1
|
Chen Y, Zhao J, Zhong C, Kang Y, Xiong Z, Huang J, Li Z, Liu Q, Shi D, Li X, Wang J, Li H. Enhancer Enh483 regulates myoblast proliferation and differentiation of buffalo myoblasts by targeting FAXC. Cell Tissue Res 2024:10.1007/s00441-024-03944-0. [PMID: 39688691 DOI: 10.1007/s00441-024-03944-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024]
Abstract
A detailed understanding of the precise regulatory mechanisms governing buffalo skeletal muscle is crucial for improving meat quality and yield. Proper skeletal muscle fate decisions necessitate the accurate regulation of key enhancers. This study screened nine potential enhancers linked to muscle development by analysing ATAC-seq data from buffalo myoblasts during the proliferative and differentiative phases. The enhancer activity of these candidates was confirmed in buffalo myoblasts, C2C12, and human skeletal muscle myoblasts using a dual-luciferase reporter system. The CRISPRi system and RT-qPCR were used to test the effects of 9 candidate enhancers on buffalo myoblasts. The active enhancer, Enh483, was selected based on its significant impact. Upon successful inhibition of Enh483 using CRISPRi, decreases in the expression of buffalo myogenic proliferation marker genes (PCNA, CyclinD1, and CDK2) were observed via RT-qPCR and Western blot. Subsequent proliferation assays using CCK-8 and EdU confirmed the promotive effect of Enh483 on buffalo myogenic cell proliferation. Following a 5-day differentiation induction period, changes in the expression of differentiation marker genes (MyoD1, MyoG, and MyHC) were analysed using RT-qPCR and Western blot. Additionally, fused myotube numbers were quantified, and the impact of Enh483 on buffalo myogenic cell differentiation was assessed through immunofluorescence. Our findings indicate that Enh483 facilitates buffalo myogenic cell differentiation. Further interaction analysis utilising 3C-PCR revealed a direct association between Enh483 and the FAXC promoter. In summary, the results from this study lay a foundational framework for deciphering the intricate regulatory mechanisms underpinning buffalo muscle development.
Collapse
Affiliation(s)
- Yaling Chen
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Jiahui Zhao
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Cuiwei Zhong
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Yujin Kang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Zhaocheng Xiong
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Jieping Huang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Zhipeng Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Qingyou Liu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Deshun Shi
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Xinxin Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Jian Wang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Hui Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
2
|
Afzal J, Liu Y, Du W, Suhail Y, Zong P, Feng J, Ajeti V, Sayyad WA, Nikolaus J, Yankova M, Deymier AC, Yue L, Kshitiz. Cardiac ultrastructure inspired matrix induces advanced metabolic and functional maturation of differentiated human cardiomyocytes. Cell Rep 2022; 40:111146. [PMID: 35905711 DOI: 10.1016/j.celrep.2022.111146] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/26/2022] [Accepted: 07/07/2022] [Indexed: 12/21/2022] Open
Abstract
The vast potential of human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) in preclinical models of cardiac pathologies, precision medicine, and drug screening remains to be fully realized because hiPSC-CMs are immature without adult-like characteristics. Here, we present a method to accelerate hiPSC-CM maturation on a substrate, cardiac mimetic matrix (CMM), mimicking adult human heart matrix ligand chemistry, rigidity, and submicron ultrastructure, which synergistically mature hiPSC-CMs rapidly within 30 days. hiPSC-CMs matured on CMM exhibit systemic transcriptomic maturation toward an adult heart state, are aligned with high strain energy, metabolically rely on oxidative phosphorylation and fatty acid oxidation, and display enhanced redox handling capability, efficient calcium handling, and electrophysiological features of ventricular myocytes. Endothelin-1-induced pathological hypertrophy is mitigated on CMM, highlighting the role of a native cardiac microenvironment in withstanding hypertrophy progression. CMM is a convenient model for accelerated development of ventricular myocytes manifesting highly specialized cardiac-specific functions.
Collapse
Affiliation(s)
- Junaid Afzal
- Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Yamin Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA
| | - Wenqiang Du
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA; Center for Cellular Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA
| | - Pengyu Zong
- Department of Cell Biology, University of Connecticut Health, Farmington, CT 06032, USA; Calhoun Cardiology Center, University of Connecticut Health, Farmington, CT 06032, USA
| | - Jianlin Feng
- Department of Cell Biology, University of Connecticut Health, Farmington, CT 06032, USA; Calhoun Cardiology Center, University of Connecticut Health, Farmington, CT 06032, USA
| | - Visar Ajeti
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA; Center for Cellular Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA
| | - Wasim A Sayyad
- Department of Cell Biology, Yale University, New Haven, CT 06510, USA
| | - Joerg Nikolaus
- West Campus Imaging Core, Yale University, New Haven, CT 06477, USA
| | - Maya Yankova
- Electron Microscopy Core, University of Connecticut Health, Farmington, CT 06032, USA
| | - Alix C Deymier
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA
| | - Lixia Yue
- Department of Cell Biology, University of Connecticut Health, Farmington, CT 06032, USA; Calhoun Cardiology Center, University of Connecticut Health, Farmington, CT 06032, USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA; Center for Cellular Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA; Department of Cell Biology, University of Connecticut Health, Farmington, CT 06032, USA.
| |
Collapse
|
3
|
Iwaki H, Sasaki S, Matsushita A, Ohba K, Matsunaga H, Misawa H, Oki Y, Ishizuka K, Nakamura H, Suda T. Essential role of TEA domain transcription factors in the negative regulation of the MYH 7 gene by thyroid hormone and its receptors. PLoS One 2014; 9:e88610. [PMID: 24781449 PMCID: PMC4004540 DOI: 10.1371/journal.pone.0088610] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 01/14/2014] [Indexed: 12/20/2022] Open
Abstract
MYH7 (also referred to as cardiac myosin heavy chain β) gene expression is known to be repressed by thyroid hormone (T3). However, the molecular mechanism by which T3 inhibits the transcription of its target genes (negative regulation) remains to be clarified, whereas those of transcriptional activation by T3 (positive regulation) have been elucidated in detail. Two MCAT (muscle C, A, and T) sites and an A/T-rich region in the MYH7 gene have been shown to play a critical role in the expression of this gene and are known to be recognized by the TEAD/TEF family of transcription factors (TEADs). Using a reconstitution system with CV-1 cells, which has been utilized in the analysis of positive as well as negative regulation, we demonstrate that both T3 receptor (TR) β1 and α1 inhibit TEAD-dependent activation of the MYH7 promoter in a T3 dose-dependent manner. TRβ1 bound with GC-1, a TRβ-selective T3 analog, also repressed TEAD-induced activity. Although T3-dependent inhibition required the DNA-binding domain (DBD) of TRβ1, it remained after the putative negative T3-responsive elements were mutated. A co-immunoprecipitation study demonstrated the in vivo association of TRβ1 with TEAD-1, and the interaction surfaces were mapped to the DBD of the TRβ1 and TEA domains of TEAD-1, both of which are highly conserved among TRs and TEADs, respectively. The importance of TEADs in MYH7 expression was also validated with RNA interference using rat embryonic cardiomyocyte H9c2 cells. These results indicate that T3-bound TRs interfere with transactivation by TEADs via protein-protein interactions, resulting in the negative regulation of MYH7 promoter activity.
Collapse
Affiliation(s)
- Hiroyuki Iwaki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Shigekazu Sasaki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
- * E-mail:
| | - Akio Matsushita
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kenji Ohba
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hideyuki Matsunaga
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hiroko Misawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Yutaka Oki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Keiko Ishizuka
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | | | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
4
|
van Empel VPM, De Windt LJ, da Costa Martins PA. Circulating miRNAs: reflecting or affecting cardiovascular disease? Curr Hypertens Rep 2013; 14:498-509. [PMID: 22996205 DOI: 10.1007/s11906-012-0310-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
MicroRNAs are a class of small, noncoding RNAs encoded by the metazoan genome that regulate protein expression. A collection of studies point to vital roles for microRNAs in the onset and development of cardiovascular diseases. So far, microRNAs have been considered as important intracellular mediators in maintaining proper cardiac function and hemostasis, and have been proposed as potential therapeutic targets in cardiovascular disease. The recent discovery that microRNAs circulate in a stable form in many body fluids, including blood, suggests that circulating microRNAs can serve as a new generation of biomarkers for cardiovascular diseases. In this review, we summarize the findings of studies focusing on circulating microRNAs present in human blood cells or plasma/serum, where they potentially could serve as diagnostic or prognostic markers for a variety of cardiovascular pathologies, including acute myocardial infarction, heart failure, coronary artery disease, stroke, diabetes and hypertension. The significance and limitations of microRNAs as the new biomarker generation for cardiovascular disease are also discussed.
Collapse
Affiliation(s)
- Vanessa P M van Empel
- Department of Cardiology, Heart Vessel Center, Maastricht University Medical Centre, The Netherlands
| | | | | |
Collapse
|
5
|
Zacharaki P, Stephanou G, Demopoulos NA. Comparison of the aneugenic properties of nocodazole, paclitaxel and griseofulvin in vitro. Centrosome defects and alterations in protein expression profiles. J Appl Toxicol 2012; 33:869-79. [PMID: 22431130 DOI: 10.1002/jat.2745] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 02/03/2012] [Accepted: 02/03/2012] [Indexed: 11/08/2022]
Abstract
We have comparatively investigated the aneugenic activity of two anticancer drugs, nocodazole (NOC) and paclitaxel (PTX), and the antifungal griseofulvin with promising role in cancer treatment (GF), which affect microtubule dynamics in different ways. The comparison was achieved in HFFF2 human fibroblasts, MCF-7 human breast cancer cells and C2C12 mouse myoblasts, and focused on three issues: (i) induction of chromosome delay by estimation of MN frequency using CREST analysis; (ii) disturbance of spindle organization with Aurora-A/β-tubulin immunofluorescence; and (iii) alterations in the expression of Aurora-A, β- and γ-tubulin by western blotting. They induced chromosome delay, provoked metaphase arrest and promoted microtubule disorganization, reflecting their common characteristic of generating aneuploidy. In particular, NOC induced mainly monopolar metaphases, although PTX induced only multipolar metaphases. GF generated different types of abnormal metaphases, exhibiting cell specificity. Additionally, NOC decreased the expression of Aurora-A and β-tubulin, while the opposite held true for PTX and GF. γ-Tubulin expression was not modulated owing to NOC treatment, whereas PTX and GF increased γ-tubulin expression. Our findings throw a light on the manifestation of the aneugenicity of the studied compounds through centrosome proliferation/separation and protein expression, reflecting their different effects on microtubule dynamics.
Collapse
Affiliation(s)
- Polyxeni Zacharaki
- Division of Genetics, Cell and Developmental Biology, Department of Biology, University of Patras, 26 500, Patras, Greece
| | | | | |
Collapse
|
6
|
Xu J, Zhao J, Evan G, Xiao C, Cheng Y, Xiao J. Circulating microRNAs: novel biomarkers for cardiovascular diseases. J Mol Med (Berl) 2011; 90:865-75. [PMID: 22159451 DOI: 10.1007/s00109-011-0840-5] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 11/26/2011] [Accepted: 11/28/2011] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are a novel class of small, non-coding, single-stranded RNAs that negatively regulate gene expression via translational inhibition or mRNA degradation followed by protein synthesis repression. Many miRNAs are expressed in a tissue- and/or cell-specific manner and their expression patterns are reflective of underlying patho-physiologic processes. miRNAs can be detected in serum or in plasma in a remarkably stable form, making them attractive biomarkers for human diseases. This review describes the progress of identifying circulating miRNAs as novel biomarkers for diverse cardiovascular diseases, including acute myocardial infarction, heart failure, coronary artery disease, diabetes, stroke, essential hypertension, and acute pulmonary embolism. In addition, the origin and function and the different strategies to identify circulating miRNAs as novel biomarkers for cardiovascular diseases are also discussed. Rarely has an opportunity arisen to advance such new biology for the diagnosis of cardiac diseases.
