1
|
Kovacs AM, Zimmer WE. Cell-specific transcription of the smooth muscle gamma-actin gene requires both positive- and negative-acting cis elements. Gene Expr 2018; 7:115-29. [PMID: 9699483 PMCID: PMC6190202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We have characterized the function of putative regulatory sequences upon the smooth muscle transcription of the SMGA gene, using promoter deletion analyses. We demonstrate that the SMGA promoter contains four domains: a basal promoter (-1 to -100), a smooth muscle specifier sequence (-100 to -400), a negative regulator (-400 to -1000), and a smooth muscle-specific modulator (-1000 to -2000). The basal or core promoter supports equivalent transcription in both smooth and skeletal muscle cells. Addition of sequences containing a CArG motif juxtaposed to an E-box element stimulates smooth muscle transcription by five- to sixfold compared to skeletal muscle. This smooth muscle-specific segment is maintained for about 200 bp, after which is a segment of DNA that appears to inhibit the transcriptional capacity of the SMGA promoter in smooth muscle cells. Within the boundary between the smooth muscle specifier and negative regulatory sequences (-400 to -500) are three E-box elements. The smooth muscle modulator domain contains two CArG elements and multiple E-boxes. When added to the SMGA promoter it causes an additional three- to fivefold increase in smooth muscle-specific transcription over that stimulated by the smooth muscle specifier domain. Thus, our studies show that the appropriate cell-specific transcription of the SMGA gene involves complex interactions directed by multiple cis-acting elements. Moreover, our characterization of a cell culture system employing embryonic gizzard smooth muscle cells lays the foundation for further molecular analyses of factors that regulate or control SMGA and other smooth muscle genes during differentiation.
Collapse
Affiliation(s)
- Adrienne M. Kovacs
- Department of Structural and Cellular Biology, University of South Alabama, Mobile, AL 36688
| | - Warren E. Zimmer
- Department of Structural and Cellular Biology, University of South Alabama, Mobile, AL 36688
- Address correspondence to Warren E. Zimmer. Tel: (334) 460-7982; Fax: (334) 460-6771; E-mail:
| |
Collapse
|
2
|
Chaponnier C, Gabbiani G. Monoclonal antibodies against muscle actin isoforms: epitope identification and analysis of isoform expression by immunoblot and immunostaining in normal and regenerating skeletal muscle. F1000Res 2016; 5:416. [PMID: 27335638 PMCID: PMC4893938 DOI: 10.12688/f1000research.8154.2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2016] [Indexed: 11/20/2022] Open
Abstract
Higher vertebrates (mammals and birds) express six different highly conserved actin isoforms that can be classified in three subgroups: 1) sarcomeric actins, α-skeletal (α-SKA) and α-cardiac (α-CAA), 2) smooth muscle actins (SMAs), α-SMA and γ-SMA, and 3) cytoplasmic actins (CYAs), β-CYA and γ-CYA. The variations among isoactins, in each subgroup, are due to 3-4 amino acid differences located in their acetylated N-decapeptide sequence. The first monoclonal antibody (mAb) against an actin isoform (α-SMA) was produced and characterized in our laboratory in 1986 (Skalli et al., 1986) . We have further obtained mAbs against the 5 other isoforms. In this report, we focus on the mAbs anti-α-SKA and anti-α-CAA obtained after immunization of mice with the respective acetylated N-terminal decapeptides using the Repetitive Immunizations at Multiple Sites Strategy (RIMMS). In addition to the identification of their epitope by immunoblotting, we describe the expression of the 2 sarcomeric actins in mature skeletal muscle and during muscle repair after micro-lesions. In particular, we analyze the expression of α-CAA, α-SKA and α-SMA by co-immunostaining in a time course frame during the muscle repair process. Our results indicate that a restricted myocyte population expresses α-CAA and suggest a high capacity of self-regeneration in muscle cells. These antibodies may represent a helpful tool for the follow-up of muscle regeneration and pathological changes.
Collapse
Affiliation(s)
- Christine Chaponnier
- Department of Pathology-Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Giulio Gabbiani
- Department of Pathology-Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
3
|
Abstract
Mammalian skeletal muscle comprises different fiber types, whose identity is first established during embryonic development by intrinsic myogenic control mechanisms and is later modulated by neural and hormonal factors. The relative proportion of the different fiber types varies strikingly between species, and in humans shows significant variability between individuals. Myosin heavy chain isoforms, whose complete inventory and expression pattern are now available, provide a useful marker for fiber types, both for the four major forms present in trunk and limb muscles and the minor forms present in head and neck muscles. However, muscle fiber diversity involves all functional muscle cell compartments, including membrane excitation, excitation-contraction coupling, contractile machinery, cytoskeleton scaffold, and energy supply systems. Variations within each compartment are limited by the need of matching fiber type properties between different compartments. Nerve activity is a major control mechanism of the fiber type profile, and multiple signaling pathways are implicated in activity-dependent changes of muscle fibers. The characterization of these pathways is raising increasing interest in clinical medicine, given the potentially beneficial effects of muscle fiber type switching in the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Stefano Schiaffino
- Venetian Institute of Molecular Medicine, Department of Biomedical Sciences, University of Padova, Consiglio Nazionale delle Ricerche Institute of Neurosciences, and Department of Human Anatomy and Physiology, University of Padova, Padova, Italy
| | - Carlo Reggiani
- Venetian Institute of Molecular Medicine, Department of Biomedical Sciences, University of Padova, Consiglio Nazionale delle Ricerche Institute of Neurosciences, and Department of Human Anatomy and Physiology, University of Padova, Padova, Italy
| |
Collapse
|
4
|
Tondeleir D, Vandamme D, Vandekerckhove J, Ampe C, Lambrechts A. Actin isoform expression patterns during mammalian development and in pathology: insights from mouse models. ACTA ACUST UNITED AC 2009; 66:798-815. [PMID: 19296487 DOI: 10.1002/cm.20350] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The dynamic actin cytoskeleton, consisting of six actin isoforms in mammals and a variety of actin binding proteins is essential for all developmental processes and for the viability of the adult organism. Actin isoform specific functions have been proposed for muscle contraction, cell migration, endo- and exocytosis and maintaining cell shape. However, these specific functions for each of the actin isoforms during development are not well understood. Based on transgenic mouse models, we will discuss the expression patterns of the six conventional actin isoforms in mammals during development and adult life. Ablation of actin genes usually leads to lethality and affects expression of other actin isoforms at the cell or tissue level. A good knowledge of their expression and functions will contribute to fully understand severe phenotypes or diseases caused by mutations in actin isoforms.
Collapse
Affiliation(s)
- Davina Tondeleir
- Department of Medical Protein Research, Flanders Interuniversity Institute for Biotechnology (VIB), Albert Baertsoenkaai 3, Ghent, Belgium
| | | | | | | | | |
Collapse
|
5
|
Stary CM, Walsh BJ, Knapp AE, Brafman D, Hogan MC. Elevation in heat shock protein 72 mRNA following contractions in isolated single skeletal muscle fibers. Am J Physiol Regul Integr Comp Physiol 2008; 295:R642-8. [PMID: 18525012 PMCID: PMC2519928 DOI: 10.1152/ajpregu.00852.2007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Accepted: 06/02/2008] [Indexed: 11/22/2022]
Abstract
The purpose of the present study was 1) to develop a stable model for measuring contraction-induced elevations in mRNA in single skeletal muscle fibers and 2) to utilize this model to investigate the response of heat shock protein 72 (HSP72) mRNA following an acute bout of fatiguing contractions. Living, intact skeletal muscle fibers were microdissected from lumbrical muscle of Xenopus laevis and either electrically stimulated for 15 min of tetanic contractions (EX; n=26) or not stimulated to contract (REST; n=14). The relative mean developed tension of EX fibers decreased to 29+/-7% of initial peak tension at the stimulation end point. Following treatment, individual fibers were allowed to recover for 1 (n=9), 2 (n=8), or 4 h (n=9) prior to isolation of total cellular mRNA. HSP72, HSP60, and cardiac alpha-actin mRNA content were then assessed in individual fibers using quantitative PCR detection. Relative HSP72 mRNA content was significantly (P<0.05) elevated at the 2-h postcontraction time point relative to REST fibers when normalized to either HSP60 (18.5+/-7.5-fold) or cardiac alpha-actin (14.7+/-4.3-fold), although not at the 1- or 4-h time points. These data indicate that 1) extraction of RNA followed by relative quantification of mRNA of select genes in isolated single skeletal muscle fibers can be reliably performed, 2) HSP60 and cardiac alpha-actin are suitable endogenous normalizing genes in skeletal muscle following contractions, and 3) a significantly elevated content of HSP72 mRNA is detectable in skeletal muscle 2 h after a single bout of fatiguing contractions, despite minimal temperature changes and without influence from extracellular sources.
