse
Affiliation(s)
- Lin Li
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Senjie Zhong
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiahao Ye
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyuan Hu
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiajun Xiong
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Guangyu Chen
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Engineering Technology Research Center For Medicinal and Functional Food, Changsha, Hunan, China
| | - Zhixi Hu
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
26
|
Wang S, Gao L, Wang C, Bai J, Shen M, Zhao X, Lin M. Effects of internet-based cognitive behavioral therapy on anxiety and depressive symptoms among patients with cardiovascular and cerebrovascular diseases: a systematic review and meta-analysis. Front Psychiatry 2025; 15:1433558. [PMID: 39886052 PMCID: PMC11779714 DOI: 10.3389/fpsyt.2024.1433558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025] Open
Abstract
Background This study aimed to evaluate the effectiveness of Internet-based Cognitive Behavioral Therapy (ICBT) in reducing anxiety and depressive symptoms among patients with cardiovascular diseases (CVDs) and to explore how intervention characteristics, such as module number and program duration, influence treatment outcomes. Methods A systematic review and meta-analysis were conducted by searching eight databases, including PubMed, Embase, and Cochrane Library, for randomized controlled trials (RCTs) published up to December 2023. Studies involving adult CVD patients with anxiety or depressive symptoms who underwent ICBT interventions were included. Statistical analyses used random-effects models, with subgroup analyses performed to assess the impact of intervention format, module number, and program duration. Sensitivity and publication bias assessments ensured the robustness of the findings. Results Eight RCTs with 1177 participants were included. ICBT significantly reduced depressive symptoms (SMD = -0.32, 95% CI [-0.56, -0.08], p < 0.015) and anxiety symptoms (SMD = -0.37, 95% CI [-0.68, -0.06], p < 0.001). Subgroup analysis indicated that self-guided ICBT was more effective than therapist-guided ICBT. Programs with fewer than eight modules were more effective for anxiety, while those with eight or more modules were more effective for depression. Shorter programs (< 9 weeks) were better for anxiety, whereas longer programs (≥ 9 weeks) were more effective for depression. Conclusions ICBT is an effective intervention for managing anxiety and depression in CVD patients. Tailoring ICBT interventions based on symptom type, module number, and program duration can optimize outcomes. Future research should explore personalized, long-term strategies to enhance effectiveness and safety.
Collapse
Affiliation(s)
- Shuangyu Wang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lishuo Gao
- School of Nursing, Tianjin Medical University, Tianjin, China
| | - Congyu Wang
- Department of Graduate, Tianjin Medical University, Tianjin, China
| | - Jinbing Bai
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Mengshuang Shen
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuejie Zhao
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mei Lin
- Department of Nursing, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
27
|
Franz K, Markó L, Mähler A, Chakaroun R, Heinitz S, Schlögl H, Sacher J, Steckhan N, Dechend R, Adams N, Andersen M, Glintborg D, Viehweger M, Bahr LS, Forslund-Startceva SK. Sex hormone-dependent host-microbiome interactions and cardiovascular risk (XCVD): design of a longitudinal multi-omics cohort study. BMJ Open 2025; 15:e087982. [PMID: 39788783 PMCID: PMC11751863 DOI: 10.1136/bmjopen-2024-087982] [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: 04/24/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Cardiovascular diseases (CVDs) present differently in women and men, influenced by host-microbiome interactions. The roles of sex hormones in CVD outcomes and gut microbiome in modifying these effects are poorly understood. The XCVD study examines gut microbiome mediation of sex hormone effects on CVD risk markers by observing transgender participants undergoing gender-affirming hormone therapy (GAHT), with findings expected to extrapolate to cisgender populations. METHODS AND ANALYSES This observational, longitudinal cohort study includes baseline, 1- and 2-year follow-ups with transgender participants beginning GAHT. It involves comprehensive phenotyping and microbiome genotyping, integrating computational analyses of high-dimensional data. Microbial diversity will be assessed using gut, skin, and oral samples via 16S rRNA and shotgun metagenomic sequencing of gut samples. Blood measurements will include sex hormones, CVD risk markers, cardiometabolic parameters, cytokines, and immune cell counts. Hair samples will be analysed for cortisol. Participants will complete online questionnaires on physical activity, mental health, stress, quality of life, fatigue, sleep, pain, and gender dysphoria, tracking medication use and diet to control for confounders. Statistical analyses will integrate phenomic, lifestyle, and multi-omic data to model health effects, testing gut microbiome mediation of CVD risk as the endocrine environment shifts between that typical for cisgender men to women and vice versa. ETHICS AND DISSEMINATION The study adheres to Good Clinical Practice and the Declaration of Helsinki. The protocol was approved by the Charité Ethical Committee (EA1/339/21). Signed informed consent will be obtained. Results will be published in peer-reviewed journals and conferences and shared as accessible summaries for participants, community groups, and the public, with participants able to view their data securely after public and patient involvement review for accessibility. TRIAL REGISTRATION NUMBER The XCVD study was registered on ClinicalTrials.gov (NCT05334888) as 'Sex-differential host-microbiome CVD risk - a longitudinal cohort approach (XCVD)" on 4 April 2022. Data set link can be found at https://classic. CLINICALTRIALS gov/ct2/show/NCT05334888.
