1
|
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
|
2
|
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
|
3
|
Yao F, Liu C, Luo D, Zhou Y, Li Q, Huang H, Xu H. Metabolites of Microbiota: A Novel Therapy for Heart Disease. FOOD REVIEWS INTERNATIONAL 2024:1-17. [DOI: 10.1080/87559129.2024.2437410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Fei Yao
- School of Mental Health, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Chao Liu
- Department of Electrocardiogram, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Duo Luo
- Department of Geriatrics, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Youlian Zhou
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qianqing Li
- Department of Electrocardiogram, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hongli Huang
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
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
|
5
|
Yafarova AA, Dementeva EV, Zlobovskaya OA, Sheptulina AF, Lopatukhina EV, Timofeev YS, Glazunova EV, Lyundup AV, Doludin YV, Kiselev AR, Shipulin GA, Makarov VV, Drapkina OM, Yudin SM. Gut Microbiota and Metabolic Alterations Associated with Heart Failure and Coronary Artery Disease. Int J Mol Sci 2024; 25:11295. [PMID: 39457077 PMCID: PMC11508380 DOI: 10.3390/ijms252011295] [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: 09/03/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the role of gut microbiota in cardiovascular diseases, with an additional focus on pro-atherogenic metabolites. We use advanced network analysis and machine learning techniques to identify key microbial features linked to coronary artery disease (CAD) and heart failure with reduced ejection fraction (HFrEF). This cross-sectional study included 189 participants divided into three groups: coronary artery disease (n = 93), heart failure with reduced ejection fraction (n = 43), and controls (n = 53). Assessments included physical exams, echocardiography, dietary surveys, blood analysis, and fecal analysis. Gut microbiota composition was analyzed using next-generation sequencing (NGS) and quantitative polymerase chain reaction (qPCR). Statistical analysis methods for testing hypotheses and correlations, alpha and beta-diversity analyses, co-occurrence networks, and machine learning were conducted using Python libraries or R packages with multiple comparisons corrected using the Benjamini-Hochberg procedure. Significant gut microbiota alterations were observed, with higher Bacillota/Bacteroidota ratios in CAD and HFrEF groups compared to controls (p < 0.001). Significant differences were observed in α-diversity indices (Pielou, Chao1, Faith) between disease groups and controls (p < 0.001). β-diversity analyses also revealed distinct microbial profiles (p = 0.0015). Interestingly, trimethylamine N-oxide (TMAO) levels were lower in CAD and HFrEF groups compared to controls (p < 0.05), while indoxyl sulfate (IS) levels were comparable between the study groups. Co-occurrence network analysis and machine learning identified key microbial features linked to these conditions, highlighting complex interactions within the gut microbiota associated with cardiovascular disease.
Collapse
Affiliation(s)
- Adel A. Yafarova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Dementeva
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Olga A. Zlobovskaya
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Anna F. Sheptulina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Lopatukhina
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Yuriy S. Timofeev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Evgeniya V. Glazunova
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Aleksey V. Lyundup
- Endocrinology Research Centre, Dmitry Ulyanov St. 19, 117036 Moscow, Russia
| | - Yuriy V. Doludin
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Anton R. Kiselev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - German A. Shipulin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Valentin V. Makarov
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Sergey M. Yudin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| |
Collapse
|
6
|
Fang Z, Zang Q, Chen J, Li Z, Yang D, Wu C, Yang H, Guo N. Whole-body mass spectrometry imaging reveals the systemic metabolic disorder and catecholamines biosynthesis alteration on heart-gut axis in heart failure rat. J Adv Res 2024:S2090-1232(24)00385-0. [PMID: 39270978 DOI: 10.1016/j.jare.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/28/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
INTRODUCTION Heart failure (HF) is a systemic metabolic disorder disease, across multiorgan investigations advancing knowledge of progression and treatment of HF. Whole-body MSI provides spatiotemporal information of metabolites in multiorgan and is expected to be a potent tool to dig out the complex mechanism of HF. OBJECTIVES This study aimed at exploring the systemic metabolic disorder in multiorgan and catecholamines biosynthesis alteration on heart-gut axis after HF. METHODS Whole-body MSI was used to characterize metabolic disorder of the whole rat body after HF. An integrated method by MSI, LC-MS/MS and ELISA was utilized to analyze key metabolites and enzymes on heart, small intestine, cecum and colon tissues of rat. Gut microbiota dysbiosis was investigated by 16S rDNA sequencing and metagenomic sequencing. Validation experiments and in vitro experiments were performed to verify the effect of catecholamines biosynthesis alteration on heart-gut axis after HF. RESULTS Whole-body MSI exhibited varieties of metabolites alteration in multiple organs. Remarkably, catecholamine biosynthesis was significantly altered in the serum, heart and intestines of rats. Furthermore, catecholamines and tyrosine hydroxylase were obviously upregulated in heart and colon tissue. Turicibacter_sanguinis was relevant to catecholamines of heart and colon. Validation experiments demonstrated excessive norepinephrine induced cardio-intestinal injury, including significantly elevating the levels of BNP, pro-BNP, LPS, DAO, and increased the abundance of Turicibacter_sanguinis. These alterations could be reversed by metoprolol treatment blocking the effect of norepinephrine. Additionally, in vitro studies demonstrated that norepinephrine promoted the growth of Turicibacter_sanguinis and Turicibacter_sanguinis could import and metabolize norepinephrine. Collectively, excessive norepinephrine exerted bidirectional effects on cardio-intestinal function to participate in the progression of HF. CONCLUSION Our study provides a new approach to elucidate multiorgan metabolic disorder and proposes new insights into heart-gut axis in HF development.
Collapse
Affiliation(s)
- Zhengyu Fang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingce Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiemei Chen
- Department of Pharmacy, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zeyu Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Dawei Yang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Na Guo
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
7
|
Zhou XD, Xu CF, Chen QF, Shapiro MD, Lip GYH, Chen LL, Targher G, Byrne CD, Tian N, Xiao T, Huang CX, Ni Y, Zheng MH. Serum bile acid profiles are associated with heart failure with preserved ejection fraction in patients with metabolic dysfunction-associated fatty liver disease: An exploratory study. Diabetes Obes Metab 2024; 26:3684-3695. [PMID: 38874096 DOI: 10.1111/dom.15709] [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: 04/28/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024]
Abstract
AIM To analyse the association between serum bile acid (BA) profile and heart failure (HF) with preserved ejection fraction (HFpEF) in patients with metabolic dysfunction-associated fatty liver disease (MAFLD). METHODS We enrolled 163 individuals with biopsy-proven MAFLD undergoing transthoracic echocardiography for any indication. HFpEF was defined as left ventricular ejection fraction >50% with at least one echocardiographic feature of HF (left ventricular diastolic dysfunction, abnormal left atrial size) and at least one HF sign or symptom. Serum levels of 38 BAs were analysed using ultra-performance liquid chromatography coupled with tandem mass spectrometry. RESULTS Among the 163 patients enrolled (mean age 47.0 ± 12.8 years, 39.3% female), 52 (31.9%) and 43 (26.4%) met the HFpEF and pre-HFpEF criteria, and 38 serum BAs were detected. Serum ursodeoxycholic acid (UDCA) and hyocholic acid (HCA) species were lower in patients with HFpEF and achieved statistical significance after correction for multiple comparisons. Furthermore, decreases in glycoursodeoxycholic acid and tauroursodeoxycholic acid were associated with HF status. CONCLUSIONS In this exploratory study, specific UDCA and HCA species were associated with HFpEF status in adults with biopsy-confirmed MAFLD.
Collapse
Affiliation(s)
- Xiao-Dong Zhou
- Department of Cardiovascular Medicine, The Heart Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cui-Fang Xu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qin-Fen Chen
- Medical Care Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, Wenzhou Medical University, Wenzhou, China
| | - Michael D Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Li-Li Chen
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Na Tian
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tie Xiao
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chen-Xiao Huang
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Ni
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| |
Collapse
|
8
|
Zheng H, Zhang X, Li C, Wang D, Shen Y, Lu J, Zhao L, Li X, Gao H. BCAA mediated microbiota-liver-heart crosstalk regulates diabetic cardiomyopathy via FGF21. MICROBIOME 2024; 12:157. [PMID: 39182099 PMCID: PMC11344321 DOI: 10.1186/s40168-024-01872-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/10/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is one of leading causes of diabetes-associated mortality. The gut microbiota-derived branched-chain amino acids (BCAA) have been reported to play a central role in the onset and progression of DCM, but the potential mechanisms remain elusive. RESULTS We found the type 1 diabetes (T1D) mice had higher circulating BCAA levels due to a reduced BCAA degradation ability of the gut microbiota. Excess BCAA decreased hepatic FGF21 production by inhibiting PPARα signaling pathway and thereby resulted in a higher expression level of cardiac LAT1 via transcription factor Zbtb7c. High cardiac LAT1 increased the levels of BCAA in the heart and then caused mitochondrial damage and myocardial apoptosis through mTOR signaling pathway, leading to cardiac fibrosis and dysfunction in T1D mice. Additionally, transplant of faecal microbiota from healthy mice alleviated cardiac dysfunction in T1D mice, but this effect was abolished by FGF21 knockdown. CONCLUSIONS Our study sheds light on BCAA-mediated crosstalk among the gut microbiota, liver and heart to promote DCM and FGF21 serves as a key mediator. Video Abstract.
Collapse
Affiliation(s)
- Hong Zheng
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xi Zhang
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chen Li
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Die Wang
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuying Shen
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jiahui Lu
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Liangcai Zhao
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaokun Li
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hongchang Gao
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
9
|
Li X, Lu C, Mao X, Fan J, Yao J, Jiang J, Wu L, Ren J, Shen J. Bibliometric analysis of research on gut microbiota and bile acids: publication trends and research frontiers. Front Microbiol 2024; 15:1433910. [PMID: 39234549 PMCID: PMC11371755 DOI: 10.3389/fmicb.2024.1433910] [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: 06/10/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
The gut microbiota is widely regarded as a "metabolic organ" that could generate myriad metabolites to regulate human metabolism. As the microbiota metabolites, bile acids (BAs) have recently been identified as the critical endocrine molecules that mediate the cross-talk between the host and intestinal microbiota. This study provided a comprehensive insight into the gut microbiota and BA research through bibliometric analysis from 2003 to 2022. The publications on this subject showed a dramatic upward trend. Although the USA and China have produced the most publications, the USA plays a dominant role in this expanding field. Specifically, the University of Copenhagen was the most productive institution. Key research hotspots are the gut-liver axis, short-chain fatty acids (SCFAs), cardiovascular disease (CVD), colorectal cancer (CRC), and the farnesoid x receptor (FXR). The molecular mechanisms and potential applications of the gut microbiota and BAs in cardiometabolic disorders and gastrointestinal cancers have significant potential for further research.
