1
|
Song R, Yin X, Zhu M, Chen X, Zhang J, Liu D, Wang S, Jiang S, Liu Z, Wang L, Feng K, Li Y. Innovative application of confocal Raman spectroscopy and Machine learning in cardiovascular diseases identification. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 340:126343. [PMID: 40347774 DOI: 10.1016/j.saa.2025.126343] [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: 02/24/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
Myocardial hypertrophy and heart failure are leading causes of mortality in cardiovascular diseases, yet current diagnostic techniques lack the resolution to monitor molecular changes effectively. In this study, we employed confocal Raman spectroscopy combined with machine learning to evaluate myocardial tissue in a transverse aortic constriction (TAC) mouse model. Mice were divided into three groups: control (CON), myocardial hypertrophy (TAC 2 W), and heart failure (TAC 4 W). The model was validated using echocardiography, histopathology, and transmission electron microscopy. Raman spectroscopy revealed significant changes in chemical composition, including decreased peak intensities at 748, 1309, 1584, 1170, 1359, 1222, 1636, 1335, and 1393 cm-1, increased intensity at 2847 cm-1, and a rightward shift of the peak at 2927 cm-1, which correlated with disease progression. Machine learning analysis demonstrated that the random forest model achieved 85 % accuracy in classifying normal, hypertrophic, and failing myocardial tissues. This study highlights the potential of confocal Raman spectroscopy combined with machine learning for the identification and real-time monitoring of cardiovascular diseases, offering a novel approach to understanding disease mechanisms and improving patient outcomes. Although the model achieved high classification accuracy, the small sample size may limit generalizability and requires further validation in larger cohorts.
Collapse
Affiliation(s)
- Renxing Song
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Xunyi Yin
- The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Manlin Zhu
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Xinyu Chen
- Department of Organic Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Jingqi Zhang
- The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Dongmei Liu
- The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Shimei Wang
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Shuang Jiang
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Zhehan Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Lin Wang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118 Guangdong, China
| | - Kun Feng
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118 Guangdong, China.
| | - Yang Li
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin 150081 Heilongjiang, China; Research Unit of Health Sciences and Technology (HST), Faculty of Medicine University of Oulu 90220, Finland; The Fourth Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150081 Heilongjiang, China; Heilongjiang Eye Hospital, Harbin, China.
| |
Collapse
|
2
|
He X, Zhang J, Zhang Y, Li H, Chen Y, Zhang H, Pan J, Zhou Y, Zhang S, Cheng L. L-Kynurenine regulates immune response in ICIs-associated myocarditis via JAK/STAT pathway. Int Immunopharmacol 2025; 156:114676. [PMID: 40267722 DOI: 10.1016/j.intimp.2025.114676] [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: 02/05/2025] [Revised: 03/24/2025] [Accepted: 04/12/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Immune checkpoint inhibitor associated myocarditis (ICIAM) is a drug-induced myocarditis characterized by the infiltration of immune cells into cardiac. However, the mechanisms are unknown and effective drug therapies are lacking in clinical practice. This study aims to explore the relationship between plasma metabolites and the treatment of ICIAM. METHODS Human plasma metabolites were analyzed using untargeted metabolomics for characteristic metabolites. For in vivo experiments, Male Balb/c mice were divided into six groups: PBS, L-Kynurenine, cTNI+PD-1 inhibitor (ICIs), ICIs+L-Kynurenine, ICIs+RO8191, ICIs+RO8191+L-Kynurenine. On day 21 post-modeling, echocardiography, ELISA and histopathology were employed to evaluate the therapeutic effect of L-Kynurenine. Flow cytometry was used to determine the proportion of immune cells in the heart and spleen. Bulk-RNAseq was conducted to analyze differential genes, and q-PCR, immunofluorescence and western blot were performed for validation experiments. RESULTS Untargeted metabolomics verified that indoleamine 2,3 dioxygenase-1 (IDO1)-derived L-Kynurenine level was higher in the serum of ICIAM compared to non-ICIAM patients. Meanwhile, in vitro and in vivo experiments showed that L-Kynurenine exhibited a therapeutic ability in ICIAM by inhibiting the pro-inflammatory polarization of immune cells and the secretion of pro-inflammatory cytokines. Mechanistically, L-Kynurenine improved cardiac functions majorly by the inhibition of the JAK1/STAT3 signaling pathway. CONCLUSION L-Kynurenine exhibits significant therapeutic potential in ICIAM. The multi-roles of L-Kynurenine in regulating immune responses make it possible to be used as a targeted drug for ICIAM therapy.
Collapse
Affiliation(s)
- Xiaozhen He
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Jian Zhang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Yerui Zhang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Huishan Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Chen
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Hui Zhang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Jianan Pan
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China
| | - Yan Zhou
- State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shilong Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Leilei Cheng
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
3
|
Lai C, Li Y, Luo W, Zhang B, Liu C, Peng L, Li H, Liu JE, Xiao X, Zhong S. Plasma metabolomics differentiating and predicting prognosis of coronary artery disease patients with distinct nutritional status. Nutr Res 2025; 137:1-13. [PMID: 40188579 DOI: 10.1016/j.nutres.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] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 04/08/2025]
Abstract
This study investigated the metabolic mechanisms underlying the association between malnutrition and poor prognosis in coronary artery disease (CAD). We hypothesized that specific metabolites associated with nutritional status impact all-cause mortality and Major Adverse Cardiovascular Events in CAD patients. To test this hypothesis, we evaluated the nutritional status of 5182 CAD patients from multiple centers using three nutritional risk screening tools and analyzed the impact on CAD outcomes with restricted cubic splines and Cox regression. Poor nutritional status was found to be linked to increased adverse outcomes. Further analysis using multiple linear regression and mediation analysis identified elevated concentrations of β-pseudouridine and dulcitol, and decreased concentrations of l-tryptophan and LPC (18:2/0:0), among other metabolites, as mediators of this association. Employing Least Absolute Shrinkage and Selection Operator for variable selection, we integrated these metabolites with clinical variables, which significantly improved the predictive accuracy for adverse outcomes. Our results highlight significant metabolic disparities in CAD patients based on nutritional status and provide novel insights into the role of nutrition-associated metabolites in CAD prognosis. These findings suggest that customized nutritional interventions targeting these metabolites could positively influence the progression of CAD.
Collapse
Affiliation(s)
- Chengyang Lai
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yangchen Li
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Wenwei Luo
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Bin Zhang
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Chen Liu
- Department of Cardiology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liming Peng
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Hanping Li
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ju-E Liu
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao Xiao
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shilong Zhong
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China; Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
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
|
5
|
Mohiti S, Christensen J, Landler NE, Sørensen IM, Thomassen JQ, Bjergfelt SS, Hansen D, Feldt-Rasmussen B, Bro S, Ebrahimi-Mameghani M, Biering-Sørensen T, Bisgaard LS, Christoffersen C. Serum tryptophan and kynurenine levels and risk of heart failure among patients with chronic kidney disease. Clin Nutr 2025; 47:14-20. [PMID: 39978230 DOI: 10.1016/j.clnu.2025.01.028] [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: 08/12/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND AND AIMS Chronic kidney disease (CKD) is often complicated by heart failure (HF), leading to increased mortality. Emerging evidence suggests that Tryptophan metabolites, through the Kynurenine pathway (KP), play a significant role in HF pathophysiology. Therefore, we explored the association of Tryptophan (TRP), Kynurenine (KYN), and the Kynurenine to Tryptophan ratio (KTR) with HF in CKD, hypothesizing a link between KP alterations and HF occurrence in this population. METHODS 673 non-dialysis patients aged 30 to 75 with CKD stages 1-5 were included. Incident HF data were collected through medical record reviews, and the median follow-up time was 3.9 years. Serum concentrations of KYN and TRP were measured using High-Performance Liquid Chromatography (HPLC). RESULTS Patients with more advanced stages of CKD had higher levels of KYN and KTR, and lower levels of TRP (p < 0.001). Following adjustments for age, sex, BMI, hypertension, and hypercholesterolemia, serum KYN and KTR remained significantly associated with prevalent HF in patients with CKD (p = 0.012, p = 0.028 respectively). Furthermore, Cox-regression analysis indicated that KTR concentration was associated with incident HF after adjusting for confounders such as age, sex, BMI, hypertension, hypercholesterolemia and diabetes (p = 0.019). CONCLUSION In conclusion, the present analysis suggests that changes in the kynurenine pathway may be a new biomarker for HF in patients with CKD. Thus, KTR concentration might be associated with prevalent and future HF in patients with CKD. Further research is needed to understand the mechanisms and potential of these metabolites in refining HF risk prediction and prevention in CKD patients.
Collapse
Affiliation(s)
- Sara Mohiti
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Student Research Committee, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jacob Christensen
- Center for Translational Cardiology and Pragmatic Randomized Trials (CTCPR), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Cardiovascular Non-Invasive Imaging Research Laboratory, Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | - Nino E Landler
- Center for Translational Cardiology and Pragmatic Randomized Trials (CTCPR), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Cardiovascular Non-Invasive Imaging Research Laboratory, Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Denmark
| | - Ida Mh Sørensen
- Department of Nephrology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Jesper Qvist Thomassen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Sasha S Bjergfelt
- Department of Nephrology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Ditte Hansen
- Department of Nephrology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Bo Feldt-Rasmussen
- Department of Nephrology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Susanne Bro
- Department of Nephrology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Mehrangiz Ebrahimi-Mameghani
- Nutrition Research Center, Department of Biochemistry and Diet Therapy, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tor Biering-Sørensen
- Center for Translational Cardiology and Pragmatic Randomized Trials (CTCPR), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Cardiovascular Non-Invasive Imaging Research Laboratory, Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Denmark; Steno Diabetes Center Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Denmark
| | - Line S Bisgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Denmark.
