1
|
Xia K, Zhou Y, Xie Y, Cai Y. Role of SMYD2 in gastrointestinal cancer progression (Review). Oncol Lett 2025; 29:282. [PMID: 40242267 PMCID: PMC12001312 DOI: 10.3892/ol.2025.15028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Gastrointestinal cancer is one of the most prevalent malignancies in humans and is often associated with a poor prognosis. Understanding the molecular mechanisms underlying cancer progression and severity is essential for the development of effective cancer therapies. Abnormal protein methylation is associated with the occurrence and advancement of cancer, highlighting the importance of protein methyltransferase research. SET and MYND domain-containing protein 2 (SMYD2), a lysine methyltransferase, has emerged as a promising small molecule target for cancer treatment. Notably, SMYD2 is implicated in the pathogenesis of several diseases, including gastrointestinal cancer. SMYD2 is closely associated with the tumorigenesis, proliferation, migration and other biological processes of gastrointestinal cancer, indicating its potential as a novel therapeutic target. The present review offers an in-depth analysis of SMYD2, covering its structural characteristics, regulatory pathways and functional significance. By assessing the biological roles and therapeutic potential of SMYD2, the current review presents fresh insights and perspectives for advancing research in different types of gastrointestinal cancer.
Collapse
Affiliation(s)
- Kun Xia
- Department of General Surgery, People's Hospital of Ningxiang City, Ningxiang, Hunan 410600, P.R. China
| | - Yaoxiang Zhou
- Department of General Surgery, People's Hospital of Ningxiang City, Ningxiang, Hunan 410600, P.R. China
| | - Youping Xie
- Department of General Surgery, People's Hospital of Ningxiang City, Ningxiang, Hunan 410600, P.R. China
| | - Yinzhong Cai
- Department of General Surgery, People's Hospital of Ningxiang City, Ningxiang, Hunan 410600, P.R. China
| |
Collapse
|
2
|
Fang W, Song L, Li Z, Meng P, Zuo S, Liu S. Effect of miRNA-200b on the proliferation of liver cancer cells via targeting SMYD2/p53 signaling pathway. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1303-1314. [PMID: 36411681 PMCID: PMC10930361 DOI: 10.11817/j.issn.1672-7347.2022.210521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
OBJECTIVES Our previous study has verified that high level of SET and MYND domain-containing protein 2 (SMYD2) plays an important role in acquiring aggressive ability for liver cancer cells in hepatocellular carcinoma. MiR-200b as a tumor suppressor gene involves in a variety of cancers. This study aims to investigate the correlation between miR-200b and SMYD2 in hepatocellular carcinoma and the underlying mechanism. METHODS Firstly, the levels of SMYD2 and miR-200b in hepatocellular carcinoma tissues and matched adjacent non-tumor liver tissues were tested with real-time reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting. Secondly, we evaluated the interaction between miR-200b and SMYD2 using dual-luciferase reporter assay. Thirdly, we elucidated the effect of miR-200b on SMYD2 and its downstream targets p53/CyclinE1. Finally, we silenced SMYD2 in hepatocellular carcinoma cell lines to investigate its effect on tumor proliferation and cell cycle progression, and further confirmed the correlation among SMYD2 and p53/CyclinE1. RESULTS Compared with the matched adjacent non-tumor liver tissues, miR-200b was obviously decreased, and SMYD2 was significantly increased in hepatocellular carcinoma (both P<0.05). Spearman's rank correlation revealed that miR-200b expression was negatively correlated with SMYD2 (P<0.01). Computer algorithm and dual-luciferase reporter assay revealed that miR-200b directly targeted and suppressed SMYD2 in HEK 293T cells. The down-regulated miR-200b expression promoted hepatoma cell proliferation (P<0.05) and increased SMYD2 expression(P<0.01), while the up-regulated expression of miR-200b had an opposite effect. The knockdown of SMYD2 suppressed the proliferation of MHCC-97L cells (P<0.01), down-regulated CyclinE1, and up-regulated p53 expression (both P<0.05). CONCLUSIONS MiR-200b is involved in hepatocellular carcinoma progression via targeting SMYD2 and regulating SMYD2/p53/CyclinE1 signaling pathway and may be used as a potential target for hepatocellular carcinoma treatment.
Collapse
Affiliation(s)
- Weijin Fang
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha 410013.
| | - Liying Song
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Zuojun Li
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Peipei Meng
- Department of Pharmacy, Women and Children's Health Care Hospital of Linyi, Linyi Shandong 276000, China
| | - Shanru Zuo
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Shikun Liu
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha 410013.