Collapse
Affiliation(s)
- Xin Li
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
- Department of General Practice, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Can Lu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xue Mao
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiahong Fan
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianting Yao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjie Jiang
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lele Wu
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingjing Ren
- Department of General Practice, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun Shen
- Department of General Medicine and Geriatrics, Linping Campus, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Bilson J, Scorletti E, Swann JR, Byrne CD. Bile Acids as Emerging Players at the Intersection of Steatotic Liver Disease and Cardiovascular Diseases. Biomolecules 2024; 14:841. [PMID: 39062555 PMCID: PMC11275019 DOI: 10.3390/biom14070841] [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: 06/30/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Affecting approximately 25% of the global population, steatotic liver disease (SLD) poses a significant health concern. SLD ranges from simple steatosis to metabolic dysfunction-associated steatohepatitis and fibrosis with a risk of severe liver complications such as cirrhosis and hepatocellular carcinoma. SLD is associated with obesity, atherogenic dyslipidaemia, and insulin resistance, increasing cardiovascular risks. As such, identifying SLD is vital for cardiovascular disease (CVD) prevention and treatment. Bile acids (BAs) have critical roles in lipid digestion and are signalling molecules regulating glucose and lipid metabolism and influencing gut microbiota balance. BAs have been identified as critical mediators in cardiovascular health, influencing vascular tone, cholesterol homeostasis, and inflammatory responses. The cardio-protective or harmful effects of BAs depend on their concentration and composition in circulation. The effects of certain BAs occur through the activation of a group of receptors, which reduce atherosclerosis and modulate cardiac functions. Thus, manipulating BA receptors could offer new avenues for treating not only liver diseases but also CVDs linked to metabolic dysfunctions. In conclusion, this review discusses the intricate interplay between BAs, metabolic pathways, and hepatic and extrahepatic diseases. We also highlight the necessity for further research to improve our understanding of how modifying BA characteristics affects or ameliorates disease.
Collapse
Affiliation(s)
- Josh Bilson
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (E.S.)
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO16 6YD, UK
| | - Eleonora Scorletti
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (E.S.)
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO16 6YD, UK
- Division of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan R. Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (E.S.)
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO16 6YD, UK
| | - Christopher D. Byrne
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (E.S.)
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
11
|
Trøseid M, Nielsen SD, Vujkovic-Cvijin I. Gut microbiome and cardiometabolic comorbidities in people living with HIV. MICROBIOME 2024; 12:106. [PMID: 38877521 PMCID: PMC11177534 DOI: 10.1186/s40168-024-01815-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/12/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Despite modern antiretroviral therapy (ART), people living with HIV (PLWH) have increased relative risk of inflammatory-driven comorbidities, including cardiovascular disease (CVD). The gut microbiome could be one of several driving factors, along with traditional risk factors and HIV-related risk factors such as coinfections, ART toxicity, and past immunodeficiency. RESULTS PLWH have an altered gut microbiome, even after adjustment for known confounding factors including sexual preference. The HIV-related microbiome has been associated with cardiometabolic comorbidities, and shares features with CVD-related microbiota profiles, in particular reduced capacity for short-chain fatty acid (SCFA) generation. Substantial inter-individual variation has so far been an obstacle for applying microbiota profiles for risk stratification. This review covers updated knowledge and recent advances in our understanding of the gut microbiome and comorbidities in PLWH, with specific focus on cardiometabolic comorbidities and inflammation. It covers a comprehensive overview of HIV-related and comorbidity-related dysbiosis, microbial translocation, and microbiota-derived metabolites. It also contains recent data from studies in PLWH on circulating metabolites related to comorbidities and underlying gut microbiota alterations, including circulating levels of the SCFA propionate, the histidine-analogue imidazole propionate, and the protective metabolite indole-3-propionic acid. CONCLUSIONS Despite recent advances, the gut microbiome and related metabolites are not yet established as biomarkers or therapeutic targets. The review gives directions for future research needed to advance the field into clinical practice, including promises and pitfalls for precision medicine. Video Abstract.
Collapse
Affiliation(s)
- Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.
- Section for Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Susanne Dam Nielsen
- Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen, 2200, Denmark
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, Copenhagen Oe, 2100, Denmark
| | - Ivan Vujkovic-Cvijin
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Karsh Division of Gastroenterology & Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
12
|
Suslov AV, Panas A, Sinelnikov MY, Maslennikov RV, Trishina AS, Zharikova TS, Zharova NV, Kalinin DV, Pontes-Silva A, Zharikov YO. Applied physiology: gut microbiota and antimicrobial therapy. Eur J Appl Physiol 2024; 124:1631-1643. [PMID: 38683402 DOI: 10.1007/s00421-024-05496-1] [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: 02/09/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
The gut microbiota plays an important role in maintaining human health and in the pathogenesis of several diseases. Antibiotics are among the most commonly prescribed drugs and have a significant impact on the structure and function of the gut microbiota. The understanding that a healthy gut microbiota prevents the development of many diseases has also led to its consideration as a potential therapeutic target. At the same time, any factor that alters the gut microbiota becomes important in this approach. Exercise and antibacterial therapy have a direct effect on the microbiota. The review reflects the current state of publications on the mechanisms of intestinal bacterial involvement in the pathogenesis of cardiovascular, metabolic, and neurodegenerative diseases. The physiological mechanisms of the influence of physical activity on the composition of the gut microbiota are considered. The mechanisms of the common interface between exercise and antibacterial therapy will be considered using the example of several socially important diseases. The aim of the study is to show the physiological relationship between the effects of exercise and antibiotics on the gut microbiota.
Collapse
Affiliation(s)
- Andrey V Suslov
- Russian National Centre of Surgery, Avtsyn Research Institute of Human Morphology, Moscow, 117418, Russia
- Pirogov Russian National Research Medical University (RNRMU), Moscow, 117997, Russia
| | - Alin Panas
- N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Bld. 2, Moscow, 119991, Russia
| | - Mikhail Y Sinelnikov
- Department of Oncology, Radiotherapy and Reconstructive Surgery, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119048, Russia
| | - Roman V Maslennikov
- Department of Internal Medicine, Gastroenterology and Hepatology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Aleksandra S Trishina
- N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Bld. 2, Moscow, 119991, Russia
| | - Tatyana S Zharikova
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 125009, Russia
- Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nataliya V Zharova
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 125009, Russia
| | - Dmitry V Kalinin
- Pathology Department, A.V. Vishnevsky National Medical Research Center of Surgery, Moscow, 115093, Russia
| | - André Pontes-Silva
- Postgraduate Program in Physical Therapy (PPGFT), Department of Physical Therapy (DFisio), Universidade Federal de São Carlos (UFSCar), São Carlos (SP), Brazil.
| | - Yury O Zharikov
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 125009, Russia
| |
Collapse
|
13
|
Bijla M, Saini SK, Pathak AK, Bharadwaj KP, Sukhavasi K, Patil A, Saini D, Yadav R, Singh S, Leeuwenburgh C, Kumar P. Microbiome interactions with different risk factors in development of myocardial infarction. Exp Gerontol 2024; 189:112409. [PMID: 38522483 DOI: 10.1016/j.exger.2024.112409] [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: 02/16/2024] [Revised: 03/10/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Among all non-communicable diseases, Cardiovascular Diseases (CVDs) stand as the leading global cause of mortality. Within this spectrum, Myocardial Infarction (MI) strikingly accounts for over 15 % of all deaths. The intricate web of risk factors for MI, comprising family history, tobacco use, oral health, hypertension, nutritional pattern, and microbial infections, is firmly influenced by the human gut and oral microbiota, their diversity, richness, and dysbiosis, along with their respective metabolites. Host genetic factors, especially allelic variations in signaling and inflammatory markers, greatly affect the progression or severity of the disease. Despite the established significance of the human microbiome-nutrient-metabolite interplay in associations with CVDs, the unexplored terrain of the gut-heart-oral axis has risen as a critical knowledge gap. Moreover, the pivotal role of the microbiome and the complex interplay with host genetics, compounded by age-related changes, emerges as an area of vital importance in the development of MI. In addition, a distinctive disease susceptibility and severity influenced by gender-based or ancestral differences, adds a crucial insights to the association with increased mortality. Here, we aimed to provide an overview on interactions of microbiome (oral and gut) with major risk factors (tobacco use, alcohol consumption, diet, hypertension host genetics, gender, and aging) in the development of MI and therapeutic regulation.
Collapse
Affiliation(s)
- Manisha Bijla
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, India
| | - Sunil Kumar Saini
- Department of Zoology, Swami Shraddhanand College, Delhi University, India
| | - Ajai Kumar Pathak
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia; Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | | | - Katyayani Sukhavasi
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital & Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
| | - Ayurshi Patil
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | - Diksha Saini
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | - Rakesh Yadav
- Department of Cardiology, AIIMS, New Delhi, India
| | - Shalini Singh
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | | | - Pramod Kumar
- ICMR-National Institute of Cancer Prevention and Research, Noida, India.
| |
Collapse
|
14
|
Shabnaz S, Nguyen TN, Williams R, Rubinstein SM, Garrett TJ, Tantawy M, Fradley MG, Alomar ME, Shain KH, Baz RC, Lenihan D, Cornell RF, Lu Q, Gong Y. Metabolomic signatures of carfilzomib-related cardiotoxicity in patients with multiple myeloma. Clin Transl Sci 2024; 17:e13828. [PMID: 38783568 PMCID: PMC11116757 DOI: 10.1111/cts.13828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/12/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
As a treatment for relapsed or refractory multiple myeloma (MM), carfilzomib has been associated with a significant risk of cardiovascular adverse events (CVAE). The goals of our study were to evaluate the metabolomic profile of MM patients to identify those at high risk prior to carfilzomib treatment and to explore the mechanisms of carfilzomib-CVAE to inform potential strategies to protect patients from this cardiotoxicity. Global metabolomic profiling was performed on the baseline and post-baseline plasma samples of 60 MM patients treated with carfilzomib-based therapy, including 31 who experienced CVAE, in a prospective cohort study. Baseline metabolites and post-baseline/baseline metabolite ratios that differ between the CVAE and no-CVAE patients were identified using unadjusted and adjusted methods. A baseline metabolomic risk score was created to stratify patients. We observed a lower abundance of tauroursodeoxycholic acid (T-UDCA) in CVAE patients at baseline (odds ratio [OR] = 0.47, 95% confidence interval [CI] = 0.21-0.94, p = 0.044) compared with the no-CVAE patients. A metabolite risk score was able to stratify patients into three risk groups. The area under the receiver-operating curve of the model with clinical predictors and metabolite risk score was 0.93. Glycochenodeoxycholic acid (OR = 0.56, 95% CI = 0.31-0.87, p = 0.023) was significantly lower in post-baseline/baseline ratios of CVAE patients compared with no-CVAE patients. Following metabolomic analysis, we created a baseline metabolite risk score that can stratify MM patients into different risk groups. The result also provided intriguing clues about the mechanism of carfilzomib-CVAE and potential cardioprotective strategies.