Collapse
Affiliation(s)
- Jiahong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
| | | | | | | | | | | |
Collapse
|
7
|
Efthimiou M, Stephanou G, Demopoulos NA, Nikolaropoulos SS. Aneugenic potential of the anticancer drugs melphalan and chlorambucil. The involvement of apoptosis and chromosome segregation regulating proteins. J Appl Toxicol 2011; 33:537-45. [PMID: 22025197 DOI: 10.1002/jat.1743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 08/20/2011] [Accepted: 08/20/2011] [Indexed: 12/31/2022]
Abstract
Previous findings showed that the anticancer drugs p-N,N-bis(2-chloroethyl) amino-l-phenylalanine (melphalan, MEL) and p-N,N-bis(2-chloroethyl)aminophenylbutyric acid (chlorambucil, CAB) belonging to the nitrogen mustard group, in addition to their clastogenic activity, also exert aneugenic potential, nondisjunction and chromosome delay. Their aneugenic potential is mainly mediated through centrosome defects. To further investigate their aneugenicity we (a) studied whether apoptosis is a mechanism responsible for the elimination of damaged cells generated by MEL and CAB and (b) investigated if proteins that regulate chromosome segregation are involved in the modulation of their aneugenic potential. Apoptosis was studied by Annexin-V/Propidium Iodide staining and fluorescence microscopy. The involvement of apoptosis on the exclusion of cells with genetic damage and centrosome disturbances was analyzed by DAPI staining and immunofluorescence of β- and γ-tubulin in the presence of pan-caspase inhibitor. The expressions of Aurora-A, Aurora-B, survivin and γ-tubulin were studied by western blot. We found that (a) apoptosis is not the mechanism of choice for selectively eliminating cells with supernumerary centrosomes, and (b) the proteins Aurora-A, Aurora-B and survivin are involved in the modulation of MEL and CAB aneugenicity. These findings are important for the understanding of the mechanism responsible for the aneugenic activity of the anticancer drugs melphalan and chlorambucil.
Collapse
Affiliation(s)
- Maria Efthimiou
- Division of Genetics, Cell and Developmental Biology, Department of Biology, University of Patras, 26 500, Patras, Greece
| | | | | | | |
Collapse
|
8
|
|
9
|
Davis J, Westfall MV, Townsend D, Blankinship M, Herron TJ, Guerrero-Serna G, Wang W, Devaney E, Metzger JM. Designing heart performance by gene transfer. Physiol Rev 2008; 88:1567-651. [PMID: 18923190 DOI: 10.1152/physrev.00039.2007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The birth of molecular cardiology can be traced to the development and implementation of high-fidelity genetic approaches for manipulating the heart. Recombinant viral vector-based technology offers a highly effective approach to genetically engineer cardiac muscle in vitro and in vivo. This review highlights discoveries made in cardiac muscle physiology through the use of targeted viral-mediated genetic modification. Here the history of cardiac gene transfer technology and the strengths and limitations of viral and nonviral vectors for gene delivery are reviewed. A comprehensive account is given of the application of gene transfer technology for studying key cardiac muscle targets including Ca(2+) handling, the sarcomere, the cytoskeleton, and signaling molecules and their posttranslational modifications. The primary objective of this review is to provide a thorough analysis of gene transfer studies for understanding cardiac physiology in health and disease. By comparing results obtained from gene transfer with those obtained from transgenesis and biophysical and biochemical methodologies, this review provides a global view of cardiac structure-function with an eye towards future areas of research. The data presented here serve as a basis for discovery of new therapeutic targets for remediation of acquired and inherited cardiac diseases.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Hagiwara N, Yeh M, Liu A. Sox6 is required for normal fiber type differentiation of fetal skeletal muscle in mice. Dev Dyn 2007; 236:2062-76. [PMID: 17584907 DOI: 10.1002/dvdy.21223] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Sox6, a member of the Sox family of transcription factors, is highly expressed in skeletal muscle. Despite its abundant expression, the role of Sox6 in muscle development is not well understood. We hypothesize that, in fetal muscle, Sox6 functions as a repressor of slow fiber type-specific genes. In the wild-type mouse, differentiation of fast and slow fibers becomes apparent during late fetal stages (after approximately embryonic day 16). However, in the Sox6 null-p(100H) mutant mouse, all fetal muscle fibers maintain slow fiber characteristics, as evidenced by expression of the slow myosin heavy chain MyHC-beta. Knockdown of Sox6 expression in wild-type myotubes results in a significant increase in MyHC-beta expression, supporting our hypothesis. Analysis of the MyHC-beta promoter revealed a Sox consensus sequence that likely functions as a negative cis-regulatory element. Together, our results suggest that Sox6 plays a critical role in the fiber type differentiation of fetal skeletal muscle.
Collapse
Affiliation(s)
- Nobuko Hagiwara
- University of California, Davis, Division of Cardiovascular Medicine/Rowe Program in Human Genetics, Davis, California 95616, USA.
| | | | | |
Collapse
|
11
|
Chen HH, Stewart AFR. Characterization of cardiac gene promoter activity: reporter constructs and heterologous promoter studies. Methods Mol Biol 2007; 366:217-25. [PMID: 17568127 DOI: 10.1007/978-1-59745-030-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Cardiac gene promoter analysis remains an integral method in molecular cardiology and continues to provide novel insights into the transcriptional mechanisms that regulate gene expression in the myocardium. Initial studies focused on the regulated expression of contractile genes, since their transcripts are abundant and their cDNAs were among the first to be cloned. More recent studies have focused on the promoters of genes expressed at much lower levels, including those that encode ion channels, signaling proteins, and the cardiac transcription factors. The standard approach to analyze myocardial gene promoters has been to transfect reporter plasmids into cultured neonatal rat cardiac myocytes. This approach has the unique advantage of allowing the exploration of different signaling mechanisms by supplementing culture media with different agonists and inhibitors. In addition, cis-elements that control gene expression under different physiological stresses have been further characterized in the context of heterologous promoters to demonstrate their "stand-alone" functional properties in the absence of confounding influences from other cis-elements and their cognate transcription factors. Here we illustrate the characterization of cardiac gene promoter activity using reporter constructs and heterologous promoter studies in cultured cardiac myocytes.
Collapse
|
12
|
Azakie A, Lamont L, Fineman JR, He Y. Divergent transcriptional enhancer factor-1 regulates the cardiac troponin T promoter. Am J Physiol Cell Physiol 2005; 289:C1522-34. [PMID: 16049055 DOI: 10.1152/ajpcell.00126.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
MCAT elements are essential for cardiac gene expression during development. Avian transcriptional enhancer factor-1 (TEF-1) proteins are muscle-enriched and contribute to MCAT binding activities. However, direct activation of MCAT-driven promoters by TEF-1-related proteins has not been uniformly achieved. Divergent TEF (DTEF)-1 is a unique member of the TEF-1 multigene family with abundant transcripts in the heart but not in skeletal muscle. Herein we show that DTEF-1 proteins are highly expressed in the heart. Protein expression is activated at very early stages of chick embryogenesis (Hamburger-Hamilton stage 4, 16–18 h), after which DTEF-1 becomes abundant in the sinus venosus and is expressed in the trabeculated ventricular myocardium and ventricular outflow tracts. By chromatin immunoprecipitation, DTEF-1 interacts with the cardiac troponin T (cTnT) promoter in vivo. DTEF-1 also interacts with MEF- 2 by coimmunoprecipitation and independently or cooperatively (with MEF-2) trans-activates the cTnT promoter. DTEF-1 isoforms do not activate the cTnT promoter in fibroblasts or skeletal muscle. DTEF-1 expression occurs very early in chick embryogenesis (16–18 h), preceding sarcomeric protein expression, and it activates cardiac promoters. As such, DTEF-1 may be an early marker of the myocardial phenotype. DTEF-1 trans-activates the cTnT promoter in a tissue-specific fashion independent of AT-rich, MEF-2, or GATA sites. The observed spatial pattern suggests decreasing levels of expression from the cardiac inlet to the ventricular outflow tracts, which may mark a cardiogenic or differentiation pathway that parallels the direction of flow through the developing chick heart.
Collapse
Affiliation(s)
- Anthony Azakie
- Department of Surgery, Univ. of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
13
|
Koitabashi N, Arai M, Tomaru K, Takizawa T, Watanabe A, Niwano K, Yokoyama T, Wuytack F, Periasamy M, Nagai R, Kurabayashi M. Carvedilol effectively blocks oxidative stress-mediated downregulation of sarcoplasmic reticulum Ca2+-ATPase 2 gene transcription through modification of Sp1 binding. Biochem Biophys Res Commun 2005; 328:116-24. [PMID: 15670758 DOI: 10.1016/j.bbrc.2004.12.139] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Indexed: 11/16/2022]
Abstract
Carvedilol is a beta-adrenoceptor blocker and a potent antioxidant that improves cardiac function in patients with heart failure. The restoration of sarcoplasmic reticulum Ca2+-ATPase (SERCA2) gene expression may be an underlying mechanism of its beneficial effects on cardiac function. In primary cultured neonatal rat cardiac myocytes, treatment with either carvedilol or its beta-receptor inactive metabolite, BM910228, attenuated the hydrogen peroxide-mediated decrease in SERCA2 mRNA and protein levels, while metoprolol, a pure beta-blocker, had no effect. Moreover, carvedilol itself significantly enhanced SERCA2 gene transcription, suggesting that carvedilol specifically restores SERCA2 gene transcription. Site-directed mutagenesis revealed that two Sp1 sites in the SERCA2 gene promoter region mediated the response to carvedilol under oxidative stress. Further, electrophoretic mobility shift assays revealed that Sp1 and Sp3 transcription factors correlated with carvedilol-mediated changes in the promoter assays. These studies may provide a mechanistic explanation for the beneficial effects of carvedilol in heart failure.
Collapse
Affiliation(s)
- Norimichi Koitabashi
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Li CY, Zhu J, Wang JYJ. Ectopic Expression of p73α, but Not p73β, Suppresses Myogenic Differentiation. J Biol Chem 2005; 280:2159-64. [PMID: 15545283 DOI: 10.1074/jbc.m411194200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The TRP73 gene, a member of the p53 family, encodes several variants through differential splicing and use of alternative promoters. At the N terminus, two different promoters generate the full-length and the DeltaN isoforms, with or without the transactivating domain. At the C terminus, seven isoforms generated through alternative splicing have been cloned. Previous studies have demonstrated that DeltaN-p73 interferes with p73-induced apoptosis. However, there has been no evidence for functional diversity of the C-terminal p73 variants. In this study, we found that p73alpha and p73beta exerted differential effect on the differentiation of C2C12 myoblasts. Although p73beta lacked any detectable effect on differentiation, p73alpha caused a substantial delay in the expression of muscle-specific genes. In co-transfection experiments p73alpha, but not p73beta, attenuated the transcriptional activity of MyoD. Microarray-based gene profiling confirmed the protraction of MyoD-dependent gene expression in C2C12 cells stably expressing p73alpha. Notwithstanding the differential effect on differentiation, p73alpha and p73beta showed similar activity in sensitizing C2C12 myoblasts to cisplatin-induced cell death. These results demonstrated a functional diversity between the two C-terminal variants of p73 and suggested that p73alpha can regulate cellular differentiation in addition to its role in stimulating cell death.
