1
|
Trujillo EM, Lee SR, Aguayo A, Torosian TC, Cripps RM. Enhanced expression of the myogenic factor Myocyte enhancer factor-2 in imaginal disc myoblasts activates a partial, but incomplete, muscle development program. Dev Biol 2024; 516:82-95. [PMID: 39111615 DOI: 10.1016/j.ydbio.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/19/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024]
Abstract
The Myocyte enhancer factor-2 (MEF2) transcription factor plays a vital role in orchestrating muscle differentiation. While MEF2 cannot effectively induce myogenesis in naïve cells, it can potently accelerate myogenesis in mesodermal cells. This includes in Drosophila melanogaster imaginal disc myoblasts, where triggering premature muscle gene expression in these adult muscle progenitors has become a paradigm for understanding the regulation of the myogenic program. Here, we investigated the global consequences of MEF2 overexpression in the imaginal wing disc myoblasts, by combining RNA-sequencing with RT-qPCR and immunofluorescence. We observed the formation of sarcomere-like structures that contained both muscle and cytoplasmic myosin, and significant upregulation of muscle gene expression, especially genes essential for myofibril formation and function. These transcripts were functional since numerous myofibrillar proteins were detected in discs using immunofluorescence. Interestingly, muscle genes whose expression is restricted to the adult stages were not activated in these adult myoblasts. These studies confirm a broad activation of the myogenic program in response to MEF2 expression and suggest that additional regulatory factors are required for promoting the adult muscle-specific program. Our findings contribute to understanding the regulatory mechanisms governing muscle development and highlight the multifaceted role of MEF2 in orchestrating this intricate process.
Collapse
Affiliation(s)
| | - Samuel R Lee
- Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - Antonio Aguayo
- Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - Tylee C Torosian
- Department of Biology, San Diego State University, San Diego, CA, 92182, USA
| | - Richard M Cripps
- Department of Biology, San Diego State University, San Diego, CA, 92182, USA.
| |
Collapse
|
2
|
Michael AH, Hana TA, Mousa VG, Ormerod KG. Muscle-fiber specific genetic manipulation of Drosophila sallimus severely impacts neuromuscular development, morphology, and physiology. Front Physiol 2024; 15:1429317. [PMID: 39351283 PMCID: PMC11439786 DOI: 10.3389/fphys.2024.1429317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
The ability of skeletal muscles to contract is derived from the unique genes and proteins expressed within muscles, most notably myofilaments and elastic proteins. Here we investigated the role of the sallimus (sls) gene, which encodes a structural homologue of titin, in regulating development, structure, and function of Drosophila melanogaster. Knockdown of sls using RNA interference (RNAi) in all body-wall muscle fibers resulted in embryonic lethality. A screen for muscle-specific drivers revealed a Gal4 line that expresses in a single larval body wall muscle in each abdominal hemisegment. Disrupting sls expression in single muscle fibers did not impact egg or larval viability nor gross larval morphology but did significantly alter the morphology of individual muscle fibers. Ultrastructural analysis of individual muscles revealed significant changes in organization. Surprisingly, muscle-cell specific disruption of sls also severely impacted neuromuscular junction (NMJ) formation. The extent of motor-neuron (MN) innervation along disrupted muscles was significantly reduced along with the number of glutamatergic boutons, in MN-Is and MN-Ib. Electrophysiological recordings revealed a 40% reduction in excitatory junctional potentials correlating with the extent of motor neuron loss. Analysis of active zone (AZ) composition revealed changes in presynaptic scaffolding protein (brp) abundance, but no changes in postsynaptic glutamate receptors. Ultrastructural changes in muscle and NMJ development at these single muscle fibers were sufficient to lead to observable changes in neuromuscular transduction and ultimately, locomotory behavior. Collectively, the data demonstrate that sls mediates critical aspects of muscle and NMJ development and function, illuminating greater roles for sls/titin.
Collapse
Affiliation(s)
| | | | | | - Kiel G. Ormerod
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, United States
| |
Collapse
|
3
|
Nikonova E, DeCata J, Canela M, Barz C, Esser A, Bouterwek J, Roy A, Gensler H, Heß M, Straub T, Forne I, Spletter ML. Bruno 1/CELF regulates splicing and cytoskeleton dynamics to ensure correct sarcomere assembly in Drosophila flight muscles. PLoS Biol 2024; 22:e3002575. [PMID: 38683844 PMCID: PMC11081514 DOI: 10.1371/journal.pbio.3002575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 05/09/2024] [Accepted: 03/04/2024] [Indexed: 05/02/2024] Open
Abstract
Muscles undergo developmental transitions in gene expression and alternative splicing that are necessary to refine sarcomere structure and contractility. CUG-BP and ETR-3-like (CELF) family RNA-binding proteins are important regulators of RNA processing during myogenesis that are misregulated in diseases such as Myotonic Dystrophy Type I (DM1). Here, we report a conserved function for Bruno 1 (Bru1, Arrest), a CELF1/2 family homolog in Drosophila, during early muscle myogenesis. Loss of Bru1 in flight muscles results in disorganization of the actin cytoskeleton leading to aberrant myofiber compaction and defects in pre-myofibril formation. Temporally restricted rescue and RNAi knockdown demonstrate that early cytoskeletal defects interfere with subsequent steps in sarcomere growth and maturation. Early defects are distinct from a later requirement for bru1 to regulate sarcomere assembly dynamics during myofiber maturation. We identify an imbalance in growth in sarcomere length and width during later stages of development as the mechanism driving abnormal radial growth, myofibril fusion, and the formation of hollow myofibrils in bru1 mutant muscle. Molecularly, we characterize a genome-wide transition from immature to mature sarcomere gene isoform expression in flight muscle development that is blocked in bru1 mutants. We further demonstrate that temporally restricted Bru1 rescue can partially alleviate hypercontraction in late pupal and adult stages, but it cannot restore myofiber function or correct structural deficits. Our results reveal the conserved nature of CELF function in regulating cytoskeletal dynamics in muscle development and demonstrate that defective RNA processing due to misexpression of CELF proteins causes wide-reaching structural defects and progressive malfunction of affected muscles that cannot be rescued by late-stage gene replacement.
Collapse
Affiliation(s)
- Elena Nikonova
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Jenna DeCata
- School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, Missouri, United States of America
| | - Marc Canela
- Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Christiane Barz
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, München, Germany
| | - Alexandra Esser
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Jessica Bouterwek
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Akanksha Roy
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Heidemarie Gensler
- Department of Systematic Zoology, Biocenter, Faculty of Biology, Ludwig-Maximilians-Universität München, München, Germany
| | - Martin Heß
- Department of Systematic Zoology, Biocenter, Faculty of Biology, Ludwig-Maximilians-Universität München, München, Germany
| | - Tobias Straub
- Biomedical Center, Bioinformatics Core Unit, Ludwig-Maximilians-Universität München, München, Germany
| | - Ignasi Forne
- Biomedical Center, Protein Analysis Unit, Ludwig-Maximilians-Universität München, München, Germany
| | - Maria L. Spletter
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
- School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, Missouri, United States of America
| |
Collapse
|
4
|
Fujita N, Girada S, Vogler G, Bodmer R, Kiger AA. PI(4,5)P 2 role in Transverse-tubule membrane formation and muscle function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578124. [PMID: 38352484 PMCID: PMC10862868 DOI: 10.1101/2024.01.31.578124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Transverse (T)-tubules - vast, tubulated domains of the muscle plasma membrane - are critical to maintain healthy skeletal and heart contractions. How the intricate T-tubule membranes are formed is not well understood, with challenges to systematically interrogate in muscle. We established the use of intact Drosophila larval body wall muscles as an ideal system to discover mechanisms that sculpt and maintain the T-tubule membrane network. A muscle-targeted genetic screen identified specific phosphoinositide lipid regulators necessary for T-tubule organization and muscle function. We show that a PI4KIIIα - Skittles/PIP5K pathway is needed for T-tubule localized PI(4)P to PI(4,5)P 2 synthesis, T-tubule organization, calcium regulation, and muscle and heart rate functions. Muscles deficient for PI4KIIIα or Amphiphysin , the homolog of human BIN1 , similarly exhibited specific loss of transversal T-tubule membranes and dyad junctions, yet retained longitudinal membranes and the associated dyads. Our results highlight the power of live muscle studies, uncovering distinct mechanisms and functions for sub-compartments of the T-tubule network relevant to human myopathy. Summary T-tubules - vast, tubulated domains of the muscle plasma membrane - are critical to maintain skeletal and heart contractions. Fujita et al . establish genetic screens and assays in intact Drosophila muscles that uncover PI(4,5)P 2 regulation critical for T-tubule maintenance and function. Key Findings PI4KIIIα is required for muscle T-tubule formation and larval mobility. A PI4KIIIα-Sktl pathway promotes PI(4)P and PI(4,5)P 2 function at T-tubules. PI4KIIIα is necessary for calcium dynamics and transversal but not longitudinal dyads. Disruption of PI(4,5)P 2 function in fly heart leads to fragmented T-tubules and abnormal heart rate.