Collapse
Affiliation(s)
- Samia Shabnaz
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Trang N Nguyen
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Roy Williams
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Samuel M Rubinstein
- Department of Medicine, Division of Hematology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Marwa Tantawy
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mohammed E Alomar
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Kenneth H Shain
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Rachid C Baz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Daniel Lenihan
- Cape Cardiology Group, Saint Francis Medical Center, Cape Girardeau, Missouri, USA
| | - Robert F Cornell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Qing Lu
- Department of Biostatistics, College of Public Health and Health Professions & College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
- Cardio-Oncology Working Group, UF Health Cancer Center, Gainesville, Florida, USA
| |
Collapse
|
15
|
Mahenthiran A, Wilcox J, Tang WHW. Heart Failure: a Punch from the Gut. Curr Heart Fail Rep 2024; 21:73-80. [PMID: 38300390 PMCID: PMC10924029 DOI: 10.1007/s11897-024-00648-y] [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] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
PURPOSE OF REVIEW This article seeks to elucidate the mechanisms underlying the bidirectional relationship between the gut and the heart, focusing on the pathophysiology of heart failure. We have previously demonstrated that Heart failure (HF) has significant effects on splanchnic vasculature and leads to key alterations in the gut microbiome, portending greater comorbidity with HF. RECENT FINDINGS A growing field of research is focused on the effects of a "leaky gut" in the development of disease across organ systems. The leaky gut hypothesis centers on intestinal epithelial barrier dysfunction causing increased permeability of the gut and subsequent alterations to gut composition by endotoxins and microbial metabolites. Changes in the quantities of metabolites including short-chain fatty acids, trimethylamine N-oxide and other amino acid metabolites, and various bile acid species have been shown to result in gut dysbiosis and worsening HF. The gut plays a highly significant role in HF prognosis and requires greater attention for future therapeutic interventions. Treatments targeting gut composition could have very beneficial effects on HF prognosis.
Collapse
Affiliation(s)
| | - Jennifer Wilcox
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - W H Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44195, USA.
| |
Collapse
|
16
|
Raju SC, Molinaro A, Awoyemi A, Jørgensen SF, Braadland PR, Nendl A, Seljeflot I, Ueland PM, McCann A, Aukrust P, Vestad B, Mayerhofer C, Broch K, Gullestad L, Lappegård KT, Halvorsen B, Kristiansen K, Hov JR, Trøseid M. Microbial-derived imidazole propionate links the heart failure-associated microbiome alterations to disease severity. Genome Med 2024; 16:27. [PMID: 38331891 PMCID: PMC10854170 DOI: 10.1186/s13073-024-01296-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Interactions between the gut microbiota, diet, and host metabolism contribute to the development of cardiovascular disease, but a firm link between disease-specific gut microbiota alterations and circulating metabolites is lacking. METHODS We performed shot-gun sequencing on 235 samples from 166 HF patients and 69 healthy control samples. Separate plasma samples from healthy controls (n = 53) were used for the comparison of imidazole propionate (ImP) levels. Taxonomy and functional pathways for shotgun sequencing data was assigned using MetaPhlAn3 and HUMAnN3 pipelines. RESULTS Here, we show that heart failure (HF) is associated with a specific compositional and functional shift of the gut microbiota that is linked to circulating levels of the microbial histidine-derived metabolite ImP. Circulating ImP levels are elevated in chronic HF patients compared to controls and associated with HF-related gut microbiota alterations. Contrary to the microbiota composition, ImP levels provide insight into etiology and severity of HF and also associate with markers of intestinal permeability and systemic inflammation. CONCLUSIONS Our findings establish a connection between changes in the gut microbiota, the presence, etiology, and severity of HF, and the gut-microbially produced metabolite ImP. While ImP appears promising as a circulating biomarker reflecting gut dysbiosis related to HF, further studies are essential to demonstrate its causal or contributing role in HF pathogenesis. TRIAL REGISTRATION NCT02637167, registered December 22, 2015.
Collapse
Affiliation(s)
- Sajan C Raju
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Antonio Molinaro
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ayodeji Awoyemi
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Silje F Jørgensen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Peder R Braadland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andraz Nendl
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Ingebjørg Seljeflot
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | | | | | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Beate Vestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Cristiane Mayerhofer
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Knut T Lappegård
- Division of Internal Medicine, Nordlandssykehuset, 8005, Bodø, Norway
- Institute of Clinical Medicine, University of Tromsø, 9037, Tromsø, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen, 518083, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Johannes R Hov
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Transplantation Medicine, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Marius Trøseid
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.
| |
Collapse
|
17
|
Yuan L, Li Y, Chen M, Xue L, Wang J, Ding Y, Gu Q, Zhang J, Zhao H, Xie X, Wu Q. Therapeutic applications of gut microbes in cardiometabolic diseases: current state and perspectives. Appl Microbiol Biotechnol 2024; 108:156. [PMID: 38244075 PMCID: PMC10799778 DOI: 10.1007/s00253-024-13007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Cardiometabolic disease (CMD) encompasses a range of diseases such as hypertension, atherosclerosis, heart failure, obesity, and type 2 diabetes. Recent findings about CMD's interaction with gut microbiota have broadened our understanding of how diet and nutrition drive microbes to influence CMD. However, the translation of basic research into the clinic has not been smooth, and dietary nutrition and probiotic supplementation have yet to show significant evidence of the therapeutic benefits of CMD. In addition, the published reviews do not suggest the core microbiota or metabolite classes that influence CMD, and systematically elucidate the causal relationship between host disease phenotypes-microbiome. The aim of this review is to highlight the complex interaction of the gut microbiota and their metabolites with CMD progression and to further centralize and conceptualize the mechanisms of action between microbial and host disease phenotypes. We also discuss the potential of targeting modulations of gut microbes and metabolites as new targets for prevention and treatment of CMD, including the use of emerging technologies such as fecal microbiota transplantation and nanomedicine. KEY POINTS: • To highlight the complex interaction of the gut microbiota and their metabolites with CMD progression and to further centralize and conceptualize the mechanisms of action between microbial and host disease phenotypes. • We also discuss the potential of targeting modulations of gut microbes and metabolites as new targets for prevention and treatment of CMD, including the use of emerging technologies such as FMT and nanomedicine. • Our study provides insight into identification-specific microbiomes and metabolites involved in CMD, and microbial-host changes and physiological factors as disease phenotypes develop, which will help to map the microbiome individually and capture pathogenic mechanisms as a whole.
Collapse
Affiliation(s)
- Lin Yuan
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Moutong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Liang Xue
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Ding
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Science & Engineering, Jinan University, Guangzhou, 510632, China
| | - Qihui Gu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Hui Zhao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| |
Collapse
|
18
|
Usman I, Anwar A, Shukla S, Pathak P. Mechanistic Review on the Role of Gut Microbiota in the Pathology of Cardiovascular Diseases. Cardiovasc Hematol Disord Drug Targets 2024; 24:13-39. [PMID: 38879769 DOI: 10.2174/011871529x310857240607103028] [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: 02/16/2024] [Revised: 04/30/2024] [Accepted: 05/17/2024] [Indexed: 07/31/2024]
Abstract
Cardiovascular diseases (CVDs), which stand as the primary contributors to illness and death on a global scale, include vital risk factors like hyperlipidemia, hypertension, diabetes, and smoking, to name a few. However, conventional cardiovascular risk factors offer only partial insight into the complexity of CVDs. Lately, a growing body of research has illuminated that the gut microbiome and its by-products are also of paramount importance in the initiation and progression of CVDs. The gastrointestinal tract houses trillions of microorganisms, commonly known as gut microbiota, that metabolize nutrients, yielding substances like trimethylamine-N-oxide (TMAO), bile acids (BAs), short-chain fatty acids (SCFAs), indoxyl sulfate (IS), and so on. Strategies aimed at addressing these microbes and their correlated biological pathways have shown promise in the management and diagnosis of CVDs. This review offers a comprehensive examination of how the gut microbiota contributes to the pathogenesis of CVDs, particularly atherosclerosis, hypertension, heart failure (HF), and atrial fibrillation (AF), explores potential underlying mechanisms, and highlights emerging therapeutic prospects in this dynamic domain.
Collapse
Affiliation(s)
- Iqra Usman
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Aamir Anwar
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Shivang Shukla
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Priya Pathak
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| |
Collapse
|
19
|
Nowiński A, Chabowski D, Giebułtowicz J, Aleksandrowicz M, Ufnal M. Deoxycholic Acid, a Secondary Bile Acid, Increases Cardiac Output and Blood Pressure in Rats. Nutrients 2023; 16:32. [PMID: 38201862 PMCID: PMC10781055 DOI: 10.3390/nu16010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Deoxycholic acid (DCA) is a secondary bile acid produced by gut bacteria. Elevated serum concentrations of DCA are observed in cardiovascular disease (CVD). We hypothesized that DCA might influence hemodynamic parameters in rats. METHODS The concentration of DCA in systemic blood was measured with liquid chromatography coupled with mass spectrometry. Arterial blood pressure (BP), heart rate (HR) and echocardiographic parameters were evaluated in anesthetized, male, 3-4-month-old Sprague-Dawley rats administered intravenously (IV) or intracerebroventricularly (ICV) with investigated compounds. Mesenteric artery (MA) reactivity was tested ex vivo. RESULTS The baseline plasma concentration of DCA was 0.24 ± 0.03 mg/L. The oral antibiotic treatment produced a large decrease in the concentration. Administered IV, the compound increased BP and HR in a dose-dependent manner. DCA also increased heart contractility and cardiac output. None of the tested compounds-prazosin (an alpha-blocker), propranolol (beta-adrenolytic), atropine (muscarinic receptor antagonist), glibenclamide (K-ATP inhibitor) or DY 268 (FXR antagonist), glycyrrhetinic acid (11HSD2 inhibitor)-significantly diminished the DCA-induced pressor effect. ICV infusion did not exert significant HR or BP changes. DCA relaxed MAs. Systemic vascular resistance did not change significantly. CONCLUSIONS DCA elevates BP primarily by augmenting cardiac output. As a metabolite derived from gut bacteria, DCA potentially serves as a mediator in the interaction between the gut microbiota and the host's circulatory system.
