1
|
Hu Y, Zou Y, Qiao L, Lin L. Integrative proteomic and metabolomic elucidation of cardiomyopathy with in vivo and in vitro models and clinical samples. Mol Ther 2024; 32:3288-3312. [PMID: 39233439 PMCID: PMC11489546 DOI: 10.1016/j.ymthe.2024.08.030] [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: 04/30/2024] [Revised: 07/16/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Cardiomyopathy is a prevalent cardiovascular disease that affects individuals of all ages and can lead to life-threatening heart failure. Despite its variety in types, each with distinct characteristics and causes, our understanding of cardiomyopathy at a systematic biology level remains incomplete. Mass spectrometry-based techniques have emerged as powerful tools, providing a comprehensive view of the molecular landscape and aiding in the discovery of biomarkers and elucidation of mechanisms. This review highlights the significant potential of integrating proteomic and metabolomic approaches with specialized databases to identify biomarkers and therapeutic targets across different types of cardiomyopathies. In vivo and in vitro models, such as genetically modified mice, patient-derived or induced pluripotent stem cells, and organ chips, are invaluable in exploring the pathophysiological complexities of this disease. By integrating omics approaches with these sophisticated modeling systems, our comprehension of the molecular underpinnings of cardiomyopathy can be greatly enhanced, facilitating the development of diagnostic markers and therapeutic strategies. Among the promising therapeutic targets are those involved in extracellular matrix remodeling, sarcomere damage, and metabolic remodeling. These targets hold the potential to advance precision therapy in cardiomyopathy, offering hope for more effective treatments tailored to the specific molecular profiles of patients.
Collapse
Affiliation(s)
- Yiwei Hu
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China
| | - Yunzeng Zou
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| | - Liang Qiao
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| | - Ling Lin
- Department of Chemistry, Zhongshan Hospital, and Minhang Hospital, Fudan University, Shanghai 200000, China.
| |
Collapse
|
2
|
Cho SW. Clinical Implications of a Proteomics-Based Approach for Cardiomyopathy and Heart Failure. Korean Circ J 2024; 54:482-484. [PMID: 39109596 PMCID: PMC11306424 DOI: 10.4070/kcj.2024.0194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Affiliation(s)
- Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea.
| |
Collapse
|
3
|
Zhao S, Liu J, Wu Q, Zhou X. Lactate regulates pathological cardiac hypertrophy via histone lactylation modification. J Cell Mol Med 2024; 28:e70022. [PMID: 39205384 PMCID: PMC11358213 DOI: 10.1111/jcmm.70022] [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: 11/06/2023] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Under the long-term pressure overload stimulation, the heart experiences embryonic gene activation, leading to myocardial hypertrophy and ventricular remodelling, which can ultimately result in the development of heart failure. Identifying effective therapeutic targets is crucial for the prevention and treatment of myocardial hypertrophy. Histone lysine lactylation (HKla) is a novel post-translational modification that connects cellular metabolism with epigenetic regulation. However, the specific role of HKla in pathological cardiac hypertrophy remains unclear. Our study aims to investigate whether HKla modification plays a pathogenic role in the development of cardiac hypertrophy. The results demonstrate significant expression of HKla in cardiomyocytes derived from an animal model of cardiac hypertrophy induced by transverse aortic constriction surgery, and in neonatal mouse cardiomyocytes stimulated by Ang II. Furthermore, research indicates that HKla is influenced by glucose metabolism and lactate generation, exhibiting significant phenotypic variability in response to various environmental stimuli. In vitro experiments reveal that exogenous lactate and glucose can upregulate the expression of HKla and promote cardiac hypertrophy. Conversely, inhibition of lactate production using glycolysis inhibitor (2-DG), LDH inhibitor (oxamate) and LDHA inhibitor (GNE-140) reduces HKla levels and inhibits the development of cardiac hypertrophy. Collectively, these findings establish a pivotal role for H3K18la in pathological cardiac hypertrophy, offering a novel target for the treatment of this condition.
Collapse
Affiliation(s)
- Shuai‐Shuai Zhao
- Department of Cardiac Surgery, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Jinlong Liu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
| | - Qi‐Cai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xue‐Liang Zhou
- Department of Cardiac Surgery, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
4
|
Jurida L, Werner S, Knapp F, Niemann B, Li L, Grün D, Wirth S, Weber A, Beuerlein K, Liebetrau C, Wiedenroth CB, Guth S, Kojonazarov B, Jafari L, Weissmann N, Günther S, Braun T, Bartkuhn M, Schermuly RT, Dorfmüller P, Yin X, Mayr M, Schmitz ML, Czech L, Schlüter KD, Schulz R, Rohrbach S, Kracht M. A common gene signature of the right ventricle in failing rat and human hearts. NATURE CARDIOVASCULAR RESEARCH 2024; 3:819-840. [PMID: 39196177 PMCID: PMC11358011 DOI: 10.1038/s44161-024-00485-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 05/02/2024] [Indexed: 08/29/2024]
Abstract
The molecular mechanisms of progressive right heart failure are incompletely understood. In this study, we systematically examined transcriptomic changes occurring over months in isolated cardiomyocytes or whole heart tissues from failing right and left ventricles in rat models of pulmonary artery banding (PAB) or aortic banding (AOB). Detailed bioinformatics analyses resulted in the identification of gene signature, protein and transcription factor networks specific to ventricles and compensated or decompensated disease states. Proteomic and RNA-FISH analyses confirmed PAB-mediated regulation of key genes and revealed spatially heterogeneous mRNA expression in the heart. Intersection of rat PAB-specific gene sets with transcriptome datasets from human patients with chronic thromboembolic pulmonary hypertension (CTEPH) led to the identification of more than 50 genes whose expression levels correlated with the severity of right heart disease, including multiple matrix-regulating and secreted factors. These data define a conserved, differentially regulated genetic network associated with right heart failure in rats and humans.
Collapse
Affiliation(s)
- Liane Jurida
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Sebastian Werner
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Fabienne Knapp
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Bernd Niemann
- Department of Cardiac and Vascular Surgery, Justus Liebig University, Giessen, Germany
| | - Ling Li
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Dimitri Grün
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Stefanie Wirth
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Axel Weber
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Knut Beuerlein
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Christoph Liebetrau
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | | | - Stefan Guth
- Department of Thoracic Surgery, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Baktybek Kojonazarov
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Leili Jafari
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Norbert Weissmann
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Günther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marek Bartkuhn
- Cardio-Pulmonary Institute, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Peter Dorfmüller
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Department of Internal Medicine, Justus Liebig University Giessen, Giessen, Germany
- Institute of Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Xiaoke Yin
- National Heart and Lung Institute, Faculty of Medicine,Imperial College London, London, UK
| | - Manuel Mayr
- National Heart and Lung Institute, Faculty of Medicine,Imperial College London, London, UK
| | - M Lienhard Schmitz
- German Center for Lung Research (DZL), Giessen, Germany
- Institute of Biochemistry, Justus Liebig University, Giessen, Germany
| | - Laureen Czech
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | | | - Rainer Schulz
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Susanne Rohrbach
- Department of Physiology, Justus Liebig University, Giessen, Germany.
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany.
- Cardio-Pulmonary Institute, Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.
- German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
5
|
Zhang F, Zhou H, Xue J, Zhang Y, Zhou L, Leng J, Fang G, Liu Y, Wang Y, Liu H, Wu Y, Qi L, Duan R, He X, Wang Y, Liu Y, Li L, Yang J, Liang D, Chen YH. Deficiency of Transcription Factor Sp1 Contributes to Hypertrophic Cardiomyopathy. Circ Res 2024; 134:290-306. [PMID: 38197258 DOI: 10.1161/circresaha.123.323272] [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: 06/28/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most prevalent monogenic heart disorder. However, the pathogenesis of HCM, especially its nongenetic mechanisms, remains largely unclear. Transcription factors are known to be involved in various biological processes including cell growth. We hypothesized that SP1 (specificity protein 1), the first purified TF in mammals, plays a role in the cardiomyocyte growth and cardiac hypertrophy of HCM. METHODS Cardiac-specific conditional knockout of Sp1 mice were constructed to investigate the role of SP1 in the heart. The echocardiography, histochemical experiment, and transmission electron microscope were performed to analyze the cardiac phenotypes of cardiac-specific conditional knockout of Sp1 mice. RNA sequencing, chromatin immunoprecipitation sequencing, and adeno-associated virus experiments in vivo were performed to explore the downstream molecules of SP1. To examine the therapeutic effect of SP1 on HCM, an SP1 overexpression vector was constructed and injected into the mutant allele of Myh6 R404Q/+ (Myh6 c. 1211C>T) HCM mice. The human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from a patient with HCM were used to detect the potential therapeutic effects of SP1 in human HCM. RESULTS The cardiac-specific conditional knockout of Sp1 mice developed a typical HCM phenotype, displaying overt myocardial hypertrophy, interstitial fibrosis, and disordered myofilament. In addition, Sp1 knockdown dramatically increased the cell area of hiPSC-CMs and caused intracellular myofibrillar disorganization, which was similar to the hypertrophic cardiomyocytes of HCM. Mechanistically, Tuft1 was identified as the key target gene of SP1. The hypertrophic phenotypes induced by Sp1 knockdown in both hiPSC-CMs and mice could be rescued by TUFT1 (tuftelin 1) overexpression. Furthermore, SP1 overexpression suppressed the development of HCM in the mutant allele of Myh6 R404Q/+ mice and also reversed the hypertrophic phenotype of HCM hiPSC-CMs. CONCLUSIONS Our study demonstrates that SP1 deficiency leads to HCM. SP1 overexpression exhibits significant therapeutic effects on both HCM mice and HCM hiPSC-CMs, suggesting that SP1 could be a potential intervention target for HCM.