Collapse
Affiliation(s)
- Kristina Franz
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) partner site Berlin, DZHK, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lajos Markó
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) partner site Berlin, DZHK, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Anja Mähler
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rima Chakaroun
- Medical Department III Endocrinology Nephrology Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
- Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg Wallenberg Laboratory for Cardiovascular and Metabolic Research, Goteborg, Sweden
| | - Sascha Heinitz
- Medical Department III Endocrinology Nephrology Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Haiko Schlögl
- Medical Department III Endocrinology Nephrology Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
- HI-MAG, Helmholtz Institute for Metabolic Obesity and Vascular Research, Leipzig, Germany
| | - Julia Sacher
- Clinic for Cognitive Neurology, University of Leipzig Medical Center, and Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Max-Planck-Institut fur molekulare Physiologie, Dortmund, Germany
| | - Nico Steckhan
- Digital Health - Connected Healthcare, Hasso Plattner Institute, University of Potsdam, Potsdam, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) partner site Berlin, DZHK, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Helios Clinic Berlin-Buch, Berlin, Germany
| | - Noah Adams
- University of Toronto, Toronto, Ontario, Canada
- Center for Applied Transgender Studies (CATS), Chicago, Illinois, USA
- Transgender Professional Association for Transgender Health, TPATH, Toronto, Ontario, Canada
| | - Marianne Andersen
- Department of Endocrinology, Odense Universitetshospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Syddanmark, Denmark
| | - Dorte Glintborg
- Institute of Clinical Research, University of Southern Denmark, Odense, Syddanmark, Denmark
- Body Identity Clinic, Odense Universitetshospital Endokrinologisk Afdeling M, Odense, Denmark
| | | | - Lina Samira Bahr
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) partner site Berlin, DZHK, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sofia Kirke Forslund-Startceva
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) partner site Berlin, DZHK, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Transgender Professional Association for Transgender Health, TPATH, Toronto, Ontario, Canada
- European Molecular Biology Laboratory Structural and Computational Biology Unit, Heidelberg, Baden-Württemberg, Germany
| |
Collapse
|
28
|
Abdulrahim AO, Doddapaneni NSP, Salman N, Giridharan A, Thomas J, Sharma K, Abboud E, Rochill K, Shreelakshmi B, Gupta V, Lakkimsetti M, Mowo-Wale A, Ali N. The gut-heart axis: a review of gut microbiota, dysbiosis, and cardiovascular disease development. Ann Med Surg (Lond) 2025; 87:177-191. [PMID: 40109640 PMCID: PMC11918638 DOI: 10.1097/ms9.0000000000002789] [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: 06/25/2024] [Accepted: 11/20/2024] [Indexed: 03/22/2025] Open
Abstract
Background Cardiovascular diseases (CVDs) are a major cause of morbidity and mortality worldwide and there are strong links existing between gut health and cardiovascular health. Gut microbial diversity determines gut health. Dysbiosis, described as altered gut microbiota, causes bacterial translocations and abnormal gut byproducts resulting in systemic inflammation. Objective To review the current literature on the relationships between gut microbiota, dysbiosis, and CVD development, and explore therapeutic methods to prevent dysbiosis and support cardiovascular health. Summary Dysbiosis increases levels of pro-inflammatory substances while reducing those of anti-inflammatory substances. This accumulative inflammatory effect negatively modulates the immune system and promotes vascular dysfunction and atherosclerosis. High Firmicutes to Bacteroidetes ratios, high trimethylamine-n-oxide to short-chain fatty acid ratios, high indole sulfate levels, low cardiac output, and polypharmacy are all associated with worse cardiovascular outcomes. Supplementation with prebiotics and probiotics potentially alleviates some CVD risk. Blood and stool samples may be used in clinical practice to quantify and qualify gut bacterial ratios and byproducts, assess patients' risk for adverse cardiovascular outcomes, and track their gut health progress. Further research is required to set population-based cutoffs for normal and abnormal gut microbiota and byproduct ratios.
Collapse
Affiliation(s)
| | | | - Nadhra Salman
- Department of Internal Medicine, Baqai Medical University, Karachi, Pakistan
| | | | | | - Kavya Sharma
- Maharishi Markandeshwar Medical College and Hospital, Himachal Pradesh, India
| | - Elias Abboud
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | | | - B Shreelakshmi
- Navodaya Medical College Hospital & Research Centre, Karnataka, India
| | | | | | | | - Noor Ali
- Dubai Medical College, Dubai, United Arab Emirates
| |
Collapse
|
29
|
Escobar C, Aldeguer X, Vivas D, Manzano Fernández S, Gonzalez Caballero E, Garcia Martín A, Barrios V, Freixa-Pamias R. The gut microbiota and its role in the development of cardiovascular disease. Expert Rev Cardiovasc Ther 2025; 23:23-34. [PMID: 39915986 DOI: 10.1080/14779072.2025.2463366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION The pathophysiology of cardiovascular diseases encompasses a complex interplay of genetic and environmental risk factors. Even if traditional risk factors are treated to target, there remains a residual risk. AREAS COVERED This manuscript reviews the potential role of gut microbiota in the development of cardiovascular disease, and as potential target. A systematic search was conducted until 30 October 2024 on PubMed (MEDLINE), using the MeSH terms [Gut microbiota] + [Dysbiosis] + [Cardiovascular] + [TMAO] + [bile acids] + [short-chain fatty acids]. EXPERT OPINION The term dysbiosis implies changes in equilibrium, with modifications in the composition and functionality of microbiota and a series of additional factors: reduced diversity and uniformity of microorganisms; reduced short-chain fatty acid-producing bacteria; increased gut permeability; release of metabolites, such as trimethylamine N-oxide, betaine, phenylalanine, tryptophan-kynurenine, phenylacetylglutamine, and lipopolysaccharides; and reduced secondary bile acid excretion, leading to inflammation, oxidative stress, and endothelial dysfunction and facilitating the onset of pathological conditions, including obesity, hypertension, diabetes, atherosclerosis, and heart failure. Attempts to restore gut microbiota balance through different interventions, mainly changes in diet, have been shown to positively affect individual components and metabolites and reduce the risk of cardiovascular disease. In addition, probiotics and prebiotics are potentially useful. Fecal microbiota transplantation is a promising therapy.
Collapse
Affiliation(s)
- Carlos Escobar
- Cardiology Department, University Hospital La Paz, Madrid, Spain
| | - Xavier Aldeguer
- Gastroenterology Department, Hospital Doctor Josep Trueta i Santa Caterina, Institut d'investigació Biomèdica de Girona IDIBGI, Girona/Salt, Spain
| | - David Vivas
- Cardiovascular Institute, San Carlos University Hospital, Madrid, Spain
- Cardiology Department, Cardiovascular Institute Vithas Milagrosa and Aravaca, Madrid, Spain
| | | | | | - Ana Garcia Martín
- Cardiology Department, University Hospital Ramón y Cajal, Alcalá University, Madrid, Spain
| | - Vivencio Barrios
- Cardiology Department, University Hospital Ramón y Cajal, Alcalá University, Madrid, Spain
| | - Román Freixa-Pamias
- Cardiology Department, Complex Hospitalari Moisès Broggi, Sant Joan Despí, Barcelona, Spain
| |
Collapse
|
30
|
Gnanasekaran D, Pandurangan AR, Ramaswamy PS, Raghavendra SK, Raman K, Somasundaram GS. Exploring the gut-heart axis: A prospective analysis of microbiota in cardiovascular health. Bioinformation 2024; 20:1900-1903. [PMID: 40230898 PMCID: PMC11993406 DOI: 10.6026/9732063002001900] [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: 12/01/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 04/16/2025] Open
Abstract
The relationship between gut microbiota composition and the development of cardiovascular disease, with a potential role of microbial metabolites in inflammatory and metabolic pathways is of interest. We analyzed gut microbiota and markers of cardiovascular health in a cohort of 100 participants for three years to search for microbial signatures correlated with increased CVD risk. Our results show several correlations between specific microbial taxa, lipid metabolism and systemic inflammation, whereby a higher Firmicutes/Bacteroides ratio is associated with a greater incidence of CVD. These results suggest that intervention targeting the microbiome has the potential to reduce risk for CVD and point towards a role for gut microbiota in cardiovascular health.