| |
Collapse
|
3
|
Yoshida GM, Yáñez JM. Increased accuracy of genomic predictions for growth under chronic thermal stress in rainbow trout by prioritizing variants from GWAS using imputed sequence data. Evol Appl 2022; 15:537-552. [PMID: 35505881 PMCID: PMC9046923 DOI: 10.1111/eva.13240] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023] Open
Abstract
Through imputation of genotypes, genome-wide association study (GWAS) and genomic prediction (GP) using whole-genome sequencing (WGS) data are cost-efficient and feasible in aquaculture breeding schemes. The objective was to dissect the genetic architecture of growth traits under chronic heat stress in rainbow trout (Oncorhynchus mykiss) and to assess the accuracy of GP based on imputed WGS and different preselected single nucleotide polymorphism (SNP) arrays. A total of 192 and 764 fish challenged to a heat stress experiment for 62 days were genotyped using a customized 1 K and 26 K SNP panels, respectively, and then, genotype imputation was performed from a low-density chip to WGS using 102 parents (36 males and 66 females) as the reference population. Imputed WGS data were used to perform GWAS and test GP accuracy under different preselected SNP scenarios. Heritability was estimated for body weight (BW), body length (BL) and average daily gain (ADG). Estimates using imputed WGS data ranged from 0.33 ± 0.05 to 0.55 ± 0.05 for growth traits under chronic heat stress. GWAS revealed that the top five cumulatively SNPs explained a maximum of 0.94%, 0.86% and 0.51% of genetic variance for BW, BL and ADG, respectively. Some important functional candidate genes associated with growth-related traits were found among the most important SNPs, including signal transducer and activator of transcription 5B and 3 (STAT5B and STAT3, respectively) and cytokine-inducible SH2-containing protein (CISH). WGS data resulted in a slight increase in prediction accuracy compared with pedigree-based method, whereas preselected SNPs based on the top GWAS hits improved prediction accuracies, with values ranging from 1.2 to 13.3%. Our results support the evidence of the polygenic nature of growth traits when measured under heat stress. The accuracies of GP can be improved using preselected variants from GWAS, and the use of WGS marginally increases prediction accuracy.
Collapse
Affiliation(s)
| | - José M. Yáñez
- Facultad de Ciencias Veterinarias y PecuariasUniversidad de ChileSantiagoChile
- Núcleo Milenio INVASALConcepciónChile
| |
Collapse
|
4
|
van der Pijl RJ, Domenighetti AA, Sheikh F, Ehler E, Ottenheijm CAC, Lange S. The titin N2B and N2A regions: biomechanical and metabolic signaling hubs in cross-striated muscles. Biophys Rev 2021; 13:653-677. [PMID: 34745373 PMCID: PMC8553726 DOI: 10.1007/s12551-021-00836-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Muscle specific signaling has been shown to originate from myofilaments and their associated cellular structures, including the sarcomeres, costameres or the cardiac intercalated disc. Two signaling hubs that play important biomechanical roles for cardiac and/or skeletal muscle physiology are the N2B and N2A regions in the giant protein titin. Prominent proteins associated with these regions in titin are chaperones Hsp90 and αB-crystallin, members of the four-and-a-half LIM (FHL) and muscle ankyrin repeat protein (Ankrd) families, as well as thin filament-associated proteins, such as myopalladin. This review highlights biological roles and properties of the titin N2B and N2A regions in health and disease. Special emphasis is placed on functions of Ankrd and FHL proteins as mechanosensors that modulate muscle-specific signaling and muscle growth. This region of the sarcomere also emerged as a hotspot for the modulation of passive muscle mechanics through altered titin phosphorylation and splicing, as well as tethering mechanisms that link titin to the thin filament system.
Collapse
Affiliation(s)
| | - Andrea A. Domenighetti
- Shirley Ryan AbilityLab, Chicago, IL USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL USA
| | - Farah Sheikh
- Division of Cardiology, School of Medicine, UC San Diego, La Jolla, CA USA
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Coen A. C. Ottenheijm
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ USA
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Stephan Lange
- Division of Cardiology, School of Medicine, UC San Diego, La Jolla, CA USA
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Function of the MYND Domain and C-Terminal Region in Regulating the Subcellular Localization and Catalytic Activity of the SMYD Family Lysine Methyltransferase Set5. Mol Cell Biol 2020; 40:MCB.00341-19. [PMID: 31685550 DOI: 10.1128/mcb.00341-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/01/2019] [Indexed: 11/20/2022] Open
Abstract
SMYD lysine methyltransferases target histones and nonhistone proteins for methylation and are critical regulators of muscle development and implicated in neoplastic transformation. They are characterized by a split catalytic SET domain and an intervening MYND zinc finger domain, as well as an extended C-terminal domain. Saccharomyces cerevisiae contains two SMYD proteins, Set5 and Set6, which share structural elements with the mammalian SMYD enzymes. Set5 is a histone H4 lysine 5, 8, and 12 methyltransferase, implicated in the regulation of stress responses and genome stability. While the SMYD proteins have diverse roles in cells, there are many gaps in our understanding of how these enzymes are regulated. Here, we performed mutational analysis of Set5, combined with phosphoproteomics, to identify regulatory mechanisms for its enzymatic activity and subcellular localization. Our results indicate that the MYND domain promotes Set5 chromatin association in cells and is required for its role in repressing subtelomeric genes. Phosphoproteomics revealed extensive phosphorylation of Set5, and phosphomimetic mutations enhance Set5 catalytic activity but diminish its ability to interact with chromatin in cells. These studies uncover multiple regions within Set5 that regulate its localization and activity and highlight potential avenues for understanding mechanisms controlling the diverse roles of SMYD enzymes.