Collapse
Affiliation(s)
- Fulei Zhang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Huixing Zhou
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Jinfeng Xue
- Department of Regenerative Medicine (J.X., L.Q.), Tongji University School of Medicine, Shanghai, China
| | - Yuemei Zhang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Liping Zhou
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Junwei Leng
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Guojian Fang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yuanyuan Liu
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Yan Wang
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Hongyu Liu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yahan Wu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Lingbin Qi
- Department of Regenerative Medicine (J.X., L.Q.), Tongji University School of Medicine, Shanghai, China
| | - Ran Duan
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Xiaoyu He
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yan Wang
- Jinzhou Medical University, China (Yuanyuan Liu, Y. Wang, Yan Wang)
| | - Yi Liu
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Li Li
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Jian Yang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Dandan Liang
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| | - Yi-Han Chen
- State Key Laboratory of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Shanghai Arrhythmias Research Center (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., Yuanyuan Liu, Y. Wang, H.L., Y. Wu, R.D., X.H., Yi Liu, L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Cardiology (F.Z., H.Z., Y.Z., L.Z., J.L., G.F., H.L., Y. Wu, R.D., X.H., L.L., J.Y., D.L., Y.-H.C.), Shanghai East Hospital, Tongji University School of Medicine, China
- Department of Pathology and Pathophysiology (L.L., J.Y., Y.-H.C.), Tongji University School of Medicine, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China (L.L., J.Y., D.L., Y.-H.C.)
| |
Collapse
|
6
|
Russo MA, Garaci E, Frustaci A, Fini M, Costantini C, Oikonomou V, Nunzi E, Puccetti P, Romani L. Host-microbe tryptophan partitioning in cardiovascular diseases. Pharmacol Res 2023; 198:106994. [PMID: 37972721 DOI: 10.1016/j.phrs.2023.106994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
The functional interdependencies between the molecular components of a biological process demand for a network medicine platform that integrates systems biology and network science, to explore the interactions among biological components in health and disease. Access to large-scale omics datasets (genomics, transcriptomics, proteomics, metabolomics, metagenomics, phenomics, etc.) has significantly advanced our opportunity along this direction. Studies utilizing these techniques have begun to provide us with a deeper understanding of how the interaction between the intestinal microbes and their host affects the cardiovascular system in health and disease. Within the framework of a multiomics network approach, we highlight here how tryptophan metabolism may orchestrate the host-microbes interaction in cardiovascular diseases and the implications for precision medicine and therapeutics, including nutritional interventions.
Collapse
Affiliation(s)
- Matteo Antonio Russo
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Andrea Frustaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Massimo Fini
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Vasileios Oikonomou
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Luigina Romani
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, 00166 Rome, Italy; Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy.
| |
Collapse
|
7
|
Spoladore R, Pinto G, Daus F, Pezzini S, Kolios D, Fragasso G. Metabolic Approaches for the Treatment of Dilated Cardiomyopathy. J Cardiovasc Dev Dis 2023; 10:287. [PMID: 37504543 PMCID: PMC10380730 DOI: 10.3390/jcdd10070287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
In dilated cardiomyopathy (DCM), where the heart muscle becomes stretched and thin, heart failure (HF) occurs, and the cardiomyocytes suffer from an energetic inefficiency caused by an abnormal cardiac metabolism. Although underappreciated as a potential therapeutic target, the optimal metabolic milieu of a failing heart is still largely unknown and subject to debate. Because glucose naturally has a lower P/O ratio (the ATP yield per oxygen atom), the previous studies using this strategy to increase glucose oxidation have produced some intriguing findings. In reality, the vast majority of small-scale pilot trials using trimetazidine, ranolazine, perhexiline, and etomoxir have demonstrated enhanced left ventricular (LV) function and, in some circumstances, myocardial energetics in chronic ischemic and non-ischemic HF with a reduced ejection fraction (EF). However, for unidentified reasons, none of these drugs has ever been tested in a clinical trial of sufficient size. Other pilot studies came to the conclusion that because the heart in severe dilated cardiomyopathy appears to be metabolically flexible and not limited by oxygen, the current rationale for increasing glucose oxidation as a therapeutic target is contradicted and increasing fatty acid oxidation is supported. As a result, treating metabolic dysfunction in HF may benefit from raising ketone body levels. Interestingly, treatment with sodium-glucose cotransporter-2 inhibitors (SGLT2i) improves cardiac function and outcomes in HF patients with or without type 2 diabetes mellitus (T2DM) through a variety of pleiotropic effects, such as elevated ketone body levels. The improvement in overall cardiac function seen in patients receiving SGLT2i could be explained by this increase, which appears to be a reflection of an adaptive process that optimizes cardiac energy metabolism. This review aims to identify the best metabolic therapeutic approach for DCM patients, to examine the drugs that directly affect cardiac metabolism, and to outline all the potential ancillary metabolic effects of the guideline-directed medical therapy. In addition, a special focus is placed on SGLT2i, which were first studied and prescribed to diabetic patients before being successfully incorporated into the pharmacological arsenal for HF patients.
Collapse
Affiliation(s)
- Roberto Spoladore
- Department of Cardiology, Heart Failure Clinic, Alessandro Manzoni Hospital, ASST Lecco, 23900 Lecco, Italy
| | - Giuseppe Pinto
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Francesca Daus
- Post-Graduate School of Cardiovascular Medicine, Milan-Bicocca University, 20126 Milan, Italy
| | - Sara Pezzini
- Post-Graduate School of Cardiovascular Medicine, Milan-Bicocca University, 20126 Milan, Italy
| | - Damianos Kolios
- Department of Clinical Cardiology, Heart Failure Clinic, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy (G.F.)
| | - Gabriele Fragasso
- Department of Clinical Cardiology, Heart Failure Clinic, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy (G.F.)
| |
Collapse
|
8
|
Visconti G, de Figueiredo M, Salamin O, Boccard J, Vuilleumier N, Nicoli R, Kuuranne T, Rudaz S. Straightforward quantification of endogenous steroids with liquid chromatography-tandem mass spectrometry: Comparing calibration approaches. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1226:123778. [PMID: 37393882 DOI: 10.1016/j.jchromb.2023.123778] [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: 02/14/2023] [Revised: 05/23/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023]
Abstract
Different calibration strategies are used in liquid chromatography hyphenated to mass spectrometry (LC-MS) bioanalysis. Currently, the surrogate matrix and surrogate analyte represent the most widely used approaches to compensate for the lack of analyte-free matrices in endogenous compounds quantification. In this context, there is a growing interest in rationalizing and simplifying quantitative analysis using a one-point concentration level of stable isotope-labeled (SIL) standards as surrogate calibrants. Accordingly, an internal calibration (IC) can be applied when the instrument response is translated into analyte concentration via the analyte-to-SIL ratio performed directly in the study sample. Since SILs are generally used as internal standards to normalize variability between authentic study sample matrix and surrogate matrix used for the calibration, IC can be calculated even if the calibration protocol was achieved for an external calibration (EC). In this study, a complete dataset of a published and fully validated method to quantify an extended steroid profile in serum was recomputed by adapting the role of SIL internal standards as surrogate calibrants. Using the validation samples, the quantitative performances for IC were comparable with the original method, showing acceptable trueness (79%-115%) and precision (0.8%-11.8%) for the 21 detected steroids. The IC methodology was then applied to human serum samples (n = 51) from healthy women and women diagnosed with mild hyperandrogenism, showing high agreement (R2 > 0.98) with the concentrations obtained using the conventional quantification based on EC. For IC, Passing-Bablok regression showed proportional biases between -15.0% and 11.3% for all quantified steroids, with an average difference of -5.8% compared to EC. These results highlight the reliability and the advantages of implementing IC in clinical laboratories routine to simplify quantification in LC-MS bioanalysis, especially when a large panel of analytes is monitored.
Collapse
Affiliation(s)
- Gioele Visconti
- School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel-Servet 1, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel-Servet 1, Geneva, Switzerland
| | - Miguel de Figueiredo
- School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel-Servet 1, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel-Servet 1, Geneva, Switzerland
| | - Olivier Salamin
- Center of Research and Expertise in Anti-Doping Sciences - REDs, Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland; Swiss Laboratory for Doping Analyses, University Center of Legal Medicine, Lausanne and Geneva, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Julien Boccard
- School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel-Servet 1, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel-Servet 1, Geneva, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland
| | - Nicolas Vuilleumier
- Department of Genetic and Laboratory Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Raul Nicoli
- Swiss Laboratory for Doping Analyses, University Center of Legal Medicine, Lausanne and Geneva, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Tiia Kuuranne
- Swiss Laboratory for Doping Analyses, University Center of Legal Medicine, Lausanne and Geneva, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel-Servet 1, Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel-Servet 1, Geneva, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
| |
Collapse
|
9
|
Autore C, Bariani R, Bauce B, Biagini E, Canepa M, Castelletti S, Crotti L, Limongelli G, Merlo M, Monda E, Pio Loco Detto Gava C, Parisi V, Tini G, Imazio M. From the phenotype to precision medicine: an update on the cardiomyopathies diagnostic workflow. J Cardiovasc Med (Hagerstown) 2023; 24:e178-e186. [PMID: 37186568 DOI: 10.2459/jcm.0000000000001424] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Cardiomyopathies are disease of the cardiac muscle largely due to genetic alterations of proteins with 'structural' or 'functional' roles within the cardiomyocyte, going from the regulation of contraction-relaxation, metabolic and energetic processes to ionic fluxes. Modifications occurring to these proteins are responsible, in the vast majority of cases, for the phenotypic manifestations of the disease, including hypertrophic, dilated, arrhythmogenic and restrictive cardiomyopathies. Secondary nonhereditary causes to be excluded include infections, toxicity from drugs or alcohol or medications, hormonal imbalance and so on. Obtaining a phenotypic definition and an etiological diagnosis is becoming increasingly relevant and feasible, thanks to the availability of new tailored treatments and the diagnostic advancements made particularly in the field of genetics. This is, for example, the case for transthyretin cardiac amyloidosis, Fabry disease or dilated cardiomyopathies due to laminopathies. For these diseases, specific medications have been developed, and a more tailored arrhythmic risk stratification guides the implantation of a defibrillator. In addition, new medications directly targeting the altered protein responsible for the phenotype are becoming available (including the myosin inhibitors mavacantem and aficamten, monoclonal antibodies against Ras-MAPK, genetic therapies for sarcoglycanopathies), thus making a precision medicine approach less unrealistic even in the field of cardiomyopathies. For these reasons, a contemporary approach to cardiomyopathies must consider diagnostic algorithms founded on the clinical suspicion of the disease and developed towards a more precise phenotypic definition and etiological diagnosis, based on a multidisciplinary methodology putting together specialists from different disciplines, facilities for advanced imaging testing and genetic and anatomopathological competencies.
