1
|
Pal Chowdhury M, Makarewicz C, Piezonka H, Buckley M. Novel Deep Eutectic Solvent-Based Protein Extraction Method for Pottery Residues and Archeological Implications. J Proteome Res 2022; 21:2619-2634. [PMID: 36268809 PMCID: PMC9639204 DOI: 10.1021/acs.jproteome.2c00340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Proteomic analysis of absorbed residues is increasingly
used to
identify the foodstuffs processed in ancient ceramic vessels, but
detailed methodological investigations in this field remain rare.
Here, we present three interlinked methodological developments with
important consequences in paleoproteomics: the comparative absorption
and identification of various food proteins, the application of a
deep eutectic solvent (DES) for extracting ceramic-bound proteins,
and the role of database choice in taxonomic identification. Our experiments
with modern and ethnoarcheological ceramics show that DES is generally
more effective at extracting ceramic-bound proteins than guanidine
hydrochloride (GuHCl), and cereal proteins are absorbed and subsequently
extracted and identifiedat least as readily as meat proteins. We also
highlight some of the challenges in cross-species proteomics, whereby
species that are less well-represented in databases can be attributed
an incorrect species-level taxonomic assignment due to interspecies
similarities in protein sequence. This is particularly problematic
in potentially mixed samples such as cooking-generated organic residues
deposited in pottery. Our work demonstrates possible proteomic separation
of fishes and birds, the latter of which have so far eluded detection
through lipidomic analyses of organic residue deposits in pottery,
which has important implications for tracking the exploitation of
avian species in various ancient communities around the globe.
Collapse
Affiliation(s)
- Manasij Pal Chowdhury
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K.,Interdisciplinary Centre for Ancient Life, Department of Earth and Environmental Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Cheryl Makarewicz
- Institute for Prehistoric and Protohistoric Archaeology, Kiel University, Johanna-Mestorf Strasse 2-6, Kiel D-24118, Germany.,Cluster of Excellence ROOTS: Social, Environmental, and Cultural Connectivity in Past Societies, Kiel University, Leibniz Strasse 1, Kiel 24118, Germany
| | - Henny Piezonka
- Institute for Prehistoric and Protohistoric Archaeology, Kiel University, Johanna-Mestorf Strasse 2-6, Kiel D-24118, Germany.,Cluster of Excellence ROOTS: Social, Environmental, and Cultural Connectivity in Past Societies, Kiel University, Leibniz Strasse 1, Kiel 24118, Germany
| | - Michael Buckley
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K.,Interdisciplinary Centre for Ancient Life, Department of Earth and Environmental Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| |
Collapse
|
2
|
Morla S, Deguchi H, Griffin JH. Skeletal muscle myosin and cardiac myosin attenuate heparin's antithrombin-dependent anticoagulant activity. J Thromb Haemost 2021; 19:470-477. [PMID: 33176060 PMCID: PMC7902397 DOI: 10.1111/jth.15169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/23/2020] [Accepted: 11/03/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Heparin enhances the ability of the plasma protease inhibitor, antithrombin, to neutralize coagulation factor Xa and thrombin. Skeletal muscle myosin binds unfractionated heparin. OBJECTIVES The aim of this study was to investigate the influence of myosin binding to heparin on antithrombin's anticoagulant activity. METHODS Inhibition of factor Xa and thrombin by antithrombin in the presence of different heparins and skeletal muscle myosin or cardiac myosin was studied by measuring inhibition of each enzyme's chromogenic substrate hydrolysis. RESULTS AND CONCLUSIONS Skeletal muscle myosin and cardiac myosin neutralized unfractionated heparin's enhancement of antithrombin's inhibition of purified factor Xa and thrombin. Skeletal muscle myosin also reduced the inhibition of factor Xa and thrombin by antithrombin in the presence of heparan sulfate. These two myosins did not protect factor Xa from antithrombin inhibition when tested in the presence of smaller heparins (eg, low molecular weight heparin, heparin pentasaccharide). This chain length dependence for skeletal muscle myosin's ability to reduce heparin's anticoagulant activity might have potential implications for therapy for patients who experience increases in plasma myosin levels (eg, acute trauma patients). In addition to the chain length, the type and extent of sulfation of glycosaminoglycans influenced the ability of skeletal muscle myosin to neutralize the polysaccharide's ability to enhance antithrombin's activity. In summary, these studies show that skeletal muscle myosin and cardiac myosin can influence antithrombin's anticoagulant activity against factor Xa and thrombin, implying that they may significantly influence the hemostatic balance involving bleeding vs clotting.
Collapse
Affiliation(s)
- Shravan Morla
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Hiroshi Deguchi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Division of Hematology, Department of Medicine, University of California, San Diego CA 92094
| |
Collapse
|
3
|
Thermal acclimation to cold alters myosin content and contractile properties of rainbow smelt, Osmerus mordax, red muscle. Comp Biochem Physiol A Mol Integr Physiol 2016; 196:46-53. [DOI: 10.1016/j.cbpa.2016.02.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/26/2016] [Accepted: 02/29/2016] [Indexed: 11/20/2022]
|
4
|
Khodabukus A, Baehr LM, Bodine SC, Baar K. Role of contraction duration in inducing fast-to-slow contractile and metabolic protein and functional changes in engineered muscle. J Cell Physiol 2015; 230:2489-97. [PMID: 25857846 DOI: 10.1002/jcp.24985] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/04/2015] [Indexed: 12/26/2022]
Abstract
The role of factors such as frequency, contraction duration and active time in the adaptation to chronic low-frequency electrical stimulation (CLFS) is widely disputed. In this study we explore the ability of contraction duration (0.6, 6, 60, and 600 sec) to induce a fast-to-slow shift in engineered muscle while using a stimulation frequency of 10 Hz and keeping active time constant at 60%. We found that all contraction durations induced similar slowing of time-to-peak tension. Despite similar increases in total myosin heavy (MHC) levels with stimulation, increasing contraction duration resulted in progressive decreases in total fast myosin. With contraction durations of 60 and 600 sec, MHC IIx levels decreased and MHC IIa levels increased. All contraction durations resulted in fast-to-slow shifts in TnT and TnC but increased both fast and slow TnI levels. Half-relaxation slowed to a greater extent with contraction durations of 60 and 600 sec despite similar changes in the calcium sequestering proteins calsequestrin and parvalbumin and the calcium uptake protein SERCA. All CLFS groups resulted in greater fatigue resistance than control. Similar increases in GLUT4, mitochondrial enzymes (SDH and ATPsynthase), the fatty acid transporter CPT-1, and the metabolic regulators PGC-1α and MEF2 were found with all contraction durations. However, the mitochondrial enzymes cytochrome C and citrate synthase were increased to greater levels with contraction durations of 60 and 600 sec. These results demonstrate that contraction duration plays a pivotal role in dictating the level of CLFS-induced contractile and metabolic adaptations in tissue-engineered skeletal muscle.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Division of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| | - Leslie M Baehr
- Division of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| | - Sue C Bodine
- Division of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| | - Keith Baar
- Division of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| |
Collapse
|
5
|
Ojima K, Ichimura E, Yasukawa Y, Wakamatsu JI, Nishimura T. Dynamics of myosin replacement in skeletal muscle cells. Am J Physiol Cell Physiol 2015; 309:C669-79. [PMID: 26377314 DOI: 10.1152/ajpcell.00170.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/06/2015] [Indexed: 01/04/2023]
Abstract
Highly organized thick filaments in skeletal muscle cells are formed from ~300 myosin molecules. Each thick-filament-associated myosin molecule is thought to be constantly exchanged. However, the mechanism of myosin replacement remains unclear, as does the source of myosin for substitution. Here, we investigated the dynamics of myosin exchange in the myofibrils of cultured myotubes by fluorescent recovery after photobleaching and found that myofibrillar myosin is actively replaced with an exchange half-life of ~3 h. Myosin replacement was not disrupted by the absence of the microtubule system or by actomyosin interactions, suggesting that known cytoskeletal systems are dispensable for myosin substitution. Intriguingly, myosin replacement was independent of myosin binding protein C, which links myosin molecules together to form thick filaments. This implies that an individual myosin molecule rather than a thick filament functions as an exchange unit. Furthermore, the myosin substitution rate was decreased by the inhibition of protein synthesis, suggesting that newly synthesized myosin, as well as preexisting cytosolic myosin, contributes to myosin replacement in myofibrils. Notably, incorporation and release of myosin occurred simultaneously in myofibrils, but rapid myosin release from myofibrils was observed without protein synthesis. Collectively, our results indicate that myosin shuttles between myofibrils and the nonmyofibrillar cytosol to maintain a dynamic equilibrium in skeletal muscle cells.
Collapse
Affiliation(s)
- Koichi Ojima
- Animal Products Research Division, NARO Institute of Livestock and Grassland Science, Ibaraki, Japan; and
| | - Emi Ichimura
- Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Yuya Yasukawa
- Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Jun-Ichi Wakamatsu
- Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Takanori Nishimura
- Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
6
|
Recher G, Coumailleau P, Rouède D, Tiaho F. Structural origin of the drastic modification of second harmonic generation intensity pattern occurring in tail muscles of climax stages xenopus tadpoles. J Struct Biol 2015; 190:1-10. [PMID: 25770062 DOI: 10.1016/j.jsb.2015.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 02/08/2015] [Accepted: 03/04/2015] [Indexed: 01/27/2023]
Abstract
Second harmonic generation (SHG) microscopy is a powerful tool for studying submicron architecture of muscles tissues. Using this technique, we show that the canonical single frequency sarcomeric SHG intensity pattern (SHG-IP) of premetamorphic xenopus tadpole tail muscles is converted to double frequency (2f) sarcomeric SHG-IP in metamorphic climax stages due to massive physiological muscle proteolysis. This conversion was found to rise from 7% in premetamorphic muscles to about 97% in fragmented muscular apoptotic bodies. Moreover a 66% conversion was also found in non-fragmented metamorphic tail muscles. Also, a strong correlation between predominant 2f sarcomeric SHG-IPs and myofibrillar misalignment is established with electron microscopy. Experimental and theoretical results demonstrate the higher sensitivity and the supra resolution power of SHG microscopy over TPEF to reveal 3D myofibrillar misalignment. From this study, we suggest that 2f sarcomeric SHG-IP could be used as signature of triad defect and disruption of excitation-contraction coupling. As the mechanism of muscle proteolysis is similar to that found in mdx mouse muscles, we further suggest that xenopus tadpole tail resorption at climax stages could be used as an alternative or complementary model of Duchene muscular dystrophy.
