1
|
Amanollahi R, Holman SL, Meakin AS, Padhee M, Botting-Lawford KJ, Zhang S, MacLaughlin SM, Kleemann DO, Walker SK, Kelly JM, Rudiger SR, McMillen IC, Wiese MD, Lock MC, Morrison JL. In Vitro Embryo Culture Impacts Heart Mitochondria in Male Adolescent Sheep. J Dev Biol 2025; 13:17. [PMID: 40407686 PMCID: PMC12101424 DOI: 10.3390/jdb13020017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/17/2025] [Accepted: 05/09/2025] [Indexed: 05/26/2025] Open
Abstract
Assisted reproductive technology (ART)such as in vitro embryo culture (IVC), is widely used in human infertility treatments; however, its long-term effects on the cardiac health of offspring remain unclear. This study aimed to determine whether the effects of IVC on cardiac metabolism and associated signaling pathways persist after birth into adolescence. Embryos were either transferred to an intermediate ewe (ET) or cultured in vitro in the absence (IVC) or presence of human serum (IVCHS) with methionine supplementation (IVCHS+M) for 6 days after mating. Naturally mated (NM) ewes were used as controls. Protein expression and hormone concentrations in the left ventricle (LV) were analyzed using Western blot and LC-MS/MS analyses, respectively. IVC was associated with sex-specific alterations in cardiac mitochondria, with males exhibiting reduced mitochondrial abundance. Cardiac protein expression of oxidative phosphorylation (OXPHOS) complexes 1 and 4 was reduced by IVC. Additionally, IVC reduced protein expression of PDK-4 and Mn-SOD in the IVCHS+M group, which may impact energy efficiency and defense against oxidative stress. These changes may predispose IVC offspring to cardiac oxidative stress and mitochondrial dysfunction, particularly in males. This study provides insights into the sex-dependent effects of IVC on cardiac health, emphasizing the importance of evaluating long-term cardiovascular risks associated with IVC protocols.
Collapse
Affiliation(s)
- Reza Amanollahi
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - Stacey L. Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - Monalisa Padhee
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - Kimberley J. Botting-Lawford
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - Song Zhang
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - Severence M. MacLaughlin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - David O. Kleemann
- South Australian Research and Development Institute, Turretfield Research Centre, Rosedale, SA 5350, Australia; (D.O.K.); (S.K.W.); (J.M.K.); (S.R.R.)
| | - Simon K. Walker
- South Australian Research and Development Institute, Turretfield Research Centre, Rosedale, SA 5350, Australia; (D.O.K.); (S.K.W.); (J.M.K.); (S.R.R.)
| | - Jennifer M. Kelly
- South Australian Research and Development Institute, Turretfield Research Centre, Rosedale, SA 5350, Australia; (D.O.K.); (S.K.W.); (J.M.K.); (S.R.R.)
| | - Skye R. Rudiger
- South Australian Research and Development Institute, Turretfield Research Centre, Rosedale, SA 5350, Australia; (D.O.K.); (S.K.W.); (J.M.K.); (S.R.R.)
| | - I. Caroline McMillen
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - Michael D. Wiese
- Centre for Pharmaceutical Innovation, Clinical & Health Sciences University of South Australia, Adelaide, SA 5001, Australia;
| | - Mitchell C. Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (R.A.); (S.L.H.); (A.S.M.); (M.P.); (K.J.B.-L.); (S.Z.); (S.M.M.); (I.C.M.)
| |
Collapse
|
2
|
Zhu L, Dong Y, Guo H, Qiu J, Guo J, Hu Y, Pan C. Murine Model Insights: Identifying Dusp15 as a Novel Biomarker for Diabetic Cardiomyopathy Uncovered Through Integrated Omics Analysis and Experimental Validation. Diabetes Metab Syndr Obes 2025; 18:515-527. [PMID: 39990179 PMCID: PMC11847420 DOI: 10.2147/dmso.s501563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
Background Diabetic Cardiomyopathy (DCM) is a heart condition that arises specifically from diabetes mellitus, characterized by cardiac dysfunction in the absence of coronary artery disease or hypertension. The prevalence of DCM is rising in tandem with the global increase in diabetes, necessitating the development of early diagnostic markers and therapeutic targets. This study integrates bioinformatics analysis with experimental validation to identify potential biomarkers for DCM. Methods We performed gene expression data mining from the Gene Expression Omnibus (GEO) database. We employed Weighted Gene Co-expression Network Analysis (WGCNA) coupled with machine learning techniques to sift through hub differentially expressed genes (DEGs). Functional enrichment and protein-protein interaction (PPI) network analysis were also conducted to pinpoint key genes functions. Subsequent in vitro and in vivo experiments were performed to validate the findings. Results Our analysis revealed six core genes significantly associated with DCM. The expression of Dusp15 was notably downregulated and validated in both high-glucose cultured cardiomyocytes and DCM animal models, suggesting its potential role in DCM pathogenesis. Conclusion The integration of bioinformatics with experimental approaches has identified Dusp15 as a promising candidate for a DCM biomarker, offering valuable insights for early diagnosis and potential therapeutic development.
Collapse
Affiliation(s)
- Lingling Zhu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| | - Ya Dong
- Department of Endocrinology, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen Clinical Research Center for Metabolic Diseases, Shenzhen Center for Diabetes Control and Prevention, Shenzhen, Guangdong Province, People’s Republic of China
| | - Hang Guo
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| | - Jie Qiu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| | - Jun Guo
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| | - Yonghui Hu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Congqing Pan
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People’s Republic of China
| |
Collapse
|
3
|
Yang F, He Y, Zhao L, Huang J, Du F, Tian S, Zhang Y, Liu X, Chen B, Ge J, Jiang Z. Leptin drives glucose metabolism to promote cardiac protection via OPA1-mediated HDAC5 translocation and Glut4 transcription. Funct Integr Genomics 2025; 25:28. [PMID: 39875704 PMCID: PMC11774999 DOI: 10.1007/s10142-024-01515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025]
Abstract
Metabolic reprogramming, the shifting from fatty acid oxidation to glucose utilization, improves cardiac function as heart failure (HF) progresses. Leptin plays an essential role in regulating glucose metabolism. However, the crosstalk between leptin and metabolic reprogramming is poorly understood. We tested the hypothesis that leptin improves cardiac function after myocardial infarction via enhancing glucose metabolism. In the isoproterenol (ISO)-induced heart failure model in vitro, H9c2 cell apoptosis was assessed by the TUNEL and Annexin V/PI staining assay. Leptin-mediated mitochondrial fusion was performed via TEM, and glucose oxidation was explored, as well as the ECAR, OCR, and protein expression of the vital metabolic enzymes. By blocking OPA1 expression or HDAC5 inhibition, the mitochondrial dynamic and glucose metabolic were detected to evaluate the role of OPA1 and HDAC5 in leptin-stimulated glucose metabolism. In the mouse model of HF in vivo, intraperitoneal leptin administration appreciably increased glucose oxidation and preserved cardiac function 56 days after coronary artery ligation. In vitro, we identified the OPA1-dependent HDAC5 nucleus export as a crucial process in boosting glucose utilization by activating MEF2 to upregulate Glut4 expression using the RNA interference technique in H9c2 cells. In vivo, leptin promotes glucose utilization and confers heart functional and survival benefits in chronic ischemic HF. The current study provided a novel insight into the role of leptin in metabolic reprogramming and revealed potential therapeutic targets for chronic HF.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Youfu He
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Ling Zhao
- Health Management Center, Guizhou International General Hospital, Guizhou Province, China
| | - Jing Huang
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Fawang Du
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Shui Tian
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Yang Zhang
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Xinghui Liu
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Baolin Chen
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China
- Guizhou University Medical College, Guizhou Province, China
| | - Junhua Ge
- Department of Cardiology, Qingdao Municipal Key Laboratory of Hypertension (Key Laboratory of Cardiovascular Medicine), The Affiliated Hospital of Qingdao University, Shandong Province, China.
| | - Zhi Jiang
- Department of Cardiology, Guizhou Provincial People`s Hospital, 83 Zhongshan East Road, Guiyang City, 550002, Guizhou Province, China.
- Guizhou University Medical College, Guizhou Province, China.
| |
Collapse
|
4
|
Alhasan KA, King MA, Pattar BSB, Lewis IA, Lopaschuk GD, Greenway SC. Anaplerotic filling in heart failure: a review of mechanism and potential therapeutics. Cardiovasc Res 2024; 120:2166-2178. [PMID: 39570879 PMCID: PMC11687400 DOI: 10.1093/cvr/cvae248] [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: 06/18/2024] [Revised: 09/08/2024] [Accepted: 11/12/2024] [Indexed: 01/01/2025] Open
Abstract
Heart failure (HF) is a complex syndrome and a leading cause of mortality worldwide. While current medical treatment is based on known pathophysiology and is effective for many patients, the underlying cellular mechanisms are poorly understood. Energy deficiency is a characteristic of HF, marked by complex alterations in metabolism. Within the tricarboxylic acid cycle, anaplerosis emerges as an essential metabolic process responsible for replenishing lost intermediates, thereby playing a crucial role in sustaining energy metabolism and consequently cardiac function. Alterations in cardiac anaplerosis are commonly observed in HF, demonstrating potential for therapeutic intervention. This review discusses recent advances in understanding the anaplerotic adaptations that occur in HF. We also explore therapeutics that can directly modulate anaplerosis or are likely to confer cardioprotective effects through anaplerosis, which could potentially be implemented to rescue the failing heart.
Collapse
Affiliation(s)
- Karm A Alhasan
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
- Department of Pediatrics and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 1N4
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Melissa A King
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada T2N 1N4
| | - Badal S B Pattar
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Ian A Lewis
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada T2N 1N4
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | - Steven C Greenway
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
- Department of Pediatrics and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 1N4
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| |
Collapse
|
5
|
Hunter B, Li M, Parker BL, Koay YC, Harney DJ, Pearson E, Cao J, Chen GT, Guneratne O, Smyth GK, Larance M, O'Sullivan JF, Lal S. Proteomic and metabolomic analyses of the human adult myocardium reveal ventricle-specific regulation in end-stage cardiomyopathies. Commun Biol 2024; 7:1666. [PMID: 39702518 DOI: 10.1038/s42003-024-07306-y] [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: 06/07/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
The left and right ventricles of the human heart are functionally and developmentally distinct such that genetic or acquired insults can cause dysfunction in one or both ventricles resulting in heart failure. To better understand ventricle-specific molecular changes influencing heart failure development, we first performed unbiased quantitative mass spectrometry on pre-mortem non-diseased human myocardium to compare the metabolome and proteome between the normal left and right ventricles. Constituents of gluconeogenesis, glycolysis, lipogenesis, lipolysis, fatty acid catabolism, the citrate cycle and oxidative phosphorylation were down-regulated in the left ventricle, while glycogenesis, pyruvate and ketone metabolism were up-regulated. Inter-ventricular significance of these metabolic pathways was then found to be diminished within end-stage dilated cardiomyopathy and ischaemic cardiomyopathy, while heart failure-associated pathways were increased in the left ventricle relative to the right within ischaemic cardiomyopathy, such as fluid sheer-stress, increased glutamine-glutamate ratio, and down-regulation of contractile proteins, indicating a left ventricular pathological bias.