Collapse
Affiliation(s)
- Camillo Autore
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome
| | - Riccardo Bariani
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua
| | - Barbara Bauce
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua
| | - Elena Biagini
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy and European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart
| | - Marco Canepa
- Department of Internal Medicine, Università degli Studi di Genova
- Cardiovascular Unit, IRCCS Ospedale Policlinico San Martino, Genova
| | - Silvia Castelletti
- Istituto Auxologico Italiano, IRCCS San Luca Hospital, Cardiology Department Milan
| | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS San Luca Hospital, Cardiology Department Milan
- University of Milano-Bicocca, Department of Medicine and Surgery, Milan
| | - Giuseppe Limongelli
- Dipartimento di Scienze Mediche Traslazionali -Università della Campania 'Luigi Vanvitelli' - Osp. Monaldi, AORN Colli, Ospedale Monaldi, Napoli
| | - Marco Merlo
- Centre for Diagnosis and Management of Cardiomyopathy, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and University of Trieste, Trieste
| | - Emanuele Monda
- Dipartimento di Scienze Mediche Traslazionali -Università della Campania 'Luigi Vanvitelli' - Osp. Monaldi, AORN Colli, Ospedale Monaldi, Napoli
| | - Carola Pio Loco Detto Gava
- Centre for Diagnosis and Management of Cardiomyopathy, Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and University of Trieste, Trieste
| | - Vanda Parisi
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy and European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart-ERN GUARD-Heart
| | - Giacomo Tini
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome
| | - Massimo Imazio
- Dipartimento Cardiotoracico, Ospedale Santa Maria della Misericordia, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| |
Collapse
|
10
|
Li X, Shen Y, Xu X, Guo G, Chen Y, Wei Q, Li H, He K, Liu C. Genomic and RNA-Seq profiling of patients with HFrEF unraveled OAS1 mutation and aggressive expression. Int J Cardiol 2023; 375:44-54. [PMID: 36414043 DOI: 10.1016/j.ijcard.2022.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Heart failure (HF) is a complex pathophysiological state characterized by inadequate delivery of blood and nutrients to the cardiac tissues. It is rarely curable and is commonly associated with a poor prognosis. In this study, we aimed to analyse exomic and RNA-Seq data from patients with HF to identify the key altered pathways in HF. METHODS Whole blood samples were collected from patients with HF and subjected to whole exome sequencing (WES) and RNA-Seq analysis. The gene expression and RNA-Seq data obtained were verified using gene chip analysis and RT-PCR. RESULTS Both exomic and RNA-Seq data confirmed the dysregulation of phosphorylation and immune signalling in patients with HF. Specifically, exomic analysis showed that TITIN, OBSCURIN, NOD2, CDH2, MAP3K5, and SLC17A4 mutations were associated with HF, and RNA-Seq revealed that S100A12, S100A8, S100A9, PFDN5, and TMCC2, were upregulated in patients with HF. Additionally, comparison between RNA-seq and WES data showed that OAS1 mutations are associated with HF. CONLCUSION Our findings indicated that patients with HF show an overall disruption of key phosphorylation and immune signalling pathways. Based on RNA-seq and WES, OAS1 mutations may be primarily responsible for these changes.
Collapse
Affiliation(s)
- Xin Li
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China; Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing 100853, China
| | - Yanying Shen
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiang Xu
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Ge Guo
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Yibing Chen
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Qingxia Wei
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Hanlu Li
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Kunlun He
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing 100853, China; Medical Big Data Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China.
| | - Chunlei Liu
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China; Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing 100853, China.
| |
Collapse
|
11
|
Joshua J, Caswell J, O’Sullivan ML, Wood G, Fonfara S. Feline myocardial transcriptome in health and in hypertrophic cardiomyopathy-A translational animal model for human disease. PLoS One 2023; 18:e0283244. [PMID: 36928240 PMCID: PMC10019628 DOI: 10.1371/journal.pone.0283244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heart disease in cats, characterized by primary left ventricular hypertrophy. Feline HCM closely resembles human HCM and is suggested as translational animal model for the human disease. A genetic cause is established in humans and suspected for cats, but little is known about the gene expression and pathways involved in the pathogenesis of HCM. To investigate the myocardial transcriptome changes in HCM, RNA sequencing was conducted on left ventricle (LV) and left atrium (LA) samples of healthy cats and cats with HCM (each n = 5; 20 samples). Ingenuity Pathway Analysis was used to determine functional pathways, regulators, and networks. Distinct gene expression profiles were identified in the LV and LA of the feline healthy and HCM myocardium. Analysis of differentially expressed mRNAs (>2 fold; FDR < 0.01) found chamber-specific (LV vs. LA) expression in both healthy and HCM groups, with higher transcriptional activity in the LA. Genes that contribute to the distinct structure and function of each chamber in health and HCM were identified in the regional comparison. The gene expression profiles of HCM compared to healthy hearts revealed disease related genes, including THBS4 and KLHL33 (LV), FAM177B and THRSP (LA), the latter 3 have not been reported for the myocardium so far, as the top differently expressed genes in the HCM heart. Differently expressed genes and functional pathways found in the HCM heart are associated with cardiac remodeling and fibrosis, inflammation, microvascular changes, calcium signaling and cardiac metabolism, with some regional differences. RhoGDI-RhoGTPase signaling, integrin and ILK signaling pathways, the LXR/RXR pathway in the LA, and the PPARα/RXRα, HIF1α and CXCR4 pathways in the LV might be of particular importance in the HCM disease process. This study identified region-specific myocardial gene transcription patterns as well as novel genes and pathways associated with HCM.
Collapse
Affiliation(s)
- Jessica Joshua
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph, Ontario, Canada
| | - Jeff Caswell
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
| | - M. Lynne O’Sullivan
- University of Prince Edward Island, Department of Companion Animals, Charlottetown, Prince Edward Island, Canada
| | - Geoffrey Wood
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
| | - Sonja Fonfara
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph, Ontario, Canada
- * E-mail:
| |
Collapse
|
12
|
Valero-Muñoz M, Saw EL, Hekman RM, Blum BC, Hourani Z, Granzier H, Emili A, Sam F. Proteomic and phosphoproteomic profiling in heart failure with preserved ejection fraction (HFpEF). Front Cardiovasc Med 2022; 9:966968. [PMID: 36093146 PMCID: PMC9452734 DOI: 10.3389/fcvm.2022.966968] [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: 06/12/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Although the prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing, evidence-based therapies for HFpEF remain limited, likely due to an incomplete understanding of this disease. This study sought to identify the cardiac-specific features of protein and phosphoprotein changes in a murine model of HFpEF using mass spectrometry. HFpEF mice demonstrated moderate hypertension, left ventricle (LV) hypertrophy, lung congestion and diastolic dysfunction. Proteomics analysis of the LV tissue showed that 897 proteins were differentially expressed between HFpEF and Sham mice. We observed abundant changes in sarcomeric proteins, mitochondrial-related proteins, and NAD-dependent protein deacetylase sirtuin-3 (SIRT3). Upregulated pathways by GSEA analysis were related to immune modulation and muscle contraction, while downregulated pathways were predominantly related to mitochondrial metabolism. Western blot analysis validated SIRT3 downregulated cardiac expression in HFpEF vs. Sham (0.8 ± 0.0 vs. 1.0 ± 0.0; P < 0.001). Phosphoproteomics analysis showed that 72 phosphosites were differentially regulated between HFpEF and Sham LV. Aberrant phosphorylation patterns mostly occurred in sarcomere proteins and nuclear-localized proteins associated with contractile dysfunction and cardiac hypertrophy. Seven aberrant phosphosites were observed at the z-disk binding region of titin. Additional agarose gel analysis showed that while total titin cardiac expression remained unaltered, its stiffer N2B isoform was significantly increased in HFpEF vs. Sham (0.144 ± 0.01 vs. 0.127 ± 0.01; P < 0.05). In summary, this study demonstrates marked changes in proteins related to mitochondrial metabolism and the cardiac contractile apparatus in HFpEF. We propose that SIRT3 may play a role in perpetuating these changes and may be a target for drug development in HFpEF.
Collapse
Affiliation(s)
- María Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Eng Leng Saw
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| | - Ryan M. Hekman
- Department of Biology, Boston University, Boston, MA, United States
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
| | - Benjamin C. Blum
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Zaynab Hourani
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, United States
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, United States
| | - Andrew Emili
- Department of Biology, Boston University, Boston, MA, United States
- Department of Biochemistry, Cell Biology and Genomics, Boston University, Boston, MA, United States
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
13
|
Weighted Gene Co-Expression Network Analysis to Identify Potential Biological Processes and Key Genes in COVID-19-Related Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4526022. [PMID: 35557984 PMCID: PMC9088964 DOI: 10.1155/2022/4526022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 12/11/2022]
Abstract
The purpose of this research was to explore the underlying biological processes causing coronavirus disease 2019- (COVID-19-) related stroke. The Gene Expression Omnibus (GEO) database was utilized to obtain four COVID-19 datasets and two stroke datasets. Thereafter, we identified key modules via weighted gene co-expression network analysis, following which COVID-19- and stroke-related crucial modules were crossed to identify the common genes of COVID-19-related stroke. The common genes were intersected with the stroke-related hub genes screened via Cytoscape software to discover the critical genes associated with COVID-19-related stroke. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis for common genes associated with COVID-19-related stroke, and the Reactome database was used to annotate and visualize the pathways involved in the key genes. Two COVID-19-related crucial modules and one stroke-related crucial module were identified. Subsequently, the top five genes were screened as hub genes after visualizing the genes of stroke-related critical module using Cytoscape. By intersecting the COVID-19- and stroke-related crucial modules, 28 common genes for COVID-19-related stroke were identified. ITGA2B and ITGB3 have been further identified as crucial genes of COVID-19-related stroke. Functional enrichment analysis indicated that both ITGA2B and ITGB3 were involved in integrin signaling and the response to elevated platelet cytosolic Ca2+, thus regulating platelet activation, extracellular matrix- (ECM-) receptor interaction, the PI3K-Akt signaling pathway, and hematopoietic cell lineage. Therefore, platelet activation, ECM-receptor interaction, PI3K-Akt signaling pathway, and hematopoietic cell lineage may represent the potential biological processes associated with COVID-19-related stroke, and ITGA2B and ITGB3 may be potential intervention targets for COVID-19-related stroke.
