1
|
Zhu S, Chen Z, Liu C, Duong J, Tran T, Liang Z, Fang X, Ouyang K. The essential role of MED27 in stabilizing the mediator complex for cardiac development and function. Life Sci 2024; 356:123020. [PMID: 39209248 DOI: 10.1016/j.lfs.2024.123020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
AIM Transcriptional regulation of gene expression plays a crucial role in orchestrating complex morphogenetic and molecular events during heart development and function. Mediator complex is an essential multi-subunit protein complex that governs gene expression in eukaryotic cells. Although Mediator subunits (MEDs) work integrally in the complex, individual MED component displays specialized functions. MED27, categorized as an Upper Tail subunit, possesses an as-yet-uncharacterized function. In this study, we aimed to investigate the physiological role of MED27 in cardiomyocytes. MATERIALS AND METHODS we generated a Med27 floxed mouse line, which was further used to generate constitutive (cKO) and inducible (icKO) cardiomyocyte-specific Med27 knockout mouse models. Morphological, histological analysis and cardiac physiological studies were performed in Med27 cKO and icKO mutants. Transcriptional profiles were determined by RNA sequencing (RNAseq) analysis. KEY FUNDINGS Ablation of MED27 in developing mouse cardiomyocytes results in embryonic lethality, while its deletion in adult cardiomyocytes leads to heart failure and mortality. Similar to the ablation of another Upper Tail subunit, MED30 in cardiomyocytes, deletion of MED27 leads to decreased protein levels of most MEDs in cardiomyocytes. Interestingly, overexpression of MED30 fails to restore the protein levels of Mediator subunits in MED27-deficient cardiomyocytes, demonstrating that the role of MED27 in maintaining the integrity and stability of the Mediator complex is independent of MED30. SIGNIFICANCE Our results revealed an essential role of MED27 in cardiac development and function by maintaining the stability of the Mediator core.
Collapse
Affiliation(s)
- Siting Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China; Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ze'e Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China; Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Canzhao Liu
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Cardiovascular Medicine, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Janelle Duong
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Tiana Tran
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Zhengyu Liang
- Department of Systems Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, California, USA.
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
2
|
Mao S, Song C, Huang H, Nie Y, Ding K, Cui J, Tian J, Tang H. Role of transcriptional cofactors in cardiovascular diseases. Biochem Biophys Res Commun 2024; 706:149757. [PMID: 38490050 DOI: 10.1016/j.bbrc.2024.149757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/16/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
Cardiovascular disease is a main cause of mortality in the world and the highest incidence of all diseases. However, the mechanism of the pathogenesis of cardiovascular disease is still unclear, and we need to continue to explore its mechanism of action. The occurrence and development of cardiovascular disease is significantly associated with genetic abnormalities, and gene expression is affected by transcriptional regulation. In this complex process, the protein-protein interaction promotes the RNA polymerase II to the initiation site. And in this process of transcriptional regulation, transcriptional cofactors are responsible for passing cues from enhancers to promoters and promoting the binding of RNA polymerases to promoters, so transcription cofactors playing a key role in gene expression regulation. There is growing evidence that transcriptional cofactors play a critical role in cardiovascular disease. Transcriptional cofactors can promote or inhibit transcription by affecting the function of transcription factors. It can affect the initiation and elongation process of transcription by forming complexes with transcription factors, which are important for the stabilization of DNA rings. It can also act as a protein that interacts with other proteins to affect the expression of other genes. Therefore, the aim of this overview is to summarize the effect of some transcriptional cofactors such as BRD4, EP300, MED1, EZH2, YAP, SIRT6 in cardiovascular disease and to provide a promising therapeutic strategy for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Shuqing Mao