Collapse
|
6
|
Zuo SR, Zuo XC, He Y, Fang WJ, Wang CJ, Zou H, Chen P, Huang LF, Huang LH, Xiang H, Liu SK. Positive Expression of SMYD2 is Associated with Poor Prognosis in Patients with Primary Hepatocellular Carcinoma. J Cancer 2018; 9:321-330. [PMID: 29344279 PMCID: PMC5771340 DOI: 10.7150/jca.22218] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose: SET and MYND domain-containing protein2 (SMYD2), a histone lysine methyltransferases, is a candidate human oncogene in multiple tumors. However, the expression dynamics of SMYD2 in hepatocellular carcinoma (HCC) and its clinical/prognostic significance are unclear. Methods: The SMYD2 expression profile was examined by quantitative real-time polymerase chain reaction (qRT-PCR), and immunohistochemistry (IHC) in HCC tissues and matched adjacent non-tumorous tissues. SMYD2 was silenced in HCC cell lines to determine its role in tumor proliferation and cell cycle progression, and the possible mechanism. Spearman's rank correlation, Kaplan-Meier plots and Cox proportional hazards regression model were used to analyze the data. Results: The SMYD2 expression in HCC tissues were significantly up-regulated at both mRNA and protein levels as compared with the matched adjacent non-tumorous tissues. By IHC, positive expression of SMYD2 was examined in 122/163 (74.85%) of HCC and in 10/59 (16.95%) of tumor-adjacent tissues. Positive expression of SMYD2 was correlated with tumor size, vascular invasion, differentiation and TNM stage (P < 0.05). In univariate survival analysis, a significant association between positive expression of SMYD2 and shortened patients' survival was found (P < 0.05). Importantly, SMYD2 expression together with vascular invasion (P < 0.05) provided significant independent prognostic parameters in multivariate analysis. Functionally, SMYD2 silenced markedly inhibited cell proliferation and cell cycle progression in SMMC-7721 cell. Conclusions: Our findings provide evidences that positive expression of SMYD2 in HCC may be important in the acquisition of an aggressive phenotype, and it is an independent biomarker for poor prognosis of patients with HCC.
Collapse
Affiliation(s)
- Shan-Ru Zuo
- Department of Center Clinical Pharmacology and Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Cong Zuo
- Department of Center Clinical Pharmacology and Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yang He
- Department of Center Clinical Pharmacology and Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei-Jin Fang
- Department of Center Clinical Pharmacology and Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chun-Jiang Wang
- Department of Center Clinical Pharmacology and Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Heng Zou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Pan Chen
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ling-Fei Huang
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhe Jiang Province, Taizhou, China
| | - Li-Hua Huang
- Center for Experimental Medicine Research, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hong Xiang
- Center for Experimental Medicine Research, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shi-Kun Liu
- Department of Center Clinical Pharmacology and Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
7
|
Stewart MD, Lopez S, Nagandla H, Soibam B, Benham A, Nguyen J, Valenzuela N, Wu HJ, Burns AR, Rasmussen TL, Tucker HO, Schwartz RJ. Mouse myofibers lacking the SMYD1 methyltransferase are susceptible to atrophy, internalization of nuclei and myofibrillar disarray. Dis Model Mech 2016; 9:347-59. [PMID: 26935107 PMCID: PMC4833328 DOI: 10.1242/dmm.022491] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The Smyd1 gene encodes a lysine methyltransferase specifically expressed in striated muscle. Because Smyd1-null mouse embryos die from heart malformation prior to formation of skeletal muscle, we developed a Smyd1 conditional-knockout allele to determine the consequence of SMYD1 loss in mammalian skeletal muscle. Ablation of SMYD1 specifically in skeletal myocytes after myofiber differentiation using Myf6(cre) produced a non-degenerative myopathy. Mutant mice exhibited weakness, myofiber hypotrophy, prevalence of oxidative myofibers, reduction in triad numbers, regional myofibrillar disorganization/breakdown and a high percentage of myofibers with centralized nuclei. Notably, we found broad upregulation of muscle development genes in the absence of regenerating or degenerating myofibers. These data suggest that the afflicted fibers are in a continual state of repair in an attempt to restore damaged myofibrils. Disease severity was greater for males than females. Despite equivalent expression in all fiber types, loss of SMYD1 primarily affected fast-twitch muscle, illustrating fiber-type-specific functions for SMYD1. This work illustrates a crucial role for SMYD1 in skeletal muscle physiology and myofibril integrity.
