1
|
Li J, Wu Q, Ling X, Ma X, Gan X, Wei W, Du J, Zhou L, Jia X, Kan J, Zhao M. Unripe apple polyphenols extract improves intestinal inflammation and restructures gut microbiota in spontaneously hypertensive rats. Food Res Int 2025; 212:116418. [PMID: 40382036 DOI: 10.1016/j.foodres.2025.116418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/28/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025]
Abstract
Natural polyphenolic extracts have been recognized to reduce the risk of hypertension. Coupled with evidence that gut dysbiosis is tightly linked to the development of hypertension, we hypothesized that modulating gut microbiota may be associated with the benefits of unripe apple polyphenols extract (UAPE). This study aimed to explore the effects of UAPE on hypertension and its complications, while elucidating the underlying mechanisms in spontaneously hypertensive rats (SHR). SHR received either vehicle (ddH2O), captopril (30 mg/kg body weight/day), or low-dose (10 mg/kg body weight/day), middle-dose (50 mg/kg body weight/day), or high-dose (250 mg/kg body weight/day) UAPE by oral gavage daily for 8 weeks. Concurrently, Wistar-Kyoto (WKY) rats received vehicle to serve as normotensive controls. We observed that UAPE offered protective effects against hypertension-induced blood pressure elevation (systolic blood pressure, diastolic blood pressure), glycolipid metabolic disorders (serum lipids, glucose), and renal damage (serum creatinine, renal histopathology) in SHR. Additionally, UAPE exerted gut health benefits via enhancing intestinal barrier integrity (colonic and ileal histopathology, colonic tight junction protein 1 and Occludin mRNA and protein) and mitigating intestinal inflammation (colonic TNFα and IL-6 mRNA) in SHR. Moreover, UAPE effectively alleviated the development of left ventricular hypertrophy (cardiac histopathology, echocardiography) and endothelial dysfunction (serum endothelial nitric oxide synthase, endothelin-1), both critical markers of hypertensive progression. Mechanistically, the anti-inflammatory effects of UAPE may be linked to the colonic inhibition of the HMGB1-TLR4-NF-κB signaling pathway (mRNA and protein for colonic HMGB1, TLR4, and P-P65) in SHR. Notably, UAPE elevated microbial richness and diversity, normalizing the Firmicutes/Bacteroidetes ratio. Besides, UAPE increased the beneficial bacteria linked to healthy states, including Intestinimonas_butyriciproducens, Lactobacillus_intestinalis, Ruminiclostridium, Oscillibacter_sp., and Bifidobacterium, reduced the harmful bacteria related to hypertension, upregulated health-promoting microbial function, and elevated the concentrations of gut microbiota-derived short chain fatty acids, including acetic acid and butyric acid, in SHR. Collectively, these observations support the antihypertensive effects of UAPE in the SHR model, highlighting the intimate link between UAPE, gut microbiota, and hypertension. Our findings provide novel insights into the UAPE-mediated improvements in hypertension and its complications, which may be intricately linked to the modulation of the microbiota-gut axis.
Collapse
Affiliation(s)
- Juan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Qiming Wu
- Nutrilite Health Institute, Shanghai 201203, China.
| | - Xiaomeng Ling
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Xiaomin Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Center for Experimental Public Health and Preventive Medicine Education, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Xiaona Gan
- Nutrilite Health Institute, Shanghai 201203, China.
| | - Wei Wei
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze 274108, China; College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China..
| | - Jun Du
- Nutrilite Health Institute, Shanghai 201203, China.
| | - Leyan Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Xue Jia
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Juntao Kan
- Nutrilite Health Institute, Shanghai 201203, China.
| | - Min Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
2
|
Okunlola FO, Okunlola AR, Adetuyi BO, Soliman MES, Alexiou A, Papadakis M, Fawzy MN, El-Saber Batiha G. Beyond the gut: Unraveling the multifaceted influence of microbiome on cardiovascular health. Clin Nutr ESPEN 2025; 67:71-89. [PMID: 40064239 DOI: 10.1016/j.clnesp.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Cardiovascular disease is one of the leading causes of death worldwide. Even while receiving adequate pharmacological treatment for their hypertension, people are nonetheless at greater risk for cardiovascular disease. There is growing evidence that the gut microbiota may have major positive and negative effects on blood pressure and illnesses related with it as more study into this topic is conducted. Trimethylamine n-oxide (TMAO) and short-chain fatty acids (SCFA) are two major by-products of the gut microbiota. TMAO is involved in the formation of other coronary artery diseases, including atherosclerosis and hypertension, while SCFAs play an important role in controlling blood pressure. Numerous investigations have confirmed the established link between dietary salt intake and hypertension. Reducing sodium in the diet is linked to lower rates of cardiovascular disease morbidity and mortality as well as lower rates of blood pressure and hypertension. In both human and animal research, high salt diets increase local and systemic tissue inflammation and compromise gut architecture. Given that the gut microbiota constantly interacts with the immune system and is required for the correct maturation of immune cells, it is scientifically conceivable that it mediates the inflammatory response. This review highlights the therapeutic possibilities for focusing on intestinal microbiomes as well as the potential functions of the gut microbiota and its metabolites in the development of hypertension.
Collapse
Affiliation(s)
- Felix Oladele Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Abimbola Rafiat Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Babatunde Oluwafemi Adetuyi
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
3
|
Wen J, Li L, Ou D, Li J, Yang Y, Duan L, Zhang X, Zhu Y, Hao J, Tong Y. Higenamine protects against doxorubicin-induced heart failure by attenuating ferroptosis via modulating the Nrf2/GPX4 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156670. [PMID: 40220414 DOI: 10.1016/j.phymed.2025.156670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/24/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Higenamine (HG), a benzylisoquinoline alkaloid in Aconiti Lateralis Radix Praeparata (ALRP), has cardioprotective effects. Prior research indicated its potential anti-heart failure (HF) function, yet the molecular mechanism remained elusive. PURPOSE This study aimed to explore the underlying mechanism of HG against doxorubicin (DOX)-induced HF via an integrated approach involving gut microbiota, untargeted metabolomics, network pharmacology, and molecular biology. METHODS DOX was employed to induce HF in rats and H9c2 cardiomyocytes injury models. Cardiac injury was assessed using hemodynamic indices, cardiac injury biomarkers, and oxidative stress markers. Cell counting kit-8 (CCK-8) method and high-content analysis were used to investigate the effects of HG on the cell proliferation, morphology and mitochondrial function of H9c2 cardiomyocytes. 16S rDNA sequencing analysis, untargeted metabolomics, and network pharmacology were performed to identify the multi-target and multi-pathway mechanisms of HG in treating HF. Furthermore, reverse transcription quantitative polymerase chain reaction (RT-qPCR), immunohistochemistry, and Western Blotting was used to investigate its intervention on the nuclear factor erythroid 2-related factor 2 (Nrf2)/glutathione peroxidase 4 (GPX4) ferroptosis pathway. RESULTS HG alleviated DOX-mediated myocardial injury by enhancing cardiac and mitochondrial function, reducing oxidative stress levels, and promoting cell proliferation. Effects of HG on changes in the gut microbiota of rats is characterized by a low abundance of Firmicutes and Proteobacteria, along with a high abundance of Bacteroidetes and Actinobacteria, indicating an improvement in DOX-induced dysbiosis. Untargeted metabolomics combined with network pharmacology showed that HG exerted anti-HF effects by regulating eight metabolites, eight pathways, and interacting with ferroptosis-related targets. Molecular biology studies revealed its cardioprotective effects via regulating the Nrf2/GPX4 ferroptosis pathway. CONCLUSION HG could inhibit ferroptosis and protect against HF by regulating the Nrf2/GPX4-mediated "mitochondrial-ferroptosis" pathway, offering a potential treatment strategy for HF.
Collapse
Affiliation(s)
- Jianxia Wen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Lu Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Dinglin Ou
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Jianling Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yi Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Liting Duan
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming, China
| | - Xinghai Zhang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yichan Zhu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Junjie Hao
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming, China.
| | - Yuling Tong
- School of Medicine and Food, Sichuan Vocational College of Health and Rehabilitation, Zigong, China.
| |
Collapse
|
4
|
Gao ZK, Fan CY, Zhang BW, Geng JX, Han X, Xu DQ, Arshad M, Sun HX, Li JY, Jin X, Mu XQ. Cardiac function of colorectal cancer mice is remotely controlled by gut microbiota: regulating serum metabolites and myocardial cytokines. Anim Microbiome 2025; 7:53. [PMID: 40448218 PMCID: PMC12123981 DOI: 10.1186/s42523-025-00405-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/06/2025] [Indexed: 06/02/2025] Open
Abstract
Several studies have indicated that the dysregulation of microbial metabolites and the inflammatory environment resulting from microbial dysbiosis may contribute to the occurrence and progression of cardiovascular diseases. Therefore, restoring the disordered gut microbiota in patients with colorectal cancer by fecal microbiota transplantation (FMT) has the potential to reduce the incidence of cardiac disease. In this study, we identified cardiac dysfunction in azomethane and dextran sodium sulfate-induced colorectal cancer mice. Intestinal microbes from healthy mice were transferred to colorectal cancer mice, which vastly reversed the disorder of the gut microbiota and effectively alleviated cardiac dysfunction. Moreover, FMT regulated the expression of serum metabolites such as uridine triphosphate (UTP), tiamulin, andrographolide, and N-Acetyl-D-glucosamine, as well as cytokines like TGF-β, IRF5, and β-MHC in the heart. These findings uncover that the disturbed gut microbiota causes cardiac dysfunction in colorectal cancer mice by modulating the expression of serum metabolites and cytokines, which could be alleviated by treatment with FMT.
Collapse
Affiliation(s)
- Zhan-Kui Gao
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Chao-Yuan Fan
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Bo-Wen Zhang
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Jia-Xin Geng
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Xing Han
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Dan-Qi Xu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Muhammad Arshad
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China
| | - Hao-Xuan Sun
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jiong-Yi Li
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiangyuan Jin
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Xiao-Qin Mu
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, 150081, China.
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, 150081, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, China.
| |
Collapse
|
5
|
Zhou ZK, Yu MM, Shou ST, Chai YF, Liu YC. Interaction Between Gut-Heart Axis in Sepsis-Induced Cardiomyopathy. Pharmacol Res 2025; 217:107806. [PMID: 40449812 DOI: 10.1016/j.phrs.2025.107806] [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: 03/24/2025] [Revised: 05/11/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
The gut microbiota and its metabolites profoundly influence cardiac function, emerging as critical players in the pathophysiology of Sepsis-Induced Cardiomyopathy (SIC). Conversely, therapeutic interventions for SIC and the resultant cardiac alterations can reciprocally modulate gut microbial composition and function. To systematically elucidate this complex bidirectional relationship during SIC, this review delineates two key aspects: the 'forward gut-heart axis', defined as influences originating from the gut microbiota and its metabolites directed towards the cardiovascular system, and the 'reverse gut-heart axis', encompassing the reciprocal effects of cardiovascular drugs and cardiac factors on the gut microbiota. Furthermore, we explore potential therapeutic strategies for SIC centered on the targeted modulation of this intricate gut-heart interplay.
Collapse
Affiliation(s)
- Zi-Kang Zhou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Mu-Ming Yu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Song-Tao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yan-Fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Yan-Cun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
6
|
Fan L, Chen J, Zhang Q, Ren J, Chen Y, Yang J, Wang L, Guo Z, Bu P, Zhu B, Zhao Y, Wang Y, Liu X, Wang W, Chen Z, Gao Q, Zheng L, Cai J. Fecal microbiota transplantation for hypertension: an exploratory, multicenter, randomized, blinded, placebo-controlled trial. MICROBIOME 2025; 13:133. [PMID: 40410854 PMCID: PMC12100813 DOI: 10.1186/s40168-025-02118-6] [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] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/17/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND On the basis of the contribution of the gut microbiota to hypertension development, a novel strategy involving fecal microbiota transplantation (FMT) has been proposed to treat hypertension, but its efficacy has not been investigated in the clinic. METHODS In a randomized, blinded, placebo-controlled clinical trial (2021/03-2021/12, ClinicalTrials.gov, NCT04406129), hypertensive patients were recruited from seven centers in China, and received FMT or placebo capsules orally at three visits. The patients were randomized at a 1:1 ratio in blocks of four and stratified by center by an independent statistician. The intention-to-treat principle was implemented, as all randomized participants who received at least one intervention were included. The primary outcome was the decrease in office systolic blood pressure (SBP) from baseline to the day 30 visit. Adverse events (AEs) were recorded through the 3-month follow-up to assess safety measures. Alterations in BP, the fecal microbiome, and the plasma metabolome were assessed via exploratory analyses. RESULTS This study included 124 patients (mean age 43 years, 73.4% men) who received FMT (n = 63) or placebo (n = 61) capsules. The numbers of participants who experienced AEs (13 (20.6%) vs. 9 (14.8%), p = 0.39) and the primary outcome (6.28 (11.83) vs. 5.77 (10.06) mmHg, p = 0.62) were comparable between the groups. The FMT group presented a decrease in SBP after 1 week of FMT, with a between-arm difference of - 4.34 (95% CI, - 8.1 to - 0.58; p = 0.024) mmHg, but this difference did not persist even after repeated intervention. After FMT, shifts in microbial richness and structure were identified and the abundance of the phyla Firmicutes and Bacteroidetes was altered. Decreases in the abundances of Eggerthella lenta, Erysipelatoclostridium ramosum, Anaerostipes hadrus, Gemella haemolysans, and Streptococcus vestibularis and increases in the abundances of Parabacteroides merdae, Prevotella copri, Bacteroides galacturonicus, Eubacterium sp. CAG 180, Desulfovibrio piger, Megamonas hypermegale, Collinsella stercoris, Coprococcus catus, and Allisonella histaminiformans were identified and correlated with office SBP. Those species were also correlated with responding and inversely office SBP-associated metabolites including tyrosine, glutamine, aspartate, phenylalanine, methionine, serine, sarcosine, and/or asparagine. CONCLUSIONS Safety but unsustainable BP reduction was observed in the first trial of the effects of FMT on hypertension. Additional intervention studies on specific microbes with metabolite-targeting and BP-modulating features are needed. Video Abstract.