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Chao Song
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hong Huang
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yali Nie
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Kai Ding
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jian Cui
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Huifang Tang
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
3
|
Luo H, Lao L, Au KS, Northrup H, He X, Forget D, Gauthier MS, Coulombe B, Bourdeau I, Shi W, Gagliardi L, Fragoso MCBV, Peng J, Wu J. ARMC5 controls the degradation of most Pol II subunits, and ARMC5 mutation increases neural tube defect risks in mice and humans. Genome Biol 2024; 25:19. [PMID: 38225631 PMCID: PMC10789052 DOI: 10.1186/s13059-023-03147-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Neural tube defects (NTDs) are caused by genetic and environmental factors. ARMC5 is part of a novel ubiquitin ligase specific for POLR2A, the largest subunit of RNA polymerase II (Pol II). RESULTS We find that ARMC5 knockout mice have increased incidence of NTDs, such as spina bifida and exencephaly. Surprisingly, the absence of ARMC5 causes the accumulation of not only POLR2A but also most of the other 11 Pol II subunits, indicating that the degradation of the whole Pol II complex is compromised. The enlarged Pol II pool does not lead to generalized Pol II stalling or a generalized decrease in mRNA transcription. In neural progenitor cells, ARMC5 knockout only dysregulates 106 genes, some of which are known to be involved in neural tube development. FOLH1, critical in folate uptake and hence neural tube development, is downregulated in the knockout intestine. We also identify nine deleterious mutations in the ARMC5 gene in 511 patients with myelomeningocele, a severe form of spina bifida. These mutations impair the interaction between ARMC5 and Pol II and reduce Pol II ubiquitination. CONCLUSIONS Mutations in ARMC5 increase the risk of NTDs in mice and humans. ARMC5 is part of an E3 controlling the degradation of all 12 subunits of Pol II under physiological conditions. The Pol II pool size might have effects on NTD pathogenesis, and some of the effects might be via the downregulation of FOLH1. Additional mechanistic work is needed to establish the causal effect of the findings on NTD pathogenesis.
Collapse
Affiliation(s)
- Hongyu Luo
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.
| | - Linjiang Lao
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Kit Sing Au
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth) and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Hope Northrup
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth) and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Xiao He
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Diane Forget
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
| | - Marie-Soleil Gauthier
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
| | - Benoit Coulombe
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
| | - Isabelle Bourdeau
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
- Division of Endocrinology, CHUM, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Wei Shi
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Lucia Gagliardi
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, Australia
- Endocrine and Diabetes Unit, Queen Elizabeth Hospital, Adelaide, Australia
| | - Maria Candida Barisson Villares Fragoso
- Unidade de Suprarrenal Disciplina de Endocrinologia E Metabologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Junzheng Peng
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Jiangping Wu
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.
- Division of Nephrology, CHUM, Montreal, QC, Canada.
| |
Collapse
|
4
|
Yu W, Xie Z, Li J, Lin J, Su Z, Che Y, Ye F, Zhang Z, Xu P, Zeng Y, Xu X, Li Z, Feng P, Mi R, Wu Y, Shen H. Super enhancers targeting ZBTB16 in osteogenesis protect against osteoporosis. Bone Res 2023; 11:30. [PMID: 37280207 DOI: 10.1038/s41413-023-00267-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 06/08/2023] Open
Abstract