Collapse
Affiliation(s)
- Gaëlle Recher
- UMR CNRS 6026, Université de Rennes1, Campus de Beaulieu, Rennes F-35000, France; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Pascal Coumailleau
- UMR CNRS 6026, Université de Rennes1, Campus de Beaulieu, Rennes F-35000, France; IRSET, INSERM, U1085, Université de Rennes1, Campus de Beaulieu, Rennes F-35000, France
| | - Denis Rouède
- IPR, CNRS, UMR-CNRS UR1-6251, Université de Rennes1, Campus de Beaulieu, Rennes F-35000, France
| | - François Tiaho
- UMR CNRS 6026, Université de Rennes1, Campus de Beaulieu, Rennes F-35000, France; IRSET, INSERM, U1085, Université de Rennes1, Campus de Beaulieu, Rennes F-35000, France.
| |
Collapse
|
7
|
Coughlin DJ, Akhtar M. Contractile properties of the myotomal muscle of sheepshead, Archosargus probatocephalus. ACTA ACUST UNITED AC 2015; 323:169-78. [PMID: 25676176 DOI: 10.1002/jez.1904] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/17/2014] [Accepted: 10/21/2014] [Indexed: 11/12/2022]
Abstract
Swimming in fishes is powered by myotomal red, white and pink skeletal muscle. Slow swimming is powered by the red (slow-twitch muscle), fast speeds are achieved by the white (fast-twitch) muscle and pink muscle apparently serves an intermediate function. In recent years, the physiological properties and molecular composition of red (slow) and white (fast) muscle fibers have been well studied, while the intermediate pink muscle, which falls in a thin sheet between the superficial red muscle and deeper white muscle, has received less attention. The goal of this study is to determine the contractile properties of red, pink, and white muscle and to establish the molecular basis of fiber type variations in contractile properties in a sheepshead (Archosargus probatocephalus). Isometric and isovelocity muscle mechanics experiments demonstrated a general pattern of increasing contractile speed from red to pink to white muscle, although red and pink muscle did not differ significantly for most contraction kinetics variables. As myosin heavy chain (MyHC) is the most important structural protein found in the muscle fibers, MyHC content was examined through immunohistochemistry. Myosin antibodies suggest a gradient in myosin content corresponding to differences in muscle contraction kinetics.
Collapse
Affiliation(s)
- David J Coughlin
- Department of Biology, Widener University, Chester, Pennsylvania
| | | |
Collapse
|
8
|
Saita S, Shirane M, Ishitani T, Shimizu N, Nakayama KI. Role of the ANKMY2-FKBP38 axis in regulation of the Sonic hedgehog (Shh) signaling pathway. J Biol Chem 2014; 289:25639-54. [PMID: 25077969 DOI: 10.1074/jbc.m114.558635] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sonic hedgehog (Shh) is a secreted morphogen that controls the patterning and growth of various tissues in the developing vertebrate embryo, including the central nervous system. Ablation of the FK506-binding protein 38 (FKBP38) gene results in activation of the Shh signaling pathway in mouse embryos, but the molecular mechanism by which FKBP38 suppresses Shh signaling has remained unclear. With the use of a proteomics approach, we have now identified ANKMY2, a protein with three ankyrin repeats and a MYND (myeloid, Nervy, and DEAF-1)-type Zn(2+) finger domain, as a molecule that interacts with FKBP38. Co-immunoprecipitation analysis confirmed that endogenous FKBP38 and ANKMY2 interact in the mouse brain. Depletion or overexpression of ANKMY2 resulted in down- and up-regulation of Shh signaling, respectively, in mouse embryonic fibroblasts. Furthermore, combined depletion of both FKBP38 and ANKMY2 attenuated Shh signaling in these cells, suggesting that ANKMY2 acts downstream of FKBP38 to activate the Shh signaling pathway. Targeting of the zebrafish ortholog of mouse Ankmy2 (ankmy2a) in fish embryos with an antisense morpholino oligonucleotide conferred a phenotype reflecting loss of function of the Shh pathway, suggesting that the regulation of Shh signaling by ANKMY2 is conserved between mammals and fish. Our findings thus indicate that the FKBP38-ANKMY2 axis plays a key role in regulation of Shh signaling in vivo.
Collapse
Affiliation(s)
| | | | - Tohru Ishitani
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Nobuyuki Shimizu
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | | |
Collapse
|
9
|
Ha K, Buchan JG, Alvarado DM, McCall K, Vydyanath A, Luther PK, Goldsmith MI, Dobbs MB, Gurnett CA. MYBPC1 mutations impair skeletal muscle function in zebrafish models of arthrogryposis. Hum Mol Genet 2013; 22:4967-77. [PMID: 23873045 DOI: 10.1093/hmg/ddt344] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Myosin-binding protein C1 (MYBPC1) is an abundant skeletal muscle protein that is expressed predominantly in slow-twitch muscle fibers. Human MYBPC1 mutations are associated with distal arthrogryposis type 1 and lethal congenital contracture syndrome type 4. As MYBPC1 function is incompletely understood, the mechanism by which human mutations result in contractures is unknown. Here, we demonstrate using antisense morpholino knockdown, that mybpc1 is required for embryonic motor activity and survival in a zebrafish model of arthrogryposis. Mybpc1 morphant embryos have severe body curvature, cardiac edema, impaired motor excitation and are delayed in hatching. Myofibril organization is selectively impaired in slow skeletal muscle and sarcomere numbers are greatly reduced in mybpc1 knockdown embryos, although electron microscopy reveals normal sarcomere structure. To evaluate the effects of human distal arthrogryposis mutations, mybpc1 mRNAs containing the corresponding human W236R and Y856H MYBPC1 mutations were injected into embryos. Dominant-negative effects of these mutations were suggested by the resultant mild bent body curvature, decreased motor activity, as well as impaired overall survival compared with overexpression of wild-type RNA. These results demonstrate a critical role for mybpc1 in slow skeletal muscle development and establish zebrafish as a tractable model of human distal arthrogryposis.
Collapse
|
10
|
Campion LA, Choi S, Mistry HL, Coughlin DJ. Myosin heavy chain and parvalbumin expression in swimming and feeding muscles of centrarchid fishes: the molecular basis of the scaling of contractile properties. Comp Biochem Physiol A Mol Integr Physiol 2012; 163:223-30. [PMID: 22705556 DOI: 10.1016/j.cbpa.2012.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 06/04/2012] [Accepted: 06/04/2012] [Indexed: 11/19/2022]
Abstract
In centrarchid fishes, such as bluegill (Lepomis macrochirus, Rafinesque) and largemouth bass (Micropterus salmoides, Lacepède), the contractile properties of feeding and swimming muscles show different scaling patterns. While the maximum shortening velocity (V(max)) and rate of relaxation from tetanus of swimming or myotomal muscle slow with growth, the feeding muscle shows distinctive scaling patterns. Cranial epaxial muscle, which is used to elevate the head during feeding strikes, retains fast contractile properties across a range of fish sizes in both species. In bass, the sternohyoideous muscle, which depresses the floor of the mouth during feeding strikes, shows faster contractile properties with growth. The objective of this study was to determine the molecular basis of these different scaling patterns. We examined the expression of two muscle proteins, myosin heavy chain (MyHC) and parvalbumin (PV), that affect contractile properties. We hypothesized that the relative contribution of slow and fast MyHC isoforms will modulate V(max) in these fishes, while the presence of PV in muscle will enhance rates of muscle relaxation. Myotomal muscle displays an increase in sMyHC expression with growth, in agreement with its physiological properties. Feeding muscles such as epaxial and sternohyoideus show no change or a decrease in sMyHC expression with growth, again as predicted from contractile properties. PV expression in myotomal muscle decreases with growth in both species, as has been seen in other fishes. The feeding muscles again show no change or an increase in PV expression with growth, contributing to faster contractile properties in these fishes. Both MyHC and PV appear to play important roles in modulating muscle contractile properties of swimming and feeding muscles in centrarchid fishes.
Collapse
Affiliation(s)
- L A Campion
- Department of Biology, Widener University, Chester, PA 19013, USA
| | | | | | | |
Collapse
|
11
|
Bernick EP, Zhang PJ, Du S. Knockdown and overexpression of Unc-45b result in defective myofibril organization in skeletal muscles of zebrafish embryos. BMC Cell Biol 2010; 11:70. [PMID: 20849610 PMCID: PMC2954953 DOI: 10.1186/1471-2121-11-70] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 09/17/2010] [Indexed: 02/01/2023] Open
Abstract
Background Unc-45 is a myosin chaperone and a Hsp90 co-chaperone that plays a key role in muscle development. Genetic and biochemical studies in C. elegans have demonstrated that Unc-45 facilitates the process of myosin folding and assembly in body wall muscles. Loss or overexpression of Unc-45 in C. elegans results in defective myofibril organization. In the zebrafish Danio rerio, unc-45b, a homolog of C. elegans unc-45, is expressed in both skeletal and cardiac muscles. Earlier studies indicate that mutation or knockdown of unc-45b expression in zebrafish results in a phenotype characterized by a loss of both thick and thin filament organization in skeletal and cardiac muscle. The effects of unc-45b knockdown on other sarcomeric structures and the phenotype of Unc-45b overexpression, however, are poorly understood in vertebrates. Results Both knockdown and overexpression provide useful tools to study gene function during animal development. Using such methods, we characterized the role of Unc-45b in myofibril assembly of skeletal muscle in Danio rerio. We showed that, in addition to thick and thin filament defects, knockdown of unc-45b expression disrupted sarcomere organization in M-lines and Z-lines of skeletal muscles in zebrafish embryos. Western blotting analysis showed that myosin protein levels were significantly decreased in unc-45b knockdown embryos. Similarly, embryos overexpressing Unc-45b also exhibited severely disorganized myosin thick filaments. Disruption of thick filament organization by Unc-45b overexpression depends on the C-terminal UCS domain in Unc-45b required for interaction with myosin. Deletion of the C-terminal UCS domain abolished the disruptive activity of Unc-45b in myosin thick filament organization. In contrast, deletion of the N-terminal TPR domain required for binding with Hsp90α had no effect. Conclusion Collectively, these studies indicate that the expression levels of Unc-45b must be precisely regulated to ensure normal myofibril organization. Loss or overexpression of Unc-45b leads to defective myofibril organization.