Collapse
Affiliation(s)
- Benjamin Hunter
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Mengbo Li
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Yen Chin Koay
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Heart Research Institute, Newtown, NSW, Australia
| | - Dylan J Harney
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Evangeline Pearson
- Paediatric Oncology and Haematology, Oxford Children's Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, England
| | - Jacob Cao
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Gavin T Chen
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Oneka Guneratne
- Kolling Institute, Royal North Shore Hospital, and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, VIC, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, Australia
| | - Mark Larance
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John F O'Sullivan
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Heart Research Institute, Newtown, NSW, Australia.
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
- Faculty of Medicine, TU Dresden, Dresden, Germany.
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, NSW, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
- The Baird Institute for Applied Heart and Lung Surgical Research, Sydney, NSW, Australia.
| |
Collapse
|
6
|
Ge T, Ning B, Wu Y, Chen X, Qi H, Wang H, Zhao M. MicroRNA-specific therapeutic targets and biomarkers of apoptosis following myocardial ischemia-reperfusion injury. Mol Cell Biochem 2024; 479:2499-2521. [PMID: 37878166 DOI: 10.1007/s11010-023-04876-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/05/2023] [Indexed: 10/26/2023]
Abstract
MicroRNAs are single-stranded non-coding RNAs that participate in post-transcriptional regulation of gene expression, it is involved in the regulation of apoptosis after myocardial ischemia-reperfusion injury. For example, the alteration of mitochondrial structure is facilitated by MicroRNA-1 through the regulation of apoptosis-related proteins, such as Bax and Bcl-2, thereby mitigating cardiomyocyte apoptosis. MicroRNA-21 not only modulates the expression of NF-κB to suppress inflammatory signals but also activates the PI3K/AKT pathway to mitigate ischemia-reperfusion injury. Overexpression of MicroRNA-133 attenuates reactive oxygen species (ROS) production and suppressed the oxidative stress response, thereby mitigating cellular apoptosis. MicroRNA-139 modulates the extrinsic death signal of Fas, while MicroRNA-145 regulates endoplasmic reticulum calcium overload, both of which exert regulatory effects on cardiomyocyte apoptosis. Therefore, the article categorizes the molecular mechanisms based on the three classical pathways and multiple signaling pathways of apoptosis. It summarizes the targets and pathways of MicroRNA therapy for ischemia-reperfusion injury and analyzes future research directions.
Collapse
Affiliation(s)
- Teng Ge
- School of Graduate, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Bo Ning
- School of Graduate, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Yongqing Wu
- School of Graduate, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Xiaolin Chen
- School of Pharmacy, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Hongfei Qi
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Institute of Integrative Medicine, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Haifang Wang
- Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Institute of Integrative Medicine, Shaanxi University of Chinese Medicine, No. 1 Middle Section of Shiji Avenue, Xianyang, 712046, China
| | - Mingjun Zhao
- Department of Cardiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Deputy 2, Weiyang West Road, Weicheng District, Xianyang, 712000, China.
| |
Collapse
|
7
|
Guo B, Zhang F, Yin Y, Ning X, Zhang Z, Meng Q, Yang Z, Jiang W, Liu M, Wang Y, Sun L, Yu L, Mu N. Post-translational modifications of pyruvate dehydrogenase complex in cardiovascular disease. iScience 2024; 27:110633. [PMID: 39224515 PMCID: PMC11367490 DOI: 10.1016/j.isci.2024.110633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Pyruvate dehydrogenase complex (PDC) is a crucial enzyme that connects glycolysis and the tricarboxylic acid (TCA) cycle pathway. It plays an essential role in regulating glucose metabolism for energy production by catalyzing the oxidative decarboxylation of pyruvate to acetyl coenzyme A. Importantly, the activity of PDC is regulated through post-translational modifications (PTMs), phosphorylation, acetylation, and O-GlcNAcylation. These PTMs have significant effects on PDC activity under both physiological and pathophysiological conditions, making them potential targets for metabolism-related diseases. This review specifically focuses on the PTMs of PDC in cardiovascular diseases (CVDs) such as myocardial ischemia/reperfusion injury, diabetic cardiomyopathy, obesity-related cardiomyopathy, heart failure (HF), and vascular diseases. The findings from this review offer theoretical references for the diagnosis, treatment, and prognosis of CVD.
Collapse
Affiliation(s)
- Bo Guo
- Department of Pharmacy, Northwest Woman’s and Children’s Hospital, Xi’an, China
| | - Fujiao Zhang
- College of Life Sciences, Northwest University, Xi’an, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Xingmin Ning
- College of Life Sciences, Northwest University, Xi’an, China
| | - Zihui Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Qinglei Meng
- College of Life Sciences, Yan’an University, Yan’an, China
| | - Ziqi Yang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Wenhua Jiang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Lijuan Sun
- Eye Institute of Chinese PLA and Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
8
|
Chen T, Xie Q, Tan B, Yi Q, Xiang H, Wang R, Zhou Q, He B, Tian J, Zhu J, Xu H. Inhibition of Pyruvate Dehydrogenase Kinase 4 Protects Cardiomyocytes from lipopolysaccharide-Induced Mitochondrial Damage by Reducing Lactate Accumulation. Inflammation 2024; 47:1356-1370. [PMID: 38401019 DOI: 10.1007/s10753-024-01981-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/22/2023] [Accepted: 01/19/2024] [Indexed: 02/26/2024]
Abstract
Mitochondrial dysfunction is considered one of the major pathogenic mechanisms of sepsis-induced cardiomyopathy (SIC). Pyruvate dehydrogenase kinase 4 (PDK4), a key regulator of mitochondrial metabolism, is essential for maintaining mitochondrial function. However, its specific role in SIC remains unclear. To investigate this, we established an in vitro model of septic cardiomyopathy using lipopolysaccharide (LPS)-induced H9C2 cardiomyocytes. Our study revealed a significant increase in PDK4 expression in LPS-treated H9C2 cardiomyocytes. Inhibiting PDK4 with dichloroacetic acid (DCA) improved cell survival, reduced intracellular lipid accumulation and calcium overload, and restored mitochondrial structure and respiratory capacity while decreasing lactate accumulation. Similarly, Oxamate, a lactate dehydrogenase inhibitor, exhibited similar effects to DCA in LPS-treated H9C2 cardiomyocytes. To further validate whether PDK4 causes cardiomyocyte and mitochondrial damage in SIC by promoting lactate production, we upregulated PDK4 expression using PDK4-overexpressing lentivirus in H9C2 cardiomyocytes. This resulted in elevated lactate levels, impaired mitochondrial structure, and reduced mitochondrial respiratory capacity. However, inhibiting lactate production reversed the mitochondrial dysfunction caused by PDK4 upregulation. In conclusion, our study highlights the pathogenic role of PDK4 in LPS-induced cardiomyocyte and mitochondrial damage by promoting lactate production. Therefore, targeting PDK4 and its downstream product lactate may serve as promising therapeutic approaches for treating SIC.
Collapse
Affiliation(s)
- Tangtian Chen
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Qiumin Xie
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Bin Tan
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Qin Yi
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Han Xiang
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Rui Wang
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Qin Zhou
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Bolin He
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jie Tian
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Cardiovascular (Internal Medicine), Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jing Zhu
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Hao Xu
- Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Department of Clinical Laboratory, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Children's Hospital of Chongqing Medical University, Box 136, No. 3 Zhongshan RD, Yuzhong District, Chongqing, 400014, China.
| |
Collapse
|
9
|
Xu L, Yang M, Wei A, Wei Z, Qin Y, Wang K, Li B, Chen K, Liu C, Li C, Wang T. Aerobic exercise-induced HIF-1α upregulation in heart failure: exploring potential impacts on MCT1 and MPC1 regulation. Mol Med 2024; 30:83. [PMID: 38867145 PMCID: PMC11167843 DOI: 10.1186/s10020-024-00854-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND The terminal stage of ischemic heart disease develops into heart failure (HF), which is characterized by hypoxia and metabolic disturbances in cardiomyocytes. The hypoxic failing heart triggers hypoxia-inducible factor-1α (HIF-1α) actions in the cells sensitized to hypoxia and induces metabolic adaptation by accumulating HIF-1α. Furthermore, soluble monocarboxylic acid transporter protein 1 (MCT1) and mitochondrial pyruvate carrier 1 (MPC1), as key nodes of metabolic adaptation, affect metabolic homeostasis in the failing rat heart. Aerobic exercise training has been reported to retard the progression of HF due to enhancing HIF-1α levels as well as MCT1 expressions, whereas the effects of exercise on MCT1 and MPC1 in HF (hypoxia) remain elusive. This research aimed to investigate the action of exercise associated with MCT1 and MPC1 on HF under hypoxia. METHODS The experimental rat models are composed of four study groups: sham stented (SHAM), HF sedentary (HF), HF short-term exercise trained (HF-E1), HF long-term exercise trained (HF-E2). HF was initiated via left anterior descending coronary artery ligation, the effects of exercise on the progression of HF were analyzed by ventricular ultrasound (ejection fraction, fractional shortening) and histological staining. The regulatory effects of HIF-1α on cell growth, MCT1 and MPC1 protein expression in hypoxic H9c2 cells were evaluated by HIF-1α activatort/inhibitor treatment and plasmid transfection. RESULTS Our results indicate the presence of severe pathological remodelling (as evidenced by deep myocardial fibrosis, increased infarct size and abnormal hypertrophy of the myocardium, etc.) and reduced cardiac function in the failing hearts of rats in the HF group compared to the SHAM group. Treadmill exercise training ameliorated myocardial infarction (MI)-induced cardiac pathological remodelling and enhanced cardiac function in HF exercise group rats, and significantly increased the expression of HIF-1α (p < 0.05), MCT1 (p < 0.01) and MPC1 (p < 0.05) proteins compared to HF group rats. Moreover, pharmacological inhibition of HIF-1α in hypoxic H9c2 cells dramatically downregulated MCT1 and MPC1 protein expression. This phenomenon is consistent with knockdown of HIF-1α at the gene level. CONCLUSION The findings propose that long-term aerobic exercise training, as a non- pharmacological treatment, is efficient enough to debilitate the disease process, improve the pathological phenotype, and reinstate cardiac function in HF rats. This benefit is most likely due to activation of myocardial HIF-1α and upregulation of MCT1 and MPC1.