Collapse
Affiliation(s)
- Luyun Fan
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Chinese Institutes for Medical Research, Beijing, 100029, China
| | - Junru Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Qi Zhang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing, 100191, China
| | - Jie Ren
- Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Youren Chen
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515000, Guangdong, China
| | - Jinfeng Yang
- The People's Hospital of Ji Xian District, Tianjin, 301900, China
| | - Lu Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Chinese Institutes for Medical Research, Beijing, 100029, China
| | - Zihong Guo
- Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, China
| | - Peili Bu
- Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Bingpo Zhu
- Southern University of Science and Technology Hospital, Shenzhen, China
| | - Yanyan Zhao
- Medical Research & Biometrics Center, National Center for Cardiovascular Dieases, Fuwai Hospital Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Yang Wang
- Medical Research & Biometrics Center, National Center for Cardiovascular Dieases, Fuwai Hospital Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Xiaoyan Liu
- Department of Cardiology, Heart Center, Beijing, Key Laboratory of Hypertension Research, Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Wenjie Wang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zhenzhen Chen
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Chinese Institutes for Medical Research, Beijing, 100029, China
| | - Qiannan Gao
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Lemin Zheng
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China.
| | - Jun Cai
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Chinese Institutes for Medical Research, Beijing, 100029, China.
| |
Collapse
|
7
|
Sen I, Trzaskalski NA, Hsiao YT, Liu PP, Shimizu I, Derumeaux GA. Aging at the Crossroads of Organ Interactions: Implications for the Heart. Circ Res 2025; 136:1286-1305. [PMID: 40403108 DOI: 10.1161/circresaha.125.325637] [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: 02/12/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 05/24/2025]
Abstract
Aging processes underlie common chronic cardiometabolic diseases such as heart failure and diabetes. Cross-organ/tissue interactions can accelerate aging through cellular senescence, tissue wasting, accelerated atherosclerosis, increased vascular stiffness, and reduction in blood flow, leading to organ remodeling and premature failure. This interorgan/tissue crosstalk can accelerate aging-related dysfunction through inflammation, senescence-associated secretome, and metabolic and mitochondrial changes resulting in increased oxidative stress, microvascular dysfunction, cellular reprogramming, and tissue fibrosis. This may also underscore the rising incidence and co-occurrence of multiorgan dysfunction in cardiometabolic aging in the population. Examples include interactions between the heart and the lungs, kidneys, liver, muscles, and brain, among others. However, this phenomenon can also present new translational opportunities for identifying diagnostic biomarkers to define early risks of multiorgan dysfunction, gain mechanistic insights, and help to design precision-directed therapeutic interventions. Indeed, this opens new opportunities for therapeutic development in targeting multiple organs simultaneously to disrupt the crosstalk-driven process of mutual disease acceleration. New therapeutic targets could provide synergistic benefits across multiple organ systems in the same at-risk patient. Ultimately, these approaches may together slow the aging process itself throughout the body. In the future, with patient-centered multisystem coordinated approaches, we can initiate a new paradigm of multiorgan early risk prediction and tailored intervention. With emerging tools including artificial intelligence-assisted risk profiling and novel preventive strategies (eg, RNA-based therapeutics), we may be able to mitigate multiorgan cardiometabolic dysfunction much earlier and, perhaps, even slow the aging process itself.
Collapse
Affiliation(s)
- Ilke Sen
- Department of Physiology, INSERM U955 (Institut national de la santé et de la recherche médicale, Unité 955), Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Fédération Hospitalo-Universitaire (FHU SENCODE), Ecole Universitaire de Recherche LIVE (EUR LIVE), Université Paris-Est Créteil, France (I. Sen, G.A.D.)
| | - Natasha A Trzaskalski
- University of Ottawa Heart Institute, Brain-Heart Interconnectome, University of Ottawa, Ontario, Canada (N.A.T., P.P.L.)
| | - Yung-Ting Hsiao
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (Y.-T.H., I. Shimizu)
| | - Peter P Liu
- University of Ottawa Heart Institute, Brain-Heart Interconnectome, University of Ottawa, Ontario, Canada (N.A.T., P.P.L.)
| | - Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (Y.-T.H., I. Shimizu)
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (I. Shimizu)
| | - Geneviève A Derumeaux
- Department of Physiology, INSERM U955 (Institut national de la santé et de la recherche médicale, Unité 955), Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Fédération Hospitalo-Universitaire (FHU SENCODE), Ecole Universitaire de Recherche LIVE (EUR LIVE), Université Paris-Est Créteil, France (I. Sen, G.A.D.)
| |
Collapse
|
8
|
Snelson M, Muralitharan RR, Liu CF, Markó L, Forslund SK, Marques FZ, Tang WHW. Gut-Heart Axis: The Role of Gut Microbiota and Metabolites in Heart Failure. Circ Res 2025; 136:1382-1406. [PMID: 40403109 PMCID: PMC12101525 DOI: 10.1161/circresaha.125.325516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 05/24/2025]
Abstract
Heart failure is a global health issue with significant mortality and morbidity. There is increasing evidence that alterations in the gastrointestinal microbiome, gut epithelial permeability, and gastrointestinal disorders contribute to heart failure progression through various pathways, including systemic inflammation, metabolic dysregulation, and modulation of cardiac function. Moreover, several medications used to treat heart failure directly impact the microbiome. The relationship between the gastrointestinal tract and the heart is bidirectional, termed the gut-heart axis. It is increasingly understood that diet-derived microbial metabolites are key mechanistic drivers of the gut-heart axis. This includes, for example, trimethylamine N-oxide and short-chain fatty acids. This review discusses current insights into the interplay between heart failure, its associated risk factors, and the gut microbiome, focusing on key metabolic pathways, the role of dietary interventions, and the potential for gut-targeted therapies. Understanding these complex interactions could pave the way for novel strategies to mitigate heart failure progression and improve patient outcomes.
Collapse
Affiliation(s)
- Matthew Snelson
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Rikeish R. Muralitharan
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Chia-Feng Liu
- Center for Microbiome and Human Health, 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, Cleveland OH, USA
| | - Lajos Markó
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
| | - Sofia K. Forslund
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Francine Z. Marques
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - W. H. Wilson Tang
- Center for Microbiome and Human Health, 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, Cleveland OH, USA
| |
Collapse
|
9
|
Yamashita T. The role of gut microbiota in cardiovascular diseases and their potential as novel therapeutic targets. J Cardiol 2025:S0914-5087(25)00126-1. [PMID: 40409712 DOI: 10.1016/j.jjcc.2025.05.002] [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: 01/19/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/25/2025]
Abstract
Cardiovascular diseases (CVDs) including heart failure (HF) is a major health, medical and social issue that needs to be resolved in Japan's super-aged society. Recent clinical and basic studies suggest that the gut microbiota and their metabolites play critical roles in the onset and progression of CVDs. We explored changes in gut microbiota composition and metabolite levels among Japanese patients to investigate their association with CVDs. Changes in specific bacteria were observed, with a decrease in phylum Bacteroidetes and increases in order Lactobacillus or genus Streptococcus in coronary artery disease patients. For HF patients, a reduction in phylum Bacteroidetes and increases in phylum Actinobacteria (e.g. Bifidobacterium) and Proteobacteria (e.g. Escherichia, Shigella, and Klebsiella) were noted. Elevated levels of gut microbiota-associated metabolites, such as trimethylamine N-oxide (TMAO) and indoxyl sulfate, were observed in CVD patients, suggesting potential effects on organ functions. Many studies have linked higher plasma TMAO levels to worse prognoses in CVDs, including HF and renal failure. However, the clinical significance and therapeutic potential of these findings require further investigation. In this manuscript, the author aims to review the current status of research on gut microbiota in CVDs, with a primary focus on the microbes themselves and their related metabolites. Further research is essential to comprehensively understand these intricacies and establish clear cause-and-effect relationships, ultimately paving the way for the development of innovative therapies for CVDs.
Collapse
Affiliation(s)
- Tomoya Yamashita
- Division of Advanced Medical and Pharmaceutical Sciences, Graduate School of Science, Technology and Innovation, Kobe University, Japan.
| |
Collapse
|
10
|
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 2025; 41:1099-1115. [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
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Medical University
| | | | - Duo Luo
- Guangzhou Medical University
| | | | | | | | | |
Collapse
|
11
|
Mukhopadhya I, Louis P. Gut microbiota-derived short-chain fatty acids and their role in human health and disease. Nat Rev Microbiol 2025:10.1038/s41579-025-01183-w. [PMID: 40360779 DOI: 10.1038/s41579-025-01183-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/15/2025]
Abstract
Short-chain fatty acids (SCFAs) are a group of organic compounds produced by the fermentation of dietary fibre by the human gut microbiota. They play diverse roles in different physiological processes of the host with implications for human health and disease. This Review provides an overview of the complex microbial metabolism underlying SCFA formation, considering microbial interactions and modulating factors of the gut environment. We explore the multifaceted mechanistic interactions between SCFAs and the host, with a particular focus on the local actions of SCFAs in the gut and their complex interactions with the immune system. We also discuss how these actions influence intestinal and extraintestinal diseases and emerging therapeutic strategies using SCFAs.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Petra Louis
- Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
12
|
Sousa A, Oliveira N, Conde R, Morais E, Amaral AP, Embade N, Millet O, Verde I. Nuclear Magnetic Resonance Analysis Seeking for Metabolic Markers of Hypertension in Human Serum. Molecules 2025; 30:2145. [PMID: 40430317 PMCID: PMC12113710 DOI: 10.3390/molecules30102145] [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: 04/01/2025] [Revised: 05/08/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Hypertension is a highly prevalent medical condition that occurs when blood pressure is too high, which greatly increases the risk of developing other cardiovascular diseases and is generally associated with higher rates of morbidity and mortality. Due to the silent/asymptomatic nature of hypertension, although the methods currently available to diagnose it are easy, they generally do not allow for an early diagnosis and an efficient prognosis to avoid irreversible damage in the medium or long term. In fact, an early diagnosis of hypertension would be crucial to decrease hypertension-associated mortality. Since metabolomics using NMR can provide a global measurement of various serum metabolites, it is very suitable for detecting novel biomarkers. We therefore analyzed serum metabolomic profiles among normotensive and hypertensive elderly individuals by NMR and identified new potential biomarkers for hypertension and associated diseases. We found higher levels of acetate, formate, and glycerol, and lower levels of glutamine, glycine, and sarcosine in individuals with hypertension. Therefore, these metabolites could be used for early diagnosis of hypertension to avoid comorbidities derived from hypertension and associated mortality.
Collapse
Affiliation(s)
- Adriana Sousa
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
- RISE-Health, Faculty of Health Sciences, University of Beira Interior (UBI), Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Nádia Oliveira
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
- RISE-Health, Faculty of Health Sciences, University of Beira Interior (UBI), Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ricardo Conde
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
- Center for Cooperative Research in Biosciences (CIC-BioGUNE), Bizkaia Science and Technology Park, 48160 Derio, Spain; (N.E.); (O.M.)
| | - Elisabete Morais
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
| | - Ana Paula Amaral
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
| | - Nieves Embade
- Center for Cooperative Research in Biosciences (CIC-BioGUNE), Bizkaia Science and Technology Park, 48160 Derio, Spain; (N.E.); (O.M.)
| | - Oscar Millet
- Center for Cooperative Research in Biosciences (CIC-BioGUNE), Bizkaia Science and Technology Park, 48160 Derio, Spain; (N.E.); (O.M.)
| | - Ignacio Verde
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior (UBI), 6200-506 Covilhã, Portugal; (A.S.); (N.O.); (R.C.); (E.M.); (A.P.A.)