Collapse
Affiliation(s)
- Elena P Bernick
- University of Maryland School of Medicine Interdisciplinary Training Program in Muscle Biology, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
12
|
Beermann ML, Ardelt M, Girgenrath M, Miller JB. Prdm1 (Blimp-1) and the expression of fast and slow myosin heavy chain isoforms during avian myogenesis in vitro. PLoS One 2010; 5:e9951. [PMID: 20376350 PMCID: PMC2848592 DOI: 10.1371/journal.pone.0009951] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 03/09/2010] [Indexed: 01/07/2023] Open
Abstract
Background Multiple types of fast and slow skeletal muscle fibers form during early embryogenesis in vertebrates. In zebrafish, formation of the earliest slow myofibers in fin muscles requires expression of the zinc-finger transcriptional repressor Prdm1 (also known as Blimp1). To further understand how the role of Prdm1 in early myogenesis may vary through evolution and during development, we have now analyzed Prdm1 expression in the diverse types of myotubes that form in culture from somitic, embryonic, and fetal chicken myoblasts. Principal Findings In cultures of somitic, embryonic limb, and fetal limb chicken cells, we found that Prdm1 was expressed in all of the differentiated muscle cells that formed, including those that expressed only fast myosin heavy chain isoforms, as well as those that co-expressed both fast and slow myosin heavy chain isoforms. Prdm1 was also expressed in Pax7-positive myoblasts, as well as in non-myogenic cells in the cultures. Furthermore, though all differentiated cells in control somite cultures co-expressed fast and slow myosin heavy chains, antisense knockdown of Prdm1 expression inhibited the formation of these co-expressing cells in somite cultures. Conclusions In chicken myogenic cell cultures, Prdm1 was expressed in most Pax7-positive myoblasts and in all differentiated muscle cells, irrespective of the developmental stage of cell donor or the pattern of fast and slow myosin heavy chains expressed in the differentiated cells that were formed. Thus, Prdm1 was expressed in myogenic cells prior to terminal differentiation; and, after differentiation, Prdm1 expression was not limited to cells that expressed slow myosin heavy chain isoforms. In addition, Prdm1 appeared to be required for differentiation of the somitic myocytes, which are the earliest myocytes to form in the avian embryo.
Collapse
Affiliation(s)
- Mary Lou Beermann
- Neuromuscular Biology & Disease Group, Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America
| | - Magdalena Ardelt
- Neuromuscular Biology & Disease Group, Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America
| | - Mahasweta Girgenrath
- Department of Health Science, Boston University, Boston, Massachusetts, United States of America
| | - Jeffrey Boone Miller
- Neuromuscular Biology & Disease Group, Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, Boston Biomedical Research Institute, Watertown, Massachusetts, United States of America
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
13
|
Vishnudas VK, Miller JB. Ku70 regulates Bax-mediated pathogenesis in laminin-alpha2-deficient human muscle cells and mouse models of congenital muscular dystrophy. Hum Mol Genet 2009; 18:4467-77. [PMID: 19692349 DOI: 10.1093/hmg/ddp399] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The severely debilitating disease Congenital Muscular Dystrophy Type 1A (MDC1A) is caused by mutations in the gene encoding laminin-alpha2. Bax-mediated muscle cell death is a significant contributor to the severe neuromuscular pathology seen in the Lama2-null mouse model of MDC1A. To extend our understanding of pathogenesis due to laminin-alpha2-deficiency, we have now analyzed molecular mechanisms of Bax regulation in normal and laminin-alpha2-deficient muscles and cells, including myogenic cells obtained from patients with a clinical diagnosis of MDC1A. In mouse myogenic cells, we found that, as in non-muscle cells, Bax co-immunoprecipitated with the multifunctional protein Ku70. In addition, cell permeable pentapeptides designed from Ku70, termed Bax-inhibiting peptides (BIPs), inhibited staurosporine-induced Bax translocation and cell death in mouse myogenic cells. We also found that acetylation of Ku70, which can inhibit binding to Bax and can be an indicator of increased susceptibility to cell death, was more abundant in Lama2-null than in normal mouse muscles. Furthermore, myotubes formed in culture from human laminin-alpha2-deficient patient myoblasts produced high levels of activated caspase-3 when grown on poly-L-lysine, but not when grown on a laminin-alpha2-containing substrate or when treated with BIPs. Finally, cytoplasmic Ku70 in human laminin-alpha2-deficient myotubes was both reduced in amount and more highly acetylated than in normal myotubes. Increased susceptibility to cell death thus appears to be an intrinsic property of human laminin-alpha2-deficient myotubes. These results identify Ku70 as a regulator of Bax-mediated pathogenesis and a therapeutic target in laminin-alpha2-deficiency.
Collapse
Affiliation(s)
- Vivek K Vishnudas
- Neuromuscular Biology & Disease Group, Boston Biomedical Research Institute, 64 Grove Street, Watertown, MA 02478, USA
| | | |
Collapse
|
14
|
Welch KC, Altshuler DL. Fiber type homogeneity of the flight musculature in small birds. Comp Biochem Physiol B Biochem Mol Biol 2009; 152:324-31. [PMID: 19162216 DOI: 10.1016/j.cbpb.2008.12.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 12/16/2008] [Accepted: 12/17/2008] [Indexed: 10/21/2022]
Abstract
Studies of medium- and large-bodied avian species have suggested that variation in flight muscle composition is related to differences in flight behavior. For example, slow-twitch or tonic fibers are generally found only in the flight muscles of non-volant or soaring/gliding birds. However, we know comparatively little about fiber composition of the muscles of the smallest birds. Here we describe the fiber composition of muscles from the wings, shoulders, and legs of two small avian species, which also display very high wingbeat frequencies: Anna's hummingbirds (Calypte anna) and zebra finches (Taeniopygia guttata). All flight muscles examined in both species contained exclusively fast oxidative glycolytic (FOG) fibers. These unique results suggest that fast oxidative fibers are both necessary and sufficient for the full range of flight behaviors in these small-bodied birds. Like all other studied birds, the zebra finch gastrocnemius, a tarsometatarsal extensor, contained a mixture of FOG (27.1%), slow oxidative (SO, 12.7%), and fast glycolytic (FG, 60.2%) fibers. By contrast, the hummingbird gastrocnemius lacked FG fibers (85.5% FOG, 14.5% SO), which may reflect the reduced role of the hindlimb during take-off. We further hypothesize that thermogenic requirements constrain fiber type heterogeneity in these small endothermic vertebrates.
Collapse
Affiliation(s)
- Kenneth C Welch
- Department of Biology, University of California, Riverside, 92521-0427, USA
| | | |
Collapse
|
15
|
Ahhmed AM, Kuroda R, Kawahara S, Ohta K, Nakade K, Aoki T, Muguruma M. Dependence of microbial transglutaminase on meat type in myofibrillar proteins cross-linking. Food Chem 2009. [DOI: 10.1016/j.foodchem.2008.05.078] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
Menelaou E, Husbands EE, Pollet RG, Coutts CA, Ali DW, Svoboda KR. Embryonic motor activity and implications for regulating motoneuron axonal pathfinding in zebrafish. Eur J Neurosci 2008; 28:1080-96. [PMID: 18823502 PMCID: PMC2741004 DOI: 10.1111/j.1460-9568.2008.06418.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Zebrafish embryos exhibit spontaneous contractions of the musculature as early as 18-19 h post fertilization (hpf) when removed from their protective chorion. These movements are likely initiated by early embryonic central nervous system activity. We have made the observation that narrowminded mutant embryos (hereafter, nrd(-/-)) lack normal embryonic motor output upon dechorionation. However, these mutants can swim and respond to tactile stimulation by larval stages of development. nrd(-/-) embryos exhibit defects in neural crest development, slow muscle development and also lack spinal mechanosensory neurons known as Rohon-Beard (RB) neurons. At early developmental stages (i.e. 21-22 hpf) and while still in their chorions, nrd siblings (nrd(+/?)) exhibited contractions of the musculature at a rate similar to wild-type embryos. Anatomical analysis indicated that RB neurons were present in the motile embryos, but absent in the non-motile embryos, indicating that the non-motile embryos were nrd(-/-) embryos. Further anatomical analysis of nrd(-/-) embryos revealed errors in motoneuron axonal pathfinding that persisted into the larval stage of development. These errors were reversed when nrd(-/-) embryos were raised in high [K(+)] beginning at 21 hpf, indicating that the abnormal axonal phenotypes may be related to a lack of depolarizing activity early in development. When activity was blocked with tricaine in wild-type embryos, motoneuron phenotypes were similar to the motoneuron phenotypes in nrd(-/-) embryos. These results implicate early embryonic activity in conjunction with other factors as necessary for normal motoneuron development.