Collapse
Affiliation(s)
- Deepika Gnanasekaran
- Department of Internal Medicine, Hillingdon Hospital, Uxbridge, London, United Kingdom
| | | | | | - Shrunga Kandhi Raghavendra
- Department of Internal Medicine, Chamarajanagar Institute of Medical Sciences (CIMS), Kasaba Hobli, Chamarajanagar, Karnataka, India
| | - Karthick Raman
- Department of Cardiology, Government Sivagangai Medical College Hospital, Sivagangai, Tamil Nadu, India
| | | |
Collapse
|
31
|
Rohun J, Dudzik D, Raczak-Gutknecht J, Wabich E, Młodziński K, Markuszewski MJ, Daniłowicz-Szymanowicz L. Metabolomics in Atrial Fibrillation: Unlocking Novel Biomarkers and Pathways for Diagnosis, Prognosis, and Personalized Treatment. J Clin Med 2024; 14:34. [PMID: 39797116 PMCID: PMC11722095 DOI: 10.3390/jcm14010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Atrial fibrillation (AF) is the most frequent arrhythmia in the adult population associated with a high rate of severe consequences leading to significant morbidity and mortality worldwide. Therefore, its prompt recognition is of high clinical importance. AF detection often remains challenging due to unspecific symptoms and a lack of reliable biomarkers for its prediction. Herein, novel bioanalytical methodologies, such as metabolomics, offer new opportunities for a better understanding of the underlying pathological mechanisms of cardiovascular diseases, including AF. The metabolome, considered a complete set of small molecules present in the organism, directly reflects the current phenotype of the studied system and is highly sensitive to any changes, including arrhythmia's onset. A growing body of evidence suggests that metabolite profiling has prognostic value in AF prediction, highlighting its potential role not only in early diagnosis but also in guiding therapeutic interventions. By identifying specific metabolites as a disease biomarker or recognising particular metabolomic pathways involved in the AF pathomechanisms, metabolomics could be of great clinical value for further clinical decision-making, risk stratification, and an individual personalised approach. The presented narrative review aims to summarise the current state of knowledge on metabolomics in AF with a special emphasis on its implications for clinical practice and personalised medicine.
Collapse
Affiliation(s)
- Justyna Rohun
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Danuta Dudzik
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Joanna Raczak-Gutknecht
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Elżbieta Wabich
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Krzysztof Młodziński
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Michał J. Markuszewski
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Ludmiła Daniłowicz-Szymanowicz
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| |
Collapse
|
32
|
Wang Q, Liu M, Liu T, Li L, Wang C, Wang X, Rong S, Zhou X. Alterations in the gut microbiome and metabolism with doxorubicin-induced heart failure severity. Front Microbiol 2024; 15:1348403. [PMID: 39777147 PMCID: PMC11703658 DOI: 10.3389/fmicb.2024.1348403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/14/2024] [Indexed: 01/11/2025] Open
Abstract
Objective This study aimed to explore the changes in gut microbiota and its metabolites in different pathophysiological stages of doxorubicin (DOX)-induced heart failure (DIHF) and the relationship between gut microbiota and metabolites in various degrees of DIHF. Materials and methods C57BL/6 J mice were injected intraperitoneally with 5 mg/kg of DOX once a week for 5 consecutive weeks. At different times after injection, the cardiac function and histopathological analysis was conducted, the serum levels of creatine kinase (CK), CK-MB, lactic dehydrogenase, and cardiac troponin T were determined. 16S rRNA gene sequencing of feces and the nontargeted metabolomics analysis of serum were performed. Multi-omics analyses were used to explore the correlation between gut microbiota and serum metabolites. Results The results showed that DOX caused cardiac contractile dysfunction and left ventricular (LV) dilation. The levels of myocardial enzymes significantly increase in 3 and 5 weeks after DOX injection. DOX-treated mice showed significant differences in the composition and abundance of gut microorganisms, and the levels of serum metabolites at different times of treatment. Multi-omics analyses showed that intestinal bacteria were significantly correlated with the differential metabolites. Some bacteria and metabolites can be used as biomarkers of DIHF (AUC > 0.8). KEGG analyses showed the involvement of different metabolic pathways in various degrees of DIHF. Conclusion Marked differences were found in the composition and abundance of gut microorganisms, the levels of serum metabolites and metabolic pathways in different degrees of DIHF. The intestinal bacteria were significantly correlated with differential metabolites in different degrees of DIHF. The gut microbiota may serve as new targets for the treatment of DIHF.
Collapse
Affiliation(s)
- Qian Wang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Meihua Liu
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Academy of Medical Sciences, The Shanxi Medical University, Taiyuan, China
| | - Tianpei Liu
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Academy of Medical Sciences, The Shanxi Medical University, Taiyuan, China
| | - Long Li
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chenyang Wang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaolin Wang
- Department of Neonatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuling Rong
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Chen W, Xiao L, Guo W, Li H, Chen R, Duan Z, Chen Q, Lei Q. Research progress of traditional Chinese medicine regulating intestinal flora in the treatment of hypertension. Front Pharmacol 2024; 15:1449972. [PMID: 39717555 PMCID: PMC11664361 DOI: 10.3389/fphar.2024.1449972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Hypertension is a common disease; however, it is more prevalent in older adults, and its prevalence is increasing in younger populations. Numerous studies have revealed that hypertension and the composition and functionality of the intestinal flora are closely correlated. The balance of the intestinal flora, intestinal barrier integrity, and metabolite content of the intestinal flora play significant roles in the occurrence and progression of hypertension. Therefore, we performed a comprehensive review of Traditional Chinese medicine (TCM) for hypertension, focusing on the role of the intestinal flora to understand the mechanism by which TCM regulates hypertension through its effects on the intestinal flora. We analyzed the findings using the terms "traditional Chinese medicine," "hypertension," "high blood pressure," "blood pressure," "intestinal flora," "intestinal barrier function," "intestinal flora metabolites," and other keywords from the China National Knowledge Infrastructure, VIP Chinese Science and Technology, Wanfang Data, PubMed, and ScienceDirect databases. We found that TCM treats hypertension by regulating the balance of the intestinal microbiota, increasing the abundance of beneficial bacteria, reducing the abundance of harmful bacteria, improving intestinal barrier function, increasing compact proteins, reducing intestinal permeability, and regulating the content of intestinal flora metabolites. The use of TCM to treat hypertension by regulating the intestinal flora is a promising therapeutic strategy. However, most studies are limited by small sample sizes and there is a lack of large-scale randomized controlled trials. In the future, multi-center controlled clinical trials are needed to verify the efficacy and safety of TCM, optimize therapeutic protocols, and establish a foundation for the standardized and personalized application of TCM in hypertension management.