Collapse
Affiliation(s)
- Longfei Xu
- Military Medical Sciences Academy, Tianjin, 300050, China
| | - Miaomiao Yang
- Military Medical Sciences Academy, Tianjin, 300050, China
| | - Aili Wei
- Military Medical Sciences Academy, Tianjin, 300050, China
| | - Zilin Wei
- Military Medical Sciences Academy, Tianjin, 300050, China
| | - Yingkai Qin
- Military Medical Sciences Academy, Tianjin, 300050, China
| | - Kun Wang
- Military Medical Sciences Academy, Tianjin, 300050, China
| | - Bin Li
- No. 950 Hospital of the Chinese People's Liberation Army, Yecheng, 844999, China
| | - Kang Chen
- Military Medical Sciences Academy, Tianjin, 300050, China
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China
| | - Chen Liu
- Military Medical Sciences Academy, Tianjin, 300050, China
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China
| | - Chao Li
- Military Medical Sciences Academy, Tianjin, 300050, China.
| | - Tianhui Wang
- Military Medical Sciences Academy, Tianjin, 300050, China.
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China.
| |
Collapse
|
10
|
Han X, Ashraf M, Shi H, Nkembo AT, Tipparaju SM, Xuan W. Combined Endurance and Resistance Exercise Mitigates Age-Associated Cardiac Dysfunction. Adv Biol (Weinh) 2024:e2400137. [PMID: 38773896 DOI: 10.1002/adbi.202400137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/07/2024] [Indexed: 05/24/2024]
Abstract
Aging is associated with a decline in cardiac function. Exercise has been shown to effectively reduce the risks of cardiovascular diseases. Here whether a combination of endurance and resistance exercises can improve cardiac function in aged mice during late life is investigated. Through transcriptome analysis, several signaling pathways activated in the hearts of 22-month-old mice after combined exercise, including cardiac muscle contraction, mitophagy, and longevity regulation are identified. Combined exercise training mitigated age-associated pathological cardiac hypertrophy, reduced oxidative stress, cardiac senescence, and enhanced cardiac function. Upstream stimulatory factor 2 (Usf2) is upregulated in the aged mouse hearts with combined exercise compared to sedentary mice. In the human cardiomyocytes senescent model, overexpression of Usf2 led to anti-senescence effects, while knockdown of Usf2 exacerbated cellular senescence. The results suggest that a combination of endurance and resistance exercises, such as swimming and resistance running, can mitigate age-related pathological cardiac remodeling and cardiac dysfunction in late life. These cardioprotective effects are likely due to the activation of Usf2 and its anti-senescence effect. Therefore, Usf2 can potentially be a novel therapeutic target for mitigating age-related cardiac dysfunction.
Collapse
Affiliation(s)
- Xiaowei Han
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Muhammad Ashraf
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Hong Shi
- Division of Rheumatology, Department of Internal Medicine, Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Augustine T Nkembo
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Srinivas M Tipparaju
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Wanling Xuan
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| |
Collapse
|
11
|
Liang Z, Ralph-Epps T, Schmidtke MW, Lazcano P, Denis SW, Balážová M, Teixeira da Rosa N, Chakkour M, Hazime S, Ren M, Schlame M, Houtkooper RH, Greenberg ML. Upregulation of the AMPK-FOXO1-PDK4 pathway is a primary mechanism of pyruvate dehydrogenase activity reduction in tafazzin-deficient cells. Sci Rep 2024; 14:11497. [PMID: 38769106 PMCID: PMC11106297 DOI: 10.1038/s41598-024-62262-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024] Open
Abstract
Barth syndrome (BTHS) is a rare disorder caused by mutations in the TAFAZZIN gene. Previous studies from both patients and model systems have established metabolic dysregulation as a core component of BTHS pathology. In particular, features such as lactic acidosis, pyruvate dehydrogenase (PDH) deficiency, and aberrant fatty acid and glucose oxidation have been identified. However, the lack of a mechanistic understanding of what causes these conditions in the context of BTHS remains a significant knowledge gap, and this has hindered the development of effective therapeutic strategies for treating the associated metabolic problems. In the current study, we utilized tafazzin-knockout C2C12 mouse myoblasts (TAZ-KO) and cardiac and skeletal muscle tissue from tafazzin-knockout mice to identify an upstream mechanism underlying impaired PDH activity in BTHS. This mechanism centers around robust upregulation of pyruvate dehydrogenase kinase 4 (PDK4), resulting from hyperactivation of AMP-activated protein kinase (AMPK) and subsequent transcriptional upregulation by forkhead box protein O1 (FOXO1). Upregulation of PDK4 in tafazzin-deficient cells causes direct phospho-inhibition of PDH activity accompanied by increased glucose uptake and elevated intracellular glucose concentration. Collectively, our findings provide a novel mechanistic framework whereby impaired tafazzin function ultimately results in robust PDK4 upregulation, leading to impaired PDH activity and likely linked to dysregulated metabolic substrate utilization. This mechanism may underlie previously reported findings of BTHS-associated metabolic dysregulation.
Collapse
Affiliation(s)
- Zhuqing Liang
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Tyler Ralph-Epps
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | | | - Pablo Lazcano
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Simone W Denis
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism Institute, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | - Mária Balážová
- Department of Membrane Biochemistry, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, 84005, Bratislava, Slovakia
| | | | - Mohamed Chakkour
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Sanaa Hazime
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Mindong Ren
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, Grossman School of Medicine, New York University, New York, NY, USA
| | - Michael Schlame
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, Grossman School of Medicine, New York University, New York, NY, USA
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism Institute, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | - Miriam L Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
12
|
Chi K, Liu J, Li X, Wang H, Li Y, Liu Q, Zhou Y, Ge Y. Biomarkers of heart failure: advances in omics studies. Mol Omics 2024; 20:169-183. [PMID: 38224222 DOI: 10.1039/d3mo00173c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Heart failure is a complex syndrome characterized by progressive circulatory dysfunction, manifesting clinically as pulmonary and systemic venous congestion, alongside inadequate tissue perfusion. The early identification of HF, particularly at the mild and moderate stages (stages B and C), presents a clinical challenge due to the overlap of signs, symptoms, and natriuretic peptide levels with other cardiorespiratory pathologies. Nonetheless, early detection coupled with timely pharmacological intervention is imperative for enhancing patient outcomes. Advances in high-throughput omics technologies have enabled researchers to analyze patient-derived biofluids and tissues, discovering biomarkers that are sensitive and specific for HF diagnosis. Due to the diversity of HF etiology, it is insufficient to study the diagnostic data of early HF using a single omics technology. This study reviewed the latest progress in genomics, transcriptomics, proteomics, and metabolomics for the identification of HF biomarkers, offering novel insights into the early clinical diagnosis of HF. However, the validity of biomarkers depends on the disease status, intervention time, genetic diversity and comorbidities of the subjects. Moreover, biomarkers lack generalizability in different clinical settings. Hence, it is imperative to conduct multi-center, large-scale and standardized clinical trials to enhance the diagnostic accuracy and utility of HF biomarkers.
Collapse
Affiliation(s)
- Kuo Chi
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Jing Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Xinghua Li
- Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| | - He Wang
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yanliang Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yabin Zhou
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| | - Yuan Ge
- Department of Cardiovascular Disease II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
13
|
Frisk C, Das S, Eriksson MJ, Walentinsson A, Corbascio M, Hage C, Kumar C, Ekström M, Maret E, Persson H, Linde C, Persson B. Cardiac biopsies reveal differences in transcriptomics between left and right ventricle in patients with or without diagnostic signs of heart failure. Sci Rep 2024; 14:5811. [PMID: 38461325 PMCID: PMC10924960 DOI: 10.1038/s41598-024-56025-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/29/2024] [Indexed: 03/11/2024] Open
Abstract
New or mild heart failure (HF) is mainly caused by left ventricular dysfunction. We hypothesised that gene expression differ between the left (LV) and right ventricle (RV) and secondly by type of LV dysfunction. We compared gene expression through myocardial biopsies from LV and RV of patients undergoing elective coronary bypass surgery (CABG). Patients were categorised based on LV ejection fraction (EF), diastolic function and NT-proBNP into pEF (preserved; LVEF ≥ 45%), rEF (reduced; LVEF < 45%) or normal LV function. Principal component analysis of gene expression displayed two clusters corresponding to LV and RV. Up-regulated genes in LV included natriuretic peptides NPPA and NPPB, transcription factors/coactivators STAT4 and VGLL2, ion channel related HCN2 and LRRC38 associated with cardiac muscle contraction, cytoskeleton, and cellular component movement. Patients with pEF phenotype versus normal differed in gene expression predominantly in LV, supporting that diastolic dysfunction and structural changes reflect early LV disease in pEF. DKK2 was overexpressed in LV of HFpEF phenotype, potentially leading to lower expression levels of β-catenin, α-SMA (smooth muscle actin), and enhanced apoptosis, and could be a possible factor in the development of HFpEF. CXCL14 was down-regulated in both pEF and rEF, and may play a role to promote development of HF.
Collapse
Affiliation(s)
- Christoffer Frisk
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Box 596, 751 24, Uppsala, Sweden
| | - Sarbashis Das
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Box 596, 751 24, Uppsala, Sweden
| | - Maria J Eriksson
- Department of Clinical Physiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Anna Walentinsson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 83, Gothenburg, Sweden
| | - Matthias Corbascio
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Thoracic Surgery, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Camilla Hage
- Department of Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Chanchal Kumar
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 83, Gothenburg, Sweden
- Department of Medicine, Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, 141 57, Huddinge, Sweden
| | - Mattias Ekström
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, 182 88, Stockholm, Sweden
- Department of Cardiology, Danderyd Hospital, 182 88, Stockholm, Sweden
| | - Eva Maret
- Department of Clinical Physiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Hans Persson
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, 182 88, Stockholm, Sweden
- Department of Cardiology, Danderyd Hospital, 182 88, Stockholm, Sweden
| | - Cecilia Linde
- Department of Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Bengt Persson
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Box 596, 751 24, Uppsala, Sweden.