Collapse
Affiliation(s)
- Creed M Stary
- Division of Physiology, Department of Medicine, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0623, USA
| | | | | | | | | |
Collapse
|
6
|
Hong SE, Park I, Cha H, Rho SH, Park WJ, Cho C, Kim DH. Identification of mouse heart transcriptomic network sensitive to various heart diseases. Biotechnol J 2008; 3:648-58. [PMID: 18320566 DOI: 10.1002/biot.200700250] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Exploring biological systems from highly complex datasets is an important task for systems biology. The present study examined co-expression dynamics of mouse heart transcriptome by spectral graph clustering (SGC) to identify a heart transcriptomic network. SGC of microarray data produced 17 classified biological conditions (called condition spectrum, CS) and co-expression patterns by generating bi-clusters. The results showed dynamic co-expression patterns with a modular structure enriched in heart-related CS (CS-1 and -13) containing abundant heart-related microarray data. Consequently, a mouse heart transcriptomic network was constructed by clique analysis from the gene clusters exclusively present in the heart-related CS; 31 cliques were used for constructing the network. The participating genes in the network were closely associated with important cardiac functions (e. g., development, lipid and glycogen metabolisms). Online Mendelian Inheritance in Man (OMIM) database indicates that mutations of the genes in the network induced serious heart diseases. Many of the tested genes in the network showed significantly altered gene expression in an animal model of hypertrophy. The results suggest that the present approach is critical for constructing a heart-related transcriptomic network and for deducing important genes involved in the pathogenesis of various heart diseases.
Collapse
Affiliation(s)
- Seong-Eui Hong
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju, Korea
| | | | | | | | | | | | | |
Collapse
|
7
|
Bertola LD, Ott EB, Griepsma S, Vonk FJ, Bagowski CP. Developmental expression of the alpha-skeletal actin gene. BMC Evol Biol 2008; 8:166. [PMID: 18518953 PMCID: PMC2443135 DOI: 10.1186/1471-2148-8-166] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Accepted: 06/02/2008] [Indexed: 01/01/2023] Open
Abstract
Background Actin is a cytoskeletal protein which exerts a broad range of functions in almost all eukaryotic cells. In higher vertebrates, six primary actin isoforms can be distinguished: alpha-skeletal, alpha-cardiac, alpha-smooth muscle, gamma-smooth muscle, beta-cytoplasmic and gamma-cytoplasmic isoactin. Expression of these actin isoforms during vertebrate development is highly regulated in a temporal and tissue-specific manner, but the mechanisms and the specific differences are currently not well understood. All members of the actin multigene family are highly conserved, suggesting that there is a high selective pressure on these proteins. Results We present here a model for the evolution of the genomic organization of alpha-skeletal actin and by molecular modeling, illustrate the structural differences of actin proteins of different phyla. We further describe and compare alpha-skeletal actin expression in two developmental stages of five vertebrate species (mouse, chicken, snake, salamander and fish). Our findings confirm that alpha-skeletal actin is expressed in skeletal muscle and in the heart of all five species. In addition, we identify many novel non-muscular expression domains including several in the central nervous system. Conclusion Our results show that the high sequence homology of alpha-skeletal actins is reflected by similarities of their 3 dimensional protein structures, as well as by conserved gene expression patterns during vertebrate development. Nonetheless, we find here important differences in 3D structures, in gene architectures and identify novel expression domains for this structural and functional important gene.
Collapse
Affiliation(s)
- Laura D Bertola
- Institute of Biology, Department of Integrative Zoology University of Leiden, 2333 AL Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
8
|
Jackaman C, Nowak KJ, Ravenscroft G, Lim EM, Clément S, Laing NG. Novel application of flow cytometry: determination of muscle fiber types and protein levels in whole murine skeletal muscles and heart. ACTA ACUST UNITED AC 2008; 64:914-25. [PMID: 17922482 DOI: 10.1002/cm.20239] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Conventional methods for measuring proteins within muscle samples such as immunohistochemistry and western blot analysis can be time consuming, labor intensive and subject to sampling errors. We have developed flow cytometry techniques to detect proteins in whole murine heart and skeletal muscle. Flow cytometry and immunohistochemistry were performed on quadriceps and soleus muscles from male C57BL/6J, BALB/c, CBA and mdx mice. Proteins including actins, myosins, tropomyosin and alpha-actinin were detected via single staining flow cytometric analysis. This correlated with immunohistochemistry using the same antibodies. Muscle fiber types could be determined by dual labeled flow cytometry for skeletal muscle actin and different myosins. This showed similar results to immunohistochemistry for I, IIA and IIB myosins. Flow cytometry of heart samples from C57BL/6J and BALB/c mice dual labeled with cardiac and skeletal muscle actin antibodies demonstrated the known increase in skeletal actin protein in BALB/c hearts. The membrane-associated proteins alpha-sarcoglycan and dystrophin could be detected in C57BL/6J mice, but were decreased or absent in mdx mice. With the ability to label whole muscle samples simultaneously with multiple antibodies, flow cytometry may have advantages over conventional methods for certain applications, including assessing the efficacy of potential therapies for muscle diseases.
Collapse
Affiliation(s)
- Connie Jackaman
- Centre for Medical Research, University of Western Australia, Western Australian Institute for Medical Research, Nedlands, Western Australia 6009, Australia.
| | | | | | | | | | | |
Collapse
|
9
|
Dennis RA, Przybyla B, Gurley C, Kortebein PM, Simpson P, Sullivan DH, Peterson CA. Aging alters gene expression of growth and remodeling factors in human skeletal muscle both at rest and in response to acute resistance exercise. Physiol Genomics 2007; 32:393-400. [PMID: 18073271 DOI: 10.1152/physiolgenomics.00191.2007] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The purpose of this investigation was to compare expression of genes that function in inflammation and stress, cell structure and signaling, or remodeling and growth in skeletal muscle of young (32 +/- 7 yr, n = 15) and elderly (72 +/- 5 yr, n = 16) healthy subjects before and after a bout of resistance leg exercises. A real-time RT-PCR method was used to screen 100 transcripts in v. lateralis biopsies obtained before and 72 h postexercise. The screen identified 15 candidates for differential expression due to aging and/or exercise that were measured quantitatively. The median levels of four mRNAs (insulin-like growth factor-1 and its binding protein IGFBP5, ciliary neurotrophic factor, and the metallopeptidase MMP2) were significantly affected by aging and were greater (1.6- to 2.3-fold, P </= 0.05) in the young than elderly muscle at both time points. The median levels of three mRNAs were significantly (P </= 0.05) affected by exercise in the young. The metallopeptidase inhibitor TIMP1 and alpha-cardiac actin mRNAs increased 2-fold and 6.5-fold, respectively, and GDF8 (myostatin) mRNA decreased by 50%. However, elderly muscle did not display any significant changes in gene expression postexercise. Thus, aging muscle shows decreased levels at rest and an impaired response to exercise for a number of mRNAs for factors potentially involved in muscle growth and remodeling. Future studies must determine the functional importance of these gene expression changes to protein synthesis, satellite cell activity, and other processes that are directly involved in the mechanisms of muscle hypertrophy.
Collapse
Affiliation(s)
- Richard A Dennis
- Central Arkansas Veterans Healthcare System, North Little Rock GRECC, 2200 Fort Roots Dr. (Bldg. 170, 3J/157), North Little Rock, AR 72114-1706, USA.
| | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Salmonids utilize a unique, class II isoactin in slow skeletal muscle. This actin contains 12 replacements when compared with those from salmonid fast skeletal muscle, salmonid cardiac muscle and rabbit skeletal muscle. Substitutions are confined to subdomains 1 and 3, and most occur after residue 100. Depending on the pairing, the 'fast', 'cardiac' and rabbit actins share four, or fewer, substitutions. The two salmonid skeletal actins differ nonconservatively at six positions, residues 103, 155, 278, 281, 310 and 360, the latter involving a change in charge. The heterogeneity has altered the biochemical properties of the molecule. Slow skeletal muscle actin can be distinguished on the basis of mass, hydroxylamine cleavage and electrophoretic mobility at alkaline pH in the presence of 8 m urea. Further, compared with its counterpart in fast muscle, slow muscle actin displays lower activation of myosin in the presence of regulatory proteins, and weakened affinity for nucleotide. It is also less resistant to urea- and heat-induced denaturation. The midpoints of the change in far-UV ellipticity of G-actin versus temperature are approximately 45 degrees C ('slow' actin) and approximately 56 degrees C ('fast' actin). Similar melting temperatures are observed when thermal unfolding is monitored in the aromatic region, and is suggestive of differential stability within subdomain 1. The changes in nucleotide affinity and stability correlate with substitutions at the nucleotide binding cleft (residue 155), and in the C-terminal region, two parts of actin which are allosterically coupled. Actin is concluded to be a source of skeletal muscle plasticity.
Collapse
|
11
|
Multiple non-collinear TF-map alignments of promoter regions. BMC Bioinformatics 2007; 8:138. [PMID: 17456238 PMCID: PMC1878506 DOI: 10.1186/1471-2105-8-138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Accepted: 04/24/2007] [Indexed: 12/25/2022] Open
Abstract
Background The analysis of the promoter sequence of genes with similar expression patterns is a basic tool to annotate common regulatory elements. Multiple sequence alignments are on the basis of most comparative approaches. The characterization of regulatory regions from co-expressed genes at the sequence level, however, does not yield satisfactory results in many occasions as promoter regions of genes sharing similar expression programs often do not show nucleotide sequence conservation. Results In a recent approach to circumvent this limitation, we proposed to align the maps of predicted transcription factors (referred as TF-maps) instead of the nucleotide sequence of two related promoters, taking into account the label of the corresponding factor and the position in the primary sequence. We have now extended the basic algorithm to permit multiple promoter comparisons using the progressive alignment paradigm. In addition, non-collinear conservation blocks might now be identified in the resulting alignments. We have optimized the parameters of the algorithm in a small, but well-characterized collection of human-mouse-chicken-zebrafish orthologous gene promoters. Conclusion Results in this dataset indicate that TF-map alignments are able to detect high-level regulatory conservation at the promoter and the 3'UTR gene regions, which cannot be detected by the typical sequence alignments. Three particular examples are introduced here to illustrate the power of the multiple TF-map alignments to characterize conserved regulatory elements in absence of sequence similarity. We consider this kind of approach can be extremely useful in the future to annotate potential transcription factor binding sites on sets of co-regulated genes from high-throughput expression experiments.