Collapse
|
14
|
Ampong I. Metabolic and metabolomics insights into dilated cardiomyopathy (DCM). ANNALS OF NUTRITION AND METABOLISM 2022; 78:147-155. [PMID: 35472668 DOI: 10.1159/000524722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/23/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is the most common form of heart muscle disease characterized by progressive dilatation and ventricular dysfunction. Metabolomics is an emerging and powerful discipline that provides a global information on the phenotype of mammalian systems via the study of endogenous and exogenous metabolites in cells, tissues and biofluids. These studies aid in the identification of biomarkers to prevent diseases in later life or help to early detect onset of diseases as well as aiding in the elucidation of disease mechanisms. SUMMARY Metabolomics provides a unique opportunity to discover biomarkers for DCM. This review demonstrates evidence of metabolite-based biomarkers useful for predicting, diagnosing and monitoring therapeutic interventions of DCM. Key metabolites identified as potential biomarkers for diagnosing DCM include acyl-carnitines, succinic acid, malate, methylhistidine, aspartate, methionine, phenylalanine. In terms of differentiating DCM from ICM, potential biomarkers including 1-pyrroline-2-carboxylate, norvaline, lysophosphatidylinositol (16:0/0:0), phosphatidylglycerol, fatty acid esters of hydroxy fatty acid, and phosphatidylcholine were identified. Acyl-carnitines, isoleucine and linoleic acid and tryptophan were the main biomarkers to monitor treatment response to DCM. Mapping metabolites to metabolic pathways revealed dysregulation of BCAA, glycolysis, tricarboxylic acid cycle and triacylglycerol and pentose phosphate metabolism which have therapeutic potential for DCM. This review shows several limitations including the use of small sample sizes, lack of interpretation of age and sex differences in most studies and the fact that studies have so far been limited to case-control study designs. KEY MESSAGES Metabolites have close proximity to disease phenotype. With recent advancements in metabolomics field, potential biomarkers for DCM have been identified based on studies using different biological and metabolomics technologies. However, multi-center studies with larger populations that will lead to validation of these identified biomarkers to enable their clinical translation and utilization are still needed.
Collapse
Affiliation(s)
- Isaac Ampong
- Center for Precision Medicine, Wake Forest University Baptist Medical Center, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| |
Collapse
|
15
|
Huang K, Zhang X, Duan J, Wang R, Wu Z, Yang C, Yang L. STAT4 and COL1A2 are potential diagnostic biomarkers and therapeutic targets for heart failure comorbided with depression. Brain Res Bull 2022; 184:68-75. [PMID: 35367598 DOI: 10.1016/j.brainresbull.2022.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Heart failure (HF) and depression are common disorders that markedly compromise quality of life and impose a great financial burden on the society. Although increasing evidence has supported the closely linkage between the two disorders, the comorbidity mechanisms remain to be fully illuminated. We performed a bioinformatics network analysis to understand potential diagnostic biomarkers and therapeutic targets for HF comorbided with depression. METHODS We downloaded the datasets of HF and depression from the Gene Expression Omnibus (GEO) database and constructed co-expression networks by Weighted Gene Co-Expression Network Analysis (WGCNA) to identify key modules. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed on the common genes existing in the HF and depression related modules. Then, we employed the STRING database to construct the protein-protein interaction (PPI) network and detected the hub genes in the network. Finally, we validated the expression difference of hub genes from additional datasets of HF and depression. RESULTS Functional enrichment analysis indicated that platelet activation, chemokine signaling and focal adhesion were probably involved in HF comorbided with depression. PPI network construction indicated that HF comorbided with depression is likely related to 5 hub genes, including STAT4, CD83, CX3CR1, COL1A2, and SH2D1B. In validated datasets, STAT4 and COL1A2 were especially involved in the comorbidity of HF and depression. CONCLUSION Our work indicated a total of 5 hub genes including STAT4, CD83, CX3CR1, COL1A2, and SH2D1B, in which STAT4 and COL1A2 especially underlie the comorbidity mechanisms of HF and depression. These shared pathways might provide new targets for further mechanistic studies of the pathogenesis and treatment of HF and depression.
Collapse
Affiliation(s)
- Kai Huang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xinying Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiahao Duan
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Ruting Wang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zifeng Wu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| |
Collapse
|
16
|
Xu Z, Lv B, Qin Y, Zhang B. Emerging Roles and Mechanism of m6A Methylation in Cardiometabolic Diseases. Cells 2022; 11:cells11071101. [PMID: 35406663 PMCID: PMC8997388 DOI: 10.3390/cells11071101] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/12/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
Cardiometabolic diseases (CMDs) are currently the leading cause of death and disability worldwide, and their underlying regulatory mechanisms remain largely unknown. N6-methyladenosine (m6A) methylation, the most common and abundant epigenetic modification of eukaryotic mRNA, is regulated by m6A methyltransferase, demethylase, and the m6A binding protein, which affect the transcription, cleavage, translation, and degradation of target mRNA. m6A methylation plays a vital role in the physiological and pathological processes of CMDs. In this review, we summarize the role played by m6A methylation in CMDs, including obesity, hypertension, pulmonary hypertension, ischemic heart disease, myocardial hypertrophy, heart failure, and atherosclerosis. We also describe mechanisms that potentially involve the participation of m6A methylation, such as those driving calcium homeostasis, circadian rhythm, lipid metabolism, autophagy, macrophage response, and inflammation. m6A methylation and its regulators are expected to be targets for the treatment of CMDs.
Collapse
|
17
|
Osmak G, Baulina N, Kiselev I, Favorova O. MiRNA-Regulated Pathways for Hypertrophic Cardiomyopathy: Network-Based Approach to Insight into Pathogenesis. Genes (Basel) 2021; 12:genes12122016. [PMID: 34946964 PMCID: PMC8701189 DOI: 10.3390/genes12122016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/26/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common hereditary heart disease. The wide spread of high-throughput sequencing casts doubt on its monogenic nature, suggesting the presence of mechanisms of HCM development independent from mutations in sarcomeric genes. From this point of view, HCM may arise from the interactions of several HCM-associated genes, and from disturbance of regulation of their expression. We developed a bioinformatic workflow to study the involvement of signaling pathways in HCM development through analyzing data on human heart-specific gene expression, miRNA-target gene interactions, and protein-protein interactions, available in open databases. Genes regulated by a pool of miRNAs contributing to human cardiac hypertrophy, namely hsa-miR-1-3p, hsa-miR-19b-3p, hsa-miR-21-5p, hsa-miR-29a-3p, hsa-miR-93-5p, hsa-miR-133a-3p, hsa-miR-155-5p, hsa-miR-199a-3p, hsa-miR-221-3p, hsa-miR-222-3p, hsa-miR-451a, and hsa-miR-497-5p, were considered. As a result, we pinpointed a module of TGFβ-mediated SMAD signaling pathways, enriched by targets of the selected miRNAs, that may contribute to the cardiac remodeling in HCM. We suggest that the developed network-based approach could be useful in providing a more accurate glimpse on pathological processes in the disease pathogenesis.
Collapse
Affiliation(s)
- German Osmak
- Laboratory of Functional Genomics of Cardiovascular Disorders, National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.B.); (I.K.); (O.F.)
- Laboratory of Medical Genomics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Correspondence:
| | - Natalia Baulina
- Laboratory of Functional Genomics of Cardiovascular Disorders, National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.B.); (I.K.); (O.F.)
- Laboratory of Medical Genomics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Ivan Kiselev
- Laboratory of Functional Genomics of Cardiovascular Disorders, National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.B.); (I.K.); (O.F.)
- Laboratory of Medical Genomics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Olga Favorova
- Laboratory of Functional Genomics of Cardiovascular Disorders, National Medical Research Center for Cardiology, 121552 Moscow, Russia; (N.B.); (I.K.); (O.F.)
- Laboratory of Medical Genomics, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
18
|
Ranjbarvaziri S, Kooiker KB, Ellenberger M, Fajardo G, Zhao M, Vander Roest AS, Woldeyes RA, Koyano TT, Fong R, Ma N, Tian L, Traber GM, Chan F, Perrino J, Reddy S, Chiu W, Wu JC, Woo JY, Ruppel KM, Spudich JA, Snyder MP, Contrepois K, Bernstein D. Altered Cardiac Energetics and Mitochondrial Dysfunction in Hypertrophic Cardiomyopathy. Circulation 2021; 144:1714-1731. [PMID: 34672721 PMCID: PMC8608736 DOI: 10.1161/circulationaha.121.053575] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/24/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a complex disease partly explained by the effects of individual gene variants on sarcomeric protein biomechanics. At the cellular level, HCM mutations most commonly enhance force production, leading to higher energy demands. Despite significant advances in elucidating sarcomeric structure-function relationships, there is still much to be learned about the mechanisms that link altered cardiac energetics to HCM phenotypes. In this work, we test the hypothesis that changes in cardiac energetics represent a common pathophysiologic pathway in HCM. METHODS We performed a comprehensive multiomics profile of the molecular (transcripts, metabolites, and complex lipids), ultrastructural, and functional components of HCM energetics using myocardial samples from 27 HCM patients and 13 normal controls (donor hearts). RESULTS Integrated omics analysis revealed alterations in a wide array of biochemical pathways with major dysregulation in fatty acid metabolism, reduction of acylcarnitines, and accumulation of free fatty acids. HCM hearts showed evidence of global energetic decompensation manifested by a decrease in high energy phosphate metabolites (ATP, ADP, and phosphocreatine) and a reduction in mitochondrial genes involved in creatine kinase and ATP synthesis. Accompanying these metabolic derangements, electron microscopy showed an increased fraction of severely damaged mitochondria with reduced cristae density, coinciding with reduced citrate synthase activity and mitochondrial oxidative respiration. These mitochondrial abnormalities were associated with elevated reactive oxygen species and reduced antioxidant defenses. However, despite significant mitochondrial injury, HCM hearts failed to upregulate mitophagic clearance. CONCLUSIONS Overall, our findings suggest that perturbed metabolic signaling and mitochondrial dysfunction are common pathogenic mechanisms in patients with HCM. These results highlight potential new drug targets for attenuation of the clinical disease through improving metabolic function and reducing mitochondrial injury.