Collapse
|
5
|
González Morales N, Marescal O, Szikora S, Katzemich A, Correia-Mesquita T, Bíró P, Erdelyi M, Mihály J, Schöck F. The oxoglutarate dehydrogenase complex is involved in myofibril growth and Z-disc assembly in Drosophila. J Cell Sci 2023; 136:jcs260717. [PMID: 37272588 PMCID: PMC10323237 DOI: 10.1242/jcs.260717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 05/24/2023] [Indexed: 06/06/2023] Open
Abstract
Myofibrils are long intracellular cables specific to muscles, composed mainly of actin and myosin filaments. The actin and myosin filaments are organized into repeated units called sarcomeres, which form the myofibrils. Muscle contraction is achieved by the simultaneous shortening of sarcomeres, which requires all sarcomeres to be the same size. Muscles have a variety of ways to ensure sarcomere homogeneity. We have previously shown that the controlled oligomerization of Zasp proteins sets the diameter of the myofibril. Here, we looked for Zasp-binding proteins at the Z-disc to identify additional proteins coordinating myofibril growth and assembly. We found that the E1 subunit of the oxoglutarate dehydrogenase complex localizes to both the Z-disc and the mitochondria, and is recruited to the Z-disc by Zasp52. The three subunits of the oxoglutarate dehydrogenase complex are required for myofibril formation. Using super-resolution microscopy, we revealed the overall organization of the complex at the Z-disc. Metabolomics identified an amino acid imbalance affecting protein synthesis as a possible cause of myofibril defects, which is supported by OGDH-dependent localization of ribosomes at the Z-disc.
Collapse
Affiliation(s)
- Nicanor González Morales
- Department of Biology, McGill University, Quebec H3A 1B1, Canada
- Department of Biology, Dalhousie University, Nova Scotia B3H 4R2, Canada
| | - Océane Marescal
- Department of Biology, McGill University, Quebec H3A 1B1, Canada
| | - Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged 6726, Hungary
| | - Anja Katzemich
- Department of Biology, McGill University, Quebec H3A 1B1, Canada
| | | | - Péter Bíró
- Department of Optics and Quantum Electronics, University of Szeged, Szeged 6720, Hungary
| | - Miklos Erdelyi
- Department of Optics and Quantum Electronics, University of Szeged, Szeged 6720, Hungary
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged 6726, Hungary
- Department of Genetics, University of Szeged, Szeged 6726, Hungary
| | - Frieder Schöck
- Department of Biology, McGill University, Quebec H3A 1B1, Canada
| |
Collapse
|
6
|
Loreau V, Rees R, Chan EH, Taxer W, Gregor K, Mußil B, Pitaval C, Luis NM, Mangeol P, Schnorrer F, Görlich D. A nanobody toolbox to investigate localisation and dynamics of Drosophila titins and other key sarcomeric proteins. eLife 2023; 12:79343. [PMID: 36645120 PMCID: PMC9886281 DOI: 10.7554/elife.79343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 12/16/2022] [Indexed: 01/17/2023] Open
Abstract
Measuring the positions and dynamics of proteins in intact tissues or whole animals is key to understanding protein function. However, to date, this is challenging, as the accessibility of large antibodies to dense tissues is often limited, and fluorescent proteins inserted close to a domain of interest may affect protein function. These complications apply in particular to muscle sarcomeres, arguably one of the most protein-dense assemblies in nature, which complicates studying sarcomere morphogenesis at molecular resolution. Here, we introduce a toolbox of nanobodies recognising various domains of the two Drosophila titin homologs, Sallimus and Projectin, as well as the key sarcomeric proteins Obscurin, α-Actinin, and Zasp52. We verified the superior labelling qualities of our nanobodies in muscle tissue as compared to antibodies. By applying our toolbox to larval muscles, we found a gigantic Sallimus isoform stretching more than 2 µm to bridge the sarcomeric I-band, while Projectin covers almost the entire myosin filaments in a polar orientation. Transgenic expression of tagged nanobodies confirmed their high affinity-binding without affecting target protein function. Finally, adding a degradation signal to anti-Sallimus nanobodies suggested that it is difficult to fully degrade Sallimus in mature sarcomeres; however, expression of these nanobodies caused developmental lethality. These results may inspire the generation of similar toolboxes for other large protein complexes in Drosophila or mammals.
Collapse
Affiliation(s)
- Vincent Loreau
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Renate Rees
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Eunice HoYee Chan
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Waltraud Taxer
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Kathrin Gregor
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Bianka Mußil
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Christophe Pitaval
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Nuno Miguel Luis
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Pierre Mangeol
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Frank Schnorrer
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Dirk Görlich
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| |
Collapse
|
7
|
Schueder F, Mangeol P, Chan EH, Rees R, Schünemann J, Jungmann R, Görlich D, Schnorrer F. Nanobodies combined with DNA-PAINT super-resolution reveal a staggered titin nanoarchitecture in flight muscles. eLife 2023; 12:e79344. [PMID: 36645127 PMCID: PMC9886278 DOI: 10.7554/elife.79344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/22/2022] [Indexed: 01/17/2023] Open
Abstract
Sarcomeres are the force-producing units of all striated muscles. Their nanoarchitecture critically depends on the large titin protein, which in vertebrates spans from the sarcomeric Z-disc to the M-band and hence links actin and myosin filaments stably together. This ensures sarcomeric integrity and determines the length of vertebrate sarcomeres. However, the instructive role of titins for sarcomeric architecture outside of vertebrates is not as well understood. Here, we used a series of nanobodies, the Drosophila titin nanobody toolbox, recognising specific domains of the two Drosophila titin homologs Sallimus and Projectin to determine their precise location in intact flight muscles. By combining nanobodies with DNA-PAINT super-resolution microscopy, we found that, similar to vertebrate titin, Sallimus bridges across the flight muscle I-band, whereas Projectin is located at the beginning of the A-band. Interestingly, the ends of both proteins overlap at the I-band/A-band border, revealing a staggered organisation of the two Drosophila titin homologs. This architecture may help to stably anchor Sallimus at the myosin filament and hence ensure efficient force transduction during flight.
Collapse
Affiliation(s)
- Florian Schueder
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian UniversityMunichGermany
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Pierre Mangeol
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| | - Eunice HoYee Chan
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| | - Renate Rees
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | | | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian UniversityMunichGermany
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Dirk Görlich
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| |
Collapse
|
8
|
Wishard R, Jayaram M, Ramesh SR, Nongthomba U. Spatial and temporal requirement of Mlp60A isoforms during muscle development and function in Drosophila melanogaster. Exp Cell Res 2023; 422:113430. [PMID: 36423661 DOI: 10.1016/j.yexcr.2022.113430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022]
Abstract
Many myofibrillar proteins undergo isoform switching in a spatio-temporal manner during muscle development. The biological significance of the variants of several of these myofibrillar proteins remains elusive. One such myofibrillar protein, the Muscle LIM Protein (MLP), is a vital component of the Z-discs. In this paper, we show that one of the Drosophila MLP encoding genes, Mlp60A, gives rise to two isoforms: a short (279 bp, 10 kDa) and a long (1461 bp, 54 kDa) one. The short isoform is expressed throughout development, but the long isoform is adult-specific, being the dominant of the two isoforms in the indirect flight muscles (IFMs). A concomitant, muscle-specific knockdown of both isoforms leads to partial developmental lethality, with most of the surviving flies being flight defective. A global loss of both isoforms in a Mlp60A-null background also leads to developmental lethality, with muscle defects in the individuals that survive to the third instar larval stage. This lethality could be rescued partially by a muscle-specific overexpression of the short isoform. Genetic perturbation of only the long isoform, through a P-element insertion in the long isoform-specific coding sequence, leads to defective flight, in around 90% of the flies. This phenotype was completely rescued when the P-element insertion was precisely excised from the locus. Hence, our data show that the two Mlp60A isoforms are functionally specialized: the short isoform being essential for normal embryonic muscle development and the long isoform being necessary for normal adult flight muscle function.
