1
|
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
|
2
|
Li Q, Xie H, Guo Y, Li J, Qian J, Wu W. Formoterol Reduces the Pro-Inflammatory Phenotype by Enhancing the Activity of Glutaminase in Monocyte-Derived Macrophages in the CVB3-Induced Viral Myocarditis. Immun Inflamm Dis 2024; 12:e70073. [PMID: 39601476 PMCID: PMC11600452 DOI: 10.1002/iid3.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/13/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Viral myocarditis (VMC) plays a significant role in heart failure, and there is currently a shortage of available targeted treatments. Macrophage phenotype and function are closely associated with the beta-2 adrenergic receptor (β2-AR). METHOD This research employed a BALB/c mouse model of VMC generated using Coxsackievirus B3 (CVB3), and the β2-AR agonist formoterol was administered as treatment. A bioinformatic analysis was conducted to identify the β2-AR in CCR2+MHCIIhigh monocyte-derived macrophages (MoMFs). Echocardiography and histopathological assessments were utilized to evaluate cardiac function and inflammation. The enzymatic activity of glutaminase (GLS) was quantified. Flow cytometry was employed to characterize the phenotype and function of the macrophages. RESULT Our study revealed that formoterol treatment effectively mitigated cardiac inflammation and fibrosis, improved cardiac function, and prolonged survival compared to the VMC group. Formoterol reduced the infiltration of CCR2+MHCIIhigh MoMFs in the heart, inhibited M1 phenotypic expression and activity, and reduced the percentage of Ly6Chigh monocytes in circulation. Additionally, formoterol stimulated M2 phenotypic expression and activity and increased the percentage of Ly6Clow monocytes in circulation. Additionally, the combination of NICB3344, a C-C motif chemokine receptor 2 inhibitor, with formoterol did not exhibit synergistic effects on reducing cardiac pathological scores or enhancing cardiac function. In vitro studies involving the use of lipopolysaccharide (LPS)-induced bone marrow-derived macrophages, revealed the ability of formoterol to suppress the M1 phenotype and functions induced by LPS while promoting the M2 phenotype and functions. Nevertheless, the observed effects were negated by the introduction of the GLS inhibitor BPTES. CONCLUSION Formoterol potentially serves as a significant metabolic regulator in the differentiation process of cardiac MoMFs, influencing this process by controlling GLS activity. Targeting β2-AR exhibits potential as an effective approach for managing VMC. It is essential to acknowledge that these findings were derived under specific experimental conditions, with the current conclusions predominantly based on animal models. Future research is necessary to further investigate the feasibility of formoterol in clinical practice.
Collapse
Affiliation(s)
- Quan‐liang Li
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Hua‐bao Xie
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Ying‐xin Guo
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Juan‐fen Li
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jing Qian
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Wei‐Feng Wu
- Department of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and Ministry, Guangxi Medical UniversityNanningChina
| |
Collapse
|
3
|
Duan S, Zhang W, Li Y, Li Y, Zhao Y, Jin W, Liu Q, Li M, Sun W, Chen L, Xu H, Tang J, Hou J, Deng Z, Yang F, Ma S, He Z. Coxsackievirus B3 HFMD animal models in Syrian hamster and rhesus monkey. Virol Sin 2024; 39:290-300. [PMID: 38331038 DOI: 10.1016/j.virs.2024.02.001] [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: 05/05/2023] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Coxsackievirus B3 (CVB3) is the pathogen causing hand, foot and mouth disease (HFMD), which manifests across a spectrum of clinical severity from mild to severe. However, CVB3-infected mouse models mainly demonstrate viral myocarditis and pancreatitis, failing to replicate human HFMD symptoms. Although several enteroviruses have been evaluated in Syrian hamsters and rhesus monkeys, there is no comprehensive data on CVB3. In this study, we have first tested the susceptibility of Syrian hamsters to CVB3 infection via different routes. The results showed that Syrian hamsters were successfully infected with CVB3 by intraperitoneal injection or nasal drip, leading to nasopharyngeal colonization, acute severe pathological injury, and typical HFMD symptoms. Notably, the nasal drip group exhibited a longer viral excretion cycle and more severe pathological damage. In the subsequent study, rhesus monkeys infected with CVB3 through nasal drips also presented signs of HFMD symptoms, viral excretion, serum antibody conversion, viral nucleic acids and antigens, and the specific organ damages, particularly in the heart. Surprisingly, there were no significant differences in myocardial enzyme levels, and the clinical symptoms resembled those often associated with common, mild infections. In summary, the study successfully developed severe Syrian hamsters and mild rhesus monkey models for CVB3-induced HFMD. These models could serve as a basis for understanding the disease pathogenesis, conducting pre-trial prevention and evaluation, and implementing post-exposure intervention.
Collapse
Affiliation(s)
- Suqin Duan
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Wei Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Yongjie Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Yanyan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Yuan Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Weihua Jin
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Quan Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Mingxue Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Wenting Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Lixiong Chen
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Hongjie Xu
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Jie Tang
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Jinghan Hou
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Zijun Deng
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China.
| | - Shaohui Ma
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China.