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
6
|
Wang Y, Wu P, Chen Z, Li Z, Wang Y, Yan M, Zhang Y, Wang S, Fang S, Yu B. Prognostic value of tryptophan catabolism-base scores in acute myocardial infarction patients. J Adv Res 2025:S2090-1232(25)00185-7. [PMID: 40147625 DOI: 10.1016/j.jare.2025.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
AIMS Tryptophan catabolism is implicated in the progression of cardiovascular disease. We sought to investigate the prognostic value of tryptophan catabolism-related features in patients with acute myocardial infarction (AMI). METHODS AND RESULTS A prospective cohort of 4071 patients (mean age: 60.7 years; 69.1 % men) with AMI between February 2017 and June 2019 was included and followed up for a median of 5.6 years (IQR 5.1-6.2). There were 666 all-cause deaths, 365 cardiovascular deaths, and 559 HF events. Plasma levels of tryptophan-related metabolites were measured using liquid chromatography tandem mass spectrometry (LC-MS/MS), and were repeatedly determined in 1044 patients after discharge. Tryptophan, kynurenine, indole-3-propionic acid, and indole-3-lactic acid were screened to construct tryptophan metabolites combination (TMC) score using coefficients from predictive models for MACE. Patients were divided into 3 groups by TMC tertiles. Patients with higher TMC score were older, more likely to be male and have hypertension. Compared to those with TMC tertile 1, patients in TMC tertile 3 had significant associations with the risk of all-cause death (HR: 1.90; 95 %CI: 1.54-2.34), cardiovascular death (HR: 2.32; 95 %CI: 1.71-3.15) and incident HF (HR: 1.77; 95 %CI: 1.40-2.24). The incremental prognostic value of TMC score over the Grace score was measured by the likelihood ratio, C-statistic, continuous net reclassification improvement (NRI), and integrated discrimination improvement (IDI) for prediction, discrimination, and reclassification of outcomes. CONCLUSIONS In this hospital-based AMI cohort, the TMC score was significantly associated with all-cause mortality, cardiovascular mortality, and incident HF, and improved risk stratification beyond established clinical risk factors. The TMC score provided a novel tool for assessment of Trp catabolism dysfunction and outcomes risk.
Collapse
Affiliation(s)
- Ye Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Pengyan Wu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Zhanchao Chen
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Zhaoying Li
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Yini Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China
| | - Miao Yan
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China
| | - Yiying Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jiamusi University, Jiamusi, China
| | - Shanjie Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China.
| | - Shaohong Fang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China.
| | - Bo Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), China; Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, China.
| |
Collapse
|
7
|
Zhou S, Zhou X, Zhang P, Zhang W, Huang J, Jia X, He X, Sun X, Su H. The gut microbiota-inflammation-HFpEF axis: deciphering the role of gut microbiota dysregulation in the pathogenesis and management of HFpEF. Front Cell Infect Microbiol 2025; 15:1537576. [PMID: 40182777 PMCID: PMC11965942 DOI: 10.3389/fcimb.2025.1537576] [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: 12/01/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Heart failure with preserved left ventricular ejection fraction (HFpEF) is a disease that affects multiple organs throughout the body, accounting for over 50% of heart failure cases. HFpEF has a significant impact on individuals' life expectancy and quality of life, but the exact pathogenesis remains unclear. Emerging evidence implicates low-grade systemic inflammation as a crucial role in the onset and progression of HFpEF. Gut microbiota dysregulation and associated metabolites alteration, including short-chain fatty acids, trimethylamine N-oxides, amino acids, and bile acids can exacerbate chronic systemic inflammatory responses and potentially contribute to HFpEF. In light of these findings, we propose the hypothesis of a "gut microbiota-inflammation-HFpEF axis", positing that the interplay within this axis could be a crucial factor in the development and progression of HFpEF. This review focuses on the role of gut microbiota dysregulation-induced inflammation in HFpEF's etiology. It explores the potential mechanisms linking dysregulation of the gut microbiota to cardiac dysfunction and evaluates the therapeutic potential of restoring gut microbiota balance in mitigating HFpEF severity. The objective is to offer novel insights and strategies for the management of HFpEF.
Collapse
Affiliation(s)
- Shenghua Zhou
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xuan Zhou
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Wei Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Jinli Huang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xuzhao Jia
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xiaole He
- Department of General Practice, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| |
Collapse
|
8
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2025; 78:263-271. [PMID: 39357800 PMCID: PMC11875914 DOI: 10.1016/j.rec.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
9
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. Rev Esp Cardiol 2025; 78:263-271. [PMID: 39357800 DOI: 10.1016/j.recesp.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 01/11/2025]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
10
|
Mitu F, Adam CA, Richter P, Costache AD, Gavril RS, Cojocaru C, Țăruș A, Enache M, Cumpăt CM, Leon MM, Tinică G. Pericardial Fluid Biomarkers as Early Predictors for Postoperative Atrial Fibrillation-A Systematic Review. Diagnostics (Basel) 2025; 15:408. [PMID: 40002559 PMCID: PMC11854266 DOI: 10.3390/diagnostics15040408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
(1) Background: Postoperative atrial fibrillation (POAF) is one of the most common complications of cardiac surgery, frequently occurring in the first 2-4 days after surgery. With a variable incidence depending on the type of surgery, research in recent years has focused on identifying predisposing factors with the aim of correcting them and thus decreasing the risk of cardiovascular and total morbidity and mortality. The analysis of pericardial fluid allowed the identification of biomarkers (interleukin-6, mitochondrial DNA, myeloperoxidase or natriuretic peptides) whose presence postoperatively was associated with increased risk of POAF. (2) Materials and Methods: We conducted a search on EMBASE and PubMed and identified 75 articles, of which 10 entered the final analysis. (3) Results: Patients who develop POAF accumulate large amounts of interleukin 6, mitochondrial DNA, myeloperoxidase, or secondary atrial natriuretic peptide as a consequence of the associated inflammatory status, atrial remodeling, or disturbance of homeostasis of various ions. There are also observations that their levels in the pericardium correlate with blood levels, but further studies on larger cohorts of patients are needed to provide new evidence in this regard. (4) Conclusions: Early recognition of patients at risk of developing POAF based on easy-to-dose and easy-to-use biochemical biomarkers, whose association with POAF has been demonstrated so far in small cohorts of patients, has both therapeutic and prognostic implications, which justifies further research on large cohorts of patients.
Collapse
Affiliation(s)
- Florin Mitu
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
- Academy of Medical Sciences, 030167 Bucharest, Romania
- Academy of Romanian Scientists, 700050 Iasi, Romania
| | - Cristina Andreea Adam
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Patricia Richter
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Rheumatology Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Alexandru-Dan Costache
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Radu Sebastian Gavril
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Clementina Cojocaru
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Andrei Țăruș
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Surgery Clinic, “Prof. Dr. George I. M. Georgescu” Cardiovascular Diseases Institute, 700503 Iasi, Romania
| | - Mihail Enache
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Surgery Clinic, “Prof. Dr. George I. M. Georgescu” Cardiovascular Diseases Institute, 700503 Iasi, Romania
| | - Carmen Marinela Cumpăt
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Maria Magdalena Leon
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Grigore Tinică
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Academy of Medical Sciences, 030167 Bucharest, Romania
- Academy of Romanian Scientists, 700050 Iasi, Romania
- Cardiovascular Surgery Clinic, “Prof. Dr. George I. M. Georgescu” Cardiovascular Diseases Institute, 700503 Iasi, Romania
| |
Collapse
|
11
|
Yin Z, Fu L, Wang Y, Tai S. Impact of gut microbiota on cardiac aging. Arch Gerontol Geriatr 2025; 128:105639. [PMID: 39312851 DOI: 10.1016/j.archger.2024.105639] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024]
Abstract
Recent research has suggested imbalances in gut microbiota composition as contributors to cardiac aging. An individual's physical condition, along with lifestyle-associated factors, including diet and medication, are significant determinants of gut microbiota composition. This review discusses evidence of bidirectional associations between aging and gut microbiota, identifying gut microbiota-derived metabolites as potential regulators of cardiac aging. It summarizes the effects of gut microbiota on cardiac aging diseases, including cardiac hypertrophy and fibrosis, heart failure, and atrial fibrillation. Furthermore, this review discusses the potential anti-aging effects of modifying gut microbiota composition through dietary and pharmacological interventions. Lastly, it underscores critical knowledge gaps and outlines future research directions. Given the current limited understanding of the direct relationship between gut microbiota and cardiac aging, there is an urgent need for preclinical and clinical investigations into the mechanistic interactions between gut microbiota and cardiac aging. Such endeavors hold promise for shedding light on the pathophysiology of cardiac aging and uncovering new therapeutic targets for cardiac aging diseases.
Collapse
Affiliation(s)
- Zhiyi Yin
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China
| | - Liyao Fu
- Hunan Key Laboratory of Cardiometabolic Medicine, Department of Cardiology, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China
| | - Yongjun Wang
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China.
| | - Shi Tai
- Hunan Key Laboratory of Cardiometabolic Medicine, Department of Cardiology, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Changsha, Hunan 410011, China.
| |
Collapse
|
12
|
Rohun J, Dudzik D, Raczak-Gutknecht J, Wabich E, Młodziński K, Markuszewski MJ, Daniłowicz-Szymanowicz L. Metabolomics in Atrial Fibrillation: Unlocking Novel Biomarkers and Pathways for Diagnosis, Prognosis, and Personalized Treatment. J Clin Med 2024; 14:34. [PMID: 39797116 PMCID: PMC11722095 DOI: 10.3390/jcm14010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Atrial fibrillation (AF) is the most frequent arrhythmia in the adult population associated with a high rate of severe consequences leading to significant morbidity and mortality worldwide. Therefore, its prompt recognition is of high clinical importance. AF detection often remains challenging due to unspecific symptoms and a lack of reliable biomarkers for its prediction. Herein, novel bioanalytical methodologies, such as metabolomics, offer new opportunities for a better understanding of the underlying pathological mechanisms of cardiovascular diseases, including AF. The metabolome, considered a complete set of small molecules present in the organism, directly reflects the current phenotype of the studied system and is highly sensitive to any changes, including arrhythmia's onset. A growing body of evidence suggests that metabolite profiling has prognostic value in AF prediction, highlighting its potential role not only in early diagnosis but also in guiding therapeutic interventions. By identifying specific metabolites as a disease biomarker or recognising particular metabolomic pathways involved in the AF pathomechanisms, metabolomics could be of great clinical value for further clinical decision-making, risk stratification, and an individual personalised approach. The presented narrative review aims to summarise the current state of knowledge on metabolomics in AF with a special emphasis on its implications for clinical practice and personalised medicine.