Collapse
Affiliation(s)
- Rohan Wishard
- Department of Molecular Reproduction, Development and Genetics; Indian Institute of Science, Bengaluru, 560012, India.
| | - Mohan Jayaram
- Department of Molecular Reproduction, Development and Genetics; Indian Institute of Science, Bengaluru, 560012, India; Department of Studies in Zoology, University of Mysore, Manasgangotri, Mysuru, 570006, India
| | - Saraf R Ramesh
- Department of Studies in Zoology, University of Mysore, Manasgangotri, Mysuru, 570006, India; Department of Life Sciences, Pooja Bhagvat Memorial Mahajana Education Center, K. R. S. Road, Mysuru, 570016, India
| | - Upendra Nongthomba
- Department of Molecular Reproduction, Development and Genetics; Indian Institute of Science, Bengaluru, 560012, India.
| |
Collapse
|
9
|
Schöck F, González-Morales N. The insect perspective on Z-disc structure and biology. J Cell Sci 2022; 135:277280. [PMID: 36226637 DOI: 10.1242/jcs.260179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myofibrils are the intracellular structures formed by actin and myosin filaments. They are paracrystalline contractile cables with unusually well-defined dimensions. The sliding of actin past myosin filaments powers contractions, and the entire system is held in place by a structure called the Z-disc, which anchors the actin filaments. Myosin filaments, in turn, are anchored to another structure called the M-line. Most of the complex architecture of myofibrils can be reduced to studying the Z-disc, and recently, important advances regarding the arrangement and function of Z-discs in insects have been published. On a very small scale, we have detailed protein structure information. At the medium scale, we have cryo-electron microscopy maps, super-resolution microscopy and protein-protein interaction networks, while at the functional scale, phenotypic data are available from precise genetic manipulations. All these data aim to answer how the Z-disc works and how it is assembled. Here, we summarize recent data from insects and explore how it fits into our view of the Z-disc, myofibrils and, ultimately, muscles.
Collapse
Affiliation(s)
- Frieder Schöck
- Department of Biology, McGill University, Montreal, Quebec, H3A 1B1, Canada
| | | |
Collapse
|
10
|
Mechanobiology of muscle and myofibril morphogenesis. Cells Dev 2021; 168:203760. [PMID: 34863916 DOI: 10.1016/j.cdev.2021.203760] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023]
Abstract
Muscles generate forces for animal locomotion. The contractile apparatus of muscles is the sarcomere, a highly regular array of large actin and myosin filaments linked by gigantic titin springs. During muscle development many sarcomeres assemble in series into long periodic myofibrils that mechanically connect the attached skeleton elements. Thus, ATP-driven myosin forces can power movement of the skeleton. Here we review muscle and myofibril morphogenesis, with a particular focus on their mechanobiology. We describe recent progress on the molecular structure of sarcomeres and their mechanical connections to the skeleton. We discuss current models predicting how tension coordinates the assembly of key sarcomeric components to periodic myofibrils that then further mature during development. This requires transcriptional feedback mechanisms that may help to coordinate myofibril assembly and maturation states with the transcriptional program. To fuel the varying energy demands of muscles we also discuss the close mechanical interactions of myofibrils with mitochondria and nuclei to optimally support powerful or enduring muscle fibers.
Collapse
|
11
|
Szikora S, Görög P, Kozma C, Mihály J. Drosophila Models Rediscovered with Super-Resolution Microscopy. Cells 2021; 10:1924. [PMID: 34440693 PMCID: PMC8391832 DOI: 10.3390/cells10081924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 11/25/2022] Open
Abstract
With the advent of super-resolution microscopy, we gained a powerful toolbox to bridge the gap between the cellular- and molecular-level analysis of living organisms. Although nanoscopy is broadly applicable, classical model organisms, such as fruit flies, worms and mice, remained the leading subjects because combining the strength of sophisticated genetics, biochemistry and electrophysiology with the unparalleled resolution provided by super-resolution imaging appears as one of the most efficient approaches to understanding the basic cell biological questions and the molecular complexity of life. Here, we summarize the major nanoscopic techniques and illustrate how these approaches were used in Drosophila model systems to revisit a series of well-known cell biological phenomena. These investigations clearly demonstrate that instead of simply achieving an improvement in image quality, nanoscopy goes far beyond with its immense potential to discover novel structural and mechanistic aspects. With the examples of synaptic active zones, centrosomes and sarcomeres, we will explain the instrumental role of super-resolution imaging pioneered in Drosophila in understanding fundamental subcellular constituents.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary;
| | - Péter Görög
- Institute of Genetics, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary;
- Doctoral School of Multidisciplinary Medical Science, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary
| | - Csaba Kozma
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, Pálfy u. 52/d, H-6725 Szeged, Hungary;
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary;
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary
| |
Collapse
|
12
|
Chae K, Valentin C, Dawson C, Jakes E, Myles KM, Adelman ZN. A knockout screen of genes expressed specifically in Ae. aegypti pupae reveals a critical role for stretchin in mosquito flight. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 132:103565. [PMID: 33716097 DOI: 10.1016/j.ibmb.2021.103565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 06/12/2023]
Abstract
Aedes aegypti is a critical vector for transmitting Zika, dengue, chikungunya, and yellow fever viruses to humans. Genetic strategies to limit mosquito survival based upon sex distortion or disruption of development may be valuable new tools to control Ae. aegypti populations. We identified six genes with expression limited to pupal development; osi8 and osi11 (Osiris protein family), CPRs and CPF (cuticle protein family), and stretchin (a muscle protein). Heritable CRISPR/Cas9-mediated gene knockout of these genes did not reveal any defects in pupal development. However, stretchin-null mutations (strnΔ35/Δ41) resulted in flightless mosquitoes with an abnormal open wing posture. The inability of adult strnΔ35/Δ41 mosquitoes to fly restricted their escape from aquatic rearing media following eclosion, and substantially reduced adult survival rates. Transgenic strains which contain the EGFP marker gene under the control of strn regulatory regions (0.8 kb, 1.4 kb, and 2.2 kb upstream, respectively), revealed the gene expression pattern of strn in muscle-like tissues in the thorax during late morphogenesis from L4 larvae to young adults. We demonstrated that Ae. aegypti pupae-specific strn is critical for adult mosquito flight capability and a key late-acting lethal target for mosquito-borne disease control.
Collapse
Affiliation(s)
- Keun Chae
- Department of Entomology, Texas A&M University, College Station, TX, 77843, USA
| | - Collin Valentin
- Department of Entomology, Texas A&M University, College Station, TX, 77843, USA
| | - Chanell Dawson
- Department of Entomology, Texas A&M University, College Station, TX, 77843, USA
| | - Emma Jakes
- Department of Entomology, Texas A&M University, College Station, TX, 77843, USA
| | - Kevin M Myles
- Department of Entomology, Texas A&M University, College Station, TX, 77843, USA
| | - Zach N Adelman
- Department of Entomology, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
13
|
The actin polymerization factor Diaphanous and the actin severing protein Flightless I collaborate to regulate sarcomere size. Dev Biol 2021; 469:12-25. [PMID: 32980309 DOI: 10.1016/j.ydbio.2020.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/15/2020] [Accepted: 09/19/2020] [Indexed: 12/22/2022]
Abstract
The sarcomere is the basic contractile unit of muscle, composed of repeated sets of actin thin filaments and myosin thick filaments. During muscle development, sarcomeres grow in size to accommodate the growth and function of muscle fibers. Failure in regulating sarcomere size results in muscle dysfunction; yet, it is unclear how the size and uniformity of sarcomeres are controlled. Here we show that the formin Diaphanous is critical for the growth and maintenance of sarcomere size: Dia sets sarcomere length and width through regulation of the number and length of the actin thin filaments in the Drosophila flight muscle. To regulate thin filament length and sarcomere size, Dia interacts with the Gelsolin superfamily member Flightless I (FliI). We suggest that these actin regulators, by controlling actin dynamics and turnover, generate uniformly sized sarcomeres tuned for the muscle contractions required for flight.