Collapse
Affiliation(s)
- Evdokia Menelaou
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | | | | | |
Collapse
|
17
|
Gofflot S, Kischel P, Thielen C, Radermacher V, Boniver J, de Leval L. Characterization of an antibody panel for immunohistochemical analysis of canine muscle cells. Vet Immunol Immunopathol 2008; 125:225-33. [DOI: 10.1016/j.vetimm.2008.05.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 05/08/2008] [Accepted: 05/27/2008] [Indexed: 10/22/2022]
|
18
|
Matsuoka Y, Inoue A. Controlled differentiation of myoblast cells into fast and slow muscle fibers. Cell Tissue Res 2008; 332:123-32. [DOI: 10.1007/s00441-008-0582-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Accepted: 01/14/2008] [Indexed: 10/22/2022]
|
19
|
The chemokine Sdf-1 and its receptor Cxcr4 are required for formation of muscle in zebrafish. BMC DEVELOPMENTAL BIOLOGY 2007; 7:54. [PMID: 17517144 PMCID: PMC1904199 DOI: 10.1186/1471-213x-7-54] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2006] [Accepted: 05/22/2007] [Indexed: 02/05/2023]
Abstract
BACKGROUND During development cell migration takes place prior to differentiation of many cell types. The chemokine receptor Cxcr4 and its ligand Sdf1 are implicated in migration of several cell lineages, including appendicular muscles. RESULTS We dissected the role of sdf1-cxcr4 during skeletal myogenesis. We demonstrated that the receptor cxcr4a is expressed in the medial-anterior part of somites, suggesting that chemokine signaling plays a role in this region of the somite. Previous reports emphasized co-operation of Sdf1a and Cxcr4b. We found that during early myogenesis Sdf1a co-operates with the second Cxcr4 of zebrafish - Cxcr4a resulting in the commitment of myoblast to form fast muscle. Disrupting this chemokine signal caused a reduction in myoD and myf5 expression and fast fiber formation. In addition, we showed that a dimerization partner of MyoD and Myf5, E12, positively regulates transcription of cxcr4a and sdf1a in contrast to that of Sonic hedgehog, which inhibited these genes through induction of expression of id2. CONCLUSION We revealed a regulatory feedback mechanism between cxcr4a-sdf1a and genes encoding myogenic regulatory factors, which is involved in differentiation of fast myofibers. This demonstrated a role of chemokine signaling during development of skeletal muscles.
Collapse
|
20
|
Wilson JE. The use of monoclonal antibodies and limited proteolysis in elucidation of structure-function relationships in proteins. METHODS OF BIOCHEMICAL ANALYSIS 2006; 35:207-50. [PMID: 2002771 DOI: 10.1002/9780470110560.ch4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- J E Wilson
- Biochemistry Department, Michigan State University, East Lansing
| |
Collapse
|
21
|
Archila S, King MA, Carlson GM, Rice NA. The cytoskeletal organizing protein Cdc42-interacting protein 4 associates with phosphorylase kinase in skeletal muscle. Biochem Biophys Res Commun 2006; 345:1592-9. [PMID: 16735024 DOI: 10.1016/j.bbrc.2006.05.073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Accepted: 05/11/2006] [Indexed: 11/18/2022]
Abstract
Phosphorylase kinase is a key enzyme in regulating glycogenolytic flux in skeletal muscle in response to changing energy demands. In the present study, we sought to identify interacting proteins of phosphorylase kinase by yeast two-hybrid screening. Screening a rabbit skeletal muscle cDNA library with the exposed C-terminus of the alpha subunit (residues 1060-1237), we identified eight independent, yet overlapping, constructs of cdc42-interacting protein 4 (CIP4). Immunocytochemistry indicated that CIP4 colocalized with phosphorylase kinase in vivo, and the cognate binding domain on CIP4 was determined to lie between residues 398 and 545. While this region of CIP4 does contain a known src homology 3 domain, transient transfections and coimmunoprecipitation experiments showed that this domain is not responsible for the dimeric interaction. Based upon sequence analysis the association is inferred to be mediated by two proline-rich sequences in CIP4, residues 436-439 and 441-444, that bind to a cognate WW domain found between residues 1107 and 1129 of PhKalpha.
Collapse
Affiliation(s)
- Soleil Archila
- Department of Biology, Western Kentucky University, Bowling Green, 42101-1080, USA
| | | | | | | |
Collapse
|
22
|
Beylkin DH, Allen DL, Leinwand LA. MyoD, Myf5, and the calcineurin pathway activate the developmental myosin heavy chain genes. Dev Biol 2006; 294:541-53. [PMID: 16584724 DOI: 10.1016/j.ydbio.2006.02.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2005] [Revised: 12/05/2005] [Accepted: 02/24/2006] [Indexed: 11/30/2022]
Abstract
Myogenesis is accompanied by the activation of two developmental myosin heavy chains (MyHCs), embryonic and perinatal, followed by a dramatic decrease in their expression during early postnatal life. The pathways that control the transcription of these genes have not been previously determined. In this study, we identified cis-acting elements and trans-acting factors that regulate the expression of these two developmental MyHCs in the mouse. Between 800 and 1000 bp of proximal promoter sequence is sufficient to drive muscle-specific expression in cell culture. Further, these same regions contain sequences that confer downregulation in postnatal life in vivo. For the embryonic MyHC gene, the region between -791 bp and -626 bp contains the majority of activating elements. In the proximal promoter regions of both genes, we identified two E-box elements that work in conjunction to activate transcription, but only the embryonic MyHC E-boxes bind a complex containing MyoD. In addition, our results reveal activation by calcineurin that is transduced only partially by its conventional downstream effectors, MEF2 and NFAT. Some common features are shared between the promoters of these two genes; however, the mechanisms of their regulation appear distinct.
Collapse
Affiliation(s)
- Doris Heidysch Beylkin
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309-0347, USA
| | | | | |
Collapse
|
23
|
Shmelkov SV, Meeus S, Moussazadeh N, Kermani P, Rashbaum WK, Rabbany SY, Hanson MA, Lane WJ, St Clair R, Walsh KA, Dias S, Jacobson JT, Hempstead BL, Edelberg JM, Rafii S. Cytokine preconditioning promotes codifferentiation of human fetal liver CD133+ stem cells into angiomyogenic tissue. Circulation 2005; 111:1175-83. [PMID: 15753226 DOI: 10.1161/01.cir.0000157155.44008.0f] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND CD133 (AC133) is a surface antigen that defines a broad population of stem cells, including myogenic and endothelial progenitors. CD133+ cells are rare in adult tissues, and the factors that support their differentiation into mature angiomyogenic cells are not known. These hurdles have hampered the use of CD133+ cells for therapeutic purposes. Because human fetal liver is a rich source of CD133+ cells, we sought to identify the growth factors that promote codifferentiation of these cells into angiogenic and myogenic cells. METHODS AND RESULTS Human fetal liver CD133+ and CD133- cell subpopulations were cultured with 5'-azacytidine or vascular endothelial growth factor (VEGF165) and/or brain-derived nerve growth factor (BDNF). CD133+ but not CD133- cells from human fetal liver codifferentiated into spindle-shaped cells, as well as flat adherent multinucleated cells capable of spontaneous contractions in culture. The resulting spindle-shaped cells were confirmed to be endothelial cells by immunohistochemistry analysis for von Willebrand factor and by acetylated LDL uptake. Multinucleated cells were characterized as striated muscles by electron microscopy and immunohistochemistry analysis for myosin heavy chain. Presence of VEGF165 and BDNF significantly enhanced angiomyogenesis in vitro. Inoculation of cells derived from CD133+ cells, but not CD133- cells, into the ear pinna of NOD/SCID mice resulted in the formation of cardiomyocytes, as identified by immunostaining with cardiac troponin-T antibody. These cells generated electrical action potentials, detectable by ECG tracing. CONCLUSIONS CD133 defines a population of human fetal liver cells capable of differentiating into both angiogenic and myogenic cells. Preconditioning of these CD133+ cells with VEGF165 and BDNF enhances the angiomyogenesis. CD133+ fetal liver cells ultimately may be used for therapeutic angiomyogenesis.
Collapse
MESH Headings
- AC133 Antigen
- Action Potentials
- Animals
- Antigens, CD/analysis
- Azacitidine/pharmacology
- Biomarkers
- Brain-Derived Neurotrophic Factor/pharmacology
- Cell Differentiation/drug effects
- Cell Lineage
- Cells, Cultured/cytology
- Cells, Cultured/drug effects
- Ear, External
- Endothelial Cells/chemistry
- Endothelial Cells/cytology
- Endothelium, Vascular/cytology
- Gene Expression Profiling
- Glycoproteins/analysis
- Humans
- Lipoproteins, LDL/metabolism
- Liver/cytology
- Liver/embryology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Muscle Proteins/biosynthesis
- Muscle Proteins/genetics
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/physiology
- Peptides/analysis
- Receptors, Immunologic/metabolism
- Receptors, Scavenger
- Stem Cell Transplantation
- Stem Cells/cytology
- Stem Cells/drug effects
- Transplantation, Heterologous
- Vascular Endothelial Growth Factor A/pharmacology
- von Willebrand Factor/analysis
Collapse
Affiliation(s)
- Sergey V Shmelkov
- Weill Medical College of Cornell University, Division of Hematology-Oncology, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Sheela SG, Lee WC, Lin WW, Chung BC. Zebrafish ftz-f1a (nuclear receptor 5a2) functions in skeletal muscle organization. Dev Biol 2005; 286:377-90. [PMID: 16162335 DOI: 10.1016/j.ydbio.2005.06.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2005] [Revised: 05/03/2005] [Accepted: 06/08/2005] [Indexed: 11/21/2022]
Abstract
Fushi-tarazu factor 1a (Ftz-F1a, Ff1a, Nr5a2) is a nuclear receptor with diverse functions in many tissues. Here, we report the function of ff1a in zebrafish muscle differentiation. In situ hybridization revealed that ff1a mRNA was present in the adaxial and migrating slow muscle precursors and was down-regulated when slow muscle cells matured. This expression was under the control of hedgehog genes, expanded when hedgehog was increased and missing in mutants defective in genes in the Hedgehog pathway like you-too (yot), sonic you (syu), and u-boot (ubo). Blocking ff1a activity by injecting a deleted form of ff1a or an antisense ff1a morpholino oligo into fish embryos caused thinner and disorganized fibers of both slow and fast properties. Transient expression of ff1a in syu, ubo, and yot embryos led to more fibril bundles, even when slow myoblasts were transfated into fast properties. We showed that ff1a and prox1 complemented each other in slow myofibril assembly, but they did not affect the expression of each other. These results demonstrate that ff1a functions in both slow and fast muscle morphogenesis in response to Hedgehog signaling, and this function parallels the activity of another slow muscle gene, prox1.