Collapse
|
12
|
Moll R, Holzhausen HJ, Mennel HD, Kuhn C, Baumann R, Taege C, Franke WW. The cardiac isoform of α-actin in regenerating and atrophic skeletal muscle, myopathies and rhabdomyomatous tumors: an immunohistochemical study using monoclonal antibodies. Virchows Arch 2006; 449:175-91. [PMID: 16715231 DOI: 10.1007/s00428-006-0220-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Accepted: 04/08/2006] [Indexed: 11/29/2022]
Abstract
The two sarcomeric isoforms of actins, cardiac and skeletal muscle alpha-actin, are highly homologous so that their immunohistochemical distinction is extremely difficult. Taking advantage of monoclonal antibodies distinguishing the two conservative amino acid exchanges near the aminoterminus, we have performed an extended immunohistochemical analysis of the cardiac alpha-actin (CAA) isoform in normal, regenerating, diseased and neoplastic human muscle tissues. Intense and uniform CAA staining is seen in fetal and adult myocardium and in fetal skeletal muscle while adult skeletal muscle is essentially negative, except for muscle spindle myocytes and a few scattered muscle fibres with overall reduced diameter. By contrast, CAA synthesis is markedly induced in regenerating skeletal muscle cells, in Duchenne muscular dystrophy and upon degenerative atrophy. CAA has also been detected in certain vascular and visceral smooth muscle cells. Among tumors, CAA has consistently been seen in rhabdomyosarcomas and rhabdomyomatous cells of nephroblastomas, whereas, smooth muscle tumors have shown only occasional staining. While the synthesis of this actin isoform is less restricted than previously thought, monoclonal antibodies against CAA provide a well-defined, reliable and sensitive diagnostic tool for the definition and detection of aberrant differentiation in diseased skeletal muscle and of striated muscle differentiation in rhabdomyosarcomas.
Collapse
Affiliation(s)
- Roland Moll
- Institute of Pathology, Philipp University of Marburg, Baldingerstrasse, D-35033 Marburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
13
|
Ilkovski B, Clement S, Sewry C, North KN, Cooper ST. Defining α-skeletal and α-cardiac actin expression in human heart and skeletal muscle explains the absence of cardiac involvement in ACTA1 nemaline myopathy. Neuromuscul Disord 2005; 15:829-35. [PMID: 16288873 DOI: 10.1016/j.nmd.2005.08.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Revised: 07/26/2005] [Accepted: 08/05/2005] [Indexed: 11/16/2022]
Abstract
Mutations in alpha-skeletal actin (ACTA1) underlie several congenital muscle disorders including nemaline myopathy (NM). Almost all ACTA1-NM patients have normal cardiac function, and, even lethally affected congenital NM patients exhibit an unremarkable gestation with decreased foetal movement just prior to birth. Although alpha-skeletal actin is thought to be the predominant sarcomeric actin in human heart (Boheler KR, Carrier L, de la Bastie D, et al. Skeletal actin mRNA increases in the human heart during ontogenic development and is the major isoform of control and failing adult hearts. J Clin Invest 1991;88:323-30 ), ACTA1-NM patients almost never exhibit a cardiac phenotype. In this study, we define the relative expression of skeletal and cardiac actin proteins in human heart and skeletal muscle. We show that alpha-cardiac actin is the predominant sarcomeric isoform in human donor hearts and in early foetal skeletal muscle development. Skeletal actin is the predominant isoform from 25 to 27 weeks gestation and is the exclusive isoform expressed in muscle from infancy through to adulthood. These findings are consistent with clinical observations of NM patients and assist us to better understand the pathogenesis of inherited myopathies and cardiomyopathies with mutations in actin.
Collapse
Affiliation(s)
- Biljana Ilkovski
- Institute for Neuromuscular Research, Children's Hospital at Westmead, Sydney, Australia
| | | | | | | | | |
Collapse
|
14
|
Segev H, Kenyagin-Karsenti D, Fishman B, Gerecht-Nir S, Ziskind A, Amit M, Coleman R, Itskovitz-Eldor J. Molecular analysis of cardiomyocytes derived from human embryonic stem cells. Dev Growth Differ 2005; 47:295-306. [PMID: 16026538 DOI: 10.1111/j.1440-169x.2005.00803.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
During early embryogenesis, the cardiovascular system is the first system to be established and is initiated by a process involving the hypoblastic cells of the primitive endoderm. Human embryonic stem (hES) cells provide a model to investigate the early developmental stages of this system. When removed from their feeder layer, hESC create embryoid bodies (EB) which, when plated, develop areas of beating cells in 21.5% of the EB. These spontaneously contracting cells were demonstrated using histology, immunostaining and reverse transcription-polymerase chain reaction (RT-PCR), to possess morphological and molecular characteristics consistent with cardiomyocytic phenotypes. In addition, the expression pattern of specific cardiomyocytic genes in human EB (hEB) was demonstrated and analyzed for the first time. GATA-4 is the first gene to be expressed in 6-day-old EB. Alpha cardiac actin and atrial natriuretic factor are expressed in older hEB at 10 and 20 days, respectively. Light chain ventricular myosin (MLC-2V) was expressed only in EB with beating areas and its expression increased with time. Alpha heavy chain myosin (alpha-MHC) expression declined in the pulsating hEB with time, in contrast to events in EB derived from mice. We conclude that human embryonic stem cells can provide a useful tool for research on embryogenesis in general and cardiovascular development in particular.
Collapse
Affiliation(s)
- Hanna Segev
- Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Vlahopoulos S, Zimmer WE, Jenster G, Belaguli NS, Balk SP, Brinkmann AO, Lanz RB, Zoumpourlis VC, Schwartz RJ. Recruitment of the androgen receptor via serum response factor facilitates expression of a myogenic gene. J Biol Chem 2004; 280:7786-92. [PMID: 15623502 DOI: 10.1074/jbc.m413992200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Androgen receptor (AR) induced precocious myogenesis in culture and myogenic specified gene activity. Increased levels of AR expression in replicating C2C12 myoblasts stimulated fusion into post-differentiated multinucleated myotubes and the appearance of skeletal alpha-actin transcripts, even in the absence of ligand. Furthermore, AR activated the skeletal alpha-actin promoter, which lacks GRE sites, in co-transfected C2C12 cells. AR co-activation of the skeletal alpha-actin promoter required co-expressed full-length serum response factor (SRF). In vitro, AR associated with SRF and was recruited by SRF to a alpha-actin promoter SRF binding site. Our data suggest that AR is capable of activating myogenic genes devoid of consensus AR binding sites via its recruitment by the myogenic enriched transcription factor, SRF.
Collapse
Affiliation(s)
- Spiros Vlahopoulos
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Lemonnier M, Buckingham ME. Characterization of a cardiac-specific enhancer, which directs {alpha}-cardiac actin gene transcription in the mouse adult heart. J Biol Chem 2004; 279:55651-8. [PMID: 15491989 DOI: 10.1074/jbc.m411082200] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Expression of the mouse alpha-cardiac actin gene in skeletal and cardiac muscle is regulated by enhancers lying 5' to the proximal promoter. Here we report the characterization of a cardiac-specific enhancer located within -2.354/-1.36 kbp of the gene, which is active in cardiocytes but not in C2 skeletal muscle cells. In vivo it directs reporter gene expression to the adult heart, where the proximal promoter alone is inactive. An 85-bp region within the enhancer is highly conserved between human and mouse and contains a central AT-rich site, which is essential for enhancer activity. This site binds myocyte enhancer factor (MEF)2 factors, principally MEF2D and MEF2A in cardiocyte nuclear extracts. These results are discussed in the context of MEF2 activity and of the regulation of the alpha-cardiac actin locus.
