1
|
Jin Q, Wang S, Yao Y, Jiang Q, Li K. The gut-eye axis: from brain neurodegenerative diseases to age-related macular degeneration. Neural Regen Res 2025; 20:2741-2757. [PMID: 39435619 PMCID: PMC11826455 DOI: 10.4103/nrr.nrr-d-24-00531] [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: 05/09/2024] [Revised: 07/25/2024] [Accepted: 09/21/2024] [Indexed: 10/23/2024] Open
Abstract
Age-related macular degeneration is a serious neurodegenerative disease of the retina that significantly impacts vision. Unfortunately, the specific pathogenesis remains unclear, and effective early treatment options are consequently lacking. The microbiome is defined as a large ecosystem of microorganisms living within and coexisting with a host. The intestinal microbiome undergoes dynamic changes owing to age, diet, genetics, and other factors. Such dysregulation of the intestinal flora can disrupt the microecological balance, resulting in immunological and metabolic dysfunction in the host, and affecting the development of many diseases. In recent decades, significant evidence has indicated that the intestinal flora also influences systems outside of the digestive tract, including the brain. Indeed, several studies have demonstrated the critical role of the gut-brain axis in the development of brain neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Similarly, the role of the "gut-eye axis" has been confirmed to play a role in the pathogenesis of many ocular disorders. Moreover, age-related macular degeneration and many brain neurodegenerative diseases have been shown to share several risk factors and to exhibit comparable etiologies. As such, the intestinal flora may play an important role in age-related macular degeneration. Given the above context, the present review aims to clarify the gut-brain and gut-eye connections, assess the effect of intestinal flora and metabolites on age-related macular degeneration, and identify potential diagnostic markers and therapeutic strategies. Currently, direct research on the role of intestinal flora in age-related macular degeneration is still relatively limited, while studies focusing solely on intestinal flora are insufficient to fully elucidate its functional role in age-related macular degeneration. Organ-on-a-chip technology has shown promise in clarifying the gut-eye interactions, while integrating analysis of the intestinal flora with research on metabolites through metabolomics and other techniques is crucial for understanding their potential mechanisms.
Collapse
Affiliation(s)
- Qianzi Jin
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Suyu Wang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yujia Yao
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qin Jiang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Keran Li
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
2
|
Chen L, Jiang Y, Cheng X. Associations Between CYP3A5 (c.6986A>G) Gene Polymorphism and Kidney Impairment in Hypertensive Adults Without Cystatin C Elevation. Cardiovasc Toxicol 2024; 24:1047-1052. [PMID: 39095622 DOI: 10.1007/s12012-024-09902-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE This study aimed to explore the potential role of CYP3A5 (c. 6986A>G) gene polymorphism in predicting kidney function impairment in patients with hypertension who did not have elevated serum cystatin C. METHODS We recruited a group of patients with hypertension who did not have elevated cystatin C and analyzed the CYP3A5 (c. 6986A>G) gene polymorphism. Chi-square tests were used to compare the clinical characteristics and genotypic distribution between the two groups. Logistic regression analysis was used to explore the association between CYP3A5 (c.6986A>G) gene polymorphism and renal function impairment in hypertension with non-elevated cystatin. RESULTS In patients with hypertension who participated in the study, there was a significant association between CYP3A5 (c. 6986A>G) gene polymorphism and kidney function impairment (p < 0.05). Patients with the CYP3A5 (c. 6986A>G) mutation display a greater risk of kidney function impairment. CONCLUSION CYP3A5 (c. 6986A>G) gene AA homozygote polymorphism significantly increases risk of kidney function impairment in patients with hypertension with normal cystatin C. However, further studies are needed to validate this association and to further understand the mechanism of CYP3A5 (c. 6986A>G) gene polymorphism in kidney function impairment in patients with hypertension.