Collapse
|
14
|
Dahl-Halvarsson M, Olive M, Pokrzywa M, Norum M, Ejeskär K, Tajsharghi H. Impaired muscle morphology in a Drosophila model of myosin storage myopathy was supressed by overexpression of an E3 ubiquitin ligase. Dis Model Mech 2020; 13:dmm047886. [PMID: 33234710 PMCID: PMC7790189 DOI: 10.1242/dmm.047886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/02/2020] [Indexed: 11/29/2022] Open
Abstract
Myosin is vital for body movement and heart contractility. Mutations in MYH7, encoding slow/β-cardiac myosin heavy chain, are an important cause of hypertrophic and dilated cardiomyopathy, as well as skeletal muscle disease. A dominant missense mutation (R1845W) in MYH7 has been reported in several unrelated cases of myosin storage myopathy. We have developed a Drosophila model for a myosin storage myopathy in order to investigate the dose-dependent mechanisms underlying the pathological roles of the R1845W mutation. This study shows that a higher expression level of the mutated allele is concomitant with severe impairment of muscle function and progressively disrupted muscle morphology. The impaired muscle morphology associated with the mutant allele was suppressed by expression of Thin (herein referred to as Abba), an E3 ubiquitin ligase. This Drosophila model recapitulates pathological features seen in myopathy patients with the R1845W mutation and severe ultrastructural abnormalities, including extensive loss of thick filaments with selective A-band loss, and preservation of I-band and Z-disks were observed in indirect flight muscles of flies with exclusive expression of mutant myosin. Furthermore, the impaired muscle morphology associated with the mutant allele was suppressed by expression of Abba. These findings suggest that modification of the ubiquitin proteasome system may be beneficial in myosin storage myopathy by reducing the impact of MYH7 mutation in patients.
Collapse
Affiliation(s)
- Martin Dahl-Halvarsson
- Department of Pathology, Institute of Biomedicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Montse Olive
- Institute of Neuropathology, Department of Pathology and Neuromuscular Unit, Department of Neurology, IDIBELL-Hospital de Bellvitge, 08907 Hospitalet de Llobregat, Barcelona, Spain
| | - Malgorzata Pokrzywa
- Department of Pathology, Institute of Biomedicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Michaela Norum
- Department of Pathology, Institute of Biomedicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Katarina Ejeskär
- Translational Medicine, School of Health Sciences, University of Skövde, SE-541 28, Skövde, Sweden
| | - Homa Tajsharghi
- Translational Medicine, School of Health Sciences, University of Skövde, SE-541 28, Skövde, Sweden
| |
Collapse
|
15
|
Gong H, Ma W, Chen S, Wang G, Khairallah R, Irving T. Localization of the Elastic Proteins in the Flight Muscle of Manduca sexta. Int J Mol Sci 2020; 21:ijms21155504. [PMID: 32752103 PMCID: PMC7432240 DOI: 10.3390/ijms21155504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 11/16/2022] Open
Abstract
The flight muscle of Manduca sexta (DLM1) is an emerging model system for biophysical studies of muscle contraction. Unlike the well-studied indirect flight muscle of Lethocerus and Drosophila, the DLM1 of Manduca is a synchronous muscle, as are the vertebrate cardiac and skeletal muscles. Very little has been published regarding the ultrastructure and protein composition of this muscle. Previous studies have demonstrated that DLM1 express two projectin isoform, two kettin isoforms, and two large Salimus (Sls) isoforms. Such large Sls isoforms have not been observed in the asynchronous flight muscles of Lethocerus and Drosophila. The spatial localization of these proteins was unknown. Here, immuno-localization was used to show that the N-termini of projectin and Salimus are inserted into the Z-band. Projectin spans across the I-band, and the C-terminus is attached to the thick filament in the A-band. The C-terminus of Sls was also located in the A-band. Using confocal microscopy and experimental force-length curves, thin filament lengths were estimated as ~1.5 µm and thick filament lengths were measured as ~2.5 µm. This structural information may help provide an interpretive framework for future studies using this muscle system.
Collapse
Affiliation(s)
- Henry Gong
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA; (H.G.); (W.M.); (S.C.); (G.W.)
| | - Weikang Ma
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA; (H.G.); (W.M.); (S.C.); (G.W.)
| | - Shaoshuai Chen
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA; (H.G.); (W.M.); (S.C.); (G.W.)
| | - Geng Wang
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA; (H.G.); (W.M.); (S.C.); (G.W.)
| | - Ramzi Khairallah
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA;
| | - Thomas Irving
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA; (H.G.); (W.M.); (S.C.); (G.W.)
- Correspondence: ; Tel.: +1-(312)-567-3489; Fax: +1-(312)-566-3494
| |
Collapse
|
16
|
Szikora S, Gajdos T, Novák T, Farkas D, Földi I, Lenart P, Erdélyi M, Mihály J. Nanoscopy reveals the layered organization of the sarcomeric H-zone and I-band complexes. J Cell Biol 2020; 219:132617. [PMID: 31816054 PMCID: PMC7039190 DOI: 10.1083/jcb.201907026] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/04/2019] [Accepted: 10/22/2019] [Indexed: 01/18/2023] Open
Abstract
Sarcomeres are extremely highly ordered macromolecular assemblies where structural organization is intimately linked to their functionality as contractile units. Although the structural basis of actin and Myosin interaction is revealed at a quasiatomic resolution, much less is known about the molecular organization of the I-band and H-zone. We report the development of a powerful nanoscopic approach, combined with a structure-averaging algorithm, that allowed us to determine the position of 27 sarcomeric proteins in Drosophila melanogaster flight muscles with a quasimolecular, ∼5- to 10-nm localization precision. With this protein localization atlas and template-based protein structure modeling, we have assembled refined I-band and H-zone models with unparalleled scope and resolution. In addition, we found that actin regulatory proteins of the H-zone are organized into two distinct layers, suggesting that the major place of thin filament assembly is an M-line-centered narrow domain where short actin oligomers can form and subsequently anneal to the pointed end.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Tamás Gajdos
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Tibor Novák
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Dávid Farkas
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - István Földi
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Peter Lenart
- Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| |
Collapse
|
17
|
Poovathumkadavil P, Jagla K. Genetic Control of Muscle Diversification and Homeostasis: Insights from Drosophila. Cells 2020; 9:cells9061543. [PMID: 32630420 PMCID: PMC7349286 DOI: 10.3390/cells9061543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022] Open
Abstract
In the fruit fly, Drosophila melanogaster, the larval somatic muscles or the adult thoracic flight and leg muscles are the major voluntary locomotory organs. They share several developmental and structural similarities with vertebrate skeletal muscles. To ensure appropriate activity levels for their functions such as hatching in the embryo, crawling in the larva, and jumping and flying in adult flies all muscle components need to be maintained in a functionally stable or homeostatic state despite constant strain. This requires that the muscles develop in a coordinated manner with appropriate connections to other cell types they communicate with. Various signaling pathways as well as extrinsic and intrinsic factors are known to play a role during Drosophila muscle development, diversification, and homeostasis. In this review, we discuss genetic control mechanisms of muscle contraction, development, and homeostasis with particular emphasis on the contractile unit of the muscle, the sarcomere.
Collapse
|
18
|
Szikora S, Novák T, Gajdos T, Erdélyi M, Mihály J. Superresolution Microscopy of Drosophila Indirect Flight Muscle Sarcomeres. Bio Protoc 2020; 10:e3654. [PMID: 33659324 DOI: 10.21769/bioprotoc.3654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 11/02/2022] Open
Abstract
Sarcomeres are extremely highly ordered macromolecular assemblies where proper structural organization is an absolute prerequisite to the functionality of these contractile units. Despite the wealth of information collected, the exact spatial arrangement of many of the H-zone and Z-disk proteins remained unknown. Recently, we developed a powerful nanoscopic approach to localize the sarcomeric protein components with a resolution well below the diffraction limit. The ease of sample preparation and the near crystalline structure of the Drosophila flight muscle sarcomeres make them ideally suitable for single molecule localization microscopy and structure averaging. Our approach allowed us to determine the position of dozens of H-zone and Z-disk proteins with a quasi-molecular, ~5-10 nm localization precision. The protocol described below provides an easy and reproducible method to prepare individual myofibrils for dSTORM imaging. In addition, it includes an in-depth description of a custom made and freely available software toolbox to process and quantitatively analyze the raw localization data.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Szeged, Hungary.,Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Tibor Novák
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Tamás Gajdos
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Szeged, Hungary.,Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| |
Collapse
|
19
|
Tune TC, Ma W, Irving T, Sponberg S. Nanometer-scale structure differences in the myofilament lattice spacing of two cockroach leg muscles correspond to their different functions. J Exp Biol 2020; 223:jeb212829. [PMID: 32205362 PMCID: PMC7225125 DOI: 10.1242/jeb.212829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 03/09/2020] [Indexed: 01/21/2023]
Abstract
Muscle is highly organized across multiple length scales. Consequently, small changes in the arrangement of myofilaments can influence macroscopic mechanical function. Two leg muscles of a cockroach have identical innervation, mass, twitch responses, length-tension curves and force-velocity relationships. However, during running, one muscle is dissipative (a 'brake'), while the other dissipates and produces significant positive mechanical work (bifunctional). Using time-resolved X-ray diffraction in intact, contracting muscle, we simultaneously measured the myofilament lattice spacing, packing structure and macroscopic force production of these muscles to test whether structural differences in the myofilament lattice might correspond to the muscles' different mechanical functions. While the packing patterns are the same, one muscle has 1 nm smaller lattice spacing at rest. Under isometric stimulation, the difference in lattice spacing disappeared, consistent with the two muscles' identical steady-state behavior. During periodic contractions, one muscle undergoes a 1 nm greater change in lattice spacing, which correlates with force. This is the first identified structural feature in the myofilament lattice of these two muscles that shares their whole-muscle dynamic differences and quasi-static similarities.