Collapse
|
25
|
Guo Y, Cheong N, Zhang Z, De Rose R, Deng Y, Farber SA, Fernandes-Alnemri T, Alnemri ES. Tim50, a component of the mitochondrial translocator, regulates mitochondrial integrity and cell death. J Biol Chem 2004; 279:24813-25. [PMID: 15044455 DOI: 10.1074/jbc.m402049200] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In yeast, Tim50 along with Tim23 regulate translocation of presequence-containing proteins across the mitochondrial inner membrane. Here, we describe the identification and characterization of a novel human mitochondrial inner membrane protein homologous to the yeast Tim50. We demonstrate that human Tim50 possesses phosphatase activity and is present in a complex with human Tim23. Down-regulation of human Tim50 expression by RNA interference increases the sensitivity of human cell lines to death stimuli by accelerating the release of cytochrome c from the mitochondria. Furthermore, injection of Tim50-specific morpholino antisense oligonucleotides during early zebrafish embryonic development causes neurodegeneration, dysmorphic hearts, and reduced motility as a result of increased cell death. These observations indicate that loss of Tim50 in vertebrates causes mitochondrial membrane permeabilization and dysfunction followed by cytoplasmic release of cytochrome c along with other mitochondrial inducers of cell death. Thus Tim50 is important for both mitochondrial function and early neuronal development.
Collapse
Affiliation(s)
- Yin Guo
- Center for Apoptosis Research and the Department of Microbiology and Immunology, Kimmel Cancer Institute, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Rice NA, Leinwand LA. Skeletal myosin heavy chain function in cultured lung myofibroblasts. J Cell Biol 2003; 163:119-29. [PMID: 14557251 PMCID: PMC2173446 DOI: 10.1083/jcb.200303194] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2003] [Accepted: 08/26/2003] [Indexed: 11/22/2022] Open
Abstract
Myofibroblasts are unique contractile cells with both muscle and nonmuscle properties. Typically myofibroblasts are identified by the expression of alpha smooth muscle actin (ASMA); however some myofibroblasts also express sarcomeric proteins. In this study, we show that pulmonary myofibroblasts express three of the eight known sarcomeric myosin heavy chains (MyHCs) (IIa, IId, and embryonic) and that skeletal muscle myosin enzymatic activity is required for pulmonary myofibroblast contractility. Furthermore, inhibition of skeletal myosin activity and myofibroblast contraction results in a decrease in both ASMA and skeletal MyHC promoter activity and ASMA protein expression, suggesting a potential coupling of skeletal myosin activity and ASMA expression in myofibroblast differentiation. To understand the molecular mechanisms whereby skeletal muscle genes are regulated in myofibroblasts, we have found that members of the myogenic regulatory factor family of transcription factors and Ca(2+) - regulated pathways are involved in skeletal MyHC promoter activity. Interestingly, the regulation of skeletal myosin expression in myofibroblasts is distinct from that observed in muscle cells and suggests that cell context is important in its control.
Collapse
Affiliation(s)
- Nancy A Rice
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309, USA
| | | |
Collapse
|
27
|
Welikson RE, Fischman DA. The C-terminal IgI domains of myosin-binding proteins C and H (MyBP-C and MyBP-H) are both necessary and sufficient for the intracellular crosslinking of sarcomeric myosin in transfected non-muscle cells. J Cell Sci 2002; 115:3517-26. [PMID: 12154082 DOI: 10.1242/jcs.115.17.3517] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Using the COS cell transfection assay developed previously, we examined which domains of myosin-binding proteins C and H (MyBP-C and MyBP-H) are involved in intracellular interactions with sarcomeric myosin heavy chain(MyHC). Earlier studies demonstrated that overexpression of sarcomeric MyHC in COS cells results in the cytoplasmic assembly of anisotropic, spindle-like aggregates of myosin-containing filaments in the absence of other myofibrillar proteins. When the same sarcomeric MyHC was co-expressed with either MyBP-C or MyBP-H, prominent cable-like co-polymers of MyHC and the MyBPs formed in the cytoplasm instead of the spindle-like aggregates formed by MyHC alone. In vitro binding assays have shown that the C-terminal IgI domain of both MyBP-C(domain C10) and MyBP-H (domain H4) contains the light meromyosin(LMM)-binding sites of each molecule, but this domain cannot explain all of the intracellular properties of the molecules. For example, domains C7-C10 of MyBP-C and domains H1-H4 of MyBP-H are required for the faithful targeting of these proteins to the A-bands of myofibrils in skeletal muscle. Using truncation mutants of both MyBPs tagged with either green fluorescent protein(GFP) or c-myc, we now demonstrate that the last four domains of both MyBP-C and MyBP-H colocalize with the full-length proteins in the MyHC/MyBP cable polymers when co-transfected with MyHC in COS cells. Deletion of the C-terminal IgI domain in either MyBP-C or MyBP-H abrogated cable formation,but the expressed proteins could still colocalize with MyHC-containing filament aggregates. Co-expression of only the C-terminal IgI domain of MyBP-C with sarcomeric MyHC was sufficient for cable formation and colocalization with myosin. We conclude that the C-terminal IgI domains of both MyBP-H and MyBP-C are both necessary and sufficient for inducing MyHC/MyBP cable formation in this COS cell system. However, there must be other myosin-binding sites in MyBP-C and MyBP-H that explain the co-distribution of these proteins with myosin filaments in the absence of cable formation. These latter sites are neither sufficient nor required for cable formation.
Collapse
Affiliation(s)
- Robert E Welikson
- Department of Cell Biology, Weill Medical College of Cornell University, 1300 York Avenue, York Avenue, New York, NY 10021, USA
| | | |
Collapse
|
28
|
Behra M, Cousin X, Bertrand C, Vonesch JL, Biellmann D, Chatonnet A, Strähle U. Acetylcholinesterase is required for neuronal and muscular development in the zebrafish embryo. Nat Neurosci 2002; 5:111-8. [PMID: 11753420 DOI: 10.1038/nn788] [Citation(s) in RCA: 287] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The neurotransmitter acetylcholine (ACh) has a crucial role in central and neuromuscular synapses of the cholinergic system. After release into the synaptic cleft, ACh is rapidly degraded by acetylcholinesterase (AChE). We have identified a mutation in the ache gene of the zebrafish, which abolishes ACh hydrolysis in homozygous animals completely. Embryos are initially motile but subsequently develop paralysis. Mutant embryos show defects in muscle fiber formation and innervation, and primary sensory neurons die prematurely. The neuromuscular phenotype in ache mutants is suppressed by a homozygous loss-of-function allele of the alpha-subunit of the nicotinic acetylcholine receptor (nAChR), indicating that the impairment of neuromuscular development is mediated by activation of nAChR in the mutant. Here we provide genetic evidence for non-classical functions of AChE in vertebrate development.
Collapse
Affiliation(s)
- Martine Behra
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, BP 163, 67404 Illkirch Cedex, C.U. de Strasbourg, France
| | | | | | | | | | | | | |
Collapse
|
29
|
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
|
30
|
Ojima K, Lin Z, Bang ML, Holtzer S, Matsuda R, Labeit S, Sweeney H, Holtzer H. Distinct families of Z-line targeting modules in the COOH-terminal region of nebulin. J Cell Biol 2000; 150:553-66. [PMID: 10931867 PMCID: PMC2175182 DOI: 10.1083/jcb.150.3.553] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
To learn how nebulin functions in the assembly and maintenance of I-Z-I bands, MYC- and GFP- tagged nebulin fragments were expressed in primary cultured skeletal myotubes. Their sites of incorporation were visualized by double staining with anti-MYC, antibodies to myofibrillar proteins, and FITC- or rhodamine phalloidin. Contrary to expectations based on in vitro binding studies, none of the nebulin fragments expressed in maturing myotubes were incorporated selectively into I-band approximately 1.0-micrometer F-alpha-actin-containing thin filaments. Four of the MYC/COOH-terminal nebulin fragments were incorporated exclusively into periodic approximately 0.1-micrometer Z-bands. Whereas both anti-MYC and Rho-phalloidin stained intra-Z-band F-alpha-actin oligomers, only the latter stained the pointed ends of the polarized approximately 1.0-micrometer thin filaments. Z-band incorporation was independent of the nebulin COOH-terminal Ser or SH3 domains. In vitro cosedimentation studies also demonstrated that nebulin SH3 fragments did not bind to F-alpha-actin or alpha-actinin. The remaining six fragments were not incorporated into Z-bands, but were incorporated (a) diffusely throughout the sarcoplasm and into (b) fibrils/patches of varying lengths and widths nested among normal striated myofibrils. Over time, presumably in response to the mediation of muscle-specific homeostatic controls, many of the ectopic MYC-positive structures were resorbed. None of the tagged nebulin fragments behaved as dominant negatives; they neither blocked the assembly nor induced the disassembly of mature striated myofibrils. Moreover, they were not cytotoxic in myotubes, as they were in the fibroblasts and presumptive myoblasts in the same cultures.