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
14
|
Lopez-Vazquez P, Fernandez-Caggiano M, Barge-Caballero E, Barge-Caballero G, Couto-Mallon D, Grille-Cancela Z, Blanco-Canosa P, Paniagua-Martin MJ, Enriquez-Vazquez D, Vazquez-Rodriguez JM, Domenech N, Crespo-Leiro MG. Reduced mitochondrial pyruvate carrier expression in hearts with heart failure and reduced ejection fraction patients: ischemic vs. non-ischemic origin. Front Cardiovasc Med 2024; 11:1349417. [PMID: 38525191 PMCID: PMC10957580 DOI: 10.3389/fcvm.2024.1349417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/21/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction and objectives Mitochondrial pyruvate carrier (MPC) mediates the entry of pyruvate into mitochondria, determining whether pyruvate is incorporated into the Krebs cycle or metabolized in the cytosol. In heart failure (HF), a large amount of pyruvate is metabolized to lactate in the cytosol rather than being oxidized inside the mitochondria. Thus, MPC activity or expression might play a key role in the fate of pyruvate during HF. The purpose of this work was to study the levels of the two subunits of this carrier, named MPC1 and MPC2, in human hearts with HF of different etiologies. Methods Protein and mRNA expression analyses were conducted in cardiac tissues from three donor groups: patients with HF with reduced ejection fraction (HFrEF) with ischemic cardiomyopathy (ICM) or idiopathic dilated cardiomyopathy (IDC), and donors without cardiac pathology (Control). MPC2 plasma levels were determined by ELISA. Results Significant reductions in the levels of MPC1, MPC2, and Sirtuin 3 (SIRT3) were observed in ICM patients compared with the levels in the Control group. However, no statistically significant differences were revealed in the analysis of MPC1 and MPC2 gene expression among the groups. Interestingly, Pyruvate dehydrogenase complex (PDH) subunits expression were increased in the ICM patients. In the case of IDC patients, a significant decrease in MPC1 was observed only when compared with the Control group. Notably, plasma MPC2 levels were found to be elevated in both disease groups compared with that in the Control group. Conclusion Decreases in MPC1 and/or MPC2 levels were detected in the cardiac tissues of HFrEF patients, with ischemic or idiopatic origen, indicating a potential reduction in mitochondrial pyruvate uptake in the heart, which could be linked to unfavorable clinical features.
Collapse
Affiliation(s)
- Paula Lopez-Vazquez
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Mariana Fernandez-Caggiano
- Barts & The London School of Medicine & Dentistry, William Harvey Research Institute, Queen Mary University of London, London, England
| | - Eduardo Barge-Caballero
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Gonzalo Barge-Caballero
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - David Couto-Mallon
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Zulaika Grille-Cancela
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Paula Blanco-Canosa
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Maria J. Paniagua-Martin
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Enriquez-Vazquez
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose M. Vazquez-Rodriguez
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Nieves Domenech
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Maria G. Crespo-Leiro
- Servicio de Cardiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
15
|
Liang Z, Ralph-Epps T, Schmidtke MW, Lazcano P, Denis SW, Balážová M, Chakkour M, Hazime S, Ren M, Schlame M, Houtkooper RH, Greenberg ML. Upregulation of the AMPK-FOXO1-PDK4 pathway is a primary mechanism of pyruvate dehydrogenase activity reduction and leads to increased glucose uptake in tafazzin-deficient cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.03.578755. [PMID: 38352304 PMCID: PMC10862887 DOI: 10.1101/2024.02.03.578755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Barth syndrome (BTHS) is a rare disorder caused by mutations in the TAFAZZIN gene. Previous studies from both patients and model systems have established metabolic dysregulation as a core component of BTHS pathology. In particular, features such as lactic acidosis, pyruvate dehydrogenase (PDH) deficiency, and aberrant fatty acid and glucose oxidation have been identified. However, the lack of a mechanistic understanding of what causes these conditions in the context of BTHS remains a significant knowledge gap, and this has hindered the development of effective therapeutic strategies for treating the associated metabolic problems. In the current study, we utilized tafazzin-knockout C2C12 mouse myoblasts (TAZ-KO) and cardiac and skeletal muscle tissue from tafazzin-knockout mice to identify an upstream mechanism underlying impaired PDH activity in BTHS. This mechanism centers around robust upregulation of pyruvate dehydrogenase kinase 4 (PDK4), resulting from hyperactivation of AMP-activated protein kinase (AMPK) and subsequent transcriptional upregulation by forkhead box protein O1 (FOXO1). Upregulation of PDK4 in tafazzin-deficient cells causes direct phospho-inhibition of PDH activity accompanied by increased glucose uptake and elevated intracellular glucose concentration. Collectively, our findings provide a novel mechanistic framework whereby impaired tafazzin function ultimately results in robust PDK4 upregulation, leading to impaired PDH activity and likely linked to dysregulated metabolic substrate utilization. This mechanism may underlie previously reported findings of BTHS-associated metabolic dysregulation.
Collapse
|
16
|
Meng S, Yu Y, Yu S, Zhu S, Shi M, Xiang M, Ma H. Advances in Metabolic Remodeling and Intervention Strategies in Heart Failure. J Cardiovasc Transl Res 2024; 17:36-55. [PMID: 37843752 DOI: 10.1007/s12265-023-10443-0] [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: 06/29/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
The heart is the most energy-demanding organ throughout the whole body. Perturbations or failure in energy metabolism contributes to heart failure (HF), which represents the advanced stage of various heart diseases. The poor prognosis and huge economic burden associated with HF underscore the high unmet need to explore novel therapies targeting metabolic modulators beyond conventional approaches focused on neurohormonal and hemodynamic regulators. Emerging evidence suggests that alterations in metabolic substrate reliance, metabolic pathways, metabolic by-products, and energy production collectively regulate the occurrence and progression of HF. In this review, we provide an overview of cardiac metabolic remodeling, encompassing the utilization of free fatty acids, glucose metabolism, ketone bodies, and branched-chain amino acids both in the physiological condition and heart failure. Most importantly, the latest advances in pharmacological interventions are discussed as a promising therapeutic approach to restore cardiac function, drawing insights from recent basic research, preclinical and clinical studies.
Collapse
Affiliation(s)
- Simin Meng
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Yi Yu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Shiyu Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Mengjia Shi
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
17
|
Rabkin SW, Wong CN. Epigenetics in Heart Failure: Role of DNA Methylation in Potential Pathways Leading to Heart Failure with Preserved Ejection Fraction. Biomedicines 2023; 11:2815. [PMID: 37893188 PMCID: PMC10604152 DOI: 10.3390/biomedicines11102815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
This review will focus on epigenetic modifications utilizing the DNA methylation mechanism, which is potentially involved in the pathogenesis of heart failure with preserved ejection fraction (HFpEF). The putative pathways of HFpEF will be discussed, specifically myocardial fibrosis, myocardial inflammation, sarcoplasmic reticulum Ca2+-ATPase, oxidative-nitrosative stress, mitochondrial and metabolic defects, as well as obesity. The relationship of HFpEF to aging and atrial fibrillation will be examined from the perspective of DNA methylation.
Collapse
Affiliation(s)
- Simon W. Rabkin
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Division of Cardiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Chenille N. Wong
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
18
|
Olou AA, Ambrose J, Jack JL, Walsh M, Ruckert MT, Eades AE, Bye BA, Dandawate P, VanSaun MN. SHP2 regulates adipose maintenance and adipocyte-pancreatic cancer cell crosstalk via PDHA1. J Cell Commun Signal 2023; 17:575-590. [PMID: 36074246 PMCID: PMC10409927 DOI: 10.1007/s12079-022-00691-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
Adipocytes are the most abundant cell type in the adipose tissue, and their dysfunction is a significant driver of obesity-related pathologies, such as cancer. The mechanisms that (1) drive the maintenance and secretory activity of adipocytes and (2) mediate the cancer cellular response to the adipocyte-derived factors are not fully understood. To address that gap of knowledge, we investigated how alterations in Src homology region 2-containing protein (SHP2) activity affect adipocyte function and tumor crosstalk. We found that phospho-SHP2 levels are elevated in adipose tissue of obese mice, obese patients, and differentiating adipocytes. Immunofluorescence and immunoprecipitation analyses as well as in-silico protein-protein interaction modeling demonstrated that SHP2 associates with PDHA1, and that a positive association promotes a reactive oxygen species (ROS)-driven adipogenic program. Accordingly, this SHP2-PDHA1-ROS regulatory axis was crucial for adipocyte maintenance and secretion of interleukin-6 (IL-6), a key cancer-promoting cytokine. Mature adipocytes treated with an inhibitor for SHP2, PDHA1, or ROS exhibited an increased level of pro-lipolytic and thermogenic proteins, corresponding to an increased glycerol release, but a suppression of secreted IL-6. A functional analysis of adipocyte-cancer cell crosstalk demonstrated a decreased migration, invasion, and a slight suppression of cell cycling, corresponding to a reduced growth of pancreatic cancer cells exposed to conditioned media (CM) from mature adipocytes previously treated with inhibitors for SHP2/PDHA1/ROS. Importantly, PDAC cell growth stimulation in response to adipocyte CM correlated with PDHA1 induction but was suppressed by a PDHA1 inhibitor. The data point to a novel role for (1) SHP2-PDHA1-ROS in adipocyte maintenance and secretory activity and (2) PDHA1 as a regulator of the pancreatic cancer cells response to adipocyte-derived factors.
Collapse
Affiliation(s)
- Appolinaire A Olou
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| | - Joe Ambrose
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Jarrid L Jack
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - McKinnon Walsh
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Mariana T Ruckert
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Austin E Eades
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Bailey A Bye
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Prasad Dandawate
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Michael N VanSaun
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
19
|
Shen M, Zheng C, Chen L, Li M, Huang X, He M, Liu C, Lin H, Liao W, Bin J, Cao S, Liao Y. LCZ696 (sacubitril/valsartan) inhibits pulmonary hypertension induced right ventricular remodeling by targeting pyruvate dehydrogenase kinase 4. Biomed Pharmacother 2023; 162:114569. [PMID: 37001183 DOI: 10.1016/j.biopha.2023.114569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Right ventricular (RV) function is a major prognostic factor in patients with cardiopulmonary disease. Effective medical therapies are available for left heart failure, but they are usually less effective or even ineffective in right heart failure. Here, we tested the hypothesis that LCZ696 (sacubitril/valsartan) can attenuate pressure overload-induced RV remodeling by inhibiting pyruvate dehydrogenase kinase 4 (PDK4). METHODS Adult male C57 mice were subjected to transverse aortic constriction (TAC), pulmonary artery constriction (PAC), or sham surgery. Bioinformatics analysis was used to screen for common differentially expressed genes (DEGs) between TAC and PAC. Chemical compounds targeting DEGs were predicted by molecular docking analysis. Effects of LCZ696 on PAC-induced RV remodeling and the associated PDK4-related mechanisms were investigated. RESULTS We found 60 common DEGs between PAC and TAC, and Pdk4 was one of the downregulated DEGs. From 47 chemical compounds with potential cardiovascular activity and PDK4 protein binding ability, we selected LCZ696 to treat PAC-induced RV remodeling because of its high docking score for binding PDK4. Compared with vehicle-treated PAC mice, LCZ696-treated mice had significantly smaller RV wall thickness and RV diameters, less myocardial fibrosis, lower expression of PDK4 protein, and less phosphorylation of glycogen synthase kinase-3β (p-GSK3β). In PAC mice, overexpression of Pdk4 blocked the inhibitory effect of LCZ696 on RV remodeling, whereas conditional knockout of Pdk4 attenuated PAC-induced RV remodeling. CONCLUSIONS Pdk4 is a common therapeutic target for pressure overload-induced left ventricular and RV remodeling, and LCZ696 attenuates RV remodeling by downregulating Pdk4 and inhibiting PDK4/p-GSK3β signal.