Collapse
Affiliation(s)
- Wenjun Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Longfei Xiao
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Wenlong Guo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Hailin Li
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Rong Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhongyu Duan
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qinghua Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qing Lei
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
34
|
Wang H, Shi C, Jiang L, Liu X, Tang R, Tang M. Neuroimaging techniques, gene therapy, and gut microbiota: frontier advances and integrated applications in Alzheimer's Disease research. Front Aging Neurosci 2024; 16:1485657. [PMID: 39691161 PMCID: PMC11649678 DOI: 10.3389/fnagi.2024.1485657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder marked by cognitive decline, for which effective treatments remain elusive due to complex pathogenesis. Recent advances in neuroimaging, gene therapy, and gut microbiota research offer new insights and potential intervention strategies. Neuroimaging enables early detection and staging of AD through visualization of biomarkers, aiding diagnosis and tracking of disease progression. Gene therapy presents a promising approach for modifying AD-related genetic expressions, targeting amyloid and tau pathology, and potentially repairing neuronal damage. Furthermore, emerging evidence suggests that the gut microbiota influences AD pathology through the gut-brain axis, impacting inflammation, immune response, and amyloid metabolism. However, each of these technologies faces significant challenges, including concerns about safety, efficacy, and ethical considerations. This article reviews the applications, advantages, and limitations of neuroimaging, gene therapy, and gut microbiota research in AD, with a particular focus on their combined potential for early diagnosis, mechanistic insights, and therapeutic interventions. We propose an integrated approach that leverages these tools to provide a multi-dimensional framework for advancing AD diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Haitao Wang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Shi
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ling Jiang
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaozhu Liu
- Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Rui Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxi Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Yaan People’s Hospital (Yaan Hospital of West China Hospital of Sichuan University), Yaan, Sichuan, China
| |
Collapse
|
35
|
Zhu T, Chen J, Zhang M, Tang Z, Tong J, Hao X, Li H, Xu J, Yang J. Tanshinone IIA Exerts Cardioprotective Effects Through Improving Gut-Brain Axis Post-Myocardial Infarction. Cardiovasc Toxicol 2024; 24:1317-1334. [PMID: 39377990 PMCID: PMC11564317 DOI: 10.1007/s12012-024-09928-4] [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: 05/29/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024]
Abstract
Myocardial infarction (MI) is a lethal cardiovascular disease worldwide. Emerging evidence has revealed the critical role of gut dysbiosis and impaired gut-brain axis in the pathological progression of MI. Tanshinone IIA (Tan IIA), a traditional Chinese medicine, has been demonstrated to exert therapeutic effects for MI. However, the effects of Tan IIA on gut-brain communication and its potential mechanisms post-MI are still unclear. In this study, we initially found that Tan IIA significantly reduced myocardial inflammation, apoptosis and fibrosis, therefore alleviating hypertrophy and improving cardiac function following MI, suggesting the cardioprotective effect of Tan IIA against MI. Additionally, we observed that Tan IIA improved the gut microbiota as evidenced by changing the α-diversity and β-diversity, and reduced histopathological impairments by decreasing inflammation and permeability in the intestinal tissues, indicating the substantial improvement of Tan IIA in gut function post-MI. Lastly, Tan IIA notably reduced lipopolysaccharides (LPS) level in serum, inflammation responses in paraventricular nucleus (PVN) and sympathetic hyperexcitability following MI, suggesting that restoration of Tan IIA on MI-induced brain alterations. Collectively, these results indicated that the cardioprotective effects of Tan IIA against MI might be associated with improvement in gut-brain axis, and LPS might be the critical factor linking gut and brain. Mechanically, Tan IIA-induced decreased intestinal damage reduced LPS release into serum, and reduced serum LPS contributes to decreased neuroinflammation with PVN and sympathetic inactivation, therefore protecting the myocardium against MI-induced injury.
Collapse
Affiliation(s)
- Tong Zhu
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China
| | - Jie Chen
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Mingxia Zhang
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China
| | - Zheng Tang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Jie Tong
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiuli Hao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Hongbao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, 710061, China
| | - Jin Xu
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China.
| | - Jinbao Yang
- The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710161, Shaanxi, China.
| |
Collapse
|
36
|
Meng Y, Sun J, Zhang G, Yu T, Piao H. Fasting: A Complex, Double-Edged Blade in the Battle Against Doxorubicin-Induced Cardiotoxicity. Cardiovasc Toxicol 2024; 24:1395-1409. [PMID: 39354217 DOI: 10.1007/s12012-024-09925-7] [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: 12/23/2023] [Accepted: 09/23/2024] [Indexed: 10/03/2024]
Abstract
In recent years, there has been a surge in the popularity of fasting as a method to enhance one's health and overall well-being. Fasting is a customary practice characterized by voluntary refraining from consuming food and beverages for a specified duration, ranging from a few hours to several days. The potential advantages of fasting, including enhanced insulin sensitivity, decreased inflammation, and better cellular repair mechanisms, have been well documented. However, the effects of fasting on cancer therapy have been the focus of recent scholarly investigations. Doxorubicin (Dox) is one of the most widely used chemotherapy medications for cancer treatment. Unfortunately, cardiotoxicity, which may lead to heart failure and other cardiovascular issues, has been linked to Dox usage. This study aims to comprehensively examine the possible advantages and disadvantages of fasting concerning Dox-induced cardiotoxicity. Researchers have investigated the potential benefits of fasting in lowering the risk of Dox-induced cardiac damage to solve this problem. Nevertheless, new studies indicate that prolonged alternate-day fasting may adversely affect the heart's capacity to manage the cardiotoxic properties of Dox. Though fasting may benefit overall health, it is essential to proceed cautiously and consider the potential risks in certain circumstances.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Jing Sun
- Department of Biobank, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Tao Yu
- Department of Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Province Cancer Hospital, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Haozhe Piao
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology, Liaoning Province Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| |
Collapse
|
37
|
Matacchione G, Piacenza F, Pimpini L, Rosati Y, Marcozzi S. The role of the gut microbiota in the onset and progression of heart failure: insights into epigenetic mechanisms and aging. Clin Epigenetics 2024; 16:175. [PMID: 39614396 PMCID: PMC11607950 DOI: 10.1186/s13148-024-01786-9] [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: 10/25/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND The gut microbiota (GM) plays a critical role in regulating human physiology, with dysbiosis linked to various diseases, including heart failure (HF). HF is a complex syndrome with a significant global health impact, as its incidence doubles with each decade of life, and its prevalence peaks in individuals over 80 years. A bidirectional interaction exists between GM and HF, where alterations in gut health can worsen the disease's progression. MAIN BODY The "gut hypothesis of HF" suggests that HF-induced changes, such as reduced intestinal perfusion and altered gut motility, negatively impact GM composition, leading to increased intestinal permeability, the release of GM-derived metabolites into the bloodstream, and systemic inflammation. This process creates a vicious cycle that further deteriorates heart function. GM-derived metabolites, including trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and secondary bile acids (BAs), can influence gene expression through epigenetic mechanisms, such as DNA methylation and histone modifications. These epigenetic changes may play a crucial role in mediating the effects of dysbiotic gut microbial metabolites, linking them to altered cardiac health and contributing to the progression of HF. This process is particularly relevant in older individuals, as the aging process itself has been associated with both dysbiosis and cumulative epigenetic alterations, intensifying the interplay between GM, epigenetic changes, and HF, and further increasing the risk of HF in the elderly. CONCLUSION Despite the growing body of evidence, the complex interplay between GM, epigenetic modifications, and HF remains poorly understood. The dynamic nature of epigenetics and GM, shaped by various factors such as age, diet, and lifestyle, presents significant challenges in elucidating the precise mechanisms underlying this complex relationship. Future research should prioritize innovative approaches to overcome these limitations. By identifying specific metabolite-induced epigenetic modifications and modulating the composition and function of GM, novel and personalized therapeutic strategies for the prevention and/or treatment of HF can be developed. Moreover, targeted research focusing specifically on older individuals is crucial for understanding the intricate connections between GM, epigenetics, and HF during aging.