Collapse
Affiliation(s)
- Sara Ranjbarvaziri
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristina B. Kooiker
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA, USA
| | - Mathew Ellenberger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Giovanni Fajardo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Mingming Zhao
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Alison Schroer Vander Roest
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Rahel A. Woldeyes
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Robyn Fong
- Department of Cardiothoracic Surgery, Stanford University, CA, USA
| | - Ning Ma
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
| | - Lei Tian
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
| | - Gavin M. Traber
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Frandics Chan
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - John Perrino
- Cell Sciences Imaging Facility, Stanford University, Stanford, CA, USA
| | - Sushma Reddy
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Wah Chiu
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Division of Cryo-EM and Bioimaging, SLAC National Accelerator Laboratory, Stanford University, Stanford, CA, USA
| | - Joseph C. Wu
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
| | - Joseph Y. Woo
- Department of Cardiothoracic Surgery, Stanford University, CA, USA
| | - Kathleen M. Ruppel
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Bernstein
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
19
|
Liu C, Chen S, Zhang H, Chen Y, Gao Q, Chen Z, Liu Z, Wang J. Bioinformatic analysis for potential biological processes and key targets of heart failure-related stroke. J Zhejiang Univ Sci B 2021; 22:718-732. [PMID: 34514752 DOI: 10.1631/jzus.b2000544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This study aimed to uncover underlying mechanisms and promising intervention targets of heart failure (HF)-related stroke. HF-related dataset GSE42955 and stroke-related dataset GSE58294 were obtained from the Gene Expression Omnibus (GEO) database. Weighted gene co-expression network analysis (WGCNA) was conducted to identify key modules and hub genes. Gene Ontology (GO) and pathway enrichment analyses were performed on genes in the key modules. Genes in HF- and stroke-related key modules were intersected to obtain common genes for HF-related stroke, which were further intersected with hub genes of stroke-related key modules to obtain key genes in HF-related stroke. Key genes were functionally annotated through GO in the Reactome and Cytoscape databases. Finally, key genes were validated in these two datasets and other datasets. HF- and stroke-related datasets each identified two key modules. Functional enrichment analysis indicated that protein ubiquitination, Wnt signaling, and exosomes were involved in both HF- and stroke-related key modules. Additionally, ten hub genes were identified in stroke-related key modules and 155 genes were identified as common genes in HF-related stroke. OTU deubiquitinase with linear linkage specificity(OTULIN) and nuclear factor interleukin 3-regulated(NFIL3) were determined to be the key genes in HF-related stroke. Through functional annotation, OTULIN was involved in protein ubiquitination and Wnt signaling, and NFIL3 was involved in DNA binding and transcription. Importantly, OTULIN and NFIL3 were also validated to be differentially expressed in all HF and stroke groups. Protein ubiquitination, Wnt signaling, and exosomes were involved in HF-related stroke. OTULIN and NFIL3 may play a key role in HF-related stroke through regulating these processes, and thus serve as promising intervention targets.
Collapse
Affiliation(s)
- Chiyu Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Sixu Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Haifeng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China
| | - Yangxin Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China
| | - Qingyuan Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China
| | - Zhiteng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China
| | - Zhaoyu Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Jingfeng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China. .,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China.
| |
Collapse
|
20
|
Shen Q, Hiebert JB, Rahman FK, Krueger KJ, Gupta B, Pierce JD. Understanding Obesity-Related High Output Heart Failure and Its Implications. INTERNATIONAL JOURNAL OF HEART FAILURE 2021; 3:160-171. [PMID: 36262639 PMCID: PMC9536652 DOI: 10.36628/ijhf.2020.0047] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/17/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Morbid obesity remains most common cause of high output failure. The prevalence of the obesity is growing when two-thirds of American adults already are overweight or obese. Obesity is the risk factor for heart disease and eventually leads to heart failure. High output heart failure is common in obese patients and is characterized by high cardiac output, decreased systemic vascular resistance, and increased oxygen consumption. It often occurs in patients with chronic severe anemia, hyperthyroidism, pregnancy, arterial-venous fistulas, and liver disease. However, the pathogenesis of obesity-related high output heart failure is not fully understood. The clinical management of obesity-related high output heart failure follows conventional heart failure regimens due to lack of specific clinical recommendations. This article reviews the possible pathophysiological mechanisms and causes that contribute to obesity-related high output heart failure. This review also focuses on the implications for clinical practice and future research involved with omics technologies to explore possible molecular pathways associated with obesity-related high output heart failure.
Collapse
Affiliation(s)
- Qiuhua Shen
- University of Kansas Medical Center, School of Nursing, Kansas City, KS, USA
| | - John B. Hiebert
- University of Kansas Medical Center, School of Nursing, Kansas City, KS, USA
| | - Faith K. Rahman
- University of Kansas Medical Center, School of Nursing, Kansas City, KS, USA
| | - Kathryn J. Krueger
- University of Kansas Medical Center, School of Nursing, Kansas City, KS, USA
| | - Bhanu Gupta
- Department of Cardiovascular Medicine, The University of Kansas Health System, Kansas City, KS, USA
| | - Janet D. Pierce
- University of Kansas Medical Center, School of Nursing, Kansas City, KS, USA
| |
Collapse
|
21
|
Ramirez Flores RO, Lanzer JD, Holland CH, Leuschner F, Most P, Schultz J, Levinson RT, Saez‐Rodriguez J. Consensus Transcriptional Landscape of Human End-Stage Heart Failure. J Am Heart Assoc 2021; 10:e019667. [PMID: 33787284 PMCID: PMC8174362 DOI: 10.1161/jaha.120.019667] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Background Transcriptomic studies have contributed to fundamental knowledge of myocardial remodeling in human heart failure (HF). However, the key HF genes reported are often inconsistent between studies, and systematic efforts to integrate evidence from multiple patient cohorts are lacking. Here, we aimed to provide a framework for comprehensive comparison and analysis of publicly available data sets resulting in an unbiased consensus transcriptional signature of human end-stage HF. Methods and Results We curated and uniformly processed 16 public transcriptomic studies of left ventricular samples from 263 healthy and 653 failing human hearts. First, we evaluated the degree of consistency between studies by using linear classifiers and overrepresentation analysis. Then, we meta-analyzed the deregulation of 14 041 genes to extract a consensus signature of HF. Finally, to functionally characterize this signature, we estimated the activities of 343 transcription factors, 14 signaling pathways, and 182 micro RNAs, as well as the enrichment of 5998 biological processes. Machine learning approaches revealed conserved disease patterns across all studies independent of technical differences. These consistent molecular changes were prioritized with a meta-analysis, functionally characterized and validated on external data. We provide all results in a free public resource (https://saezlab.shinyapps.io/reheat/) and exemplified usage by deciphering fetal gene reprogramming and tracing the potential myocardial origin of the plasma proteome markers in patients with HF. Conclusions Even though technical and sampling variability confound the identification of differentially expressed genes in individual studies, we demonstrated that coordinated molecular responses during end-stage HF are conserved. The presented resource is crucial to complement findings in independent studies and decipher fundamental changes in failing myocardium.
Collapse
Affiliation(s)
- Ricardo O. Ramirez Flores
- Faculty of Medicine, and Heidelberg University HospitalInstitute for Computational BiomedicineBioquantHeidelberg UniversityHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- Informatics for LifeHeidelbergGermany
| | - Jan D. Lanzer
- Faculty of Medicine, and Heidelberg University HospitalInstitute for Computational BiomedicineBioquantHeidelberg UniversityHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- Informatics for LifeHeidelbergGermany
- Department of General Internal Medicine and PsychosomaticsHeidelberg University HospitalHeidelbergGermany
| | - Christian H. Holland
- Faculty of Medicine, and Heidelberg University HospitalInstitute for Computational BiomedicineBioquantHeidelberg UniversityHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Florian Leuschner
- Department of CardiologyMedical University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/MannheimHeidelbergGermany
| | - Patrick Most
- Department of CardiologyMedical University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/MannheimHeidelbergGermany
- Center for Translational MedicineJefferson UniversityPhiladelphiaPA
| | - Jobst‐Hendrik Schultz
- Department of General Internal Medicine and PsychosomaticsHeidelberg University HospitalHeidelbergGermany
| | - Rebecca T. Levinson
- Informatics for LifeHeidelbergGermany
- Department of General Internal Medicine and PsychosomaticsHeidelberg University HospitalHeidelbergGermany
| | - Julio Saez‐Rodriguez
- Faculty of Medicine, and Heidelberg University HospitalInstitute for Computational BiomedicineBioquantHeidelberg UniversityHeidelbergGermany
- Informatics for LifeHeidelbergGermany
- Faculty of MedicineJoint Research Centre for Computational Biomedicine (JRC‐COMBINE)RWTH Aachen UniversityAachenGermany
| |
Collapse
|
22
|
Tannous C, Deloux R, Karoui A, Mougenot N, Burkin D, Blanc J, Coletti D, Lavery G, Li Z, Mericskay M. NMRK2 Gene Is Upregulated in Dilated Cardiomyopathy and Required for Cardiac Function and NAD Levels during Aging. Int J Mol Sci 2021; 22:3534. [PMID: 33805532 PMCID: PMC8036583 DOI: 10.3390/ijms22073534] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/09/2021] [Accepted: 03/25/2021] [Indexed: 01/16/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a disease of multifactorial etiologies, the risk of which is increased by male sex and age. There are few therapeutic options for patients with DCM who would benefit from identification of common targetable pathways. We used bioinformatics to identify the Nmrk2 gene involved in nicotinamide adenine dinucleotde (NAD) coenzyme biosynthesis as activated in different mouse models and in hearts of human patients with DCM while the Nampt gene controlling a parallel pathway is repressed. A short NMRK2 protein isoform is also known as muscle integrin binding protein (MIBP) binding the α7β1 integrin complex. We investigated the cardiac phenotype of Nmrk2-KO mice to establish its role in cardiac remodeling and function. Young Nmrk2-KO mice developed an eccentric type of cardiac hypertrophy in response to pressure overload rather than the concentric hypertrophy observed in controls. Nmrk2-KO mice developed a progressive DCM-like phenotype with aging, associating eccentric remodeling of the left ventricle and a decline in ejection fraction and showed a reduction in myocardial NAD levels at 24 months. In agreement with involvement of NMRK2 in integrin signaling, we observed a defect in laminin deposition in the basal lamina of cardiomyocytes leading to increased fibrosis at middle age. The α7 integrin was repressed at both transcript and protein level at 24 months. Nmrk2 gene is required to preserve cardiac structure and function, and becomes an important component of the NAD biosynthetic pathways during aging. Molecular characterization of compounds modulating this pathway may have therapeutic potential.
Collapse
Affiliation(s)
- Cynthia Tannous
- Inserm Unit UMR-S 1180 CARPAT, Faculty of Pharmacy, Université Paris-Saclay, 92296 Châtenay-Malabry, France; (C.T.); (R.D.); (A.K.)
- INSERM Unit U1164 / CNRS UMR 8256, Biologie de l’Adaptation et du Vieillissement, Institut de Biologie Paris-Seine, Sorbonne Université, 75006 Paris, France; (J.B.); (D.C.); (Z.L.)