Collapse
MESH Headings
- Actins/chemistry
- Animals
- Animals, Genetically Modified
- Base Sequence
- Binding Sites
- Binding, Competitive
- Cell Line
- Cell Nucleus/metabolism
- Cells, Cultured
- DNA/metabolism
- DNA-Binding Proteins/metabolism
- Enhancer Elements, Genetic
- Gene Expression Regulation
- Gene Expression Regulation, Developmental
- Genes, Reporter
- Genome
- Heart/embryology
- Heart/physiology
- Humans
- MADS Domain Proteins
- MEF2 Transcription Factors
- Mice
- Mice, Inbred C3H
- Mice, Transgenic
- Models, Genetic
- Molecular Sequence Data
- Muscle, Skeletal/metabolism
- Mutagenesis, Site-Directed
- Mutation
- Myocardium/metabolism
- Myogenic Regulatory Factors
- Promoter Regions, Genetic
- Rats
- Rats, Wistar
- Time Factors
- Transcription Factors/metabolism
- Transcription, Genetic
- Transfection
- beta-Galactosidase/metabolism
Collapse
Affiliation(s)
- Marguerite Lemonnier
- CNRS URA 2578, Département de Biologie du Développement, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | | |
Collapse
|
17
|
dos Remedios CG, Chhabra D, Kekic M, Dedova IV, Tsubakihara M, Berry DA, Nosworthy NJ. Actin binding proteins: regulation of cytoskeletal microfilaments. Physiol Rev 2003; 83:433-73. [PMID: 12663865 DOI: 10.1152/physrev.00026.2002] [Citation(s) in RCA: 706] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The actin cytoskeleton is a complex structure that performs a wide range of cellular functions. In 2001, significant advances were made to our understanding of the structure and function of actin monomers. Many of these are likely to help us understand and distinguish between the structural models of actin microfilaments. In particular, 1) the structure of actin was resolved from crystals in the absence of cocrystallized actin binding proteins (ABPs), 2) the prokaryotic ancestral gene of actin was crystallized and its function as a bacterial cytoskeleton was revealed, and 3) the structure of the Arp2/3 complex was described for the first time. In this review we selected several ABPs (ADF/cofilin, profilin, gelsolin, thymosin beta4, DNase I, CapZ, tropomodulin, and Arp2/3) that regulate actin-driven assembly, i.e., movement that is independent of motor proteins. They were chosen because 1) they represent a family of related proteins, 2) they are widely distributed in nature, 3) an atomic structure (or at least a plausible model) is available for each of them, and 4) each is expressed in significant quantities in cells. These ABPs perform the following cellular functions: 1) they maintain the population of unassembled but assembly-ready actin monomers (profilin), 2) they regulate the state of polymerization of filaments (ADF/cofilin, profilin), 3) they bind to and block the growing ends of actin filaments (gelsolin), 4) they nucleate actin assembly (gelsolin, Arp2/3, cofilin), 5) they sever actin filaments (gelsolin, ADF/cofilin), 6) they bind to the sides of actin filaments (gelsolin, Arp2/3), and 7) they cross-link actin filaments (Arp2/3). Some of these ABPs are essential, whereas others may form regulatory ternary complexes. Some play crucial roles in human disorders, and for all of them, there are good reasons why investigations into their structures and functions should continue.
Collapse
Affiliation(s)
- C G dos Remedios
- Institute for Biomedical Research, Muscle Research Unit, Department of Anatomy and Histology, University of Sydney, Australia.
| | | | | | | | | | | | | |
Collapse
|
18
|
Royer CL, Howell JC, Morrison PR, Srour EF, Yoder MC. Muscle-derived CD45-SCA-1+c-kit- progenitor cells give rise to skeletal muscle myotubes in vitro. In Vitro Cell Dev Biol Anim 2002; 38:512-7. [PMID: 12703978 DOI: 10.1290/1071-2690(2002)038<0512:mcpcgr>2.0.co;2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A stem cell population isolated from murine skeletal muscle has recently been shown to differentiate into hematopoietic cells after transplantation in vivo. In the present study, we tested the hypothesis that this cell population would also, under appropriate culture conditions, differentiate into skeletal muscle cells in vitro. Lower-extremity skeletal muscle tissue isolated from 3- to 4-wk-old mice was dissected free from bone and vessels, enzymatically digested, and flow cytometrically sorted to yield CD45(-)Sca-1(+)c-Kit(-) (S+) cells. These cells were further sorted into CD34(+) and CD34(-) fractions and examined for skeletal, cardiac, and hematopoietic lineage-specific messenger RNA (mRNA) transcripts immediately after isolation and after a 10- to 14-d culture period. Freshly isolated S(+)CD34(+) cells lacked expression of skeletal-, cardiac-, or hematopoietic-specific mRNA transcripts, whereas S(+)CD34(-) cells expressed c-met, a marker for skeletal muscle satellite cells. During 10-14 d in culture, both S(+)CD34(+) and S(+)CD34(-) cell populations underwent a period of attachment followed by elongation and, ultimately, fusion to create large multinucleated contractile myotubes expressing skeletal muscle lineage mRNA transcripts but not hematopoietic or cardiac lineage transcripts. We conclude that murine skeletal muscle possesses two populations of progenitor cells that can be directly isolated. One population expressing the phenotype S(+)CD34(-) may contain satellite cells, whereas the S(+)CD34(+) population is devoid of satellite cell markers. Both populations possess the ability to differentiate into skeletal muscle cells in vitro.
Collapse
Affiliation(s)
- Cassandre L Royer
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | |
Collapse
|
19
|
Springer ML, Ozawa CR, Blau HM. Transient production of alpha-smooth muscle actin by skeletal myoblasts during differentiation in culture and following intramuscular implantation. CELL MOTILITY AND THE CYTOSKELETON 2002; 51:177-86. [PMID: 11977092 DOI: 10.1002/cm.10022] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
alpha-smooth muscle actin (SMA) is typically not present in post-embryonic skeletal muscle myoblasts or skeletal muscle fibers. However, both primary myoblasts isolated from neonatal mouse muscle tissue, and C2C12, an established myoblast cell line, produced SMA in culture within hours of exposure to differentiation medium. The SMA appeared during the cells' initial elongation, persisted through differentiation and fusion into myotubes, remained abundant in early myotubes, and was occasionally observed in a striated pattern. SMA continued to be present during the initial appearance of sarcomeric actin, but disappeared shortly thereafter leaving only sarcomeric actin in contractile myotubes derived from primary myoblasts. Within one day after implantation of primary myoblasts into mouse skeletal muscle, SMA was observed in the myoblasts; but by 9 days post-implantation, no SMA was detectable in myoblasts or muscle fibers. Thus, both neonatal primary myoblasts and an established myoblast cell line appear to similarly reprise an embryonic developmental program during differentiation in culture as well as differentiation within adult mouse muscles.
Collapse
Affiliation(s)
- Matthew L Springer
- Baxter Laboratory for Genetic Pharmacology, Stanford University, Stanford, California 94305-5175, USA
| | | | | |
Collapse
|
20
|
|
21
|
Bottinelli R, Reggiani C. Human skeletal muscle fibres: molecular and functional diversity. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2001; 73:195-262. [PMID: 10958931 DOI: 10.1016/s0079-6107(00)00006-7] [Citation(s) in RCA: 374] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Contractile and energetic properties of human skeletal muscle have been studied for many years in vivo in the body. It has been, however, difficult to identify the specific role of muscle fibres in modulating muscle performance. Recently it has become possible to dissect short segments of single human muscle fibres from biopsy samples and make them work in nearly physiologic conditions in vitro. At the same time, the development of molecular biology has provided a wealth of information on muscle proteins and their genes and new techniques have allowed analysis of the protein isoform composition of the same fibre segments used for functional studies. In this way the histological identification of three main human muscle fibre types (I, IIA and IIX, previously called IIB) has been followed by a precise description of molecular composition and functional and biochemical properties. It has become apparent that the expression of different protein isoforms and therefore the existence of distinct muscle fibre phenotypes is one of the main determinants of the muscle performance in vivo. The present review will first describe the mechanisms through which molecular diversity is generated and how fibre types can be identified on the basis of structural and functional characteristics. Then the molecular and functional diversity will be examined with regard to (1) the myofibrillar apparatus; (2) the sarcolemma and the sarcoplasmic reticulum; and (3) the metabolic systems devoted to producing ATP. The last section of the review will discuss the advantage that fibre diversity can offer in optimizing muscle contractile performance.
Collapse
Affiliation(s)
- R Bottinelli
- Institute of Human Physiology, University of Pavia, Via Forlanni 6, 27100, Pavia, Italy.
| | | |
Collapse
|
22
|
Takai E, Akita H, Shiga N, Kanazawa K, Yamada S, Terashima M, Matsuda Y, Iwai C, Kawai K, Yokota Y, Yokoyama M. Mutational analysis of the cardiac actin gene in familial and sporadic dilated cardiomyopathy. AMERICAN JOURNAL OF MEDICAL GENETICS 1999; 86:325-7. [PMID: 10494087 DOI: 10.1002/(sici)1096-8628(19991008)86:4<325::aid-ajmg5>3.0.co;2-u] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dilated cardiomyopathy (DCM) results in part from genetic disorders. Recently, missense mutations of the cardiac actin gene have been reported to cause DCM. We studied 136 Japanese DCM cases to elucidate how frequently the gene mutations are involved in its pathogenesis. Genomic DNA samples were obtained from 136 DCM cases (107 males, 29 females), containing 30 familial DCM (5 confirmed and 25 suspected). All six exons of the cardiac actin gene were analyzed by polymerase chain reaction, single-strand conformation polymorphism, and sequencing. We detected no mutations of the disease causation previously reported (G867A or A1014G) but two silent mutations (G979C and C1018T) in exon 6 and one point mutation (T1080A) in the 3'-untranslated region. As a result of screening 128 healthy subjects, these novel silent mutations were found to be mere genetic polymorphisms, not responsible for the disease. Although some genetic polymorphisms exist in the cardiac actin gene, mutations of the gene are rarely responsible for DCM, at least in the Japanese patients.