Collapse
Affiliation(s)
- K. Ojima
- Department of Cell and Developmental Biology, The School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Z.X. Lin
- Department of Cell Biology, Beijing Institute for Cancer Research, Beijing Medical University, Beijing 100034, China
| | | | - S. Holtzer
- Department of Cell and Developmental Biology, The School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - R. Matsuda
- Department of Life Science, University of Tokyo, Tokyo, Japan 153-8092
| | - S. Labeit
- Department of Anesthesiology and Intensive Operative Care, Klinikum, Mannheim, Germany
| | - H.L. Sweeney
- Department of Physiology, The School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - H. Holtzer
- Department of Cell and Developmental Biology, The School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
31
|
Chimori Y, Hayashi K, Kimura K, Nishida W, Funahashi S, Miyata S, Shimane M, Matsuzawa Y, Sobue K. Phenotype-dependent expression of cadherin 6B in vascular and visceral smooth muscle cells. FEBS Lett 2000; 469:67-71. [PMID: 10708758 DOI: 10.1016/s0014-5793(00)01220-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We used mRNA subtraction of differentiated and dedifferentiated smooth muscle cells (SMCs) to reveal the molecular mechanisms underlying the phenotypic modulation of SMCs. With this approach, we found that a 10 kb mRNA encoding a homotypic cell adhesion molecule, cadherin 6B, was strongly expressed in differentiated vascular and visceral SMCs, but not in the dedifferentiated SMCs derived from them. In vivo, cadherin 6B was expressed in vascular and visceral SMCs, in addition to brain, spinal cord, retina and kidney, at a late stage of chicken embryonic development. These results suggest that cadherin 6B is a novel molecular marker for vascular and visceral SMC phenotypes and is involved in the late differentiation of SMCs.
Collapse
Affiliation(s)
- Y Chimori
- Division of Neurochemistry and Neuropharmacology, Department of Neuroscience, Biomedical Research Center, Osaka University Graduate School of Medicine (C13), 2-2 Yamadaoka, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Miyata S, Minobe W, Bristow MR, Leinwand LA. Myosin heavy chain isoform expression in the failing and nonfailing human heart. Circ Res 2000; 86:386-90. [PMID: 10700442 DOI: 10.1161/01.res.86.4.386] [Citation(s) in RCA: 361] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the heart, the relative proportions of the 2 forms of the motor protein myosin heavy chain (MyHC) have been shown to be affected by a wide variety of pathological and physiological stimuli. Hearts that express the faster MyHC motor protein, alpha, produce more power than those expressing the slower MyHC motor protein, beta, leading to the hypothesis that MyHC isoforms play a major role in the determination of cardiac contractility. We showed previously that a significant amount of alphaMyHC mRNA is expressed in nonfailing human ventricular myocardium and that alphaMyHC mRNA expression is decreased 15-fold in end-stage failing left ventricles. In the present study, we determined the MyHC protein isoform content of human heart samples of known MyHC mRNA composition. We demonstrate that alphaMyHC protein was easily detectable in 12 nonfailing hearts. alphaMyHC protein represented 7.2+/-3.2% of total MyHC protein (compared with approximately 35% of the MyHC mRNA), suggesting that translational regulation may be operative; in contrast, there was effectively no detectable alphaMyHC protein in the left ventricles of 10 end-stage failing human hearts.
Collapse
Affiliation(s)
- S Miyata
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309-0347, USA
| | | | | | | |
Collapse
|
33
|
Rathgeber B, Boles J, Xavier I, McCurdy A, Shand P. Thermally Induced Changes in Protein Extractability of Postrigor Turkey Breast Meat Measured by Two Methods. J Food Sci 1999. [DOI: 10.1111/j.1365-2621.1999.tb12269.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
34
|
Takke C, Campos-Ortega JA. her1, a zebrafish pair-rule like gene, acts downstream of notch signalling to control somite development. Development 1999; 126:3005-14. [PMID: 10357943 DOI: 10.1242/dev.126.13.3005] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During vertebrate embryonic development, the paraxial mesoderm becomes subdivided into metameric units known as somites. In the zebrafish embryo, genes encoding homologues of the proteins of the Drosophila Notch signalling pathway are expressed in the presomitic mesoderm and expression is maintained in a segmental pattern during somitogenesis. This expression pattern suggests a role for these genes during somite development. We misexpressed various zebrafish genes of this group by injecting mRNA into early embryos. RNA encoding a constitutively active form of notch1a (notch1a-intra) and a truncated variant of deltaD [deltaD(Pst)], as well as transcripts of deltaC and deltaD, the hairy-E(spl) homologues her1 and her4, and groucho2 were tested for their effects on somite formation, myogenesis and on the pattern of transcription of putative downstream genes. In embryos injected with any of these RNAs, with the exception of groucho2 RNA, the paraxial mesoderm differentiated normally into somitic tissue, but failed to segment correctly. Activation of notch results in ectopic activation of her1 and her4. This misregulation of the expression of her genes might be causally related to the observed mesodermal defects, as her1 and her4 mRNA injections led to effects similar to those seen with notch1a-intra. deltaC and deltaD seem to function after subdivision of the presomitic mesoderm, since the her gene transcription pattern in the presomitic mesoderm remains essentially normal after misexpression of delta genes. Whereas notch signalling alone apparently does not affect myogenesis, zebrafish groucho2 is involved in differentiation of mesodermal derivatives.
Collapse
Affiliation(s)
- C Takke
- Institut für Entwicklungsbiologie, Universität zu Köln, Germany
| | | |
Collapse
|
35
|
Takke C, Dornseifer P, v Weizsäcker E, Campos-Ortega JA. her4, a zebrafish homologue of the Drosophila neurogenic gene E(spl), is a target of NOTCH signalling. Development 1999; 126:1811-21. [PMID: 10101116 DOI: 10.1242/dev.126.9.1811] [Citation(s) in RCA: 121] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
her4 encodes a zebrafish bHLH protein of the hairy-E(spl) family. The gene is transcribed in a complex pattern in the developing nervous system and in the hypoblast. During early neurogenesis, her4 expression domains include the regions of the neural plate from which primary neurons arise, suggesting that the gene is involved in directing their development. Indeed, misexpression of specific her4 variants leads to a reduction in the number of primary neurons formed. The amino-terminal region of her4, including the basic domain, and the region between the putative helix IV and the carboxy-terminal tetrapeptide wrpw are essential for this effect, since her4 variants lacking either of these regions are non-functional. However, the carboxy-terminal wrpw itself is dispensable. We have examined the interrelationships between deltaD, deltaA, notch1, her4 and neurogenin1 by means of RNA injections. her4 is involved in a regulatory feedback loop which modulates the activity of the proneural gene neurogenin, and as a consequence, of deltaA and deltaD. Activation of notch1 leads to strong activation of her4, to suppression of neurogenin transcription and, ultimately, to a reduction in the number of primary neurons. These results suggest that her4 acts as a target of notch-mediated signals that regulate primary neurogenesis.
Collapse
Affiliation(s)
- C Takke
- Institut für Entwicklungsbiologie, Universität zu Köln, Germany
| | | | | | | |
Collapse
|
36
|
Gilbert R, Cohen JA, Pardo S, Basu A, Fischman DA. Identification of the A-band localization domain of myosin binding proteins C and H (MyBP-C, MyBP-H) in skeletal muscle. J Cell Sci 1999; 112 ( Pt 1):69-79. [PMID: 9841905 DOI: 10.1242/jcs.112.1.69] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although major constituents of the thick filaments of vertebrate striated muscles, the myosin binding proteins (MyBP-C and MyBP-H) are still of uncertain function. Distributed in the cross-bridge bearing zone of the A-bands of myofibrils, in a series of transverse 43 nm stripes, the proteins are constructed of a tandem series of small globular domains, each composed of approximately 90–100 amino acids, which have sequence similarities to either the C2-set of immunoglobulins (IgC2) and the fibronectin type III (FnIII) motifs. MyBP-C is composed of ten globular domains (approximately 130 kDa) whereas MyBP-H is smaller (approximately 58 kDa) and consists of a unique N-terminal segment followed by four globular domains, the order of which is identical to that of MyBP-C (FnIII-IgC2-FnIII-IgC2). To improve our understanding of this protein family we have characterized the domains in each of these two proteins which are required for targeting the proteins to their native site(s) in the sarcomere during myogenesis. Cultures of skeletal muscle myoblasts were transfected with expression plasmids encoding mutant constructs of the MyBPs bearing an N-terminal myc epitope, and their localization to the A-band examined by immunofluorescence microscopy. Based on the clarity and intensity of the myc A-band signals we concluded that constructs encoding the four C-terminal motifs of MyBP-C and MyBP-H (approximately 360 amino acids) were all that was necessary to efficiently localize each of these peptides to the A-band. Truncation mutants lacking one of these 4 domains were less efficiently targeted to the C-zone of the sarcomere. Deletion of the last C-terminal motif of MyBP-H, its myosin binding domain, abolished all localization to the A-band. A chimeric construct, HU-3C10, in which the C-terminal motif of MyBP-H was replaced by the myosin binding domain of MyBP-C, efficiently localized to the A-band. Taken together, these observations indicate that MyBP-C and MyBP-H are localized to the A-band by the same C-terminal domain, composed of two IgC2 and two FnIII motifs. A model has been proposed for the interaction and positioning of the MyBPs in the thick filament through a ternary complex of the four C-terminal motifs with the myosin rods and titin.