Collapse
Affiliation(s)
- M David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Suhujey Lopez
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Harika Nagandla
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Benjamin Soibam
- Department of Computer Science and Engineering Technology, University of Houston-Downtown, Houston, TX 77002, USA
| | - Ashley Benham
- Stem Cell Engineering Department, Texas Heart Institute at St Luke's Episcopal Hospital, Houston, TX 77030, USA
| | - Jasmine Nguyen
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Nicolas Valenzuela
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Harry J Wu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Alan R Burns
- College of Optometry, University of Houston, Houston, TX 77204, USA
| | - Tara L Rasmussen
- Department of Molecular Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haley O Tucker
- Department of Molecular Biosciences, Institute for Cellular Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA Stem Cell Engineering Department, Texas Heart Institute at St Luke's Episcopal Hospital, Houston, TX 77030, USA
| |
Collapse
|
8
|
Nagandla H, Lopez S, Yu W, Rasmussen TL, Tucker HO, Schwartz RJ, Stewart MD. Defective myogenesis in the absence of the muscle-specific lysine methyltransferase SMYD1. Dev Biol 2015; 410:86-97. [PMID: 26688546 DOI: 10.1016/j.ydbio.2015.12.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/07/2015] [Accepted: 12/07/2015] [Indexed: 11/19/2022]
Abstract
The SMYD (SET and MYND domain) family of lysine methyltransferases harbor a unique structure in which the methyltransferase (SET) domain is intervened by a zinc finger protein-protein interaction MYND domain. SMYD proteins methylate both histone and non-histone substrates and participate in diverse biological processes including transcriptional regulation, DNA repair, proliferation and apoptosis. Smyd1 is unique among the five family members in that it is specifically expressed in striated muscles. Smyd1 is critical for development of the right ventricle in mice. In zebrafish, Smyd1 is necessary for sarcomerogenesis in fast-twitch muscles. Smyd1 is expressed in the skeletal muscle lineage throughout myogenesis and in mature myofibers, shuttling from nucleus to cytosol during myoblast differentiation. Because of this expression pattern, we hypothesized that Smyd1 plays multiple roles at different stages of myogenesis. To determine the role of Smyd1 in mammalian myogenesis, we conditionally eliminated Smyd1 from the skeletal muscle lineage at the myoblast stage using Myf5(cre). Deletion of Smyd1 impaired myoblast differentiation, resulted in fewer myofibers and decreased expression of muscle-specific genes. Muscular defects were temporally restricted to the second wave of myogenesis. Thus, in addition to the previously described functions for Smyd1 in heart development and skeletal muscle sarcomerogenesis, these results point to a novel role for Smyd1 in myoblast differentiation.
Collapse
Affiliation(s)
- Harika Nagandla
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Suhujey Lopez
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Wei Yu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Tara L Rasmussen
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA; Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, TX, USA
| | - Haley O Tucker
- Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, TX, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA; Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX, USA
| | - M David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| |
Collapse
|
9
|
Sharma M, McFarlane C, Kambadur R, Kukreti H, Bonala S, Srinivasan S. Myostatin: expanding horizons. IUBMB Life 2015; 67:589-600. [PMID: 26305594 DOI: 10.1002/iub.1392] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 05/29/2015] [Indexed: 12/13/2022]
Abstract
Myostatin is a secreted growth and differentiation factor that belongs to the TGF-β superfamily. Myostatin is predominantly synthesized and expressed in skeletal muscle and thus exerts a huge impact on muscle growth and function. In keeping with its negative role in myogenesis, myostatin expression is tightly regulated at several levels including epigenetic, transcriptional, post-transcriptional, and post-translational. New revelations regarding myostatin regulation also offer mechanisms that could be exploited for developing myostatin antagonists. Increasingly, it is becoming clearer that besides its conventional role in muscle, myostatin plays a critical role in metabolism. Hence, molecular mechanisms by which myostatin regulates several key metabolic processes need to be further explored.
Collapse
Affiliation(s)
- Mridula Sharma
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Department of Cell & Molecular Biology, Brenner Centre for Molecular Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
| | - Craig McFarlane
- Department of Cell & Molecular Biology, Brenner Centre for Molecular Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
| | - Ravi Kambadur
- Department of Cell & Molecular Biology, Brenner Centre for Molecular Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Himani Kukreti
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Sabeera Bonala
- Department of Cell & Molecular Biology, Brenner Centre for Molecular Medicine, Singapore Institute of Clinical Sciences (SICS), Singapore
| | - Shruti Srinivasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| |
Collapse
|
10
|
Du SJ, Tan X, Zhang J. SMYD proteins: key regulators in skeletal and cardiac muscle development and function. Anat Rec (Hoboken) 2015; 297:1650-62. [PMID: 25125178 DOI: 10.1002/ar.22972] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 04/28/2014] [Accepted: 04/28/2014] [Indexed: 11/07/2022]
Abstract
Muscle fibers are composed of myofibrils, one of the most highly ordered macromolecular assemblies in cells. Recent studies demonstrate that members of the Smyd family play critical roles in myofibril assembly of skeletal and cardiac muscle during development. The Smyd family consists of five members including Smyd1, Smyd2, Smyd3, Smyd4, and Smyd5. They share two highly conserved structural and functional domains, namely the SET and MYND domains involved in lysine methylation and protein-protein interaction, respectively. Smyd1 is specifically expressed in muscle cells under the regulation of myogenic transcriptional factors of the MyoD and Mef2 families and the serum responsive factor. Loss of function studies reveal that Smyd1 is required for cardiomyogenesis and sarcomere assembly in skeletal and cardiac muscles. Smyd2, on another hand, is dispensable for heart development in mice. However, Smyd2 appears to play a role in myofilament organization in both skeletal and cardiac muscles via Hsp90 methylation. A Drosophila Smyd4 homologue is a muscle-specific transcriptional modulator involved in the development or function of adult muscle. The molecular mechanisms by which Smyd family proteins function in muscle cells are not well understood. It has been suggested that members of the Smyd family may use multiple mechanisms to control muscle development and cell differentiation, including transcriptional regulation, epigenetic regulation via histone methylation, and methylation of proteins other than histones, such as molecular chaperone Hsp90.