Collapse
Affiliation(s)
- Travis Carver Tune
- School of Physics, Georgia Institute of Technology, Atlanta, GA, 30332 USA
| | - Weikang Ma
- Biophysics Collaborative Access Team and CSRRI, Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, 60616 USA
| | - Thomas Irving
- Biophysics Collaborative Access Team and CSRRI, Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, 60616 USA
| | - Simon Sponberg
- School of Physics, Georgia Institute of Technology, Atlanta, GA, 30332 USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332 USA
| |
Collapse
|
20
|
Nikonova E, Kao SY, Spletter ML. Contributions of alternative splicing to muscle type development and function. Semin Cell Dev Biol 2020; 104:65-80. [PMID: 32070639 DOI: 10.1016/j.semcdb.2020.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/30/2022]
Abstract
Animals possess a wide variety of muscle types that support different kinds of movements. Different muscles have distinct locations, morphologies and contractile properties, raising the question of how muscle diversity is generated during development. Normal aging processes and muscle disorders differentially affect particular muscle types, thus understanding how muscles normally develop and are maintained provides insight into alterations in disease and senescence. As muscle structure and basic developmental mechanisms are highly conserved, many important insights into disease mechanisms in humans as well as into basic principles of muscle development have come from model organisms such as Drosophila, zebrafish and mouse. While transcriptional regulation has been characterized to play an important role in myogenesis, there is a growing recognition of the contributions of alternative splicing to myogenesis and the refinement of muscle function. Here we review our current understanding of muscle type specific alternative splicing, using examples of isoforms with distinct functions from both vertebrates and Drosophila. Future exploration of the vast potential of alternative splicing to fine-tune muscle development and function will likely uncover novel mechanisms of isoform-specific regulation and a more holistic understanding of muscle development, disease and aging.
Collapse
Affiliation(s)
- Elena Nikonova
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, 82152 Martinsried-Planegg, Germany
| | - Shao-Yen Kao
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, 82152 Martinsried-Planegg, Germany
| | - Maria L Spletter
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, 82152 Martinsried-Planegg, Germany; Center for Integrated Protein Science Munich (CIPSM) at the Department of Chemistry, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
21
|
Ono K, Qin Z, Johnsen RC, Baillie DL, Ono S. Kettin, the large actin-binding protein with multiple immunoglobulin domains, is essential for sarcomeric actin assembly and larval development in Caenorhabditis elegans. FEBS J 2019; 287:659-670. [PMID: 31411810 DOI: 10.1111/febs.15039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/29/2019] [Accepted: 08/12/2019] [Indexed: 12/30/2022]
Abstract
Among many essential genes in the nematode Caenorhabditis elegans, let-330 is located on the left arm of chromosome V and was identified as the largest target of a mutagen in this region. However, let-330 gene has not been characterized at the molecular level. Here, we report that two sequenced let-330 alleles are nonsense mutations of ketn-1, a previously characterized gene encoding kettin. Kettin is a large actin-binding protein of 472 kDa with 31 immunoglobulin domains and is expressed in muscle cells in C. elegans. let-330/ketn-1 mutants are homozygous lethal at the first larval stage with mild defects in body elongation. These mutants have severe defects in sarcomeric actin and myosin assembly in striated muscle. However, α-actinin and vinculin, which are components of the dense bodies anchoring actin to the membranes, were not significantly disorganized by let-330/ketn-1 mutation. Kettin localizes to embryonic myofibrils before α-actinin is expressed, and α-actinin deficiency does not affect kettin localization in larval muscle. Depletion of vinculin minimally affects kettin localization but significantly reduces colocalization of actin with kettin in embryonic muscle cells. These results indicate that kettin is an essential protein for sarcomeric assembly of actin filaments in muscle cells.
Collapse
Affiliation(s)
- Kanako Ono
- Department of Pathology, Emory University, Atlanta, GA, USA.,Department of Cell Biology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Zhaozhao Qin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Robert C Johnsen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - David L Baillie
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Shoichiro Ono
- Department of Pathology, Emory University, Atlanta, GA, USA.,Department of Cell Biology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
22
|
Phillips MA, Rutledge GA, Kezos JN, Greenspan ZS, Talbott A, Matty S, Arain H, Mueller LD, Rose MR, Shahrestani P. Effects of evolutionary history on genome wide and phenotypic convergence in Drosophila populations. BMC Genomics 2018; 19:743. [PMID: 30305018 PMCID: PMC6180417 DOI: 10.1186/s12864-018-5118-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 09/26/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Studies combining experimental evolution and next-generation sequencing have found that adaptation in sexually reproducing populations is primarily fueled by standing genetic variation. Consequently, the response to selection is rapid and highly repeatable across replicate populations. Some studies suggest that the response to selection is highly repeatable at both the phenotypic and genomic levels, and that evolutionary history has little impact. Other studies suggest that even when the response to selection is repeatable phenotypically, evolutionary history can have significant impacts at the genomic level. Here we test two hypotheses that may explain this discrepancy. Hypothesis 1: Past intense selection reduces evolutionary repeatability at the genomic and phenotypic levels when conditions change. Hypothesis 2: Previous intense selection does not reduce evolutionary repeatability, but other evolutionary mechanisms may. We test these hypotheses using D. melanogaster populations that were subjected to 260 generations of intense selection for desiccation resistance and have since been under relaxed selection for the past 230 generations. RESULTS We find that, with the exception of longevity and to a lesser extent fecundity, 230 generations of relaxed selection has erased the extreme phenotypic differentiation previously found. We also find no signs of genetic fixation, and only limited evidence of genetic differentiation between previously desiccation resistance selected populations and their controls. CONCLUSION Our findings suggest that evolution in our system is highly repeatable even when populations have been previously subjected to bouts of extreme selection. We therefore conclude that evolutionary repeatability can overcome past bouts of extreme selection in Drosophila experimental evolution, provided experiments are sufficiently long and populations are not inbred.
Collapse
Affiliation(s)
- Mark A Phillips
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, USA.