As the major cell precursors in osteogenesis, mesenchymal stem cells (MSCs) are indispensable for bone homeostasis and development. However, the primary mechanisms regulating osteogenic differentiation are controversial. Composed of multiple constituent enhancers, super enhancers (SEs) are powerful cis-regulatory elements that identify genes that ensure sequential differentiation. The present study demonstrated that SEs were indispensable for MSC osteogenesis and involved in osteoporosis development. Through integrated analysis, we identified the most common SE-targeted and osteoporosis-related osteogenic gene, ZBTB16. ZBTB16, positively regulated by SEs, promoted MSC osteogenesis but was expressed at lower levels in osteoporosis. Mechanistically, SEs recruited bromodomain containing 4 (BRD4) at the site of ZBTB16, which then bound to RNA polymerase II-associated protein 2 (RPAP2) that transported RNA polymerase II (POL II) into the nucleus. The subsequent synergistic regulation of POL II carboxyterminal domain (CTD) phosphorylation by BRD4 and RPAP2 initiated ZBTB16 transcriptional elongation, which facilitated MSC osteogenesis via the key osteogenic transcription factor SP7. Bone-targeting ZBTB16 overexpression had a therapeutic effect on the decreased bone density and remodeling capacity of Brd4fl/fl Prx1-cre mice and osteoporosis (OP) models. Therefore, our study shows that SEs orchestrate the osteogenesis of MSCs by targeting ZBTB16 expression, which provides an attractive focus and therapeutic target for osteoporosis. Without SEs located on osteogenic genes, BRD4 is not able to bind to osteogenic identity genes due to its closed structure before osteogenesis. During osteogenesis, histones on osteogenic identity genes are acetylated, and OB-gain SEs appear, enabling the binding of BRD4 to the osteogenic identity gene ZBTB16. RPAP2 transports RNA Pol II from the cytoplasm to the nucleus and guides Pol II to target ZBTB16 via recognition of the navigator BRD4 on SEs. After the binding of the RPAP2-Pol II complex to BRD4 on SEs, RPAP2 dephosphorylates Ser5 at the Pol II CTD to terminate the transcriptional pause, and BRD4 phosphorylates Ser2 at the Pol II CTD to initiate transcriptional elongation, which synergistically drives efficient transcription of ZBTB16, ensuring proper osteogenesis. Dysregulation of SE-mediated ZBTB16 expression leads to osteoporosis, and bone-targeting ZBTB16 overexpression is efficient in accelerating bone repair and treating osteoporosis.
Collapse
Affiliation(s)
- Wenhui Yu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
- Shenzhen Key Laboratory of Ankylosing Spondylitis, Shenzhen, 518003, PR China
| | - Jinteng Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Jiajie Lin
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Zepeng Su
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Yunshu Che
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Feng Ye
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Zhaoqiang Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Peitao Xu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Yipeng Zeng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Xiaojun Xu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Zhikun Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Pei Feng
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Rujia Mi
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China
| | - Yanfeng Wu
- Shenzhen Key Laboratory of Ankylosing Spondylitis, Shenzhen, 518003, PR China.
- Center for Biotherapy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China.
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, PR China.
- Shenzhen Key Laboratory of Ankylosing Spondylitis, Shenzhen, 518003, PR China.
| |
Collapse
|
5
|
Ilchuk LA, Kubekina MV, Okulova YD, Silaeva YY, Tatarskiy VV, Filatov MA, Bruter AV. Genetically Engineered Mice Unveil In Vivo Roles of the Mediator Complex. Int J Mol Sci 2023; 24:ijms24119330. [PMID: 37298278 DOI: 10.3390/ijms24119330] [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: 04/28/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
The Mediator complex is a multi-subunit protein complex which plays a significant role in the regulation of eukaryotic gene transcription. It provides a platform for the interaction of transcriptional factors and RNA polymerase II, thus coupling external and internal stimuli with transcriptional programs. Molecular mechanisms underlying Mediator functioning are intensively studied, although most often using simple models such as tumor cell lines and yeast. Transgenic mouse models are required to study the role of Mediator components in physiological processes, disease, and development. As constitutive knockouts of most of the Mediator protein coding genes are embryonically lethal, conditional knockouts and corresponding activator strains are needed for these studies. Recently, they have become more easily available with the development of modern genetic engineering techniques. Here, we review existing mouse models for studying the Mediator, and data obtained in corresponding experiments.