Collapse
Affiliation(s)
- Artur Nowiński
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-106 Warsaw, Poland; (D.C.); (M.U.)
| | - Dawid Chabowski
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-106 Warsaw, Poland; (D.C.); (M.U.)
| | - Joanna Giebułtowicz
- Department of Bioanalysis and Drugs Analysis, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha, 02-097 Warsaw, Poland;
| | - Marta Aleksandrowicz
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Marcin Ufnal
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-106 Warsaw, Poland; (D.C.); (M.U.)
| |
Collapse
|
20
|
Liu Z, Luo G, Du R, Kan G, Han X, Zhong G, Xing W, Cui Y, Sun W, Li J, Li Y, Zhao D, Yuan X, Jin X, Han Y, Sun H, Ling S, Li Y. Simulated spaceflight-induced cardiac remodeling is modulated by gut microbial-derived trimethylamine N-oxide. iScience 2023; 26:108556. [PMID: 38125015 PMCID: PMC10730869 DOI: 10.1016/j.isci.2023.108556] [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: 08/28/2023] [Revised: 10/19/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Spaceflight is physically demanding and can negatively affect astronauts' health. It has been shown that the human gut microbiota and cardiac function are affected by spaceflight and simulated spaceflight. This study investigated the effects of the gut microbiota on simulated spaceflight-induced cardiac remodeling using 10° of head-down bed rest (HDBR) in rhesus macaques and 30° of hindlimb unloading (HU) in mice. The gut microbiota, fecal metabolites, and cardiac remodeling were markedly affected by HDBR in macaques and HU in mice, cardiac remodeling in control mice was affected by the gut microbiota of HU mice and that of HU mice was protected by the gut microbiota of control mice, and there was a correlation between cardiac remodeling and the gut microbial-derived metabolite trimethylamine N-oxide. These findings suggest that spaceflight can affect cardiac remodeling by modulating the gut microbiota and fecal metabolites.
Collapse
Affiliation(s)
- Zizhong Liu
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Gui Luo
- Department of Rheumatology and Immunology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ruikai Du
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Guanghan Kan
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xuan Han
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Guohui Zhong
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Wenjuan Xing
- School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the Ministry of Education, Fourth Military Medical University, Xi’an, China
| | - Ying Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weijia Sun
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Jianwei Li
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Yuheng Li
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Dingsheng Zhao
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xinxin Yuan
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoyan Jin
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Yanping Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Huiyuan Sun
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Shukuan Ling
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - Yingxian Li
- National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
21
|
Martemucci G, Fracchiolla G, Muraglia M, Tardugno R, Dibenedetto RS, D’Alessandro AG. Metabolic Syndrome: A Narrative Review from the Oxidative Stress to the Management of Related Diseases. Antioxidants (Basel) 2023; 12:2091. [PMID: 38136211 PMCID: PMC10740837 DOI: 10.3390/antiox12122091] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Metabolic syndrome (MS) is a growing disorder affecting thousands of people worldwide, especially in industrialised countries, increasing mortality. Oxidative stress, hyperglycaemia, insulin resistance, inflammation, dysbiosis, abdominal obesity, atherogenic dyslipidaemia and hypertension are important factors linked to MS clusters of different pathologies, such as diabesity, cardiovascular diseases and neurological disorders. All biochemical changes observed in MS, such as dysregulation in the glucose and lipid metabolism, immune response, endothelial cell function and intestinal microbiota, promote pathological bridges between metabolic syndrome, diabesity and cardiovascular and neurodegenerative disorders. This review aims to summarise metabolic syndrome's involvement in diabesity and highlight the link between MS and cardiovascular and neurological diseases. A better understanding of MS could promote a novel strategic approach to reduce MS comorbidities.
Collapse
Affiliation(s)
- Giovanni Martemucci
- Department of Agricultural and Environmental Sciences, University of Bari Aldo Moro, 70126 Bari, Italy;
| | - Giuseppe Fracchiolla
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Marilena Muraglia
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Roberta Tardugno
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Roberta Savina Dibenedetto
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | | |
Collapse
|
22
|
Russo MA, Garaci E, Frustaci A, Fini M, Costantini C, Oikonomou V, Nunzi E, Puccetti P, Romani L. Host-microbe tryptophan partitioning in cardiovascular diseases. Pharmacol Res 2023; 198:106994. [PMID: 37972721 DOI: 10.1016/j.phrs.2023.106994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
The functional interdependencies between the molecular components of a biological process demand for a network medicine platform that integrates systems biology and network science, to explore the interactions among biological components in health and disease. Access to large-scale omics datasets (genomics, transcriptomics, proteomics, metabolomics, metagenomics, phenomics, etc.) has significantly advanced our opportunity along this direction. Studies utilizing these techniques have begun to provide us with a deeper understanding of how the interaction between the intestinal microbes and their host affects the cardiovascular system in health and disease. Within the framework of a multiomics network approach, we highlight here how tryptophan metabolism may orchestrate the host-microbes interaction in cardiovascular diseases and the implications for precision medicine and therapeutics, including nutritional interventions.
Collapse
Affiliation(s)
- Matteo Antonio Russo
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Andrea Frustaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Massimo Fini
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Vasileios Oikonomou
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Luigina Romani
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy; Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy.
| |
Collapse
|
23
|
Ahmad AF, Caparrós-Martin JA, Gray N, Lodge S, Wist J, Lee S, O'Gara F, Shah A, Ward NC, Dwivedi G. Insights into the associations between the gut microbiome, its metabolites, and heart failure. Am J Physiol Heart Circ Physiol 2023; 325:H1325-H1336. [PMID: 37737730 DOI: 10.1152/ajpheart.00436.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/05/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
Heart failure (HF) is the end stage of most cardiovascular diseases and remains a significant health problem globally. We aimed to assess whether patients with left ventricular ejection fraction ≤45% had alterations in both the gut microbiome profile and production of associated metabolites when compared with a healthy cohort. We also examined the associated inflammatory, metabolomic, and lipidomic profiles of patients with HF. This single center, observational study, recruited 73 patients with HF and 59 healthy volunteers. Blood and stool samples were collected at baseline and 6-mo follow-up, along with anthropometric and clinical data. When compared with healthy controls, patients with HF had reduced gut bacterial alpha diversity at follow-up (P = 0.004) but not at baseline. The stool microbiota of patients with HF was characterized by a depletion of operational taxonomic units representing commensal Clostridia at both baseline and follow-up. Patients with HF also had significantly elevated baseline plasma acetate (P = 0.007), plasma trimethylamine-N-oxide (TMAO) (P = 0.003), serum soluble CD14 (sCD14; P = 0.005), and soluble CD163 (sCD163; P = 0.004) levels compared with healthy controls. Furthermore, patients with HF had a distinct metabolomic and lipidomic profile at baseline when compared with healthy controls. Differences in the composition of the gut microbiome and the levels of associated metabolites were observed in patients with HF when compared with a healthy cohort. This was also associated with an altered metabolomic and lipidomic profile. Our study identifies microorganisms and metabolites that could represent new therapeutic targets and diagnostic tools in the pathogenesis of HF.NEW & NOTEWORTHY We found a reduction in gut bacterial alpha diversity in patients with heart failure (HF) and that the stool microbiota of patients with HF was characterized by depletion of operational taxonomic units representing commensal Clostridia at both baseline and follow-up. Patients with HF also had altered bacterial metabolites and increased inflammatory profiles compared with healthy controls. A distinct metabolomic and lipidomic profile was present in patients with HF at baseline when compared with healthy controls.
Collapse
Affiliation(s)
- Adilah F Ahmad
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Jose A Caparrós-Martin
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Nicola Gray
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Microbiology, PathWest Laboratory Medicine, Perth, Western Australia, Australia
| | - Fergal O'Gara
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
- BIOMERIT Research Centre, School of Microbiology, University College Cork, Cork, Ireland
| | - Amit Shah
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Natalie C Ward
- Dobney Hypertension Centre, Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| |
Collapse
|
24
|
Liu S, He Y, Zhang Y, Zhang Z, Huang K, Deng L, Liao B, Zhong Y, Feng J. Targeting gut microbiota in aging-related cardiovascular dysfunction: focus on the mechanisms. Gut Microbes 2023; 15:2290331. [PMID: 38073096 PMCID: PMC10730151 DOI: 10.1080/19490976.2023.2290331] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
The global population is aging and age-related cardiovascular disease is increasing. Even after controlling for cardiovascular risk factors, readmission and mortality rates remain high. In recent years, more and more in-depth studies have found that the composition of the gut microbiota and its metabolites, such as trimethylamine N-oxide (TMAO), bile acids (BAs), and short-chain fatty acids (SCFAs), affect the occurrence and development of age-related cardiovascular diseases through a variety of molecular pathways, providing a new target for therapy. In this review, we discuss the relationship between the gut microbiota and age-related cardiovascular diseases, and propose that the gut microbiota could be a new therapeutic target for preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Siqi Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yali Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Zhaolun Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Keming Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
25
|
Shi M, Wei J, Yuan H, Li Y, Guo Z. The role of the gut microbiota and bile acids in heart failure: A review. Medicine (Baltimore) 2023; 102:e35795. [PMID: 37960774 PMCID: PMC10637566 DOI: 10.1097/md.0000000000035795] [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: 02/27/2023] [Accepted: 10/04/2023] [Indexed: 11/15/2023] Open
Abstract
Heart failure (HF) is the terminal manifestation of various cardiovascular diseases. Recently, accumulating evidence has demonstrated that gut microbiota are involved in the development of various cardiovascular diseases. Gut microbiota and their metabolites might play a pivotal role in the development of HF. However, previous studies have rarely described the complex role of gut microbiota and their metabolites in HF. In this review, we mainly discussed bile acids (BAs), the metabolites of gut microbiota. We explained the mechanisms by which BAs are involved in the pathogenesis of HF. We also discussed the use of gut microbiota and BAs for treating HF in Chinese medicine, highlighting the advantages of Chinese medicine in treating HF.
Collapse
Affiliation(s)
- Min Shi
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan, Changsha, China
| | - Jiaming Wei
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan, Changsha, China
| | - Hui Yuan
- Hunan University of Chinese Medicine, Changsha, China
| | - Ya Li
- Hunan University of Chinese Medicine, Changsha, China
| | - Zhihua Guo
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Key Laboratory of Colleges and Universities of Intelligent Traditional Chinese Medicine Diagnosis and Preventive Treatment of Chronic Diseases of Hunan, Changsha, China
| |
Collapse
|
26
|
Liu Y, Huang Y, He Q, Dou Z, Zeng M, Wang X, Li S. From heart to gut: Exploring the gut microbiome in congenital heart disease. IMETA 2023; 2:e144. [PMID: 38868221 PMCID: PMC10989834 DOI: 10.1002/imt2.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 06/14/2024]
Abstract
Congenital heart disease (CHD) is a prevalent birth defect and a significant contributor to childhood mortality. The major characteristics of CHD include cardiovascular malformations and hemodynamical disorders. However, the impact of CHD extends beyond the circulatory system. Evidence has identified dysbiosis of the gut microbiome in patients with CHD. Chronic hypoxia and inflammation associated with CHD affect the gut microbiome, leading to alterations in its number, abundance, and composition. The gut microbiome, aside from providing essential nutrients, engages in direct interactions with the host immune system and indirect interactions via metabolites. The abnormal gut microbiome or its products can translocate into the bloodstream through an impaired gut barrier, leading to an inflammatory state. Metabolites of the gut microbiome, such as short-chain fatty acids and trimethylamine N-oxide, also play important roles in the development, treatment, and prognosis of CHD. This review discusses the role of the gut microbiome in immunity, gut barrier, neurodevelopment, and perioperative period in CHD. By fostering a better understanding of the cross-talk between CHD and the gut microbiome, this review aims to contribute to improve clinical management and outcomes for CHD patients.