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Robin Deloux
- Inserm Unit UMR-S 1180 CARPAT, Faculty of Pharmacy, Université Paris-Saclay, 92296 Châtenay-Malabry, France; (C.T.); (R.D.); (A.K.)
| | - Ahmed Karoui
- Inserm Unit UMR-S 1180 CARPAT, Faculty of Pharmacy, Université Paris-Saclay, 92296 Châtenay-Malabry, France; (C.T.); (R.D.); (A.K.)
| | - Nathalie Mougenot
- Plateau d’Expérimentation Cœur, Muscle, Vaisseaux PECMV, UMS28, Sorbonne Université, 75013 Paris, France;
| | - Dean Burkin
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV 89102, USA;
| | - Jocelyne Blanc
- INSERM Unit U1164 / CNRS UMR 8256, Biologie de l’Adaptation et du Vieillissement, Institut de Biologie Paris-Seine, Sorbonne Université, 75006 Paris, France; (J.B.); (D.C.); (Z.L.)
| | - Dario Coletti
- INSERM Unit U1164 / CNRS UMR 8256, Biologie de l’Adaptation et du Vieillissement, Institut de Biologie Paris-Seine, Sorbonne Université, 75006 Paris, France; (J.B.); (D.C.); (Z.L.)
| | - Gareth Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, 2nd Floor IBR Tower, Edgbaston, Birmingham B15 2TT, UK;
| | - Zhenlin Li
- INSERM Unit U1164 / CNRS UMR 8256, Biologie de l’Adaptation et du Vieillissement, Institut de Biologie Paris-Seine, Sorbonne Université, 75006 Paris, France; (J.B.); (D.C.); (Z.L.)
| | - Mathias Mericskay
- Inserm Unit UMR-S 1180 CARPAT, Faculty of Pharmacy, Université Paris-Saclay, 92296 Châtenay-Malabry, France; (C.T.); (R.D.); (A.K.)
| |
Collapse
|
23
|
Fernandes Vileigas D, Cicogna AC. Effects of obesity on the cardiac proteome. ENDOCRINE AND METABOLIC SCIENCE 2021. [DOI: 10.1016/j.endmts.2020.100076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
24
|
Baulina NM, Kiselev IS, Chumakova OS, Favorova OO. Hypertrophic Cardiomyopathy as an Oligogenic Disease: Transcriptomic Arguments. Mol Biol 2021. [DOI: 10.1134/s0026893320060023] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
Pappone C, Micaglio E, Locati ET, Monasky MM. The omics of channelopathies and cardiomyopathies: what we know and how they are useful. Eur Heart J Suppl 2020; 22:L105-L109. [PMID: 33654474 PMCID: PMC7904073 DOI: 10.1093/eurheartj/suaa146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sudden cardiac death results from arrhythmias commonly caused by channelopathies and cardiomyopathies, often due to several genetic factors. An emerging concept is that these disease states may in fact overlap, with variants in traditionally classified ‘cardiomyopathy genes’ resulting in ‘channelopathies phenotypes’. Another important concept is the influence of both genetic and non-genetic factors in disease expression, leading to the utilization of systems biology approaches, such as genomics/epigenomics, transcriptomics, proteomics, metabolomics, lipidomics, and glycomics, to understand the disease severity and progression and to determine the prognosis and the best course of treatment. In fact, our group has discovered significant differences in metabolites, proteins, and lipids between controls and Brugada syndrome patients. Omics approaches are useful in overcoming the dogma that both channelopathies and cardiomyopathies exist as Mendelian disorders (caused by a mutation in a single gene). This shift in understanding could lead to new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Carlo Pappone
- Arrhythmology and Electrophysiology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Emanuele Micaglio
- Arrhythmology and Electrophysiology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Emanuela T Locati
- Arrhythmology and Electrophysiology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Michelle M Monasky
- Arrhythmology and Electrophysiology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW The goal of this review is to summarize the state of big data analyses in the study of heart failure (HF). We discuss the use of big data in the HF space, focusing on "omics" and clinical data. We address some limitations of this data, as well as their future potential. RECENT FINDINGS Omics are providing insight into plasmal and myocardial molecular profiles in HF patients. The introduction of single cell and spatial technologies is a major advance that will reshape our understanding of cell heterogeneity and function as well as tissue architecture. Clinical data analysis focuses on HF phenotyping and prognostic modeling. Big data approaches are increasingly common in HF research. The use of methods designed for big data, such as machine learning, may help elucidate the biology underlying HF. However, important challenges remain in the translation of this knowledge into improvements in clinical care.
Collapse
Affiliation(s)
- Jan D Lanzer
- Institute for Computational Biomedicine, Bioquant, Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Internal Medicine II, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Leuschner
- Department of Cardiology, Medical University Hospital, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Rebecca T Levinson
- Institute for Computational Biomedicine, Bioquant, Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Heidelberg, Germany
- Internal Medicine II, Heidelberg University Hospital, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Bioquant, Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Heidelberg, Germany.
- Joint Research Centre for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
27
|
Mollanoori H, Rahmati Y, Hassani B, Esmaeili S, Amini K, Teimourian S. Screening the underlying molecular mechanisms involved in the development of heart failure. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
28
|
Guan F, Yang X, Li J, Dong W, Zhang X, Liu N, Gao S, Wang J, Zhang L, Lu D. New Molecular Mechanism Underlying Myc-Mediated Cytochrome P450 2E1 Upregulation in Apoptosis and Energy Metabolism in the Myocardium. J Am Heart Assoc 2020; 8:e009871. [PMID: 30563421 PMCID: PMC6405704 DOI: 10.1161/jaha.118.009871] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Canonical studies indicate that cytochrome P450 2E1 (CYP2E1) plays a critical role in the metabolism of xenobiotics and ultimately participates in tissue damage. CYP2E1 upregulates in the pathophysiological development of multiple diseases; however, the mechanism of CYP2E1 upregulation, particularly in heart disease, remains elusive. Methods and Results We found that the level of CYP2E1 increased in heart tissues from patients with hypertrophic cardiomyopathy; multiple mouse models of heart diseases, including dilated cardiomyopathy, hypertrophic cardiomyopathy, and myocardial ischemia; and HL‐1 myocytes under stress. We determined that Myc bound to the CYP2E1 promoter and activated its transcription by bioinformatics analysis, luciferase activity, and chromatin immunoprecipitation, and Myc expression was modulated by extracellular signal–regulated kinases 1/2 and phosphatidylinositol 3 kinase/protein kinase B pathways under stress or injury in myocardium by signal transduction analysis. In addition, the level of oxidative stress and apoptosis gradually worsened with age in transgenic mice overexpressing CYP2E1, which was significantly inhibited with CYP2E1 knockdown. Conclusions Our results demonstrated that CYP2E1 is likely a sensor of diverse pathophysiological factors and states in the myocardium. Upregulated CYP2E1 has multiple pathophysiological roles in the heart, including increased oxidative stress and apoptosis as well as energy supply to meet the energy demand of the heart in certain disease states. Our discovery thus provides a basis for a therapeutic strategy for heart diseases targeting Myc and CYP2E1.
Collapse
Affiliation(s)
- Feifei Guan
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| | - Xinlan Yang
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| | - Jing Li
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| | - Wei Dong
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| | - Xu Zhang
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| | - Ning Liu
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| | - Shan Gao
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| | - Jizheng Wang
- 2 State Key Laboratory of Cardiovascular Disease Fuwai Hospital National Center for Cardiovascular Disease Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Lianfeng Zhang
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| | - Dan Lu
- 1 Key Laboratory of Human Disease Comparative Medicine NHFPC Institute of Laboratory Animal Science Chinese Academy of Medical Sciences & Comparative Medical Center Peking Union Medical College Beijing China
| |
Collapse
|
29
|
Kumar G, Saleem N, Kumar S, Maulik SK, Ahmad S, Sharma M, Goswami SK. Transcriptomic Validation of the Protective Effects of Aqueous Bark Extract of Terminalia arjuna (Roxb.) on Isoproterenol-Induced Cardiac Hypertrophy in Rats. Front Pharmacol 2019; 10:1443. [PMID: 31920643 PMCID: PMC6916006 DOI: 10.3389/fphar.2019.01443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022] Open
Abstract
Aqueous extract of the bark of Terminalia arjuna (TA) is used by a large population in the Indian subcontinent for treating various cardiovascular conditions. Animal experiments have shown its anti-atherogenic, anti-hypertensive, and anti-inflammatory effects. It has several bioactive ingredients with hemodynamic, ROS scavenging, and anti-inflammatory properties. Earlier we have done limited proteomic and transcriptomic analysis to show its efficacy in ameliorating cardiac hypertrophy induced by isoproterenol (ISO) in rats. In the present study we have used high-throughput sequencing of the mRNA from control and treated rat heart to further establish its efficacy. ISO (5 mg/kg/day s.c.) was administered in male adult rats for 14 days to induce cardiac hypertrophy. Standardized aqueous extract TA bark extract was administered orally. Total RNA were isolated from control, ISO, ISO + TA, and TA treated rat hearts and subjected to high throughput sequence analysis. The modulations of the transcript levels were then subjected to bio-informatics analyses using established software. Treatment with ISO downregulated 1,129 genes and upregulated 204 others. Pre-treatment with the TA bark extracts markedly restored that expression pattern with only 97 genes upregulated and 85 genes downregulated. The TA alone group had only 88 upregulated and 26 downregulated genes. The overall profile of expression in ISO + TA and TA alone groups closely matched with the control group. The genes that were modulated included those involved in metabolism, activation of receptors and cell signaling, and cardiovascular and other diseases. Networks associated with those genes included those involved in angiogenesis, extracellular matrix organization, integrin binding, inflammation, drug metabolism, redox metabolism, oxidative phosphorylation, and organization of myofibril. Overlaying of the networks in ISO and ISO_TA group showed that those activated in ISO group were mostly absent in ISO_TA and TA group, suggesting a global effect of the TA extracts. This study for the first time reveals that TA partially or completely restores the gene regulatory network perturbed by ISO treatment in rat heart; signifying its efficacy in checking ISO-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Gaurav Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Nikhat Saleem
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Santosh Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Subir K Maulik
- Department of Pharmacology, All India Institute of Medical Sciences (A.I.I.M.S.), New Delhi, India
| | - Sayeed Ahmad
- Bioactive Natural Product Laboratory, Department of Pharmacognosy & Phytochemistry, School of Pharmaceutical Education & Research, New Delhi, India
| | - Manish Sharma
- Peptide and Proteomics Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organisation, New Delhi, India
| | - Shyamal K Goswami
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
30
|
Feig MA, Pop C, Bhardwaj G, Sappa PK, Dörr M, Ameling S, Weitmann K, Nauck M, Lehnert K, Beug D, Kühl U, Schultheiss HP, Völker U, Felix SB, Hammer E. Global plasma protein profiling reveals DCM characteristic protein signatures. J Proteomics 2019; 209:103508. [DOI: 10.1016/j.jprot.2019.103508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 12/28/2022]
|
31
|
Abstract
Aging is associated with a progressive decline in cardiovascular structure and function. Accumulating evidence links cardiovascular aging to epigenetic alterations encompassing a complex interplay of DNA methylation, histone posttranslational modifications, and dynamic nucleosome occupancy governed by numerous epigenetic factors. Advances in genomics technology have led to a profound understanding of chromatin reorganization in both cardiovascular aging and diseases. This review summarizes recent discoveries in epigenetic mechanisms involved in cardiovascular aging and diseases and discusses potential therapeutic strategies to retard cardiovascular aging and conquer related diseases through the rejuvenation of epigenetic signatures to a young state.