Collapse
Affiliation(s)
- Shao Jun Du
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | | | | |
Collapse
|
11
|
Rasmussen TL, Ma Y, Park CY, Harriss J, Pierce SA, Dekker JD, Valenzuela N, Srivastava D, Schwartz RJ, Stewart MD, Tucker HO. Smyd1 facilitates heart development by antagonizing oxidative and ER stress responses. PLoS One 2015; 10:e0121765. [PMID: 25803368 PMCID: PMC4372598 DOI: 10.1371/journal.pone.0121765] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 02/18/2015] [Indexed: 12/21/2022] Open
Abstract
Smyd1/Bop is an evolutionary conserved histone methyltransferase previously shown by conventional knockout to be critical for embryonic heart development. To further explore the mechanism(s) in a cell autonomous context, we conditionally ablated Smyd1 in the first and second heart fields of mice using a knock-in (KI) Nkx2.5-cre driver. Robust deletion of floxed-Smyd1 in cardiomyocytes and the outflow tract (OFT) resulted in embryonic lethality at E9.5, truncation of the OFT and right ventricle, and additional defects consistent with impaired expansion and proliferation of the second heart field (SHF). Using a transgenic (Tg) Nkx2.5-cre driver previously shown to not delete in the SHF and OFT, early embryonic lethality was bypassed and both ventricular chambers were formed; however, reduced cardiomyocyte proliferation and other heart defects resulted in later embryonic death at E11.5-12.5. Proliferative impairment prior to both early and mid-gestational lethality was accompanied by dysregulation of transcripts critical for endoplasmic reticulum (ER) stress. Mid-gestational death was also associated with impairment of oxidative stress defense—a phenotype highly similar to the previously characterized knockout of the Smyd1-interacting transcription factor, skNAC. We describe a potential feedback mechanism in which the stress response factor Tribbles3/TRB3, when directly methylated by Smyd1, acts as a co-repressor of Smyd1-mediated transcription. Our findings suggest that Smyd1 is required for maintaining cardiomyocyte proliferation at minimally two different embryonic heart developmental stages, and its loss leads to linked stress responses that signal ensuing lethality.
Collapse
Affiliation(s)
- Tara L. Rasmussen
- Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, Texas, United States of America
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Affiliated Hospital of Hainan Medical University, Haikou, Hainan, P.R. China
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Chong Yon Park
- Gladstone Institute of Cardiovascular Disease and Departments of Pediatrics and Biochemistry and Biophysics, University of California, San Francisco, California, United States of America
| | - June Harriss
- Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, Texas, United States of America
| | - Stephanie A. Pierce
- Gladstone Institute of Cardiovascular Disease and Departments of Pediatrics and Biochemistry and Biophysics, University of California, San Francisco, California, United States of America
| | - Joseph D. Dekker
- Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, Texas, United States of America
| | - Nicolas Valenzuela
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease and Departments of Pediatrics and Biochemistry and Biophysics, University of California, San Francisco, California, United States of America
| | - Robert J. Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, United States of America
| | - M. David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, United States of America
- * E-mail: (MDS); (HT)
| | - Haley O. Tucker
- Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas, Austin, Texas, United States of America
- * E-mail: (MDS); (HT)
| |
Collapse
|
12
|
Voelkel T, Andresen C, Unger A, Just S, Rottbauer W, Linke WA. Lysine methyltransferase Smyd2 regulates Hsp90-mediated protection of the sarcomeric titin springs and cardiac function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:812-22. [DOI: 10.1016/j.bbamcr.2012.09.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/17/2012] [Accepted: 09/18/2012] [Indexed: 11/16/2022]
|
13
|
Cho HS, Hayami S, Toyokawa G, Maejima K, Yamane Y, Suzuki T, Dohmae N, Kogure M, Kang D, Neal DE, Ponder BAJ, Yamaue H, Nakamura Y, Hamamoto R. RB1 methylation by SMYD2 enhances cell cycle progression through an increase of RB1 phosphorylation. Neoplasia 2012; 14:476-86. [PMID: 22787429 PMCID: PMC3394190 DOI: 10.1593/neo.12656] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 05/05/2012] [Accepted: 05/07/2012] [Indexed: 12/13/2022]
Abstract
It is well known that RB functions are regulated by posttranslational modifications such as phosphorylation and acetylation, but the significance of lysine methylation on RB has not been fully elucidated. Our expression analysis of SMYD2 by quantitative real-time polymerase chain reaction showed that expression levels of SMYD2 are significantly elevated in human bladder carcinomas compared with nonneoplastic bladder tissues (P < .0001), and its expression levels in tumor tissues were much higher than those of any other normal tissues. SMYD2 knockdown resulted in the suppression of cancer cell growth, and cell cycle analysis indicated that SMYD2 might play a crucial role in the G(1)/S transition. According to an in vitro methyltransferase assay, we found that SMYD2 methylates RB1 protein, and liquid chromatography-tandem mass spectrometry analysis revealed lysine 810 of RB1 to be methylated by SMYD2. Importantly, this methylation enhanced Ser 807/811 phosphorylation of RB1 both in vitro and in vivo. Furthermore, we demonstrated that methylated RB1 accelerates E2F transcriptional activity and promotes cell cycle progression. SMYD2 is an important oncoprotein in various types of cancer, and SMYD2-dependent RB1 methylation at lysine 810 promotes cell cycle progression of cancer cells. Further study may explore SMYD2-dependent RB1 methylation as a potential therapeutic target in human cancer.