Collapse
Affiliation(s)
- Yuze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yuan Huang
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Qiyu He
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Zheng Dou
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Min Zeng
- Department of Pediatric Intensive Care Unit, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Xu Wang
- Department of Pediatric Intensive Care Unit, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Shoujun Li
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| |
Collapse
|
27
|
Almeida C, Gonçalves-Nobre JG, Alpuim Costa D, Barata P. The potential links between human gut microbiota and cardiovascular health and disease - is there a gut-cardiovascular axis? FRONTIERS IN GASTROENTEROLOGY 2023; 2. [DOI: 10.3389/fgstr.2023.1235126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut-heart axis is an emerging concept highlighting the crucial link between gut microbiota and cardiovascular diseases (CVDs). Recent studies have demonstrated that gut microbiota is pivotal in regulating host metabolism, inflammation, and immune function, critical drivers of CVD pathophysiology. Despite a strong link between gut microbiota and CVDs, this ecosystem’s complexity still needs to be fully understood. The short-chain fatty acids, trimethylamine N-oxide, bile acids, and polyamines are directly or indirectly involved in the development and prognosis of CVDs. This review explores the relationship between gut microbiota metabolites and CVDs, focusing on atherosclerosis and hypertension, and analyzes personalized microbiota-based modulation interventions, such as physical activity, diet, probiotics, prebiotics, and fecal microbiota transplantation, as a promising strategy for CVD prevention and treatment.
Collapse
|
28
|
Gundogdu A, Nalbantoglu OU. The role of the Mediterranean diet in modulating the gut microbiome: A review of current evidence. Nutrition 2023; 114:112118. [PMID: 37437419 DOI: 10.1016/j.nut.2023.112118] [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: 02/06/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 07/14/2023]
Abstract
The Mediterranean diet (MedDiet) is recognized as one of the United Nations Educational, Scientific and Cultural Organization Intangible Cultural Heritage assets associated with lower rates of cardiometabolic diseases; lower prevalence of cancer, Alzheimer's disease, depression, and onset of inflammatory bowel disease; and more generally low-grade inflammation and mortality risks. Beyond being an input source of beneficial micronutrients, it recently has been discovered that the MedDiet plays a role in a more complex human microbiome-mediated mechanism. An interesting hypothesis suggests a bidirectional relationship between the MedDiet and the gut microbiome, where gut microbiota assembly and biosynthetic capacity are responsive to the diet; in return, the microbiome-reachable nutrients shape and modulate the microbiome toward a characteristic probiotic state. It can be speculated that that primary health benefits of the MedDiet exerted via the gut microbiome are mediated by the bioactive compounds transformed by the microbiome. Furthermore, it is possible that additional probiotic properties of the organisms promoted by diet adherence have secondary benefits. As more detailed omic-based studies take place, more evidence on the MedDiet as a core generic probiotic microbiome modulation strategy surface. However, individual-specific microbiome compositions might impose personal variations on the diet outcome. Therefore, a prospective strategy of a fine-tuned precision nutrition approach might deliver optimized benefits of the MedDiet.
Collapse
Affiliation(s)
- Aycan Gundogdu
- Department of Microbiology and Clinical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey; Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey.
| | - Ozkan Ufuk Nalbantoglu
- Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey; Department of Computer Engineering, Faculty of Engineering, Erciyes University, Kayseri, Turkey
| |
Collapse
|
29
|
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
|
30
|
Abstract
PURPOSE OF REVIEW The role and underlying mechanisms mediated by dietary salt in modulating the gut microbiota and contributing to heart failure (HF) are not clear. This review summarizes the mechanisms of dietary salt and the gut-heart axis in HF. RECENT FINDINGS The gut microbiota has been implicated in several cardiovascular diseases (CVDs) including HF. Dietary factors including high consumption of salt play a role in influencing the gut microbiota, resulting in dysbiosis. An imbalance of microbial species due to a reduction in microbial diversity with accompanying immune cell activation has been implicated in the pathogenesis of HF via several mechanisms. The gut microbiota and gut-associated metabolites contribute to HF by reducing gut microbiota biodiversity and activating several signaling pathways. High dietary salt modulates the gut microbiota composition and exacerbate or induce HF by increasing the expression of the epithelial sodium/hydrogen exchanger isoform 3 in the gut, cardiac expression of beta myosin heavy chain, activation of the myocyte enhancer factor/nuclear factor of activated T cell, and salt-inducible kinase 1. These mechanisms explain the resulting structural and functional derangements in patients with HF.
Collapse
Affiliation(s)
- Sepiso K Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Zambia
- Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37232-6602, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37232-6602, USA.
| |
Collapse
|
31
|
Dela Cruz M, Lin H, Han J, Adler E, Boissiere J, Khalid M, Sidebottom A, Sundararajan A, Lehmann C, Moran A, Odenwald M, Stutz M, Kim G, Pinney S, Jeevanandam V, Alegre ML, Pamer E, Nguyen AB. Reduced immunomodulatory metabolite concentrations in peri-transplant fecal samples from heart allograft recipients. FRONTIERS IN TRANSPLANTATION 2023; 2:1182534. [PMID: 38993864 PMCID: PMC11235359 DOI: 10.3389/frtra.2023.1182534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2024]
Abstract
Background Emerging evidence is revealing the impact of the gut microbiome on hematopoietic and solid organ transplantation. Prior studies postulate that this influence is mediated by bioactive metabolites produced by gut-dwelling commensal bacteria. However, gut microbial metabolite production has not previously been measured among heart transplant (HT) recipients. Methods In order to investigate the potential influence of the gut microbiome and its metabolites on HT, we analyzed the composition and metabolite production of the fecal microbiome among 48 HT recipients at the time of HT. Results Compared to 20 healthy donors, HT recipients have significantly reduced alpha, i.e. within-sample, microbiota diversity, with significantly lower abundances of key anaerobic commensal bacteria and higher abundances of potentially pathogenic taxa that have been correlated with adverse outcomes in other forms of transplantation. HT recipients have a wide range of microbiota-derived fecal metabolite concentrations, with significantly reduced levels of immune modulatory metabolites such as short chain fatty acids and secondary bile acids compared to healthy donors. These differences were likely due to disease severity and prior antibiotic exposures but were not explained by other demographic or clinical factors. Conclusions Key potentially immune modulatory gut microbial metabolites are quantifiable and significantly reduced among HT recipients compared to healthy donors. Further study is needed to understand whether this wide range of gut microbial dysbiosis and metabolite alterations impact clinical outcomes and if they can be used as predictive biomarkers or manipulated to improve transplant outcomes.
Collapse
Affiliation(s)
- Mark Dela Cruz
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| | - Huaiying Lin
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Jiho Han
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| | - Emerald Adler
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Jaye Boissiere
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Maryam Khalid
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Ashley Sidebottom
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Anitha Sundararajan
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Christopher Lehmann
- Department of Medicine, Section of Infectious Diseases, University of Chicago Medicine, Chicago, IL, United States
| | - Angelica Moran
- Department of Pathology, University of Chicago Medicine, Chicago, IL, United States
| | - Matthew Odenwald
- Department of Medicine, Section of Gastroenterology, University of Chicago Medicine, Chicago, IL, United States
| | - Matthew Stutz
- Department of Medicine, Section of Pulmonary and Critical Care, University of Chicago Medicine, Chicago, IL, United States
| | - Gene Kim
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| | - Sean Pinney
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| | - Valluvan Jeevanandam
- Department of Surgery, Section of Cardiac Surgery, University of Chicago Medicine, Chicago, IL, United States
| | - Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, United States
| | - Eric Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Ann B. Nguyen
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| |
Collapse
|
32
|
Wegermann K, Fudim M, Henao R, Howe CF, McGarrah R, Guy C, Abdelmalek MF, Diehl AM, Moylan CA. Serum Metabolites Are Associated With HFpEF in Biopsy-Proven Nonalcoholic Fatty Liver Disease. J Am Heart Assoc 2023:e029873. [PMID: 37421270 PMCID: PMC10382080 DOI: 10.1161/jaha.123.029873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/05/2023] [Indexed: 07/10/2023]
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) and heart failure with preserved ejection fraction (HFpEF) share common risk factors, including obesity and diabetes. They are also thought to be mechanistically linked. The aim of this study was to define serum metabolites associated with HFpEF in a cohort of patients with biopsy-proven NAFLD to identify common mechanisms. Methods and Results We performed a retrospective, single-center study of 89 adult patients with biopsy-proven NAFLD who had transthoracic echocardiography performed for any indication. Metabolomic analysis was performed on serum using ultrahigh performance liquid and gas chromatography/tandem mass spectrometry. HFpEF was defined as ejection fraction >50% plus at least 1 echocardiographic feature of HFpEF (diastolic dysfunction, abnormal left atrial size) and at least 1 heart failure sign or symptom. We performed generalized linear models to evaluate associations between individual metabolites, NAFLD, and HFpEF. Thirty-seven out of 89 (41.6%) patients met criteria for HFpEF. A total of 1151 metabolites were detected; 656 were analyzed after exclusion of unnamed metabolites and those with >30% missing values. Fifty-three metabolites were associated with the presence of HFpEF with unadjusted P value <0.05; none met statistical significance after adjustment for multiple comparisons. The majority (39/53, 73.6%) were lipid metabolites, and levels were generally increased. Two cysteine metabolites (cysteine s-sulfate and s-methylcysteine) were present at significantly lower levels in patients with HFpEF. Conclusions We identified serum metabolites associated with HFpEF in patients with biopsy-proven NAFLD, with increased levels of multiple lipid metabolites. Lipid metabolism could be an important pathway linking HFpEF to NAFLD.