| | - Grant A Rutledge
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, USA
| | - James N Kezos
- Department of Development, Aging, and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, San Diego, USA
| | - Zachary S Greenspan
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, USA
| | - Andrew Talbott
- Department of Biological Science, California State University Fullerton, 800 N State College Blvd, Fullerton, CA, 92831, USA
| | - Sara Matty
- Department of Biological Science, California State University Fullerton, 800 N State College Blvd, Fullerton, CA, 92831, USA
| | - Hamid Arain
- Department of Biological Science, California State University Fullerton, 800 N State College Blvd, Fullerton, CA, 92831, USA
| | - Laurence D Mueller
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, USA
| | - Michael R Rose
- Department of Ecology and Evolutionary Biology, University of California Irvine, Irvine, USA
| | - Parvin Shahrestani
- Department of Biological Science, California State University Fullerton, 800 N State College Blvd, Fullerton, CA, 92831, USA
| |
Collapse
|
23
|
Palandri A, Martin E, Russi M, Rera M, Tricoire H, Monnier V. Identification of cardioprotective drugs by medium-scale in vivo pharmacological screening on a Drosophila cardiac model of Friedreich's ataxia. Dis Model Mech 2018; 11:dmm033811. [PMID: 29898895 PMCID: PMC6078405 DOI: 10.1242/dmm.033811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/06/2018] [Indexed: 12/30/2022] Open
Abstract
Friedreich's ataxia (FA) is caused by reduced levels of frataxin, a highly conserved mitochondrial protein. There is currently no effective treatment for this disease, which is characterized by progressive neurodegeneration and cardiomyopathy, the latter being the most common cause of death in patients. We previously developed a Drosophila melanogaster cardiac model of FA, in which the fly frataxin is inactivated specifically in the heart, leading to heart dilatation and impaired systolic function. Methylene Blue (MB) was highly efficient to prevent these cardiac dysfunctions. Here, we used this model to screen in vivo the Prestwick Chemical Library, comprising 1280 compounds. Eleven drugs significantly reduced the cardiac dilatation, some of which may possibly lead to therapeutic applications in the future. The one with the strongest protective effects was paclitaxel, a microtubule-stabilizing drug. In parallel, we characterized the histological defects induced by frataxin deficiency in cardiomyocytes and observed strong sarcomere alterations with loss of striation of actin fibers, along with full disruption of the microtubule network. Paclitaxel and MB both improved these structural defects. Therefore, we propose that frataxin inactivation induces cardiac dysfunction through impaired sarcomere assembly or renewal due to microtubule destabilization, without excluding additional mechanisms. This study is the first drug screening of this extent performed in vivo on a Drosophila model of cardiac disease. Thus, it also brings the proof of concept that cardiac functional imaging in adult Drosophila flies is usable for medium-scale in vivo pharmacological screening, with potent identification of cardioprotective drugs in various contexts of cardiac diseases.
Collapse
Affiliation(s)
- Amandine Palandri
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA) UMR8251 CNRS, 75205, Paris Cedex 13, France
| | - Elodie Martin
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA) UMR8251 CNRS, 75205, Paris Cedex 13, France
| | - Maria Russi
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA) UMR8251 CNRS, 75205, Paris Cedex 13, France
| | - Michael Rera
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA) UMR8251 CNRS, 75205, Paris Cedex 13, France
| | - Hervé Tricoire
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA) UMR8251 CNRS, 75205, Paris Cedex 13, France
| | - Véronique Monnier
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA) UMR8251 CNRS, 75205, Paris Cedex 13, France
| |
Collapse
|
24
|
Green HJ, Griffiths AGM, Ylänne J, Brown NH. Novel functions for integrin-associated proteins revealed by analysis of myofibril attachment in Drosophila. eLife 2018; 7:e35783. [PMID: 30028294 PMCID: PMC6092120 DOI: 10.7554/elife.35783] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/19/2018] [Indexed: 01/18/2023] Open
Abstract
We use the myotendinous junction of Drosophila flight muscles to explore why many integrin associated proteins (IAPs) are needed and how their function is coordinated. These muscles revealed new functions for IAPs not required for viability: Focal Adhesion Kinase (FAK), RSU1, tensin and vinculin. Genetic interactions demonstrated a balance between positive and negative activities, with vinculin and tensin positively regulating adhesion, while FAK inhibits elevation of integrin activity by tensin, and RSU1 keeps PINCH activity in check. The molecular composition of myofibril termini resolves into 4 distinct layers, one of which is built by a mechanotransduction cascade: vinculin facilitates mechanical opening of filamin, which works with the Arp2/3 activator WASH to build an actin-rich layer positioned between integrins and the first sarcomere. Thus, integration of IAP activity is needed to build the complex architecture of the myotendinous junction, linking the membrane anchor to the sarcomere.
Collapse
Affiliation(s)
- Hannah J Green
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
- Department of Biological and Environmental SciencesUniversity of JyväskyläJyväskyläFinland
- Nanoscience CenterUniversity of JyväskyläJyväskyläFinland
| | - Annabel GM Griffiths
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| | - Jari Ylänne
- Department of Biological and Environmental SciencesUniversity of JyväskyläJyväskyläFinland
- Nanoscience CenterUniversity of JyväskyläJyväskyläFinland
| | - Nicholas H Brown
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
25
|
Drosophila model of myosin myopathy rescued by overexpression of a TRIM-protein family member. Proc Natl Acad Sci U S A 2018; 115:E6566-E6575. [PMID: 29946036 DOI: 10.1073/pnas.1800727115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Myosin is a molecular motor indispensable for body movement and heart contractility. Apart from pure cardiomyopathy, mutations in MYH7 encoding slow/β-cardiac myosin heavy chain also cause skeletal muscle disease with or without cardiac involvement. Mutations within the α-helical rod domain of MYH7 are mainly associated with Laing distal myopathy. To investigate the mechanisms underlying the pathology of the recurrent causative MYH7 mutation (K1729del), we have developed a Drosophila melanogaster model of Laing distal myopathy by genomic engineering of the Drosophila Mhc locus. Homozygous MhcK1728del animals die during larval/pupal stages, and both homozygous and heterozygous larvae display reduced muscle function. Flies expressing only MhcK1728del in indirect flight and jump muscles, and heterozygous MhcK1728del animals, were flightless, with reduced movement and decreased lifespan. Sarcomeres of MhcK1728del mutant indirect flight muscles and larval body wall muscles were disrupted with clearly disorganized muscle filaments. Homozygous MhcK1728del larvae also demonstrated structural and functional impairments in heart muscle, which were not observed in heterozygous animals, indicating a dose-dependent effect of the mutated allele. The impaired jump and flight ability and the myopathy of indirect flight and leg muscles associated with MhcK1728del were fully suppressed by expression of Abba/Thin, an E3-ligase that is essential for maintaining sarcomere integrity. This model of Laing distal myopathy in Drosophila recapitulates certain morphological phenotypic features seen in Laing distal myopathy patients with the recurrent K1729del mutation. Our observations that Abba/Thin modulates these phenotypes suggest that manipulation of Abba/Thin activity levels may be beneficial in Laing distal myopathy.
Collapse
|
26
|
Spletter ML, Barz C, Yeroslaviz A, Zhang X, Lemke SB, Bonnard A, Brunner E, Cardone G, Basler K, Habermann BH, Schnorrer F. A transcriptomics resource reveals a transcriptional transition during ordered sarcomere morphogenesis in flight muscle. eLife 2018; 7:34058. [PMID: 29846170 PMCID: PMC6005683 DOI: 10.7554/elife.34058] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/26/2018] [Indexed: 01/07/2023] Open
Abstract
Muscles organise pseudo-crystalline arrays of actin, myosin and titin filaments to build force-producing sarcomeres. To study sarcomerogenesis, we have generated a transcriptomics resource of developing Drosophila flight muscles and identified 40 distinct expression profile clusters. Strikingly, most sarcomeric components group in two clusters, which are strongly induced after all myofibrils have been assembled, indicating a transcriptional transition during myofibrillogenesis. Following myofibril assembly, many short sarcomeres are added to each myofibril. Subsequently, all sarcomeres mature, reaching 1.5 µm diameter and 3.2 µm length and acquiring stretch-sensitivity. The efficient induction of the transcriptional transition during myofibrillogenesis, including the transcriptional boost of sarcomeric components, requires in part the transcriptional regulator Spalt major. As a consequence of Spalt knock-down, sarcomere maturation is defective and fibers fail to gain stretch-sensitivity. Together, this defines an ordered sarcomere morphogenesis process under precise transcriptional control - a concept that may also apply to vertebrate muscle or heart development.