Collapse
Affiliation(s)
- E Takai
- First Department of Internal Medicine, Kobe University School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wadhwa R, Sugihara T, Yoshida A, Duncan EL, Hardeman EC, Nomura H, Reddel RR, Kaul SC. Cloning and characterization of a novel gene, striamin, that interacts with the tumor suppressor protein p53. J Biol Chem 1999; 274:14948-55. [PMID: 10329696 DOI: 10.1074/jbc.274.21.14948] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Expression analysis of a novel cDNA isolated from immortal murine fibroblasts revealed a single transcript of 3.0 kilobase pairs that was highly expressed in mouse and human striated muscle and in mouse heart. The gene has therefore been named striamin. Its expression was confined to skeletal muscle types with a fast glycolytic (2B) contractile phenotype. It was also detected in C2C12 mouse myoblasts and was down-regulated during in vitro myogenesis. The cDNA has a single open reading frame encoding a predicted 16.8-kDa protein of 149 amino acids with no homology to known proteins. Microinjection and transfection of green fluorescence protein-tagged striamin demonstrated that it localizes to the nucleus. Coimmunoprecipitations revealed that it can interact with p53 (a positive marker for myoblast differentiation) in vivo and in vitro. Furthermore, it repressed p53 activity in p53-mediated reporter assays. Fluorescence in situ hybridization with a mouse P1 genomic clone localized the gene to chromosome 12C3, which is syntenic to human chromosome 14q21-22.
Collapse
Affiliation(s)
- R Wadhwa
- Chugai Research Institute for Molecular Medicine, 153-2 Nagai, Niihari-Mura, Niihari-Gun, Ibaraki 300-41, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Rolland AP, Mumper RJ. Plasmid delivery to muscle: Recent advances in polymer delivery systems. Adv Drug Deliv Rev 1998; 30:151-172. [PMID: 10837608 DOI: 10.1016/s0169-409x(97)00113-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Preclinical studies involving intramuscular injection of plasmid into animals have revealed at least four significant variables that effect levels of gene expression (i.e., >fivefold effect over controls), including the formulation, injection technique, species and pretreatment of the muscle with myotoxic agents to induce muscle damage. The uptake of plasmid formulated in saline has been shown to be a saturable process, most likely via a receptor-mediated event involving the T tubules and caveolae. Pharmacokinetic studies have demonstrated that the bioavailability of injected plasmid to muscle cells is very low, due to rapid and extensive plasmid degradation by extracellular nucleases. We have developed protective, interactive, non-condensing (PINC) delivery systems designed to complex plasmids and to (i) protect plasmids from rapid nuclease degradation, (ii) disperse and retain intact plasmid in the muscle and (iii) facilitate the uptake of plasmid by muscle cells. PINC systems result in up to at least a one log increase in both the extent and levels of gene expression over plasmid formulated in saline. We have combined the PINC delivery systems with two different muscle-specific expression plasmids. After direct intramuscular injection of these gene medicines, we have shown both local myotrophic and neurotrophic effects of expressed human insulin-like growth factor (hIGF-I) and the secretion of biologically active human growth hormone (hGH) into the systemic circulation.
Collapse
Affiliation(s)
- AP Rolland
- GeneMedicine, Inc., 8301 New Trails Drive, The Woodlands, TX 77381-4248, USA
| | | |
Collapse
|
25
|
Developmental Expression of Mouse Erythrocyte Protein 4.2 mRNA: Evidence for Specific Expression in Erythroid Cells. Blood 1998. [DOI: 10.1182/blood.v91.2.695] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractErythrocyte protein 4.2 (P4.2) is an important component of the erythrocyte membrane skeletal network with an undefined biologic function. Presently, very little is known about the expression of the P4.2 gene during mouse embryonic development and in adult animals. By using the Northern blot and in situ hybridization techniques, we have examined the spatial and temporal expression of the P4.2 gene during mouse development. We show that expression of the mouse P4.2 gene is temporally regulated during embryogenesis and that the P4.2 mRNA expression pattern coincides with the timing of erythropoietic activity in hematopoietic organs. P4.2 transcripts are first detected in embryos on day 7.5 of gestation and are localized exclusively in primitive erythroid cells of yolk sac origin. These erythroid cells remain to be the only source for P4.2 expression until the switch of the hematopoietic producing site to fetal liver. In mid- and late-gestation periods, P4.2 mRNA expression is restricted to the erythroid cells in fetal liver and to circulating erythrocytes. Around and after birth, the site for P4.2 expression is switched from liver to spleen and bone marrow, and P4.2 transcripts are only detected in cells of the erythroid lineage. These results provide the evidence for specific P4.2 expression in erythroid cells. In addition, the timing and pattern of expression of the P4.2 gene suggest the specific regulation of the P4.2 gene.
Collapse
|
26
|
Developmental Expression of Mouse Erythrocyte Protein 4.2 mRNA: Evidence for Specific Expression in Erythroid Cells. Blood 1998. [DOI: 10.1182/blood.v91.2.695.695_695_705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Erythrocyte protein 4.2 (P4.2) is an important component of the erythrocyte membrane skeletal network with an undefined biologic function. Presently, very little is known about the expression of the P4.2 gene during mouse embryonic development and in adult animals. By using the Northern blot and in situ hybridization techniques, we have examined the spatial and temporal expression of the P4.2 gene during mouse development. We show that expression of the mouse P4.2 gene is temporally regulated during embryogenesis and that the P4.2 mRNA expression pattern coincides with the timing of erythropoietic activity in hematopoietic organs. P4.2 transcripts are first detected in embryos on day 7.5 of gestation and are localized exclusively in primitive erythroid cells of yolk sac origin. These erythroid cells remain to be the only source for P4.2 expression until the switch of the hematopoietic producing site to fetal liver. In mid- and late-gestation periods, P4.2 mRNA expression is restricted to the erythroid cells in fetal liver and to circulating erythrocytes. Around and after birth, the site for P4.2 expression is switched from liver to spleen and bone marrow, and P4.2 transcripts are only detected in cells of the erythroid lineage. These results provide the evidence for specific P4.2 expression in erythroid cells. In addition, the timing and pattern of expression of the P4.2 gene suggest the specific regulation of the P4.2 gene.
Collapse
|
27
|
Collins T, Joya JE, Arkell RM, Ferguson V, Hardeman EC. Reappearance of the minor alpha-sarcomeric actins in postnatal muscle. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 273:C1801-10. [PMID: 9435483 DOI: 10.1152/ajpcell.1997.273.6.c1801] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The postnatal expression profiles of alpha-sarcomeric actin transcripts and protein are quantified in mouse striated muscles from birth to postnatal day 56 by Northern and Western blot analyses. alpha-Cardiac actin (alpha-CA) transcripts transiently increase between 12 and 21 days after birth in the quadriceps muscle, reaching approximately 90% that found in the adult mouse heart. Although alpha-CA is the alpha-sarcomeric actin isoform expressed in the immature fiber, the expression profiles of other contractile protein isoforms indicate that this postnatal period is not reflective of an immature phenotype. alpha-Skeletal actin (alpha-SA) transcripts accumulate to approximately 32% of the total alpha-sarcomeric actin transcripts in the adult heart. Our study shows that 1) there is a simultaneous reappearance of alpha-CA and alpha-SA in postnatal skeletal and heart muscles, respectively, and 2) the contractile protein gene expression profile characteristic of adult skeletal muscle is not achieved until after 42 days postnatal in the mouse. We propose there is a previously uncharacterized period of postnatal striated muscle maturation marked by the reappearance of the minor alpha-sarcomeric actins.
Collapse
Affiliation(s)
- T Collins
- Muscle Development Unit, Children's Medical Research Institute, Westmead, New South Wales, Australia
| | | | | | | | | |
Collapse
|
28
|
Reecy JM, Bidwell CA, Briley GP, Grant AL. Structure and regulation of the porcine skeletal alpha-actin-encoding gene. Gene 1996; 180:23-8. [PMID: 8973342 DOI: 10.1016/s0378-1119(96)00394-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The postnatal increase in skeletal alpha-actin (Sk-alpha-Act) synthesis in pigs is due, in part, to increased transcription. To characterize the factors responsible for its transcriptional regulation, we have cloned and determined the nucleotide sequence of a 5.2-kb HindIII genomic DNA fragment which contains the complete coding region of Sk-alpha-Act distributed over seven exons, plus 1.9 kb of 5' flanking region and 0.5 kb of 3' flanking sequence. The major transcription start point (tsp) of Sk-alpha-Act was determined to be 840 bp 5' to the ATG start codon by primer extension and RNase protection analysis. To demonstrate that the Sk-alpha-Act promoter was functional, L6 myoblasts, C2C12 myoblasts and HeLa cells were transfected with a construct (pPSKAFL-CAT) linking the 5' Sk-alpha-Act promoter to the chloramphenicol acetyltransferase reporter gene (cat). Cell lysates from L6 myoblasts, L6 myotubes, C2C12 myoblasts, C2C12 myotubes, and HeLa cells were analyzed for CAT activity. CAT activity was detected only in C2C12 myotubes. Thus, the porcine Sk-alpha-Act promoter is regulated in a developmental and cell-type specific manner.