Collapse
Affiliation(s)
- Justyna Rohun
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Danuta Dudzik
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Joanna Raczak-Gutknecht
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Elżbieta Wabich
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Krzysztof Młodziński
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| | - Michał J. Markuszewski
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdańsk, Poland; (D.D.); (J.R.-G.); (M.J.M.)
| | - Ludmiła Daniłowicz-Szymanowicz
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland; (J.R.); (E.W.); (K.M.)
| |
Collapse
|
13
|
Ajoolabady A, Pratico D, Dunn WB, Lip GYH, Ren J. Metabolomics: Implication in cardiovascular research and diseases. Obes Rev 2024; 25:e13825. [PMID: 39370721 DOI: 10.1111/obr.13825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 10/08/2024]
Abstract
Cellular metabolism influences all aspects of cellular function and is crucial for overall organismal health. Metabolic disorders related to cardiovascular health can lead to cardiovascular diseases (CVDs). Moreover, associated comorbidities may also damage cardiovascular metabolism, exacerbating CVD and perpetuating a vicious cycle. Given the prominent role of metabolic alterations in CVD, metabolomics has emerged as an imperative technique enabling a comprehensive assessment of metabolites and metabolic architecture within the body. Metabolite profile and metabolic pathways help to deepen and broaden our understanding of complex genomic landscape and pathophysiology of CVD. Here in this review, we aim to overview the experimental and clinical applications of metabolomics in pathogenesis, diagnosis, prognosis, and management of various CVD plus future perspectives and limitations.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Warwick B Dunn
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| |
Collapse
|
14
|
Rovira J, Ramirez-Bajo MJ, Bañon-Maneus E, Ventura-Aguiar P, Arias-Guillén M, Romano-Andrioni B, Ojeda R, Revuelta I, García-Calderó H, Barberà JA, Dantas AP, Diaz-Ricart M, Crispi F, García-Pagán JC, Campistol JM, Diekmann F. Mediterranean Diet Pattern: Potential Impact on the Different Altered Pathways Related to Cardiovascular Risk in Advanced Chronic Kidney Disease. Nutrients 2024; 16:3739. [PMID: 39519573 PMCID: PMC11547550 DOI: 10.3390/nu16213739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) remains the most common cause of mortality in chronic kidney disease (CKD) patients. Several studies suggest that the Mediterranean diet reduces the risk of CVD due to its influence on endothelial function, inflammation, lipid profile, and blood pressure. Integrating metabolomic and proteomic analyses of CKD could provide insights into the pathways involved in uremia-induced CVD and those pathways modifiable by the Mediterranean diet. METHODS We performed metabolomic and proteomic analyses on serum samples from 19 patients with advanced CKD (aCKD) and 27 healthy volunteers. The metabolites were quantified using four different approaches, based on their properties. Proteomic analysis was performed after depletion of seven abundant serum proteins (Albumin, IgG, antitrypsin, IgA, transferrin, haptoglobin, and fibrinogen). Integrative analysis was performed using MetaboAnalyst 4.0 and STRING 11.0 software to identify the dysregulated pathways and biomarkers. RESULTS A total of 135 metabolites and 75 proteins were differentially expressed in aCKD patients, compared to the controls. Pathway enrichment analysis showed significant alterations in the innate immune system pathways, including complement, coagulation, and neutrophil degranulation, along with disrupted linoleic acid and cholesterol metabolism. Additionally, certain key metabolites and proteins were altered in aCKD patients, such as glutathione peroxidase 3, carnitine, homocitrulline, 3-methylhistidine, and several amino acids and derivatives. CONCLUSIONS Our findings reveal significant dysregulation of the serum metabolome and proteome in aCKD, particularly in those pathways associated with endothelial dysfunction and CVD. These results suggest that CVD prevention in CKD may benefit from a multifaceted approach, including dietary interventions such as the Mediterranean diet.
Collapse
Affiliation(s)
- Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomètiques August Pi i Sunyer (IDIBAPS), 08027 Barcelona, Spain; (M.J.R.-B.); (E.B.-M.); (P.V.-A.); (M.A.-G.); (I.R.); (J.M.C.)
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), 28029 Madrid, Spain
| | - María José Ramirez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomètiques August Pi i Sunyer (IDIBAPS), 08027 Barcelona, Spain; (M.J.R.-B.); (E.B.-M.); (P.V.-A.); (M.A.-G.); (I.R.); (J.M.C.)
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), 28029 Madrid, Spain
| | - Elisenda Bañon-Maneus
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomètiques August Pi i Sunyer (IDIBAPS), 08027 Barcelona, Spain; (M.J.R.-B.); (E.B.-M.); (P.V.-A.); (M.A.-G.); (I.R.); (J.M.C.)
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), 28029 Madrid, Spain
| | - Pedro Ventura-Aguiar
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomètiques August Pi i Sunyer (IDIBAPS), 08027 Barcelona, Spain; (M.J.R.-B.); (E.B.-M.); (P.V.-A.); (M.A.-G.); (I.R.); (J.M.C.)
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), 28029 Madrid, Spain
- Department of Nephrology and Kidney Transplantation, Clínic’s Institute of Nephrology and Urology (ICNU), Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain; (B.R.-A.); (R.O.)
| | - Marta Arias-Guillén
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomètiques August Pi i Sunyer (IDIBAPS), 08027 Barcelona, Spain; (M.J.R.-B.); (E.B.-M.); (P.V.-A.); (M.A.-G.); (I.R.); (J.M.C.)
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), 28029 Madrid, Spain
- Department of Nephrology and Kidney Transplantation, Clínic’s Institute of Nephrology and Urology (ICNU), Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain; (B.R.-A.); (R.O.)
| | - Barbara Romano-Andrioni
- Department of Nephrology and Kidney Transplantation, Clínic’s Institute of Nephrology and Urology (ICNU), Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain; (B.R.-A.); (R.O.)
| | - Raquel Ojeda
- Department of Nephrology and Kidney Transplantation, Clínic’s Institute of Nephrology and Urology (ICNU), Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain; (B.R.-A.); (R.O.)
| | - Ignacio Revuelta
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomètiques August Pi i Sunyer (IDIBAPS), 08027 Barcelona, Spain; (M.J.R.-B.); (E.B.-M.); (P.V.-A.); (M.A.-G.); (I.R.); (J.M.C.)
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), 28029 Madrid, Spain
- Department of Nephrology and Kidney Transplantation, Clínic’s Institute of Nephrology and Urology (ICNU), Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain; (B.R.-A.); (R.O.)
| | - Héctor García-Calderó
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic_Clínic Barcelona, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN-RareLiver), Department of Medicine and Health Sciences, University of Barcelona, CSUR_EVH, 08036 Barcelona, Spain; (H.G.-C.); (J.C.G.-P.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), 28200 Madrid, Spain
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain;
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), 30627 Madrid, Spain
| | - Ana Paula Dantas
- Cardiovascular Institute, Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08007 Barcelona, Spain;
| | - Maribel Diaz-Ricart
- Hematopathology, Centre Diagnòstic Biomèdic (CDB), Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08007 Barcelona, Spain;
- Barcelona Endothelium Team (BET), 08036 Barcelona, Spain
| | - Fàtima Crispi
- BCNatal|Fetal Medicine Research Center, Hospital Clínic and Hospital Sant Joan de Déu, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08007 Barcelona, Spain;
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), 28029 Madrid, Spain
| | - Juan Carlos García-Pagán
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic_Clínic Barcelona, Institut de Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN-RareLiver), Department of Medicine and Health Sciences, University of Barcelona, CSUR_EVH, 08036 Barcelona, Spain; (H.G.-C.); (J.C.G.-P.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), 28200 Madrid, Spain
| | - Josep M. Campistol
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomètiques August Pi i Sunyer (IDIBAPS), 08027 Barcelona, Spain; (M.J.R.-B.); (E.B.-M.); (P.V.-A.); (M.A.-G.); (I.R.); (J.M.C.)
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), 28029 Madrid, Spain
- Department of Nephrology and Kidney Transplantation, Clínic’s Institute of Nephrology and Urology (ICNU), Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain; (B.R.-A.); (R.O.)
| | - Fritz Diekmann
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d’Investigacions Biomètiques August Pi i Sunyer (IDIBAPS), 08027 Barcelona, Spain; (M.J.R.-B.); (E.B.-M.); (P.V.-A.); (M.A.-G.); (I.R.); (J.M.C.)
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), 28029 Madrid, Spain
- Department of Nephrology and Kidney Transplantation, Clínic’s Institute of Nephrology and Urology (ICNU), Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036 Barcelona, Spain; (B.R.-A.); (R.O.)
| |
Collapse
|
15
|
Gawałko M, Middeldorp ME, Saljic A, Penders J, Jespersen T, Albert CM, Marcus GM, Wong CX, Sanders P, Linz D. Diet and risk of atrial fibrillation: a systematic review. Eur Heart J 2024; 45:4259-4274. [PMID: 39288159 DOI: 10.1093/eurheartj/ehae551] [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: 04/16/2024] [Revised: 06/24/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Atrial fibrillation (AF) is the most prevalent sustained cardiac arrhythmia. Comprehensive modification of established AF risk factors combined with dietary interventions and breaking deleterious habits has been shown to reduce AF burden and recurrence. Numerous AF risk factors, such as diabetes, obesity or hypertension can be partially related to dietary and lifestyle choices. Therefore, dietary interventions may have potential as a therapeutic approach in AF. Based on available data, current guidelines recommend alcohol abstinence or reduction to decrease AF symptoms, burden, and progression, and do not indicate the need for caffeine abstention to prevent AF episodes (unless it is a trigger for AF symptoms). Uncertainty persists regarding harms or benefits of other dietary factors including chocolate, fish, salt, polyunsaturated and monounsaturated fatty acids, vitamins, and micronutrients. This article provides a systematic review of the association between AF and both dietary patterns and components. Additionally, it discusses potentially related mechanisms and introduces different strategies to assess patients' nutrition patterns, including mobile health solutions and diet indices. Finally, it highlights the gaps in knowledge requiring future investigation.