Collapse
Affiliation(s)
- Chun-Ying Li
- Division of Biological Sciences and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0322, USA
| | | | | |
Collapse
|
15
|
Giger JM, Haddad F, Qin AX, Zeng M, Baldwin KM. Effect of unloading on type I myosin heavy chain gene regulation in rat soleus muscle. J Appl Physiol (1985) 2004; 98:1185-94. [PMID: 15591287 DOI: 10.1152/japplphysiol.01099.2004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Slow-twitch soleus, a weight-bearing hindlimb muscle, predominantly expresses the type I myosin heavy chain (MHC) isoform. However, under unloading conditions, a transition in MHC expression occurs from slow type I toward the fast-type isoforms. Transcriptional processes are believed to be involved in this adaptation. To test the hypothesis that the downregulation of MHC1 in soleus muscle following unloading is controlled through cis element(s) in the proximal region of the promoter, the MHC1 promoter was injected into soleus muscles of control rats and those subjected to 7 days of hindlimb suspension. Mutation analyses of six putative regulatory elements within the -408-bp region demonstrated that three elements, an A/T-rich, the proximal muscle-type CAT (betae3), and an E-box (-63 bp), play an important role in the basal level of MHC1 gene activity in the control soleus and function as unloading-responsive elements. Gel mobility shift assays revealed a diminished level of complex formation of the betae3 and E-box probes with nuclear extract from hindlimb suspension soleus compared with control soleus. Supershift assays indicated that transcriptional enhancer factor 1 and myogenin factors bind the betae3 and E-box elements, respectively, in the control soleus. Western blots showed that the relative concentrations of the transcriptional enhancer factor 1 and myogenin factors were significantly attenuated in the unloaded soleus compared with the control muscle. We conclude that the downregulation of MHC1 in response to unloading is due, in part, to a significant decrease in the concentration of these transcription factors available for binding the positive regulatory elements.
Collapse
Affiliation(s)
- Julia M Giger
- Dept. of Physiology and Biophysics, Univ. of California-Irvine, D-346, Med Sci I, Irvine, CA 92697, USA
| | | | | | | | | |
Collapse
|
16
|
Allegra S, Li J, Saez J, Langlois D. Terminal differentiation of Sol 8 myoblasts is retarded by a transforming growth factor-beta autocrine regulatory loop. Biochem J 2004; 381:429-36. [PMID: 15056073 PMCID: PMC1133849 DOI: 10.1042/bj20031008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2003] [Revised: 03/24/2004] [Accepted: 03/31/2004] [Indexed: 11/17/2022]
Abstract
In DM (differentiation medium), Sol 8 myoblasts spontaneously form myotubes and express the betaMHC (beta-myosin heavy chain), their main marker of terminal differentiation. This marker is detectable at 24 h, and increases up to 72 h. Our aim was to define temporal effects of TGFbeta (transforming growth factor beta) on betaMHC expression in Sol 8 cells. TGFbeta1 (1 ng/ml) added at time zero to DM decreased MyoD expression and completely inhibited betaMHC expression in Sol 8 cells. This inhibition of betaMHC expression was progressively lost when TGFbeta1 was added from 8 to 34 h. After 34 h, the cells were irreversibly differentiated, and TGFbeta1 did not inhibit betaMHC accumulation any longer. Two independent approaches showed that a TGFbeta autocrine regulatory loop retarded and partially impaired Sol 8 cell terminal differentiation. First, permanent immunoneutralization of the active TGFbetas released by the cells into DM increased betaMHC levels at 72 h compared with controls. Secondly, a dominant-negative mutant of the TGFbeta type II receptor was overexpressed in Sol 8 cells under the control of the betaMHC promoter. Both the dominant-negative receptor and the betaMHC gene were expressed after 24 h in DM. The delayed blocking of the TGFbeta signalling pathway by the dominant-negative receptor was as effective as permanent immunoneutralization to promote betaMHC expression. To conclude, TGFbeta inhibits Sol 8 cell terminal differentiation within a narrow time interval (24-34 h) that coincides with the onset of betaMHC expression.
Collapse
Key Words
- autocrine regulatory loop
- immunoneutralization
- sol 8 myoblasts
- terminal differentiation
- transforming growth factor β (tgfβ)
- dominant-negative tgfβ type ii receptor
- cmv, cytomegalovirus
- dm, differentiation medium
- dmem, dulbecco's modified eagle's medium
- egfp, enhanced green fluorescent protein
- fbs, fetal bovine serum
- gm, growth medium
- hs, horse serum
- βmhc, β-myosin heavy chain
- mrf, myogenic regulatory factor
- tgfβ, transforming growth factor β
- tβr(i/ii), type i/ii tgfβ receptor
- wt, wild-type
Collapse
Affiliation(s)
- Séverine Allegra
- UMR 369 INSERM/UCBL and IFR 62 Laënnec, Faculté de médecine, R.T.H. Laënnec, 7 rue G. Paradin, 69372 Lyon, Cedex 08, France
| | - Jacques Yuan Li
- UMR 369 INSERM/UCBL and IFR 62 Laënnec, Faculté de médecine, R.T.H. Laënnec, 7 rue G. Paradin, 69372 Lyon, Cedex 08, France
| | - José Maria Saez
- UMR 369 INSERM/UCBL and IFR 62 Laënnec, Faculté de médecine, R.T.H. Laënnec, 7 rue G. Paradin, 69372 Lyon, Cedex 08, France
| | - Dominique Langlois
- UMR 369 INSERM/UCBL and IFR 62 Laënnec, Faculté de médecine, R.T.H. Laënnec, 7 rue G. Paradin, 69372 Lyon, Cedex 08, France
| |
Collapse
|
17
|
Shie JL, Wu G, Wu J, Liu FF, Laham RJ, Oettgen P, Li J. RTEF-1, a Novel Transcriptional Stimulator of Vascular Endothelial Growth Factor in Hypoxic Endothelial Cells. J Biol Chem 2004; 279:25010-6. [PMID: 15073166 DOI: 10.1074/jbc.m403103200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic growth factor known to be up-regulated in ischemic heart and hypoxic endothelial cells. However, the transcriptional regulation of VEGF in hypoxia-induced angiogenesis is not fully understood. Transcriptional enhancer factor-1 (TEF-1) is a transcriptional factor family that can regulate many genes expressed in cardiac and skeletal muscle cells by binding to myocyte-specific chloramphenicol acetyltransferase heptamer elements in the promoters of these genes. In this study, we demonstrated that related TEF-1 (RTEF-1), a member of the TEF-1 family, is up-regulated in hypoxic endothelial cells. Overexpression of RTEF-1 increases VEGF promoter activity and VEGF expression. Sequential deletion and site-directed mutation analyses of the VEGF promoter demonstrated that a GC-rich region containing four Sp1 response elements, located between -114 and -50, was essential for RTEF-1 function. This region is beyond the hypoxia-inducible factor-1alpha binding site and does not consist of M-CAT-related elements. By electrophoretic mobility shift assay, RTEF-1 was found to interact with the first Sp1 residue (-97 to -87) of the four consecutive Sp1 elements. Binding activity of RTEF-1 to VEGF promoter is also confirmed by chromatin immunoprecipitation. In addition, induction of VEGF promoter activity by RTEF-1 results in an increase of angiogenic processes including endothelial cells proliferation and vascular structure formation. These results indicate that RTEF-1 acts as a transcriptional stimulator of VEGF by regulating VEGF promoter activity through binding to Sp1 site. In addition, RTEF-1-induced VEGF promoter activity was enhanced in a hypoxic condition, indicating that RTEF-1 may play an important role in the regulation of VEGF under hypoxia.
Collapse
Affiliation(s)
- Jue-Lon Shie
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Moreno H, Serrano AL, Santalucía T, Gumá A, Cantó C, Brand NJ, Palacin M, Schiaffino S, Zorzano A. Differential regulation of the muscle-specific GLUT4 enhancer in regenerating and adult skeletal muscle. J Biol Chem 2003; 278:40557-64. [PMID: 12893821 DOI: 10.1074/jbc.m306609200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have reported a novel functional co-operation among MyoD, myocyte enhancer factor-2 (MEF2), and the thyroid hormone receptor in a muscle-specific enhancer of the rat GLUT4 gene in muscle cells. Here, we demonstrate that the muscle-specific enhancer of the GLUT4 gene operates in skeletal muscle and is muscle fiber-dependent and innervation-independent. Under normal conditions, both in soleus and in extensor digitorum longus muscles, the activity of the enhancer required the integrity of the MEF2-binding site. Cancellation of the binding site of thyroid hormone receptor enhanced its activity, suggesting an inhibitory role. Muscle regeneration of the soleus and extensor digitorum longus muscles caused a marked induction of GLUT4 and stimulation of the enhancer activity, which was independent of innervation. During muscle regeneration, the enhancer activity was markedly inhibited by cancellation of the binding sites of MEF2, MyoD, or thyroid hormone receptors. Different MEF2 isoforms expressed in skeletal muscle (MEF2A, MEF2C, and MEF2D) and all members of the MyoD family had the capacity to participate in the activity of the GLUT4 enhancer as assessed by transient transfection in cultured cells. Our data indicate that the GLUT4 enhancer operates in muscle fibers and its activity contributes to the differences in GLUT4 gene expression between oxidative and glycolytic muscle fibers and to the GLUT4 up-regulation that occurs during muscle regeneration. The activity of the enhancer is maintained in adult muscle by MEF2, whereas during regeneration the operation of the enhancer depends on MEF2, myogenic transcription factors of the MyoD family, and thyroid hormone receptors.
Collapse
Affiliation(s)
- Horacio Moreno
- Parc Científic de Barcelona and Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Avda. Diagonal 645, E-08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Huey KA, Haddad F, Qin AX, Baldwin KM. Transcriptional regulation of the type I myosin heavy chain gene in denervated rat soleus. Am J Physiol Cell Physiol 2003; 284:C738-48. [PMID: 12444021 DOI: 10.1152/ajpcell.00389.2002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Denervation (DEN) of rat soleus is associated with a decreased expression of slow type I myosin heavy chain (MHC) and an increased expression of the faster MHC isoforms. The molecular mechanisms behind these shifts remain unclear. We first investigated endogenous transcriptional activity of the type I MHC gene in normal and denervated soleus muscles via pre-mRNA analysis. Our results suggest that the type I MHC gene is regulated via transcriptional processes in the denervated soleus. Deletion and mutational analysis of the rat type I MHC promoter was then used to identify cis elements or regions of the promoter involved in this response. DEN significantly decreased in vivo activity of the -3,500, -2,500, -914, -408, -299, and -215 bp type I MHC promoters, relative to the alpha-skeletal actin promoter. In contrast, normalized -171 promoter activity was unchanged. Mutation of the betae3 element (-214/-190) in the -215 promoter and deletion of this element (-171 promoter) blunted type I downregulation with DEN. In contrast, betae3 mutation in the -408 promoters was not effective in attenuating the DEN response, suggesting the existence of additional DEN-responsive sites between -408 and -215. Western blotting and gel mobility supershift assays demonstrated decreased expression and DNA binding of transcription enhancer factor 1 (TEF-1) with DEN, suggesting that this decrease may contribute to type I MHC downregulation in denervated muscle.