Collapse
Affiliation(s)
- Kara Wegermann
- Division of Gastroenterology, Department of Medicine Duke University Health System Durham NC
| | - Marat Fudim
- Division of Cardiology, Department of Medicine Duke University Health System Durham NC
| | - Ricardo Henao
- Department of Biostatistics and Bioinformatics Duke University Durham NC
| | | | - Robert McGarrah
- Division of Cardiology, Department of Medicine Duke University Health System Durham NC
| | - Cynthia Guy
- Department of Pathology Duke University Hospital Durham NC
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic Rochester MN
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine Duke University Health System Durham NC
| | - Cynthia A Moylan
- Division of Gastroenterology, Department of Medicine Duke University Health System Durham NC
- Department of Medicine, Durham Veterans Affairs Medical Center Durham NC
| |
Collapse
|
33
|
Guiducci L, Nicolini G, Forini F. Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease. Metabolites 2023; 13:760. [PMID: 37367916 DOI: 10.3390/metabo13060760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
The cardiovascular and metabolic disorders, collectively known as cardiometabolic disease (CMD), are high morbidity and mortality pathologies associated with lower quality of life and increasing health-care costs. The influence of the gut microbiota (GM) in dictating the interpersonal variability in CMD susceptibility, progression and treatment response is beginning to be deciphered, as is the mutualistic relation established between the GM and diet. In particular, dietary factors emerge as pivotal determinants shaping the architecture and function of resident microorganisms in the human gut. In turn, intestinal microbes influence the absorption, metabolism, and storage of ingested nutrients, with potentially profound effects on host physiology. Herein, we present an updated overview on major effects of dietary components on the GM, highlighting the beneficial and detrimental consequences of diet-microbiota crosstalk in the setting of CMD. We also discuss the promises and challenges of integrating microbiome data in dietary planning aimed at restraining CMD onset and progression with a more personalized nutritional approach.
Collapse
Affiliation(s)
- Letizia Guiducci
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124 Pisa, Italy
| | | | - Francesca Forini
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
34
|
Cui H, Han S, Dai Y, Xie W, Zheng R, Sun Y, Xia X, Deng X, Cao Y, Zhang M, Shang H. Gut microbiota and integrative traditional Chinese and western medicine in prevention and treatment of heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154885. [PMID: 37302262 DOI: 10.1016/j.phymed.2023.154885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Heart failure (HF) is the terminal stage of multiple cardiovascular diseases, with high mortality and morbidity. More and more studies have proved that gut microbiota may play a role in the process of HF, which is expected to become a new therapeutic target. The combination of traditional Chinese and Western medicine has vast therapeutic potential of complementation against HF. PURPOSE This manuscript expounds on the research progress of mechanisms of gut microbiota participating in the occurrence and prognosis of HF and the role of integrative traditional Chinese and Western medicine from 1987 to 2022. The combination of traditional Chinese and Western medicine in the prevention and treatment of HF from the perspective of gut microbiota has been discussed. METHODS Studies focusing on the effects and their mechanisms of gut microbiota in HF and the role of integrative traditional Chinese and Western medicine were identified and summarized, including contributions from February 1987 until August 2022. The investigation was carried out in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. We searched PubMed, Embase, Cochrane Library, CNKI, Wanfang, and VIP databases up to April 2023 by using the relevant keywords and operators. RESULTS A total of 34 articles were finally included in this review.16 RCTs and 13 basic researches, and 3 clinical research studies involving 7 relevant outcome indicators(cardiac function evaluation index, changes in gut microbiota, inflammatory factors, metabolites of gut microbiota, serum nutritional index protein, quality of life score, intestinal permeability and all-cause mortality). Compared with healthy controls, serum TNF-α and TMAO levels were significantly higher in patients with heart failure [MD = 5.77, 95%CI(4.97, 6.56), p < 0.0001; SMD = 1.92, 95%CI(1.70, 2.14), p < 0.0001]. Escherichia coli and Thick-walled bacteria increased significantly [SMD = -0.99, 95%CI(-1.38, -0.61), p < 0.0001, SMD = 2.58, 95%CI(2.23, 2.93), p < 0.0001];The number of bacteroides and lactobacillus decreased [SMD = -2.29, 95%CI(-2.54, -2.04), p < 0.0001; SMD = -1.55, 95%CI(-1.8, -1.3), p < 0.0001]. There was no difference in bifidobacterium [SMD = 0.16, 95%CI(-0.22, 0.54), p = 0.42]. In the published literature, it is not difficult to see that most of the results are studied and proved based on animal experiments or clinical trials, involving the cellular level, while the mechanism and mode of action of the molecular biology of traditional Chinese medicine are less elaborated, which is related to the characteristics of multi-components and multi-targets of traditional Chinese medicine. The above are the shortcomings of published literature, which can also be the direction of future research. CONCLUSION Heart failure patients have decreased beneficial bacteria such as Bacillus mimics and Lactobacillus in the intestinal flora and increased harmful flora like thick-walled flora. And increase the inflammatory response of the body and the expression of trimethylamine oxide (TMAO) in the serum. And The prevention and treatment of integrative traditional Chinese and Western medicine against heart failure based on gut microbiota and its metabolites is a promising research direction.
Collapse
Affiliation(s)
- Herong Cui
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Songjie Han
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yanan Dai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wei Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rui Zheng
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yang Sun
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaofeng Xia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaopeng Deng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yaru Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mei Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
35
|
Guan XQ, Wang CH, Cheng P, Fu LY, Wu QJ, Cheng G, Guan L, Sun ZJ. Effects of Empagliflozin on Gut Microbiota in Heart Failure with a Preserved Ejection Fraction: The Design of a Pragmatic Randomized, Open-Label Controlled Trial (EMPAGUM). Drug Des Devel Ther 2023; 17:1495-1502. [PMID: 37223722 PMCID: PMC10202117 DOI: 10.2147/dddt.s404479] [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] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/01/2023] [Indexed: 05/25/2023] Open
Abstract
Although empagliflozin has been recommended for individuals with heart failure, its effects on heart failure with preserved ejection fraction (HFpEF) remain uncertain from a physiopathological standpoint. The metabolites produced by gut microbiota have been shown to have a crucial role in the development of heart failure. Sodium-glucose cotransporter-2 inhibitors (SGLT2) have been shown to change the make-up of the gut microbiota in rodent studies. There is mixed evidence from similar studies investigating whether or not SGLT2 can affect the microbiota in the human gut. This trial is a pragmatic, randomized, open-label controlled study with empagliflozin as an intervention. We will enroll 100 patients with HFpEF and randomly assign them to one of two groups to receive either empagliflozin or a placebo. Patients in the Empagliflozin group will be given 10 mg of the drug daily, while those in the Control group will not be given empagliflozin or any other SGLT2. The purpose of the trial is to validate the changes that occur in gut microbiota in patients with HFpEF who take empagliflozin and to investigate the function of gut microbiota and their metabolites in the process.
Collapse
Affiliation(s)
- Xue-Qing Guan
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
| | - Chuan-He Wang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
| | - Peng Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
| | - Ling-Yu Fu
- Department of Clinical Epidemiology and Evidence-Based Medicine, The First Affiliated Hospital, China Medical University, Shenyang, 110021, People’s Republic of China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
| | - Gong Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
| | - Lin Guan
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
| | - Zhi-Jun Sun
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110021, People’s Republic of China
| |
Collapse
|
36
|
Mamic P, Snyder M, Tang WHW. Gut Microbiome-Based Management of Patients With Heart Failure: JACC Review Topic of the Week. J Am Coll Cardiol 2023; 81:1729-1739. [PMID: 37100490 DOI: 10.1016/j.jacc.2023.02.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 04/28/2023]
Abstract
Despite therapeutic advances, chronic heart failure (HF) is still associated with significant risk of morbidity and mortality. The course of disease and responses to therapies vary widely among individuals with HF, highlighting the need for precision medicine approaches. Gut microbiome stands to be an important aspect of precision medicine in HF. Exploratory clinical studies have revealed shared patterns of gut microbiome dysregulation in this disease, with mechanistic animal studies providing evidence for active involvement of the gut microbiome in development and pathophysiology of HF. Deeper insights into gut microbiome-host interactions in patients with HF promise to deliver novel disease biomarkers, preventative and therapeutic targets, and improve disease risk stratification. This knowledge may enable a paradigm shift in how we care for patients with HF, and pave the path toward improved clinical outcomes through personalized HF care.
Collapse
Affiliation(s)
- Petra Mamic
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford University, Stanford, California, USA; Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, California, USA.
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, California, USA
| | - W H Wilson Tang
- Kaufman Center for Heart Failure Treatment and Recovery, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
37
|
Fan Y, Ying J, Ma H, Cui H. Microbiota-related metabolites fueling the understanding of ischemic heart disease. IMETA 2023; 2:e94. [PMID: 38868424 PMCID: PMC10989774 DOI: 10.1002/imt2.94] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/04/2023] [Accepted: 01/21/2023] [Indexed: 06/14/2024]
Abstract
Up-to-date knowledge of gut microbial taxa associated with ischemic heart disease (IHD). Microbial metabolites for mechanistic dissection of IHD pathology. Microbiome-based therapies in IHD prevention and treatment.
Collapse
Affiliation(s)
- Yong Fan
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Jiajun Ying
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
- Department of Cardiology, Ningbo First HospitalNingbo UniversityNingboChina
| | - Hongchuang Ma
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Hanbin Cui
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
- Department of Cardiology, Ningbo First HospitalNingbo UniversityNingboChina
- Ningbo Clinical Research Center for Cardiovascular DiseaseNingboChina
| |
Collapse
|
38
|
Matsiras D, Bezati S, Ventoulis I, Verras C, Parissis J, Polyzogopoulou E. Gut Failure: A Review of the Pathophysiology and Therapeutic Potentials in the Gut-Heart Axis. J Clin Med 2023; 12:2567. [PMID: 37048650 PMCID: PMC10095379 DOI: 10.3390/jcm12072567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Despite considerable advances in the field, heart failure (HF) still poses a significant disease burden among affected individuals since it continues to cause high morbidity and mortality rates. Inflammation is considered to play a key role in disease progression, but the exact underlying pathophysiological mechanisms involved have not yet been fully elucidated. The gut, as a potential source of inflammation, could feasibly explain the state of low-grade inflammation seen in patients with chronic HF. Several derangements in the composition of the microbiota population, coupled with an imbalance between favorable and harmful metabolites and followed by gut barrier disruption and eventually bacterial translocation, could contribute to cardiac dysfunction and aggravate HF. On the other hand, HF-associated congestion and hypoperfusion alters intestinal function, thereby creating a vicious cycle. Based on this evidence, novel pharmaceutical agents have been developed and their potential therapeutic use has been tested in both animal and human subjects. The ultimate goal in these efforts is to reverse the aforementioned intestinal derangements and block the inflammation cascade. This review summarizes the gut-related causative pathways implicated in HF pathophysiology, as well as the associated therapeutic interventions described in the literature.