Collapse
Affiliation(s)
- Giulia Matacchione
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60127, Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121, Ancona, Italy
| | | | - Yuri Rosati
- Pneumologia, IRCCS INRCA, 60027, Osimo, Italy
| | - Serena Marcozzi
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121, Ancona, Italy.
| |
Collapse
|
38
|
Młynarska E, Wasiak J, Gajewska A, Bilińska A, Steć G, Jasińska J, Rysz J, Franczyk B. Gut Microbiota and Gut-Brain Axis in Hypertension: Implications for Kidney and Cardiovascular Health-A Narrative Review. Nutrients 2024; 16:4079. [PMID: 39683474 DOI: 10.3390/nu16234079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
INTRODUCTION Arterial hypertension is a major contributor to a wide range of health complications, with cardiac hypertrophy and chronic kidney disease being among the most prevalent. Consequently, novel strategies for the treatment and prevention of hypertension are actively being explored. Recent research has highlighted a potential link between hypertension and the gut-brain axis. A bidirectional communication between the microbiota and the brain via the vagus nerve, enteric nervous system, hypothalamus-pituitary-adrenal axis, secreted short-chain fatty acids, and neurotransmitter metabolism. MATERIALS AND METHODS A comprehensive literature search was conducted using databases such as PubMed to identify studies exploring the relationship between gut microbiota and hypertension, along with the effects of dietary interventions and probiotics on blood pressure regulation. DISCUSSION Studies in both animal models and human subjects have demonstrated a strong correlation between alterations in gut microbiota composition and the development of hypertension. By influencing blood pressure, the gut microbiota can potentially affect the progression of cardiovascular and kidney disorders. Modulating gut microbiota through dietary interventions and probiotics has shown promise in regulating blood pressure and reducing systemic inflammation, offering a novel approach to managing hypertension. Diets such as the Mediterranean diet, which is rich in polyphenols and omega-3 fatty acids and low in sodium, promote the growth of beneficial gut bacteria that support cardiovascular health. Additionally, probiotics have been found to enhance gut barrier function, reduce inflammation, and modulate the Renin-Angiotensin System, all of which contribute to lowering blood pressure. CONCLUSIONS Further research is needed to determine the mechanisms of action of the microbiota in hypertension. The aim of this study was to evaluate the influence of gut microbiota on blood pressure regulation and the progression of hypertension-related complications, such as cardiovascular and kidney disorders.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Aleksandra Bilińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Greta Steć
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Jasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
39
|
Alexandrescu L, Suceveanu AP, Stanigut AM, Tofolean DE, Axelerad AD, Iordache IE, Herlo A, Nelson Twakor A, Nicoara AD, Tocia C, Dumitru A, Dumitru E, Condur LM, Aftenie CF, Tofolean IT. Intestinal Insights: The Gut Microbiome's Role in Atherosclerotic Disease: A Narrative Review. Microorganisms 2024; 12:2341. [PMID: 39597729 PMCID: PMC11596410 DOI: 10.3390/microorganisms12112341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Recent advances have highlighted the gut microbiota as a significant contributor to the development and progression of atherosclerosis, which is an inflammatory cardiovascular disease (CVD) characterized by plaque buildup within arterial walls. The gut microbiota, consisting of a diverse collection of microorganisms, impacts the host's metabolism, immune responses, and lipid processing, all of which contribute to atherosclerosis. This review explores the complex mechanisms through which gut dysbiosis promotes atherogenesis. We emphasize the potential of integrating microbiota modulation with traditional cardiovascular care, offering a holistic approach to managing atherosclerosis. Important pathways involve the translocation of inflammatory microbial components, modulation of lipid metabolism through metabolites such as trimethylamine-N-oxide (TMAO), and the production of short-chain fatty acids (SCFAs) that influence vascular health. Studies reveal distinct microbial profiles in atherosclerosis patients, with increased pathogenic bacteria (Megamonas, Veillonella, Streptococcus) and reduced anti-inflammatory genera (Bifidobacterium, Roseburia), highlighting the potential of these profiles as biomarkers and therapeutic targets. Probiotics are live microorganisms that have health benefits on the host. Prebiotics are non-digestible dietary fibers that stimulate the growth and activity of beneficial gut bacteria. Interventions targeting microbiota, such as probiotics, prebiotics, dietary modifications, and faecal microbiota transplantation (FMT), present effective approaches for restoring microbial equilibrium and justifying cardiovascular risk. Future research should focus on longitudinal, multi-omics studies to clarify causal links and refine therapeutic applications.
Collapse
Affiliation(s)
- Luana Alexandrescu
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Adrian Paul Suceveanu
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Alina Mihaela Stanigut
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
- Nephrology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
| | - Doina Ecaterina Tofolean
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
- Pneumology Department, Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
| | - Ani Docu Axelerad
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Ionut Eduard Iordache
- Department of General Surgery, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania;
| | - Alexandra Herlo
- Department XIII, Discipline of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Andreea Nelson Twakor
- Internal Medicine Department, Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania;
| | - Alina Doina Nicoara
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
- Internal Medicine Department, Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania;
| | - Cristina Tocia
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Andrei Dumitru
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
| | - Eugen Dumitru
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
- Academy of Romanian Scientist, 3 Ilfov Street, 050044 Bucharest, Romania
| | - Laura Maria Condur
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Cristian Florentin Aftenie
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Ioan Tiberiu Tofolean
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| |
Collapse
|
40
|
Guo X, Chen Z, Gao C, Zhang L, Liu Y, Lin M, Zhu P, Yang J, Wang Z, Zhang J, Sun H. 20S-O-Glc-DM treats metabolic syndrome-induced heart failure through regulating gut flora. Eur J Pharmacol 2024; 982:176946. [PMID: 39182541 DOI: 10.1016/j.ejphar.2024.176946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/19/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
Heart failure is a multifactorial disease, the percentage of patients with heart failure caused by metabolic syndrome is increasing year by year. The effect of gut flora dysbiosis on metabolic syndrome and heart failure has received widespread attention in recent years. Drugs to treat the condition urgently need to be discovered. C20DM, as a precursor compound of ginsenoside, is a small molecule compound obtained by biosynthetic means and is not available in natural products. In this project, we found that C20DM could improve the diversity of gut flora and elevate the expression of intestinal tight junction proteins-Occludin, Claudin, ZO-1, which inhibited the activity of the TLR4-MyD88-NF-kB pathway, and as a result, reduced myocardial inflammation and slowed down heart failure in metabolic syndrome mice. In conclusion, our study suggests that C20DM can treat heart failure by regulating gut flora, and it may be a candidate drug for treating metabolic syndrome-induced heart failure.