Collapse
Affiliation(s)
- K A Huey
- Department of Physiology and Biophysics, University of California, Irvine 92697, USA
| | | | | | | |
Collapse
|
20
|
Tsika RW, McCarthy J, Karasseva N, Ou Y, Tsika GL. Divergence in species and regulatory role of beta -myosin heavy chain proximal promoter muscle-CAT elements. Am J Physiol Cell Physiol 2002; 283:C1761-75. [PMID: 12388056 DOI: 10.1152/ajpcell.00278.2002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We examined the functional role of distinct muscle-CAT (MCAT) elements during non-weight-bearing (NWB) regulation of a wild-type 293-base pair beta-myosin heavy chain (beta MyHC) transgene. Electrophoretic mobility shift assays (EMSA) revealed decreased NTEF-1, poly(ADP-ribose) polymerase, and Max binding at the human distal MCAT element when using NWB soleus vs. control soleus nuclear extract. Compared with the wild-type transgene, expression assays revealed that distal MCAT element mutation decreased basal transgene expression, which was decreased further in response to NWB. EMSA analysis of the human proximal MCAT (pMCAT) element revealed low levels of NTEF-1 binding that did not differ between control and NWB extract, whereas the rat pMCAT element displayed robust NTEF-1 binding that decreased when using NWB soleus extracts. Differences in binding between human and rat pMCAT elements were consistent whether using rat or mouse nuclear extract or in vitro synthesized human TEF-1 proteins. Our results provide the first evidence that 1) different binding properties and likely regulatory functions are served by the human and rat pMCAT elements, and 2) previously unrecognized beta MyHC proximal promoter elements contribute to NWB regulation.
Collapse
Affiliation(s)
- Richard W Tsika
- Department of Biochemistry, School of Medicine, University of Missouri-Columbia, Columbia, Missouri 65211, USA.
| | | | | | | | | |
Collapse
|
21
|
Willoughby DS, Nelson MJ. Myosin heavy-chain mRNA expression after a single session of heavy-resistance exercise. Med Sci Sports Exerc 2002; 34:1262-9. [PMID: 12165680 DOI: 10.1097/00005768-200208000-00006] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE This study determined the effects of a single session of heavy-resistance exercise on myosin heavy-chain (MHC) mRNA expression, along with the expression of the transcription factors Myo-D, myogenin, and Id-1. METHODS Four male subjects participated in a control group (CON) and seven in a resistance-exercise group (REX). REX performed one resistance-exercise session employing three sets of 8-10 repetitions at 75-80% one-repetition maximum on the squat, leg press, and leg extension. Vastus lateralis biopsies were obtained pre, post, and at 6 h postexercise. RESULTS In regard to CON, no significant differences were located for any criterion variable (P > 0.05). For REX, elevations of 38.19%, 45.61%, and 74.24% (P < 0.05), respectively, occurred at 6 h-post for Type I, IIa, and IIx MHC mRNA. Myo-D and myogenin mRNA were elevated 27.28% and 23.58% postexercise (P < 0.05), respectively, but no change was observed in Id-1 mRNA. Elevations at 6 h-post of 46.85% and 46.41% (P < 0.05), respectively, occurred for Myo-D and myogenin mRNA with no change in Id-1. Myo-D and myogenin protein increased 57.91% and 52.30%, respectively, postexercise and 317.56% and 254.08 at 6 h-post (P < 0.05), whereas no change was noted for Id-1. Myofibrillar protein was elevated 84.52% at 6 h-post (P < 0.05). Type I and IIa MHC mRNA at 6 h-post were correlated with myogenin mRNA and protein postexercise and 6 h-post, whereas Type IIx at 6 h-post was correlated with Myo-D mRNA and protein postexercise and 6 h-post (P < 0.05). CONCLUSIONS These results indicate that the mRNA expression of all three MHC isoforms is up-regulated after a single session of heavy-resistance exercise and that Myo-D and myogenin seem to play a role in MHC isoform gene expression.
Collapse
Affiliation(s)
- Darryn S Willoughby
- Department of Kinesiology, Texas Christian University, TCU Box 297730, Fort Worth, TX 76129, USA.
| | | |
Collapse
|
22
|
Maeda T, Gupta MP, Stewart AFR. TEF-1 and MEF2 transcription factors interact to regulate muscle-specific promoters. Biochem Biophys Res Commun 2002; 294:791-7. [PMID: 12061776 DOI: 10.1016/s0006-291x(02)00556-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Many muscle-specific genes are regulated by transcriptional enhancer factor-1 (TEF-1), serum response factor (SRF), and myocyte enhancer factor-2 (MEF2) transcription factors. TEF-1 interacts with the MADS domain of SRF and together SRF and TEF-1 co-activate the skeletal alpha-actin promoter. MEF2 factors also contain a MADS domain with 50% amino acid identity to the SRF MADS domain. Because of this sequence divergence, some SRF co-factors do not interact with MEF2. To demonstrate that TEF-1 factors could also interact with MEF2 through its MADS domain, we used co-immunoprecipitation and GST pull-down assays in vitro and a mammalian two-hybrid assay in vivo. The MADS domain was not sufficient for MEF2 interaction with TEF-1, because additional sequences in the activation domains of both proteins were required for in vivo association. The physiological significance of this interaction was also demonstrated by transient transfection assays using muscle-specific promoters. Our results suggest that by their interaction with MEF2 factors, TEF-1 factors can control MEF2-dependent muscle-specific gene expression.
Collapse
Affiliation(s)
- Tomoji Maeda
- Cardiovascular Institute, School of Medicine, University of Pittsburgh, BST 1704.3, PA 15213, USA
| | | | | |
Collapse
|
23
|
Giger JM, Haddad F, Qin AX, Baldwin KM. Functional overload increases beta-MHC promoter activity in rodent fast muscle via the proximal MCAT (betae3) site. Am J Physiol Cell Physiol 2002; 282:C518-27. [PMID: 11832337 DOI: 10.1152/ajpcell.00444.2001] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Functional overload (OL) of the rat plantaris muscle by the removal of synergistic muscles induces a shift in the myosin heavy chain (MHC) isoform expression profile from the fast isoforms toward the slow type I, or, beta-MHC isoform. Different length rat beta-MHC promoters were linked to a firefly luciferase reporter gene and injected in control and OL plantaris muscles. Reporter activities of -3,500, -914, -408, and -215 bp promoters increased in response to 1 wk of OL. The smallest -171 bp promoter was not responsive to OL. Mutation analyses of putative regulatory elements within the -171 and -408 bp region were performed. The -408 bp promoters containing mutations of the betae1, distal muscle CAT (MCAT; betae2), CACC, or A/T-rich (GATA), were still responsive to OL. Only the proximal MCAT (betae3) mutation abolished the OL response. Gel mobility shift assays revealed a significantly higher level of complex formation of the betae3 probe with nuclear protein from OL plantaris compared with control plantaris. These results suggest that the betae3 site functions as a putative OL-responsive element in the rat beta-MHC gene promoter.
Collapse
Affiliation(s)
- Julia M Giger
- Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA 92697, USA
| | | | | | | |
Collapse
|
24
|
Huey KA, Roy RR, Haddad F, Edgerton VR, Baldwin KM. Transcriptional regulation of the type I myosin heavy chain promoter in inactive rat soleus. Am J Physiol Cell Physiol 2002; 282:C528-37. [PMID: 11832338 DOI: 10.1152/ajpcell.00355.2001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic muscle inactivity with spinal cord isolation (SI) decreases expression of slow type I myosin heavy chain (MHC) while increasing expression of the faster MHC isoforms, primarily IIx. The purpose of this study was to determine whether type I MHC downregulation in the soleus muscle of SI rats is regulated transcriptionally and to identify cis-acting elements or regions of the rat type I MHC gene promoter involved in this response. One week of SI significantly decreased in vivo activity of the -3500-, -408-, -299-, -215-, and -171-bp type I MHC promoters. The activity of all tested deletions of the type I MHC promoter, relative to the human skeletal alpha-actin promoter, were significantly reduced in the SI soleus, except activity of the -171-bp promoter, which increased. Mutation of the betae3 element (-214/-190 bp) in the -215- and -408-bp promoters and deletion of this element (-171-bp promoter) attenuated type I downregulation with SI. Gel mobility shift assays demonstrated a decrease in transcription enhancer factor-1 binding to the betae3 element with SI, despite an increase in total binding to this region. These results demonstrate that type I MHC downregulation with SI is transcriptionally regulated and suggest that interactions between transcription enhancer factor-1 and the betae3 element are likely involved in this response.
Collapse
Affiliation(s)
- K A Huey
- Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA 92697, USA
| | | | | | | | | |
Collapse
|
25
|
Allen DL, Sartorius CA, Sycuro LK, Leinwand LA. Different pathways regulate expression of the skeletal myosin heavy chain genes. J Biol Chem 2001; 276:43524-33. [PMID: 11551968 DOI: 10.1074/jbc.m108017200] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian skeletal muscles are a mosaic of different fiber types largely defined by differential myosin heavy chain (MyHC) expression. Little is known about the molecular mechanisms regulating expression of the MyHC gene family members in different fiber types. In this work, we identified several cis- and trans-elements that regulate expression of the three adult fast MyHC genes. Despite multiple DNA-binding motifs for well characterized muscle transcription factors upstream of all three fast MyHC genes, expression of MyoD/Myf-5, calcineurin, or NFAT3 had different effects on the three promoters. MyoD or Myf-5 overexpression preferentially activated the IIb promoter, whereas NFAT or activated calcineurin overexpression preferentially activated the IIa promoter. Calcineurin had a 50-100-fold stimulatory effect on the IIa promoter, and the known downstream effectors of calcineurin (myocyte enhancer factor-2 and NFAT) cannot completely account for this activation. Finally, we identified two elements critical for regulating MyHC-IId/x expression: a 130-base pair enhancer element and a CArG-like element that inhibited IId/x promoter activity in vitro. Thus, we have found specific regulatory pathways that are distinct for the three adult fast MyHC genes. These elements are logical candidates for fiber-specific control of skeletal muscle gene expression in vivo.