Collapse
Affiliation(s)
- Monika Gawałko
- 1st Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, 1 Port Road, SA 5000 Adelaide, Australia
| | - Melissa E Middeldorp
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, 1 Port Road, SA 5000 Adelaide, Australia
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S San Vincente Blvd, AHSP 3100 Los Angeles, CA, USA
- Cardiology Department, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Arnela Saljic
- Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Christine M Albert
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S San Vincente Blvd, AHSP 3100 Los Angeles, CA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Gregory M Marcus
- Division of Cardiology, University of California San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Christopher X Wong
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, 1 Port Road, SA 5000 Adelaide, Australia
- Division of Cardiology, University of California San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, 1 Port Road, SA 5000 Adelaide, Australia
| | - Dominik Linz
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- Department of Cardiology, Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
16
|
Zhang J, Jiang X, Pang B, Li D, Kang L, Zhou T, Wang B, Zheng L, Zhou CM, Zhang L. Association between tryptophan concentrations and the risk of developing cardiovascular diseases: a systematic review and meta-analysis. Nutr Metab (Lond) 2024; 21:82. [PMID: 39407297 PMCID: PMC11476920 DOI: 10.1186/s12986-024-00857-1] [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: 07/17/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Metabolic regulation of various amino acids have been proven to be effective in preventing cardiovascular disease (CVD). The impact of tryptophan, an essential amino acid, on the risk of developing CVD has not been fully elucidated. AIMS The aim of this meta-analysis was to systematically review evidence of the effects of tryptophan on CVD risk. METHODS The PubMed, Embase, Web of Science, Cochrane Library, and China National Knowledge Infrastructure (CNKI) databases were searched to collect relevant trials from inception to August 2024. The means and hazard ratios (HRs) were extracted and pooled. Subgroup analysis was performed to identify pooled effect estimates, and sensitivity analysis was conducted to assess the robustness of the pooled estimates. RESULTS Data were collected from 34,370 people under follow-up for CVD events in 13 studies, including cohort studies and case-control studies. They were categorized into three groups on the basis of sample type and indicators: the plasma tryptophan level group, the plasma tryptophan CVD hazard group, and the urinary tryptophan CVD hazard group. The CVD included in this study were coronary artery disease, heart failure, and peripheral artery disease. Twelve studies on plasma tryptophan were meta-analyzed. The plasma tryptophan levels in CVD patients were generally lower than those in individuals without CVD (SMD = -8.57, 95%CI (-15.77, -1.37), P = 0.02). Decreased circulating tryptophan levels are associated with cardiovascular disease risk (HR = 0.85, 95%CI (0.78, 0.92), P < 0.00001). CONCLUSIONS Decreased circulating tryptophan levels are associated with an increased risk of CVD events. Intervention in circulating tryptophan levels may be indicated to help prevent CVD.
Collapse
Affiliation(s)
- Jing Zhang
- Department of General Surgery, Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xia Jiang
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bo Pang
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dongyun Li
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Longfei Kang
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tengda Zhou
- Department of General Surgery, Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Boyu Wang
- Department of General Surgery, Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihua Zheng
- Department of General Surgery, Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chuan-Min Zhou
- Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Lei Zhang
- Department of General Surgery, Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
17
|
Wang T, Ning M, Mo Y, Tian X, Fu Y, Laher I, Li S. Metabolomic Profiling Reveals That Exercise Lowers Biomarkers of Cardiac Dysfunction in Rats with Type 2 Diabetes. Antioxidants (Basel) 2024; 13:1167. [PMID: 39456421 PMCID: PMC11505272 DOI: 10.3390/antiox13101167] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
The increasing prevalence of type 2 diabetes mellitus (T2DM) leads to significant global health challenges, including cardiac structural and functional deficits, which in severe cases can progress to heart failure that can further strain healthcare resources. Aerobic exercise can ameliorate cardiac dysfunction in individuals with diabetes, although a comprehensive understanding of its underlying mechanisms remains elusive. This study utilizes untargeted metabolomics to reveal aerobic-exercise-activated metabolic biomarkers in the cardiac tissues of Sprague Dawley rats with T2DM. Metabolomics analysis revealed that diabetes altered 1029 myocardial metabolites, while aerobic exercise reversed 208 of these metabolites, of which 112 were upregulated and 96 downregulated. Pathway topology analysis suggested that these metabolites predominantly contributed to purine metabolism and arginine biosynthesis. Furthermore, receiver operating characteristic curve analysis identified 10 potential biomarkers, including xanthine, hypoxanthine, inosine, dGMP, l-glutamic acid, l-arginine, l-tryptophan, (R)-3-hydroxybutyric acid, riboflavin, and glucolepidiin. Finally, data from Pearson correlation analysis indicated that some metabolic biomarkers strongly correlated with cardiac function. Our data suggest that certain metabolic biomarkers play an important role in ameliorating diabetes-related cardiac dysfunction by aerobic exercise.
Collapse
Affiliation(s)
- Tutu Wang
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Miaomiao Ning
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Yurou Mo
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Xinyu Tian
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Yu Fu
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| | - Shunchang Li
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| |
Collapse
|
18
|
Nabil MA, Rychlik L, Nicholson A, Cheung P, Olsovsky GD, Molden J, Tripuraneni A, Hajivandi SS, Banchs JE. Dietary interventions in the management of atrial fibrillation. Front Cardiovasc Med 2024; 11:1418059. [PMID: 39149585 PMCID: PMC11324562 DOI: 10.3389/fcvm.2024.1418059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024] Open
Abstract
Atrial fibrillation (AF) represents the most common cardiac arrhythmia with significant morbidity and mortality implications. It is a common cause of hospital admissions, significantly impacts quality of life, increases morbidity and decreases life expectancy. Despite advancements in treatment options, prevalence of AF remains exceptionally high. AF is a challenging disease to manage, not just clinically but also financially. Evidence suggests lifestyle modification, including dietary changes, plays a significant role in the treatment of AF. This review aims to analyze the existing literature on the effects of dietary modifications on the incidence, progression, and outcomes of atrial fibrillation. It examines various dietary components, including alcohol, caffeine, omega-3 polyunsaturated fatty acids and minerals, and their impact on AF incidence, progression, and outcomes. The evidence surrounding the effects of dietary patterns, such as the Mediterranean and low carbohydrate diets, on AF is also evaluated. Overall, this review underscores the importance of dietary interventions as part of a comprehensive approach to AF management and highlights the need for further research in this emerging field.
Collapse
Affiliation(s)
- Muhammad Ahad Nabil
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Round Rock, TX, United States
| | - Leanne Rychlik
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Temple, TX, United States
| | - Audrey Nicholson
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Round Rock, TX, United States
| | - Peter Cheung
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Round Rock, TX, United States
| | - Gregory D Olsovsky
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Temple, TX, United States
| | - Jaime Molden
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Temple, TX, United States
| | - Ajay Tripuraneni
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Temple, TX, United States
| | - Shayan-Salehi Hajivandi
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Round Rock, TX, United States
| | - Javier E Banchs
- Department of Medicine, Division of Cardiology, Baylor Scott & White Health, Temple, TX, United States
| |
Collapse
|
19
|
Linna-Kuosmanen S, Vuori M, Kiviniemi T, Palmu J, Niiranen T. Genetics, transcriptomics, metagenomics, and metabolomics in the pathogenesis and prediction of atrial fibrillation. Eur Heart J Suppl 2024; 26:iv33-iv40. [PMID: 39099578 PMCID: PMC11292413 DOI: 10.1093/eurheartjsupp/suae072] [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] [Indexed: 08/06/2024]
Abstract
The primary cellular substrates of atrial fibrillation (AF) and the mechanisms underlying AF onset remain poorly characterized and therefore, its risk assessment lacks precision. While the use of omics may enable discovery of novel AF risk factors and narrow down the cellular pathways involved in AF pathogenesis, the work is far from complete. Large-scale genome-wide association studies and transcriptomic analyses that allow an unbiased, non-candidate-gene-based delineation of molecular changes associated with AF in humans have identified at least 150 genetic loci associated with AF. However, only few of these loci have been thoroughly mechanistically dissected, indicating that much remains to be discovered for targeted diagnostics and therapeutics. Metabolomics and metagenomics, on the other hand, add to the understanding of AF downstream of the primary substrate and integrate the signalling of environmental and host factors, respectively. These two rapidly developing fields have already provided several correlates of prevalent and incident AF that require additional validation in external cohorts and experimental studies. In this review, we take a look at the recent developments in genetics, transcriptomics, metagenomics, and metabolomics and how they may aid in improving the discovery of AF risk factors and shed light into the molecular mechanisms leading to AF onset.