Collapse
Affiliation(s)
- Leonid A Ilchuk
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Marina V Kubekina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Yulia D Okulova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Yulia Yu Silaeva
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Maxim A Filatov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Alexandra V Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology", Ministry of Health of the Russian Federation, Kashirskoe Sh. 24, 115478 Moscow, Russia
| |
Collapse
|
6
|
Wei R, Guo S, Meng Z, Li Z, Liu J, Hu L, Sui L. Mediator1 involved in functional integration of Smad3 and Notch1 promoting enamel mineralization. Biochem Biophys Res Commun 2023; 663:47-53. [PMID: 37119765 DOI: 10.1016/j.bbrc.2023.04.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/27/2023] [Accepted: 04/17/2023] [Indexed: 05/01/2023]
Abstract
Enamel hypoplasia is a tooth development defection due to the disruption of enamel matrix mineralization, manifesting as chalky white phenotype. Multiple genes may be involved in this tooth agenesis. It has been proved that ablation of coactivator Mediator1 (Med1) switches the cell fate of dental epithelia, resulting in abnormal tooth development via Notch1 signaling. Smad3 (-/-) mice displays the similar chalky white incisors. However, the expression of Smad3 in Med1 ablation mice and the impact of Med1 on functional integration between Smad3 and Notch1 remains unclear. Cre-loxP-based C57/BL6 mice with epithelial-specific Med1 knockout (Med1 KO) backgrounds were generated. Mandibles and dental epithelial stem cells (DE-SCs) from incisors cervical loop (CL) were isolated from wild-type (CON) mice and Med1 KO mice. Transcriptome sequencing was used to analyze the differences of CL tissue between KO and CON mice. The results revealed the enrichment of TGF-β signaling pathway. qRT-PCR and western blot were performed to show the gene and protein expression of Smad3, pSmad3, Notch1 and NICD, the key regulators of TGF-β and Notch1 signaling pathway. Expression of Notch1 and Smad3 was confirmed to be down-regulated in Med1 KO cells. Using activators of Smad3 and Notch1 on Med1 KO cells, both pSmad3 and NICD were rescued. Moreover, adding inhibitors and activators of Smad3 and Notch1 to cells of CON groups respectively, the protein expressions of Smad3, pSmad3, Notch1 and NICD were synergistically affected. In summary, Med1 participates in the functional integration of Smad3 and Notch1, thus promoting enamel mineralization.
Collapse
Affiliation(s)
- Ran Wei
- School of Stomatology, Tianjin Medical University, Tianjin, 300014, China.
| | - Shuling Guo
- School of Stomatology, Tianjin Medical University, Tianjin, 300014, China.
| | - Zhaosong Meng
- School of Stomatology, Tianjin Medical University, Tianjin, 300014, China.
| | - Zhe Li
- School of Stomatology, Tianjin Medical University, Tianjin, 300014, China.
| | - Jiacheng Liu
- School of Stomatology, Tianjin Medical University, Tianjin, 300014, China.
| | - Lizhi Hu
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Tianjin Medical University, Tianjin, 300014, China.
| | - Lei Sui
- School of Stomatology, Tianjin Medical University, Tianjin, 300014, China.
| |
Collapse
|
7
|
Kraus L, Beavens B. The Current Therapeutic Role of Chromatin Remodeling for the Prognosis and Treatment of Heart Failure. Biomedicines 2023; 11:biomedicines11020579. [PMID: 36831115 PMCID: PMC9953583 DOI: 10.3390/biomedicines11020579] [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: 01/24/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Cardiovascular diseases are a major cause of death globally, with no cure to date. Many interventions have been studied and suggested, of which epigenetics and chromatin remodeling have been the most promising. Over the last decade, major advancements have been made in the field of chromatin remodeling, particularly for the treatment of heart failure, because of innovations in bioinformatics and gene therapy. Specifically, understanding changes to the chromatin architecture have been shown to alter cardiac disease progression via variations in genomic sequencing, targeting cardiac genes, using RNA molecules, and utilizing chromatin remodeler complexes. By understanding these chromatin remodeling mechanisms in an injured heart, treatments for heart failure have been suggested through individualized pharmaceutical interventions as well as biomarkers for major disease states. By understanding the current roles of chromatin remodeling in heart failure, a potential therapeutic approach may be discovered in the future.