- RISE-Health, Faculty of Health Sciences, University of Beira Interior (UBI), Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
13
|
Saeedi Saravi SS, Pugin B, Constancias F, Shabanian K, Spalinger M, Thomas A, Le Gludic S, Shabanian T, Karsai G, Colucci M, Menni C, Attaye I, Zhang X, Allemann MS, Lee P, Visconti A, Falchi M, Alimonti A, Ruschitzka F, Paneni F, Beer JH. Gut microbiota-dependent increase in phenylacetic acid induces endothelial cell senescence during aging. NATURE AGING 2025:10.1038/s43587-025-00864-8. [PMID: 40355758 DOI: 10.1038/s43587-025-00864-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/02/2025] [Indexed: 05/15/2025]
Abstract
Endothelial cell senescence is a key driver of cardiovascular aging, yet little is known about the mechanisms by which it is induced in vivo. Here we show that the gut bacterial metabolite phenylacetic acid (PAA) and its byproduct, phenylacetylglutamine (PAGln), are elevated in aged humans and mice. Metagenomic analyses reveal an age-related increase in PAA-producing microbial pathways, positively linked to the bacterium Clostridium sp. ASF356 (Clos). We demonstrate that colonization of young mice with Clos increases blood PAA levels and induces endothelial senescence and angiogenic incompetence. Mechanistically, we find that PAA triggers senescence through mitochondrial H2O2 production, exacerbating the senescence-associated secretory phenotype. By contrast, we demonstrate that fecal acetate levels are reduced with age, compromising its function as a Sirt1-dependent senomorphic, regulating proinflammatory secretion and redox homeostasis. These findings define PAA as a mediator of gut-vascular crosstalk in aging and identify sodium acetate as a potential microbiome-based senotherapy to promote healthy aging.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland.
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland.
| | - Benoit Pugin
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Florentin Constancias
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Khatereh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Marianne Spalinger
- Department for Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Aurélien Thomas
- Faculty Unit of Toxicology, University Center of Legal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, University Center of Legal Medicine, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne, Geneva, Switzerland
| | - Sylvain Le Gludic
- Faculty Unit of Toxicology, University Center of Legal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, University Center of Legal Medicine, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne, Geneva, Switzerland
| | - Taraneh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland
| | - Manuel Colucci
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera Italiana, Lugano, Switzerland
| | - Cristina Menni
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Ilias Attaye
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
- Amsterdam Cardiovascular Sciences, Diabetes & Metabolism, Amsterdam, Netherlands
| | - Xinyuan Zhang
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
| | - Meret Sarah Allemann
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Pratintip Lee
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Alessia Visconti
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinial and Biological Sciences, University of Turin, Turin, Italy
| | - Mario Falchi
- Department of Twin Research, King's College London, St Thomas' Hospital Campus, London, UK
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medicine, University of Padova, Padova, Italy
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Zurich, Switzerland
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.
- Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland.
| |
Collapse
|
14
|
Zhao Z, Xiang L, Hong JS, Wang Y, Feng J. Mechanisms of Acetate in Alleviating SETDB1-Linked Neuroinflammation and Cognitive Impairment in a Mouse Model of OSA. J Inflamm Res 2025; 18:5931-5950. [PMID: 40357375 PMCID: PMC12067661 DOI: 10.2147/jir.s510690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Background Microglia-mediated neuroinflammation is crucial for obstructive sleep apnea (OSA)-induced cognitive impairment. We aimed to investigate roles of acetate (ACE) and SET domain bifurcated histone lysine methyltransferase 1 (SETDB1) in neuroinflammation of OSA. Methods After C57BL/6J mice were exposed to OSA-associated intermittent hypoxia (IH) or normoxia for four weeks, the composition of the gut microbiota (GM) and the levels of serum short-chain fatty acids (SCFAs) were measured by 16S rRNA and GC-MS methods, respectively. To assess the effect of ACE on IH mice, glyceryl triacetate (GTA) was gavaged in IH-exposed mice and the cognitive function, microglial activation, and hippocampal neuronal death were examined. Moreover, ACE-treated BV2 microglia cells were also utilized for further mechanistic studies. Results IH disrupts the gut microbiome, reduces microbiota-SCFAs, and impairs cognitive function. Gavage with GTA significantly mitigated these cognitive deficits. Following IH exposure, we observed substantial increases in SETDB1 both in vivo and in vitro, along with elevated levels of histone H3 lysine 9 trimethylation (H3K9me3). Genetic or pharmacological inhibition of SETDB1 in microglia led to decreased induction of proinflammatory factors, as well as reduced reactive oxygen species (ROS) generation. Mechanistically, SETDB1 was found to upregulate the transcription factors p-signal transducer and activator of transcription 3 (p-STAT3) and p-NF-κB. In vitro, ACE supplementation effectively repressed high SETDB1 and H3K9me3 levels, thereby inhibiting microglial pro-inflammatory responses induced by IH. In vivo, ACE supplementation significantly reduced hippocampal levels of p-STAT3, p-NF-κB, and pro-inflammatory cytokines while also protecting neuronal integrity. Conclusion This study provides the first evidence that H3K9 methyltransferase SETDB1 promotes microglial pro-inflammatory response distinct from its previously shown role in macrophages. Our findings also identify ACE supplementation as a promising dietary intervention for OSA-related cognitive impairment with SETDB1 serving as both a mechanistic biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Zhan Zhao
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Li Xiang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Yubao Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| |
Collapse
|
15
|
Pan Y, Chen B, Xie J, Chen D, Cai Y, Zhao D, Cao Y, Lian F, Yan X. Lentinan alleviates angiotensin II-induced myocardial remodeling through LMP7-SOCS3 signaling. Int J Biol Macromol 2025; 308:142146. [PMID: 40101827 DOI: 10.1016/j.ijbiomac.2025.142146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Myocardial remodeling is a major pathological mechanism causing heart failure. As a critical negative modulator of cardiac remodeling, suppressor of cytokine signaling 3 (SOCS3) is regulated by immunoproteasome subunit large multifunctional peptidase 7 (LMP7). Lentinan (LNT), a β-polysaccharide extracted from Lentinus edodes, has anti-inflammatory and antioxidant properties. However, the role and molecular mechanisms of LNT in angiotensin II (Ang II)-triggered myocardial remodeling are unclear. Myocardial remodeling was established using Ang II infusion (1000 or 200 ng/kg/min) for 2 weeks. Cardiomyocytes and cardiac fibroblasts were triggered by Ang II. LNT was administered daily by oral gavage to mice starting 1 day before Ang II or saline treatment. Here, we found that LNT supplementation dose-dependently ameliorated Ang II-triggered myocardial dysfunction and remodeling (hypertrophy, fibrosis, inflammation, and superoxide production). Mechanistically, LNT suppressed SOCS3 protein degradation by downregulating immunoproteasome LMP7 activity and expression, thereby inactivating downstream signaling, such as STAT3, ERK, AKT, NF-κB, and TGF-β. Conversely, SOCS3 knockdown significantly blocked the protective effect of LNT on myocardial remodeling in Ang II-infused mice. Together, our findings suggest that LNT may be a new therapeutic approach for myocardial remodeling and heart failure.
Collapse
Affiliation(s)
- Yu Pan
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Bingqi Chen
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Jiawen Xie
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Danni Chen
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Yuwei Cai
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Denghui Zhao
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Yifei Cao
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Fuzhi Lian
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China; Engineering Research Center of Mobile Health Management System, Ministry of Education, Hangzhou, China
| | - Xiao Yan
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou 311121, China.
| |
Collapse
|
16
|
Barrea L, Caprio M, Perrini S, Camajani E, Crafa A, Gangitano E, Gorini S, Sciacca L, Verde L, Albertelli M, Ferone D, Giorgino F, Colao A, Aimaretti G, Muscogiuri G. Diabetes mellitus secondary to endocrine diseases: a position statement of the working group of the club of the Italian society of endocrinology (SIE)-Nutrition hormones and metabolism. J Endocrinol Invest 2025:10.1007/s40618-025-02589-2. [PMID: 40293649 DOI: 10.1007/s40618-025-02589-2] [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: 01/04/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE This position statement addressed the limited scientific literature on the management of diabetes mellitus secondary to endocrinopathies, despite its frequent occurrence in hormonal diseases such as acromegaly, Cushing's syndrome, primary hyperaldosteronism, pheochromocytoma, hyperthyroidism, and neuroendocrine tumors. The aim was to review the pathophysiological mechanisms, clinical features, and management strategies, focusing on nutritional and pharmacological approaches. METHODS A comprehensive review of existing literature was conducted regarding studies on diabetes secondary to endocrinopathies and the effects of treatments for these conditions, such as somatostatin analogues and pancreatic surgery. Particular emphasis was placed on understanding glucose metabolism derangements and the interplay between endocrine excess and therapeutic interventions. RESULTS Secondary diabetes arises not only from hormone excess but also as a consequence of treatments for endocrine disorders. For instance, somatostatin analogues, while effective in resolving hormone hypersecretion, impair glucose metabolism by inhibiting pancreatic insulin secretion. Similarly, pancreatic surgery for neuroendocrine tumors often exacerbates glycemic disturbances. The management of secondary diabetes requires a multidisciplinary approach that includes treating the underlying endocrine disorder, tailoring antidiabetic therapy, and optimizing nutritional strategies to mitigate metabolic disruptions. CONCLUSION Diabetes secondary to endocrinopathies presents unique challenges due to its complex etiology and the metabolic effects of treatments. This position statement underscores the importance of an integrated management approach, offering guidance for clinicians in addressing this multifaceted condition. Further research is needed to develop evidence-based guidelines for optimal care.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Psicologia e Scienze della Salute, Università Telematica Pegaso, Centro Direzionale Isola F2, Via Porzio, 80143, Naples, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Rome, Italy
| | - Sebastio Perrini
- Section of Endocrinology, Department of Medicine and Surgery, LUM University, Casamassima, BA, Italy
| | - Elisabetta Camajani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Rome, Italy
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, 95123, Italy
| | - Elena Gangitano
- Department of Experimental Medicine, Sapienza University of Rome, Rome, 00185, Italy
| | - Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Rome, Italy
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, University of Catania, Catania, 95123, Italy
| | - Ludovica Verde
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
| | - Manuela Albertelli
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, Genova, Italy
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Diego Ferone
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, Genova, Italy
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, 70124, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia ed Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Università degli Studi di Napoli Federico II, Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", Via Sergio Pansini 5, Naples, 80131, Italy
| | - Gianluca Aimaretti
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA.
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia ed Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.
- Università degli Studi di Napoli Federico II, Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", Via Sergio Pansini 5, Naples, 80131, Italy.
| |
Collapse
|
17
|
Mahgoup EM. "Gut Microbiota as a Therapeutic Target for Hypertension: Challenges and Insights for Future Clinical Applications" "Gut Microbiota and Hypertension Therapy". Curr Hypertens Rep 2025; 27:14. [PMID: 40261509 DOI: 10.1007/s11906-025-01331-w] [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] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW Systemic hypertension is a major risk factor for cardiovascular disease and remains challenging to manage despite the widespread use of antihypertensive medications and lifestyle modifications. This review explores the role of gut microbiota in hypertension development and regulation, highlighting key mechanisms such as inflammation, gut-brain axis modulation, and bioactive metabolite production. We also assess the potential of microbiota-targeted therapies for hypertension management. RECENT FINDINGS Emerging evidence indicates that microbial dysbiosis, high-salt diets, and gut-derived metabolites such as short-chain fatty acids (SCFAs) and bile acids significantly influence blood pressure regulation. Preclinical and early clinical studies suggest that interventions targeting gut microbiota, including probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), and dietary modifications, may help modulate hypertension. However, variability in gut microbiota composition among individuals and limited human trial data pose challenges to translating these findings into clinical practice. While microbiota-based therapies show promise for hypertension management, further research is needed to establish their efficacy and long-term effects. Large-scale, standardized clinical trials are crucial for understanding the therapeutic potential and limitations of gut microbiota interventions. A deeper understanding of the gut-hypertension axis could lead to novel, personalized treatment strategies for hypertension.
Collapse
Affiliation(s)
- Elsayed M Mahgoup
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt.
- Department of Internal Medicine, Division of Cardiovascular Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
18
|
Su X, Tian Z, Fang Y, Zhou S, Ma S. Effects of high-dose glucocorticoids on gut microbiota in the treatment of Graves' ophthalmopathy. Microbiol Spectr 2025:e0246724. [PMID: 40261021 DOI: 10.1128/spectrum.02467-24] [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: 09/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
Many studies indicate the gut microbiome is associated with diseases caused by administering high-dose glucocorticoids (GCs), such as hypertension, hyperglycemia, and osteoporosis. However, the association between intestinal flora and the use of high-dose GCs remains elusive. We aimed to characterize gut microbiome in Graves' ophthalmopathy (GO) patients after administering high-dose GCs. In this study, 20 primary GO patients were recruited. The differences in gut microbiota of GO patients before and after administering high-dose GCs were analyzed by 16S rDNA sequencing technology. Untargeted metabolomic analysis was used to examine the differences in gut metabolites between two groups. There were significant differences in α and β diversities of gut microbiota in GO patients before and after administering high-dose GCs. The random forest analysis indicated that three intestinal bacteria (Faecalibacterium, Streptococcus, and Prevotella) could distinguish the two groups with the highest accuracy, which was proven by receiver operator characteristic curve and linear discriminant analysis effect size analysis. The short-chain fatty acid-producing ability in GO patients' gut after high-dose GC administration was significantly decreased. The 5-hydroxytryptamine levels significantly increased in the gut of GO patients after administering high-dose GCs. Our study suggests that high-dose GC administration causes the changes in gut microbiome and metabolites. Moreover, the altered flora and metabolites are related to hypertension, hyperglycemia, and osteoporosis. These findings can help understand the development of side effects caused by high-dose GCs and can be further used to develop potential probiotics to facilitate the prevention for those side effects.IMPORTANCEFor the first time, we revealed that gut microbiome and metabolome in Graves' ophthalmopathy patients after high-dose glucocorticoid (GC) administration significantly changed, and the altered flora and metabolites are related to hypertension, hyperglycemia, and osteoporosis. These findings can help understand the development of side effects caused by high-dose GCs and can be further used to develop potential probiotics to facilitate the prevention for those side effects.