Collapse
Affiliation(s)
- D L Allen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309-0347, USA
| | | | | | | |
Collapse
|
26
|
Santalucía T, Moreno H, Palacín M, Yacoub MH, Brand NJ, Zorzano A. A novel functional co-operation between MyoD, MEF2 and TRalpha1 is sufficient for the induction of GLUT4 gene transcription. J Mol Biol 2001; 314:195-204. [PMID: 11718554 DOI: 10.1006/jmbi.2001.5091] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We report tripartite co-operation between MyoD, myocyte enhancer factor-2 (MEF2) and the thyroid hormone receptor (TRalpha1) that takes place in the context of an 82-bp muscle-specific enhancer in the rat insulin-responsive glucose transporter (GLUT4) gene that is active in both cardiac and skeletal muscle. In the L6E9 skeletal muscle cell line and in 10T1/2 fibroblasts, a powerful synergistic activation of the GLUT4 enhancer relied on the over-expression of MyoD, MEF2 and TRalpha1 and the integrity of their respective binding sites, and occurred when linked to either a heterologous promoter or in the context of the native GLUT4 promoter. In cardiac myocytes, enhancer activity was dependent on the binding sites for MEF2 and TRalpha1. Furthermore, we show that in 10T1/2 fibroblasts, the forced expression of MyoD, MEF2 and TRalpha1 induced the expression of the endogenous, otherwise silent, GLUT4 gene. In all, our results indicate a novel functional co-operation between these three factors which is required for full activation of GLUT4 transcription.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites
- Cell Line
- Cells, Cultured
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Enhancer Elements, Genetic/genetics
- Fibroblasts/metabolism
- Genes, Reporter/genetics
- Glucose Transporter Type 4
- Humans
- MEF2 Transcription Factors
- Mice
- Monosaccharide Transport Proteins/genetics
- Monosaccharide Transport Proteins/metabolism
- Muscle Proteins
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- MyoD Protein/genetics
- MyoD Protein/metabolism
- Myocardium/cytology
- Myocardium/metabolism
- Myogenic Regulatory Factors
- Precipitin Tests
- Promoter Regions, Genetic/genetics
- Protein Binding
- Rats
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Thyroid Hormone
- Response Elements/genetics
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic/genetics
- Transcriptional Activation
- Transfection
- Troponin I/genetics
Collapse
Affiliation(s)
- T Santalucía
- Department of Cardiothoracic Surgery, National Heart and Lung Institute, Faculty of Medicine, Imperial College of Science, Technology and Medicine, London, SW3 6LY, UK
| | | | | | | | | | | |
Collapse
|
27
|
Horton MJ, Brandon CA, Morris TJ, Braun TW, Yaw KM, Sciote JJ. Abundant expression of myosin heavy-chain IIB RNA in a subset of human masseter muscle fibres. Arch Oral Biol 2001; 46:1039-50. [PMID: 11543711 PMCID: PMC3848045 DOI: 10.1016/s0003-9969(01)00066-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Type IIB fast fibres are typically demonstrated in human skeletal muscle by histochemical staining for the ATPase activity of myosin heavy-chain (MyHC) isoforms. However, the monoclonal antibody specific for the mammalian IIB isoform does not detect MyHC IIB protein in man and MyHC IIX RNA is found in histochemically identified IIB fibres, suggesting that the IIB protein isoform may not be present in man; if this is not so, jaw-closing muscles, which express a diversity of isoforms, are likely candidates for their presence. ATPase histochemistry, immunohistochemistry polyacrylamide gel electrophoresis and in situ hybridization, which included a MyHC IIB-specific mRNA riboprobe, were used to compare the composition and RNA expression of MyHC isoforms in a human jaw-closing muscle, the masseter, an upper limb muscle, the triceps, an abdominal muscle, the external oblique, and a lower limb muscle, the gastrocnemius. The external oblique contained a mixture of histochemically defined type I, IIA and IIB fibres distributed in a mosaic pattern, while the triceps and gastrocnemius contained only type I and IIA fibres. Typical of limb muscle fibres, the MyHC I-specific mRNA probes hybridized with histochemically defined type I fibres, the IIA-specific probes with type IIA fibres and the IIX-specific probes with type IIB fibres. The MyHC IIB mRNA probe hybridized only with a few histochemically defined type I fibres in the sample from the external oblique; in addition to this IIB message, these fibres also expressed RNAs for MyHC I, IIA and IIX. MyHC IIB RNA was abundantly expressed in histochemical and immunohistochemical type IIA fibres of the masseter, together with transcripts for IIA and in some cases IIX. No MyHC IIB protein was detected in fibres and extracts of either the external oblique or masseter by immunohistochemistry, immunoblotting and electrophoresis. Thus, IIB RNA, but not protein, was found in the fibres of two different human skeletal muscles. It is believed this is the first report of the substantial expression of IIB mRNA in man as demonstrated in a subset of masseter fibres, but rarely in limb muscle, and in only a few fibres of the external oblique. These findings provide further evidence for the complexity of myosin gene expression, especially in jaw-closing muscles.
Collapse
MESH Headings
- Abdominal Muscles/metabolism
- Abdominal Muscles/ultrastructure
- Adenosine Triphosphatases
- Adolescent
- Adult
- Electrophoresis, Polyacrylamide Gel
- Female
- Gene Expression
- Humans
- Immunoblotting
- In Situ Hybridization
- Male
- Masseter Muscle/metabolism
- Masseter Muscle/ultrastructure
- Middle Aged
- Muscle Fibers, Fast-Twitch/metabolism
- Muscle Fibers, Fast-Twitch/ultrastructure
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/ultrastructure
- Muscle Fibers, Slow-Twitch/metabolism
- Muscle Fibers, Slow-Twitch/ultrastructure
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- Myosin Heavy Chains/analysis
- Myosin Heavy Chains/genetics
- Myosin Type I/analysis
- Myosin Type I/genetics
- Myosin Type II/analysis
- Myosin Type II/genetics
- Nonmuscle Myosin Type IIA/analysis
- Nonmuscle Myosin Type IIA/genetics
- Nonmuscle Myosin Type IIB/analysis
- Nonmuscle Myosin Type IIB/genetics
- Phenotype
- Protein Isoforms/analysis
- Protein Isoforms/genetics
- RNA Probes
- RNA, Messenger/analysis
- RNA, Messenger/genetics
Collapse
Affiliation(s)
- Michael J. Horton
- Department of Orthodontics, School of Dental Medicine, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA 15261, USA
| | - Carla A. Brandon
- Department of Orthodontics, School of Dental Medicine, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA 15261, USA
| | - Terence J. Morris
- Department of Orthodontics, School of Dental Medicine, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA 15261, USA
| | - Thomas W. Braun
- Department of Oral Surgery, School of Dental Medicine, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA 15261, USA
| | - Kenneth M. Yaw
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, 3471 Fifth Ave., Pittsburgh, PA 15213, USA
| | - James J. Sciote
- Department of Orthodontics, School of Dental Medicine, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA 15261, USA
- Corresponding author. Tel.: +1-412-648-8419; fax: +1-412-648-8817. (J.J. Sciote)
| |
Collapse
|
28
|
Campos-Caro A, Carrasco-Serrano C, Valor LM, Ballesta JJ, Criado M. Activity of the nicotinic acetylcholine receptor alpha5 and alpha7 subunit promoters in muscle cells. DNA Cell Biol 2001; 20:657-66. [PMID: 11749724 DOI: 10.1089/104454901753340640] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The acetylcholine receptor alpha5 and alpha7 subunits are components of different nicotinic receptor subtypes expressed in the nervous system. However, they are also present in non-neuronal tissues. We have detected alpha5 and alpha7 transcripts in mouse C2C12 muscle cells. Moreover, on differentiation of myoblasts into myotubes, the amount of alpha7 transcripts increased significantly, whereas alpha5 remained unchanged. In order to explore how the expression of these neuronal genes is regulated in muscle, we have characterized their promoter activities. Deletion and mutagenesis analysis with transfected reporter genes showed that transcriptional activity was controlled by regulatory elements also operative in neuronal-like cells. Thus, the activity of the alpha5 subunit core promoter decreased to approximately 50% on alteration of one, two, or three of the five Sp1 binding sites present in this region and was almost abolished when four or five sites were mutated simultaneously. In the case of the alpha7 subunit promoter, the upstream stimulatory factor and the early growth response gene transcription factor were involved in regulating its transcriptional activity. In addition, the alpha7 promoter was activated during the differentiation process, in a mechanism partially dependent on the mentioned factors.
Collapse
Affiliation(s)
- A Campos-Caro
- Instituto de Neurociencias, Centro Mixto CSIC-Universidad Miguel Hernández, Alicante, Spain
| | | | | | | | | |
Collapse
|
29
|
Hiroi Y, Hiroi J, Kudoh S, Yazaki Y, Nagai R, Komuro I. Two distinct mechanisms of angiotensin II-induced negative regulation of the mitogen-activated protein kinases in cultured cardiac myocytes. Hypertens Res 2001; 24:385-94. [PMID: 11510751 DOI: 10.1291/hypres.24.385] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Increasing evidence has suggested that mitogen-activated protein kinases (MAPKs) play important roles in the development of cardiac hypertrophy. We and others have reported that the activity of MAPKs is tightly regulated by angiotensin II (Ang II) in cardiac myocytes. In the present study, we determined the molecular mechanism of Ang II-induced inactivation of MAPKs in rat neonatal cardiac myocytes. Ang II increased MAPK phosphatase 1 (MKP-1) gene expressions within 10 min. Levels of MKP-1 transcripts peaked at 30 min and gradually decreased thereafter. The increase in MKP-1 mRNA levels was Ang II-concentration dependent. An Ang II type 1 receptor (AT1)-specific antagonist, CV-11974, completely suppressed the Ang II-induced increase in MKP-1 gene expression, while a type 2 receptor (AT2)-specific antagonist, PD-123319, had no significant effects. Induction of MKP-1 gene expressions by Ang II was inhibited by pretreatment with an intracellular Ca2+ chelator, BAPTA-AM, or with the protein kinase C inhibitors, H-7 and Calphostin C. Phorbol ester and Ca2+ ionophore both significantly increased MKP-1 mRNA levels and showed synergistic action. Overexpression of MKP-1 cDNA blocked the Ang II-induced increase in expressions of immediate early response genes. In addition, Ang II-induced MAPK activation was significantly inhibited by pretreatment with CV-11974, but significantly enhanced by pretreatment with PD-123319. Addition of the AT2 agonist, CGP42112A, reduced basal MAPK activities, and pretreatment with PD-123319 abolished MAPK inactivation by CGP42112A. In conclusion, these observations suggest that Ang II negatively regulates MAPKs through AT1 receptors by increasing MKP-1 mRNA levels and through AT2 receptors by unknown mechanisms.
Collapse
Affiliation(s)
- Y Hiroi
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Wright CE, Bodell PW, Haddad F, Qin AX, Baldwin KM. In vivo regulation of the beta-myosin heavy chain gene in hypertensive rodent heart. Am J Physiol Cell Physiol 2001; 280:C1262-76. [PMID: 11287340 DOI: 10.1152/ajpcell.2001.280.5.c1262] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The main goal of this study was to examine the transcriptional activity of different-length beta-myosin heavy chain (beta-MHC) promoters in the hypertensive rodent heart using the direct gene transfer approach. A hypertensive state was induced by abdominal aortic constriction (AbCon) sufficient to elevate mean arterial pressure by approximately 45% relative to control. Results show that beta-MHC promoter activity of all tested wild-type constructs, i.e., -3500, -408, -299, -215, -171, and -71 bp, was significantly increased in AbCon hearts. In the normal control hearts, expression of the -71-bp construct was comparable to that of the promoterless vector, but its induction by AbCon was comparable to that of the other constructs. Additional results, based on mutation analysis and DNA gel mobility shift assays targeting betae1, betae2, GATA, and betae3 elements, show that these previously defined cis-elements in the proximal promoter are indeed involved in maintaining basal promoter activity; however, none of these elements, either individually or collectively, appear to be major players in mediating the hypertension response of the beta-MHC gene. Collectively, these results indicate that three separate regions on the beta-MHC promoter are involved in the induction of the gene in response to hypertension: 1) a distal region between -408 and -3500 bp, 2) a proximal region between -299 and -215 bp, and 3) a basal region within -71 bp of the transcription start site. Future research needs to further characterize these responsive regions to more fully delineate beta-MHC transcriptional regulation in response to pressure overload.