Collapse
|
20
|
Shi W, Zhou X, Li X, Peng X, Chen G, Li Y, Zhang C, Yu H, Feng Z, Gou X, Fan J. Human Umbilical Cord Mesenchymal Stem Cells Protect against Renal Ischemia-Reperfusion Injury by Secreting Extracellular Vesicles Loaded with miR-148b-3p That Target Pyruvate Dehydrogenase Kinase 4 to Inhibit Endoplasmic Reticulum Stress at the Reperfusion Stages. Int J Mol Sci 2023; 24:ijms24108899. [PMID: 37240246 DOI: 10.3390/ijms24108899] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Renal ischemia-reperfusion (I/R) injury is a leading cause of acute kidney injury (AKI), with high mortality. Recent studies have reported that human umbilical cord mesenchymal stem cells (HucMSCs) play an important role in repairing organ and tissue injuries because of their unique characteristics. However, the potential of HucMSC extracellular vesicles (HucMSC-EVs) to promote the repair of renal tubular cells remains to be explored. This study found that HucMSC-EVs derived from HucMSCs played a protective role and were associated with kidney I/R injury. We found that miR-148b-3p in HucMSC-EVs had a protective effect against kidney I/R injury. HK-2 cells overexpressing miR-148b-3p were protected against I/R injury by inhibiting apoptosis. Next, the target mRNA of miR-148b-3p was predicted online, and the target mRNA, pyruvate dehydrogenase kinase 4 (PDK4), was identified and verified using dual luciferase. We discovered that I/R injury significantly increased endoplasmic reticulum (ER) stress, whereas siR-PDK4 inhibited these effects and protected against I/R injury. Interestingly, after administrating HucMSC-EVs to HK-2 cells, PDK4 expression and ER stress induced by I/R injury were significantly inhibited. HK-2 ingested miR-148b-3p from HucMSC-EVs, and its ER induced by I/R injury was significantly deregulated. This study suggests that HucMSC-EVs protect kidneys from I/R injury during the early I/R stage. These results suggest a new mechanism for HucMSC-EVs in treating AKI and provide a new treatment strategy for I/R injury.
Collapse
Affiliation(s)
- Wei Shi
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiang Zhou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xinyuan Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiang Peng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Guo Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yang Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chunlin Zhang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haitao Yu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhenwei Feng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jing Fan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
21
|
Shen X, Chang P, Zhang X, Zhang J, Wang X, Quan Z, Wang P, Liu T, Niu Y, Zheng R, Chen B, Yu J. The landscape of N6-methyladenosine modification patterns and altered transcript profiles in the cardiac-specific deletion of natriuretic peptide receptor A. Mol Omics 2023; 19:105-125. [PMID: 36412146 DOI: 10.1039/d2mo00201a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The atrial natriuretic peptide (ANP) and the brain natriuretic peptide (BNP) are critical biological makers and regulators of cardiac functions. Our previous results show that NPRA (natriuretic peptide receptor A)-deficient mice have distinct metabolic patterns and expression profiles compared with the control. Still, the molecular mechanism that could account for this observation remains to be elucidated. Here, methylation alterations were detected by mazF-digestion, and differentially expressed genes of transcriptomes were detected by a Genome Oligo Microarray using the myocardium from NPRA-deficient (NPRA-/-) mice and wild-type (NPRA+/+) mice as the control. Comprehensive analysis of m6A methylation data gave an altered landscape of m6A modification patterns and altered transcript profiles in cardiac-specific NPRA-deficient mice. The m6A "reader" igf2bp3 showed a clear trend of increase, suggesting a function in altered methylation and expression in cardiac-specific NPRA-deficient mice. Intriguingly, differentially m6A-methylated genes were enriched in the metabolic process and insulin resistance pathway, suggesting a regulatory role in cardiac metabolism of m6A modification regulated by NPRA. Notably, it was confirmed that the pyruvate dehydrogenase kinase 4 (Pdk4) gene upregulated the gene expression and the hypermethylation level simultaneously, which may be the key factor for the cardiac metabolic imbalance and insulin resistance caused by natriuretic peptide signal resistance. Taken together, cardiac metabolism might be regulated by natriuretic peptide signaling, with decreased m6A methylation and a decrease of Pdk4.
Collapse
Affiliation(s)
- Xi Shen
- Clinical Experimental Centre, Xi'an International Medical Centre Hospital, 777, Xitai Road, Hightech-zone, Xi'an, Shaanxi 710100, P. R. China. .,Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, P. R. China
| | - Pan Chang
- Department of Cardiology, the Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Xiaomeng Zhang
- Department of Cardiology, the Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Jing Zhang
- Department of Cardiology, the Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Xihui Wang
- Department of Cardiology, the Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Zhuo Quan
- Clinical Experimental Centre, Xi'an International Medical Centre Hospital, 777, Xitai Road, Hightech-zone, Xi'an, Shaanxi 710100, P. R. China. .,Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, P. R. China
| | - Pengli Wang
- Clinical Experimental Centre, Xi'an International Medical Centre Hospital, 777, Xitai Road, Hightech-zone, Xi'an, Shaanxi 710100, P. R. China. .,Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, P. R. China
| | - Tian Liu
- Clinical Experimental Centre, Xi'an International Medical Centre Hospital, 777, Xitai Road, Hightech-zone, Xi'an, Shaanxi 710100, P. R. China. .,Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, P. R. China
| | - Yan Niu
- Clinical Experimental Centre, Xi'an International Medical Centre Hospital, 777, Xitai Road, Hightech-zone, Xi'an, Shaanxi 710100, P. R. China. .,Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, P. R. China
| | - Rong Zheng
- Clinical Experimental Centre, Xi'an International Medical Centre Hospital, 777, Xitai Road, Hightech-zone, Xi'an, Shaanxi 710100, P. R. China. .,Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, P. R. China
| | - Baoying Chen
- Imaging Diagnosis and Treatment Centre, Xi'an International Medical Centre Hospital, 777, Xitai Road, Hightech-zone, Xi'an, Shaanxi 710100, P. R. China.
| | - Jun Yu
- Clinical Experimental Centre, Xi'an International Medical Centre Hospital, 777, Xitai Road, Hightech-zone, Xi'an, Shaanxi 710100, P. R. China. .,Xi'an Engineering Technology Research Center for Cardiovascular Active Peptides, P. R. China
| |
Collapse
|
22
|
Wang X, Huang Y, Zhang K, Chen F, Nie T, Zhao Y, He F, Ni J. Changes of energy metabolism in failing heart and its regulation by SIRT3. Heart Fail Rev 2023:10.1007/s10741-023-10295-5. [PMID: 36708431 DOI: 10.1007/s10741-023-10295-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 01/29/2023]
Abstract
Heart failure (HF) is the leading cause of hospitalization in elderly patients and a disease with extremely high morbidity and mortality rate worldwide. Although there are some existing treatment methods for heart failure, due to its complex pathogenesis and often accompanied by various comorbidities, there is still a lack of specific drugs to treat HF. The mortality rate of patients with HF is still high, highlighting an urgent need to elucidate the pathophysiological mechanisms of HF and seek new therapeutic approaches. The heart is an organ with a very high metabolic intensity, mainly using fatty acids, glucose, ketone bodies, and branched-chain amino acids as energy substrates to supply energy for the heart. Loss of metabolic flexibility and metabolic remodeling occurs with HF. Sirtuin3 (SIRT3) is a member of the NAD+-dependent Sirtuin family located in mitochondria, and can participate in mitochondrial physiological functions through the deacetylation of metabolic and respiratory enzymes in mitochondria. As the center of energy metabolism, mitochondria are involved in many physiological processes. Maintaining stable metabolic and physiological functions of the heart depends on normal mitochondrial function. The damage or loss of SIRT3 can lead to various cardiovascular diseases. Therefore, we summarize the recent progress of SIRT3 in cardiac mitochondrial protection and metabolic remodeling.
Collapse
Affiliation(s)
- Xiao Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yuting Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, 341000, China
| | - Kai Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Feng Chen
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Tong Nie
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yun Zhao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Feng He
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, 438000, China.
| | - Jingyu Ni
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
23
|
Huang X, Zeng Z, Li S, Xie Y, Tong X. The Therapeutic Strategies Targeting Mitochondrial Metabolism in Cardiovascular Disease. Pharmaceutics 2022; 14:pharmaceutics14122760. [PMID: 36559254 PMCID: PMC9788260 DOI: 10.3390/pharmaceutics14122760] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is a group of systemic disorders threatening human health with complex pathogenesis, among which mitochondrial energy metabolism reprogramming has a critical role. Mitochondria are cell organelles that fuel the energy essential for biochemical reactions and maintain normal physiological functions of the body. Mitochondrial metabolic disorders are extensively involved in the progression of CVD, especially for energy-demanding organs such as the heart. Therefore, elucidating the role of mitochondrial metabolism in the progression of CVD is of great significance to further understand the pathogenesis of CVD and explore preventive and therapeutic methods. In this review, we discuss the major factors of mitochondrial metabolism and their potential roles in the prevention and treatment of CVD. The current application of mitochondria-targeted therapeutic agents in the treatment of CVD and advances in mitochondria-targeted gene therapy technologies are also overviewed.