Collapse
Affiliation(s)
- Suvi Linna-Kuosmanen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland
| | - Matti Vuori
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Tuomas Kiviniemi
- Department of Internal Medicine, University of Turku, Turku, Finland
- Heart Center, Turku University Hospital, Turku, Finland
| | - Joonatan Palmu
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Teemu Niiranen
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Internal Medicine, University of Turku, Turku, Finland
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Turku, Finland
| |
Collapse
|
20
|
Grishanova AY, Perepechaeva ML. Kynurenic Acid/AhR Signaling at the Junction of Inflammation and Cardiovascular Diseases. Int J Mol Sci 2024; 25:6933. [PMID: 39000041 PMCID: PMC11240928 DOI: 10.3390/ijms25136933] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Persistent systemic chronic inflammatory conditions are linked with many pathologies, including cardiovascular diseases (CVDs), a leading cause of death across the globe. Among various risk factors, one of the new possible contributors to CVDs is the metabolism of essential amino acid tryptophan. Proinflammatory signals promote tryptophan metabolism via the kynurenine (KYN) pathway (KP), thereby resulting in the biosynthesis of several immunomodulatory metabolites whose biological effects are associated with the development of symptoms and progression of various inflammatory diseases. Some participants in the KP are agonists of aryl hydrocarbon receptor (AhR), a central player in a signaling pathway that, along with a regulatory influence on the metabolism of environmental xenobiotics, performs a key immunomodulatory function by triggering various cellular mechanisms with the participation of endogenous ligands to alleviate inflammation. An AhR ligand with moderate affinity is the central metabolite of the KP: KYN; one of the subsequent metabolites of KYN-kynurenic acid (KYNA)-is a more potent ligand of AhR. Understanding the role of AhR pathway-related metabolites of the KP that regulate inflammatory factors in cells of the cardiovascular system is interesting and important for achieving effective treatment of CVDs. The purpose of this review was to summarize the results of studies about the participation of the KP metabolite-KYNA-and of the AhR signaling pathway in the regulation of inflammation in pathological conditions of the heart and blood vessels and about the possible interaction of KYNA with AhR signaling in some CVDs.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, Novosibirsk 630060, Russia;
| |
Collapse
|
21
|
Li Y, Sun T, Chen J, Liu X, Fu R, Xue F, Liu W, Ju M, Dai X, Li H, Wang W, Chi Y, Li T, Shao S, Yang R, Chen Y, Zhang L. Metabolomics profile and machine learning prediction of treatment responses in immune thrombocytopenia: A prospective cohort study. Br J Haematol 2024; 204:2405-2417. [PMID: 38438130 DOI: 10.1111/bjh.19391] [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: 11/06/2023] [Revised: 02/06/2024] [Accepted: 02/25/2024] [Indexed: 03/06/2024]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease characterized by antibody-mediated platelet destruction and impaired platelet production. The mechanisms underlying ITP and biomarkers predicting the response of drug treatments are elusive. We performed a metabolomic profiling of bone marrow biopsy samples collected from ITP patients admission in a prospective study of the National Longitudinal Cohort of Hematological Diseases. Machine learning algorithms were conducted to discover novel biomarkers to predict ITP patient treatment responses. From the bone marrow biopsies of 91 ITP patients, we quantified a total of 4494 metabolites, including 1456 metabolites in the positive mode and 3038 metabolites in the negative mode. Metabolic patterns varied significantly between groups of newly diagnosed and chronic ITP, with a total of 876 differential metabolites involved in 181 unique metabolic pathways. Enrichment factors and p-values revealed the top metabolically enriched pathways to be sphingolipid metabolism, the sphingolipid signalling pathway, ubiquinone and other terpenoid-quinone biosynthesis, thiamine metabolism, tryptophan metabolism and cofactors biosynthesis, the phospholipase D signalling pathway and the phosphatidylinositol signalling system. Based on patient responses to five treatment options, we screened several metabolites using the Boruta algorithm and ranked their importance using the random forest algorithm. Lipids and their metabolism, including long-chain fatty acids, oxidized lipids, glycerophospholipids, phosphatidylcholine and phosphatidylethanolamine biosynthesis, helped differentiate drug treatment responses. In conclusion, this study revealed metabolic alterations associated with ITP in bone marrow supernatants and a potential biomarker predicting the response to ITP.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ting Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jia Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaofan Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Rongfeng Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Feng Xue
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mankai Ju
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xinyue Dai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huiyuan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wentian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ying Chi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ting Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuai Shao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yunfei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Gene Therapy for Blood Diseases, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Gan J, Wang Y, Deng Y, Zhang J, Wang S, Jiang X, Guo M, Song L. Sacubitril/valsartan ameliorates cardiac function and ventricular remodeling in CHF rats via the inhibition of the tryptophan/kynurenine metabolism and inflammation. Sci Rep 2024; 14:12377. [PMID: 38811632 PMCID: PMC11136956 DOI: 10.1038/s41598-024-62472-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
Sacubitril/valsartan has been highly recognized as a treatment for Chronic heart failure (CHF). Its potential cardioprotective benefits and mechanisms, however, remain to be explored. Metabolomics can be used to identify the metabolic characteristics and related markers, as well as the influence of drugs, thereby opening up the new mechanism for sacubitril/valsartan therapy in CHF disease. In this study, the ligation of left anterior descending and exhaustive swimming were used to induce a rat model of CHF after myocardial infarction. The efficacy was appraised with echocardiography, serum NT-proBNP, and histopathologica. UPLC-Q/TOF-MS combined with multivariate statistical analysis approach were used to analyze the effect of sacubitril/valsartan on CHF rats. RT-qPCR and western blot were performed to investigate the tryptophan/kynurenine metabolism pathway. Accordingly, the basal cardiac function were increased, while the serum NT-proBNP and collagen volume fraction decreased in CHF rats with sacubitril/valsartan. Sacubitril/valsartan regulated the expression of kynurenine et.al 8 metabolomic biomarkers in CHF rats serum, and it contributed to the cardioprotective effects through tryptophan metabolism pathway. In addition, the mRNA and protein expression of the indoleamine 2,3-dioxygenase (IDO) in the myocardial tissue of CHF rats, were down-regulated by sacubitril/valsartan, which was the same with the IL-1β, IFN-γ, TNF-α, COX-2, and IL-6 mRNA expression, and IL-1β, IFN-γ, and TNF-α expression in serum. In conclusion, sacubitril/valsartan can ameliorate cardiac function and ventricular remodeling in CHF rats, at least in part through inhibition of tryptophan/kynurenine metabolism.
Collapse
Affiliation(s)
- Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuli Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yun Deng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiaqi Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuangcui Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Lili Song
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
23
|
Neves LS, Saraiva F, Ferreira R, Leite-Moreira A, Barros AS, Diaz SO. Metabolomics and Cardiovascular Risk in Patients with Heart Failure: A Systematic Review and Meta-Analysis. Int J Mol Sci 2024; 25:5693. [PMID: 38891881 PMCID: PMC11172189 DOI: 10.3390/ijms25115693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
The associations of plasma metabolites with adverse cardiovascular (CV) outcomes are still underexplored and may be useful in CV risk stratification. We performed a systematic review and meta-analysis to establish correlations between blood metabolites and adverse CV outcomes in patients with heart failure (HF). Four cohorts were included, involving 83 metabolites and 37 metabolite ratios, measured in 1158 HF patients. Hazard ratios (HR) of 42 metabolites and 3 metabolite ratios, present in at least two studies, were combined through meta-analysis. Higher levels of histidine (HR 0.74, 95% CI [0.64; 0.86]) and tryptophan (HR 0.82 [0.71; 0.96]) seemed protective, whereas higher levels of symmetric dimethylarginine (SDMA) (HR 1.58 [1.30; 1.93]), N-methyl-1-histidine (HR 1.56 [1.27; 1.90]), SDMA/arginine (HR 1.38 [1.14; 1.68]), putrescine (HR 1.31 [1.06; 1.61]), methionine sulfoxide (HR 1.26 [1.03; 1.52]), and 5-hydroxylysine (HR 1.25 [1.05; 1.48]) were associated with a higher risk of CV events. Our findings corroborate important associations between metabolic imbalances and a higher risk of CV events in HF patients. However, the lack of standardization and data reporting hampered the comparison of a higher number of studies. In a future clinical scenario, metabolomics will greatly benefit from harmonizing sample handling, data analysis, reporting, and sharing.
Collapse
Affiliation(s)
- Leonel Sousa Neves
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (L.S.N.); (F.S.); (A.L.-M.)
| | - Francisca Saraiva
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (L.S.N.); (F.S.); (A.L.-M.)
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Adelino Leite-Moreira
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (L.S.N.); (F.S.); (A.L.-M.)
| | - António S. Barros
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (L.S.N.); (F.S.); (A.L.-M.)
| | - Sílvia O. Diaz
- Cardiovascular R&D Centre, UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (L.S.N.); (F.S.); (A.L.-M.)
| |
Collapse
|
24
|
Cienkowski K, Cienkowska A, Kupczynska K, Bielecka-Dabrowa A. The Role of Gut Microbiota and Its Metabolites in Patients with Heart Failure. Biomedicines 2024; 12:894. [PMID: 38672248 PMCID: PMC11048107 DOI: 10.3390/biomedicines12040894] [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: 02/29/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Heart failure (HF) is a significant health concern; early detection and prevention are crucial. Recent studies suggest that the gut microbiota and its metabolites may influence HF development and risk factors. We explored this relationship by examining changes in gut microbiota composition and metabolite levels in HF patients. HF patients often exhibit decreased alpha and beta diversity compared to controls, suggesting lower bacterial richness and community variation. Changes in specific bacterial phyla were observed, with decreases in Firmicutes (e.g., Ruminococcus) and Bacteroidetes (e.g., Prevotella) and increases in Proteobacteria (e.g., Escherichia, Shigella, and Klebsiella) and Actinobacteria. Gut-microbiota-related metabolites have been identified, potentially affecting various body systems, including the cardiovascular system. Among these are short-chain fatty acids (SCFAs), betaine, trimethylamine N-oxide (TMAO), phenylalanine, tryptophan-kynurenine, and phenylacetylgutamine (PAGIn). Although SCFAs positively affect our organisms, patients with HF have been observed to experience a decline in bacteria responsible for producing these chemical compounds. There have been indications of possible links between betaine, TMAO, phenylalanine, tryptophan-kynurenine, PAGIn, and heart failure. TMAO and phenylalanine, in particular, show promise as potential prognostic factors. However, their clinical significance has not yet been thoroughly evaluated and requires further investigation.
Collapse
Affiliation(s)
- Krzysztof Cienkowski
- Faculty of Medicine, Medical University of Lodz (MUL), al. Tadeusza Kosciuszki 4, 90419 Lodz, Poland
| | - Alicja Cienkowska
- Faculty of Biology and Environmental Protection, University of Lodz, ul. Gabriela Narutowicza 68, 90136 Lodz, Poland
| | - Karolina Kupczynska
- Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Rzgowska 281/289, 93338 Lodz, Poland; (K.K.)
| | - Agata Bielecka-Dabrowa
- Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Rzgowska 281/289, 93338 Lodz, Poland; (K.K.)