Collapse
Affiliation(s)
- C E Wright
- Department of Physiology and Biophysics, University of California, Irvine, California 92697, USA
| | | | | | | | | |
Collapse
|
31
|
|
32
|
Abstract
The alpha- and beta-myosin genes extend over 51 kb on chromosome 14 in human and 11 in mouse separated by about 4.5 kb of intergenic sequence. They are located in tandem in the order of their expression during development. Transcription of each gene is independently controlled but coordinately regulated. During each embryogenesis, the beta-MHC gene is expressed as part of the cardiac myogenic program under the control of NKX-2.5, MEF-2C, and GATA-4/5/6. After birth, thyroid hormone induces expression of alpha-MHC mRNA and inhibits expression of the beta-MHC gene. While a large number of physiological stimuli are capable of modifying this basic paradigm, thyroid hormone is required for expression of alpha-MHC in ventricular muscle. The positive TRE for T(3)-stimulation of alpha-MHC is an imperfect direct repeat located in the proximal promoter of the gene. The negative TRE for the beta-MHC gene is probably a binding half-site that is located adjacent to the TATA box. Binding of TEF-1 to a strong positive element in the proximal promoter is important in basal expression of beta-MHC gene and in the response to alpha(1)-adrenergic stimulation. The beta-MHC gene also is induced together with several other "fetal" genes during cardiac hypertrophy by a mechanism involving Ca(2+)-mediated activation of calcineurin and NF-AT3. Upon activation, NF-AT3 translocates to the nucleus and interacts with GATA-4 to stimulate beta-MHC expression. Changes in chromatin structure mediated by the association of histone acetylases and deacetylases with transcription factors are essential in regulating cell-specific expression of MHC genes.
Collapse
Affiliation(s)
- E Morkin
- Departments of Medicine, Physiology, Pharmacology, and the Sarver Heart Center, University of Arizona College of Medicine, Tucson, Arizona 85724, USA
| |
Collapse
|
33
|
Giger JM, Haddad F, Qin AX, Baldwin KM. In vivo regulation of the beta-myosin heavy chain gene in soleus muscle of suspended and weight-bearing rats. Am J Physiol Cell Physiol 2000; 278:C1153-61. [PMID: 10837343 DOI: 10.1152/ajpcell.2000.278.6.c1153] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the weight-bearing hindlimb soleus muscle of the rat, approximately 90% of muscle fibers express the beta-myosin heavy chain (beta-MHC) isoform protein. Hindlimb suspension (HS) causes the MHC isoform population to shift from beta toward the fast MHC isoforms. Our aim was to establish a model to test the hypothesis that this shift in expression is transcriptionally regulated through specific cis elements of the beta-MHC promoter. With the use of a direct gene transfer approach, we determined the activity of different length beta-MHC promoter fragments, linked to a firefly luciferase reporter gene, in soleus muscle of control and HS rats. In weight-bearing rats, the relative luciferase activity of the longest beta-promoter fragment (-3500 bp) was threefold higher than the shorter promoter constructs, which suggests that an enhancer sequence is present in the upstream promoter region. After 1 wk of HS, the reporter activities of the -3500-, -914-, and -408-bp promoter constructs were significantly reduced ( approximately 40%), compared with the control muscles. However, using the -215-bp construct, no differences in promoter activity were observed between HS and control muscles, which indicates that the response to HS in the rodent appears to be regulated within the -408 and -215 bp of the promoter.
Collapse
Affiliation(s)
- J M Giger
- Department of Physiology and Biophysics, University of California, Irvine 92697, USA
| | | | | | | |
Collapse
|
34
|
Zhu W, Zou Y, Shiojima I, Kudoh S, Aikawa R, Hayashi D, Mizukami M, Toko H, Shibasaki F, Yazaki Y, Nagai R, Komuro I. Ca2+/calmodulin-dependent kinase II and calcineurin play critical roles in endothelin-1-induced cardiomyocyte hypertrophy. J Biol Chem 2000; 275:15239-45. [PMID: 10809760 DOI: 10.1074/jbc.275.20.15239] [Citation(s) in RCA: 123] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Endothelin-1 (ET-1) induces cardiac hypertrophy. Because Ca(2+) is a major second messenger of ET-1, the role of Ca(2+) in ET-1-induced hypertrophic responses in cultured cardiac myocytes of neonatal rats was examined. ET-1 activated the promoter of the beta-type myosin heavy chain gene (beta-MHC) (-354 to +34 base pairs) by about 4-fold. This activation was inhibited by chelation of Ca(2+) and the blocking of protein kinase C activity. Similarly, the beta-MHC promoter was activated by Ca(2+) ionophores and a protein kinase C activator. beta-MHC promoter activation induced by ET-1 was suppressed by pretreatment with the calmodulin inhibitor, W7, the Ca(2+)/calmodulin-dependent kinase II (CaMKII) inhibitor, KN62, and the calcineurin inhibitor, cyclosporin A. beta-MHC promoter activation by ET-1 was also attenuated by overexpression of dominant-negative mutants of CaMKII and calcineurin. ET-1 increased the activity of CaMKII and calcineurin in cardiac myocytes. Pretreatment with KN62 and cyclosporin A strongly suppressed ET-1-induced increases in [(3)H]phenylalanine uptake and in cell size. These results suggest that Ca(2+) plays a critical role in ET-1-induced cardiomyocyte hypertrophy by activating CaMKII- and calcineurin-dependent pathways.
Collapse
Affiliation(s)
- W Zhu
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Schreiber KL, Calderone A, Rindt H. Distant upstream regulatory domains direct high levels of beta -myosin heavy chain gene expression in differentiated embryonic stem cells. J Mol Cell Cardiol 2000; 32:585-98. [PMID: 10756116 DOI: 10.1006/jmcc.2000.1100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Eukaryotic gene transcription takes place in the context of chromatin. In order to study the expression of the beta -myosin heavy chain (MyHC) gene in its appropriate cardiac environment in vitro, embryonic stem cell lines were generated and induced to differentiate into the cardiac lineage. We show that the upstream region of the beta -MyHC gene (-5518 to -2490 relative to the transcriptional start site) directed high levels of transcriptional activity only when stably integrated, but not when expressed extrachromosomally in transient assays. These results are consistent with earlier findings using an in vivo transgenic approach. The expression of beta -MyHC reporter gene constructs was strictly correlated to differentiation status and coincided with the expression of endogenous cardiac marker genes and with morphological differentiation of embryoid bodies in vitro. Using populations of stably transfected cell clones, two domains important for high level expression were identified. The analysis of individual cell clones suggested that the positive regulatory domains act according to the graded model of enhancement. These results show that chromosomal integration is necessary for the appropriate function of the beta -MyHC gene's upstream regulatory region.
Collapse
Affiliation(s)
- K L Schreiber
- Department of Medicine, University of Montreal and Montreal Heart Institute, 5000 Belanger Street, Montreal, Quebec, H1T 1C8, Canada
| | | | | |
Collapse
|
36
|
Adams GR, Haddad F, McCue SA, Bodell PW, Zeng M, Qin L, Qin AX, Baldwin KM. Effects of spaceflight and thyroid deficiency on rat hindlimb development. II. Expression of MHC isoforms. J Appl Physiol (1985) 2000; 88:904-16. [PMID: 10710385 DOI: 10.1152/jappl.2000.88.3.904] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Both slow-twitch and fast-twitch muscles are undifferentiated after birth as to their contractile protein phenotype. Thus we examined the separate and combined effects of spaceflight (SF) and thyroid deficiency (TD) on myosin heavy chain (MHC) gene expression (protein and mRNA) in muscles of neonatal rats (7 and 14 days of age at launch) exposed to SF for 16 days. Spaceflight markedly reduced expression of the slow, type I MHC gene by approximately 55%, whereas it augmented expression of the fast IIx and IIb MHCs in antigravity skeletal muscles. In fast muscles, SF caused subtle increases in the fast IIb MHC relative to the other adult MHCs. In contrast, TD prevented the normal expression of the fast MHC phenotype, particularly the IIb MHC, whereas TD maintained expression of the embryonic/neonatal MHC isoforms; this response occurred independently of gravity. Collectively, these results suggest that normal expression of the type I MHC gene requires signals associated with weight-bearing activity, whereas normal expression of the IIb MHC requires an intact thyroid state acting independently of the weight-bearing activities typically encountered during neonatal development of laboratory rodents. Finally, MHC expression in developing muscles is chiefly regulated by pretranslational processes based on the tight relationship between the MHC protein and mRNA data.
Collapse
Affiliation(s)
- G R Adams
- Department of Physiology and Biophysics, University of California, Irvine, California 92697-4560, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
McCarthy JJ, Vyas DR, Tsika GL, Tsika RW. Segregated regulatory elements direct beta-myosin heavy chain expression in response to altered muscle activity. J Biol Chem 1999; 274:14270-9. [PMID: 10318848 DOI: 10.1074/jbc.274.20.14270] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Our previous transgenic analyses revealed that a 600-base pair beta-myosin heavy chain (betaMyHC) promoter conferred mechanical overload (MOV) and non-weight-bearing (NWB) responsiveness to a chloramphenicol acetyltransferase reporter gene. Whether the same DNA regulatory element(s) direct betaMyHC expression following MOV or NWB activity in vivo remains unknown. We now show that a 293-base pair betaMyHC promoter fused to chloramphenicol acetyltransferase (beta293) responds to MOV, but not NWB activity, indicating a segregation of these two diverse elements. Inclusion of the betaMyHC negative regulatory element (-332 to -300; betaNRE) within transgene beta350 repressed expression in all transgenic lines. Electrophoretic mobility shift assays showed highly enriched binding activity only in NWB soleus nuclear extracts that was specific to the distal region of the betaNRE sense strand (dbetaNRE-S; -332 to -311). Supershift electrophoretic mobility shift assay revealed that the binding at the distal region of the betaNRE sense strand was antigenically distinct from cellular nucleic acid-binding protein and Y-box-binding factor 1, two proteins shown to bind this element. Two-dimensional UV cross-linking and shift Southwestern blotting analyses detected two proteins (50 and 52 kDa) that bind to this element. These in vivo results demonstrate that segregated betaMyHC promoter elements transcriptionally regulate betaMyHC transgene expression in response to two diverse modes of neuromuscular activity.
Collapse
Affiliation(s)
- J J McCarthy
- Department of Veterinary Biomedical Sciences, School of Veterinary Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | | |
Collapse
|
38
|
Morimoto T, Hasegawa K, Kaburagi S, Kakita T, Masutani H, Kitsis RN, Matsumori A, Sasayama S. GATA-5 is involved in leukemia inhibitory factor-responsive transcription of the beta-myosin heavy chain gene in cardiac myocytes. J Biol Chem 1999; 274:12811-8. [PMID: 10212267 DOI: 10.1074/jbc.274.18.12811] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Leukemia inhibitory factor is a member of a family of structurally related cytokines sharing the receptor component gp130. Activation of gp130 by leukemia inhibitory factor is sufficient to induce myocardial cell hypertrophy accompanied by specific changes in the pattern of gene expression. However, the molecular mechanisms that link gp130 activation to these changes have not been clarified. The present study investigated the transcriptional pathways by which leukemia inhibitory factor activates beta-myosin heavy chain expression during myocardial cell hypertrophy. Mutation of the GATA motif in the beta-myosin heavy chain promoter totally abolished leukemia inhibitory factor-responsive transcription without changing basal transcriptional activity. In contrast, endothelin-1 responsiveness was unaffected by the GATA mutation. Among members of the cardiac GATA transcription factor subfamily (GATA-4, -5, and -6), GATA-5 was the sole and potent transactivator for the beta-myosin heavy chain promoter. This transactivation was dependent on sequence-specific binding of GATA-5 to the beta-myosin heavy chain GATA element. Cardiac nuclear factors that bind to to the beta-myosin heavy chain GATA element were induced by leukemia inhibitory factor stimulation. Last, leukemia inhibitory factor stimulation markedly increased transcripts of cardiac GATA-5, the expression of which is normally restricted to the early embryo. Thus, GATA-5 may be involved in gp130 signaling in cardiac myocytes.