Collapse
Affiliation(s)
- Xiaoyang Huang
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Zhenhua Zeng
- Biomedical Research Center, Hunan University of Medicine, Huaihua 418000, China
| | - Siqi Li
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Yufei Xie
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoyong Tong
- Department of Pharmacology and Pharmacy, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
- Jinfeng Laboratory, Chongqing 401329, China
- Correspondence:
| |
Collapse
|
24
|
Abdullah CS, Remex NS, Aishwarya R, Nitu S, Kolluru GK, Traylor J, Hartman B, King J, Bhuiyan MAN, Hall N, Murnane KS, Goeders NE, Kevil CG, Orr AW, Bhuiyan MS. Mitochondrial dysfunction and autophagy activation are associated with cardiomyopathy developed by extended methamphetamine self-administration in rats. Redox Biol 2022; 58:102523. [PMID: 36335762 PMCID: PMC9641018 DOI: 10.1016/j.redox.2022.102523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
The recent rise in illicit use of methamphetamine (METH), a highly addictive psychostimulant, is a huge health care burden due to its central and peripheral toxic effects. Mounting clinical studies have noted that METH use in humans is associated with the development of cardiomyopathy; however, preclinical studies and animal models to dissect detailed molecular mechanisms of METH-associated cardiomyopathy development are scarce. The present study utilized a unique very long-access binge and crash procedure of METH self-administration to characterize the sequelae of pathological alterations that occur with METH-associated cardiomyopathy. Rats were allowed to intravenously self-administer METH for 96 h continuous weekly sessions over 8 weeks. Cardiac function, histochemistry, ultrastructure, and biochemical experiments were performed 24 h after the cessation of drug administration. Voluntary METH self-administration induced pathological cardiac remodeling as indicated by cardiomyocyte hypertrophy, myocyte disarray, interstitial and perivascular fibrosis accompanied by compromised cardiac systolic function. Ultrastructural examination and native gel electrophoresis revealed altered mitochondrial morphology and reduced mitochondrial oxidative phosphorylation (OXPHOS) supercomplexes (SCs) stability and assembly in METH exposed hearts. Redox-sensitive assays revealed significantly attenuated mitochondrial respiratory complex activities with a compensatory increase in pyruvate dehydrogenase (PDH) activity reminiscent of metabolic remodeling. Increased autophagy flux and increased mitochondrial antioxidant protein level was observed in METH exposed heart. Treatment with mitoTEMPO reduced the autophagy level indicating the involvement of mitochondrial dysfunction in the adaptive activation of autophagy in METH exposed hearts. Altogether, we have reported a novel METH-associated cardiomyopathy model using voluntary drug seeking behavior. Our studies indicated that METH self-administration profoundly affects mitochondrial ultrastructure, OXPHOS SCs assembly and redox activity accompanied by increased PDH activity that may underlie observed cardiac dysfunction.
Collapse
Affiliation(s)
- Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Richa Aishwarya
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Sadia Nitu
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Gopi K Kolluru
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - James Traylor
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Brandon Hartman
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Judy King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Mohammad Alfrad Nobel Bhuiyan
- Department of Medicine, Division of Clinical Informatics, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Nicole Hall
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Kevin Sean Murnane
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Psychiatry, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Nicholas E Goeders
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
25
|
Liu M, Lv J, Pan Z, Wang D, Zhao L, Guo X. Mitochondrial dysfunction in heart failure and its therapeutic implications. Front Cardiovasc Med 2022; 9:945142. [PMID: 36093152 PMCID: PMC9448986 DOI: 10.3389/fcvm.2022.945142] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022] Open
Abstract
The ATP consumption in heart is very intensive to support muscle contraction and relaxation. Mitochondrion is the power plant of the cell. Mitochondrial dysfunction has long been believed as the primary mechanism responsible for the inability of energy generation and utilization in heart failure. In addition, emerging evidence has demonstrated that mitochondrial dysfunction also contributes to calcium dysregulation, oxidative stress, proteotoxic insults and cardiomyocyte death. These elements interact with each other to form a vicious circle in failing heart. The role of mitochondrial dysfunction in the pathogenesis of heart failure has attracted increasing attention. The complex signaling of mitochondrial quality control provides multiple targets for maintaining mitochondrial function. Design of therapeutic strategies targeting mitochondrial dysfunction holds promise for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Miaosen Liu
- Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jialan Lv
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhicheng Pan
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongfei Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liding Zhao
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Xiaogang Guo,
| |
Collapse
|
26
|
Meng R, Song J, Guan L, Li Q, Shi C, Su D, Ma X. Genome-wide analysis of methylation in rat fetal heart under hyperglycemia by methylation-dependent restriction site–associated DNA sequencing. PLoS One 2022; 17:e0268117. [PMID: 35544480 PMCID: PMC9094537 DOI: 10.1371/journal.pone.0268117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/24/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetes mellitus causes an increased incidence of congenital heart malformations. However, the pathogenesis and potential epigenetic mechanism involved in this process are unclear. In this study, we used MethylRAD sequencing to compare changes in methylation levels in the genomic landscapes in the fetal heart in a rat model of hyperglycemia. Our results showed that methylation of CCGG/CCNGG sites were mostly enriched in intergenic regions, followed by intron, exon, upstream and the 5′ and 3′ untranslated regions. qRT-PCR results confirmed the MethylRAD sequencing findings, suggesting that abnormal CCGG/CCNGG methylation in the upstream region regulated gene expression. The differential methylation genes (DMGs) based on the CCGG and CCNGG sites in the upstream region were examined by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis. Gene Ontology indicated that the CCGG-based DMGs involved in biological process and function were mainly related to transcription and co-SMAD binding. The CCNGG-based DMGs were mainly related to transcription and cytokine-mediated signaling pathways. Kyoto Encyclopedia of Genes and Genomes analysis indicated that CCGG-based DMGs were mainly involved in the Wnt signaling and TGF-β signaling pathways. CCNGG-based DMGs were involved in the TNF signaling and apoptosis pathways. These genes may play dominant roles in cardiomyocyte apoptosis and heart disease and require further study. These genes may also serve as potential molecular targets or diagnostic biomarkers for heart malformations under hyperglycemia.
Collapse
Affiliation(s)
- Rui Meng
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Department of Genetics, National Research Institute for Family Planning, Health Department, Beijing, China
| | - Junxian Song
- Department of Cardiology, Peking University People’s Hospital, Beijing, China
| | - Lina Guan
- Department of Genetics, National Research Institute for Family Planning, Health Department, Beijing, China
| | - Qian Li
- Department of Genetics, National Research Institute for Family Planning, Health Department, Beijing, China
| | - Cuige Shi
- Department of Genetics, National Research Institute for Family Planning, Health Department, Beijing, China
| | - Dongmei Su
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Department of Genetics, National Research Institute for Family Planning, Health Department, Beijing, China
- * E-mail: (DS); , (XM)
| | - Xu Ma
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Department of Genetics, National Research Institute for Family Planning, Health Department, Beijing, China
- * E-mail: (DS); , (XM)
| |
Collapse
|
27
|
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the general population. Energy metabolism disturbance is one of the early abnormalities in CVDs, such as coronary heart disease, diabetic cardiomyopathy, and heart failure. To explore the role of myocardial energy homeostasis disturbance in CVDs, it is important to understand myocardial metabolism in the normal heart and their function in the complex pathophysiology of CVDs. In this article, we summarized lipid metabolism/lipotoxicity and glucose metabolism/insulin resistance in the heart, focused on the metabolic regulation during neonatal and ageing heart, proposed potential metabolic mechanisms for cardiac regeneration and degeneration. We provided an overview of emerging molecular network among cardiac proliferation, regeneration, and metabolic disturbance. These novel targets promise a new era for the treatment of CVDs.
Collapse
Affiliation(s)
- Lu-Yun WANG
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen CHEN
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Piquereau J, Boitard SE, Ventura-Clapier R, Mericskay M. Metabolic Therapy of Heart Failure: Is There a Future for B Vitamins? Int J Mol Sci 2021; 23:30. [PMID: 35008448 PMCID: PMC8744601 DOI: 10.3390/ijms23010030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 01/17/2023] Open
Abstract
Heart failure (HF) is a plague of the aging population in industrialized countries that continues to cause many deaths despite intensive research into more effective treatments. Although the therapeutic arsenal to face heart failure has been expanding, the relatively short life expectancy of HF patients is pushing towards novel therapeutic strategies. Heart failure is associated with drastic metabolic disorders, including severe myocardial mitochondrial dysfunction and systemic nutrient deprivation secondary to severe cardiac dysfunction. To date, no effective therapy has been developed to restore the cardiac energy metabolism of the failing myocardium, mainly due to the metabolic complexity and intertwining of the involved processes. Recent years have witnessed a growing scientific interest in natural molecules that play a pivotal role in energy metabolism with promising therapeutic effects against heart failure. Among these molecules, B vitamins are a class of water soluble vitamins that are directly involved in energy metabolism and are of particular interest since they are intimately linked to energy metabolism and HF patients are often B vitamin deficient. This review aims at assessing the value of B vitamin supplementation in the treatment of heart failure.
Collapse
Affiliation(s)
- Jérôme Piquereau
- UMR-S 1180, Inserm Unit of Signaling and Cardiovascular Pathophysiology, Faculty of Pharmacy, Université Paris-Saclay, 92296 Chatenay-Malabry, France; (S.E.B.); (R.V.-C.)
| | | | | | - Mathias Mericskay
- UMR-S 1180, Inserm Unit of Signaling and Cardiovascular Pathophysiology, Faculty of Pharmacy, Université Paris-Saclay, 92296 Chatenay-Malabry, France; (S.E.B.); (R.V.-C.)
| |
Collapse
|
29
|
Liu J, Wang X, Xue F, Zheng M, Luan Q. Abnormal mitochondrial structure and function are retained in gingival tissues and human gingival fibroblasts from patients with chronic periodontitis. J Periodontal Res 2021; 57:94-103. [PMID: 34826335 DOI: 10.1111/jre.12941] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/17/2021] [Accepted: 09/25/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND OBJECTIVE The abnormal structure and function of mitochondria in cells is closely associated with inflammatory diseases. However, the physiology of mitochondria within gingival tissues and human gingival fibroblasts (HGFs) in patients with chronic periodontitis (CP) remains unclear. The objective of this study was to investigate the structure profile and function of mitochondria in gingival tissues and in HGFs derived from patients with or without CP. These features of mitochondria in HGFs were further analyzed when HGFs were induced by lipopolysaccharide (LPS) from Porphyromonas gingivalis (P.g). METHODS Gingival tissues and HGFs were collected from CP and healthy patients. Mitochondrial structure was assessed by transmission electron microscopy. Tissues or cells lysis was performed for mitochondrial DNA (mtDNA) quantification, and real-time polymerase chain reaction (RT-PCR) tests were used to determine mtDNA copy numbers. Western blot analysis was used to evaluate autophagy-related protein (ATG)-5, microtubule-associated protein light chain 3 (LC3), and mitochondrial matrix protein pyruvate dehydrogenase kinase isozyme 2 (PDK2) levels in tissues and HGFs from CP and healthy individuals. RESULTS Tissues and HGFs from CP showed a significant greater mitochondrial structure destruction, lower mtDNA level, increased ATG5, LC3-II, and lower PDK2 protein levels than those of healthy individuals. In addition, LPS from P.g also triggered the same results in HGFs from healthy donors. Moreover, the challenge of HGFs from CP with LPS worsened these parameters. CONCLUSION Mitochondrial structure and function within gingival tissues and HGFs from CP individuals were abnormal compared to those from healthy donors, and LPS could promote mitochondrial destruction.