Collapse
Affiliation(s)
- Dionysis Matsiras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Sofia Bezati
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece
| | - Christos Verras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - John Parissis
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Effie Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| |
Collapse
|
39
|
Ren H, Zhu B, An Y, Xie F, Wang Y, Tan Y. Immune communication between the intestinal microbiota and the cardiovascular system. Immunol Lett 2023; 254:13-20. [PMID: 36693435 DOI: 10.1016/j.imlet.2023.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/27/2022] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
The intestine hosts a large number of microbial communities. Recent studies have shown that gut microbiota-mediated immune responses play a vital role in developing cardiovascular diseases (CVD). Immune cells are extensively infiltrated in the gut and heart tissues, such as T cells, B cells, and macrophages. They play a crucial role in the crosstalk between the heart and gut microbiota. And the microbiota influences the bidirectional function of immune cells in CVD such as myocardial infarction and atherosclerosis, including through metabolites. The mapping of immune cell-mediated immune networks in the heart and gut provides us with new targets for treating CVD. This review discusses the role of immune cells in gut microbiota and cardiac communication during health and CVD.
Collapse
Affiliation(s)
- Hao Ren
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Botao Zhu
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Yuze An
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Feng Xie
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Yichuan Wang
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Yurong Tan
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China.
| |
Collapse
|
40
|
Guan X, Sun Z. The Role of Intestinal Flora and Its Metabolites in Heart Failure. Infect Drug Resist 2023; 16:51-64. [PMID: 36636378 PMCID: PMC9830706 DOI: 10.2147/idr.s390582] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/25/2022] [Indexed: 01/05/2023] Open
Abstract
Intestinal flora is a complex collection of microbial communities that participate in the physiological and pathological activities of the human body through various pathways. In recent years, numerous studies have reported that intestinal flora are involved in the occurrence and development of heart failure (HF) and its metabolic products could play an important role in this progression, suggesting a great value in the clinical treatment of this condition. This study reported the interaction between intestinal flora and HF, and with intestinal flora metabolites, such as short-chain fatty acids, trimethylamine N-oxide and bile acids and urotoxins, considered as the starting point, the mechanism of the roles in HF was summarized. Additionally, the current research status and the development prospects of applying flora and metabolites to the clinical therapeutic decision of HF were discussed.
Collapse
Affiliation(s)
- Xueqing Guan
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Zhijun Sun
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China,Correspondence: Zhijun Sun, Department of Cardiology, Shengjing Hospital, No. 39 of Huaxiang Road, Tiexi District, Shenyang, 110021, People’s Republic of China, Tel +86 18940251218, Fax +86 18940251218, Email
| |
Collapse
|
41
|
Liu S, Xing J, Zheng Z, Liu Z, Song F, Liu S. Effect of Qishen granules on isoproterenol-induced chronic heart failure in rats evaluated by comprehensive metabolomics. Phytother Res 2022; 36:4573-4586. [PMID: 35906729 DOI: 10.1002/ptr.7576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022]
Abstract
Qishen granules (QSG), a Chinese herbal formula, has been widely used in the treatment of myocardial ischemic chronic heart failure (CHF) for many years, but its mechanism of action is still unclear. In this study, comprehensive metabolomics was used to investigate the underlying protective mechanisms of QSG in an isoproterenol-induced CHF rat model. A total of 14 biomarkers were identified in serum and 34 biomarkers in urine, which were mainly related to fatty acid metabolism, bile acid metabolism, amino acid metabolism, purine metabolism, vitamin metabolism, and inflammation. Finally, 22 markers were selected for quantitative analysis of serum, urine, and fecal samples to verify the reliability of the results of untargeted metabolomics, and the results were similar to those of untargeted metabolomics. The correlation analysis showed that the targeted quantitative endogenous metabolites and CHF-related indexes were closely related. QSG might alleviate myocardial inflammatory response, oxidative stress, and amino acid metabolism disorder in CHF by regulating the level of endogenous metabolites. This study revealed QSG could regulate potential biomarkers and correlated metabolic pathway, which provided support for the further application of QSG.
Collapse
Affiliation(s)
- Shuxin Liu
- National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Junpeng Xing
- National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Zhong Zheng
- National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Zhiqiang Liu
- National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Fengrui Song
- National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Shu Liu
- National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| |
Collapse
|
42
|
Bhat MA, Mishra AK, Tantray JA, Alatawi HA, Saeed M, Rahman S, Jan AT. Gut Microbiota and Cardiovascular System: An Intricate Balance of Health and the Diseased State. Life (Basel) 2022; 12:1986. [PMID: 36556351 PMCID: PMC9780831 DOI: 10.3390/life12121986] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
Gut microbiota encompasses the resident microflora of the gut. Having an intricate relationship with the host, it plays an important role in regulating physiology and in the maintenance of balance between health and disease. Though dietary habits and the environment play a critical role in shaping the gut, an imbalance (referred to as dysbiosis) serves as a driving factor in the occurrence of different diseases, including cardiovascular disease (CVD). With risk factors of hypertension, diabetes, dyslipidemia, etc., CVD accounts for a large number of deaths among men (32%) and women (35%) worldwide. As gut microbiota is reported to have a direct influence on the risk factors associated with CVDs, this opens up new avenues in exploring the possible role of gut microbiota in regulating the gross physiological aspects along the gut-heart axis. The present study elaborates on different aspects of the gut microbiota and possible interaction with the host towards maintaining a balance between health and the occurrence of CVDs. As the gut microbiota makes regulatory checks for these risk factors, it has a possible role in shaping the gut and, as such, in decreasing the chances of the occurrence of CVDs. With special emphasis on the risk factors for CVDs, this paper includes information on the prominent bacterial species (Firmicutes, Bacteriodetes and others) towards an advance in our understanding of the etiology of CVDs and an exploration of the best possible therapeutic modules for implementation in the treatment of different CVDs along the gut-heart axis.
Collapse
Affiliation(s)
- Mujtaba Aamir Bhat
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Javeed Ahmad Tantray
- Department of Zoology, Central University of Kashmir, Ganderbal 191131, Jammu and Kashmir, India
| | - Hanan Ali Alatawi
- Department of Biological Sciences, University College of Haqel, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail 55476, Saudi Arabia
| | - Safikur Rahman
- Department of Botany, MS College, BR Ambedkar Bihar University, Muzaffarpur 842001, Bihar, India
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India
| |
Collapse
|
43
|
Liu H, Sun Y, Nie C, Xie X, Yuan X, Ma Q, Zhang M, Chen Z, Hu X, Li J. Highland barley β-glucan alleviated western diet-induced non-alcoholic fatty liver disease via increasing energy expenditure and regulating bile acid metabolism in mice. Food Funct 2022; 13:11664-11675. [PMID: 36278802 DOI: 10.1039/d2fo01167k] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become a public health burden. Controlling bile acids (BAs) metabolism and energy expenditure are potential therapies for NAFLD. Because one of the main health effects of cereal β-glucan (BG) is its ability to lower cholesterol by interacting with BAs, BG may regulate imbalances of the metabolism of BAs during NAFLD. Therefore, by using metabolic tests coupled with the profiling of hepatic BAs, we have assessed the effect of BG from highland barley on western diet (WD) induced NAFLD mice. BG treatment prevented fat accumulation and increased adipose lipolysis. These moderating effects were associated with an increased energy expenditure. Moreover, BG-treated mice enhanced the production of hepatic BAs, which may be connected with the activation of farnesoid X receptor (FXR) signaling in the liver and inhibition of FXR signaling in the ileum. Our results suggest that BG prevents fat accumulation by increasing energy expenditure, a mechanism associated with major changes in the composition of hepatic BAs.
Collapse
Affiliation(s)
- Huicui Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Yanli Sun
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Chenxi Nie
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Xiaoqing Xie
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Xiaojin Yuan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Qingyu Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Min Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Zhifei Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
| | - Xinzhong Hu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, People's Republic of China
| | - Juxiu Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, People's Republic of China.
- Engineering Research Center of Grain and Oil Functionalized Processing, Universities of Shaanxi Province, 712100, People's Republic of China
| |
Collapse
|
44
|
Qian B, Zhang K, Li Y, Sun K. Update on gut microbiota in cardiovascular diseases. Front Cell Infect Microbiol 2022; 12:1059349. [PMID: 36439214 PMCID: PMC9684171 DOI: 10.3389/fcimb.2022.1059349] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
In recent years, due to the development and widespread utilization of metagenomic sequencing and metabolomics, the relationship between gut microbiota and human cardiovascular diseases (CVDs) has received extensive attention. A growing number of studies have shown a strong relationship between gut microbiota and CVDs, such as coronary atherosclerosis, hypertension (HTN) and heart failure (HF). It has also been revealed that intestinal flora-related metabolites, such as trimethylamine-N-oxide (TMAO), short-chain fatty acids (SCFA) and bile acids (BAs), are also related to the development, prevention, treatment and prognosis of CVDs. In this review, we presented and summarized the recent findings on the relationship between gut microbiota and CVDs, and concluded several currently known gut microbiota-related metabolites and the occurrence and development of CVDs.
Collapse
Affiliation(s)
| | | | - Yuan Li
- *Correspondence: Kangyun Sun, ; Yuan Li,
| | | |
Collapse
|
45
|
Alka Ahuja, Saraswathy Mp, Nandakumar S, Prakash F A, Kn G, Um D. Role of the Gut Microbiome in Diabetes and Cardiovascular Diseases Including Restoration and Targeting Approaches- A Review. DRUG METABOLISM AND BIOANALYSIS LETTERS 2022; 15:133-149. [PMID: 36508273 DOI: 10.2174/2949681015666220615120300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022]
Abstract
Metabolic diseases, including cardiovascular diseases (CVD) and diabetes, have become the leading cause of morbidity and mortality worldwide. Gut microbiota appears to play a vital role in human disease and health, according to recent scientific reports. The gut microbiota plays an important role in sustaining host physiology and homeostasis by creating a cross-talk between the host and microbiome via metabolites obtained from the host's diet. Drug developers and clinicians rely heavily on therapies that target the microbiota in the management of metabolic diseases, and the gut microbiota is considered the biggest immune organ in the human body. They are highly associated with intestinal immunity and systemic metabolic disorders like CVD and diabetes and are reflected as potential therapeutic targets for the management of metabolic diseases. This review discusses the mechanism and interrelation between the gut microbiome and metabolic disorders. It also highlights the role of the gut microbiome and microbially derived metabolites in the pathophysiological effects related to CVD and diabetes. It also spotlights the reasons that lead to alterations of microbiota composition and the prominence of gut microbiota restoration and targeting approaches as effective treatment strategies in diabetes and CVD. Future research should focus onunderstanding the functional level of some specific microbial pathways that help maintain physiological homeostasis, multi-omics, and develop novel therapeutic strategies that intervene with the gut microbiome for the prevention of CVD and diabetes that contribute to a patient's well-being.