Collapse
|
8
|
Fatima M, Gao J, Han T, Ding Y, Zhang Y, Wen E, Jia L, Wang R, Wang W, Zhao S, Bai L, Liu E. MED1 Deficiency in Macrophages Aggravates Isoproterenol-Induced Cardiac Fibrosis in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1016-1027. [PMID: 35461855 DOI: 10.1016/j.ajpath.2022.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/24/2022] [Accepted: 03/31/2022] [Indexed: 06/14/2023]
Abstract
Mediator 1 (MED1), a key subunit of the mediator complex, interacts with various nuclear receptors and functions in lipid metabolism and energy homeostasis. Dilated cardiomyopathy-related ventricular dilatation and heart failure have been reported in mice with cardiomyocyte-specific Med1 deficiency. However, the contribution of macrophage-specific MED1 in cardiac remodeling remains unclear. In this study, macrophage-specific Med1 knockout (Med1ΔMac) mice were generated and exposed to isoproterenol (ISO) to induce cardiac fibrosis; these mice showed aggravated cardiac fibrosis compared with Med1fl/fl mice. The levels of expression of marker genes for myofibroblast transdifferentiation [α-smooth muscle actin (SMA)] and of profibrotic genes, including Col1a1, Col3a1, Postn, Mmp2, Timp1, and Fn1, were significantly increased in the cardiac tissues of Med1ΔMac mice with ISO-induced myocardial fibrosis. In particular, the transforming growth factor (TGF)-β-Smad2/3 signaling pathway was activated. In bone marrow-derived and peritoneal macrophages, Med1 deficiency was also associated with elevated levels of expression of proinflammatory genes, including Il6, Tnfa, and Il1b. These findings indicate that macrophage-specific MED1 deficiency may aggravate ISO-induced cardiac fibrosis via the regulation of the TGF-β-SMAD2/3 pathway, and the underlying mechanism may involve MED1 deficiency triggering the activation of inflammatory cytokines in macrophages, which in turn may stimulate phenotypic switch of cardiac fibroblasts and accelerate cardiac fibrosis. Thus, MED1 is a potential therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Mehreen Fatima
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Jie Gao
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Tuo Han
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yiming Ding
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yali Zhang
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Ergang Wen
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Linying Jia
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Rong Wang
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Weirong Wang
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Sihai Zhao
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Liang Bai
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.
| | - Enqi Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
9
|
Predictive Biomarkers for Postmyocardial Infarction Heart Failure Using Machine Learning: A Secondary Analysis of a Cohort Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2903543. [PMID: 34938340 PMCID: PMC8687817 DOI: 10.1155/2021/2903543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022]
Abstract
Background There are few biomarkers with an excellent predictive value for postacute myocardial infarction (MI) patients who developed heart failure (HF). This study aimed to screen candidate biomarkers to predict post-MI HF. Methods This is a secondary analysis of a single-center cohort study including nine post-MI HF patients and eight post-MI patients who remained HF-free over a 6-month follow-up. Transcriptional profiling was analyzed using the whole blood samples collected at admission, discharge, and 1-month follow-up. We screened differentially expressed genes and identified key modules using weighted gene coexpression network analysis. We confirmed the candidate biomarkers using the developed external datasets on post-MI HF. The receiver operating characteristic curves were created to evaluate the predictive value of these candidate biomarkers. Results A total of 6,778, 1,136, and 1,974 genes (dataset 1) were differently expressed at admission, discharge, and 1-month follow-up, respectively. The white and royal blue modules were most significantly correlated with post-MI HF (dataset 2). After overlapping dataset 1, dataset 2, and external datasets (dataset 3), we identified five candidate biomarkers, including FCGR2A, GSDMB, MIR330, MED1, and SQSTM1. When GSDMB and SQSTM1 were combined, the area under the curve achieved 1.00, 0.85, and 0.89 in admission, discharge, and 1-month follow-up, respectively. Conclusions This study demonstrates that FCGR2A, GSDMB, MIR330, MED1, and SQSTM1 are the candidate predictive biomarker genes for post-MI HF, and the combination of GSDMB and SQSTM1 has a high predictive value.