Collapse
Affiliation(s)
- Jia Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xiaoxuan Wang
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Fei Xue
- National Clinical Research Center for Oral Diseases, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Beijing, China.,First Clinical Division, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ming Zheng
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Qingxian Luan
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, China.,Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
30
|
Mi S, Jiang H, Zhang L, Xie Z, Zhou J, Sun A, Jin H, Ge J. Regulation of Cardiac-Specific Proteins Expression by Moderate-Intensity Aerobic Exercise Training in Mice With Myocardial Infarction Induced Heart Failure Using MS-Based Proteomics. Front Cardiovasc Med 2021; 8:732076. [PMID: 34692783 PMCID: PMC8531249 DOI: 10.3389/fcvm.2021.732076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/18/2021] [Indexed: 01/14/2023] Open
Abstract
This study aims to systematically reveal the changes in protein levels induced by regular exercise in mice with ischemic-induced heart failure (HF). Aerobic exercise training for the ischemic-induced HF mice lasted for 4 weeks and then we used the liquid chromatography-mass spectrometry method to identify and quantify the protein profile in the myocardium of mice. As a whole, 1,304 proteins (597 proteins up-regulated; 707 proteins down-regulated) were differentially expressed between the exercise group and the sedentary group, including numerous proteins related to energy metabolism. The significant alteration of the component (E1 component subunit alpha and subunit beta) and the activity-regulating enzyme (pyruvate dehydrogenase kinase 2 and pyruvate dehydrogenase kinase 4) of pyruvate dehydrogenase complex and poly [ADP-ribose] polymerase 3, a nicotinamide adenine dinucleotide(+)-consuming enzymes, was further verified in targeted analysis. Generally, this proteomics profiling furnishes a systematic insight of the influence of aerobic exercise on HF.
Collapse
Affiliation(s)
- Shouling Mi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Hao Jiang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Lei Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhonglei Xie
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jingmin Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hong Jin
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Stomatological Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Perez DM. The promise and problems of metabolic-based therapies for heart failure. Interv Cardiol 2021; 13:415-424. [PMID: 34970333 PMCID: PMC8715677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023] Open
Abstract
Despite standard therapies, heart failure patients have high rates of morbidity highlighting the need to develop alternative therapeutic approaches. Heart failure has been described as an energy-starved condition that is hypothesized to drive the pathological remodeling of the heart. Numerous studies have described the metabolic defects that occur when the heart fails and adaptive changes that take place to maintain the energy needed for the heart to function properly. In this review we will summarize the metabolic requirements of a normal heart and what happens during failure. We will also summarize the various metabolic therapeutic strategies that have been developed over the years to treat heart failure and their results from clinical trials.
Collapse
Affiliation(s)
- Dianne M. Perez
- Author for correspondence: Dianne M. Perez, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA,
| |
Collapse
|
32
|
Yin Q, Wang P, Wu X. MicroRNA -148 alleviates cardiac dysfunction, immune disorders and myocardial apoptosis in myocardial ischemia-reperfusion (MI/R) injury by targeting pyruvate dehydrogenase kinase (PDK4). Bioengineered 2021; 12:5552-5565. [PMID: 34517782 PMCID: PMC8806724 DOI: 10.1080/21655979.2021.1965812] [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] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Ischemic heart disease in children may be induced by varied factors, and there is no corresponding systematic treatment up to now. This study aims to investigate the effects of microRNA (miR)-148 on myocardial injury in immature rats with myocardial ischemia-reperfusion (MI/R) injury. In this study, MI/R model was established by ligating the coronary artery of heart. The results showed that miR-148 alleviated myocardial injury and rescued relevant parameters (mean ventricular systolic blood pressure (MAP), left ventricular systolic blood pressure (LVSP), heart rate (HR), creatine kinase-MB (CK-MB), cTn1 and Mb in immature rats with MI/R injury. Besides, miR-148 improved the immune dysfunction induced by MI/R through increasing the number of interleukin (IL)-10+ cells and reducing the number of inducible nitric oxide synthase (iNOS)+ cells. In addition, miR-148 relieved the apoptosis of cardiomyocytes induced by MI/R through inhibiting the expression of Bax and elevating the expression of Bcl-2. Further molecular mechanism indicated that pyruvate dehydrogenase kinase 4 (PDK4) was the downstream target of miR-148, which was further confirmed by dual luciferase reporter assay and related expression detection. Accordingly, silenced PDK4 attenuated cardiac dysfunction, immune disorder and myocardial apoptosis in immature rats and enhanced the ability of antioxidant enzymes. What is more, activated SMAD pathway induced by MI/R injury was then blocked by silenced PDK4. Taken together, our study demonstrated that overexpressed miR-148 relieved cardiac dysfunction, immune disorder and cardiomyocyte apoptosis in immature MI/R rats by PDK4 inhibition, which provided novel targets for MI/R injury treatment.
Collapse
Affiliation(s)
- Qi Yin
- Department of Health care center, Hainan People's Hospital, Haikou, Hainan, China
| | - Ping Wang
- Department of Health care center, Hainan People's Hospital, Haikou, Hainan, China
| | - Xiaohua Wu
- Department of Health care center, Hainan People's Hospital, Haikou, Hainan, China
| |
Collapse
|
33
|
Feng K, Liu Y, Sun J, Zhao C, Duan Y, Wang W, Yan K, Yan X, Sun H, Hu Y, Han J. Compound Danshen Dripping Pill inhibits doxorubicin or isoproterenol-induced cardiotoxicity. Biomed Pharmacother 2021; 138:111531. [PMID: 34311530 DOI: 10.1016/j.biopha.2021.111531] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is the advanced heart disease with high morbidity and mortality. Compound DanShen Dripping Pill (CDDP) is a widely used Traditional Chinese Medicine for cardiovascular disease treatment. Herein, we investigated if CDDP can protect mice against doxorubicin (DOX) or isoprenaline (ISO)-induced HF. After 3 days feeding of normal chow containing CDDP, mice were started DOX or ISO treatment for 4 weeks or 18 days. At the end of treatment, mice were conducted electrocardiogram and echocardiographic test. Blood and heart samples were determined biochemical parameters, myocardial structure and expression of the related molecules. CDDP normalized DOX/ISO-induced heart weight changes, HF parameters and fibrogenesis. The DOX/ISO-impaired left ventricular ejection fraction and fractional shortening were restored by CDDP. Mechanistically, CDDP blocked DOX/ISO-inhibited expression of antioxidant enzymes and DOX/ISO-induced expression of pro-fibrotic molecules, inflammation and cell apoptosis. Additional DOX/ISO-impaired targets in cardiac function but protected by CDDP were identified by RNAseq, qRT-PCR and Western blot. In addition, CDDP protected cardiomyocytes against oxygen-glucose deprivation-induced injuries. Taken together, our study shows that CDDP can protect against myocardial injuries in different models, suggesting its potential application for HF treatment.
Collapse
Affiliation(s)
- Ke Feng
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yuxin Liu
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Jia Sun
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Chunlai Zhao
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, China
| | - Wenjia Wang
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Kaijing Yan
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China; The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China; Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| | - Xijun Yan
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China; The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China; Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| | - He Sun
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China; The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China; Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| | - Yunhui Hu
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China.
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.
| |
Collapse
|
34
|
Cluntun AA, Badolia R, Lettlova S, Parnell KM, Shankar TS, Diakos NA, Olson KA, Taleb I, Tatum SM, Berg JA, Cunningham CN, Van Ry T, Bott AJ, Krokidi AT, Fogarty S, Skedros S, Swiatek WI, Yu X, Luo B, Merx S, Navankasattusas S, Cox JE, Ducker GS, Holland WL, McKellar SH, Rutter J, Drakos SG. The pyruvate-lactate axis modulates cardiac hypertrophy and heart failure. Cell Metab 2021; 33:629-648.e10. [PMID: 33333007 PMCID: PMC7933116 DOI: 10.1016/j.cmet.2020.12.003] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 10/12/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022]
Abstract
The metabolic rewiring of cardiomyocytes is a widely accepted hallmark of heart failure (HF). These metabolic changes include a decrease in mitochondrial pyruvate oxidation and an increased export of lactate. We identify the mitochondrial pyruvate carrier (MPC) and the cellular lactate exporter monocarboxylate transporter 4 (MCT4) as pivotal nodes in this metabolic axis. We observed that cardiac assist device-induced myocardial recovery in chronic HF patients was coincident with increased myocardial expression of the MPC. Moreover, the genetic ablation of the MPC in cultured cardiomyocytes and in adult murine hearts was sufficient to induce hypertrophy and HF. Conversely, MPC overexpression attenuated drug-induced hypertrophy in a cell-autonomous manner. We also introduced a novel, highly potent MCT4 inhibitor that mitigated hypertrophy in cultured cardiomyocytes and in mice. Together, we find that alteration of the pyruvate-lactate axis is a fundamental and early feature of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Ahmad A Cluntun
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Rachit Badolia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sandra Lettlova
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - K Mark Parnell
- Vettore Biosciences, 1700 Owens Street Suite 515, San Francisco, CA 94158, USA
| | - Thirupura S Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Nikolaos A Diakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Kristofor A Olson
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Iosif Taleb
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Jordan A Berg
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Corey N Cunningham
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Tyler Van Ry
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA; Metabolomics, Proteomics and Mass Spectrometry Core Facility, University of Utah, Salt Lake City, UT 84112, USA
| | - Alex J Bott
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Aspasia Thodou Krokidi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sarah Fogarty
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA; Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Sophia Skedros
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Wojciech I Swiatek
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Xuejing Yu
- University of Utah, School of Medicine, Salt Lake City, UT 84132, USA; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, UT 84132, USA
| | - Bai Luo
- Drug Discovery Core Facility, University of Utah, Salt Lake City, UT 84112, USA
| | - Shannon Merx
- Vettore Biosciences, 1700 Owens Street Suite 515, San Francisco, CA 94158, USA
| | - Sutip Navankasattusas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA; Metabolomics, Proteomics and Mass Spectrometry Core Facility, University of Utah, Salt Lake City, UT 84112, USA
| | - Gregory S Ducker
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - William L Holland
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Stephen H McKellar
- University of Utah, School of Medicine, Salt Lake City, UT 84132, USA; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, UT 84132, USA; U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake VA (Veterans Affairs) Health Care System, Salt Lake City, UT, USA
| | - Jared Rutter
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA; Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA; U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake VA (Veterans Affairs) Health Care System, Salt Lake City, UT, USA.