Collapse
Affiliation(s)
- J M Reecy
- Department of Animal Science, Purdue University, West Lafayette, IN 47907-1151, USA
| | | | | | | |
Collapse
|
29
|
Qian J, Kumar A, Szucsik JC, Lessard JL. Tissue and developmental specific expression of murine smooth muscle gamma-actin fusion genes in transgenic mice. Dev Dyn 1996; 207:135-44. [PMID: 8906417 DOI: 10.1002/(sici)1097-0177(199610)207:2<135::aid-aja2>3.0.co;2-i] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Smooth muscle gamma-actin (SMGA) is an excellent marker of smooth muscle differentiation because it is essentially restricted to smooth muscle. As a first step toward unraveling the mechanisms underlying smooth muscle development and differentiation, we have examined the tissue-specific and developmental expression patterns of six constructs carrying portions of the murine SMGA gene linked to chloramphenicol acetyltransferase (CAT) in stable lines of transgenic mice. Based on the transgenic studies most, if not all, of the regulatory elements necessary for proper spatial and temporal expression of SMGA are present within a 13.7 kb segment of the SMGA gene containing 4.9 kb of upstream sequence, exon 1, intron 1, and a portion of exon 2 up to the start codon for translation. A second construct (SMGA11.6CAT) that lacks the distal 2.1 kb of upstream sequence but is otherwise identical to SMGA13.7CAT shows a similar level of smooth muscle-specific CAT activity. However, SMGA9.3CAT fusion gene containing only 571 bp of 5' flanking sequence, but otherwise identical to SMGA13.7CAT, and SMGA6.0CAT containing only the 4.9 kb upstream sequence, exon 1, and a miniintron 1 show a more than a 100-fold reduction of CAT activity in most smooth muscle-rich tissues. Furthermore, removal of most or all of intron 1 from a transgene with 571 bp of upstream sequence (SMGA2.0 CAT and SMGA0.6CAT) results in a near-complete or complete loss of activity, respectively, in all tissues. Overall, the studies suggest that upstream elements between -2.7 kb and -571 bp and elements within intron 1 are required for high levels of SMGA gene expression in an appropriate temporal-spatial fashion.
Collapse
Affiliation(s)
- J Qian
- Division of Developmental Biology, Children's Hospital Medical Center, College of Medicine, University of Cincinnati, Ohio 45229-3039, USA
| | | | | | | |
Collapse
|
30
|
Franke WW, Stehr S, Stumpp S, Kuhn C, Heid H, Rackwitz HR, Schnölzer M, Baumann R, Holzhausen HJ, Moll R. Specific immunohistochemical detection of cardiac/fetal alpha-actin in human cardiomyocytes and regenerating skeletal muscle cells. Differentiation 1996; 60:245-50. [PMID: 8765054 DOI: 10.1046/j.1432-0436.1996.6040245.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We describe three murine monoclonal antibodies (mAbs) raised against a synthetic decapeptide representing the aminoterminal sequence of the cardiac/ fetal isoform of sarcomeric alpha-actin. When used for immunoblotting or histological immunolocalization, these mAbs distinguish cardiac/fetal alpha-actin from skeletal muscle alpha-actin, and also from all other actin isoforms. We show, by immunofluorescence and immunoperoxidase microscopy of tissue sections, that cardiac/fetal alpha-actin can be localized not only in cardiomyocytes but also in skeletal muscles and their satellite cells during regeneration. These mAbs are potentially valuable in developmental biology, for the characterization of tissue and cultured myogenic cells, in pathology, and for serodiagnosis.
Collapse
Affiliation(s)
- W W Franke
- Division of Cell Biology, German Cancer Research Center, Herdelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Metzger JM, Lin WI, Samuelson LC. Vital staining of cardiac myocytes during embryonic stem cell cardiogenesis in vitro. Circ Res 1996; 78:547-52. [PMID: 8635211 DOI: 10.1161/01.res.78.4.547] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Mouse embryonic stem (ES) cells differentiate in vitro into a variety of cell types, including spontaneously contracting cardiac myocytes. The primary aim of this work was to use vital stain techniques for real-time detection of developing cardiac myocytes in ES cell differentiation cultures. The -440 to +6 human cardiac alpha-actin promoter was used to direct expression of the Escherichia coli reporter gene lacZ (pHCActlacZ) into ES cell-derived cardiac myocytes during cardiogenesis in vitro. Undifferentiated ES cells were electroporated with HCActlacZ together with a plasmid containing the neomycin gene under the direction of the phosphoglycerate kinase promoter, and stable transformants were selected in G418. Individual clones were screened for activation of lacZ gene expression in cardiac myocytes developing in vitro. Results showed that expression of the HCActlacZ reporter construct was activated very early during the ES cell differentiation program, at a time point before the appearance of spontaneous contractile activity. The earliest detection was at day 6 of differentiation, when approximately 25% of the differentiation cultures expressed the reporter construct, with expression increasing to approximately 70% at day 9 and continuing throughout the duration of spontaneous contractile activity exhibited by the ES cell-derived cardiac myocytes. Indirect immunofluorescence assays provide evidence that expression was restricted to the cardiac myocytes in culture. In the present study, we show vital staining of transgene expression in living cardiac myocytes using lipophilic fluorogenic beta-galactopyranoside substrates for real-time detection of the reporter gene during continuous contraction of the ES cell myocytes in vitro. The vital stain approach used in the present study will permit the identification of differentiating ES cells that are committed to the cardiac lineage for analysis of gene expression at early time points of ES cell cardiogenesis and, in addition, will aid in selecting genetically modified ES cell cardiac myocytes for use in functional studies.
Collapse
Affiliation(s)
- J M Metzger
- Department of Physiology, School of Medicine, University of Michigan, Ann Arbor 48109-0622, USA
| | | | | |
Collapse
|
32
|
Sidjanin D, Grdina D, Woloschak GE. UV-induced changes in cell cycle and gene expression within rabbit lens epithelial cells. Photochem Photobiol 1996; 63:79-85. [PMID: 8577869 DOI: 10.1111/j.1751-1097.1996.tb02995.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Damage to lens epithelial cells is a probable initiation process in cataract formation mediated by UV radiation. In these experiments, we investigated the effects of exposure to 254 nm radiation on cell cycle progression and gene expression in the rabbit lens epithelial cell line N/N1003A. The RNA was harvested at various times following exposure to UV (254 nm) radiation and analyzed by dot-blot and northern blot hybridizations. These results revealed that during the first 6 h following exposure of the cells to UV, there was, associated with decreasing dose, a decrease in accumulation of transcripts specific for histones H3 and H4 and an increase in the mRNA encoding protein kinase C and beta- and gamma-actin. Using flow cytometry, we detected an accumulation of cells in G1/S phase of the cell cycle 1 h following exposure to 254 nm radiation. The observed changes in gene expression, especially the decreased accumulation of histone transcripts reported here, may play a role in UV-induced inhibition of cell cycle progression.
Collapse
Affiliation(s)
- D Sidjanin
- Department of Biological Sciences, Northern Illinois University, Dekalb, USA
| | | | | |
Collapse
|
33
|
Tanaka M, Hiroe M, Ito H, Nishikawa T, Adachi S, Aonuma K, Marumo F. Differential localization of atrial natriuretic peptide and skeletal alpha-actin messenger RNAs in left ventricular myocytes of patients with dilated cardiomyopathy. J Am Coll Cardiol 1995; 26:85-92. [PMID: 7797780 DOI: 10.1016/0735-1097(95)00145-p] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVES This study was designed to determine whether atrial natriuretic peptide and skeletal alpha-actin messenger RNAs (mRNAs) are co-localized in ventricular myocytes of patients with dilated cardiomyopathy. BACKGROUND Atrial natriuretic peptide and skeletal alpha-actin are known as augmented genes with cardiac hypertrophy. However, the expression and localization of both genes in chronic failing heart remain unclear. METHODS Left ventricular biopsy specimens were obtained from 14 patients with dilated cardiomyopathy. Atrial natriuretic peptide and skeletal alpha-actin mRNAs were detected by in situ hybridization with specific sulfur-35 uridine triphosphate-labeled RNA probes in the serial sections. RESULTS Atrial natriuretic peptide mRNA was detected in 10 patients, and intense signals were localized in the myocytes located in the subendocardium and around the interstitial fibrous area. By contrast, skeletal alpha-actin mRNA was homogeneously detected in all myocytes in seven patients. By left ventriculography, patients with skeletal alpha-actin-positive findings had a lower ejection fraction (37.1 +/- 6.0%) than those with negative findings (46.3 +/- 5.8%, p < 0.05), but atrial natriuretic peptide mRNA expression was not related to left ventricular function. CONCLUSIONS These results indicate that the expression of atrial natriuretic peptide and skeletal alpha-actin mRNAs are not always co-localized in the left ventricle of patients with dilated cardiomyopathy and suggest that the mechanisms of the regulation of these two genes in the chronic failing heart are different.