Collapse
Affiliation(s)
- T Morimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Wright CE, Haddad F, Qin AX, Bodell PW, Baldwin KM. In vivo regulation of beta-MHC gene in rodent heart: role of T3 and evidence for an upstream enhancer. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:C883-91. [PMID: 10199819 DOI: 10.1152/ajpcell.1999.276.4.c883] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac beta-myosin heavy chain (beta-MHC) gene expression is mainly regulated through transcriptional processes. Although these results are based primarily on in vitro cell culture models, relatively little information is available concerning the interaction of key regulatory factors thought to modulate MHC expression in the intact rodent heart. Using a direct gene transfer approach, we studied the in vivo transcriptional activity of different-length beta-MHC promoter fragments in normal control and in altered thyroid states. The test beta-MHC promoter was fused to a firefly luciferase reporter gene, whereas the control alpha-MHC promoter was fused to the Renilla luciferase reporter gene and was used to account for variations in transfection efficiency. Absolute reporter gene activities showed that beta- and alpha-MHC genes were individually and reciprocally regulated by thyroid hormone. The beta-to-alpha ratios of reporter gene expression demonstrated an almost threefold larger beta-MHC gene expression in the longest than in the shorter promoter fragments in normal control animals, implying the existence of an upstream enhancer. A mutation in the putative thyroid response element of the -408-bp beta-MHC promoter construct caused transcriptional activity to drop to null. When studied in the -3, 500-bp beta-MHC promoter, construct activity was reduced ( approximately 100-fold) while thyroid hormone responsiveness was retained. These findings suggest that, even though the bulk of the thyroid hormone responsiveness of the gene is contained within the first 215 bp of the beta-MHC promoter sequence, the exact mechanism of triiodothyronine (T3) action remains to be elucidated.
Collapse
Affiliation(s)
- C E Wright
- Department of Physiology and Biophysics, University of California, Irvine, California 92697, USA
| | | | | | | | | |
Collapse
|
40
|
Qian Q, Kuo L, Yu YT, Rottman JN. A concise promoter region of the heart fatty acid-binding protein gene dictates tissue-appropriate expression. Circ Res 1999; 84:276-89. [PMID: 10024301 DOI: 10.1161/01.res.84.3.276] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The heart fatty acid-binding protein (HFABP) is a member of a family of binding proteins with distinct tissue distributions and diverse roles in fatty acid metabolism, trafficking, and signaling. Other members of this family have been shown to possess concise promoter regions that direct appropriate tissue-specific expression. The basis for the specific expression of the HFABP has not been previously evaluated, and the mechanisms governing expression of metabolic genes in the heart are not completely understood. We used transient and permanent transfections in ventricular myocytes, skeletal myocytes, and nonmyocytic cells to map regulatory elements in the HFABP promoter, and audited results in transgenic mice. Appropriate tissue-specific expression in cell culture and in transgenic mice was dictated by 1.2 kb of the 5'-flanking sequence of FABP3, the HFABP gene. Comparison of orthologous murine and human genomic sequences demonstrated multiple regions of near-identity within this promoter region, including a CArG-like element close to the TATA box. Binding and transactivation studies demonstrated that this element can function as an atypical myocyte enhancer-binding factor 2 site. Interactions with adjacent sites are likely to be necessary for fully appropriate, tissue-specific, developmental and metabolic regulation.
Collapse
Affiliation(s)
- Q Qian
- Departments of Internal Medicine (Cardiology), Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | |
Collapse
|
41
|
Haddad F, Qin AX, Zeng M, McCue SA, Baldwin KM. Interaction of hyperthyroidism and hindlimb suspension on skeletal myosin heavy chain expression. J Appl Physiol (1985) 1998; 85:2227-36. [PMID: 9843547 DOI: 10.1152/jappl.1998.85.6.2227] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We examined the novel interaction of hyperthyroidism and hindlimb suspension on the pattern of myosin heavy chain (MHC) expression (mRNA and protein) in skeletal muscles. Female Sprague-Dawley rats were assigned to four groups: 1) normal control (Con); 2) thyroid hormone treated [150 micrograms 3,5,3'-triiodothyronine (T3). kg-1. day-1] (T3); 3) hindlimb suspension (HS); or 4) T3-treated and HS (T3 + HS). Results show for the first time the novel observation that the combination T3 + HS induces a rapid and sustained, marked (80-90%) downregulation of type I MHC gene expression that is mirrored temporally by concomitant marked upregulation of type IIb MHC gene expression, as evidenced by the de novo synthesis of type IIb MHC protein in the soleus. The fast type IIx MHC isoform showed a differential response among the experimental groups, generally increasing with the separate and combined treatments in both the soleus and vastus intermedius muscles while decreasing in the plantaris muscles. The fast type IIa MHC was the least responsive to suspension of the MHCs and reflected its greatest responsiveness to T3 treatment while also undergoing differential adaptations in slow vs. fast muscle (increases vs. decreases, respectively). These results confirm previous findings that all four adult MHC genes are sensitive to T3 and suspension in a muscle-specific manner. In addition, we show that T3 + HS can interact synergistically to create novel adaptations in MHC expression that could not be observed when each factor was imposed separately.
Collapse
Affiliation(s)
- F Haddad
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, Calfornia 92697, USA
| | | | | | | | | |
Collapse
|
42
|
Gupta M, Zak R, Libermann TA, Gupta MP. Tissue-restricted expression of the cardiac alpha-myosin heavy chain gene is controlled by a downstream repressor element containing a palindrome of two ets-binding sites. Mol Cell Biol 1998; 18:7243-58. [PMID: 9819411 PMCID: PMC109306 DOI: 10.1128/mcb.18.12.7243] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The expression of the alpha-myosin heavy chain (MHC) gene is restricted primarily to cardiac myocytes. To date, several positive regulatory elements and their binding factors involved in alpha-MHC gene regulation have been identified; however, the mechanism restricting the expression of this gene to cardiac myocytes has yet to be elucidated. In this study, we have identified by using sequential deletion mutants of the rat cardiac alpha-MHC gene a 30-bp purine-rich negative regulatory (PNR) element located in the first intronic region that appeared to be essential for the tissue-specific expression of the alpha-MHC gene. Removal of this element alone elevated (20- to 30-fold) the expression of the alpha-MHC gene in cardiac myocyte cultures and in heart muscle directly injected with plasmid DNA. Surprisingly, this deletion also allowed a significant expression of the alpha-MHC gene in HeLa and other nonmuscle cells, where it is normally inactive. The PNR element required upstream sequences of the alpha-MHC gene for negative gene regulation. By DNase I footprint analysis of the PNR element, a palindrome of two high-affinity Ets-binding sites (CTTCCCTGGAAG) was identified. Furthermore, by analyses of site-specific base-pair mutation, mobility gel shift competition, and UV cross-linking, two different Ets-like proteins from cardiac and HeLa cell nuclear extracts were found to bind to the PNR motif. Moreover, the activity of the PNR-binding factor was found to be increased two- to threefold in adult rat hearts subjected to pressure overload hypertrophy, where the alpha-MHC gene is usually suppressed. These data demonstrate that the PNR element plays a dual role, both downregulating the expression of the alpha-MHC gene in cardiac myocytes and silencing the muscle gene activity in nonmuscle cells. Similar palindromic Ets-binding motifs are found conserved in the alpha-MHC genes from different species and in other cardiac myocyte-restricted genes. These results are the first to reveal a role of the Ets class of proteins in controlling the tissue-specific expression of a cardiac muscle gene.
Collapse
Affiliation(s)
- M Gupta
- The Heart Institute for Children, Hope Children's Hospital, Oak Lawn, Illinois 60453, USA.
| | | | | | | |
Collapse
|
43
|
Swartz EA, Johnson AD, Owens GK. Two MCAT elements of the SM alpha-actin promoter function differentially in SM vs. non-SM cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:C608-18. [PMID: 9688616 DOI: 10.1152/ajpcell.1998.275.2.c608] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Transcriptional activity of the smooth muscle (SM) alpha-actin gene is differentially regulated in SM vs. non-SM cells. Contained within the rat SM alpha-actin promoter are two MCAT motifs, binding sites for transcription enhancer factor 1 (TEF-1) transcriptional factors implicated in the regulation of many muscle-specific genes. Transfections of SM alpha-actin promoter-CAT constructs containing wild-type or mutagenized MCAT elements were performed to evaluate their functional significance. Mutation of the MCAT elements resulted in increased transcriptional activity in SM cells, whereas these mutations either had no effect or decreased activity in L6 myotubes or endothelial cells. High-resolution gel shift assays resolved several complexes of different mobilities that were formed between MCAT oligonucleotides and nuclear extracts from the different cell types, although no single band was unique to SM. Western blot analysis of nuclear extracts with polyclonal antibodies to conserved domains of the TEF-1 gene family revealed multiple reactive bands, some that were similar and others that differed between SM and non-SM. Supershift assays with a polyclonal antibody to the TEF-related protein family demonstrated that TEF-1 or TEF-1-related proteins were contained in the shifted complexes. Results suggest that the MCAT elements may contribute to cell type-specific regulation of the SM alpha-actin gene. However, it remains to be determined whether the differential transcriptional activity of MCAT elements in SM vs. non-SM is due to differences in expression of TEF-1 or TEF-1-related proteins or to unique (cell type specific) combinatorial interactions of the MCAT elements with other cis-elements and trans-factors.
Collapse
Affiliation(s)
- E A Swartz
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22906, USA
| | | | | |
Collapse
|
44
|
Abstract
Thrombin has been shown to mediate neurite retraction in neurons and synapse elimination at the neuromuscular junction. The presence of prothrombin mRNA has been demonstrated in brain and in muscle, but extra-hepatic regulation of the prothrombin gene has not been investigated. To identify cis-acting DNA elements involved in the expression of the prothrombin gene in muscle, we have isolated and analyzed a 1.3-kilobase pair promoter region of the mouse prothrombin gene. Using a series of transiently transfected plasmid constructs in which gene segments of the prothrombin promoter were linked to the luciferase gene, we have identified a sequence, -302 to -210, essential for prothrombin promoter activity in C2-myotubes. Fine analysis revealed that deletion of nucleotides between -248 and -235 eliminated prothrombin promoter activity in C2-myotubes. Furthermore, electrophoretic mobility shift assays demonstrated that a nuclear factor present in C2-myotubes, but not in C2-myoblasts or HepG2 hepatocytes, specifically binds to the sequence -241 to -225. Substitutional mutation of nucleotides -237 to -231 abolished myotube-specific promoter activity and inhibited the nuclear factor binding. Quantitative reverse transcription polymerase chain reaction demonstrated the expression of prothrombin mRNA in myotubes, but not in myoblasts, of primary, C2, and G8 muscle cells. This result correlates with the lack of prothrombin promoter activity in C2-myoblasts. The data thus suggest that a myotube-specific nuclear factor binds to a cis-acting sequence encompassing the core nucleotides -237 to -231 and plays a critical role in muscle-specific, differentiation-dependent expression of the mouse prothrombin gene.