Collapse
Affiliation(s)
- Xinhuan Su
- Department of Endocrinology, Department of Geriatrics, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhenyu Tian
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yalun Fang
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Shengnan Zhou
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shizhan Ma
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
19
|
Liu W, Wang L, Ou J, Peng D, Zhang Y, Chen W, Wang Y. Gut Microbiota Metabolites and Chronic Diseases: Interactions, Mechanisms, and Therapeutic Strategies. Int J Mol Sci 2025; 26:3752. [PMID: 40332366 PMCID: PMC12027615 DOI: 10.3390/ijms26083752] [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: 03/04/2025] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
The gut microbiota, shaped by factors such as diet, lifestyle, and genetics, plays a pivotal role in regulating host metabolism, immune function, and overall health. The diversity and balance of the gut microbiota are closely linked to the onset and progression of various chronic diseases. A growing body of evidence has demonstrated that alterations in the composition, function, and metabolites of the gut microbiota are significantly associated with cardiovascular diseases, including hypertension, atherosclerosis, and heart failure; metabolic disorders such as obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease; and gastrointestinal conditions like inflammatory bowel disease and colorectal cancer. Despite substantial advances in microbiome research, challenges remain in fully elucidating the causal relationships between the gut microbiota and disease, as well as in translating these insights into clinical applications. This review aims to investigate the regulatory pathways via which the gut microbiota affects cardiovascular health, metabolic function, and gastrointestinal disease. Additionally, it highlights emerging strategies for the prevention and treatment of these chronic conditions, focusing on microbiota-targeted therapies and personalized dietary interventions as promising approaches for improving health outcomes.
Collapse
Affiliation(s)
- Wenwen Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Hefei 230012, China
| | - Jinmei Ou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Anhui Key Laboratory of New Manufacturing Technology of Chinese Medicine Pieces, Hefei 230012, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Hefei 230012, China
| | - Yue Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Hefei 230012, China
- Anhui Key Laboratory of New Manufacturing Technology of Chinese Medicine Pieces, Hefei 230012, China
| | - Yanyan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Hefei 230012, China
| |
Collapse
|
20
|
Boicean A, Ichim C, Sasu SM, Todor SB. Key Insights into Gut Alterations in Metabolic Syndrome. J Clin Med 2025; 14:2678. [PMID: 40283508 PMCID: PMC12028006 DOI: 10.3390/jcm14082678] [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: 03/20/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Over time, extensive research has underscored the pivotal role of gut microbiota in the onset and progression of various diseases, with a particular focus on fecal microbiota transplantation (FMT) as a potential therapeutic approach. The practice of transferring fecal matter from a healthy donor to a patient provides valuable insights into how alterations in gut microbiota can impact disease development and how rectifying dysbiosis may offer therapeutic benefits. Re-establishing a balanced symbiotic relationship in the gastrointestinal tract has shown positive results in managing both intestinal and systemic conditions. Currently, one of the most pressing global health issues is metabolic syndrome-a cluster of conditions that includes insulin resistance, lipid imbalances, central obesity and hypertension. In this context, FMT has emerged as a promising strategy for addressing key components of metabolic syndrome, such as improving insulin sensitivity, body weight and lipid profiles. However, further well-structured studies are needed to refine treatment protocols and establish the long-term safety and efficacy of this intervention.
Collapse
Affiliation(s)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.)
| | - Sabina-Maria Sasu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.)
| | | |
Collapse
|
21
|
Graça ICR, Martins C, Ribeiro F, Nunes A. Understanding Hypertension: A Metabolomic Perspective. BIOLOGY 2025; 14:403. [PMID: 40282268 PMCID: PMC12025236 DOI: 10.3390/biology14040403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Metabolomics approaches, such as Fourier transform infrared (FTIR) spectroscopy, nuclear magnetic resonance (NMR) spectroscopy, and mass spectrometry (MS), have emerged as powerful tools for studying cardiovascular diseases (CVD), including hypertension. The use of biological fluids, like plasma and serum, has garnered significant interest due to their accessibility and potential in elucidating disease mechanisms. This review aims to summarize the current literature on the application of metabolomics techniques (FTIR, NMR, and MS) in the study of hypertension, focusing on their contributions to understanding disease pathophysiology, biomarker discovery, and therapeutic advancements. A comprehensive analysis of metabolomic studies was performed, with a particular emphasis on the diversity of altered metabolites associated with systolic blood pressure (SBP), diastolic blood pressure (DBP), and sex-related differences. Metabolomics techniques, including FTIR, NMR, and MS, provide comprehensive insights into the biochemical alterations underlying hypertension, such as amino acid and fatty acid metabolism impairment or inflammation and oxidative stress processes. This review underscores their role in advancing biomarker identification, deepening our understanding of disease mechanisms, and supporting the development of targeted therapeutic strategies. The integration of these tools highlights their potential in personalized medicine and their capacity to improve clinical outcomes.
Collapse
Affiliation(s)
- Inês C. R. Graça
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Cláudia Martins
- Institute of Biomedicine (iBiMED), Department of Medical Sciences (DCM), University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Fernando Ribeiro
- Institute of Biomedicine (iBiMED), School of Health Sciences (ESSUA), University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Alexandra Nunes
- Institute of Biomedicine (iBiMED), Department of Medical Sciences (DCM), University of Aveiro, 3810-193 Aveiro, Portugal;
| |
Collapse
|
22
|
Fountoulakis PN, Theofilis P, Vlachakis PK, Karakasis P, Pamporis K, Sagris M, Dimitroglou Y, Tsioufis P, Oikonomou E, Tsioufis K, Tousoulis D. Gut Microbiota in Heart Failure-The Role of Inflammation. Biomedicines 2025; 13:911. [PMID: 40299538 PMCID: PMC12024997 DOI: 10.3390/biomedicines13040911] [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: 01/26/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Heart failure (HF) has become an immense health concern affecting almost 1-2% of the population globally. It is a complex syndrome characterized by activation of the sympathetic nervous system and the Renin-Angiotensin-Aldosterone (RAAS) axis as well as endothelial dysfunction, oxidative stress, and inflammation. The recent literature points towards the interaction between the intestinal flora and the heart, also called the gut-heart axis. The human gastrointestinal tract is naturally inhabited by various microbes, which are distinct for each patient, regulating the functions of many organs. Alterations of the gut microbiome, a process called dysbiosis, may result in systemic diseases and have been associated with heart failure through inflammatory and autoimmune mechanisms. The disorder of intestinal permeability favors the translocation of microbes and many metabolites capable of inducing inflammation, thus further contributing to the deterioration of normal cardiac function. Besides diet modifications and exercise training, many studies have revealed possible gut microbiota targeted treatments for managing heart failure. The aim of this review is to demonstrate the impact of the inflammatory environment induced by the gut microbiome and its metabolites on heart failure and the elucidation of these novel therapeutic approaches.
Collapse
Affiliation(s)
- Petros N. Fountoulakis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| | - Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| | - Panayotis K. Vlachakis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| | - Paschalis Karakasis
- 2nd Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Konstantinos Pamporis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| | - Marios Sagris
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| | - Yannis Dimitroglou
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| | - Panagiotis Tsioufis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.F.); (P.T.); (P.K.V.); (K.P.); (M.S.); (Y.D.); (P.T.); (K.T.)
| |
Collapse
|
23
|
Guo S, Yang F, Zhang J, Liao Y, Xia N, Tang T, Wang C, Wang QK, Chen C, Hu D, Shan Z, Cheng X. Inulin Diet Alleviates Abdominal Aortic Aneurysm by Increasing Akkermansia and Improving Intestinal Barrier. Biomedicines 2025; 13:920. [PMID: 40299521 PMCID: PMC12024805 DOI: 10.3390/biomedicines13040920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Previous studies have shown varying efficacy of high-fiber diets containing different ingredients in abdominal aortic aneurysms (AAAs). This study aimed to identify which high-fiber diet protects against AAA in mice and elucidate the underlying mechanisms. Methods: This study compared inulin, cellulose, and chow diets in terms of their impact on aneurysm enlargement, elastin degradation, matrix metalloproteinase 2 and 9 expressions, CD3+ T cell and CD68+ macrophage infiltration, and macrophage differentiation. It also examined gut microbiota composition, focusing on Akkermansia, and evaluated intestinal barrier function and systemic inflammatory response. Results: The inulin diet, but not the cellulose diet, compared with the chow diet, reduced aneurysm enlargement, elastin degradation, matrix metalloproteinase 2 and 9 expressions, CD3+ T cell and CD68+ macrophage infiltration, and skewed macrophage towards M2 differentiation. The inulin diet enriched Akkermansia in both the small and large intestine. The inulin diet also enhanced the intestinal barrier by augmenting goblet cells, upregulating the gene related to the epithelial barrier and antibacterial peptides in the small intestine, and reducing circulating lipopolysaccharide and interleukin-1β levels. The inulin diet lowered the proportion of Ly6Chi monocytes and C-C chemokine receptor 2 expression on these cells in the bone marrow, reducing aneurysm infiltration. Administering Akkermansia to AAA mice decreased intestinal permeability and mitigated AAA. Conclusions: A diet rich in fermentable fiber inulin, as opposed to cellulose, alleviates AAA in mice. This beneficial effect is attributed to the enhanced presence of Akkermansia bacteria and improvement of the intestinal barrier.
Collapse
Affiliation(s)
- Shuang Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (S.G.); (F.Y.); (J.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Vascular Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100084, China
| | - Fen Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (S.G.); (F.Y.); (J.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiyu Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (S.G.); (F.Y.); (J.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuhan Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (S.G.); (F.Y.); (J.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ni Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (S.G.); (F.Y.); (J.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tingting Tang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (S.G.); (F.Y.); (J.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chaolong Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qing K. Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhilei Shan
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (S.G.); (F.Y.); (J.Z.)
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
24
|
Tonon CR, Pereira AG, Ferreira NF, Monte MG, Vieira NM, Fujimori ASS, Ballin PDS, de Paiva SAR, Zornoff LAM, Minicucci MF, Polegato BF. The Gut-Heart Axis and Its Role in Doxorubicin-Induced Cardiotoxicity: A Narrative Review. Microorganisms 2025; 13:855. [PMID: 40284691 PMCID: PMC12029146 DOI: 10.3390/microorganisms13040855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Doxorubicin is a widely used chemotherapy for the treatment of several types of cancer. However, its application is restricted due to adverse effects, particularly cardiotoxicity, which can progress to heart failure-a chronic and debilitating condition. Several mechanisms have been identified in the pathophysiology of doxorubicin-induced cardiotoxicity, including oxidative stress, mitochondrial dysfunction, inflammation, and disruption of collagen homeostasis. More recently, dysbiosis of the gut microbiota has been implicated in the development and perpetuation of cardiac injury. Studies have reported alterations in the composition and abundance of the microbiota during doxorubicin treatment. Therefore, as of recent, there is a new field of research in order to develop strategies involving the gut microbiota to prevent or attenuate cardiotoxicity since there is no effective therapy at the moment. This narrative review aims to provide an update on the role of gut microbiota and intestinal permeability in the pathophysiology of cardiovascular diseases, and more specifically doxorubicin-induced cardiotoxicity. Additionally, it seeks to establish a foundation for future research targeting gut microbiota to alleviate cardiotoxicity.
Collapse
Affiliation(s)
- Carolina Rodrigues Tonon
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, Brazil (B.F.P.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wei L, Van Beeck W, Hanlon M, DiCaprio E, Marco ML. Lacto-Fermented Fruits and Vegetables: Bioactive Components and Effects on Human Health. Annu Rev Food Sci Technol 2025; 16:289-314. [PMID: 39805038 DOI: 10.1146/annurev-food-052924-070656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Lacto-fermented fruits and vegetables (FVs) such as kimchi, sauerkraut, and fermented olives and nonalcoholic juices have a long history as dietary staples. Herein, the production steps and microbial ecology of lacto-fermented FVs are discussed alongside findings from human and laboratory studies investigating the health benefits of these foods. Lacto-fermented FVs are enriched in beneficial live microbes and bioactive compounds, including lactic and acetic acids, phenolic compounds, bacteriocins, and amino acid derivatives such as indole-3-lactic acid, phenyl-lactic acid, and γ-aminobutyric acid. At least 11 human studies have been performed on kimchi, whereas others have been investigated in only one or two trials. Besides exploring the health benefits, it is imperative to ensure that these foods made either commercially or at home have minimal risk for foodborne illness and exposure to undesired compounds like biogenic amines. Development of starter-culture strains and production protocols can lead to lacto-fermented FVs designed for specific health benefits.