Collapse
Affiliation(s)
- R Gilbert
- Department of Cell Biology, Cornell University Medical College, New York, NY 10021, USA.
| | | | | | | | | |
Collapse
|
37
|
Mayer DC, Leinwand LA. Sarcomeric gene expression and contractility in myofibroblasts. J Cell Biol 1997; 139:1477-84. [PMID: 9396753 PMCID: PMC2132619 DOI: 10.1083/jcb.139.6.1477] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/1997] [Revised: 09/25/1997] [Indexed: 02/05/2023] Open
Abstract
Myofibroblasts are unusual cells that share morphological and functional features of muscle and nonmuscle cells. Such cells are thought to control liver blood flow and kidney glomerular filtration rate by having unique contractile properties. To determine how these cells achieve their contractile properties and their resemblance to muscle cells, we have characterized two myofibroblast cell lines. Here, we demonstrate that myofibroblast cell lines from kidney mesangial cells (BHK) and liver stellate cells activate extensive programs of muscle gene expression including a wide variety of muscle structural proteins. In BHK cells, six different striated myosin heavy chain isoforms and many thin filament proteins, including troponin T and tropomyosin are expressed. Liver stellate cells express a limited subset of the muscle thick filament proteins expressed in BHK cells. Although these cells are mitotically active and do not morphologically differentiate into myotubes, we show that MyoD and myogenin are expressed and functional in both cell types. Finally, these cells contract in response to endothelin-1 (ET-1); and we show that ET-1 treatment increases the expression of sarcomeric myosin.
Collapse
Affiliation(s)
- D C Mayer
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309, USA
| | | |
Collapse
|
38
|
Chao TH, Bandman E. Cloning, nucleotide sequence and characterization of a full-length cDNA encoding the myosin heavy chain from adult chicken pectoralis major muscle. Gene X 1997; 199:265-70. [PMID: 9358064 DOI: 10.1016/s0378-1119(97)00386-7] [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: 02/05/2023] Open
Abstract
Four cDNA clones, encoding the chicken adult sarcomeric MyHC, have been isolated from a pectoralis major muscle cDNA library using gene-specific DNA probes. These clones were sequenced and then subcloned into a full-length, 6-kb, chicken adult sarcomeric MyHC cDNA. The entire cDNA consists of 5873 nucleotides with 19 bp 5'-untranslated region and 34 bp 3'-untranslated region. The complete cDNA encodes a 1939-aa polypeptide whose molecular weight is 223 kDa. The calculated isoelectric point of this protein is approximately 5.7. Analysis of the deduced amino acid sequence and comparison with a previously published amino-acid sequence of the same MyHC isoform reveals that six amino acid residues are different. Hydrophilicity analysis of this adult MyHC amino-acid sequence shows a similar pattern as the embryonic MyHC. A recombinant baculovirus, carrying this full-length adult MyHC cDNA, has also been generated and expressed in the Sf9 insect cell line. A approximately 220-kDa recombinant MyHC was synthesized and reacted specifically with chicken adult MyHC monoclonal antibodies.
Collapse
Affiliation(s)
- T H Chao
- Department of Food Science and Technology, University of California, Davis 95616, USA
| | | |
Collapse
|
39
|
Du SJ, Devoto SH, Westerfield M, Moon RT. Positive and negative regulation of muscle cell identity by members of the hedgehog and TGF-beta gene families. J Cell Biol 1997; 139:145-56. [PMID: 9314535 PMCID: PMC2139815 DOI: 10.1083/jcb.139.1.145] [Citation(s) in RCA: 170] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/1997] [Revised: 06/25/1997] [Indexed: 02/05/2023] Open
Abstract
We have examined whether the development of embryonic muscle fiber type is regulated by competing influences between Hedgehog and TGF-beta signals, as previously shown for development of neuronal cell identity in the neural tube. We found that ectopic expression of Hedgehogs or inhibition of protein kinase A in zebrafish embryos induces slow muscle precursors throughout the somite but muscle pioneer cells only in the middle of the somite. Ectopic expression in the notochord of Dorsalin-1, a member of the TGF-beta superfamily, inhibits the formation of muscle pioneer cells, demonstrating that TGF-beta signals can antagonize the induction of muscle pioneer cells by Hedgehog. We propose that a Hedgehog signal first induces the formation of slow muscle precursor cells, and subsequent Hedgehog and TGF-beta signals exert competing positive and negative influences on the development of muscle pioneer cells.
Collapse
Affiliation(s)
- S J Du
- Howard Hughes Medical Institute, University of Washington, School of Medicine, Seattle 98195, USA
| | | | | | | |
Collapse
|
40
|
Abstract
MRF4, myogenin, MyoD, and Myf-5 are the four members of the basic helix-loop-helix family of muscle-specific regulatory factors (MRFs). We examined whether MRF4 could substitute for myogenin in vivo by determining if the myofiber- and MRF4-deficient phenotype of myogenin (-/-) mice could be rescued by a myogenin promoter-MRF4 transgene. When the transgene was expressed at a physiological level in myogenin-deficient fetuses, we found that expression of the endogenous MRF4 gene was restored to normal levels, whereas MyoD levels were unchanged. Thus, MRF4 can participate in a positive autoregulatory loop and can substitute for myogenin to activate its own promoter. Myogenin-deficient fetuses that expressed the transgene also had more myosin, more and larger myofibers, and a more normal ribcage morphology than myogenin-deficient littermates without the transgene. The transgene failed, however, to restore normal numbers of myofibers or viability to myogenin-deficient mice, because the approximately 1.6 kb myogenin promoter fragment was not expressed in most late-forming myofibers. These results demonstrate that MRF4 is able to substitute for myogenin to activate MRF4 expression and promote myofiber formation during the early stages of myogenesis.
Collapse
Affiliation(s)
- Z Zhu
- Neuromuscular Laboratory, Massachusetts General Hospital, Charlestown 02129, USA
| | | |
Collapse
|
41
|
Devoto SH, Melançon E, Eisen JS, Westerfield M. Identification of separate slow and fast muscle precursor cells in vivo, prior to somite formation. Development 1996; 122:3371-80. [PMID: 8951054 DOI: 10.1242/dev.122.11.3371] [Citation(s) in RCA: 438] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have examined the development of specific muscle fiber types in zebrafish axial muscle by labeling myogenic precursor cells with vital fluorescent dyes and following their subsequent differentiation and fate. Two populations of muscle precursors, medial and lateral, can be distinguished in the segmental plate by position, morphology and gene expression. The medial cells, known as adaxial cells, are large, cuboidal cells adjacent to the notochord that express myoD. Surprisingly, after somite formation, they migrate radially away from the notochord, becoming a superficial layer of muscle cells. A subset of adaxial cells develop into engrailed-expressing muscle pioneers. Adaxial cells differentiate into slow muscle fibers of the adult fish. We have named the lateral population of cells in the segmental plate, lateral presomitic cells. They are smaller, more irregularly shaped and separated from the notochord by adaxial cells; they do not express myoD until after somite formation. Lateral presomitic cells remain deep in the myotome and they differentiate into fast muscle fibers. Thus, slow and fast muscle fiber types in zebrafish axial muscle arise from distinct populations of cells in the segmental plate that develop in different cellular environments and display distinct behaviors.
Collapse
Affiliation(s)
- S H Devoto
- Institute of Neuroscience, University of Oregon, Eugene 97403, USA
| | | | | | | |
Collapse
|
42
|
Moncman CL, Wang K. Nebulette: a 107 kD nebulin-like protein in cardiac muscle. CELL MOTILITY AND THE CYTOSKELETON 1995; 32:205-25. [PMID: 8581976 DOI: 10.1002/cm.970320305] [Citation(s) in RCA: 136] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A 107-kD protein has been identified in primary cultures of chicken embryonic cardiomyocytes by immunoprecipitations with certain anti-nebulin monoclonal antibodies (mAbs). These mAbs, prepared against a fragment of human skeletal muscle nebulin located near the carboxyl terminus, detect a 107-kD protein in extracts of adult chicken heart, adult mouse heart, and adult rabbit heart by immunoblot analysis. A partial cDNA corresponding to this protein has been isolated by immunological screening of a chicken heart cDNA expression vector library. The partial cDNA encodes a 380-amino acid open reading frame composed entirely of nebulin-like 35-residue modules marked by the highly conserved sequence motifs: SXXXYK and TPD. The open reading frame exhibits 60-85% homology with skeletal muscle nebulins from a variety of species. This cDNA recognizes an approximately 8-kb transcript in cardiac RNA and does not hybridize to skeletal muscle RNAs by northern analysis. Immunofluorescence localization of this nebulin-like protein in primary cultures of chicken cardiomyocytes and embryonic chicken cardiac myofibrils indicates that the protein is localized to the I-Z-I complex of the myofibrils, extending approximately 25% of the thin filament length. Comparisons of the distribution of this protein relative to actin, myosin, and titin in spreading cardiomyocytes suggest that the cardiac nebulin-like protein becomes aligned with the nascent myofibrils early during myofibrillogenesis. To distinguish this petite nebulin-like protein from the 600-900 kD skeletal muscle nebulin, we have named it nebulette.
Collapse
Affiliation(s)
- C L Moncman
- Department of Chemistry and Biochemistry, University of Texas, Austin 78712, USA
| | | |
Collapse
|
43
|
Winkelmann DA, Kinose F, Chung AL. Inhibition of actin filament movement by monoclonal antibodies against the motor domain of myosin. J Muscle Res Cell Motil 1993; 14:452-67. [PMID: 7693748 DOI: 10.1007/bf00121297] [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: 01/26/2023]
Abstract
Conformational transitions in defined regions of the motor domain of skeletal muscle myosin involved in ATP hydrolysis, actin binding and motility were probed with monoclonal antibodies. Competition binding assays demonstrate that three different monoclonal antibodies react with spatially related sites on the myosin heavy chain. One recognizes a sequential epitope between residues 65 and 80 and has no effect on actin filament movement in an in vitro motility assay despite tight binding to myosin and acto-S1. The other two monoclonal antibodies react with sequential epitopes between residues 29 and 60 and both inhibit actin filament movement. A fourth monoclonal antibody reacts with the N-terminus of the heavy chain (residues 1-12) at a spatially distinct site on the myosin head and also inhibits actin filament movement. These four monoclonal antibodies have been mapped by immunoelectron microscopy to the large, actin binding region of the myosin head; however, the antibody binding sites remain accessible on rigor complexes of acto-S1. Thus, this group of monoclonal antibodies identify sequential epitopes in a mobile segment of the myosin heavy chain. In addition, two conformation-sensitive monoclonal antibodies are described that are affected by ATP and actin binding to myosin S1, and display distinct and marked inhibitory effects on actin filament movement. In contrast, an anti-light chain monoclonal antibody that binds near the myosin head-rod junction has little effect on the number and velocity of moving actin filaments. These results identify mobile regions on the myosin head that are perturbed by antibody binding and that may be linked to force production and motion.