Collapse
Affiliation(s)
- Weiqi Zhang
- From the Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China (W.Z., G.-H.L.).,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics (W.Z., G.-H.L.), Chinese Academy of Sciences, Beijing.,Institute of Stem Cell and Regeneration (W.Z., M.S., J.Q., G.-H.L.), Chinese Academy of Sciences, Beijing.,University of Chinese Academy of Sciences, Beijing (W.Z., M.S., J.Q., G.-H.L.)
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology (M.S.), Chinese Academy of Sciences, Beijing.,Institute of Stem Cell and Regeneration (W.Z., M.S., J.Q., G.-H.L.), Chinese Academy of Sciences, Beijing.,University of Chinese Academy of Sciences, Beijing (W.Z., M.S., J.Q., G.-H.L.)
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology (J.Q.), Chinese Academy of Sciences, Beijing.,Institute of Stem Cell and Regeneration (W.Z., M.S., J.Q., G.-H.L.), Chinese Academy of Sciences, Beijing.,University of Chinese Academy of Sciences, Beijing (W.Z., M.S., J.Q., G.-H.L.)
| | - Guang-Hui Liu
- From the Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China (W.Z., G.-H.L.).,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics (W.Z., G.-H.L.), Chinese Academy of Sciences, Beijing.,Institute of Stem Cell and Regeneration (W.Z., M.S., J.Q., G.-H.L.), Chinese Academy of Sciences, Beijing.,University of Chinese Academy of Sciences, Beijing (W.Z., M.S., J.Q., G.-H.L.)
| |
Collapse
|
32
|
Leon-Mimila P, Wang J, Huertas-Vazquez A. Relevance of Multi-Omics Studies in Cardiovascular Diseases. Front Cardiovasc Med 2019; 6:91. [PMID: 31380393 PMCID: PMC6656333 DOI: 10.3389/fcvm.2019.00091] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death around the world. Despite the larger number of genes and loci identified, the precise mechanisms by which these genes influence risk of cardiovascular disease is not well understood. Recent advances in the development and optimization of high-throughput technologies for the generation of “omics data” have provided a deeper understanding of the processes and dynamic interactions involved in human diseases. However, the integrative analysis of “omics” data is not straightforward and represents several logistic and computational challenges. In spite of these difficulties, several studies have successfully applied integrative genomics approaches for the investigation of novel mechanisms and plasma biomarkers involved in cardiovascular diseases. In this review, we summarized recent studies aimed to understand the molecular framework of these diseases using multi-omics data from mice and humans. We discuss examples of omics studies for cardiovascular diseases focused on the integration of genomics, epigenomics, transcriptomics, and proteomics. This review also describes current gaps in the study of complex diseases using systems genetics approaches as well as potential limitations and future directions of this emerging field.
Collapse
Affiliation(s)
- Paola Leon-Mimila
- Division of Cardiology, David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jessica Wang
- Division of Cardiology, David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Adriana Huertas-Vazquez
- Division of Cardiology, David Geffen School of Medicine, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
33
|
Jie QQ, Li G, Duan JB, Li XB, Yang W, Chu YP, Yu SD, Liu XY, Wang CY, Liu FF, Ze F, Huang YW, Chen Y, Ding YS, Guo JH, Wu L. Remodeling of myocardial energy and metabolic homeostasis in a sheep model of persistent atrial fibrillation. Biochem Biophys Res Commun 2019; 517:8-14. [PMID: 31307785 DOI: 10.1016/j.bbrc.2019.05.112] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 05/16/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the most common progressive cardiac arrhythmia and is often associated with rapid contraction in both atria and ventricles. The role of atrial energy and metabolic homeostasis in AF progression is under-investigated. OBJECTIVES To determine the remodeling of energy metabolism during persistent AF and the effect of eplerenone (EPL), an aldosterone inhibitor, on metabolic homeostasis. METHODS A nonsustained atrial pacing sheep model was developed to simulate the progression of AF from paroxysmal to persistent. Metabolomic and proteomic analyses at termination of the experiment were used to analyze atrial tissues obtained from sheep in sham, sugar pill (SP) and EPL-treated groups. RESULTS Proteomic analysis indicated that compared to the sham group, in SP group, fatty acid (FA) synthesis, FA oxidation, tricarboxylic acid (TCA) cycle processes and amino acids (AAs) transport and metabolism were reduced, while glycolytic processes were increased. In metabolomic analysis, the levels of intermediate metabolites of the glycolytic pathways, including 2-phosphoglyceric acid (2 PG), 1,3-bisphosphoglyceric acid (1,3 PG), and pyruvate, HBP (uridine diphosphate-N-acetylglucosamine, UDP-GlcNAc), TCA (citrate) and AAs were greater while the levels of the majority of lipid classes, including phosphatidic acid (PA), phosphatidylcholine (PC), phosphatidylglycerol (PG), glycerophosphoglycerophosphates (PGP), glycerophosphoinositols (PI) and glycerophosphoserines (PS), were decreased in the atria of SP group than in those of sham group. EPL-pretreatment decreased the expression of glut4 and increased the content of acylcarnitines and lipids, such as lyso phospholipids, phospholipids and neutral lipids. CONCLUSION In the metabolic remodeling during AF, glucose and lipid metabolism were up- and down-regulated, respectively, to sustain TCA cycle anaplerosis. EPL partialy reversed the metabolic shifting.
Collapse
Affiliation(s)
- Qi-Qiang Jie
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Gang Li
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Jiang-Bo Duan
- Department of Cardiac Electrophysiology, Peking University People's Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Xue-Bin Li
- Department of Cardiac Electrophysiology, Peking University People's Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Yang
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yan-Peng Chu
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Shan-Dong Yu
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Xiao-Yan Liu
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Cheng-Yu Wang
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Fei-Fei Liu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Feng Ze
- Department of Cardiac Electrophysiology, Peking University People's Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yu-Wen Huang
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ying Chen
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yan-Sheng Ding
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ji-Hong Guo
- Department of Cardiac Electrophysiology, Peking University People's Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| | - Lin Wu
- Department of Cardiology, Peking University First Hospital, Beijing, China; Department of Cardiology, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China; Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| |
Collapse
|
34
|
Sex-specific differences in hepatic steatosis in obese spontaneously hypertensive (SHROB) rats. Biol Sex Differ 2018; 9:40. [PMID: 30201044 PMCID: PMC6131947 DOI: 10.1186/s13293-018-0202-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022] Open
Abstract
Background Patients with metabolic syndrome, who are characterized by co-existence of insulin resistance, hypertension, hyperlipidemia, and obesity, are also prone to develop non-alcoholic fatty liver disease (NAFLD). Although the prevalence and severity of NAFLD is significantly greater in men than women, the mechanisms by which gender modulates the pathogenesis of hepatic steatosis are poorly defined. The obese spontaneously hypertensive (SHROB) rats represent an attractive model of metabolic syndrome without overt type 2 diabetes. Although pathological manifestation caused by the absence of a functional leptin receptor has been extensively studied in SHROB rats, it is unknown whether these animals elicited sex-specific differences in the development of hepatic steatosis. Methods We compared hepatic pathology in male and female SHROB rats. Additionally, we examined key biochemical and molecular parameters of signaling pathways linked with hyperinsulinemia and hyperlipidemia. Finally, using methods of quantitative polymerase chain reaction (qPCR) and western blot analysis, we quantified expression of 45 genes related to lipid biosynthesis and metabolism in the livers of male and female SHROB rats. Results We show that all SHROB rats developed hepatic steatosis that was accompanied by enhanced expression of SREBP1, SREBP2, ACC1, and FASN proteins. The livers of male rats also elicited higher induction of Pparg, Ppara, Slc2a4, Atox1, Skp1, Angptl3, and Pnpla3 mRNAs. In contrast, the livers of female SHROB rats elicited constitutively higher levels of phosphorylated JNK and AMPK and enhanced expression of Cd36. Conclusion Based on these data, we conclude that the severity of hepatic steatosis in male and female SHROB rats was mainly driven by increased de novo lipogenesis. Moreover, male and female SHROB rats also elicited differential severity of hepatic steatosis that was coupled with sex-specific differences in fatty acid transport and esterification. Electronic supplementary material The online version of this article (10.1186/s13293-018-0202-x) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Perrino C, Barabási AL, Condorelli G, Davidson SM, De Windt L, Dimmeler S, Engel FB, Hausenloy DJ, Hill JA, Van Laake LW, Lecour S, Leor J, Madonna R, Mayr M, Prunier F, Sluijter JPG, Schulz R, Thum T, Ytrehus K, Ferdinandy P. Epigenomic and transcriptomic approaches in the post-genomic era: path to novel targets for diagnosis and therapy of the ischaemic heart? Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res 2018; 113:725-736. [PMID: 28460026 PMCID: PMC5437366 DOI: 10.1093/cvr/cvx070] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 04/27/2017] [Indexed: 01/19/2023] Open
Abstract
Despite advances in myocardial reperfusion therapies, acute myocardial ischaemia/reperfusion injury and consequent ischaemic heart failure represent the number one cause of morbidity and mortality in industrialized societies. Although different therapeutic interventions have been shown beneficial in preclinical settings, an effective cardioprotective or regenerative therapy has yet to be successfully introduced in the clinical arena. Given the complex pathophysiology of the ischaemic heart, large scale, unbiased, global approaches capable of identifying multiple branches of the signalling networks activated in the ischaemic/reperfused heart might be more successful in the search for novel diagnostic or therapeutic targets. High-throughput techniques allow high-resolution, genome-wide investigation of genetic variants, epigenetic modifications, and associated gene expression profiles. Platforms such as proteomics and metabolomics (not described here in detail) also offer simultaneous readouts of hundreds of proteins and metabolites. Isolated omics analyses usually provide Big Data requiring large data storage, advanced computational resources and complex bioinformatics tools. The possibility of integrating different omics approaches gives new hope to better understand the molecular circuitry activated by myocardial ischaemia, putting it in the context of the human ‘diseasome’. Since modifications of cardiac gene expression have been consistently linked to pathophysiology of the ischaemic heart, the integration of epigenomic and transcriptomic data seems a promising approach to identify crucial disease networks. Thus, the scope of this Position Paper will be to highlight potentials and limitations of these approaches, and to provide recommendations to optimize the search for novel diagnostic or therapeutic targets for acute ischaemia/reperfusion injury and ischaemic heart failure in the post-genomic era.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Albert-Laszló Barabási