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Sciences Medical Primate Research Center Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Kunming, 650118, China.
| |
Collapse
|
4
|
Wang L, Sun T, Liu X, Wang Y, Qiao X, Chen N, Liu F, Zhou X, Wang H, Shen H. Myocarditis: A multi-omics approach. Clin Chim Acta 2024; 554:117752. [PMID: 38184138 DOI: 10.1016/j.cca.2023.117752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
Myocarditis, an inflammatory condition of weakened heart muscles often triggered by a variety of causes, that can result in heart failure and sudden death. Novel ways to enhance our understanding of myocarditis pathogenesis is available through newer modalities (omics). In this review, we examine the roles of various biomolecules and associated functional pathways across genomics, transcriptomics, proteomics, and metabolomics in the pathogenesis of myocarditis. Our analysis further explores the reproducibility and variability intrinsic to omics studies, underscoring the necessity and significance of employing a multi-omics approach to gain profound insights into myocarditis pathogenesis. This integrated strategy not only enhances our understanding of the disease, but also confirms the critical importance of a holistic multi-omics approach in disease analysis.
Collapse
Affiliation(s)
- Lulu Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Tao Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Xiaolan Liu
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Yan Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Xiaorong Qiao
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Nuo Chen
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Fangqian Liu
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Xiaoxiang Zhou
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Hua Wang
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Hongxing Shen
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China.
| |
Collapse
|
5
|
Wang Y, Xie Y, Mahara G, Xiong Y, Xiong Y, Zheng Q, Chen J, Zhang W, Zhou H, Li Q. Intestinal microbiota and metabolome perturbations in ischemic and idiopathic dilated cardiomyopathy. J Transl Med 2024; 22:89. [PMID: 38254195 PMCID: PMC10804607 DOI: 10.1186/s12967-023-04605-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/06/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Various clinical similarities are present in ischemic (ICM) and idiopathic dilated cardiomyopathy (IDCM), leading to ambiguity on some occasions. Previous studies have reported that intestinal microbiota appeared dysbiosis in ICM, whether implicating in the IDCM remains unclear. The aim of this study was to assess the alterations in intestinal microbiota and fecal metabolites in ICM and IDCM. METHODS ICM (n = 20), IDCM (n = 22), and healthy controls (HC, n = 20) were enrolled in this study. Stool samples were collected for 16S rRNA gene sequencing and gas chromatography-mass spectrometry (GC-MS) analysis. RESULTS Both ICM and IDCM exhibited reduced alpha diversity and altered microbial community structure compared to HC. At the genus level, nine taxa including Blautia, [Ruminococcus]_torques_group, Christensenellaceae_R-7_group, UCG-002, Corynebacterium, Oceanobacillus, Gracilibacillus, Klebsiella and Citrobacter was specific to ICM, whereas one taxa Alistipes uniquely altered in IDCM. Likewise, these changes were accompanied by significant metabolic differences. Further differential analysis displayed that 18 and 14 specific metabolites uniquely changed in ICM and IDCM, respectively. The heatmap was generated to display the association between genera and metabolites. Receiver operating characteristic curve (ROC) analysis confirmed the predictive value of the distinct microbial-metabolite features in disease status. The results showed that microbial (area under curve, AUC = 0.95) and metabolic signatures (AUC = 0.84) were effective in discriminating ICM from HC. Based on the specific microbial and metabolic features, the patients with IDCM could be separated from HC with an AUC of 0.80 and 0.87, respectively. Furthermore, the gut microbial genus (AUC = 0.88) and metabolite model (AUC = 0.89) were comparable in predicting IDCM from ICM. Especially, the combination of fecal microbial-metabolic features improved the ability to differentiate IDCM from ICM with an AUC of 0.96. CONCLUSION Our findings highlighted the alterations of gut microbiota and metabolites in different types of cardiomyopathies, providing insights into the pathophysiological mechanisms of myocardial diseases. Moreover, multi-omics analysis of fecal samples holds promise as a non-invasive tool for distinguishing disease status.
Collapse
Affiliation(s)
- Yusheng Wang
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yandan Xie
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Gehendra Mahara
- Clinical Research Center, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yanling Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yalan Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qifang Zheng
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Jianqin Chen
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
6
|
Kong Q, Chen L, Zeng X, Lu F, Huang Y, Wu W. Alterations of the gut microbiome and metabolic profile in CVB3-induced mice acute viral myocarditis. BMC Microbiol 2023; 23:139. [PMID: 37202726 DOI: 10.1186/s12866-023-02863-4] [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: 09/27/2022] [Accepted: 04/16/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Acute viral myocarditis (AVMC) is an inflammatory disease of the myocardium. Evidence indicates that dysbiosis of gut microbiome and related metabolites intimately associated with cardiovascular diseases through the gut-heart axis. METHODS We built mouse models of AVMC, then applied 16 S rDNA gene sequencing and UPLC-MS/MS metabolomics to explore variations of gut microbiome and disturbances of cardiac metabolic profiles. RESULTS Compared with Control group, analysis of gut microbiota showed lower diversity in AVMC, decreased relative abundance of genera mainly belonging to the phyla Bacteroidetes, and increased of phyla Proteobacteria. Metabolomics analysis showed disturbances of cardiac metabolomics, including 62 increased and 84 decreased metabolites, and mainly assigned to lipid, amino acid, carbohydrate and nucleotide metabolism. The steroid hormone biosynthesis, cortisol synthesis and secretion pathway were particularly enriched in AVMC. Among them, such as estrone 3-sulfate, desoxycortone positively correlated with disturbed gut microbiome. CONCLUSION In summary, both the structure of the gut microbiome community and the cardiac metabolome were significantly changed in AVMC. Our findings suggest that gut microbiome may participate in the development of AVMC, the mechanism may be related to its role in dysregulated metabolites such as steroid hormone biosynthesis.