Collapse
Affiliation(s)
- M Tanaka
- Second Department of Internal Medicine, Tokyo Medical and Dental University, Tokyo Women's Medical College, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Davey HW, Wildeman AG. Molecular analysis of bovine actin gene and pseudogene sequences: expression of nonmuscle and striated muscle isoforms in adult tissues. DNA Cell Biol 1995; 14:555-63. [PMID: 7598810 DOI: 10.1089/dna.1995.14.555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Most studies on the tissue distribution of actin isoform transcripts have been done in small mammals such as rat and mouse. We have begun a characterization of the actin gene family in a large mammal, the bovine. The alpha skeletal gene was isolated, and an isoform-specific probe to the 3' untranslated region of the transcript identified. This probe, in combination with isoform specific probes for alpha cardiac, beta nonmuscle, and gamma nonmuscle actins, was used to examine expression of nonmuscle and striated muscle actin gene transcription in different tissues. In contrast to other species so far examined, striated muscle isoforms were more strictly tissue specific, with virtually no alpha cardiac isoform transcripts detected in skeletal muscle and almost no alpha skeletal transcripts in cardiac tissue. The distribution of the beta and gamma nonmuscle actins was also unique in bovine compared to other species. A partial beta-actin pseudogene, and the chromosomal DNA flanking one end of it, were also cloned and sequenced. This chromosomal site was found to be homologous to a viral integration site previously identified in simian virus 40 (SV40)-transformed rat cells, suggesting that this region of the chromosome may be a preferred target for insertion events.
Collapse
Affiliation(s)
- H W Davey
- Department of Molecular Biology and Genetics, University of Guelph, Ontario, Canada
| | | |
Collapse
|
35
|
Ontell M, Ontell MP, Buckingham M. Muscle-specific gene expression during myogenesis in the mouse. Microsc Res Tech 1995; 30:354-65. [PMID: 7787235 DOI: 10.1002/jemt.1070300503] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Over the past decade, significant advances in molecular biological techniques have substantially increased our understanding of in vivo myogenesis, supplementing the information that previously had been obtained from classical embryological and morphological studies of muscle development. In this review, we have attempted to correlate morphogenetic events in developing murine muscle with the expression of genes encoding the MyoD family of myogenic regulatory factors and the contractile proteins. Differences in the pattern of expression of these genes in murine myotomal and limb muscle are discussed in the context of muscle cell lineage and environmental factors. The differences in gene expression in these two types of muscle suggest that no single coordinated pattern of gene activation is required during the initial formation of the muscles of the mouse.
Collapse
Affiliation(s)
- M Ontell
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pennsylvania 15261, USA
| | | | | |
Collapse
|
36
|
Gene Expression in Cardiac Hypertrophy. MOLECULAR BIOLOGY OF CARDIAC DEVELOPMENT AND GROWTH 1995. [DOI: 10.1007/978-3-662-22192-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
37
|
Ohta T. Further examples of evolution by gene duplication revealed through DNA sequence comparisons. Genetics 1994; 138:1331-7. [PMID: 7896112 PMCID: PMC1206269 DOI: 10.1093/genetics/138.4.1331] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
To test the theory that evolution by gene duplication occurs as a result of positive Darwinian selection that accompanies the acceleration of mutant substitutions, DNA sequences of recent duplication were analyzed by estimating the numbers of synonymous and nonsynonymous substitutions. For the troponin C family, at the period of differentiation of the fast and slow isoforms, amino acid substitutions were shown to have been accelerated relative to synonymous substitutions. Comparison of the first exon of alpha-actin genes revealed that amino acid substitutions were accelerated when the smooth muscle, skeletal and cardiac isoforms differentiated. Analysis of members of the heat shock protein 70 gene family of mammals indicates that heat shock responsive genes including duplicated copies are evolving rapidly, contrary to the cognitive genes which have been evolutionarily conservative. For the alpha 1-antitrypsin reactive center, the acceleration of amino acid substitution has been found for gene paris of recent duplication.
Collapse
Affiliation(s)
- T Ohta
- National Institute of Genetics, Mishima, Japan
| |
Collapse
|
38
|
Dunwoodie S, Joya J, Arkell R, Hardeman E. Multiple regions of the human cardiac actin gene are necessary for maturation-based expression in striated muscle. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)32703-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
39
|
Johnson TB, Kent RL, Bubolz BA, McDermott PJ. Electrical stimulation of contractile activity accelerates growth of cultured neonatal cardiocytes. Circ Res 1994; 74:448-59. [PMID: 8118953 DOI: 10.1161/01.res.74.3.448] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
An electrical stimulation system was designed to regulate synchronized contractile activity of neonatal rat cardiocytes and to examine the effects of mechanical contraction on cardiocyte growth. Continuous electrical stimulation at a pulse duration of 5 milliseconds and frequency of 3 Hz resulted in a time-dependent accumulation of cell protein that reached 34% above initial values, as measured by the protein-to-DNA ratio. The growth response did not occur using voltage amplitudes that were subthreshold for contraction and was independent of contraction frequencies set at > or = 0.5 Hz. The RNA-to-DNA ratio increased in parallel to cell protein, indicating that the capacity for protein synthesis was enhanced by contraction. Rates of 28S rRNA synthesis were accelerated twofold in contracting cardiocytes. By comparison, protein and RNA accumulation did not occur in electrically stimulated cardiocytes in which contraction was blocked by either 10 mumol/L verapamil or by 5 mmol/L 2,3-butanedione monoxime, an inhibitor of actomyosin crossbridge cycling. Electrical stimulation of cardiocyte contraction did not enhance alpha-cardiac actin or myosin heavy chain (alpha+beta) mRNA transcript levels relative to 28S rRNA during the period of rapid growth that occurred over the first 48 hours. It is concluded that (1) electrical stimulation of contraction accelerates cardiocyte growth and RNA accumulation, (2) mechanical contraction is involved in regulating the growth of electrically stimulated cardiocytes, and (3) the levels of alpha-actin and myosin heavy chain mRNA increase in proportion to rRNA during the growth of contracting cardiocytes.
Collapse
Affiliation(s)
- T B Johnson
- Department of Medicine, Medical University of South Carolina, Charleston
| | | | | | | |
Collapse
|
40
|
Pickles JO. An analysis of actin isoforms expressed in hair-cell enriched fractions of the chick basilar papilla by the polymerase chain reaction technique. Hear Res 1993; 71:225-9. [PMID: 8113140 DOI: 10.1016/0378-5955(93)90038-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Actin mRNA was characterised in hair-cell enriched fractions of the chick basilar papilla, by means of the reverse-transcription polymerase chain reaction technique. Primers were directed against the 3' untranslated portions of the actin mRNAs. Evidence for beta-cytoplasmic and gamma-cytoplasmic actin mRNA was found; no evidence was found for alpha-skeletal, alpha-cardiac or type 5 cytoplasmic actin mRNAs. Since beta-actin is known to form bundles of filaments whereas gamma-actin does not, this suggests that the hair-cell stereocilia are composed of beta-actin.
Collapse
Affiliation(s)
- J O Pickles
- Department of Physiology and Pharmacology, University of Queensland, Brisbane, Australia
| |
Collapse
|
41
|
Dudek EJ, Peak JG, Roth RM, Peak MJ. Isolation of V79 fibroblast cell lines containing elevated metallothionein levels that have increased resistance to the cytotoxic effects of ultraviolet-A radiation. Photochem Photobiol 1993; 58:836-40. [PMID: 8310006 DOI: 10.1111/j.1751-1097.1993.tb04980.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Isolated clones of V79 Chinese hamster lung fibroblasts, selected for resistance against cadmium toxicity, were exposed to monochromatic 365 nm ultraviolet-A (UVA; 320 nm to visible light) radiation and examined for cell survival. All three of the Cd-resistant V79 clones (V79Cd) tested exhibited significant increases in survival after irradiation compared with control cultures similar to the increased survival observed in Zn acetate-induced V79 cells. Dose-modifying factors calculated for these survival experiments were all approximately 1.5. When characterized for steady-state levels of metallothionein (MT) mRNA and associated Cd-binding activity, all of the Cd-resistant V79Cd clones demonstrated elevated constitutive levels of both, implicating MT as the mechanism responsible for the observed cellular resistance to Cd and also to 365 nm UVA radiation. However, whereas levels of intracellular MT protein correlated with differences in survival against Cd, MT intracellular levels did not correlate well with protection against 365 nm UVA. Increased cell survival after exposure to 365 nm UVA radiation mediated by MT appeared to reach a threshold level and MT only provided a limited degree of protection. Since UVA radiation is known to cause cell death mediated through the intracellular generation of reactive oxygen species (ROS), these results suggest that the role of MT in ameliorating cellular photooxidative damage produced by UVA is by reducing intracellular ROS.
Collapse
Affiliation(s)
- E J Dudek
- Center for Mechanistic Biology and Biotechnology, Argonne National Laboratory, IL 60439-4833
| | | | | | | |
Collapse
|
42
|
Richardson J, Pessin J. Identification of a skeletal muscle-specific regulatory domain in the rat GLUT4/muscle-fat gene. J Biol Chem 1993. [DOI: 10.1016/s0021-9258(19)36888-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
43
|
Abstract
BACKGROUND Normal gastrointestinal development is a complex process involving the precise integration of multiple cell types. To gain a better understanding of these processes, the present study examined isoactin gene expression in the adult rat gastrointestinal tract. METHODS Northern blot analysis was performed on specified segments of the adult rat esophagus, stomach, small intestine, cecum, colon, rectum, and anus using actin isoform-specific complementary DNAs for all six vertebrate isoactins. RESULTS Smooth muscle and cytoplasmic isoactins were heterogeneously coexpressed in a segment-specific manner throughout the gastrointestinal tract. In addition, striated muscle isoactin expression was also detected in segments of the adult rat esophagus, stomach, colon, cecum, rectum, and anus. Histological analysis indicated that the adult rat esophagus, stomach, and anus contained significant quantities of skeletal muscle, providing a source for the striated muscle isoactins detected in these gut segments. A similar source of striated muscle isoactin expression in the cecum, colon, and rectum was not identified. Both coordinate and independent regulation of isoactin gene expression was observed in the gastrointestinal tract, although distinct patterns of autoregulation were absent. CONCLUSIONS This study represents the first complete analysis of isoactin gene expression in the adult rat gastrointestinal tract and provides the basis for future studies designed to investigate the factors responsible for these processes.