Collapse
Affiliation(s)
- Xinyi Guo
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhiwei Chen
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Chen Gao
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lingzhi Zhang
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yanxin Liu
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Modi Lin
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ping Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & NHC Key Laboratory of Biosynthesis of Natural Products, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jinling Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & NHC Key Laboratory of Biosynthesis of Natural Products, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Zhe Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & NHC Key Laboratory of Biosynthesis of Natural Products, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jinlan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & NHC Key Laboratory of Biosynthesis of Natural Products, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Hua Sun
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
41
|
Jiang X, Wang X, Zhang M, Yu L, He J, Wu S, Yan J, Zheng Y, Zhou Y, Chen Y. Associations between specific dietary patterns, gut microbiome composition, and incident subthreshold depression in Chinese young adults. J Adv Res 2024; 65:183-195. [PMID: 38879123 PMCID: PMC11518947 DOI: 10.1016/j.jare.2024.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/25/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION The interplay between influential factors and the incidence of subthreshold depression (SD) in young adults remains poorly understood. OBJECTIVES This study sought to understand the dietary habits, gut microbiota composition, etc. among individuals with SD in young adults and to investigate their association with SD occurrence. METHODS Employing a cross-sectional approach, 178 individuals with SD, aged 18-32 years, were matched with 114 healthy counterparts. SD status was evaluated using the Zung Self-rating Depression Scale (SDS), Zung Self-rating Anxiety Scale (SAS), Beck Depression Inventory 2nd version (BDI-II), the 17-item Hamilton Rating Scales of Depression (HAMD-17), and Pittsburgh Sleep Quality Index (PSQI). Metagenomic sequencing was utilized to identify fecal microbial profiles. Dietary patterns were discerned via factor analysis of a 25-item food frequency questionnaire (FFQ). Logistic regression analysis and mediation analysis were performed to explore the potential links between gut microbiota, dietary patterns, and incident SD. RESULTS Data on dietary habits were available for 292 participants (mean [SD] age, 22.1 [2.9] years; 216 [73.9 %] female). Logistic regression analysis revealed that dietary patterns Ⅰ (odds ratio [OR], 0.34; 95 % CI, 0.15-0.75) and IV (OR, 0.39; 95 % CI, 0.17-0.86 and OR, 0.39; 95 % CI, 0.18-0.84) were associated with reduced risk of SD. Distinct microbial profiles were observed in young adults with SD, marked by increased microbial diversity and taxonomic alterations. Moreover, mediation analysis suggested Veillonella atypica as a potential mediator linking SDS or BDI-II scores with a healthy dietary pattern rich in bean products, coarse grains, nuts, fruits, mushrooms, and potatoes (β = 0.25, 95 % CI: 0.02-0.78 and β = 0.18, 95 % CI: 0.01-0.54). CONCLUSIONS Our findings highlight the complex interplay between dietary patterns, gut microbiota, and the risk of developing SD in young adults, underscoring the potential for dietary interventions and microbiome modulation in mental health promotion.
Collapse
Affiliation(s)
- Xiumin Jiang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; South China Research Center for Acupuncture and Moxibustion, Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaotong Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China; South China Research Center for Acupuncture and Moxibustion, Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng Zhang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Yu
- Department of Traditional Chinese Medicine, the Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Sleep Research Institute of Integrative Medicine, the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun He
- Rehabilitation Center, Counseling Department, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shengwei Wu
- Department of Traditional Chinese Medicine, the Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinglan Yan
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanjia Zheng
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanyuan Zhou
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongjun Chen
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; South China Research Center for Acupuncture and Moxibustion, Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China.
| |
Collapse
|
42
|
Brusnic O, Onisor D, Boicean A, Hasegan A, Ichim C, Guzun A, Chicea R, Todor SB, Vintila BI, Anderco P, Porr C, Dura H, Fleaca SR, Cristian AN. Fecal Microbiota Transplantation: Insights into Colon Carcinogenesis and Immune Regulation. J Clin Med 2024; 13:6578. [PMID: 39518717 PMCID: PMC11547077 DOI: 10.3390/jcm13216578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Colorectal cancer (CRC) constitutes a significant global health challenge, with recent studies underscoring the pivotal role of the gut microbiome in its pathogenesis and progression. Fecal microbiota transplantation (FMT) has emerged as a compelling therapeutic approach, offering the potential to modulate microbial composition and optimize treatment outcomes. Research suggests that specific bacterial strains are closely linked to CRC, influencing both its clinical management and therapeutic interventions. Moreover, the gut microbiome's impact on immunotherapy responsiveness heralds new avenues for personalized medicine. Despite the promise of FMT, safety concerns, particularly in immunocompromised individuals, remain a critical issue. Clinical outcomes vary widely, influenced by genetic predispositions and the specific transplantation methodologies employed. Additionally, rigorous donor selection and screening protocols are paramount to minimize risks and maximize therapeutic efficacy. The current body of literature advocates for the establishment of standardized protocols and further clinical trials to substantiate FMT's role in CRC management. As our understanding of the microbiome deepens, FMT is poised to become a cornerstone in CRC treatment, underscoring the imperative for continued research and clinical validation.
Collapse
Affiliation(s)
- Olga Brusnic
- Department of Gastroenterology, University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mures, Romania; (O.B.); (D.O.)
| | - Danusia Onisor
- Department of Gastroenterology, University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mures, Romania; (O.B.); (D.O.)
| | - Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Adrian Hasegan
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Andreea Guzun
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Radu Chicea
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Samuel Bogdan Todor
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Bogdan Ioan Vintila
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Paula Anderco
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Corina Porr
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Horatiu Dura
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Sorin Radu Fleaca
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Adrian Nicolae Cristian
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| |
Collapse
|
43
|
Młynarska E, Jakubowska P, Frąk W, Gajewska A, Sornowska J, Skwira S, Wasiak J, Rysz J, Franczyk B. Associations of Microbiota and Nutrition with Cognitive Impairment in Diseases. Nutrients 2024; 16:3570. [PMID: 39458564 PMCID: PMC11510709 DOI: 10.3390/nu16203570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Recent research highlights the growing interest in the impact of nutrition on cognitive health and function in disease, as dietary habits are increasingly recognized as crucial factors in relation to brain function. This focus is especially important given the rising prevalence of neurodegenerative diseases and the cognitive decline associated with poor dietary choices. Links are now being sought between brain function and the microbiota and gut-brain axis. Mechanisms are proposed that include low-grade chronic neuroinflammation, the influence of short-chain fatty acids, or the disruption of glial cells and transmitters in the brain. METHODS We reviewed the articles on pubmed. This is not a systematic review, but of the narrative type. We wanted to outline the issue and summarise the latest information. RESULTS The axis in question has its foundation in nutrition. It has been reported that diet, particularly the components and the timing of food intake, has an impact on cognitive processes. The Mediterranean diet is most often cited in the literature as being beneficial to health. In order to obtain a more complete view, it is worth considering other dietary patterns, even those that impair our health. CONCLUSIONS Determining what is beneficial and what is not will allow us to develop a speronized strategy for the prevention of, and fight against, cognitive impairment. Appropriately selected supplements, the functions of which we have also discussed, may prove supportive.