Collapse
Affiliation(s)
- Hyun-Soo Cho
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shinya Hayami
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Gouji Toyokawa
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Maejima
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuka Yamane
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | - Naoshi Dohmae
- Biomolecular Characterization Team, RIKEN, Saitama, Japan
| | - Masaharu Kogure
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Daechun Kang
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - David E Neal
- Department of Oncology, Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Bruce AJ Ponder
- Department of Oncology, Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Hiroki Yamaue
- Second Department of Surgery, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yusuke Nakamura
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryuji Hamamoto
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Oncology, Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
14
|
Donlin LT, Andresen C, Just S, Rudensky E, Pappas CT, Kruger M, Jacobs EY, Unger A, Zieseniss A, Dobenecker MW, Voelkel T, Chait BT, Gregorio CC, Rottbauer W, Tarakhovsky A, Linke WA. Smyd2 controls cytoplasmic lysine methylation of Hsp90 and myofilament organization. Genes Dev 2012; 26:114-9. [PMID: 22241783 DOI: 10.1101/gad.177758.111] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Protein lysine methylation is one of the most widespread post-translational modifications in the nuclei of eukaryotic cells. Methylated lysines on histones and nonhistone proteins promote the formation of protein complexes that control gene expression and DNA replication and repair. In the cytoplasm, however, the role of lysine methylation in protein complex formation is not well established. Here we report that the cytoplasmic protein chaperone Hsp90 is methylated by the lysine methyltransferase Smyd2 in various cell types. In muscle, Hsp90 methylation contributes to the formation of a protein complex containing Smyd2, Hsp90, and the sarcomeric protein titin. Deficiency in Smyd2 results in the loss of Hsp90 methylation, impaired titin stability, and altered muscle function. Collectively, our data reveal a cytoplasmic protein network that employs lysine methylation for the maintenance and function of skeletal muscle.
Collapse
Affiliation(s)
- Laura T Donlin
- Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Wang L, Li L, Zhang H, Luo X, Dai J, Zhou S, Gu J, Zhu J, Atadja P, Lu C, Li E, Zhao K. Structure of human SMYD2 protein reveals the basis of p53 tumor suppressor methylation. J Biol Chem 2011; 286:38725-38737. [PMID: 21880715 PMCID: PMC3207477 DOI: 10.1074/jbc.m111.262410] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 08/17/2011] [Indexed: 11/06/2022] Open
Abstract
SMYD2 belongs to a subfamily of histone lysine methyltransferase and was recently identified to methylate tumor suppressor p53 and Rb. Here we report that SMYD2 prefers to methylate p53 Lys-370 over histone substrates in vitro. Consistently, the level of endogenous p53 Lys-370 monomethylation is significantly elevated when SMYD2 is overexpressed in vivo. We have solved the high resolution crystal structures of the full-length SMYD2 protein in binary complex with its cofactor S-adenosylmethionine and in ternary complex with cofactor product S-adenosylhomocysteine and p53 substrate peptide (residues 368-375), respectively. p53 peptide binds to a deep pocket of the interface between catalytic SET(1-282) and C-terminal domain (CTD) with an unprecedented U-shaped conformation. Subtle conformational change exists around the p53 binding site between the binary and ternary structures, in particular the tetratricopeptide repeat motif of the CTD. In addition, a unique EDEE motif between the loop of anti-parallel β7 and β8 sheets of the SET core not only interacts with p53 substrate but also forms a hydrogen bond network with residues from CTD. These observations suggest that the tetratricopeptide repeat and EDEE motif may play an important role in determining p53 substrate binding specificity. This is further verified by the findings that deletion of the CTD domain drastically reduces the methylation activity of SMYD2 to p53 protein. Meanwhile, mutation of EDEE residues impairs both the binding and the enzymatic activity of SMYD2 to p53 Lys-370. These data together reveal the molecular basis of SMYD2 in specifically recognizing and regulating functions of p53 tumor suppressor through Lys-370 monomethylation.