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Rzgowska 281/289, 93338 Lodz, Poland
| |
Collapse
|
25
|
Liu Z, Ma Z, Jin L, Nizhamuding X, Zeng J, Zhang T, Zhang J, Wang J, Zhao H, Zhou W, Zhang C. Altered neopterin and IDO in kynurenine metabolism based on LC-MS/MS metabolomics study: Novel therapeutic checkpoints for type 2 diabetes mellitus. Clin Chim Acta 2024; 557:117859. [PMID: 38518968 DOI: 10.1016/j.cca.2024.117859] [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: 11/26/2023] [Revised: 01/26/2024] [Accepted: 03/03/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND This study assessed the alternations of kynurenine pathway (KP) and neopterin in type 2 diabetes mellitus (T2DM) and explored possible differential metabolites. METHODS A fresh residual sera panel was collected from 80 healthy control (HC) individuals and 72 T2DM patients. Metabolites/ratios of interest including tryptophan (TRP), kynurenine (KYN), 5-hydroxytryptamine (5HT), kynurenic acid (KA), xanthurenic acid (XA), neopterin (NEO), KA/KYN ratio and KYN/TRP ratio were determined using a targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomics approach, and the difference between groups was assessed. Supervised orthogonal partial least squares-discriminant analysis and differential metabolite screening with fold change (FC) were performed to identify distinct biomarkers. The diagnostic performance of KP metabolites in T2DM was evaluated. RESULTS Significant decreases of TRP, 5HT, KA, XA, and KA/KYN and increases of KYN/TRP and NEO in T2DM compared to HC group were observed (P < 0.05). The KP metabolites panel significantly changed between T2DM and HC groups (Q2: 0.925, P < 0.005). 5HT (FC: 0.63, P < 0.01) and NEO (FC: 3.27, P < 0.01) were proven to be distinct differential metabolites. A combined testing of fasting plasma glucose and KYN/TRP showed good value in the prediction of T2DM (AUC: 0.904, 95% CI 0.843-0.947). CONCLUSIONS The targeted LC-MS/MS metabolomics study is a powerful tool for evaluating the status of T2DM. This study facilitated the application of KP metabolomics into future clinical practice. 5HT and NEO are promising biomarkers in T2DM. KYN/TRP was highly associated with the development of T2DM and may serve as a potential treatment target.
Collapse
Affiliation(s)
- Zhenni Liu
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Zijia Ma
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Lizi Jin
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xiaerbanu Nizhamuding
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Jie Zeng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China
| | - Tianjiao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Jiangtao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China
| | - Jing Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China
| | - Haijian Zhao
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China
| | - Weiyan Zhou
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China.
| | - Chuanbao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
26
|
Zhang Y, Huang K, Duan J, Zhao R, Yang L. Gut microbiota connects the brain and the heart: potential mechanisms and clinical implications. Psychopharmacology (Berl) 2024; 241:637-651. [PMID: 38407637 DOI: 10.1007/s00213-024-06552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/04/2024] [Indexed: 02/27/2024]
Abstract
Nowadays, high morbidity and mortality of cardiovascular diseases (CVDs) and high comorbidity rate of neuropsychiatric disorders contribute to global burden of health and economics. Consequently, a discipline concerning abnormal connections between the brain and the heart and the resulting disease states, known as psychocardiology, has garnered interest among researchers. However, identifying a common pathway that physicians can modulate remains a challenge. Gut microbiota, a constituent part of the human intestinal ecosystem, is likely involved in mutual mechanism CVDs and neuropsychiatric disorder share, which could be a potential target of interventions in psychocardiology. This review aimed to discuss complex interactions from the perspectives of microbial and intestinal dysfunction, behavioral factors, and pathophysiological changes and to present possible approaches to regulating gut microbiota, both of which are future directions in psychocardiology.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Kai Huang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Jiahao Duan
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Rong Zhao
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
| |
Collapse
|
27
|
Chi K, Liu J, Li X, Wang H, Li Y, Liu Q, Zhou Y, Ge Y. Biomarkers of heart failure: advances in omics studies. Mol Omics 2024; 20:169-183. [PMID: 38224222 DOI: 10.1039/d3mo00173c] [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: 01/16/2024]
Abstract
Heart failure is a complex syndrome characterized by progressive circulatory dysfunction, manifesting clinically as pulmonary and systemic venous congestion, alongside inadequate tissue perfusion. The early identification of HF, particularly at the mild and moderate stages (stages B and C), presents a clinical challenge due to the overlap of signs, symptoms, and natriuretic peptide levels with other cardiorespiratory pathologies. Nonetheless, early detection coupled with timely pharmacological intervention is imperative for enhancing patient outcomes. Advances in high-throughput omics technologies have enabled researchers to analyze patient-derived biofluids and tissues, discovering biomarkers that are sensitive and specific for HF diagnosis. Due to the diversity of HF etiology, it is insufficient to study the diagnostic data of early HF using a single omics technology. This study reviewed the latest progress in genomics, transcriptomics, proteomics, and metabolomics for the identification of HF biomarkers, offering novel insights into the early clinical diagnosis of HF. However, the validity of biomarkers depends on the disease status, intervention time, genetic diversity and comorbidities of the subjects. Moreover, biomarkers lack generalizability in different clinical settings. Hence, it is imperative to conduct multi-center, large-scale and standardized clinical trials to enhance the diagnostic accuracy and utility of HF biomarkers.
Collapse
Affiliation(s)
- Kuo Chi
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Jing Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Xinghua Li
- Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| | - He Wang
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yanliang Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yabin Zhou
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yuan Ge
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
28
|
Gao J, Yang T, Song B, Ma X, Ma Y, Lin X, Wang H. Abnormal tryptophan catabolism in diabetes mellitus and its complications: Opportunities and challenges. Biomed Pharmacother 2023; 166:115395. [PMID: 37657259 DOI: 10.1016/j.biopha.2023.115395] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/20/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023] Open
Abstract
In recent years, the incidence rate of diabetes mellitus (DM), including type 1 diabetes mellitus(T1DM), type 2 diabetes mellitus(T2DM), and gestational diabetes mellitus (GDM), has increased year by year and has become a major global health problem. DM can lead to serious complications of macrovascular and microvascular. Tryptophan (Trp) is an essential amino acid for the human body. Trp is metabolized in the body through the indole pathway, kynurenine (Kyn) pathway and serotonin (5-HT) pathway, and is regulated by intestinal microorganisms to varying degrees. These three metabolic pathways have extensive regulatory effects on the immune, endocrine, neural, and energy metabolism systems of the body, and are related to the physiological and pathological processes of various diseases. The key enzymes and metabolites in the Trp metabolic pathway are also deeply involved in the pathogenesis of DM, playing an important role in pancreatic function, insulin resistance (IR), intestinal barrier, and angiogenesis. In DM and its complications, there is a disruption of Trp metabolic balance. Several therapy approaches for DM and complications have been proven to modify tryptophan metabolism. The metabolism of Trp is becoming a new area of focus for DM prevention and care. This paper reviews the impact of the three metabolic pathways of Trp on the pathogenesis of DM and the alterations in Trp metabolism in these diseases, expecting to provide entry points for the treatment of DM and its complications.
Collapse
Affiliation(s)
- Jialiang Gao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ting Yang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bohan Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaojie Ma
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yichen Ma
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaowei Lin
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hongwu Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
29
|
Zhen D, Ding L, Wang B, Wang X, Hou Y, Ding W, Portha B, Liu J. Oral administration of kynurenic acid delays the onset of type 2 diabetes in Goto-Kakizaki rats. Heliyon 2023; 9:e17733. [PMID: 37424591 PMCID: PMC10328841 DOI: 10.1016/j.heliyon.2023.e17733] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 06/11/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Kynurenic acid (KYNA) is an endogenous catabolite of tryptophan that has been found to demonstrate neuroprotective properties in psychiatric disorders. Recently, accumulating data have suggested that KYNA may also play a significant role in various metabolic diseases by stimulating energy metabolism in adipose tissue and muscle. However, whether KYNA can serves as an anti-diabetes agent has yet to be studied. In this study, we investigated the potential anti-diabetic effects of administering KYNA orally through drinking water in pre-diabetic Goto-Kakizaki rats and examined how this treatment may influence energy metabolism regulation within the liver. We found that hyperglycemic Goto-Kakizaki rats showed lower plasmatic KYNA levels compared to normal rats. Oral administration of KYNA significantly delayed the onset of diabetes in Goto-Kakizaki rats compared to untreated animals. Moreover, we found that KYNA treatment significantly increased respiration exchange ratio and promoted the energy expenditure by stimulating the expression of uncoupling protein (UCP). We confirmed that KYNA stimulated the UCP expression in HepG2 cells and mouse hepatocytes at mRNA and protein levels. Our study reveals that KYNA could potentially act as an anti-diabetic agent and KYNA-induced UCP upregulation is closely associated with the regulation of energy metabolism. These results provide further evidence for the therapeutic potential of KYNA in diabetes.
Collapse
Affiliation(s)
- Delong Zhen
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Lina Ding
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Bao Wang
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaolei Wang
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yanli Hou
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wenyu Ding
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Bernard Portha
- Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251, Université Paris-Cité, Paris, France
| | - Junjun Liu
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
30
|
Matsiras D, Bezati S, Ventoulis I, Verras C, Parissis J, Polyzogopoulou E. Gut Failure: A Review of the Pathophysiology and Therapeutic Potentials in the Gut-Heart Axis. J Clin Med 2023; 12:2567. [PMID: 37048650 PMCID: PMC10095379 DOI: 10.3390/jcm12072567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Despite considerable advances in the field, heart failure (HF) still poses a significant disease burden among affected individuals since it continues to cause high morbidity and mortality rates. Inflammation is considered to play a key role in disease progression, but the exact underlying pathophysiological mechanisms involved have not yet been fully elucidated. The gut, as a potential source of inflammation, could feasibly explain the state of low-grade inflammation seen in patients with chronic HF. Several derangements in the composition of the microbiota population, coupled with an imbalance between favorable and harmful metabolites and followed by gut barrier disruption and eventually bacterial translocation, could contribute to cardiac dysfunction and aggravate HF. On the other hand, HF-associated congestion and hypoperfusion alters intestinal function, thereby creating a vicious cycle. Based on this evidence, novel pharmaceutical agents have been developed and their potential therapeutic use has been tested in both animal and human subjects. The ultimate goal in these efforts is to reverse the aforementioned intestinal derangements and block the inflammation cascade. This review summarizes the gut-related causative pathways implicated in HF pathophysiology, as well as the associated therapeutic interventions described in the literature.