Collapse
Affiliation(s)
- Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Paulina Jakubowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Weronika Frąk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Agata Gajewska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Sornowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Sylwia Skwira
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Wasiak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
44
|
Russo MA, Puccetti M, Costantini C, Giovagnoli S, Ricci M, Garaci E, Romani L. Human and gut microbiota synergy in a metabolically active superorganism: a cardiovascular perspective. Front Cardiovasc Med 2024; 11:1411306. [PMID: 39465131 PMCID: PMC11502352 DOI: 10.3389/fcvm.2024.1411306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
Despite significant advances in diagnosis and treatment over recent decades, cardiovascular disease (CVD) remains one of the leading causes of morbidity and mortality in Western countries. This persistent burden is partly due to the incomplete understanding of fundamental pathogenic mechanisms, which limits the effectiveness of current therapeutic interventions. In this context, recent evidence highlights the pivotal role of immuno-inflammatory activation by the gut microbiome in influencing cardiovascular disorders, potentially opening new therapeutic avenues. Indeed, while atherosclerosis has been established as a chronic inflammatory disease of the arterial wall, accumulating data suggest that immune system regulation and anti-inflammatory pathways mediated by gut microbiota metabolites play a crucial role in a range of CVDs, including heart failure, pericardial disease, arrhythmias, and cardiomyopathies. Of particular interest is the emerging understanding of how tryptophan metabolism-by both host and microbiota-converges on the Aryl hydrocarbon Receptor (AhR), a key regulator of immune homeostasis. This review seeks to enhance our understanding of the role of the immune system and inflammation in CVD, with a focus on how gut microbiome-derived tryptophan metabolites, such as indoles and their derivatives, contribute to cardioimmunopathology. By exploring these mechanisms, we aim to facilitate the development of novel, microbiome-centered strategies for combating CVD.
Collapse
Affiliation(s)
| | - Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Enrico Garaci
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| |
Collapse
|
45
|
Luo T, Che Q, Guo Z, Song T, Zhao J, Xu D. Modulatory effects of traditional Chinese medicines on gut microbiota and the microbiota-gut-x axis. Front Pharmacol 2024; 15:1442854. [PMID: 39444598 PMCID: PMC11497133 DOI: 10.3389/fphar.2024.1442854] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
The gut microbiota offers numerous benefits to the human body, including the promotion of nutrient absorption, participation in metabolic processes, and enhancement of immune function. Recent studies have introduced the concept of the gut-organ axis, which encompasses interactions such as the gut-brain axis, gut-liver axis, and gut-lung axis. This concept underscores the complex interplay between gut microbiota and various organs and tissues, including the brain, heart, lungs, liver, kidneys, muscles, and bones. Growing evidence indicates that gut microbiota can influence the onset and progression of multi-organ system diseases through their effects on the gut-organ axis. Traditional Chinese medicine has demonstrated significant efficacy in regulating the gastrointestinal system, leveraging its unique advantages. Considerable advancements have been made in understanding the role of gut microbiota and the gut-organ axis within the mechanisms of action of traditional Chinese medicine. This review aims to elucidate the roles of gut microbiota and the gut-organ axis in human health, explore the potential connections between traditional Chinese medicine and gut microbiota, and examine the therapeutic effects of traditional Chinese medicine on the microbiota-gut-organ axis. Furthermore, the review addresses the limitations and challenges present in current research while proposing potential directions for future investigations in this area.
Collapse
Affiliation(s)
- Tingting Luo
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
| | - Qingya Che
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
| | - Ziyi Guo
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Tingxia Song
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
| | - Juanjuan Zhao
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Delin Xu
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
46
|
Cuervo L, McAlpine PL, Olano C, Fernández J, Lombó F. Low-Molecular-Weight Compounds Produced by the Intestinal Microbiota and Cardiovascular Disease. Int J Mol Sci 2024; 25:10397. [PMID: 39408727 PMCID: PMC11477366 DOI: 10.3390/ijms251910397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is the main cause of mortality in industrialized countries, with over 500 million people affected worldwide. In this work, the roles of low-molecular-weight metabolites originating from the gut microbiome, such as short-chain fatty acids, hydrogen sulfide, trimethylamine, phenylacetic acid, secondary bile acids, indoles, different gases, neurotransmitters, vitamins, and complex lipids, are discussed in relation to their CVD-promoting or preventing activities. Molecules of mixed microbial and human hepatic origin, such as trimethylamine N-oxide and phenylacetylglutamine, are also presented. Finally, dietary agents with cardioprotective effects, such as probiotics, prebiotics, mono- and poly-unsaturated fatty acids, carotenoids, and polyphenols, are also discussed. A special emphasis is given to their gut microbiota-modulating properties.
Collapse
Affiliation(s)
- Lorena Cuervo
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Patrick L. McAlpine
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos Olano
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Javier Fernández
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Felipe Lombó
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
47
|
Józefczuk P, Biliński J, Minkowska A, Łaguna P. Gut microbiome in children undergoing hematopoietic stem cell transplantation. Best Pract Res Clin Gastroenterol 2024; 72:101955. [PMID: 39645282 DOI: 10.1016/j.bpg.2024.101955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
Hematopoietic stem cell transplantation (HSCT) is used in children as a treatment for various cancers, e.g. acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), or other diseases, e.g. severe congenital immunodeficiency, metabolic disorders, hence the patient population is quite diverse. There is an increasing interest on the role of the microbiome in peri-transplant period. In this review, concepts of HSCT with the focus on the importance of microbiome composition, its changes during treatment and possible microbiota oriented interventions will be discussed. This paper analyzes data in pediatric population, but in view of interesting results and absence of analogous data for pediatric patients, it also looks at studies performed on adult population and pre-clinical trials on animals discussing possible translation to children.
Collapse
Affiliation(s)
- Paweł Józefczuk
- Department of Oncology, Pediatric Hematology, Clinical Transplantology and Pediatrics, Medical University of Warsaw, Poland.
| | - Jarosław Biliński
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Poland; Human Biome Institute, Gdansk, Warsaw, Poland
| | - Aleksandra Minkowska
- Department of Oncology, Pediatric Hematology, Clinical Transplantology and Pediatrics, Medical University of Warsaw, Poland
| | - Paweł Łaguna
- Department of Oncology, Pediatric Hematology, Clinical Transplantology and Pediatrics, Medical University of Warsaw, Poland
| |
Collapse
|
48
|
Flori L, Benedetti G, Martelli A, Calderone V. Microbiota alterations associated with vascular diseases: postbiotics as a next-generation magic bullet for gut-vascular axis. Pharmacol Res 2024; 207:107334. [PMID: 39103131 DOI: 10.1016/j.phrs.2024.107334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
The intestinal microbiota represents a key element in maintaining the homeostasis and health conditions of the host. Vascular pathologies and other risk factors such as aging have been recently associated with dysbiosis. The qualitative and quantitative alteration of the intestinal microbiota hinders correct metabolic homeostasis, causing structural and functional changes of the intestinal wall itself. Impairment of the intestinal microbiota, combined with the reduction of the barrier function, worsen the pathological scenarios of peripheral tissues over time, including the vascular one. Several experimental evidence, collected in this review, describes in detail the changes of the intestinal microbiota in dysbiosis associated with vascular alterations, such as atherosclerosis, hypertension, and endothelial dysfunction, the resulting metabolic disorders and how these can impact on vascular health. In this context, the gut-vascular axis is considered, for the first time, as a merged unit involved in the development and progression of vascular pathologies and as a promising target. Current approaches for the management of dysbiosis such as probiotics, prebiotics and dietary modifications act mainly on the intestinal district. Postbiotics, described as preparation of inanimate microorganisms and/or their components that confers health benefits on the host, represent an innovative strategy for a dual management of intestinal dysbiosis and vascular pathologies. In this context, this review has the further purpose of defining the positive effects of the supplementation of bacterial strains metabolites (short‑chain fatty acids, exopolysaccharides, lipoteichoic acids, gallic acid, and protocatechuic acid) restoring intestinal homeostasis and acting directly on the vascular district through the gut-vascular axis.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy.
| | - Giada Benedetti
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy.