Collapse
Affiliation(s)
- D A Winkelmann
- Department of Pathology, Robert Wood Johnson Medical School, Piscataway, NJ 08854
| | | | | |
Collapse
|
44
|
Smith TH, Block NE, Rhodes SJ, Konieczny SF, Miller JB. A unique pattern of expression of the four muscle regulatory factor proteins distinguishes somitic from embryonic, fetal and newborn mouse myogenic cells. Development 1993; 117:1125-33. [PMID: 8391976 DOI: 10.1242/dev.117.3.1125] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A unique pattern of expression of the four muscle regulatory factor (MRF) proteins was found to distinguish early somitic from embryonic, fetal and newborn limb myogenic cells in vitro. Expression of the myosin heavy chain (MHC), MyoD, myogenin, Myf-5, and MRF4 proteins was examined by immunocytochemistry in cultures of four distinct types of mouse myogenic cells: somitic (E8.5), embryonic (E11.5), fetal (E16.5) and newborn limb. In embryonic, fetal and newborn cultures, the MRF proteins were expressed in generally similar patterns: MyoD was the first MRF expressed; MyoD and myogenin were expressed by more cells than Myf-5 or MRF4; and each of the four MRFs was found both in cells that expressed MHC and in cells that did not express MHC. In cultures of somitic cells, in contrast, Myf-5 was expressed first and by more cells than MyoD or myogenin; MRF4 was not detected; and the MRFs were never found to be coexpressed with MHC in the same cell. Thus, some somitic cells had the unexpected ability to maintain MHC expression in the absence of detectable MRF protein expression. The different myogenic programs of embryonic, fetal and newborn myogenic cells are not, therefore, a simple result of qualitatively different MRF expression patterns, whereas myogenesis by somitic cells does include a unique pattern of MRF expression.
Collapse
Affiliation(s)
- T H Smith
- Neuromuscular Laboratory, Massachusetts General Hospital, Charlestown 02129
| | | | | | | | | |
Collapse
|
45
|
Rindt H, Bauer BJ, Robbins J. In vitro production of enzymatically active myosin heavy chain. J Muscle Res Cell Motil 1993; 14:26-34. [PMID: 8478425 DOI: 10.1007/bf00132177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In order to initiate studies on the structural and functional relationships of the myosin heavy chain, we constructed a full-length complementary DNA encoding the isoform that is found in the fast white muscle of the embryonic chicken. The complementary DNA contained 108 basepairs of its 3'-untranslated region and was preceded by a leader sequence derived from the alfalfa mosaic virus. Similarly, a complementary DNA encoding 963 amino acids which encompass the subfragment-1 of myosin and part of the subfragment-2 was also constructed. Each was inserted into the expression vector pMT2 and transiently transfected into COS-1 cells. Both constructs directed the expression of the respective proteins, each of which was immunogenic. The full-length and subfragment-1 proteins interacted with actin and demonstrated high levels of a K(+)-activated, EDTA-resistant ATPase activity, which is characteristic of myosin.
Collapse
Affiliation(s)
- H Rindt
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, OH 45267
| | | | | |
Collapse
|
46
|
Vikstrom KL, Rovner AS, Saez CG, Bravo-Zehnder M, Straceski AJ, Leinwand LA. Sarcomeric myosin heavy chain expressed in nonmuscle cells forms thick filaments in the presence of substoichiometric amounts of light chains. CELL MOTILITY AND THE CYTOSKELETON 1993; 26:192-204. [PMID: 8293476 DOI: 10.1002/cm.970260303] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Central to the function of myosin is its ability to assemble into thick filaments which interact precisely and specifically with other myofibrillar proteins. We have established a novel experimental system for studying myofibrillogenesis using transient transfections of COS cells, a monkey kidney cell line. We have expressed both full-length rat alpha cardiac myosin heavy chain (MHC) and a truncated heavy meromyosin-like alpha MHC (sHMM) and shown that immunoreactive MHC proteins of the expected sizes were detected in lysates of transfected cells. Surprisingly, the full-length MHC formed large spindle-shaped structures throughout the cytoplasm of transfected cells as determined by immunofluorescence microscopy. The structures were not found in cells expressing the sHMM construct, indicating that their formation required an MHC rod. The spindle-shaped structures ranged in length from approximately 1 micron to over 20 microns in length and were birefringent suggesting that they are ordered arrays of thick filaments. This was confirmed by electron microscopic analysis of the transfected cells which revealed arrays of filamentous structures approximately 12 nm in diameter at their widest point. In addition, the vast majority of transfected MHC did not associate with the endogenous nonmuscle myosin light chains, demonstrating that myosin thick filaments can form in the absence of stoichiometric amounts of myosin light chains.
Collapse
Affiliation(s)
- K L Vikstrom
- Albert Einstein College of Medicine, Bronx, NY 10461-1975
| | | | | | | | | | | |
Collapse
|
47
|
Moore LA, Tidyman WE, Arrizubieta MJ, Bandman E. The evolutionary relationship of avian and mammalian myosin heavy-chain genes. J Mol Evol 1993; 36:21-30. [PMID: 8433376 DOI: 10.1007/bf02407303] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Sequence comparisons of avian and mammalian skeletal and cardiac myosin heavy-chain isoforms are used to examine the evolutionary relationships of sarcomeric myosin multigene families. Mammalian fast-myosin heavy-chain isoforms from different species, with comparable developmental expression, are more similar to each other than they are to other fast isoforms within the same genome. In contrast, the developmentally regulated chicken fast isoforms are more similar to each other than they are to myosin heavy-chain isoforms in other species. Extensive regions of nucleotide identity among the chicken fast myosin heavy chains and in the mouse and rat alpha- and beta-cardiac myosin heavy-chain sequences suggest that gene-conversion-like mechanisms have played a major role in the concerted evolution of these gene families. We also conclude that the chicken fast myosin heavy-chain multigene family has undergone recent expansion subsequent to the divergence of birds and mammals and that both the developmental regulation and the specialization of myosin isoforms have likely developed independently in birds and mammals.
Collapse
Affiliation(s)
- L A Moore
- Department of Food Science and Technology, University of California, Davis 95616
| | | | | | | |
Collapse
|
48
|
Ogata I, Sáez CG, Greenwel P, Ponce MDL, Geerts A, Leinwand LA, Rojkind M. Rat liver fat-storing cell lines express sarcomeric myosin heavy chain mRNA and protein. CELL MOTILITY AND THE CYTOSKELETON 1993; 26:125-32. [PMID: 8287498 DOI: 10.1002/cm.970260204] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Fat-storing cells (FSC, lipocytes, or Ito cells) of liver store vitamin A and are the main producers of extracellular matrix in normal and cirrhotic liver. During liver injury, FSC undergo an activation process characterized by a decrease in vitamin A storage and an increase in cell proliferation and extracellular matrix deposition. This activation process also occurs upon culturing FSC from normal liver. In contrast to most cells of nonmuscle origin, activated FSC express two cytoskeletal proteins normally found in muscle, desmin, and smooth muscle alpha-actin. Based on their strategic perisinusoidal location, it has been hypothesized that FSC play a role in regulating blood flow. However, the nature of the contractile elements involved in this process remains to be determined. In this communication we demonstrate the presence of a sarcomeric myosin in proteins solubilized from liver biomatrix. In addition we demonstrate the expression of sarcomeric myosin heavy chain (MHC) mRNA and protein in two FSC clones derived from a CCl4-cirrhotic rat liver (CFSC). Through cloning the cDNA corresponding to the MHC gene expressed in these cells we demonstrate that it encodes fast IId skeletal MHC and thus represents a marker normally seen in adult muscle. The unexpected expression of an adult stage skeletal muscle molecular motor in FSC from cirrhotic liver is consistent with the proposed specialized contractile capacity of these cells.
Collapse
Affiliation(s)
- I Ogata
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York 10461
| | | | | | | | | | | | | |
Collapse
|
49
|
Moore LA, Arrizubieta MJ, Tidyman WE, Herman LA, Bandman E. Analysis of the chicken fast myosin heavy chain family. Localization of isoform-specific antibody epitopes and regions of divergence. J Mol Biol 1992; 225:1143-51. [PMID: 1377278 DOI: 10.1016/0022-2836(92)90114-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
cDNAs encoding the rod region of four different fast myosin heavy chains (MYCHs) in the chicken were identified, using anti-MYCH monoclonal antibodies, in two expression libraries prepared from 19-day embryonic and adult chicken muscle. These clones were used to determine the amino acid sequences that encompass the epitopes of five anti-MYHC monoclonal antibodies. Additionally, the amino acid sequences were compared to each other and to a full length embryonic MYHC. Although there is extensive homology in the chicken fast myosin rods, sequences within the hinge, within the central portion of the light meromyosin fragment, and at the carboxy terminus exhibit the largest number of amino acid substitutions. We propose that divergence within these subdomains may contribute to isoform-specific properties associated with skeletal myosin rods.
Collapse
Affiliation(s)
- L A Moore
- Department of Food Science and Technology, University of California, Davis 95616
| | | | | | | | | |
Collapse
|
50
|
Stromer MH. Immunocytochemical localization of proteins in striated muscle. INTERNATIONAL REVIEW OF CYTOLOGY 1992; 142:61-144. [PMID: 1487396 DOI: 10.1016/s0074-7696(08)62075-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- M H Stromer
- Department of Animal Science, Iowa State University, Ames 50011
| |
Collapse
|