Collapse
Affiliation(s)
- Dionysis Matsiras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Sofia Bezati
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece
| | - Christos Verras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - John Parissis
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Effie Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| |
Collapse
|
31
|
Mi MY, Whitlock M, Shi X, Farrell LA, Bhambhani VM, Quadir J, Blatnik M, Wald KP, Tierney B, Kim A, Loudon P, Chen ZZ, Correa A, Gao Y, Carson AP, Bertoni AG, Roth Flach RJ, Gerszten RE. Mixed meal tolerance testing highlights in diabetes altered branched-chain ketoacid metabolism and pathways associated with all-cause mortality. Am J Clin Nutr 2023; 117:529-539. [PMID: 36811472 PMCID: PMC10356557 DOI: 10.1016/j.ajcnut.2023.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Elevated BCAA levels are strongly associated with diabetes, but how diabetes affects BCAA, branched-chain ketoacids (BCKAs), and the broader metabolome after a meal is not well known. OBJECTIVE To compare quantitative BCAA and BCKA levels in a multiracial cohort with and without diabetes after a mixed meal tolerance test (MMTT) as well as to explore the kinetics of additional metabolites and their associations with mortality in self-identified African Americans. METHODS We administered an MMTT to 11 participants without obesity or diabetes and 13 participants with diabetes (treated with metformin only) and measured the levels of BCKAs, BCAAs, and 194 other metabolites at 8 time points across 5 h. We used mixed models for repeated measurements to compare between group metabolite differences at each timepoint with adjustment for baseline. We then evaluated the association of top metabolites with different kinetics with all-cause mortality in the Jackson Heart Study (JHS) (N = 2441). RESULTS BCAA levels, after adjustment for baseline, were similar at all timepoints between groups, but adjusted BCKA kinetics were different between groups for α-ketoisocaproate (P = 0.022) and α-ketoisovalerate (P = 0.021), most notably diverging at 120 min post-MMTT. An additional 20 metabolites had significantly different kinetics across timepoints between groups, and 9 of these metabolites-including several acylcarnitines-were significantly associated with mortality in JHS, irrespective of diabetes status. The highest quartile of a composite metabolite risk score was associated with higher mortality (HR:1.57; 1.20, 2.05, P = 0.00094) than the lowest quartile. CONCLUSIONS BCKA levels remained elevated after an MMTT among participants with diabetes, suggesting that BCKA catabolism may be a key dysregulated process in the interaction of BCAA and diabetes. Metabolites with different kinetics after an MMTT may be markers of dysmetabolism and associated with increased mortality in self-identified African Americans.
Collapse
Affiliation(s)
- Michael Y Mi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | | | - Xu Shi
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Laurie A Farrell
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Juweria Quadir
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Kyle P Wald
- Early Clinical Development, Pfizer, Groton, CT, USA
| | | | - Albert Kim
- Internal Medicine Research Unit, Pfizer, Cambridge, MA, USA; Cytel, Cambridge, MA, USA
| | - Peter Loudon
- Early Clinical Development, Pfizer, Cambridge, UK; Tenpoint Therapeutics, Cambridge, UK
| | - Zsu-Zsu Chen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Adolfo Correa
- Department of Population Health Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yan Gao
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alain G Bertoni
- Department of Epidemiology & Prevention, Wake Forest School of Medicine, Winston Salem, NC, USA
| | | | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
32
|
Kynurenine Pathway in Diabetes Mellitus-Novel Pharmacological Target? Cells 2023; 12:cells12030460. [PMID: 36766803 PMCID: PMC9913876 DOI: 10.3390/cells12030460] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
The tryptophan-kynurenine pathway (Trp-KYN) is the major route for tryptophan conversion in the brain and in the periphery. Kynurenines display a wide range of biological actions (which are often contrasting) such as cytotoxic/cytoprotective, oxidant/antioxidant or pro-/anti-inflammatory. The net effect depends on their local concentration, cellular environment, as well as a complex positive and negative feedback loops. The imbalance between beneficial and harmful kynurenines was implicated in the pathogenesis of various neurodegenerative disorders, psychiatric illnesses and metabolic disorders, including diabetes mellitus (DM). Despite available therapies, DM may lead to serious macro- and microvascular complications including cardio- and cerebrovascular disease, peripheral vascular disease, chronic renal disease, diabetic retinopathy, autonomic neuropathy or cognitive impairment. It is well established that low-grade inflammation, which often coincides with DM, can affect the function of KP and, conversely, that kynurenines may modulate the immune response. This review provides a detailed summary of findings concerning the status of the Trp-KYN pathway in DM based on available animal, human and microbiome studies. We highlight the importance of the molecular interplay between the deranged (functionally and qualitatively) conversion of Trp to kynurenines in the development of DM and insulin resistance. The Trp-KYN pathway emerges as a novel target in the search for preventive and therapeutic interventions in DM.
Collapse
|
33
|
Yang Y, Du Z, Fang M, Ma Y, Liu Y, Wang T, Han Z, Peng Z, Pan Y, Qin H, Qin Y, Jiang Y, Tu P, Guo X, Lu Y, Yang X, Hua K. Metabolic signatures in pericardial fluid and serum are associated with new-onset atrial fibrillation after isolated coronary artery bypass grafting. Transl Res 2023; 256:30-40. [PMID: 36638862 DOI: 10.1016/j.trsl.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Postoperative atrial fibrillation (POAF) is a common complication of coronary artery bypass grafting (CABG) procedures. However, the molecular mechanism of POAF remains poorly understood, hence the absence of effective prevention strategies. Here we used targeted metabolomics on pericardial fluid and serum samples from CABG patients to investigate POAF-associated metabolic alterations and related risk prediction of new-onset AF. Nine differential metabolites in various metabolic pathways were found in both pericardial fluid and serum samples from patients with POAF and without POAF. By using machine learning algorithms and regression models, a 4-metabolite (aceglutamide, ornithine, methionine, and arginine) risk prediction model was constructed and showed accurate performance in predicting POAF in both discovery and validation sets. This work extends the metabolic insights of the cardiac microenvironment and blood in patients with POAF and paves the way for the use of targeted metabolomics for predicting POAF in patients with CABG surgery.
Collapse
Affiliation(s)
- Yunxiao Yang
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhiyong Du
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Meng Fang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ying Ma
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, State Key Laboratory of Dao-di Herbs, Beijing, China
| | - Yuhua Liu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Tianguang Wang
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhongyi Han
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhan Peng
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yilin Pan
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haokai Qin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanwen Qin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoyu Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yingyuan Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Xiubin Yang
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Kun Hua
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
34
|
Razquin C, Ruiz-Canela M, Toledo E, Clish CB, Guasch-Ferré M, García-Gavilán JF, Wittenbecher C, Alonso-Gómez A, Fitó M, Liang L, Corella D, Gómez-Gracia E, Estruch R, Fiol M, Santos-Lozano JM, Serra-Majem L, Ros E, Aros F, Salas-Salvadó J, Hu FB, Martínez-González MA. Circulating Amino Acids and Risk of Peripheral Artery Disease in the PREDIMED Trial. Int J Mol Sci 2022; 24:270. [PMID: 36613713 PMCID: PMC9820556 DOI: 10.3390/ijms24010270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Effective prevention and risk prediction are important for peripheral artery disease (PAD) due to its poor prognosis and the huge disease burden it produces. Circulating amino acids (AA) and their metabolites may serve as biomarkers of PAD risk, but they have been scarcely investigated. The objective was to prospectively analyze the associations of baseline levels of plasma AA (and their pathways) with subsequent risk of PAD and the potential effect modification by a nutritional intervention with the Mediterranean diet (MedDiet). A matched case-control study was nested in the PREDIMED trial, in which participants were randomized to three arms: MedDiet with tree nut supplementation group, MedDiet with extra-virgin olive oil (EVOO) supplementation group or control group (low-fat diet). One hundred and sixty-seven PAD cases were matched with 250 controls. Plasma AA was measured with liquid chromatography/mass spectrometry at the Broad Institute. Baseline tryptophan, serine and threonine were inversely associated with PAD (ORfor 1 SD increase = 0.78 (0.61-0.99); 0.67 (0.51-0.86) and 0.75 (0.59-0.95), respectively) in a multivariable-adjusted conditional logistic regression model. The kynurenine/tryptophan ratio was directly associated with PAD (ORfor 1 SD increase = 1.50 (1.14-1.98)). The nutritional intervention with the MedDiet+nuts modified the association between threonine and PAD (p-value interaction = 0.018) compared with the control group. However, subjects allocated to the MedDiet+EVOO group were protected against PAD independently of baseline threonine. Plasma tryptophan, kynurenine/tryptophan ratio, serine and threonine might serve as early biomarkers of future PAD in subjects at a high risk of cardiovascular disease. The MedDiet supplemented with EVOO exerted a protective effect, regardless of baseline levels of threonine.
Collapse
Grants
- R01HL118264 NIH HHS
- RTIC RD 06/0045 Instituto de Salud Carlos III
- CIBEROBN Instituto de Salud Carlos III
- RTIC G03/140 Instituto de Salud Carlos III
- CNIC 06/2007 Spanish National Centre for Cardiovascular Research
- PI04-2239, PI 05/2584, CP06/00100, PI07/0240, PI07/1138, PI07/0954, PI 07/0473, PI10/01407, PI10/02658, PI11/01647, P11/02505, PI13/00462, and JR17/00022 Instituto de Salud Carlos III
- AGL-2009-13906-C02, AGL2010-22319-C03 and SAF2016-80532-R Instituto de Salud Carlos III
Collapse
Affiliation(s)
- Cristina Razquin
- Department of Preventive Medicine and Public Health, IdiSNA (Instituto de Investigación Sanitaria de Navarra), University of Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, IdiSNA (Instituto de Investigación Sanitaria de Navarra), University of Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Estefania Toledo
- Department of Preventive Medicine and Public Health, IdiSNA (Instituto de Investigación Sanitaria de Navarra), University of Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Clary B. Clish
- Broad Institute and MIT, Harvard University, Cambridge, MA 02142, USA
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Public Health, Section of Epidemiology, University of Copenhagen, 1165 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 1353 Copenhagen, Denmark
| | - Jesús F. García-Gavilán
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unitat de Nutrició Humana, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari San Joan de Reus, 43204 Reus, Spain
| | - Clemens Wittenbecher
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- SciLifeLab, Food and Nutrition Science Division, Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Angel Alonso-Gómez
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Montse Fitó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain
| | - Liming Liang
- Department of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Dolores Corella
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine, University of Valencia, 46100 Valencia, Spain
| | | | - Ramon Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Miquel Fiol
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Plataforma de Ensayos Clínicos, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Jose M. Santos-Lozano
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, 41013 Sevilla, Spain
| | - Luis Serra-Majem
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Nutrition Research Group, Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain
| | - Emilio Ros
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition, Agust Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Fernando Aros
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Jordi Salas-Salvadó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unitat de Nutrició Humana, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari San Joan de Reus, 43204 Reus, Spain
| | - Frank B. Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Miguel A. Martínez-González
- Department of Preventive Medicine and Public Health, IdiSNA (Instituto de Investigación Sanitaria de Navarra), University of Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
35
|
Mediterranean Diet on Sleep: A Health Alliance. Nutrients 2022; 14:nu14142998. [PMID: 35889954 PMCID: PMC9318336 DOI: 10.3390/nu14142998] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/17/2022] [Accepted: 07/20/2022] [Indexed: 12/19/2022] Open
Abstract
The Mediterranean diet is a plant-based, antioxidant-rich, unsaturated fat dietary pattern that has been consistently associated with lower rates of noncommunicable diseases and total mortality, so that it is considered one of the healthiest dietary patterns. Clinical trials and mechanistic studies have demonstrated that the Mediterranean diet and its peculiar foods and nutrients exert beneficial effects against inflammation, oxidative stress, dysmetabolism, vascular dysfunction, adiposity, senescence, cognitive decline, neurodegeneration, and tumorigenesis, thus preventing age-associated chronic diseases and improving wellbeing and health. Nocturnal sleep is an essential physiological function, whose alteration is associated with health outcomes and chronic diseases. Scientific evidence suggests that diet and sleep are related in a bidirectional relationship, and the understanding of this association is important given their role in disease prevention. In this review, we surveyed the literature concerning the current state of evidence from epidemiological studies on the impact of the Mediterranean diet on nighttime sleep quantity and quality. The available studies indicate that greater adherence to the Mediterranean diet is associated with adequate sleep duration and with several indicators of better sleep quality. Potential mechanisms mediating the effect of the Mediterranean diet and its foods and nutrients on sleep are described, and gap-in-knowledge and new research agenda to corroborate findings are discussed.