Collapse
Affiliation(s)
- Ling Chen
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou City, 215000, Jiangsu Province, People's Republic of China
| | - Yufeng Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou City, 215125, Jiangsu Province, People's Republic of China.
| | - Xingbo Cheng
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou City, 215000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
3
|
Guivala SJ, Bode KA, Okun JG, Kartal E, Schwedhelm E, Pohl LV, Werner S, Erbs S, Thiele H, Büttner P. Interactions between the gut microbiome, associated metabolites and the manifestation and progression of heart failure with preserved ejection fraction in ZSF1 rats. Cardiovasc Diabetol 2024; 23:299. [PMID: 39143579 PMCID: PMC11325580 DOI: 10.1186/s12933-024-02398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is associated with systemic inflammation, obesity, metabolic syndrome, and gut microbiome changes. Increased trimethylamine-N-oxide (TMAO) levels are predictive for mortality in HFpEF. The TMAO precursor trimethylamine (TMA) is synthesized by the intestinal microbiome, crosses the intestinal barrier and is metabolized to TMAO by hepatic flavin-containing monooxygenases (FMO). The intricate interactions of microbiome alterations and TMAO in relation to HFpEF manifestation and progression are analyzed here. METHODS Healthy lean (L-ZSF1, n = 12) and obese ZSF1 rats with HFpEF (O-ZSF1, n = 12) were studied. HFpEF was confirmed by transthoracic echocardiography, invasive hemodynamic measurements, and detection of N-terminal pro-brain natriuretic peptide (NT-proBNP). TMAO, carnitine, symmetric dimethylarginine (SDMA), and amino acids were measured using mass-spectrometry. The intestinal epithelial barrier was analyzed by immunohistochemistry, in-vitro impedance measurements and determination of plasma lipopolysaccharide via ELISA. Hepatic FMO3 quantity was determined by Western blot. The fecal microbiome at the age of 8, 13 and 20 weeks was assessed using 16s rRNA amplicon sequencing. RESULTS Increased levels of TMAO (+ 54%), carnitine (+ 46%) and the cardiac stress marker NT-proBNP (+ 25%) as well as a pronounced amino acid imbalance were observed in obese rats with HFpEF. SDMA levels in O-ZSF1 were comparable to L-ZSF1, indicating stable kidney function. Anatomy and zonula occludens protein density in the intestinal epithelium remained unchanged, but both impedance measurements and increased levels of LPS indicated an impaired epithelial barrier function. FMO3 was decreased (- 20%) in the enlarged, but histologically normal livers of O-ZSF1. Alpha diversity, as indicated by the Shannon diversity index, was comparable at 8 weeks of age, but decreased by 13 weeks of age, when HFpEF manifests in O-ZSF1. Bray-Curtis dissimilarity (Beta-Diversity) was shown to be effective in differentiating L-ZSF1 from O-ZSF1 at 20 weeks of age. Members of the microbial families Lactobacillaceae, Ruminococcaceae, Erysipelotrichaceae and Lachnospiraceae were significantly differentially abundant in O-ZSF1 and L-ZSF1 rats. CONCLUSIONS In the ZSF1 HFpEF rat model, increased dietary intake is associated with alterations in gut microbiome composition and bacterial metabolites, an impaired intestinal barrier, and changes in pro-inflammatory and health-predictive metabolic profiles. HFpEF as well as its most common comorbidities obesity and metabolic syndrome and the alterations described here evolve in parallel and are likely to be interrelated and mutually reinforcing. Dietary adaption may have a positive impact on all entities.
Collapse
Affiliation(s)
- Salmina J Guivala
- Department of Cardiology, Angiology and Pulmonology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| | - Konrad A Bode
- Department Molecular Diagnostics, Laboratory Dr. Limbach and Colleagues, Am Breitspiel 15, 69126, Heidelberg, Germany
| | - Jürgen G Okun
- Division of Neuropediatrics and Metabolic Medicine, Department of General Pediatrics, University Children's Hospital Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Ece Kartal
- Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Luca V Pohl
- Heart Center Leipzig, University of Leipzig, Strümpellstrasse 89, 04289, Leipzig, Germany
| | - Sarah Werner
- Heart Center Leipzig, University of Leipzig, Strümpellstrasse 89, 04289, Leipzig, Germany
| | - Sandra Erbs
- Heart Center Leipzig, University of Leipzig, Strümpellstrasse 89, 04289, Leipzig, Germany
| | - Holger Thiele
- Heart Center Leipzig, University of Leipzig, Strümpellstrasse 89, 04289, Leipzig, Germany
| | - Petra Büttner