Collapse
Affiliation(s)
- S Kim
- Laboratory of Developmental Neurobiology, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
45
|
Takano H, Komuro I, Oka T, Shiojima I, Hiroi Y, Mizuno T, Yazaki Y. The Rho family G proteins play a critical role in muscle differentiation. Mol Cell Biol 1998; 18:1580-9. [PMID: 9488475 PMCID: PMC108873 DOI: 10.1128/mcb.18.3.1580] [Citation(s) in RCA: 123] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Rho family GTP-binding proteins play a critical role in a variety of cytoskeleton-dependent cell functions. In this study, we examined the role of Rho family G proteins in muscle differentiation. Dominant negative forms of Rho family proteins and RhoGDI, a GDP dissociation inhibitor, suppressed transcription of muscle-specific genes, while mutationally activated forms of Rho family proteins strongly activated their transcription. C2C12 cells overexpressing RhoGDI (C2C12RhoGDI cells) did not differentiate into myotubes, and expression levels of myogenin, MRF4, and contractile protein genes but not MyoD and myf5 genes were markedly reduced in C2C12RhoGDI cells. The promoter activity of the myogenin gene was suppressed by dominant negative mutants of Rho family proteins and was reduced in C2C12RhoGDI cells. Expression of myocyte enhancer binding factor 2 (MEF2), which has been reported to be required for the expression of the myogenin gene, was reduced at the mRNA and protein levels in C2C12RhoGDI cells. These results suggest that the Rho family proteins play a critical role in muscle differentiation, possibly by regulating the expression of the myogenin and MEF2 genes.
Collapse
Affiliation(s)
- H Takano
- Department of Medicine III, University of Tokyo School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Swoap SJ. In vivo analysis of the myosin heavy chain IIB promoter region. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:C681-7. [PMID: 9530099 DOI: 10.1152/ajpcell.1998.274.3.c681] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The myosin heavy chain (MHC) IIB gene is preferentially expressed in fast-twitch muscles of the hindlimb, such as the tibialis anterior (TA). The molecular mechanism(s) for this preferential expression are unknown. The goals of the current study were 1) to determine whether the cloned region of the MHC IIB promoter contains the necessary cis-acting element(s) to drive fiber-type-specific expression of this gene in vivo, 2) to determine which region within the promoter is responsible for fiber-type-specific expression, and 3) to determine whether transcription off of the cloned region of the MHC IIB promoter accurately mimics endogenous gene expression in a muscle undergoing a fiber-type transition. To accomplish these goals, a 2.6-kilobase fragment of the promoter-enhancer region of the MHC IIB gene was cloned upstream of the firefly luciferase reporter gene and coinjected with pRL-cytomegalovirus (CMV) (CMV promoter driving the renilla luciferase reporter) into the TA and the slow soleus muscle. Firefly luciferase activity relative to renilla luciferase activity within the TA was 35-fold greater than within the soleus. Deletional analysis demonstrated that only the proximal 295 base pairs (pGL3IIB0.3) were required to maintain this muscle-fiber-type specificity. Reporter gene expression of pGL3IIB0.3 construct was significantly upregulated twofold in unweighted soleus muscles compared with normal soleus muscles. Thus the region within the proximal 295 base pairs of the MHC IIB gene contains at least one element that can drive fiber-type-specific expression of a reporter gene.
Collapse
Affiliation(s)
- S J Swoap
- Department of Biology, Williams College, Williamstown, Massachusetts 01267, USA
| |
Collapse
|
47
|
Froman BE, Tait RC, Gorin FA. Role of E and CArG boxes in developmental regulation of muscle glycogen phosphorylase promoter during myogenesis. DNA Cell Biol 1998; 17:105-15. [PMID: 9502427 DOI: 10.1089/dna.1998.17.105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Muscle glycogen phosphorylase (MGP) transcript and protein levels increase during skeletal muscle development in tandem with the products of other muscle genes responsible for glucose and glycogen metabolism. Previous studies demonstrated that a 269 bp region 5' to exon 1 of MGP is sufficient for developmental regulation in the C2C12 myogenic cell line (Froman et al., 1994). This genomic region (-209 to +60) contains four consensus E box motifs, a CArG-like sequence, and a GC-rich domain. Native MGP transcripts were not detected in pluripotent CH310T1/2 fibroblasts, but low levels of MGP mRNA were measured in CH310T1/2 cells that were stably transfected with MyoD. Three of the E box motifs in the MGP proximal promoter interacted with C2C12 nuclear proteins. However, cotransfection of the MGP promoter with myogenic regulatory factors, including MyoD and myogenin, produced less than 2-fold activation compared with 20-fold activation of the desmin promoter. Mutational analyses of the MGP promoter demonstrated that increased expression in C2C12 myotubes did not require any of the E box motifs or the CArG-like element. A small region (-76 to -68) upstream of GC-rich domain (-64 to -51) significantly reduced promoter activities in both myoblasts and myotubes. The functional studies suggest that MGP is developmentally regulated during myogenesis by alternative pathways that utilize unidentified regulatory elements or ancillary factors.
Collapse
Affiliation(s)
- B E Froman
- Department of Neurology/Center for the Neurosciences, University of California Davis, 95616-8647, USA
| | | | | |
Collapse
|
48
|
Hasegawa K, Lee SJ, Jobe SM, Markham BE, Kitsis RN. cis-Acting sequences that mediate induction of beta-myosin heavy chain gene expression during left ventricular hypertrophy due to aortic constriction. Circulation 1997; 96:3943-53. [PMID: 9403619 DOI: 10.1161/01.cir.96.11.3943] [Citation(s) in RCA: 127] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Marked alterations in the expression of specific genes occur during the development of cardiac hypertrophy in vivo. Little is known, however, about the cis-acting elements that mediate these changes in response to clinically relevant hypertrophic stimuli, such as hemodynamic overload, in intact adult animals. METHODS AND RESULTS The left ventricular expression of a directly injected reporter gene driven by 3542 bp of rat beta-myosin heavy chain (beta-MHC) promoter was increased 3.0-fold by aortic constriction (P<.005), an increment similar to the 3.2-fold increase in the level of the endogenous beta-MHC mRNA in the same left ventricles. Subsequent analysis identified a 107-bp beta-MHC promoter sequence (-303/-197) sufficient to convert a heterologous neutral promoter to one that is activated by aortic constriction. These sequences contain two M-CAT elements, which have previously been demonstrated to mediate inducible expression during alpha1-adrenergic-stimulated hypertrophy in cultured neonatal cardiac myocytes, and a GATA element. Although simultaneous mutation of both M-CAT elements markedly decreased the basal transcriptional activity of an injected 333-bp beta-MHC promoter, it had no effect on aortic constriction-stimulated transcription (3.5-fold increase, P<.005 for both wild type and mutant). In contrast, mutation of the GATA motif markedly attenuated aortic constriction-stimulated transcription (1.6-fold, P=NS) without affecting the basal transcriptional activity. This GATA site can interact with in vitro translated GATA-4 and compete with an established GATA site for GATA-4 binding activity in nuclear extracts from aortic constricted hearts. CONCLUSIONS Basal and aortic constriction-stimulated transcription of the beta-MHC gene is mediated, at least in part, through different mechanisms. A GATA element within beta-MHC sequences -303/-197 plays a role in the transcriptional activation of this gene by aortic constriction.
Collapse
Affiliation(s)
- K Hasegawa
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
The goal of this study was to examine the effects of altered thyroid state and denervation (Den) on skeletal myosin heavy chain (MHC) expression in the plantaris and soleus muscles. Rats were subjected to unilateral denervation (Den) and randomly assigned to one of three groups: (1) euthyroid; (2) hyperthyroid; (3) and hypothyroid. Denervation caused severe muscle atrophy and muscle-type specific MHC transformation. Denervation transformed the soleus to a faster muscle, and its effects required the presence of circulating thyroid hormone. In contrast, denervation transformed the plantaris to a slower muscle independently of thyroid state. Furthermore, thyroid hormone effects did not depend upon innervation status in the soleus, while they required the presence of the nerve in the plantaris. Collectively, these findings suggest that both thyroid hormone and intact nerve (a) differentially affect MHC transformations in fast and slow muscle; and (b) are important factors in regulating the optimal expression of both type I and IIB MHC genes. This research suggests that for patients with nerve damage and/or paralysis, both muscle mass and biochemical properties can also be affected by the thyroid state.
Collapse
Affiliation(s)
- F Haddad
- Department of Physiology and Biophysics, University of California at Irvine, 92697-4560, USA
| | | | | | | |
Collapse
|
50
|
Shi H, Shigeta H, Yang N, Fu K, O'Brian G, Teng CT. Human estrogen receptor-like 1 (ESRL1) gene: genomic organization, chromosomal localization, and promoter characterization. Genomics 1997; 44:52-60. [PMID: 9286700 DOI: 10.1006/geno.1997.4850] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Estrogen receptor-like 1a (ESRL1a; same as estrogen receptor-related orphan receptors, ERR1) belongs to a subfamily of the nuclear receptor superfamily. We have previously shown that human ESRL1a modulates estrogen responsiveness of the lactoferrin gene promoter in transiently transfected endometrial carcinoma RL95-2 cells. In this study, we cloned and characterized the human ESRL1 gene. Through the fluorescence in situ hybridization method, the ESRL1 gene was localized to the centromere region of chromosome 11q12. Partial sequencing, restriction mapping, and PCR analysis revealed that the ESRL1 gene consists of seven exons and is approximately 20 kb in length. We found that the smallest exon (exon 3) contains 117 bp and the largest exon (exon 7) has 1032 bp. The smallest intron (intron 5) is only 88 bp long and the largest intron (intron 2) is 8 kb long. All introns have the conserved GT and AG dinucleotides present at the donor and acceptor sites, respectively. Like the estrogen receptor, the highly conserved DNA-binding domain of hESRL1a is encoded by exon 2 and exon 3, and the intron/exon junctions (2 and 3) are well conserved between the two genes. Primer extension analysis revealed multiple transcription initiation start sites in human uterine (HeLa, HEC, and RL95-2) cell lines. However, one major initiation start site was found by RNase protection assay. The hESRL1a mRNA is differentially expressed in various human tissues. The nucleotide sequence adjacent to the transcription start sites of the ESRL1 lacks the typical TATA and CAAT boxes but is GC rich and contains 10 consensus Sp1-binding elements and two E boxes. The region that contains these transcription factor-binding elements showed a high level of promoter activity when transiently transfected into RL95-2 cells.
Collapse
MESH Headings
- Blotting, Northern
- Blotting, Southern
- Cell Line
- Chromosome Mapping
- Chromosomes, Human, Pair 11/genetics
- Cloning, Molecular
- Consensus Sequence/genetics
- Conserved Sequence
- DNA Primers
- Exons/genetics
- Gene Expression Regulation
- Humans
- In Situ Hybridization, Fluorescence
- Introns/genetics
- Organ Specificity
- Promoter Regions, Genetic
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Estrogen/genetics
- Restriction Mapping
- Sequence Analysis, DNA
- Transcription, Genetic
- ERRalpha Estrogen-Related Receptor
Collapse
Affiliation(s)
- H Shi
- Gene Regulation Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | |
Collapse
|