Collapse
Affiliation(s)
- Qing Kong
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lili Chen
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaochun Zeng
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Feiyu Lu
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanlan Huang
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weifeng Wu
- Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
7
|
Tomar M, Sharma A, Araniti F, Pateriya A, Shrivastava A, Tamrakar AK. Distinct Metabolomic Profiling of Serum Samples from High-Fat-Diet-Induced Insulin-Resistant Mice. ACS Pharmacol Transl Sci 2023; 6:771-782. [PMID: 37200804 PMCID: PMC10186361 DOI: 10.1021/acsptsci.3c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Indexed: 05/20/2023]
Abstract
High-fat-diet (HFD)-induced obesity is associated with an elevated risk of insulin resistance (IR), which may precede the onset of type 2 diabetes mellitus and associated metabolic complications. Being a heterogeneous metabolic condition, it is pertinent to understand the metabolites and metabolic pathways that are altered during the development and progression of IR toward T2DM. Serum samples were collected from C57BL/6J mice fed with HFD or chow diet (CD) for 16 weeks. Collected samples were analyzed by gas chromatography-tandem mass spectrometry (GC-MS/MS). Data on the identified raw metabolites were evaluated using a combination of univariate and multivariate statistical methods. Mice fed with HFD had glucose and insulin intolerance associated with impairment of insulin signaling in key metabolic tissues. From the GC-MS/MS analysis of serum samples, a total of 75 common annotated metabolites were identified between HFD- and CD-fed mice. In the t-test, 22 significantly altered metabolites were identified. Out of these, 16 metabolites were up-accumulated, whereas 6 metabolites were down-accumulated. Pathway analysis identified 4 significantly altered metabolic pathways. In particular, primary bile acid biosynthesis and linoleic acid metabolism were upregulated, whereas the TCA cycle and pentose and glucuronate interconversion were downregulated in HFD-fed mice in comparison to CD-fed mice. These results show the distinct metabolic profiles associated with the onset of IR that could provide promising metabolic biomarkers for diagnostic and clinical applications.
Collapse
Affiliation(s)
- Manendra
Singh Tomar
- Centre
for Advance Research, Faculty of Medicine, King George’s Medical University, Lucknow 226003, India
| | - Aditya Sharma
- Division
of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Fabrizio Araniti
- Dipartimento
di Scienze Agrarie e Ambientali—Produzione, Territorio, Agroenergia, Università degli Studi di Milano, Via Celoria 2, 20133 Milano, Italy
| | - Ankit Pateriya
- Centre
for Advance Research, Faculty of Medicine, King George’s Medical University, Lucknow 226003, India
| | - Ashutosh Shrivastava
- Centre
for Advance Research, Faculty of Medicine, King George’s Medical University, Lucknow 226003, India
| | - Akhilesh Kumar Tamrakar
- Division
of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| |
Collapse
|
8
|
NMR-Based Metabolomic Analysis of Cardiac Tissues Clarifies Molecular Mechanisms of CVB3-Induced Viral Myocarditis and Dilated Cardiomyopathy. Molecules 2022; 27:molecules27186115. [PMID: 36144851 PMCID: PMC9500976 DOI: 10.3390/molecules27186115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Viral myocarditis (VMC), which is defined as inflammation of the myocardium with consequent myocardial injury, may develop chronic disease eventually leading to dilated cardiomyopathy (DCM). Molecular mechanisms underlying the progression from acute VMC (aVMC), to chronic VMC (cVMC) and finally to DCM, are still unclear. Here, we established mouse models of VMC and DCM with Coxsackievirus B3 infection and conducted NMR-based metabolomic analysis of aqueous metabolites extracted from cardiac tissues of three histologically classified groups including aVMC, cVMC and DCM. We showed that these three pathological groups were metabolically distinct from their normal counterparts and identified three impaired metabolic pathways shared by these pathological groups relative to normal controls, including nicotinate and nicotinamide metabolism; alanine, aspartate and glutamate metabolism; and D-glutamine and D-glutamate metabolism. We also identified two extra impaired metabolic pathways in the aVMC group, including glycine, serine and threonine metabolism; and taurine and hypotaurine metabolism Furthermore, we identified potential cardiac biomarkers for metabolically distinguishing these three pathological stages from normal controls. Our results indicate that the metabolomic analysis of cardiac tissues can provide valuable insights into the molecular mechanisms underlying the progression from acute VMC to DCM.
Collapse
|