Collapse
Affiliation(s)
- Lei Wei
- Department of Food Science and Technology, University of California, Davis, Davis, California, USA;
| | - Wannes Van Beeck
- Department of Food Science and Technology, University of California, Davis, Davis, California, USA;
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Melanie Hanlon
- Department of Food Science and Technology, University of California, Davis, Davis, California, USA;
- Department of Food Science and Technology, Oregon State University, Corvallis, Oregon, USA
| | - Erin DiCaprio
- Department of Food Science and Technology, University of California, Davis, Davis, California, USA;
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, Davis, California, USA;
| |
Collapse
|
26
|
Guan X, Sun C, Su J, Sun Z, Cheng C. Deciphering the causality of gut microbiota, circulating metabolites and heart failure: a mediation mendelian. Front Pharmacol 2025; 16:1531384. [PMID: 40235532 PMCID: PMC11996798 DOI: 10.3389/fphar.2025.1531384] [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: 11/20/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Background Growing evidence suggesting a connection between the gut microbiome, plasma metabolites, and the development of heart failure (HF). However, the causality of this relationship remains to be fully elucidated. Methods Utilizing summary statistics from extensive genome-wide association studies (GWAS), we investigated the interplay among the gut microbiome, 1,400 plasma metabolites and heart failure. We conducted bidirectional Mendelian randomization (MR) analyses and MR mediation analysis to discern the causality within these relationships. The inverse variance-weighted (IVW) method served as our primary analytical approach, supported by various MR methods and sensitivity analyses. Results We revealed casual relationships between nine microbial groups/pathways and heart failure. Additionally, 15 metabolites exhibited casual links with HF, with eight exerting protective effects. Through two-step MR analysis we also identified the metabolite, Campesterol, mediated the increasing risk from gut microbiota to HF and a metabolite ratio played the converse role. Conclusion This investigation has provided robust evidence supporting the causal links between the gut microbiome, plasma metabolites, and heart failure. The findings enhance our comprehension of the role of circulating metabolites and offer significant insights for future etiological research and therapeutic development in heart failure.
Collapse
Affiliation(s)
- Xueqing Guan
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chaonan Sun
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| | - Jianyao Su
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhijun Sun
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cheng Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
27
|
González-Correa C, Moleón J, Miñano S, Robles-Vera I, de la Visitación N, Guerra-Hernández E, Toral M, Jiménez R, Duarte J, Romero M. Protective Effect of Dietary Fiber on Blood Pressure and Vascular Dysfunction Through Regulation of Sympathetic Tone and Immune Response in Genetic Hypertension. Phytother Res 2025; 39:1858-1875. [PMID: 40122676 DOI: 10.1002/ptr.8484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
The mechanisms underlying the antihypertensive effect of dietary fibers remain poorly understood. This study investigates whether dietary fiber supplementation can prevent cardiovascular damage and high blood pressure in a genetic model of neurogenic hypertension. Six-week-old male spontaneously hypertensive rats (SHR) and their respective normotensive control, Wistar Kyoto rats (WKY), were divided into four groups: Untreated WKY, untreated SHR, SHR treated with resistant starch (SHR + RS), and SHR treated with inulin-type fructans (SHR + ITF) for 12 weeks. Additionally, a faecal microbiota transplantation (FMT) experiment was conducted, transferring faecal content from treated SHR donors to recipient SHRs. A diet rich in RS fiber reduced vascular oxidative stress, inflammation, and high blood pressure. These protective effects were associated with a reshaped gut microbiota, leading to increased short-chain fatty acid production, reduced endotoxemia, decreased sympathetic activity, and a restored balance between Th17 and Treg lymphocytes in mesenteric lymph nodes and aorta. Elevated plasma levels of acetate and butyrate in the SHR + RS group correlated with increased expression of aortic GPR41, GRP43 and PPARδ. Conversely, ITF treatment failed to prevent hypertension or endothelial dysfunction in SHR. FMT from the SHR + RS group to recipient SHR partially replicated these beneficial effects. This study highlights the antihypertensive benefits of dietary insoluble RS fiber, which are attributed to enhanced short-chain fatty acids production in the gut. This leads to improved gut permeability, reduced sympathetic tone, and diminished vascular T-cell accumulation. Therefore, dietary interventions with RS fiber may offer promising therapeutic strategies for preventing hypertension.
Collapse
Affiliation(s)
- Cristina González-Correa
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Javier Moleón
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Sofía Miñano
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Néstor de la Visitación
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | | | - Marta Toral
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
- Ciber de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy and Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, Ibs.GRANADA, Granada, Spain
| |
Collapse
|
28
|
Bikbavova G, Livzan M, Turchaninov D, Lisyutenko N, Romanyuk A, Mashnin M. Diagnostic significance of the gut microbiota and its metabolites in cardiovascular risk assessment among patients with ulcerative colitis. RUSSIAN JOURNAL OF EVIDENCE-BASED GASTROENTEROLOGY 2025; 14:39. [DOI: 10.17116/dokgastro20251401139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Cardiovascular diseases (CVDs) rank among the leading causes of mortality. Studies suggest that ulcerative colitis (UC) is associated with an increased risk of CVDs. Gut microbiota and its metabolites play a pivotal role in the development of cardiovascular pathologies, including through the proatherogenic effects of trimethylamine N-oxide (TMAO) and disruptions in intestinal permeability. Objective: To evaluate the diagnostic value of markers characterizing gut microbiota metabolism and intestinal permeability in UC patients in the context of arterial hypertension (AH) risk. Materials and Methods. The study included 80 UC patients divided into two groups: those with UC and AH (Group 1) and those without AH (Group 2). TMAO levels in blood, zonulin, and short-chain fatty acids (SCFAs) in stool were assessed. Quantitative features with an alternative distribution were described using the median (Me), upper, and lower quartiles (P25, P75). The Mann-Whitney test and Bayesian approach were applied to evaluate diagnostic significance. Results. A significant increase in TMAO (1473.4 ng/mL vs. 785.02 ng/mL, p=0.01) and zonulin levels (318.42 ng/g vs. 164.22 ng/g, p=0.039) was observed in Group 1. Reduced acetic acid levels and anaerobic index were noted in the AH group, indicating gut microbiota imbalance. Conclusion. UC is a systemic inflammatory disease associated with an increased risk of CVDs. Investigating gut microbiota and its metabolites provides new opportunities for the prevention and treatment of cardiovascular complications in UC patients.
Collapse
|
29
|
Zhang M, Zhou C, Li X, Li H, Han Q, Chen Z, Tang W, Yin J. Interactions between Gut Microbiota, Host Circadian Rhythms, and Metabolic Diseases. Adv Nutr 2025; 16:100416. [PMID: 40139315 DOI: 10.1016/j.advnut.2025.100416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
The circadian rhythm arises endogenously from genetically encoded molecular clocks, wherein the components collaborate to induce cyclic fluctuations, occurring approximately every 24 h. The rhythms synchronize biological processes with regular and predictable environmental patterns to guarantee the host metabolism and energy homeostasis function and well-being. Disruptions to circadian rhythms are widely associated with metabolic disorders. Notably, microbial rhythms are influenced by both the host's intrinsic circadian clock and external rhythmic factors (i.e., light-dark cycle, diet patterns, and diet composition), which affect the structure of microbial communities and metabolic functions. Moreover, microbiota and the metabolites also reciprocally influence host rhythms, potentially impacting host metabolic function. This review aimed to explore the bidirectional interactions between the circadian clock, factors influencing host-microbial circadian rhythms, and the effects on lipid metabolism and energy homeostasis.
Collapse
Affiliation(s)
- Mingliang Zhang
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | | | - Xinguo Li
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Hui Li
- Xiangxi Vocational and Technical College for Nationalities, Jishou, China
| | - Qi Han
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Zhong Chen
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China; Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group, Co Ltd, Chengdu, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China.
| |
Collapse
|
30
|
Kaur N, Kumar P, Dhami M, Aran KR. Antibiotic-induced gut dysbiosis: unraveling the gut-heart axis and its impact on cardiovascular health. Mol Biol Rep 2025; 52:319. [PMID: 40095156 DOI: 10.1007/s11033-025-10425-2] [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: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Cardiovascular diseases (CVDs) remain the major cause of morbidity and mortality amongst people of all ages across the world. Research suggests that the initiation and progression of CVDs are associated with antibiotic-induced gut dysbiosis. Antibiotics are primarily intended to be used to treat bacterial infections, which can alter gut microbiota (GM) composition, by lowering the abundance of beneficial bacteria, like Firmicutes, Bacteroidetes, and increasing the profusion of Enterobacteriaceae, leading to harm on gut health. Additionally, it reduces short-chain fatty acids (SCFAs) and bile acid metabolism, increases trimethylamine N-oxide (TMAO) production, intestinal permeability allowing lipopolysaccharide (LPS) and TMAO into systemic circulation. SCFAs play a key role in lipid metabolism, inflammation, and strengthening of the intestinal barrier, and participate in CVDs through FFAR2 and FFAR3 receptors, whereas dysbiosis reduces SCFAs levels and worsens these effects. TMAO enhances oxidative stress, inflammation, endothelial dysfunction, and cholesterol dysregulation, thus worsening CVDs. Furthermore, LPS develops systemic inflammation, insulin resistance, and endothelial dysfunction by activating the NF-κB pathway. Dysbiosis also affects bile acid synthesis, disrupting lipid and glucose metabolism, further participating in the progression of CVDs. This article aims to explore the role of gut dysbiosis in various CVDs, including congenital heart disease, hypertension, valvular heart disease, coronary heart disease, and heart failure. Furthermore, this article aims to bridge the knowledge gap regarding the gut-heart axis by exploring how antibiotics alter the gut microbiota homeostasis, further contributing to the development of CVDs and therapeutic interventions that reduce cardiovascular risks and restore the gut microbiota homeostasis.
Collapse
Affiliation(s)
- Navpreet Kaur
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India
| | - Pankaj Kumar
- Department of Pharmacology, Himachal Institute of Pharmaceutical Education and Research (HIPER), Tehsil-Nadaun, Hamirpur, Himachal Pradesh, 177033, India
| | - Mahadev Dhami
- Bhimdatta Polytechnic Institute, Patan, Baitadi, 10200, Nepal
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
31
|
Li J, Wei W, Ma X, Ji J, Ling X, Xu Z, Guan Y, Zhou L, Wu Q, Huang W, Liu F, Zhao M. Antihypertensive effects of rice peptides involve intestinal microbiome alterations and intestinal inflammation alleviation in spontaneously hypertensive rats. Food Funct 2025; 16:1731-1759. [PMID: 39752320 DOI: 10.1039/d4fo04251d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Gut dysbiosis serves as an underlying risk factor for the development of hypertension. The resolution of this dysbiosis has emerged as a promising strategy in improving hypertension. Food-derived bioactive protein peptides have become increasingly more attractive in ameliorating hypertension, primarily due to their anti-inflammatory and anti-oxidant activities. However, the regulatory mechanisms linking rice peptides (RP), gut dysbiosis, and hypertension remain to be fully elucidated. In our study, male spontaneously hypertensive rats (SHR) were fed with chow diet and concomitantly treated with ddH2O (Ctrl) or varying doses of rice peptides (20, 100, or 500 mg (kg bw day)-1 designated as low-dose RP, LRP; medium-dose RP, MRP; high-dose RP, HAP) or captopril (Cap) by intragastric administration. Wistar-Kyoto (WKY) rats served as the normotensive control group and were orally administered with ddH2O. We observed beneficial effects of RP in lowering blood pressure and ameliorating cardiovascular risk profiles, as evidenced by improvements in glucolipid metabolic disorders, hepatic and renal damage, left ventricular hypertrophy and endothelial dysfunction in hypertensive rats. More importantly, we found that RP attenuated intestinal pathological damage, improved impaired intestinal barrier, and reduced intestinal inflammation by inhibiting the HMGB1-TLR4-NF-κB pathway. Notably, multi-omics integrative analyses have revealed that RP altered the composition and function of the gut microbiota. This is exemplified by the observed enrichment of beneficial bacterial constituents, such as g_Lactobacillus, g_Lactococcus, s_Lactobacillus_intestinalis, and Lactococcus lactis, and elevated production of microbiota-derived short-chain fatty acid metabolites. Collectively, these studies suggest that the hypotensive effects of RP may be associated with modulation of the gut microbiota and its short-chain fatty acids metabolites. This implicates the microbiota-gut-HMGB1-TLR4-NF-κB axis as a novel venue for the amelioration of hypertension and its complications.
Collapse
Affiliation(s)
- Juan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Wei Wei
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, 274108, China.
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xiaomin Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Center for Experimental Public Health and Preventive Medicine Education, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Jing Ji
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Xiaomeng Ling
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Zhuyan Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Yutong Guan
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Leyan Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Qiming Wu
- Nutrilite Health Institute, Shanghai, 201203, China.
| | - Wenhua Huang
- AMWAY (China) R&D Center, Guangzhou, 510730, China.