- Heart Center Leipzig, University of Leipzig, Strümpellstrasse 89, 04289, Leipzig, Germany
| |
Collapse
|
4
|
Pang S, Han T, Huang X, Zhao Y, Qian J, Zhong J, Xie P, Liao L. Exploring the potential causal relationship between gut microbiota and heart failure: A two-sample mendelian randomization study combined with the geo database. Curr Probl Cardiol 2024; 49:102235. [PMID: 38040216 DOI: 10.1016/j.cpcardiol.2023.102235] [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: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
OBJECTIVE In recent years, researchers have observed a potential association between alterations in gut microbiota and the onset and progression of heart failure. Nevertheless, the causal relationship between gut microbiota and heart failure remains a topic of controversy. This study employed a two-sample Mendelian randomization approach to investigate the causal link between gut microbiota and heart failure. METHOD We extracted single nucleotide polymorphism (SNPs) data for heart failure (ebi-a-gcst009541) and gut microbiota from the publicly available genome-wide association analysis (GWAS) summary database. The primary analytical method employed was inverse variance weighting (IVW), complemented by validation using MR-PRESSO, weighted median, and MR pleiotropic residual methods. Additionally, gene pleiotropy (MR-Egger), heterogeneity testing, and a "leave-one-out" analysis were conducted to assess the robustness of the findings. Utilizing the limma package, differentially expressed genes (DEGs) from the Gut Microbiota datasets (GSE3586, GSE5406) and Heart Failure datasets (GSE47908, GSE87466) sourced from the Gene Expression Omnibus (GEO) were curated. Subsequent enrichment analysis was conducted using the Cluster Profiler and GO plot packages to validate the MR analysis outcomes. RESULTS The results of our analysis revealed seven distinct bacterial groups in the intestines that exhibited associations.with.the.risk.of.heart.failure. These.included.class.negativicutes (P = 0.02,OR:1.11,95%CI:1.02,1.21), gene.eubacterium.eligensgroup (P = 0.02,OR:1.10,95%CI:1.01,1.20),gene.eubacteriummoxidoreducensgroup (P = 0.01,OR:1.10,95%CI:1.02,1.19),Order.selenium (P = 0.02,OR:1.11,95%CI:1.02,1.21), gene.familyxiiiucg001 (P = 0.03,OR=1.09.95%CI:1.01,1.19), gene.familyxiiiad3011group (P = 0.03,OR:0.92,95%CI:0.86,0.99), and.gene.anaerostipes (P = 0.00,OR:0.87,95%CI:0.80,0.94). Nevertheless, upon conducting reverse causal MR analysis, no evidence of a causal relationship between heart failure and the aforementioned seven gut microbiota groups was found.Bioinformatics analysis reveals shared DEGs between gut microbiota and heart failure. CONCLUSION This Mendelian randomization study represents the first endeavor to explore the causal relationship between specific gut microbiota and heart failure. The findings suggest a significant correlation between these seven specific gut microbiota groups and the risk of heart failure, potentially offering valuable insights for heart failure prevention and control efforts.
Collapse
Affiliation(s)
- Shuwen Pang
- Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, No. 16 Xiantong Road, Luohu District, Shenzhen, 518001,Guangdong , China; General Hospital of the Southern Theater Command of the People's Liberation Army of China, 111 Liuhua Road, Yuexiu District, Guangzhou, 5100102, Guangdong, China; Graduate School of Guangzhou University of Traditional Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510100, Guangdong, China.
| | - Tao Han
- Wangjing Hospital, China Academy of Chinese Medical Sciences, 6 Wangjing Zhonghuan South Road, Chaoyang District, 100102, Beijing, China.
| | - Xiwei Huang
- General Hospital of the Southern Theater Command of the People's Liberation Army of China, 111 Liuhua Road, Yuexiu District, Guangzhou, 5100102, Guangdong, China; Graduate School of Guangzhou University of Traditional Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510100, Guangdong, China.
| | - Yueli Zhao
- General Hospital of the Southern Theater Command of the People's Liberation Army of China, 111 Liuhua Road, Yuexiu District, Guangzhou, 5100102, Guangdong, China.
| | - Jing Qian
- The Second Affiliated Hospital of Naval Medical University (Shanghai Long March Hospital), 415 Fengyang Road, Huangpu District, 200003, Shanghai, China.
| | - Jiahui Zhong
- Guangdong Provincial People's Hospital, No. 106 Zhongshan Second Road, Yuexiu District, Guangzhou, 510080,Guangdong, China.
| | - Pingjin Xie
- Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, No. 16 Xiantong Road, Luohu District, Shenzhen, 518001,Guangdong , China.