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa 56120, Italy; Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa 56120, Italy.
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa 56120, Italy; Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa 56120, Italy.
| |
Collapse
|
49
|
Cano R, Bermúdez V, Galban N, Garrido B, Santeliz R, Gotera MP, Duran P, Boscan A, Carbonell-Zabaleta AK, Durán-Agüero S, Rojas-Gómez D, González-Casanova J, Díaz-Vásquez W, Chacín M, Angarita Dávila L. Dietary Polyphenols and Gut Microbiota Cross-Talk: Molecular and Therapeutic Perspectives for Cardiometabolic Disease: A Narrative Review. Int J Mol Sci 2024; 25:9118. [PMID: 39201807 PMCID: PMC11354808 DOI: 10.3390/ijms25169118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
The intricate interplay between the gut microbiota and polyphenols has emerged as a captivating frontier in understanding and potentially harnessing the therapeutic potential of these bioactive compounds. Phenolic compounds, renowned for their antioxidant, anti-inflammatory, antidiabetic, and anticancer properties, are subject to intricate transformations within the gut milieu, where the diverse microbial ecosystem exerts profound effects on their metabolism and bioavailability. Conversely, polyphenols exhibit a remarkable capacity to modulate the composition and activity of the gut microbiota, fostering a bidirectional relationship that extends beyond mere nutrient processing. This symbiotic interaction holds significant implications for human health, particularly in cardiometabolic diseases such as diabetes mellitus, metabolic-dysfunction-associated steatotic liver disease, and cardiovascular disease. Through a comprehensive exploration of molecular interactions, this narrative review elucidates the reciprocal dynamics between the gut microbiota and polyphenols, unveiling novel avenues for therapeutic intervention in cardiometabolic disorders. By unravelling the intricate cross-talk between these two entities, this review underscores the multifaceted roles of polyphenols in overall health and the pivotal role of gut microbiota modulation as a promising therapeutic strategy in mitigating the burden of cardiometabolic diseases.
Collapse
Affiliation(s)
- Raquel Cano
- Centro de Investigaciones Endocrino-Metabólicas, Escuela de Medicina, Universidad del Zulia, Maracaibo 4001, Venezuela; (R.C.); (N.G.); (R.S.); (P.D.)
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080002, Colombia
| | - Nestor Galban
- Centro de Investigaciones Endocrino-Metabólicas, Escuela de Medicina, Universidad del Zulia, Maracaibo 4001, Venezuela; (R.C.); (N.G.); (R.S.); (P.D.)
| | - Bermary Garrido
- Centro de Investigaciones Endocrino-Metabólicas, Escuela de Medicina, Universidad del Zulia, Maracaibo 4001, Venezuela; (R.C.); (N.G.); (R.S.); (P.D.)
| | - Raquel Santeliz
- Centro de Investigaciones Endocrino-Metabólicas, Escuela de Medicina, Universidad del Zulia, Maracaibo 4001, Venezuela; (R.C.); (N.G.); (R.S.); (P.D.)
| | - Maria Paula Gotera
- Centro de Investigaciones Endocrino-Metabólicas, Escuela de Medicina, Universidad del Zulia, Maracaibo 4001, Venezuela; (R.C.); (N.G.); (R.S.); (P.D.)
| | - Pablo Duran
- Centro de Investigaciones Endocrino-Metabólicas, Escuela de Medicina, Universidad del Zulia, Maracaibo 4001, Venezuela; (R.C.); (N.G.); (R.S.); (P.D.)
| | - Arturo Boscan
- Escuela de Medicina, Facultad de Medicina, Universidad del Zulia, Maracaibo 4001, Venezuela;
| | | | - Samuel Durán-Agüero
- Escuela de Nutrición y Dietética, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago 7511111, Chile
| | - Diana Rojas-Gómez
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Santiago 8370321, Chile;
| | - Jorge González-Casanova
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile
| | - Waldo Díaz-Vásquez
- Escuela de Nutrición y Dietética, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago 7511111, Chile
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080002, Colombia
| | - Lissé Angarita Dávila
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Concepción 4260000, Chile
| |
Collapse
|
50
|
Li D, Li X, Zhang X, Chen J, Wang Z, Yu Z, Wu M, Liu L. Geniposide for treating atherosclerotic cardiovascular disease: a systematic review on its biological characteristics, pharmacology, pharmacokinetics, and toxicology. Chin Med 2024; 19:111. [PMID: 39164773 PMCID: PMC11334348 DOI: 10.1186/s13020-024-00981-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024] Open
Abstract
In recent years, the prevalence and fatality rates of atherosclerotic cardiovascular disease have not only shown a consistent rise that cannot be ignored, but have also become a pressing social health problem that requires urgent attention. While interventional surgery and drug therapy offer significant therapeutic results, they often come with common side effects. Geniposide, an active component extracted from the Chinese medicine Gardenia jasminoides Ellis, shows promise in the management of cardiac conditions. This review comprehensively outlines the underlying pharmacological mechanisms by which geniposide exerts its effects on atherosclerosis. Geniposide exhibits a range of beneficial effects including alleviating inflammation, inhibiting the development of macrophage foam cells, improving lipid metabolism, and preventing platelet aggregation and thrombosis. It also demonstrates mitochondrial preservation, anti-apoptotic effects, and modulation of autophagy. Moreover, geniposide shows potential in improving oxidative stress and endoplasmic reticulum stress by maintaining the body's antioxidant and oxidative balance. Additionally, this review comprehensively details the biological properties of geniposide, including methods of extraction and purification, as well as its pharmacokinetics and toxicological characteristics. It further discusses the clinical applications of related biopharmaceuticals, emphasizing the potential of geniposide in the prevention and treatment of atherosclerotic cardiovascular diseases. Furthermore, it highlights the limitations of current research, aiming to provide insights for future studies.
Collapse
Affiliation(s)
- Dexiu Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaoya Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaonan Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Jiye Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zeping Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zongliang Yu
- Beijing University of Chinese Medicine, Beijing, China
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China.
| |
Collapse
|