Collapse
Affiliation(s)
- Li Wang
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Ling Li
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Hailong Zhang
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Xiao Luo
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Jingquan Dai
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Shaolian Zhou
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Justin Gu
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Jidong Zhu
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Peter Atadja
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Chris Lu
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - En Li
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China
| | - Kehao Zhao
- Novartis Institutes for BioMedical Research, Building 8, Lane 898, Halei Road, Pudong, Shanghai 201203, China.
| |
Collapse
|
16
|
Fujii T, Tsunesumi SI, Yamaguchi K, Watanabe S, Furukawa Y. Smyd3 is required for the development of cardiac and skeletal muscle in zebrafish. PLoS One 2011; 6:e23491. [PMID: 21887258 PMCID: PMC3160858 DOI: 10.1371/journal.pone.0023491] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 07/19/2011] [Indexed: 01/17/2023] Open
Abstract
Modifications of histone tails are involved in the regulation of a wide range of biological processes including cell cycle, cell survival, cell division, and cell differentiation. Among the modifications, histone methylation plays a critical role in cardiac and skeletal muscle differentiation. In our earlier studies, we found that SMYD3 has methyltransferase activity to histone H3 lysine 4, and that its up-regulation is involved in the tumorigenesis of human colon, liver, and breast. To clarify the role of Smyd3 in development, we have studied its expression patterns in zebrafish embryos and the effect of its suppression on development using Smyd3-specific antisense morpholino-oligonucleotides. We here show that transcripts of smyd3 were expressed in zebrafish embryos at all developmental stages examined and that knockdown of smyd3 in embryos resulted in pericardial edema and defects in the trunk structure. In addition, these phenotypes were associated with abnormal expression of three heart-chamber markers including cmlc2, amhc and vmhc, and abnormal expression of myogenic regulatory factors including myod and myog. These data suggest that Smyd3 plays an important role in the development of heart and skeletal muscle.
Collapse
Affiliation(s)
- Tomoaki Fujii
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Japan
| | - Shin-ichiro Tsunesumi
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Japan
| | - Sumiko Watanabe
- Division of Molecular Developmental Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Japan
- * E-mail:
| |
Collapse
|
17
|
Jiang Y, Sirinupong N, Brunzelle J, Yang Z. Crystal structures of histone and p53 methyltransferase SmyD2 reveal a conformational flexibility of the autoinhibitory C-terminal domain. PLoS One 2011; 6:e21640. [PMID: 21738746 PMCID: PMC3125274 DOI: 10.1371/journal.pone.0021640] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 06/03/2011] [Indexed: 12/22/2022] Open
Abstract
SmyD2 belongs to a new class of chromatin regulators that control gene expression in heart development and tumorigenesis. Besides methylation of histone H3 K4, SmyD2 can methylate non-histone targets including p53 and the retinoblastoma tumor suppressor. The methyltransferase activity of SmyD proteins has been proposed to be regulated by autoinhibition via the intra- and interdomain bending of the conserved C-terminal domain (CTD). However, there has been no direct evidence of a conformational change in the CTD. Here, we report two crystal structures of SmyD2 bound either to the cofactor product S-adenosylhomocysteine or to the inhibitor sinefungin. SmyD2 has a two-lobed structure with the active site located at the bottom of a deep crevice formed between the CTD and the catalytic domain. By extensive engagement with the methyltransferase domain, the CTD stabilizes the autoinhibited conformation of SmyD2 and restricts access to the catalytic site. Unexpectedly, despite that the two SmyD2 structures are highly superimposable, significant differences are observed in the first two helices of the CTDs: the two helices bend outwards and move away from the catalytic domain to generate a less closed conformation in the sinefungin-bound structure. Although the overall fold of the individual domains is structurally conserved among SmyD proteins, SmyD2 appear to be a conformational “intermediate” between a close form of SmyD3 and an open form of SmyD1. In addition, the structures reveal that the CTD is structurally similar to tetratricopeptide repeats (TPR), a motif through which many cochaperones bind to the heat shock protein Hsp90. Our results thus provide the first evidence for the intradomain flexibility of the TPR-like CTD, which may be important for the activation of SmyD proteins by Hsp90.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Nualpun Sirinupong
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Joseph Brunzelle
- Advance Photon Source, Argonne National Lab, Argonne, Illinois, United States of America
| | - Zhe Yang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
18
|
Structural insights into the autoinhibition and posttranslational activation of histone methyltransferase SmyD3. J Mol Biol 2010; 406:149-59. [PMID: 21167177 DOI: 10.1016/j.jmb.2010.12.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Revised: 11/29/2010] [Accepted: 12/07/2010] [Indexed: 10/18/2022]
Abstract
The SmyD family represents a new class of chromatin regulators that is important in heart and skeletal muscle development. However, the critical questions regarding how they are regulated posttranslationally remain largely unknown. We previously suggested that the histone methyltransferase activity of SmyD1, a vital myogenic regulator, appears to be regulated by autoinhibition and that the possible hinge motion of the conserved C-terminal domain (CTD) might be central to the maintenance and release of the autoinhibition. However, the lack of direct evidence of the hinge motion has limited our further understanding of this autoinhibitory mechanism. Here, we report the crystal structure of full-length SmyD3 in complex with the methyltransferase inhibitor sinefungin at 1.7 Å. SmyD3 has a two-lobed structure with the substrate binding cleft located at the bottom of a 15-Å-deep crevice formed between the N- and C-terminal lobes. Comparison of SmyD3 and SmyD1 clearly suggests that the CTD can undergo a large hinge-bending motion that defines two distinct conformations: SmyD3 adopts a closed conformation with the CTD partially blocking the substrate binding cleft; in contrast, SmyD1 appears to represent an open form, where the CTD swings out by ∼12 Å from the N-terminal lobe, forming an open cleft with the active site completely exposed. Overall, these findings provide novel structural insights into the mechanism that modulates the activity of the SmyD proteins and support the observation that a posttranslational activation, such as by molecular chaperon Hsp90, is required to potentiate the proteins.