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, USA.,Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Center for Network Science, Central European University, Budapest, Hungary.,Department of Medicine, and Division of Network Medicine, Brigham and Womens Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Gianluigi Condorelli
- Department of Cardiovascular Medicine, Humanitas Research Hospital and Humanitas University, Rozzano, Italy.,Institute of Genetic and Biomedical Research, National Research Council of Italy, Rozzano, Milan, Italy
| | - Sean Michael Davidson
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Leon De Windt
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, University Frankfurt, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), RheinMain, Germany
| | - Felix Benedikt Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Derek John Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, UK.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore.,Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Joseph Addison Hill
- Departments of Medicine (Cardiology) and Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Linda Wilhelmina Van Laake
- Division of Heart and Lungs, Hubrecht Institute, University Medical Center Utrecht, Utrecht, The Netherlands.,UMC Utrecht Regenerative Medicine Center and Hubrecht Institute, Utrecht, The Netherlands
| | - Sandrine Lecour
- Hatter Cardiovascular Research Institute, University of Cape Town, Cape Town, South Africa
| | - Jonathan Leor
- Neufeld Cardiac Research Institute, Tel-Aviv University, Tel-Aviv, Israel.,Tamman Cardiovascular Research Institute, Sheba Medical Center; Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel Hashomer, Israel
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy; Institute of Cardiology, Department of Neurosciences, Imaging, and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy.,The Texas Heart Institute and Center for Cardiovascular Biology and Atherosclerosis Research, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Fabrice Prunier
- Department of Cardiology, Institut MITOVASC, University of Angers, University Hospital of Angers, Angers, France
| | - Joost Petrus Geradus Sluijter
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
36
|
|
37
|
Liang Y, Xie SB, Wu CH, Hu Y, Zhang Q, Li S, Fan YG, Leng RX, Pan HF, Xiong HB, Ye DQ. Coagulation cascade and complement system in systemic lupus erythematosus. Oncotarget 2017; 9:14862-14881. [PMID: 29599912 PMCID: PMC5871083 DOI: 10.18632/oncotarget.23206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022] Open
Abstract
This study was conducted to (1) characterize coagulation cascade and complement system in systemic lupus erythematosus (SLE); (2) evaluate the associations between coagulation cascade, complement system, inflammatory response and SLE disease severity; (3) test the diagnostic value of a combination of D-dimer and C4 for lupus activity. Transcriptomics, proteomics and metabolomics were performed in 24 SLE patients and 24 healthy controls. The levels of ten coagulations, seven complements and three cytokines were measured in 112 SLE patients. Clinical data were collected from 2025 SLE patients. The analysis of multi-omics data revealed the common links for the components of coagulation cascade and complement system. The results of ELISA showed coagulation cascade and complement system had an interaction effect on SLE disease severity, this effect was pronounced among patients with excess inflammation. The analysis of clinical data revealed a combination of D-dimer and C4 provided good diagnostic performance for lupus activity. This study suggested that coagulation cascade and complement system become 'partners in crime', contributing to SLE disease severity and identified the diagnostic value of D-dimer combined with C4for lupus activity.
Collapse
Affiliation(s)
- Yan Liang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | | | - Chang-Hao Wu
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Yuan Hu
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qin Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Yin-Guang Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Rui-Xue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Hua-Bao Xiong
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| |
Collapse
|
38
|
Kumar S, Jahangir Alam M, Prabhakar P, Ahmad S, Maulik SK, Sharma M, Goswami SK. Proteomic analysis of the protective effects of aqueous bark extract of Terminalia arjuna (Roxb.) on isoproterenol-induced cardiac hypertrophy in rats. JOURNAL OF ETHNOPHARMACOLOGY 2017; 198:98-108. [PMID: 28063919 DOI: 10.1016/j.jep.2016.12.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 12/12/2016] [Accepted: 12/31/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aqueous bark extract of Terminalia arjuna (TA) has been in use as an ethnomedicine for cardiovascular ailments in the Indian subcontinent for centuries. Studies using hemodynamic, ROS scavenging and anti-inflammatory parameters in animal models have shown its anti-atherogenic, hypotensive, inotropic, anti-inflammatory effects. However, details analysis on its effects on established molecular and cell biological markers are a prerequisite for its wider acceptance to the medical community. AIMS OF THE STUDY To test the efficacy of TA extract in ameliorating cardiac hypertrophy induced by ISO in rats. METHODS Cardiac hypertrophy was induced by ISO (5mg/kg/day s.c. for 14 days) in rats and a standardized aqueous extract of TA stem bark was orally administered by gavage. Total RNA and protein were isolated from control, ISO, ISO plus TA and TA treated rat hearts and analyzed for the transcripts for the markers of hypertrophy, signaling kinases, transcription factors and total protein profile. RESULTS TA extract reversed the induction of fetal genes like β-myosin heavy chain, skeletal α-actin and brain natriuretic peptide in hypertrophic rat hearts. While ISO slightly increased the level of phospho-ERK, TA repressed it to about one third of the base line level. Survival kinase Akt, ER stress marker Grp78 and epigenetic regulator HDAC5 were augmented by ISO and TA restored them by various extents. ISO administration moderately increased the transcription factor NFκB binding activity, while coadministration of TA further increased it. AP-1 binding activity was largely unchanged by ISO treatment but it was upregulated when administered along with TA. MEF2D binding activity was increased by ISO and TA restored it to the baseline level. Global proteomic analysis revealed that TA treatment restored a subset of proteins up- and down-regulated in the hypertrophied hearts. Amongst those restored by TA were purinergic receptor X, myosin light chain 3, tropomyosin, and kininogen; suggesting a nodal role of TA in modulating cardiac function. CONCLUSIONS This study for the first time reveals that TA partially or completely restores the marker mRNAs, signaling kinases, transcription factors and total protein profile in rat heart, thereby demonstrating its efficacy in preventing ISO-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Santosh Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India.
| | - Md Jahangir Alam
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India.
| | - Pankaj Prabhakar
- Department of Pharmacology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029, New Delhi, India.
| | - Sayeed Ahmad
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmacy, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India.
| | - Subir K Maulik
- Department of Pharmacology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029, New Delhi, India.
| | - Manish Sharma
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi 110054, India.
| | - Shyamal K Goswami
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India.
| |
Collapse
|
39
|
DeAguero JL, McKown EN, Zhang L, Keirsey J, Fischer EG, Samedi VG, Canan BD, Kilic A, Janssen PML, Delfín DA. Altered protein levels in the isolated extracellular matrix of failing human hearts with dilated cardiomyopathy. Cardiovasc Pathol 2016; 26:12-20. [PMID: 27837684 DOI: 10.1016/j.carpath.2016.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/23/2016] [Accepted: 10/10/2016] [Indexed: 01/09/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is associated with extensive pathological cardiac remodeling and involves numerous changes in the protein expression profile of the extracellular matrix of the heart. We obtained seven human, end-stage, failing hearts with DCM (DCM-failing) and nine human, nonfailing donor hearts and compared their extracellular matrix protein profiles. We first showed that the DCM-failing hearts had indeed undergone extensive remodeling of the left ventricle myocardium relative to nonfailing hearts. We then isolated the extracellular matrix from a subset of these hearts and performed a proteomic analysis on the isolated matrices. We found that the levels of 26 structural proteins were altered in the DCM-failing isolated cardiac extracellular matrix compared to nonfailing isolated cardiac extracellular matrix. Overall, most of the extracellular matrix proteins showed reduced levels in the DCM-failing hearts, while all of the contractile proteins showed increased levels. There was a mixture of increased and decreased levels of cytoskeletal and nuclear transport proteins. Using immunoprobing, we verified that collagen IV (α2 and α6 isoforms), zyxin, and myomesin protein levels were reduced in the DCM-failing hearts. We expect that these data will add to the understanding of the pathology associated with heart failure with DCM.
Collapse
Affiliation(s)
- Joshua L DeAguero
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | - Elizabeth N McKown
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | - Liwen Zhang
- The Ohio State University Mass Spectrometry and Proteomics Facility, Campus Chemical Instrument Center, 460 W. 12th Ave., Room 250 Biomedical Research Tower, Columbus, OH 43210, USA.
| | - Jeremy Keirsey
- The Ohio State University Mass Spectrometry and Proteomics Facility, Campus Chemical Instrument Center, 460 W. 12th Ave., Room 250 Biomedical Research Tower, Columbus, OH 43210, USA.
| | - Edgar G Fischer
- The University of New Mexico School of Medicine, Department of Pathology, MSC08 4640, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | - Von G Samedi
- The University of New Mexico School of Medicine, Department of Pathology, MSC08 4640, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| | - Benjamin D Canan
- The Ohio State University College of Medicine, Department of Physiology and Cell Biology and the Davis Heart Lung Research Institute, 200 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210.
| | - Ahmet Kilic
- The Ohio State University College of Medicine, Department of Surgery and the Davis Heart Lung Research Institute, Richard M. Ross Heart Hospital, 452 West 10th Ave., Columbus, OH 43210.
| | - Paul M L Janssen
- The Ohio State University College of Medicine, Department of Physiology and Cell Biology and the Davis Heart Lung Research Institute, 200 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210.
| | - Dawn A Delfín
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|