Collapse
Affiliation(s)
- Maria L Spletter
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
- Biomedical Center, Physiological ChemistryLudwig-Maximilians-Universität MünchenMartinsriedGermany
| | - Christiane Barz
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
| | - Assa Yeroslaviz
- Computational Biology GroupMax Planck Institute of BiochemistryMartinsriedGermany
| | - Xu Zhang
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
- Aix Marseille Univ, CNRS, IBDMMarseilleFrance
- School of Life Science and EngineeringFoshan UniversityGuangdongChina
| | - Sandra B Lemke
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
| | - Adrien Bonnard
- Aix Marseille Univ, CNRS, IBDMMarseilleFrance
- Aix Marseille Univ, INSERM, TAGCMarseilleFrance
| | - Erich Brunner
- Institute of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | - Giovanni Cardone
- Imaging FacilityMax Planck Institute of BiochemistryMartinsriedGermany
| | - Konrad Basler
- Institute of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | - Bianca H Habermann
- Computational Biology GroupMax Planck Institute of BiochemistryMartinsriedGermany
- Aix Marseille Univ, CNRS, IBDMMarseilleFrance
- Aix Marseille Univ, INSERM, TAGCMarseilleFrance
| | - Frank Schnorrer
- Muscle Dynamics GroupMax Planck Institute of BiochemistryMartinsriedGermany
- Aix Marseille Univ, CNRS, IBDMMarseilleFrance
| |
Collapse
|
27
|
Bulgakova NA, Wellmann J, Brown NH. Diverse integrin adhesion stoichiometries caused by varied actomyosin activity. Open Biol 2018; 7:rsob.160250. [PMID: 28446705 PMCID: PMC5413901 DOI: 10.1098/rsob.160250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/17/2017] [Indexed: 12/14/2022] Open
Abstract
Cells in an organism are subjected to numerous sources of external and internal forces, and are able to sense and respond to these forces. Integrin-mediated adhesion links the extracellular matrix outside cells to the cytoskeleton inside, and participates in sensing, transmitting and responding to forces. While integrin adhesion rapidly adapts to changes in forces in isolated migrating cells, it is not known whether similar or more complex responses occur within intact, developing tissues. Here, we studied changes in integrin adhesion composition upon different contractility conditions in Drosophila embryonic muscles. We discovered that all integrin adhesion components tested were still present at muscle attachment sites (MASs) when either cytoplasmic or muscle myosin II was genetically removed, suggesting a primary role of a developmental programme in the initial assembly of integrin adhesions. Contractility does, however, increase the levels of integrin adhesion components, suggesting a mechanism to balance the strength of muscle attachment to the force of muscle contraction. Perturbing contractility in distinct ways, by genetic removal of either cytoplasmic or muscle myosin II or eliminating muscle innervation, each caused unique alterations to the stoichiometry at MASs. This suggests that different integrin-associated proteins are added to counteract different kinds of force increase.
Collapse
Affiliation(s)
- Natalia A Bulgakova
- Department of Physiology, Development and Neuroscience and The Gurdon Institute, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Jutta Wellmann
- Department of Physiology, Development and Neuroscience and The Gurdon Institute, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience and The Gurdon Institute, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
28
|
Glasheen BM, Eldred CC, Sullivan LC, Zhao C, Reedy MK, Edwards RJ, Swank DM. Stretch activation properties of Drosophila and Lethocerus indirect flight muscle suggest similar calcium-dependent mechanisms. Am J Physiol Cell Physiol 2017; 313:C621-C631. [PMID: 28835434 DOI: 10.1152/ajpcell.00110.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Muscle stretch activation (SA) is critical for optimal cardiac and insect indirect flight muscle (IFM) power generation. The SA mechanism has been investigated for decades with many theories proposed, but none proven. One reason for the slow progress could be that multiple SA mechanisms may have evolved in multiple species or muscle types. Laboratories studying IFM SA in the same or different species have reported differing SA functional properties which would, if true, suggest divergent mechanisms. However, these conflicting results might be due to different experimental methodologies. Thus, we directly compared SA characteristics of IFMs from two SA model systems, Drosophila and Lethocerus, using two different fiber bathing solutions. Compared with Drosophila IFM, Lethocerus IFM isometric tension is 10- or 17-fold higher and SA tension was 5- or 10-fold higher, depending on the bathing solution. However, the rate of SA tension generation was 9-fold faster for Drosophila IFM. The inverse differences between rate and tension in the two species causes maximum power output to be similar, where Drosophila power is optimized in the bathing solution that favors faster muscle kinetics and Lethocerus in the solution that favors greater tension generation. We found that isometric tension and SA tension increased with calcium concentration for both species in both solutions, reaching a maximum plateau around pCa 5.0. Our results favor a similar mechanism for both species, perhaps involving a troponin complex that does not fully calcium activate the thin filament thus leaving room for further tension generation by SA.
Collapse
Affiliation(s)
- Bernadette M Glasheen
- Department of Biological Sciences, Biomedical Engineering, & Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York
| | - Catherine C Eldred
- Department of Biological Sciences, Biomedical Engineering, & Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York
| | - Leah C Sullivan
- Department of Biological Sciences, Biomedical Engineering, & Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York
| | - Cuiping Zhao
- Department of Biological Sciences, Biomedical Engineering, & Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York
| | - Michael K Reedy
- Department of Cell Biology, Duke University , Durham North Carolina
| | - Robert J Edwards
- Department of Cell Biology, Duke University , Durham North Carolina
| | - Douglas M Swank
- Department of Biological Sciences, Biomedical Engineering, & Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York
| |
Collapse
|
29
|
Randazzo D, Pierantozzi E, Rossi D, Sorrentino V. The potential of obscurin as a therapeutic target in muscle disorders. Expert Opin Ther Targets 2017; 21:897-910. [DOI: 10.1080/14728222.2017.1361931] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Davide Randazzo
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Daniela Rossi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
30
|
González-Morales N, Holenka TK, Schöck F. Filamin actin-binding and titin-binding fulfill distinct functions in Z-disc cohesion. PLoS Genet 2017; 13:e1006880. [PMID: 28732005 PMCID: PMC5521747 DOI: 10.1371/journal.pgen.1006880] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 06/21/2017] [Indexed: 01/03/2023] Open
Abstract
Many proteins contribute to the contractile properties of muscles, most notably myosin thick filaments, which are anchored at the M-line, and actin thin filaments, which are anchored at the Z-discs that border each sarcomere. In humans, mutations in the actin-binding protein Filamin-C result in myopathies, but the underlying molecular function is not well understood. Here we show using Drosophila indirect flight muscle that the filamin ortholog Cheerio in conjunction with the giant elastic protein titin plays a crucial role in keeping thin filaments stably anchored at the Z-disc. We identify the filamin domains required for interaction with the titin ortholog Sallimus, and we demonstrate a genetic interaction of filamin with titin and actin. Filamin mutants disrupting the actin- or the titin-binding domain display distinct phenotypes, with Z-discs breaking up in parallel or perpendicularly to the myofibril, respectively. Thus, Z-discs require filamin to withstand the strong contractile forces acting on them. The Z-disc is a macromolecular complex required to attach and stabilize actin thin filaments in the sarcomere, the smallest contractile unit of striated muscles. Mutations in Z-disc-associated proteins typically result in muscle disorders. Dimeric filamin organizes actin filaments, localizes at the Z-disc in vertebrates and causes muscle disorders in humans when mutated. Despite its clinical relevance, the molecular function of filamin in the sarcomere is not well understood. Here we use Drosophila muscles and an array of filamin mutations to address the molecular and cell biological function of filamin in the sarcomere. We show that filamin mainly serves as a Z-disc cohesive element, binding both thin filaments and titin. This configuration enables filamin to act as a bridge between thin filaments and the elastic scaffold protein titin from the adjacent sarcomere, maintaining sarcomere stability during muscle contraction.