| | - Lu Liao
- Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, No. 16 Xiantong Road, Luohu District, Shenzhen, 518001,Guangdong , China.
| |
Collapse
|
5
|
Paraskevaidis I, Briasoulis A, Tsougos E. Oral Cardiac Drug-Gut Microbiota Interaction in Chronic Heart Failure Patients: An Emerging Association. Int J Mol Sci 2024; 25:1716. [PMID: 38338995 PMCID: PMC10855150 DOI: 10.3390/ijms25031716] [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: 12/05/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Regardless of the currently proposed best medical treatment for heart failure patients, the morbidity and mortality rates remain high. This is due to several reasons, including the interaction between oral cardiac drug administration and gut microbiota. The relation between drugs (especially antibiotics) and gut microbiota is well established, but it is also known that more than 24% of non-antibiotic drugs affect gut microbiota, altering the microbe's environment and its metabolic products. Heart failure treatment lies mainly in the blockage of neuro-humoral hyper-activation. There is debate as to whether the administration of heart-failure-specific drugs can totally block this hyper-activation, or whether the so-called intestinal dysbiosis that is commonly observed in this group of patients can affect their action. Although there are several reports indicating a strong relation between drug-gut microbiota interplay, little is known about this relation to oral cardiac drugs in chronic heart failure. In this review, we review the contemporary data on a topic that is in its infancy. We aim to produce scientific thoughts and questions and provide reasoning for further clinical investigation.
Collapse
Affiliation(s)
- Ioannis Paraskevaidis
- Division of Cardiology, Hygeia Hospital, Erithrou Stavrou 4, 15123 Athens, Greece;
- Heart Failure Subdivision, Department of Clinical Therapeutics, Alexandra Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Vassilisis Sofias 80, 11528 Athens, Greece;
| | - Alexandros Briasoulis
- Heart Failure Subdivision, Department of Clinical Therapeutics, Alexandra Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Vassilisis Sofias 80, 11528 Athens, Greece;
| | - Elias Tsougos
- Division of Cardiology, Hygeia Hospital, Erithrou Stavrou 4, 15123 Athens, Greece;
| |
Collapse
|
6
|
Paraskevaidis I, Xanthopoulos A, Tsougos E, Triposkiadis F. Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ. Biomedicines 2023; 11:2574. [PMID: 37761015 PMCID: PMC10526035 DOI: 10.3390/biomedicines11092574] [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: 08/01/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
There is a bidirectional relationship between the heart and the gut. The gut microbiota, the community of gut micro-organisms themselves, is an excellent gut-homeostasis keeper since it controls the growth of potentially harmful bacteria and protects the microbiota environment. There is evidence suggesting that a diet rich in fatty acids can be metabolized and converted by gut microbiota and hepatic enzymes to trimethyl-amine N-oxide (TMAO), a product that is associated with atherogenesis, platelet dysfunction, thrombotic events, coronary artery disease, stroke, heart failure (HF), and, ultimately, death. HF, by inducing gut ischemia, congestion, and, consequently, gut barrier dysfunction, promotes the intestinal leaking of micro-organisms and their products, facilitating their entrance into circulation and thus stimulating a low-grade inflammation associated with an immune response. Drugs used for HF may alter the gut microbiota, and, conversely, gut microbiota may modify the pharmacokinetic properties of the drugs. The modification of lifestyle based mainly on exercise and a Mediterranean diet, along with the use of pre- or probiotics, may be beneficial for the gut microbiota environment. The potential role of gut microbiota in HF development and progression is the subject of this review.
Collapse
Affiliation(s)
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.X.); (F.T.)
| | - Elias Tsougos
- 6th Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece
| | - Filippos Triposkiadis
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.X.); (F.T.)