Collapse
|
36
|
Ala M, Eftekhar SP. The Footprint of Kynurenine Pathway in Cardiovascular Diseases. Int J Tryptophan Res 2022; 15:11786469221096643. [PMID: 35784899 PMCID: PMC9248048 DOI: 10.1177/11786469221096643] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/28/2022] [Indexed: 12/30/2022] Open
Abstract
Kynurenine pathway is the main route of tryptophan metabolism and produces several metabolites with various biologic properties. It has been uncovered that several cardiovascular diseases are associated with the overactivation of kynurenine pathway and kynurenine and its metabolites have diagnostic and prognostic value in cardiovascular diseases. Furthermore, it was found that several kynurenine metabolites can differently affect cardiovascular health. For instance, preclinical studies have shown that kynurenine, xanthurenic acid and cis-WOOH decrease blood pressure; kynurenine and 3-hydroxyanthranilic acid prevent atherosclerosis; kynurenic acid supplementation and kynurenine 3-monooxygenase (KMO) inhibition improve the outcome of stroke. Indoleamine 2,3-dioxygenase (IDO) overactivity and increased kynurenine levels improve cardiac and vascular transplantation outcomes, whereas exacerbating the outcome of myocardial ischemia, post-ischemic myocardial remodeling, and abdominal aorta aneurysm. IDO inhibition and KMO inhibition are also protective against viral myocarditis. In addition, dysregulation of kynurenine pathway is observed in several conditions such as senescence, depression, diabetes, chronic kidney disease (CKD), cirrhosis, and cancer closely connected to cardiovascular dysfunction. It is worth defining the exact effect of each metabolite of kynurenine pathway on cardiovascular health. This narrative review is the first review that separately discusses the involvement of kynurenine pathway in different cardiovascular diseases and dissects the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Seyed Parsa Eftekhar
- Student Research Committee, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
37
|
Jamshed L, Debnath A, Jamshed S, Wish JV, Raine JC, Tomy GT, Thomas PJ, Holloway AC. An Emerging Cross-Species Marker for Organismal Health: Tryptophan-Kynurenine Pathway. Int J Mol Sci 2022; 23:6300. [PMID: 35682980 PMCID: PMC9181223 DOI: 10.3390/ijms23116300] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Tryptophan (TRP) is an essential dietary amino acid that, unless otherwise committed to protein synthesis, undergoes metabolism via the Tryptophan-Kynurenine (TRP-KYN) pathway in vertebrate organisms. TRP and its metabolites have key roles in diverse physiological processes including cell growth and maintenance, immunity, disease states and the coordination of adaptive responses to environmental and dietary cues. Changes in TRP metabolism can alter the availability of TRP for protein and serotonin biosynthesis as well as alter levels of the immune-active KYN pathway metabolites. There is now considerable evidence which has shown that the TRP-KYN pathway can be influenced by various stressors including glucocorticoids (marker of chronic stress), infection, inflammation and oxidative stress, and environmental toxicants. While there is little known regarding the role of TRP metabolism following exposure to environmental contaminants, there is evidence of linkages between chemically induced metabolic perturbations and altered TRP enzymes and KYN metabolites. Moreover, the TRP-KYN pathway is conserved across vertebrate species and can be influenced by exposure to xenobiotics, therefore, understanding how this pathway is regulated may have broader implications for environmental and wildlife toxicology. The goal of this narrative review is to (1) identify key pathways affecting Trp-Kyn metabolism in vertebrates and (2) highlight consequences of altered tryptophan metabolism in mammals, birds, amphibians, and fish. We discuss current literature available across species, highlight gaps in the current state of knowledge, and further postulate that the kynurenine to tryptophan ratio can be used as a novel biomarker for assessing organismal and, more broadly, ecosystem health.
Collapse
Affiliation(s)
- Laiba Jamshed
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada; (L.J.); (A.D.); (S.J.)
| | - Amrita Debnath
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada; (L.J.); (A.D.); (S.J.)
| | - Shanza Jamshed
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada; (L.J.); (A.D.); (S.J.)
| | - Jade V. Wish
- Department of Chemistry, Centre for Oil and Gas Research and Development (COGRAD), University of Manitoba, 586 Parker Building, 144 Dysart Rd., Winnipeg, MB R3T 2N2, Canada; (J.V.W.); (G.T.T.)
| | - Jason C. Raine
- Quesnel River Research Centre, University of Northern British Columbia, Prince George, BC V2N 4Z9, Canada;
| | - Gregg T. Tomy
- Department of Chemistry, Centre for Oil and Gas Research and Development (COGRAD), University of Manitoba, 586 Parker Building, 144 Dysart Rd., Winnipeg, MB R3T 2N2, Canada; (J.V.W.); (G.T.T.)
| | - Philippe J. Thomas
- Environment and Climate Change Canada, National Wildlife Research Centre, Ottawa, ON K1A 0H3, Canada;
| | - Alison C. Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada; (L.J.); (A.D.); (S.J.)
| |
Collapse
|
38
|
Luo Q, Hu Y, Chen X, Luo Y, Chen J, Wang H. Effects of Gut Microbiota and Metabolites on Heart Failure and Its Risk Factors: A Two-Sample Mendelian Randomization Study. Front Nutr 2022; 9:899746. [PMID: 35799593 PMCID: PMC9253861 DOI: 10.3389/fnut.2022.899746] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/30/2022] [Indexed: 12/11/2022] Open
Abstract
Introduction Previous observational studies have indicated that gut microbiota and metabolites may contribute to heart failure and its risk factors. However, with the limitation of reverse causality and confounder in observational studies, such relationship remains unclear. This study aims to reveal the causal effect of gut microbiota and metabolites on heart failure and its risk factors. Methods This study collected summary statistics regarding gut microbiota and metabolites, heart failure, diabetes, hypertension, chronic kidney disease, myocardial infarction, atrial fibrillation, hypertrophic cardiomyopathy, dilated cardiomyopathy, coronary heart disease, valvular heart disease, and myocarditis. Two-sample Mendelian randomization analysis was performed using MR-Egger, inverse variance weighted (IVW), MR-PRESSO, maximum likelihood, and weighted median. Results Results from gene prediction showed that among all gut microbiota, candida, shigella, and campylobacter were not associated with higher incidence of heart failure. However, genetic prediction suggested that for every 1 unit increase in shigella concentration, the relative risk increased by 38.1% for myocarditis and 13.3% for hypertrophic cardiomyopathy. Besides, for every 1 unit increased in candida concentration, the relative risk of chronic kidney disease increased by 7.1%. As for intestinal metabolites, genetic prediction results suggested that for every 1 unit increase in betaine, the relative risk of heart failure and myocardial infarction increased by 1.4% and 1.7%, separately. Conclusions This study suggested new evidence of the relationship between gut microbiota and heart failure and its risk factors, which may shed light on designing microbiome- and microbiome-dependent metabolite interventions on heart failure and its risk factors in clinical trials in the future.
Collapse
Affiliation(s)
- Qiang Luo
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Yilan Hu
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Xin Chen
- Department of Laboratory, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Yong Luo
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Jie Chen
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| | - Han Wang
- Department of Cardiology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
39
|
Tuerhongjiang G, Guo M, Qiao X, Lou B, Wang C, Wu H, Wu Y, Yuan Z, She J. Interplay Between Gut Microbiota and Amino Acid Metabolism in Heart Failure. Front Cardiovasc Med 2021; 8:752241. [PMID: 34746265 PMCID: PMC8566708 DOI: 10.3389/fcvm.2021.752241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/07/2021] [Indexed: 11/14/2022] Open
Abstract
Heart failure (HF) is a complex clinical syndrome of which the incidence is on the rise worldwide. Cardiometabolic disorders are associated with the deterioration of cardiac function and progression of HF. Recently, there has been renewed interest in gut microbiota (GM) and its metabolites in the cardiovascular disease. HF-caused hypoperfusion could increase intestinal permeability, and a “leaky” bowel leads to bacterial translocation and make its metabolites more easily enter the circulation. Considerable evidence shows that the composition of microbiota and amino acids (AAs) has been altered in HF patients, and AAs could serve as a diagnostic and prognostic biomarker in HF. The findings indicate that the gut–amino acid–HF axis may play a key role in the progression of HF. In this paper, we focus on the interrelationship between the AA metabolism and GM alterations during the development of heart failure. We also discuss the potential prognostic and therapeutic value of the gut–amino acid–HF axis in the cortex of HF.
Collapse
Affiliation(s)
- Gulinigaer Tuerhongjiang
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Manyun Guo
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Xiangrui Qiao
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Bowen Lou
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Chen Wang
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Haoyu Wu
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Yue Wu
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Zuyi Yuan
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Jianqing She
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| |
Collapse
|