| |
Collapse
|
35
|
Cortassa S, Juhaszova M, Aon MA, Zorov DB, Sollott SJ. Mitochondrial Ca 2+, redox environment and ROS emission in heart failure: Two sides of the same coin? J Mol Cell Cardiol 2021; 151:113-125. [PMID: 33301801 PMCID: PMC7880885 DOI: 10.1016/j.yjmcc.2020.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 11/05/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022]
Abstract
Heart failure (HF) is a progressive, debilitating condition characterized, in part, by altered ionic equilibria, increased ROS production and impaired cellular energy metabolism, contributing to variable profiles of systolic and diastolic dysfunction with significant functional limitations and risk of premature death. We summarize current knowledge concerning changes of intracellular Na+ and Ca2+ control mechanisms during the disease progression and their consequences on mitochondrial Ca2+ homeostasis and the shift in redox balance. Absent existing biological data, our computational modeling studies advance a new 'in silico' analysis to reconcile existing opposing views, based on different experimental HF models, regarding variations in mitochondrial Ca2+ concentration that participate in triggering and perpetuating oxidative stress in the failing heart and their impact on cardiac energetics. In agreement with our hypothesis and the literature, model simulations demonstrate the possibility that the heart's redox status together with cytoplasmic Na+ concentrations act as regulators of mitochondrial Ca2+ levels in HF and of the bioenergetics response that will ultimately drive ATP supply and oxidative stress. The resulting model predictions propose future directions to study the evolution of HF as well as other types of heart disease, and to develop novel testable mechanistic hypotheses that may lead to improved therapeutics.
Collapse
Affiliation(s)
- Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States.
| | - Magdalena Juhaszova
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States.
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States; Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, United States.
| | - Dmitry B Zorov
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States.
| |
Collapse
|
36
|
Alam S, Abdullah CS, Aishwarya R, Morshed M, Nitu SS, Miriyala S, Panchatcharam M, Kevil CG, Orr AW, Bhuiyan MS. Dysfunctional Mitochondrial Dynamic and Oxidative Phosphorylation Precedes Cardiac Dysfunction in R120G-αB-Crystallin-Induced Desmin-Related Cardiomyopathy. J Am Heart Assoc 2020; 9:e017195. [PMID: 33208022 PMCID: PMC7763772 DOI: 10.1161/jaha.120.017195] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Background The mutated α-B-Crystallin (CryABR120G) mouse model of desmin-related myopathy (DRM) shows an age-dependent onset of pathologic cardiac remodeling and progression of heart failure. CryABR120G expression in cardiomyocytes affects the mitochondrial spatial organization within the myofibrils, but the molecular perturbation within the mitochondria in the relation of the overall course of the proteotoxic disease remains unclear. Methods and Results CryABR120G mice show an accumulation of electron-dense aggregates and myofibrillar degeneration associated with the development of cardiac dysfunction. Though extensive studies demonstrated that these altered ultrastructural changes cause cardiac contractility impairment, the molecular mechanism of cardiomyocyte death remains elusive. Here, we explore early pathological processes within the mitochondria contributing to the contractile dysfunction and determine the pathogenic basis for the heart failure observed in the CryABR120G mice. In the present study, we report that the CryABR120G mice transgenic hearts undergo altered mitochondrial dynamics associated with increased level of dynamin-related protein 1 and decreased level of optic atrophy type 1 as well as mitofusin 1 over the disease process. In association with these changes, an altered level of the components of mitochondrial oxidative phosphorylation and pyruvate dehydrogenase complex regulatory proteins occurs before the manifestation of pathologic adverse remodeling in the CryABR120G hearts. Mitochondria isolated from CryABR120G transgenic hearts without visible pathology show decreased electron transport chain complex activities and mitochondrial respiration. Taken together, we demonstrated the involvement of mitochondria in the pathologic remodeling and progression of DRM-associated cellular dysfunction. Conclusions Mitochondrial dysfunction in the form of altered mitochondrial dynamics, oxidative phosphorylation and pyruvate dehydrogenase complex proteins level, abnormal electron transport chain complex activities, and mitochondrial respiration are evident on the CryABR120G hearts before the onset of detectable pathologies and development of cardiac contractile dysfunction.
Collapse
Affiliation(s)
- Shafiul Alam
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Chowdhury S. Abdullah
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Richa Aishwarya
- Department of Molecular and Cellular PhysiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Mahboob Morshed
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Sadia S. Nitu
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Sumitra Miriyala
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences CenterShreveportLA
| | - Manikandan Panchatcharam
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences CenterShreveportLA
| | - Christopher G. Kevil
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
- Department of Molecular and Cellular PhysiologyLouisiana State University Health Sciences CenterShreveportLA
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences CenterShreveportLA
| | - A. Wayne Orr
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
- Department of Molecular and Cellular PhysiologyLouisiana State University Health Sciences CenterShreveportLA
- Department of Cellular Biology and AnatomyLouisiana State University Health Sciences CenterShreveportLA
| | - Md. Shenuarin Bhuiyan
- Department of Pathology and Translational PathobiologyLouisiana State University Health Sciences CenterShreveportLA
- Department of Molecular and Cellular PhysiologyLouisiana State University Health Sciences CenterShreveportLA
| |
Collapse
|
37
|
Ma Z, Shen Z, Gong Y, Zhou J, Chen X, Lv Q, Wang M, Chen J, Yu M, Fu G, He H, Lai D. Weighted gene co-expression network analysis identified underlying hub genes and mechanisms in the occurrence and development of viral myocarditis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1348. [PMID: 33313093 PMCID: PMC7723587 DOI: 10.21037/atm-20-3337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Myocarditis is an inflammatory myocardial disease, which may lead to heart failure and sudden death. Despite extensive research into the pathogenesis of myocarditis, effective treatments for this condition remain elusive. This study aimed to explore the potential pathogenesis and hub genes for viral myocarditis. Methods A weighted gene co-expression network analysis (WGCNA) was performed based on the gene expression profiles derived from mouse models at different stages of viral myocarditis (GSE35182). Functional annotation was executed within the key modules. Potential hub genes were predicted based on the intramodular connectivity (IC). Finally, potential microRNAs that regulate gene expression were predicted by miRNet analysis. Results Three gene co-expression modules showed the strongest correlation with the acute or chronic disease stage. A significant positive correlation was detected between the acute disease stage and the turquoise module, the genes of which were mainly enriched in antiviral response and immune-inflammatory activation. Furthermore, a significant positive correlation and a negative correlation were identified between the chronic disease stage and the brown and yellow modules, respectively. These modules were mainly associated with the cytoskeleton, phosphorylation, cellular catabolic process, and autophagy. Subsequently, we predicted the underlying hub genes and microRNAs in the three modules. Conclusions This study revealed the main biological processes in different stages of viral myocarditis and predicted hub genes in both the acute and chronic disease stages. Our results may be helpful for developing new therapeutic targets for viral myocarditis in future research.
Collapse
Affiliation(s)
- Zetao Ma
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhida Shen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingchao Gong
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoou Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Lv
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiawen Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mei Yu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong He
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongwu Lai
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
38
|
McCommis KS, Kovacs A, Weinheimer CJ, Shew TM, Koves TR, Ilkayeva OR, Kamm DR, Pyles KD, King MT, Veech RL, DeBosch BJ, Muoio DM, Gross RW, Finck BN. Nutritional modulation of heart failure in mitochondrial pyruvate carrier-deficient mice. Nat Metab 2020; 2:1232-1247. [PMID: 33106690 PMCID: PMC7957960 DOI: 10.1038/s42255-020-00296-1] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/10/2020] [Indexed: 01/04/2023]
Abstract
The myocardium is metabolically flexible; however, impaired flexibility is associated with cardiac dysfunction in conditions including diabetes and heart failure. The mitochondrial pyruvate carrier (MPC) complex, composed of MPC1 and MPC2, is required for pyruvate import into the mitochondria. Here we show that MPC1 and MPC2 expression is downregulated in failing human and mouse hearts. Mice with cardiac-specific deletion of Mpc2 (CS-MPC2-/-) exhibited normal cardiac size and function at 6 weeks old, but progressively developed cardiac dilation and contractile dysfunction, which was completely reversed by a high-fat, low-carbohydrate ketogenic diet. Diets with higher fat content, but enough carbohydrate to limit ketosis, also improved heart failure, while direct ketone body provisioning provided only minor improvements in cardiac remodelling in CS-MPC2-/- mice. An acute fast also improved cardiac remodelling. Together, our results reveal a critical role for mitochondrial pyruvate use in cardiac function, and highlight the potential of dietary interventions to enhance cardiac fat metabolism to prevent or reverse cardiac dysfunction and remodelling in the setting of MPC deficiency.
Collapse
Affiliation(s)
- Kyle S McCommis
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biochemistry & Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA.
| | - Attila Kovacs
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Carla J Weinheimer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Trevor M Shew
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy R Koves
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dakota R Kamm
- Department of Biochemistry & Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Kelly D Pyles
- Department of Biochemistry & Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - M Todd King
- Laboratory of Metabolic Control, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, USA
| | - Richard L Veech
- Laboratory of Metabolic Control, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, USA
| | - Brian J DeBosch
- Departments of Pediatrics and Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Deborah M Muoio
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Richard W Gross
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Chemistry, Washington University, St. Louis, MO, USA
| | - Brian N Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
39
|
Abstract
The heart requires a high amount of energy, in the form of adenosine triphosphate, to maintain its viability and pump function. Anaerobic glycolysis and mitochondrial oxidative phosphorylation are the two main metabolic pathways by which adenosine triphosphate is generated, using fatty acids, glucose, lactate, and ketone bodies as primary substrates. Previous studies have demonstrated that, in response to stress, the heart undergoes alterations in metabolism, ranging from changes in substrate utilization to mitochondrial function, collectively called metabolic remodeling. However, the molecular mechanism mediating metabolic remodeling in the heart remains unclear. Yes-associated protein 1 (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), which are major downstream effectors of the Hippo signaling pathway, play an important role in the regulation of heart size and cellular homeostasis of cardiomyocytes through the regulation of various transcriptional factors under both physiological and pathophysiological conditions. Recent findings in various organs and cell types have revealed that YAP and TAZ play an important role in energy metabolism. Here, we summarize what is currently known about YAP/TAZ in the regulation of metabolism of various substrates and mitochondrial function in various organs and cell types and discuss the potential role of YAP/TAZ in mediating metabolic remodeling of the heart during stress and heart failure.
Collapse
|