| |
Collapse
|
7
|
Tan Y, Xu Y, Zhang Z, Ran Z, Liu X, Jia Y, Chen Y. The Prognostic Value and Treatment Strategies of Nutritional Status in Heart Failure Patients. Curr Probl Cardiol 2023; 48:101742. [PMID: 37087080 DOI: 10.1016/j.cpcardiol.2023.101742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 04/24/2023]
Abstract
Heart failure is a complex clinical syndrome caused by a variety of reasons leading to abnormal changes in the structure and/or function of the heart, with ventricular systolic and/or diastolic dysfunction, which is a serious manifestation or late stage of various heart diseases. The overall prognosis of patients is poor, and risk assessment of patients with HF is currently a hot topic of research due to the large heterogeneity of etiology, phenotype, and genetic background of HF patients. Besides, the nutritional level and status of HF patients are affected by various aspects. Patients with malnutrition, high saturated fatty acids and cholesterol, low minerals, and other conditions tend to have a poor prognosis. So targeted improvement of the nutritional status of HF patients is important to improve the prognosis and the quality of survival of patients. We use heart failure, nutrition, and diet therapy as the keyword method to summarize the prognostic value of indicators of nutritional status in HF patients, the effects of nutritional status on HF patients with different etiology, and potential treatment strategies for HF patients with different etiology. This review is valuable for understanding the prognostic value of nutritional levels in patients with HF and guiding clinical therapeutic approaches.
Collapse
Affiliation(s)
- Yinxi Tan
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuanwei Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zixuan Zhang
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zechao Ran
- West China School of Clinical Medicine and West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xinyue Liu
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunqi Jia
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yucheng Chen
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
8
|
Mahajan A, Bandaru D, Parikh K, Gupta V, Patel M. From the inside out: understanding the gut-heart connection. Future Cardiol 2023; 19:505-514. [PMID: 37721335 DOI: 10.2217/fca-2023-0068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023] Open
Abstract
The gut microbiome was first termed as 'Animalcules' by Antonie van Leeuwenhoek in the 17th century. The diverse composition and complex interactions of gut microbes are essential for good human health. They play a crucial role in inflammation, which by itself leads to the development of cardiovascular diseases. Although the mechanisms are not fully understood, it has been studied that the gut microbiota produce several bioactive metabolites impacting cardiovascular health mainly through TMAO pathway, SCFA pathway and bile acid pathway. Moreover, studies have found that using dietary interventions like high fiber diet and probiotics to re-establish a healthy equilibrium show promising results on improving cardiovascular health and thus, could be potentially used for prevention and management of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Kinna Parikh
- G.M.E.R.S Medical College, Gandhinagar, 382007, India
| | - Vasu Gupta
- Satyam Hospital & Trauma Center, Jalandhar, 144008, India
| | - Meet Patel
- Department of Medicine, Shraddha Hospital, Gujarat, 380043, India
| |
Collapse
|
9
|
Lupu VV, Adam Raileanu A, Mihai CM, Morariu ID, Lupu A, Starcea IM, Frasinariu OE, Mocanu A, Dragan F, Fotea S. The Implication of the Gut Microbiome in Heart Failure. Cells 2023; 12:1158. [PMID: 37190067 PMCID: PMC10136760 DOI: 10.3390/cells12081158] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Heart failure is a worldwide health problem with important consequences for the overall wellbeing of affected individuals as well as for the healthcare system. Over recent decades, numerous pieces of evidence have demonstrated that the associated gut microbiota represent an important component of human physiology and metabolic homeostasis, and can affect one's state of health or disease directly, or through their derived metabolites. The recent advances in human microbiome studies shed light on the relationship between the gut microbiota and the cardiovascular system, revealing its contribution to the development of heart failure-associated dysbiosis. HF has been linked to gut dysbiosis, low bacterial diversity, intestinal overgrowth of potentially pathogenic bacteria and a decrease in short chain fatty acids-producing bacteria. An increased intestinal permeability allowing microbial translocation and the passage of bacterial-derived metabolites into the bloodstream is associated with HF progression. A more insightful understanding of the interactions between the human gut microbiome, HF and the associated risk factors is mandatory for optimizing therapeutic strategies based on microbiota modulation and offering individualized treatment. The purpose of this review is to summarize the available data regarding the influence of gut bacterial communities and their derived metabolites on HF, in order to obtain a better understanding of this multi-layered complex relationship.
Collapse
Affiliation(s)
- Vasile Valeriu Lupu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (I.M.S.)
| | - Anca Adam Raileanu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (I.M.S.)
| | | | - Ionela Daniela Morariu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ancuta Lupu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (I.M.S.)
| | - Iuliana Magdalena Starcea
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (I.M.S.)
| | - Otilia Elena Frasinariu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (I.M.S.)
| | - Adriana Mocanu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (I.M.S.)
| | - Felicia Dragan
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Silvia Fotea
- Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| |
Collapse
|