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Min Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
32
|
Hatamnejad MR, Medzikovic L, Dehghanitafti A, Rahman B, Vadgama A, Eghbali M. Role of Gut Microbial Metabolites in Ischemic and Non-Ischemic Heart Failure. Int J Mol Sci 2025; 26:2242. [PMID: 40076864 PMCID: PMC11900495 DOI: 10.3390/ijms26052242] [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: 01/22/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The effect of the gut microbiota extends beyond their habitant place from the gastrointestinal tract to distant organs, including the cardiovascular system. Research interest in the relationship between the heart and the gut microbiota has recently been emerging. The gut microbiota secretes metabolites, including Trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), bile acids (BAs), indole propionic acid (IPA), hydrogen sulfide (H2S), and phenylacetylglutamine (PAGln). In this review, we explore the accumulating evidence on the role of these secreted microbiota metabolites in the pathophysiology of ischemic and non-ischemic heart failure (HF) by summarizing current knowledge from clinical studies and experimental models. Elevated TMAO contributes to non-ischemic HF through TGF-ß/Smad signaling-mediated myocardial hypertrophy and fibrosis, impairments of mitochondrial energy production, DNA methylation pattern change, and intracellular calcium transport. Also, high-level TMAO can promote ischemic HF via inflammation, histone methylation-mediated vascular fibrosis, platelet hyperactivity, and thrombosis, as well as cholesterol accumulation and the activation of MAPK signaling. Reduced SCFAs upregulate Egr-1 protein, T-cell myocardial infiltration, and HDAC 5 and 6 activities, leading to non-ischemic HF, while reactive oxygen species production and the hyperactivation of caveolin-ACE axis result in ischemic HF. An altered BAs level worsens contractility, opens mitochondrial permeability transition pores inducing apoptosis, and enhances cholesterol accumulation, eventually exacerbating ischemic and non-ischemic HF. IPA, through the inhibition of nicotinamide N-methyl transferase expression and increased nicotinamide, NAD+/NADH, and SIRT3 levels, can ameliorate non-ischemic HF; meanwhile, H2S by suppressing Nox4 expression and mitochondrial ROS production by stimulating the PI3K/AKT pathway can also protect against non-ischemic HF. Furthermore, PAGln can affect sarcomere shortening ability and myocyte contraction. This emerging field of research opens new avenues for HF therapies by restoring gut microbiota through dietary interventions, prebiotics, probiotics, or fecal microbiota transplantation and as such normalizing circulating levels of TMAO, SCFA, BAs, IPA, H2S, and PAGln.
Collapse
Affiliation(s)
| | | | | | | | | | - Mansoureh Eghbali
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, BH-550 CHS, Los Angeles, CA 90095-7115, USA; (M.R.H.); (L.M.); (A.D.); (B.R.); (A.V.)
| |
Collapse
|
33
|
Zhou Z, Sun L, Zhou W, Gao W, Yuan X, Zhou H, Ren Y, Li B, Wu Y, She J. Probiotic Bifidobacterium reduces serum TMAO in unstable angina patients via the gut to liver to heart axis. LIVER RESEARCH (BEIJING, CHINA) 2025; 9:57-65. [PMID: 40206430 PMCID: PMC11977283 DOI: 10.1016/j.livres.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 04/11/2025]
Abstract
Background and aims Studies indicate that the gut microbiota and its metabolites are involved in the progression of cardiovascular diseases, and enterohepatic circulation plays an important role in this progression. This study aims to identify potential probiotics for the treatment of unstable angina (UA) and elucidate their mechanisms of action. Methods Initially, the gut microbiota from patients with UA and control was analyzed. To directly assess the effects of Bifidobacterium supplementation, 10 patients with UA were enrolled and administered Bifidobacterium (630 mg per intake twice a day for 1 month). The fecal metagenome, serum trimethylamine N-oxide (TMAO) levels, and other laboratory parameters were evaluated before and after Bifidobacterium supplementation. Results After supplementing with Bifidobacterium for 1 month, there were statistically significant differences (P < 0.05) in TMAO, aspartate aminotransferase, total cholesterol, and low-density lipoprotein compared to before. Additionally, the abundance of Bifidobacterium longum increased significantly, although the overall abundance of Bifidobacterium did not reach statistical significance. The gut microbiota, metabolites, and gut-liver axis are involved in the progression of UA, and potential mechanisms should be further studied. Conclusions Metagenomic analysis demonstrated a reduced abundance of Bifidobacterium in patients with UA. Supplementation with Bifidobacterium restored gut dysbiosis and decreased circulating TMAO levels in patients with UA. This study provides evidence that Bifidobacterium may exert cardiovascular-protective effects through the gut-liver-heart axis. Clinical trial number ChiCTR2400093946.
Collapse
Affiliation(s)
- Zhihong Zhou
- Department of Cardiology, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region, China
| | - Lizhe Sun
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Cardiometabolic Innovation Center of Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Wei Zhou
- Department of Cardiology, Wuzhong People’s Hospital, Wuzhong, Ningxia, China
| | - Wen Gao
- Department of Cardiology, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region, China
| | - Xiao Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Cardiometabolic Innovation Center of Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Huijuan Zhou
- Department of Cardiology, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region, China
| | - Yuzhen Ren
- Department of Cardiology, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region, China
| | - Bihua Li
- Department of Cardiology, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region, China
| | - Yue Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Cardiometabolic Innovation Center of Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jianqing She
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Cardiometabolic Innovation Center of Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
34
|
Williams EG, Alissa M, Alsugoor MH, Albakri GS, Altamimi AA, Alabdullateef AA, Almansour NM, Aldakheel FM, Alessa S, Marber M. Integrative approaches to atrial fibrillation prevention and management: Leveraging gut health for improved cardiovascular outcomes in the aging population. Curr Probl Cardiol 2025; 50:102952. [PMID: 39626858 DOI: 10.1016/j.cpcardiol.2024.102952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/14/2024]
Abstract
Atrial fibrillation (AF) is a prevalent clinical arrhythmia associated with a high incidence and severe complications such as cerebral embolism and heart failure. While the etiology and pathogenesis of AF involve numerous factors, recent research emphasizes the significant role of intestinal microbiota imbalance in the emergence and progression of AF, particularly among older adults. This review investigates the mechanisms by which intestinal flora and their metabolites contribute to the onset of AF in the elderly, highlighting novel interactions between gut health and cardiac function. Current literature often overlooks these critical connections, indicating a substantial research gap in understanding how dysbiosis may exacerbate AF and hinder recovery. Furthermore, exploring the bidirectional relationship between the gut microbiome and systemic inflammation in the context of AF provides a unique perspective that has yet to be thoroughly investigated. Future research should focus on longitudinal studies assessing gut microbiota composition and function in AF patients and consider probiotics or prebiotics as potential adjunctive therapies for mitigating AF. This comprehensive approach may pave the way for innovative treatments integrating cardiology with gastroenterology, enhancing patient outcomes through a holistic understanding of health.
Collapse
Affiliation(s)
- Emma Grace Williams
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112; 2 Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Mahdi H Alsugoor
- Department of Emergency Medical Services, Faculty of Health Sciences, AlQunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia
| | - Ghadah Shukri Albakri
- Department of Teaching and Learning, College of Education and Human Development, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Ali A Altamimi
- Department of Medical Laboratory, Prince Sultan Air Base Hospital, Al-Kharj, Saudi Arabia
| | | | - Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| | - Fahad M Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Salem Alessa
- Department of Medical Laboratory, Al Kharj Military Industries Corporation Hospital, Al-kharj, Saudi Arabia
| | - Michael Marber
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| |
Collapse
|
35
|
Li L, Yang L, Liu M. The effect of high-fiber diet based on gut microbiota in patients with chronic heart failure. Physiol Genomics 2025; 57:140-145. [PMID: 39823195 DOI: 10.1152/physiolgenomics.00163.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/12/2024] [Accepted: 12/21/2024] [Indexed: 01/19/2025] Open
Abstract
This research explored the effect of a high-fiber diet based on gut microbiota on patients with chronic heart failure (HF). Patients with chronic HF, who had undergone a dietary survey indicating a daily dietary fiber intake of less than 15 g/day, were divided into the control and study groups (n = 50). In addition to conventional heart failure treatment, the study group received dietary guidance, while the control group did not receive any dietary guidance and maintained their usual low-fiber dietary habits. After 1 yr intervention, the daily dietary fiber intake, abundance of gut microbiota, plasma trimethylamine N-oxide (TMAO), albumin (ALB), prealbumin (PA), transferrin (TF), C-reactive protein (CRP), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-8 (IL-8), left ventricular ejection fraction (LVEF), left ventricular end-diastolic index (LVEDVI), and left ventricular end-systolic volume index (LVESVI), Barthel index (BI), and 6 min walking distance (6 MWD) were compared. After the intervention, in both groups, the daily dietary fiber intake increased and the abundance of Firmicutes, Proteobacteria, Actinobacteria, and Fusobacteria decreased and that of Bacteroides increased; the plasma TMAO decreased; serum ALB, PA, and TF levels increased; serum CRP, TNF-α, IL-6, and IL-8 levels decreased, and the change was greater in the study group; LVEF elevated, LVEDVI and LVESVI reduced, and the differences between both groups were not significant; BI and 6 MWD elevated, and the study group was higher than the control group. High-fiber diet positively regulates the composition of gut microbiota, nutritional status, and microinflammatory level in patients with chronic HF, thereby improving patients' quality of life.NEW & NOTEWORTHY Traditional heart failure (HF) treatment primarily focuses on medications and surgery, whereas this study explores the improvement effects of a diet high in dietary fiber on patients with chronic HF from the perspective of gut microbiota, providing a new perspective for HF treatment.
Collapse
Affiliation(s)
- Liwei Li
- Department of Cardiovascular Medicine, Wuhan Integrated Traditional Chinese and Western Medicine Hospital (Wuhan First Hospital), Wuhan, People's Republic of China
| | - Li Yang
- Department of Cardiovascular Medicine, Wuhan Integrated Traditional Chinese and Western Medicine Hospital (Wuhan First Hospital), Wuhan, People's Republic of China
| | - Mingrong Liu
- Department of Radiology, Wuhan Integrated Traditional Chinese and Western Medicine Hospital (Wuhan First Hospital), Wuhan, People's Republic of China
| |
Collapse
|
36
|
Yang Q, Kang Y, Tang W, Li M, Zhao C. Interplay of gut microbiota in Kawasaki disease: role of gut microbiota and potential treatment strategies. Future Microbiol 2025; 20:357-369. [PMID: 40013895 PMCID: PMC11938985 DOI: 10.1080/17460913.2025.2469432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Kawasaki disease (KD) is an acute systemic immune vasculitis with predominant involvement of the medium and small arteries. It mostly affects pediatric patients, representing the most common form of pediatric vasculitis in children less than 5 years old. Numerous diseases, especially those related to the immune system, have established links with the intestinal flora. Recent studies have investigated the intestinal flora changes throughout the management of KD. There was gut microbiota dysbiosis in pediatric KD at the acute phase, particularly the downregulation of short-chain fat acids-producing microbiota and the over-proliferation of opportunistic pathogens. The relationship between the response to therapies in individuals with KD and specific microbiota remains uncertain. Targeted microbial supplements and dietary regulation may serve as potential measures to alleviate KD complications and thus improve prognosis. This review provides an overview of the current understanding of the interplay of the gut microbiota and KD. Furthermore, it discusses the possibility of altering the gut microbiota to reinstate a healthy condition.
Collapse
Affiliation(s)
- Qing Yang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Yaqing Kang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Tang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Cuifen Zhao
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
37
|
Shremo Msdi A, Haghparast A, Garey KW, Wang EM. Microbiome-Based Therapeutics for Salt-Sensitive Hypertension: A Scoping Review. Nutrients 2025; 17:825. [PMID: 40077695 PMCID: PMC11901721 DOI: 10.3390/nu17050825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 01/29/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The purpose of this scoping review was to provide a comprehensive understanding of the current knowledge concerning the gut microbiome and SCFAs as emerging treatments for salt-sensitive hypertension. Relevant animal and human studies were identified via PubMed through August 2024. Twenty-four human (n = 9) and animal (n = 15) trials were included. Most human studies were observational (n = 6), aiming to compare gut microbiota differences between hypertensive and normotensive individuals. Three human studies evaluated microbiome-based interventions either via a sodium-restricted diet (n = 2) or prebiotic supplementation (n = 1). Fifteen animal trials involving either mice or rats were identified, all of which were interventional. These included dietary changes (n = 9), probiotic treatments (n = 1), postbiotic primarily bacterial metabolites (n = 4), and live biotherapeutic products (n = 4). All interventions were effective in decreasing blood pressure. Microbiome-based therapies as biologic modifiers for salt-sensitive hypertension are emerging. Substantial knowledge gaps remain, warranting further research to fully explore this promising therapeutic avenue.