Collapse
|
10
|
Funamoto M, Sunagawa Y, Katanasaka Y, Shimizu K, Miyazaki Y, Sari N, Shimizu S, Mori K, Wada H, Hasegawa K, Morimoto T. Histone Acetylation Domains Are Differentially Induced during Development of Heart Failure in Dahl Salt-Sensitive Rats. Int J Mol Sci 2021; 22:1771. [PMID: 33578969 PMCID: PMC7916721 DOI: 10.3390/ijms22041771] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/06/2021] [Accepted: 02/07/2021] [Indexed: 12/21/2022] Open
Abstract
Histone acetylation by epigenetic regulators has been shown to activate the transcription of hypertrophic response genes, which subsequently leads to the development and progression of heart failure. However, nothing is known about the acetylation of the histone tail and globular domains in left ventricular hypertrophy or in heart failure. The acetylation of H3K9 on the promoter of the hypertrophic response gene was significantly increased in the left ventricular hypertrophy stage, whereas the acetylation of H3K122 did not increase in the left ventricular hypertrophy stage but did significantly increase in the heart failure stage. Interestingly, the interaction between the chromatin remodeling factor BRG1 and p300 was significantly increased in the heart failure stage, but not in the left ventricular hypertrophy stage. This study demonstrates that stage-specific acetylation of the histone tail and globular domains occurs during the development and progression of heart failure, providing novel insights into the epigenetic regulatory mechanism governing transcriptional activity in these processes.
Collapse
Affiliation(s)
- Masafumi Funamoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| | - Yasufumi Katanasaka
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| | - Kana Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Yusuke Miyazaki
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| | - Nurmila Sari
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
| | - Satoshi Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Kiyoshi Mori
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| | - Hiromichi Wada
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Koji Hasegawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| |
Collapse
|
11
|
Liu CF, Abnousi A, Bazeley P, Ni Y, Morley M, Moravec CS, Hu M, Tang WHW. Global analysis of histone modifications and long-range chromatin interactions revealed the differential cistrome changes and novel transcriptional players in human dilated cardiomyopathy. J Mol Cell Cardiol 2020; 145:30-42. [PMID: 32533974 DOI: 10.1016/j.yjmcc.2020.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 05/18/2020] [Accepted: 06/02/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Acetylation and methylation of histones alter the chromatin structure and accessibility that affect transcriptional regulators binding to enhancers and promoters. The binding of transcriptional regulators enables the interaction between enhancers and promoters, thus affecting gene expression. However, our knowledge of these epigenetic alternations in patients with heart failure remains limited. METHODS AND RESULTS From the comprehensive analysis of major histone modifications, 3-dimensional chromatin interactions, and transcriptome in left ventricular (LV) tissues from dilated cardiomyopathy (DCM) patients and non-heart failure (NF) donors, differential active enhancer and promoter regions were identified between NF and DCM. Moreover, the genome-wide average promoter signal is significantly lower in DCM than in NF. Super-enhancer (SE) analysis revealed that fewer SEs were found in DCM LVs than in NF ones, and three unique SE-associated genes between NF and DCM were identified. Moreover, SEs are enriched within the genomic region associated with long-range chromatin interactions. The differential enhancer-promoter interactions were observed in the known heart failure gene loci and are correlated with the gene expression levels. Motif analysis identified known cardiac factors and possible novel players for DCM. CONCLUSIONS We have established the cistrome of four histone modifications and chromatin interactome for enhancers and promoters in NF and DCM tissues. Differential histone modifications and enhancer-promoter interactions were found in DCM, which were associated with gene expression levels of a subset of disease-associated genes in human heart failure.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, USA
| | - Armen Abnousi
- Quantitative Health Sciences, Lerner Research Institute, USA
| | - Peter Bazeley
- Quantitative Health Sciences, Lerner Research Institute, USA
| | - Ying Ni
- Taussig Cancer Institute, Heart and Vascular Institute, Cleveland Clinic, OH, USA
| | | | - Christine S Moravec
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, USA
| | - Ming Hu
- Quantitative Health Sciences, Lerner Research Institute, USA
| | - W H Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, USA; Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH, USA.