Collapse
Affiliation(s)
- R A Liddell
- Department of Anatomy, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | |
Collapse
|
44
|
Ito H, Hiroe M, Hirata Y, Tsujino M, Adachi S, Shichiri M, Koike A, Nogami A, Marumo F. Insulin-like growth factor-I induces hypertrophy with enhanced expression of muscle specific genes in cultured rat cardiomyocytes. Circulation 1993; 87:1715-21. [PMID: 7683979 DOI: 10.1161/01.cir.87.5.1715] [Citation(s) in RCA: 276] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Cardiac hypertrophy is commonly observed in acromegalic patients, in whom serum insulin-like growth factor-I (IGF-I) levels are elevated. In the present study, we examined whether IGF-I induces hypertrophy in cultured neonatal rat cardiomyocytes through its specific receptor and whether IGF binding protein-3 (IGFBP-3), which is a major circulating carrier protein for IGF-I, inhibits IGF-I-induced cardiac hypertrophy in vitro. METHODS AND RESULTS Because the response of cardiac hypertrophy is characterized by the induction of expression for muscle-specific genes, the effect of IGF-I on steady-state levels of mRNA for myosin light chain-2 (MLC-2) and troponin I and for skeletal and cardiac alpha-actin isoforms was evaluated by Northern blot analysis. IGF-I (10(-7) M) increased mRNA levels for MLC-2 and troponin I as early as 60 minutes with maximum levels by 6 hours, which were maintained for as long as 24 hours. IGF-I (10(-7) M) also increased transcripts for skeletal alpha-actin but not for cardiac alpha-actin. The cell size as evaluated morphometrically was almost doubled after 48-hour treatment with IGF-I. IGF-I induction of protein synthesis was dose dependent (10(-10) to 10(-7) M) with a maximal 2.2-fold increase seen at 10(-8) M. In contrast to the hypertrophic effect of IGF-I, growth hormone affected neither protein synthesis nor expression for muscle-specific genes. Binding study using 125I-IGF-I revealed the presence of specific binding sites for IGF-I in rat cardiomyocytes. IGFBP-3 induced a dose-dependent inhibition of protein synthesis stimulated by IGF-I; IGFBP-3 (10(-7) M) completely inhibited the [3H]leucine uptake stimulated by IGF-I (10(-8) M). IGFBP-3 similarly inhibited the IGF-I-stimulated gene expressions for MLC-2 and troponin I. CONCLUSIONS These results suggest that IGF-I directly causes cardiac hypertrophy and that its effect can be blocked by IGFBP-3.
Collapse
Affiliation(s)
- H Ito
- Second Department of Internal Medicine, Tokyo Medical and Dental University, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kovilur S, Jacobson JW, Beach RL, Jeffery WR, Tomlinson CR. Evolution of the chordate muscle actin gene. J Mol Evol 1993; 36:361-8. [PMID: 8315656 DOI: 10.1007/bf00182183] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The ascidians Styela plicata, S. clava, and Mogula citrina are urochordates. The larvae of urochordates are considered to morphologically resemble the ancestral vertebrate. We asked whether larval and adult ascidian muscle actin sequences are nonmusclelike as in lower invertebrates, musclelike as in vertebrates, or possess characteristics of both. Nonmuscle and muscle actin cDNA clones from S. plicata were sequenced. Based on 27 diagnostic amino acids, which distinguish vertebrate muscle actin from other actins, we found that the deduced protein sequences of ascidian muscle actins exhibit similarities to both invertebrate and vertebrate muscle actins. A comparison to muscle actins from different vertebrate and invertebrate phylogenetic groups suggested that the urochordate muscle actins represent a transition from a nonmusclelike sequence to a vertebrate musclelike sequence. The ascidian adult muscle actin is more similar to skeletal actin and the larval muscle actin is more similar to cardiac actin, which indicates that the divergence of the skeletal and cardiac isoforms occurred before the emergence of urochordates. The muscle actin gene may be a powerful probe for investigating the chordate lineage.
Collapse
Affiliation(s)
- S Kovilur
- Department of Biology, University of Houston, TX 77204
| | | | | | | | | |
Collapse
|
46
|
|
47
|
Toyofuku T, Doyle DD, Zak R, Kordylewski L. Expression of phospholamban mRNA during early avian muscle morphogenesis is distinct from that of alpha-actin. Dev Dyn 1993; 196:103-13. [PMID: 8364220 DOI: 10.1002/aja.1001960204] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We have studied the expression of phospholamban during the early development of chick embryos by in situ hybridization and have compared it to that of alpha-cardiac and alpha-skeletal actin. In adult cross-striated muscles there is only one phospholamban gene and it is expressed exclusively in the heart and slow muscles. In the heart phospholamban transcripts were first detected at stage 14 in the region of presumptive ventricle and at stage 20 in the atrium. In the myotomal portion of the somites phospholamban mRNA was first detected at stage 20, which lagged behind the appearance of the alpha-actins. In the limb rudiments all three mRNAs were barely detectable through stage 24, but increased by stage 28+. However, quantitative analysis of signal intensity at stage 28+ indicated that less phospholamban mRNA is present in the limb bud than in the myotome since for phospholamban the ratio of the signal density in the myotome to that in the limb rudiments was about twice the value of the ratio determined for the alpha-actins. Northern blot analysis of embryonic day 11 chick fast pectoralis muscle showed that phospholamban mRNA was not detected in vivo while alpha-cardiac actin mRNA was. Moreover, no phospholamban mRNA was detected in primary cultures derived from pectoralis muscle of the same age. In concert with previous observations that phospholamban is not detectable at stage 30-32 in wing or thigh muscle, these results suggest that phospholamban mRNA is expressed independently of the alpha-actins in the limb buds during early myogenesis.
Collapse
Affiliation(s)
- T Toyofuku
- Department of Medicine, University of Chicago, Illinois 60637
| | | | | | | |
Collapse
|
48
|
Kovacs AM, Zimmer WE. Molecular cloning and expression of the chicken smooth muscle gamma-actin mRNA. CELL MOTILITY AND THE CYTOSKELETON 1993; 24:67-81. [PMID: 8319268 DOI: 10.1002/cm.970240108] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We have investigated the expression of chicken smooth muscle gamma-actin mRNA by isolation and characterization of cDNAs representing this actin isoform and utilizing the cDNA to probe RNA from adult and developing cells. Nucleotide sequence elucidated from an apparent full length smooth muscle gamma-actin cDNA revealed that it contained 94 bp of 5' non-translated sequence, an open reading frame of 1131 bp, and 97 bp of 3' non-translated sequence. Within the 376 amino acid sequence deduced from the chicken cDNA were diagnostic amino acids at the NH2- and COOH-terminal regions which provided unequivocal identification of the gamma-enteric smooth muscle actin isoform. In addition, the chicken gamma-enteric actin deduced from our cDNA clones was found to differ from the sequence reported in earlier protein studies [J. Vandekerckhove and K. Weber, FEBS Lett. 102:219, 1979] by containing a proline rather than a glutamine at position 359 of the protein, indicating that the avian gamma-enteric actin isoform is identical to its mammalian counterpart. Comparison of the 5' and 3' non-translated sequence determined from the chicken cDNA to that elucidated for rat, mouse, and human showed that there is not a high degree of cross-species sequence conservation outside of the coding regions among these mRNAs. Northern hybridization analyses demonstrated that the gamma-enteric actin mRNA is expressed in adult aorta and oviduct tissues but not in adult skeletal muscle, cardiac muscle, liver, brain, and spleen tissues. The gamma-enteric actin mRNA was first observed in measurable quantities in gizzard tissue from 4-5 day embryos and increased in content in developing smooth muscle cells through 16-17 embryonic days. Following this initial increase during embryonic development, the gamma-enteric actin mRNA exhibits a decline in content until approximately 7 days posthatching, after which there is an increase in content to maximal levels found in adult gizzard tissue. In general, the developmental appearance of the gamma-enteric mRNA parallels that observed for this protein in previous studies indicating that the developmental expression of smooth muscle gamma-actin is regulated, in part, by an increased content of mRNA in chicken visceral smooth muscle cells during myogenesis.
Collapse
Affiliation(s)
- A M Kovacs
- Department of Structural and Cellular Biology, University of South Alabama, School of Medicine, Mobile 36688
| | | |
Collapse
|
49
|
Brennan K, Hardeman E. Quantitative analysis of the human alpha-skeletal actin gene in transgenic mice. J Biol Chem 1993. [DOI: 10.1016/s0021-9258(18)54211-3] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
50
|
Affiliation(s)
- T Obinata
- Department of Biology, Faculty of Science, Chiba University, Japan
| |
Collapse
|