Collapse
|
19
|
Diehl F, Brown MA, van Amerongen MJ, Novoyatleva T, Wietelmann A, Harriss J, Ferrazzi F, Böttger T, Harvey RP, Tucker PW, Engel FB. Cardiac deletion of Smyd2 is dispensable for mouse heart development. PLoS One 2010; 5:e9748. [PMID: 20305823 PMCID: PMC2840034 DOI: 10.1371/journal.pone.0009748] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 02/25/2010] [Indexed: 01/02/2023] Open
Abstract
Chromatin modifying enzymes play a critical role in cardiac differentiation. Previously, it has been shown that the targeted deletion of the histone methyltransferase, Smyd1, the founding member of the SET and MYND domain containing (Smyd) family, interferes with cardiomyocyte maturation and proper formation of the right heart ventricle. The highly related paralogue, Smyd2 is a histone 3 lysine 4- and lysine 36-specific methyltransferase expressed in heart and brain. Here, we report that Smyd2 is differentially expressed during cardiac development with highest expression in the neonatal heart. To elucidate the functional role of Smyd2 in the heart, we generated conditional knockout (cKO) mice harboring a cardiomyocyte-specific deletion of Smyd2 and performed histological, functional and molecular analyses. Unexpectedly, cardiac deletion of Smyd2 was dispensable for proper morphological and functional development of the murine heart and had no effect on global histone 3 lysine 4 or 36 methylation. However, we provide evidence for a potential role of Smyd2 in the transcriptional regulation of genes associated with translation and reveal that Smyd2, similar to Smyd3, interacts with RNA Polymerase II as well as to the RNA helicase, HELZ.
Collapse
Affiliation(s)
- Florian Diehl
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Hessen, Germany
| | - Mark A. Brown
- Section of Molecular Genetics and Microbiology and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Machteld J. van Amerongen
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Hessen, Germany
| | - Tatyana Novoyatleva
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Hessen, Germany
| | - Astrid Wietelmann
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Hessen, Germany
| | - June Harriss
- Section of Molecular Genetics and Microbiology and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Fulvia Ferrazzi
- Dipartimento di Informatica e Sistemistica, Università degli Studi di Pavia, Pavia, Lombardia, Italia
| | - Thomas Böttger
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Hessen, Germany
| | - Richard P. Harvey
- Developmental Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Philip W. Tucker
- Section of Molecular Genetics and Microbiology and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Felix B. Engel
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Hessen, Germany
- * E-mail:
| |
Collapse
|
20
|
Li D, Niu Z, Yu W, Qian Y, Wang Q, Li Q, Yi Z, Luo J, Wu X, Wang Y, Schwartz RJ, Liu M. SMYD1, the myogenic activator, is a direct target of serum response factor and myogenin. Nucleic Acids Res 2010; 37:7059-71. [PMID: 19783823 PMCID: PMC2790895 DOI: 10.1093/nar/gkp773] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
SMYD1 is a heart and muscle specific SET-MYND domain containing protein, which functions as a histone methyltransferase and regulates downstream gene transcription. We demonstrated that the expression of SMYD1 is restricted in the heart and skeletal muscle tissues in human. To reveal the regulatory mechanisms of SMYD1 expression during myogenesis and cardiogenesis, we cloned and characterized the human SMYD1 promoter, which contains highly conserved serum response factor (SRF) and myogenin binding sites. Overexpression of SRF and myogenin significantly increased the endogenous expression level of Smyd1 in C2C12 cells, respectively. Deletion of Srf in the heart of mouse embryos dramatically decreased the expression level of Smyd1 mRNA and the expression of Smyd1 can be rescued by exogenous SRF introduction in SRF null ES cells during differentiation. Furthermore, we demonstrated that SRF binds to the CArG site and myogenin binds to the E-box element on Smyd1 promoter region using EMSA and ChIP assays. Moreover, forced expression of SMYD1 accelerates myoblast differentiation and myotube formation in C2C12 cells. Taken together, these studies demonstrated that SMYD1 is a key regulator of myogenic differentiation and acts as a downstream target of muscle regulatory factors, SRF and myogenin.
Collapse
Affiliation(s)
- Dali Li
- The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|