| |
Collapse
|
12
|
Abstract
Pathological cardiac remodeling is induced through multiple mechanisms that include neurohumoral and biomechanical stress resulting in transcriptional alterations that ultimately become maladaptive and lead to the development of heart failure (HF). Although cardiac transcriptional remodeling is mediated by the activation of numerous signaling pathways that converge on a limited number of transcription factors (TFs) that promote hypertrophy (pro-hypertrophic TFs), the current therapeutic approach to prevent HF utilizes pharmacological inhibitors that largely target specific receptors that are activated in response to pathological stimuli. Thus, there is limited efficacy with the current pharmacological approaches to inhibit transcriptional remodeling associated with the development of HF. Recent evidence suggests that these pro-hypertrophic TFs co-localize at enhancers to cooperatively activate transcription associated with pathological cardiac remodeling. In disease states, including cancer and HF, evidence suggests that the general transcriptional machinery is disproportionately bound at enhancers. Therefore, pharmacological inhibition of transcriptional machinery that integrates pro-hypertrophic TFs may represent a promising alternative therapeutic approach to limit pathological remodeling associated with the development of HF.
Collapse
|
13
|
Ponce JM, Coen G, Spitler KM, Dragisic N, Martins I, Hinton A, Mungai M, Tadinada SM, Zhang H, Oudit GY, Song L, Li N, Sicinski P, Strack S, Abel ED, Mitchell C, Hall DD, Grueter CE. Stress-Induced Cyclin C Translocation Regulates Cardiac Mitochondrial Dynamics. J Am Heart Assoc 2020; 9:e014366. [PMID: 32248761 PMCID: PMC7428645 DOI: 10.1161/jaha.119.014366] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/24/2020] [Indexed: 12/14/2022]
Abstract
Background Nuclear-to-mitochondrial communication regulating gene expression and mitochondrial function is a critical process following cardiac ischemic injury. In this study, we determined that cyclin C, a component of the Mediator complex, regulates cardiac and mitochondrial function in part by modifying mitochondrial fission. We tested the hypothesis that cyclin C functions as a transcriptional cofactor in the nucleus and a signaling molecule stimulating mitochondrial fission in response to stimuli such as cardiac ischemia. Methods and Results We utilized gain- and loss-of-function mouse models in which the CCNC (cyclin C) gene was constitutively expressed (transgenic, CycC cTg) or deleted (knockout, CycC cKO) in cardiomyocytes. The knockout and transgenic mice exhibited decreased cardiac function and altered mitochondria morphology. The hearts of knockout mice had enlarged mitochondria with increased length and area, whereas mitochondria from the hearts of transgenic mice were significantly smaller, demonstrating a role for cyclin C in regulating mitochondrial dynamics in vivo. Hearts from knockout mice displayed altered gene transcription and metabolic function, suggesting that cyclin C is essential for maintaining normal cardiac function. In vitro and in vivo studies revealed that cyclin C translocates to the cytoplasm, enhancing mitochondria fission following stress. We demonstrated that cyclin C interacts with Cdk1 (cyclin-dependent kinase 1) in vivo following ischemia/reperfusion injury and that, consequently, pretreatment with a Cdk1 inhibitor results in reduced mitochondrial fission. This finding suggests a potential therapeutic target to regulate mitochondrial dynamics in response to stress. Conclusions Our study revealed that cyclin C acts as a nuclear-to-mitochondrial signaling factor that regulates both cardiac hypertrophic gene expression and mitochondrial fission. This finding provides new insights into the regulation of cardiac energy metabolism following acute ischemic injury.