Collapse
Affiliation(s)
- Abdulwhab Shremo Msdi
- Department of Pharmacy Practice and Translational Research, College of Pharmacy, University of Houston, 4349 Martin Luther King Boulevard, Houston, TX 77204, USA
| | | | | | | |
Collapse
|
38
|
Shen S, Tian B, Zhang H, Wang YC, Li T, Cao Y. Heart Failure and Gut Microbiota: What Is Cause and Effect? RESEARCH (WASHINGTON, D.C.) 2025; 8:0610. [PMID: 39981296 PMCID: PMC11839986 DOI: 10.34133/research.0610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
Emerging evidence highlights the central role of gut microbiota in maintaining physiological homeostasis within the host. Disruptions in gut microbiota can destabilize systemic metabolism and inflammation, driving the onset and progression of cardiometabolic diseases. In heart failure (HF), intestinal dysfunction may induce the release of endotoxins and metabolites, leading to dysbiosis and exacerbating HF through the gut-heart axis. Understanding the relationship between gut microbiota and HF offers critical insights into disease mechanisms and therapeutic opportunities. Current research highlights promising potential to improve patient outcomes by restoring microbiota balance. In this review, we summarize the current studies in understanding the gut microbiota-HF connection and discuss avenues for future investigation.
Collapse
Affiliation(s)
- Shichun Shen
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Beiduo Tian
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Haizhu Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yu-Chen Wang
- Department of Medicine, Division of Cardiology, Department of Microbiology, Immunology and Molecular Genetics, and Department of Human Genetics,
University of California, Los Angeles, CA, USA
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics,
West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yang Cao
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
39
|
Khan MM, Kirabo A. Fiber, Fatty Acids, and Blood Pressure: A Gut-Level Solution. Circ Res 2025; 136:358-360. [PMID: 39946444 PMCID: PMC11839179 DOI: 10.1161/circresaha.125.326065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Affiliation(s)
- Mohd Mabood Khan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 (M.M.K., A.K.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 (M.M.K., A.K.)
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology, and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| |
Collapse
|
40
|
R Muralitharan R, Zheng T, Dinakis E, Xie L, Barbaro-Wahl A, Jama HA, Nakai M, Paterson M, Leung KC, McArdle Z, Mirabito Colafella K, Johnson C, Qin W, Salimova E, Bitto NJ, Kaparakis-Liaskos M, Kaye DM, O'Donnell JA, Mackay CR, Marques FZ. Gut Microbiota Metabolites Sensed by Host GPR41/43 Protect Against Hypertension. Circ Res 2025; 136:e20-e33. [PMID: 39840468 DOI: 10.1161/circresaha.124.325770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Fermentation of dietary fiber by the gut microbiota leads to the production of metabolites called short-chain fatty acids, which lower blood pressure and exert cardioprotective effects. Short-chain fatty acids activate host signaling responses via the functionally redundant receptors GPR41 (G-protein-coupled receptor 41) and GPR43 (G-protein-coupled receptor 43), which are highly expressed by immune cells. Whether and how these receptors protect against hypertension or mediate the cardioprotective effects of dietary fiber remains unknown. METHODS Cardiovascular phenotype was assessed in untreated and Ang II (angiotensin II) treated hypertensive wild-type and GPR41/43 knockout (KO) double knockout male mice fed diets with different levels of fiber content. Some mice received TLR4 (toll-like receptor 4)-antagonist treatment and bone marrow chimeras. SNPs (single-nucleotide polymorphisms) associated with GPR41 and GPR43 expression were assessed in UK Biobank participants. RESULTS Untreated GPR41/43KO mice had unaltered blood pressure but had greater cardiac and renal collagen deposition with higher macrophage numbers in the kidney compared with wild-type mice. Ang II-treated GPR41/43KO mice showed higher systolic blood pressure, cardiorenal weights and collagen deposition, and increased gut permeability, which allows the translocation of gastrointestinal bacterial components such as lipopolysaccharides into the circulation. The use of an antagonist to the lipopolysaccharide receptor, TLR4, a potent proinflammatory signaling molecule, restored the cardiovascular phenotype in GPR41/43KO mice. The lack of GPR41/43 expression in the immune compartment was sufficient to lead to a worsened hypertensive phenotype. We also demonstrate that GPR41/43 is, at least partially, responsible for the blood pressure-lowering and cardioprotective effects of a high-fiber diet. Finally, using the UK Biobank, we provide translational evidence that variants associated with lower expression of both GPR41 and GPR43 are more prevalent in participants with hypertension. CONCLUSIONS Our findings highlight that lack of short-chain fatty acid-receptor signaling via both GPR41 and GPR43 increases risk of high blood pressure, suggesting treatments that target these receptors could be a novel strategy to prevent or treat hypertension.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Tenghao Zheng
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Evany Dinakis
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Liang Xie
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia (L.X., C.R.M.)
- Now with Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore (L.X.)
| | - Anastasia Barbaro-Wahl
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Hamdi A Jama
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Michael Nakai
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Madeleine Paterson
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Kwan Charmaine Leung
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Zoe McArdle
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia (Z.M., K.M.C.)
| | - Katrina Mirabito Colafella
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia (Z.M., K.M.C.)
| | - Chad Johnson
- Bioimaging Platform, La Trobe University, Melbourne, Australia (C.J.)
| | - Wendy Qin
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Ekaterina Salimova
- Monash Bioimaging Facility, Monash University, Melbourne, Australia (E.S.)
| | - Natalie J Bitto
- Department of Microbiology, Anatomy, Physiology and Pharmacology (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- La Trobe Research Centre for Extracellular Vesicles (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
| | - Maria Kaparakis-Liaskos
- Department of Microbiology, Anatomy, Physiology and Pharmacology (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- La Trobe Research Centre for Extracellular Vesicles (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- Now with Department of Microbiology & Immunology, University of Melbourne, Australia (M. K-L.)
| | - David M Kaye
- Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia (D.M.K.)
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (D.M.K., F.Z.M.)
- Department of Cardiology, Alfred Hospital, Melbourne, Australia (D.M.K.)
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Charles R Mackay
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia (L.X., C.R.M.)
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China (C.R.M.)
| | - Francine Z Marques
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (D.M.K., F.Z.M.)
| |
Collapse
|
41
|
Mohammadi F, Rudkowska I. Dietary Lipids, Gut Microbiota, and Their Metabolites: Insights from Recent Studies. Nutrients 2025; 17:639. [PMID: 40004966 PMCID: PMC11858126 DOI: 10.3390/nu17040639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/30/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Dietary lipid intake can influence the gut microbiota (GM) and their metabolites, such as short-chain fatty acids (SCFA) and bile acids, which are key mediators of health. The objective is to examine how dietary lipids' quantity and quality influence the GM and metabolite profiles. A literature review of 33 studies in animals and humans was performed on the effects of saturated fatty acids (SFAs), monounsaturated fatty acids (MUFAs), polyunsaturated fatty acids (PUFAs), trans-fatty acids (TFAs), and sterols on GM composition and gut-derived metabolites. The results show that diets rich in MUFAs, n-3 PUFAs, and short-chain FAs have the potential to enhance beneficial bacteria and metabolites. In addition, trans-palmitoleic acid, conjugated linoleic acid, and phytosterols may also have potentially beneficial effects on GM, but more research is needed. Medium-chain FAs and n-6 PUFAs have variable effects on the GM. Conversely, intakes of high-fat diets, long-chain SFAs, industrial TFAs, and cholesterol disrupt GM balance. In conclusion, animal studies clearly demonstrate that dietary fats influence the GM and related metabolites. Yet, human studies are limited. Therefore, well-designed human studies that consider the whole diet and baseline health status are needed to better understand the effects of dietary lipids on GM.
Collapse
Affiliation(s)
- Farzad Mohammadi
- Endocrinology and Nephrology Unit, CHU de Québec—Université Laval Research Center, 2705 Laurier Blvd, Québec, QC G1V 4G2, Canada;
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Iwona Rudkowska
- Endocrinology and Nephrology Unit, CHU de Québec—Université Laval Research Center, 2705 Laurier Blvd, Québec, QC G1V 4G2, Canada;
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
42
|
Zhou T, Wang Z, Lv X, Guo M, Zhang N, Liu L, Geng L, Shao J, Zhang K, Gao M, Mao A, Zhu Y, Yu F, Feng L, Wang X, Zhai Q, Chen W, Ma X. Targeting gut S. aureofaciens Tü117 serves as a new potential therapeutic intervention for the prevention and treatment of hypertension. Cell Metab 2025; 37:496-513.e11. [PMID: 39908987 DOI: 10.1016/j.cmet.2025.01.004] [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: 01/03/2024] [Revised: 09/13/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025]
Abstract
Currently, the regulation of specific gut microbial metabolism for the development and/or treatment of hypertension remains largely unexplored. Here, we show that α-lipomycin, produced by Streptomyces aureofaciens (S. aureofaciens) Tü117, is upregulated in the serum of high-salt diet (HSD) mice and patients with essential hypertension. α-lipomycin causes vasodilation impairment involving transient receptor potential vanilloid 4 (TRPV4)-mediated nitric oxide and endothelium-derived hyperpolarizing factor pathways in mice. We also find that Lactobacillus plantarum (L. plantarum) CCFM639 attenuates the increase in blood pressure (BP) potentially through inhibiting the proliferation of S. aureofaciens Tü117 in mice. An exploratory intervention trial indicates that L. plantarum CCFM639 supplementation reduces BPs in subjects newly diagnosed with pre-hypertension or stage 1 hypertension without antihypertensive medication. Our findings provide evidence for a role of S. aureofaciens Tü117-associated α-lipomycin elevation in the pathogenesis of HSD-induced hypertension, highlighting that targeting gut bacteria serves as a new therapeutic intervention for hypertension.
Collapse
Affiliation(s)
- Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Zhiwei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Xiaowang Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Mengting Guo
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ning Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Liangju Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Jing Shao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Mengru Gao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Yifei Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Xiaoyan Wang
- Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Qixiao Zhai
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Food Nutrition and Human Health Interdisciplinary Center, School of Food Science and Technology, Wuxi School of medicine, Jiangnan University, Wuxi 214122, China; Affiliated Hospital of Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
43
|
Gao Y, Qin G, Liang S, Yin J, Wang B, Jiang H, Liu M, Luo F, Li X. Metagenomic Sequencing Combined with Metabolomics to Explore Gut Microbiota and Metabolic Changes in Mice with Acute Myocardial Infarction and the Potential Mechanism of Allicin. Drug Des Devel Ther 2025; 19:771-791. [PMID: 39925879 PMCID: PMC11806679 DOI: 10.2147/dddt.s504884] [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: 11/05/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025] Open
Abstract
Background Acute myocardial infarction (AMI) is a significant contributor to global morbidity and mortality. Allicin exhibits promising therapeutic potential in AMI as a primary bioactive component derived from garlic; however, its underlying mechanisms remain incompletely elucidated. Methods Our study induced AMI in mice by ligating the left coronary artery, and administered allicin orally for 28 days. The cardioprotective effects of allicin treatment were comprehensively assessed using echocardiography, histopathological examinations, intestinal barrier function, and serum inflammatory factors. The potential mechanisms of allicin were elucidated through analysis of metagenomics and serum metabolomics. Network pharmacology (NP) was used to further investigate and validate the possible molecular mechanisms of allicin. Results Our findings revealed allicin's capacity to ameliorate cardiac impairments, improve intestinal barrier integrity, and reduce serum IL-18 and IL-1β levels after AMI. Further analysis demonstrated that the administration of allicin has the potential to ameliorate intestinal flora disorder following AMI by modulating the abundance of beneficial bacteria, such as g_Lactobacillus, g_Prevotella, g_Alistipes, and g_Limosilactobacillus, while reducing the abundance of harmful bacteria g_Parasutterella. Additionally, it exhibits the ability to enhance myocardial energy metabolism flexibility through modulating metabolites and key enzymes associated with the fatty acid metabolic pathway. Mechanistically, NP and in vivo experiments indicated that allicin might suppress pyroptosis and reduce inflammatory response via blocked activation of the NF-κB-mediated NLRP3/Caspase-1/GSDMD pathway. Moreover, Spearman correlation analysis suggested a significant association between the allicin-induced alterations in microbiota and metabolites with cardiac function and inflammatory cytokines. Conclusion Our study demonstrated that allicin alleviated myocardial injury and reduced inflammatory response by inhibiting the NF-κB-mediated NLRP3/Caspase-1/GSDMD pathway while remodeling microbiota disturbance, improving serum metabolic disorder, and enhancing the intestinal barrier. These research findings offer a novel perspective on the potential therapeutic value of allicin as an adjunctive dietary supplement to conventional treatments for AMI.