Collapse
Affiliation(s)
- Alka Ahuja
- College of Pharmacy, National University of Science and Technology, PC130, Muscat, Sultanate of Oman
| | - Saraswathy Mp
- Department of Microbiology, ESIC Medical College and PGIMSR, Chennai-600078, India
| | - Nandakumar S
- Department of Biotechnology, Pondicherry University, Kalapet, Puducherry-605014, India
| | - Arul Prakash F
- Centre of Molecular Medicine and Diagnostics (COMMAND), Saveetha Dental College and Hospital, Saveetha Institute of Medical & Technical Sciences, Chennai- 600077, India
| | - Gurpreet Kn
- College of Pharmacy, National University of Science and Technology, PC130, Muscat, Sultanate of Oman
| | - Dhanalekshmi Um
- College of Pharmacy, National University of Science and Technology, PC130, Muscat, Sultanate of Oman
| |
Collapse
|
46
|
Ferro F, Azzolin F, Spelat R, Bevilacqua L, Maglione M. Assessing the Efficacy of Whole-Body Titanium Dental Implant Surface Modifications in Inducing Adhesion, Proliferation, and Osteogenesis in Human Adipose Tissue Stem Cells. J Funct Biomater 2022; 13:jfb13040206. [PMID: 36412847 PMCID: PMC9680380 DOI: 10.3390/jfb13040206] [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: 09/27/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Although the influence of titanium implants' micro-surface properties on titanium discs has been extensively investigated, the research has not taken into consideration their whole-body effect, which may be considered possible using a combinatorial approach. METHODS Five titanium dental implants with a similar moderate roughness and different surface textures were thoroughly characterized. The cell adhesion and proliferation were assessed after adipose-tissue-derived stem cells (ADSCs) were seeded on whole-body implants. The implants' inductive properties were assessed by evaluating the osteoblastic gene expression. RESULTS The surface micro-topography was analyzed, showing that hydroxyapatite (HA)-blasted and bland acid etching implants had the highest roughness and a lower number of surface particles. Cell adhesion was observed after 24 h on all the implants, with the highest score registered for the HA-blasted and bland acid etching implants. Cell proliferation was observed only on the laser-treated and double-acid-etched surfaces. The ADSCs expressed collagen type I, osteonectin, and alkaline phosphatase on all the implant surfaces, with high levels on the HA-treated surfaces, which also triggered osteocalcin expression on day seven. CONCLUSIONS The findings of this study show that the morphology and treatment of whole titanium dental implants, primarily HA-treated and bland acid etching implants, impact the adherence and activity of ADSCs in osteogenic differentiation in the absence of specific osteo-inductive signals.
Collapse
Affiliation(s)
- Federico Ferro
- Department of Medical and Biological Sciences, University of Udine, 33100 Udine, Italy
- Correspondence:
| | - Federico Azzolin
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34125 Trieste, Italy
| | - Renza Spelat
- Neurobiology Sector, International School for Advanced Studies (SISSA), 34136 Trieste, Italy
| | - Lorenzo Bevilacqua
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34125 Trieste, Italy
| | - Michele Maglione
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34125 Trieste, Italy
| |
Collapse
|
47
|
Pan Z, Hu Y, Huang Z, Han N, Li Y, Zhuang X, Yin J, Peng H, Gao Q, Zhang W, Huang Y, Cui Y, Bi Y, Xu ZZ, Yang R. Alterations in gut microbiota and metabolites associated with altitude-induced cardiac hypertrophy in rats during hypobaric hypoxia challenge. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2093-2113. [PMID: 35301705 DOI: 10.1007/s11427-021-2056-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/04/2022] [Indexed: 02/08/2023]
Abstract
The gut microbiota is involved in host responses to high altitude. However, the dynamics of intestinal microecology and their association with altitude-related illness are poorly understood. Here, we used a rat model of hypobaric hypoxia challenge to mimic plateau exposure and monitored the gut microbiome, short-chain fatty acids (SCFAs), and bile acids (BAs) over 28 d. We identified weight loss, polycythemia, and pathological cardiac hypertrophy in hypoxic rats, accompanied by a large compositional shift in the gut microbiota, which is mainly driven by the bacterial families of Prevotellaceae, Porphyromonadaceae, and Streptococcaceae. The aberrant gut microbiota was characterized by increased abundance of the Parabacteroides, Alistipes, and Lactococcus genera and a larger Bacteroides to Prevotella ratio. Trans-omics analyses showed that the gut microbiome was significantly correlated with the metabolic abnormalities of SCFAs and BAs in feces, suggesting an interaction network remodeling of the microbiome-metabolome after the hypobaric hypoxia challenge. Interestingly, the transplantation of fecal microbiota significantly increased the diversity of the gut microbiota, partially inhibited the increased abundance of the Bacteroides and Alistipes genera, restored the decrease of plasma propionate, and moderately ameliorated cardiac hypertrophy in hypoxic rats. Our results provide an insight into the longitudinal changes in intestinal microecology during the hypobaric hypoxia challenge. Abnormalities in the gut microbiota and microbial metabolites contribute to the development of high-altitude heart disease in rats.
Collapse
Affiliation(s)
- Zhiyuan Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yichen Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Zongyu Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Ni Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jiye Yin
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Hui Peng
- Tianjin Institute of Environmental & Operational Medicine, Tianjin, 300050, China
| | - Quansheng Gao
- Tianjin Institute of Environmental & Operational Medicine, Tianjin, 300050, China
| | - Wenpeng Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yong Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| | - Zhenjiang Zech Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China. .,Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| |
Collapse
|
48
|
Abstract
The metabolism of bile acids (BAs) by gut bacteria plays an important role in human health. This study identified and characterized 7α-dehydroxylating bacteria, which are majorly responsible for converting primary BAs to secondary BAs, in the human gut and investigated their association with human disease. Six 7α-dehydratase (BaiE) clusters were identified from human gut metagenomes through sequence similarity network and genome neighborhood network analyses. Abundance analyses of gut metagenomes and metatranscriptomes identified a cluster of bacteria (cluster 1) harboring baiE genes that may be key 7α-dehydroxylating bacteria in the human gut. The baiE gene abundance of cluster 1 was significantly and positively correlated with the ratio of secondary BAs to primary BAs. Furthermore, the baiE gene abundances of cluster 1 were significantly negatively correlated with inflammatory bowel disease, including Crohn's disease and ulcerative colitis, as well as advanced nonalcoholic fatty liver disease, liver cirrhosis, and ankylosing spondylitis. Phylogenetic and metagenome-assembled genome analyses showed that the 7α-dehydroxylating bacterial clade of cluster 1 was affiliated with the family Oscillospiraceae and may demonstrate efficient BA dehydroxylation ability by harboring both a complete bai operon, for proteins which produce secondary BAs from primary BAs, and a gene for bile salt hydrolase, which deconjugates BAs, in the human gut. IMPORTANCE In this study, we identified a key 7α-dehydroxylating bacterial group predicted to be largely responsible for converting primary bile acids (BAs) to secondary BAs in the human gut through sequence similarity network, genome neighborhood network, and gene abundance analyses using human gut metagenomes. The key bacterial group was phylogenetically quite different from known 7α-dehydroxylating bacteria, and their abundance was highly correlated with the occurrence of diverse diseases associated with bile acid 7α-dehydroxylation. In addition, we characterized the metabolic features of the key bacterial group using their metagenome-assembled genomes. This approach is useful to identify and characterize key gut bacteria highly associated with human health and diseases.
Collapse
|
49
|
Lu X, Liu J, Zhou B, Wang S, Liu Z, Mei F, Luo J, Cui Y. Microbial metabolites and heart failure: Friends or enemies? Front Microbiol 2022; 13:956516. [PMID: 36046023 PMCID: PMC9420987 DOI: 10.3389/fmicb.2022.956516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/19/2022] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF), a global health issue characterized by structural or functional cardiac dysfunction, which was found to be associated with the gut microbiome recently. Although multiple studies suggested that the gut microbiome may have an impact on the development of cardiovascular diseases, the underlying mechanism of the gut microbiome in HF remains unclear. The study of metabolites from gut microbiota influenced by dietary nutrition uptake suggested that gut microbiota may affect the process of HF. However, on the basis of the microbiota’s complicated roles and their interactions with metabolites, studies of microbial metabolites in HF had rarely been described so far. In this review, we focused on dietary nutrition-related factors that were involved in the development and progression of HF, such as trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and bile acids (BAs), to summarize their advances and several potential targets in HF. From a therapeutic standpoint, we discussed microbial metabolites as a potential strategy and their applications in HF as well.
Collapse
Affiliation(s)
- Xiaofeng Lu
- Department of Cardiovascular Surgery, Heart Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jingjing Liu
- Department of Cardiovascular Surgery, Heart Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Bengbu Medical College, Bengbu, China
| | - Bing Zhou
- Department of Cardiovascular Surgery, Heart Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Shuwei Wang
- Department of Cardiovascular Surgery, Heart Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Zhifang Liu
- Department of Cardiovascular Surgery, Heart Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Fuyang Mei
- Department of Cardiovascular Surgery, Heart Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Junxiang Luo
- Department of Critical Care Medicine, Lishui Hospital of Traditional Chinese Medicine, Lishui, China
- Junxiang Luom,
| | - Yong Cui
- Department of Cardiovascular Surgery, Heart Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Yong Cui,
| |
Collapse
|
50
|
Ferro F, Spelat R, Valente C, Contessotto P. Understanding How Heart Metabolic Derangement Shows Differential Stage Specificity for Heart Failure with Preserved and Reduced Ejection Fraction. Biomolecules 2022; 12:biom12070969. [PMID: 35883525 PMCID: PMC9312956 DOI: 10.3390/biom12070969] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) is a clinical condition defined by structural and functional abnormalities in the heart that gradually result in reduced cardiac output (HFrEF) and/or increased cardiac pressures at rest and under stress (HFpEF). The presence of asymptomatic individuals hampers HF identification, resulting in delays in recognizing patients until heart dysfunction is manifested, thus increasing the chance of poor prognosis. Given the recent advances in metabolomics, in this review we dissect the main alterations occurring in the metabolic pathways behind the decrease in cardiac function caused by HF. Indeed, relevant preclinical and clinical research has been conducted on the metabolite connections and differences between HFpEF and HFrEF. Despite these promising results, it is crucial to note that, in addition to identifying single markers and reliable threshold levels within the healthy population, the introduction of composite panels would strongly help in the identification of those individuals with an increased HF risk. That said, additional research in the field is required to overcome the current drawbacks and shed light on the pathophysiological changes that lead to HF. Finally, greater collaborative data sharing, as well as standardization of procedures and approaches, would enhance this research field to fulfil its potential.
Collapse
Affiliation(s)
- Federico Ferro
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34125 Trieste, Italy
- Correspondence:
| | - Renza Spelat
- Neurobiology Sector, International School for Advanced Studies (SISSA), 34136 Trieste, Italy;
| | - Camilla Valente
- Department of Molecular Medicine, University of Padova, 35122 Padova, Italy; (C.V.); (P.C.)
| | - Paolo Contessotto
- Department of Molecular Medicine, University of Padova, 35122 Padova, Italy; (C.V.); (P.C.)
| |
Collapse
|