Collapse
MESH Headings
- Animals
- CDC2 Protein Kinase/antagonists & inhibitors
- CDC2 Protein Kinase/metabolism
- Cells, Cultured
- Cyclin C/deficiency
- Cyclin C/genetics
- Cyclin C/metabolism
- Disease Models, Animal
- Energy Metabolism/drug effects
- Humans
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondrial Dynamics/drug effects
- Myocardial Reperfusion Injury/genetics
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Protein Kinase Inhibitors/pharmacology
- Protein Transport
- Rats, Wistar
- Signal Transduction
Collapse
Affiliation(s)
- Jessica M. Ponce
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
- Interdisciplinary Graduate Program in GeneticsUniversity of IowaIowa CityIA
| | - Grace Coen
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
| | - Kathryn M. Spitler
- Department of BiochemistryCarver College of MedicineUniversity of IowaIowa CityIA
| | - Nikola Dragisic
- Stead Family Department of PediatricsUniversity of IowaIowa CityIA
| | - Ines Martins
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
| | - Antentor Hinton
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
- Fraternal Order of Eagles Diabetes Research CenterDivision of Endocrinology and MetabolismCarver College of MedicineUniversity of IowaIowa CityIA
| | - Margaret Mungai
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
- Fraternal Order of Eagles Diabetes Research CenterDivision of Endocrinology and MetabolismCarver College of MedicineUniversity of IowaIowa CityIA
| | - Satya Murthy Tadinada
- Department of Pharmacology and Iowa Neuroscience InstituteCarver College of MedicineUniversity of IowaIowa CityIA
| | - Hao Zhang
- Mazankowski Alberta Heart Institute Canada Research Chair in Heart FailureDivision of Cardiology2C2 Walter Mackenzie Health Sciences Centre EdmontonAlbertaCanada
| | - Gavin Y. Oudit
- Mazankowski Alberta Heart Institute Canada Research Chair in Heart FailureDivision of Cardiology2C2 Walter Mackenzie Health Sciences Centre EdmontonAlbertaCanada
| | - Long‐Sheng Song
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
- Fraternal Order of Eagles Diabetes Research CenterDivision of Endocrinology and MetabolismCarver College of MedicineUniversity of IowaIowa CityIA
- Iowa City Veterans Affairs Medical CenterIowa CityIA
| | - Na Li
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA
- Department of GeneticsHarvard Medical SchoolBostonMA
| | - Peter Sicinski
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA
- Department of GeneticsHarvard Medical SchoolBostonMA
| | - Stefan Strack
- Department of Pharmacology and Iowa Neuroscience InstituteCarver College of MedicineUniversity of IowaIowa CityIA
| | - E. Dale Abel
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
- Fraternal Order of Eagles Diabetes Research CenterDivision of Endocrinology and MetabolismCarver College of MedicineUniversity of IowaIowa CityIA
| | - Colleen Mitchell
- Department of Mathematics and Delta CenterUniversity of IowaIowa CityIA
| | - Duane D. Hall
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
| | - Chad E. Grueter
- Abboud Cardiovascular Research CenterDivision of Cardiovascular MedicineDepartment of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA
- Interdisciplinary Graduate Program in GeneticsUniversity of IowaIowa CityIA
- Fraternal Order of Eagles Diabetes Research CenterDivision of Endocrinology and MetabolismCarver College of MedicineUniversity of IowaIowa CityIA
| |
Collapse
|