Collapse
Affiliation(s)
- Yijie Gao
- National Integrated Traditional and Western Medicine Center for Cardiovascular Disease, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Gaofeng Qin
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Shandong, People’s Republic of China
| | - Shichao Liang
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Jiajie Yin
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Baofu Wang
- National Integrated Traditional and Western Medicine Center for Cardiovascular Disease, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Hong Jiang
- National Integrated Traditional and Western Medicine Center for Cardiovascular Disease, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Mengru Liu
- National Integrated Traditional and Western Medicine Center for Cardiovascular Disease, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Fangyuan Luo
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Xianlun Li
- National Integrated Traditional and Western Medicine Center for Cardiovascular Disease, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| |
Collapse
|
44
|
Chulenbayeva L, Issilbayeva A, Sailybayeva A, Bekbossynova M, Kozhakhmetov S, Kushugulova A. Short-Chain Fatty Acids and Their Metabolic Interactions in Heart Failure. Biomedicines 2025; 13:343. [PMID: 40002756 PMCID: PMC11853371 DOI: 10.3390/biomedicines13020343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Short-chain fatty acids (SCFAs), produced through fermentation of dietary fibers by gut bacteria, play a central role in modulating cardiovascular function and heart failure (HF) development. The progression of HF is influenced by intestinal barrier dysfunction and microbial translocation, where SCFAs serve as key mediators in the gut-heart axis. This review examines the complex metabolic interactions between SCFAs and other gut microbiota metabolites in HF, including their relationships with trimethylamine N-oxide (TMAO), aromatic amino acids (AAAs), B vitamins, and bile acids (BAs). We analyze the associations between SCFA production and clinical parameters of HF, such as left ventricular ejection fraction (LVEF), N-terminal pro-B-type natriuretic peptide (NT-proBNP), and glomerular filtration rate (GFR). Gaining insights into metabolic networks offers new potential therapeutic targets and prognostic markers for managing heart failure, although their clinical significance needs further exploration.
Collapse
Affiliation(s)
- Laura Chulenbayeva
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.I.); (S.K.); (A.K.)
| | - Argul Issilbayeva
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.I.); (S.K.); (A.K.)
| | - Aliya Sailybayeva
- Heart Center, CF “University Medical Center”, Astana 010000, Kazakhstan; (A.S.); (M.B.)
| | | | - Samat Kozhakhmetov
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.I.); (S.K.); (A.K.)
| | - Almagul Kushugulova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (A.I.); (S.K.); (A.K.)
- Heart Center, CF “University Medical Center”, Astana 010000, Kazakhstan; (A.S.); (M.B.)
| |
Collapse
|
45
|
Huang L, Liu M, Shen L, Chen D, Wu T, Gao Y. Polysaccharides from Yupingfeng granules ameliorated cyclophosphamide-induced immune injury by protecting intestinal barrier. Int Immunopharmacol 2025; 146:113866. [PMID: 39709910 DOI: 10.1016/j.intimp.2024.113866] [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: 08/05/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024]
Abstract
Immune injury is the main side effect caused by cyclophosphamide and the disruption of the intestinal barrier may be an important cause. Yupingfeng granules have been reported to have immunomodulatory effects and polysaccharides are important components of them. This study aimed to investigate the ameliorative effect of polysaccharides from Yupingfeng granules (YPFP) on cyclophosphamide induced immune injury and reveal their potential mechanisms based on its protective effect on the intestine. YPFP were isolated and preliminarily characterized. Pharmacodynamic evaluation revealed that YPFP treatment could effectively mitigate lesions of immune organs, ameliorate white blood cells and downregulate IL-10 level. Further, the protective effect of intestinal barrier on the basis of intestinal tight junctions, MUC-2, microflora, endogenous metabolites, pathways and immune cells was discussed to outline mechanisms. The results showed that YPFP repaired the integrity of intestinal epithelium, enhanced the abundance of Muribaculaceae_unclassified, Bacteroide and Muribaculum, downgraded the abundance of Lachnospiraceae_NK4A136_group, improved the excretion of lipids and bile acids especially 3-oxo-LCA, increased the content of SCFAs in feces and inhibited the expression of key proteins of PI3K-AKT and MAPK-JUN pathways. More importantly, Th17 and Treg balance was remodeled after YPFP administration, which might be related to certain differential metabolites and pathways enriched by metabolomics. This study provides a rich understanding of YPFP and lays a foundation for further development of Yupingfeng granules. It was shown for the first time that the immunomodulatory effect of YPFP might be involved in multiple mechanisms of intestinal homeostasis. YPFP could be regarded as an immunomodulator to alleviate immune damage caused by cyclophosphamide.
Collapse
Affiliation(s)
- Leyi Huang
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201201, China; National Key Laboratory of Lead Druggability Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Mo Liu
- National Key Laboratory of Lead Druggability Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Longhai Shen
- National Key Laboratory of Lead Druggability Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Daofeng Chen
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201201, China.
| | - Tong Wu
- National Key Laboratory of Lead Druggability Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, China.
| | - Yongjian Gao
- Sinopharm Group Guangdong Medi-World Pharmaceutical Co., Ltd., Guangzhou, China
| |
Collapse
|
46
|
Zhang Q, Liang D, Zhang C, Ye L, Sun P, Zhu H, Zhao Y, Li Y, Guan Y, Zhang H. Integrated Microbiome and Metabolome Analysis Reveals Correlations Between Gut Microbiota Components and Metabolic Profiles in Mice With Mitoxantrone-Induced Cardiotoxicity. Drug Des Devel Ther 2025; 19:439-455. [PMID: 39867867 PMCID: PMC11766154 DOI: 10.2147/dddt.s479682] [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: 07/27/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Purpose Mitoxantrone (MTX) is largely restricted in clinical usage due to its significant cardiotoxicity. Multiple studies have shown that an imbalance in the gut-heart axis plays an important role in the development of cardiovascular disease (CVD). We aim to explore the possible correlations between gut microbiota (GM) compositions and cardiometabolic (CM) disorder in MTX-triggered cardiotoxicity mice. Methods MTX cumulative dose of 6 mg/kg was administered to healthy Kunming male mice to trigger cardiotoxicity, with 1 mg/kg twice weekly for a duration of 3 weeks. Plasma CK-MB and LDH levels were determined, and the heart tissue histopathology was assessed, followed by utilizing an integrated liquid chromatography-mass spectrometry (LC-MS)-based heart metabolomics study alongside the 16S ribosomal RNA (rRNA) sequencing method to assess MTX impact on GM and CM profiles in mice, establishing associations between GM and CM profiles through the Pearson correlation coefficient calculation. Results MTX caused CK-MB and LDH level elevations and cardiotoxicity in our mouse model. MTX primarily affected the processes of protein digestion and absorption, mineral absorption, membrane transport, production of aminoacyl-transfer RNA (tRNA), metabolism of nucleotides, lipids, and amino acids, as well as autophagy. Additionally, MTX increased Romboutsia, Enterococcus, and Turicibacter abundances and lowered norank_f__Muribaculaceae, Alistipes, Odoribacter, norank_f__Lachnospiraceae, norank_f__Ruminococcaceae, norank_f__Oscillospiraceae, unclassified_f__Ruminococcaceae, NK4A214_group, Colidextribacter, norank_f__norank_o__Clostridia_vadinBB60_group, Rikenella, and Anaerotruncus abundances. The correlation analyses showcased variations in the abundance of diverse flora, such as Romboutsia, Enterococcus, Turicibacter, and norank_f__Muribaculaceae, which were related to MTX-induced cardiac injury. Conclusion Our study supports the claim that MTX provokes cardiotoxicity by modifying CM and GM profiles. Our results offer new possibilities for controlling MTX-triggered cardiotoxicity.
Collapse
Affiliation(s)
- Qing Zhang
- College of Clinical Medicine, Jining Medical University, Jining, 272013, People’s Republic of China
| | - Deshuai Liang
- Department of Pharmacy, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Chengfang Zhang
- Department of Clinical Laboratory, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Ling Ye
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Ping Sun
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Hongli Zhu
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Yongqin Zhao
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Yuewen Li
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Yun Guan
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Haiguo Zhang
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| |
Collapse
|
47
|
Sharma SA, Oladejo SO, Kuang Z. Chemical interplay between gut microbiota and epigenetics: Implications in circadian biology. Cell Chem Biol 2025; 32:61-82. [PMID: 38776923 PMCID: PMC11569273 DOI: 10.1016/j.chembiol.2024.04.016] [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: 12/15/2023] [Revised: 03/22/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Circadian rhythms are intrinsic molecular mechanisms that synchronize biological functions with the day/night cycle. The mammalian gut is colonized by a myriad of microbes, collectively named the gut microbiota. The microbiota impacts host physiology via metabolites and structural components. A key mechanism is the modulation of host epigenetic pathways, especially histone modifications. An increasing number of studies indicate the role of the microbiota in regulating host circadian rhythms. However, the mechanisms remain largely unknown. Here, we summarize studies on microbial regulation of host circadian rhythms and epigenetic pathways, highlight recent findings on how the microbiota employs host epigenetic machinery to regulate circadian rhythms, and discuss its impacts on host physiology, particularly immune and metabolic functions. We further describe current challenges and resources that could facilitate research on microbiota-epigenetic-circadian rhythm interactions to advance our knowledge of circadian disorders and possible therapeutic avenues.
Collapse
Affiliation(s)
- Samskrathi Aravinda Sharma
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Sarah Olanrewaju Oladejo
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Zheng Kuang
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
48
|
Xu Q, Wang W, Li Y, Cui J, Zhu M, Liu Y, Liu Y. The oral-gut microbiota axis: a link in cardiometabolic diseases. NPJ Biofilms Microbiomes 2025; 11:11. [PMID: 39794340 PMCID: PMC11723975 DOI: 10.1038/s41522-025-00646-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
The oral-gut microbiota axis plays a crucial role in cardiometabolic health. This review explores the interactions between these microbiomes through enteric, hematogenous, and immune pathways, resulting in disruptions in microbial balance and metabolic processes. These disruptions contribute to systemic inflammation, metabolic disorders, and endothelial dysfunction, which are closely associated with cardiometabolic diseases. Understanding these interactions provides insights for innovative therapeutic strategies to prevent and manage cardiometabolic diseases.
Collapse
Affiliation(s)
- Qian Xu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China
| | - Wenting Wang
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China
| | - Yiwen Li
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China
| | - Jing Cui
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China
| | - Mengmeng Zhu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China
| | - Yanfei Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China
| | - Yue Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China.
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, 100091, Beijing, China.
| |
Collapse
|
49
|
Abdulrahim AO, Doddapaneni NSP, Salman N, Giridharan A, Thomas J, Sharma K, Abboud E, Rochill K, Shreelakshmi B, Gupta V, Lakkimsetti M, Mowo-Wale A, Ali N. The gut-heart axis: a review of gut microbiota, dysbiosis, and cardiovascular disease development. Ann Med Surg (Lond) 2025; 87:177-191. [PMID: 40109640 PMCID: PMC11918638 DOI: 10.1097/ms9.0000000000002789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/20/2024] [Indexed: 03/22/2025] Open
Abstract
Background Cardiovascular diseases (CVDs) are a major cause of morbidity and mortality worldwide and there are strong links existing between gut health and cardiovascular health. Gut microbial diversity determines gut health. Dysbiosis, described as altered gut microbiota, causes bacterial translocations and abnormal gut byproducts resulting in systemic inflammation. Objective To review the current literature on the relationships between gut microbiota, dysbiosis, and CVD development, and explore therapeutic methods to prevent dysbiosis and support cardiovascular health. Summary Dysbiosis increases levels of pro-inflammatory substances while reducing those of anti-inflammatory substances. This accumulative inflammatory effect negatively modulates the immune system and promotes vascular dysfunction and atherosclerosis. High Firmicutes to Bacteroidetes ratios, high trimethylamine-n-oxide to short-chain fatty acid ratios, high indole sulfate levels, low cardiac output, and polypharmacy are all associated with worse cardiovascular outcomes. Supplementation with prebiotics and probiotics potentially alleviates some CVD risk. Blood and stool samples may be used in clinical practice to quantify and qualify gut bacterial ratios and byproducts, assess patients' risk for adverse cardiovascular outcomes, and track their gut health progress. Further research is required to set population-based cutoffs for normal and abnormal gut microbiota and byproduct ratios.
Collapse
Affiliation(s)
| | | | - Nadhra Salman
- Department of Internal Medicine, Baqai Medical University, Karachi, Pakistan
| | | | | | - Kavya Sharma
- Maharishi Markandeshwar Medical College and Hospital, Himachal Pradesh, India
| | - Elias Abboud
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | | | - B Shreelakshmi
- Navodaya Medical College Hospital & Research Centre, Karnataka, India
| | | | | | | | - Noor Ali
- Dubai Medical College, Dubai, United Arab Emirates
| |
Collapse
|
50
|
Billingsley HE, Carbone S, Driggin E, Kitzman DW, Hummel SL. Dietary Interventions in Heart Failure With Preserved Ejection Fraction: A Scoping Review. JACC. ADVANCES 2025; 4:101465. [PMID: 39801812 PMCID: PMC11719370 DOI: 10.1016/j.jacadv.2024.101465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025]
Abstract
Patients with heart failure with preserved ejection fraction (HFpEF) are burdened by multiple diet-sensitive comorbidities, including obesity and malnutrition. Despite this, a low percentage of patients with HFpEF have been enrolled in dietary intervention trials in heart failure and few dietary interventions have been conducted in HFpEF exclusively. This scoping review will examine available evidence regarding dietary interventions in patients with HFpEF, highlight existing gaps in knowledge, and discuss emerging dietary therapies in this population.
Collapse
Affiliation(s)
- Hayley E. Billingsley
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Salvatore Carbone
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
- Virginia Commonwealth University Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Elissa Driggin
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
| | - Dalane W. Kitzman
- Sections on Cardiovascular Medicine and Geriatrics and Gerontology, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Scott L. Hummel
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Cardiology, Veteran’s Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|