Collapse
Affiliation(s)
| | | | - Frieder Schöck
- Department of Biology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
31
|
Lindstedt S, Nishikawa K. Huxleys’ Missing Filament: Form and Function of Titin in Vertebrate Striated Muscle. Annu Rev Physiol 2017; 79:145-166. [DOI: 10.1146/annurev-physiol-022516-034152] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Stan Lindstedt
- Center for Bioengineering Innovation, Northern Arizona University, Flagstaff, Arizona 86011-4185
| | - Kiisa Nishikawa
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona 86011-4185;
| |
Collapse
|
32
|
Fujita N, Huang W, Lin TH, Groulx JF, Jean S, Nguyen J, Kuchitsu Y, Koyama-Honda I, Mizushima N, Fukuda M, Kiger AA. Genetic screen in Drosophila muscle identifies autophagy-mediated T-tubule remodeling and a Rab2 role in autophagy. eLife 2017; 6. [PMID: 28063257 PMCID: PMC5249261 DOI: 10.7554/elife.23367] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 12/17/2016] [Indexed: 01/04/2023] Open
Abstract
Transverse (T)-tubules make-up a specialized network of tubulated muscle cell membranes involved in excitation-contraction coupling for power of contraction. Little is known about how T-tubules maintain highly organized structures and contacts throughout the contractile system despite the ongoing muscle remodeling that occurs with muscle atrophy, damage and aging. We uncovered an essential role for autophagy in T-tubule remodeling with genetic screens of a developmentally regulated remodeling program in Drosophila abdominal muscles. Here, we show that autophagy is both upregulated with and required for progression through T-tubule disassembly stages. Along with known mediators of autophagosome-lysosome fusion, our screens uncovered an unexpected shared role for Rab2 with a broadly conserved function in autophagic clearance. Rab2 localizes to autophagosomes and binds to HOPS complex members, suggesting a direct role in autophagosome tethering/fusion. Together, the high membrane flux with muscle remodeling permits unprecedented analysis both of T-tubule dynamics and fundamental trafficking mechanisms. DOI:http://dx.doi.org/10.7554/eLife.23367.001
Collapse
Affiliation(s)
- Naonobu Fujita
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States.,Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Wilson Huang
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Tzu-Han Lin
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Jean-Francois Groulx
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Steve Jean
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Jen Nguyen
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Yoshihiko Kuchitsu
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Ikuko Koyama-Honda
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Amy A Kiger
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| |
Collapse
|
33
|
Abstract
In this review we discuss the history and the current state of ideas related to the mechanism of size regulation of the thick (myosin) and thin (actin) filaments in vertebrate striated muscles. Various hypotheses have been considered during of more than half century of research, recently mostly involving titin and nebulin acting as templates or 'molecular rulers', terminating exact assembly. These two giant, single-polypeptide, filamentous proteins are bound in situ along the thick and thin filaments, respectively, with an almost perfect match in the respective lengths and structural periodicities. However, evidence still questions the possibility that the proteins function as templates, or scaffolds, on which the thin and thick filaments could be assembled. In addition, the progress in muscle research during the last decades highlighted a number of other factors that could potentially be involved in the mechanism of length regulation: molecular chaperones that may guide folding and assembly of actin and myosin; capping proteins that can influence the rates of assembly-disassembly of the myofilaments; Ca2+ transients that can activate or deactivate protein interactions, etc. The entire mechanism of sarcomere assembly appears complex and highly dynamic. This mechanism is also capable of producing filaments of about the correct size without titin and nebulin. What then is the role of these proteins? Evidence points to titin and nebulin stabilizing structures of the respective filaments. This stabilizing effect, based on linear proteins of a fixed size, implies that titin and nebulin are indeed molecular rulers of the filaments. Although the proteins may not function as templates in the assembly of the filaments, they measure and stabilize exactly the same size of the functionally important for the muscles segments in each of the respective filaments.
Collapse
|
34
|
Zhao C, Swank DM. The Drosophila indirect flight muscle myosin heavy chain isoform is insufficient to transform the jump muscle into a highly stretch-activated muscle type. Am J Physiol Cell Physiol 2016; 312:C111-C118. [PMID: 27881413 DOI: 10.1152/ajpcell.00284.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 11/22/2022]
Abstract
Stretch activation (SA) is a delayed increase in force that enables high power and efficiency from a cyclically contracting muscle. SA exists in various degrees in almost all muscle types. In Drosophila, the indirect flight muscle (IFM) displays exceptionally high SA force production (FSA), whereas the jump muscle produces only minimal FSA We previously found that expressing an embryonic (EMB) myosin heavy chain (MHC) isoform in the jump muscle transforms it into a moderately SA muscle type and enables positive cyclical power generation. To investigate whether variation in MHC isoforms is sufficient to produce even higher FSA, we substituted the IFM MHC isoform (IFI) into the jump muscle. Surprisingly, we found that IFI only caused a 1.7-fold increase in FSA, less than half the increase previously observed with EMB, and only at a high Pi concentration, 16 mM. This IFI-induced FSA is much less than what occurs in IFM, relative to isometric tension, and did not enable positive cyclical power generation by the jump muscle. Both isometric tension and FSA of control fibers decreased with increasing Pi concentration. However, for IFI-expressing fibers, only isometric tension decreased. The rate of FSA generation was ~1.5-fold faster for IFI fibers than control fibers, and both rates were Pi dependent. We conclude that MHC isoforms can alter FSA and hence cyclical power generation but that isoforms can only endow a muscle type with moderate FSA Highly SA muscle types, such as IFM, likely use a different or additional mechanism.
Collapse
Affiliation(s)
- Cuiping Zhao
- Department of Biological Sciences, Department of Biomedical Engineering, and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - Douglas M Swank
- Department of Biological Sciences, Department of Biomedical Engineering, and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
35
|
Gautel M, Djinović-Carugo K. The sarcomeric cytoskeleton: from molecules to motion. ACTA ACUST UNITED AC 2016; 219:135-45. [PMID: 26792323 DOI: 10.1242/jeb.124941] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Highly ordered organisation of striated muscle is the prerequisite for the fast and unidirectional development of force and motion during heart and skeletal muscle contraction. A group of proteins, summarised as the sarcomeric cytoskeleton, is essential for the ordered assembly of actin and myosin filaments into sarcomeres, by combining architectural, mechanical and signalling functions. This review discusses recent cell biological, biophysical and structural insight into the regulated assembly of sarcomeric cytoskeleton proteins and their roles in dissipating mechanical forces in order to maintain sarcomere integrity during passive extension and active contraction. α-Actinin crosslinks in the Z-disk show a pivot-and-rod structure that anchors both titin and actin filaments. In contrast, the myosin crosslinks formed by myomesin in the M-band are of a ball-and-spring type and may be crucial in providing stable yet elastic connections during active contractions, especially eccentric exercise.
Collapse
Affiliation(s)
- Mathias Gautel
- King's College London BHF Centre of Research Excellence, Randall Division for Cell and Molecular Biophysics, and Cardiovascular Division, New Hunt's House, London SE1 1UL, UK
| | - Kristina Djinović-Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, Vienna A-1030, Austria Department of Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Aškerčeva 5, Ljubljana 1000, Slovenia
| |
Collapse
|
36
|
Shwartz A, Dhanyasi N, Schejter ED, Shilo BZ. The Drosophila formin Fhos is a primary mediator of sarcomeric thin-filament array assembly. eLife 2016; 5. [PMID: 27731794 PMCID: PMC5061545 DOI: 10.7554/elife.16540] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/15/2016] [Indexed: 01/26/2023] Open
Abstract
Actin-based thin filament arrays constitute a fundamental core component of muscle sarcomeres. We have used formation of the Drosophila indirect flight musculature for studying the assembly and maturation of thin-filament arrays in a skeletal muscle model system. Employing GFP-tagged actin monomer incorporation, we identify several distinct phases in the dynamic construction of thin-filament arrays. This sequence includes assembly of nascent arrays after an initial period of intensive microfilament synthesis, followed by array elongation, primarily from filament pointed-ends, radial growth of the arrays via recruitment of peripheral filaments and continuous barbed-end turnover. Using genetic approaches we have identified Fhos, the single Drosophila homolog of the FHOD sub-family of formins, as a primary and versatile mediator of IFM thin-filament organization. Localization of Fhos to the barbed-ends of the arrays, achieved via a novel N-terminal domain, appears to be a critical aspect of its sarcomeric roles. DOI:http://dx.doi.org/10.7554/eLife.16540.001 Muscles owe their ability to contract to structural units called sarcomeres, and a single muscle fiber can contain many thousands of these structures, aligned one next to the other. Each mature sarcomere is made up of precisely arranged and intertwined thin filaments of actin and thicker bundles of motor proteins, surrounded by other proteins. Sliding the motors along the filaments provides the force needed to contract the muscle. However, it was far from clear how sarcomeres, especially the arrays of thin-filaments, are assembled from scratch in developing muscles. When the fruit fly Drosophila transforms from a larva into an adult, it needs to build muscles to move its newly forming wings. While smaller in size, these flight muscles closely resemble the skeletal muscles of animals with backbones, and therefore serve as a good model for muscle formation in general. New muscles require new sarcomeres too, and now Shwartz et al. have observed and monitored sarcomeres assembling in developing flight muscles of fruit flies, a process that takes about three days. The analysis made use of genetically engineered flies in which the gene for a fluorescently labeled version of actin, the building block of the thin filaments, could be switched on at specific points in time. Looking at how these green-glowing proteins become incorporated into the growing sarcomere revealed that the assembly process involves four different phases. First, a large store of unorganized and newly-made thin filaments is generated for future use. These filaments are then assembled into rudimentary structures in which the filaments are roughly aligned. Once these core structures are formed, the existing filaments are elongated, while additional filaments are brought in to expand the structure further. Finally, actin proteins are continuously added and removed at the part of the sarcomere where the thin filaments are anchored. Shwartz et al. went on to identify a protein termed Fhos as the chief player in the process. Fhos is a member of a family of proteins that are known to elongate and organize actin filaments in many different settings. Without Fhos, the thin-filament arrays cannot properly begin to assemble, and the subsequent steps of growth and expansion are blocked as well. The next challenges will be to understand what guides the initial stages in the assembly of the thin-filament array, and how the coordination between assembly of actin filament arrays and motor proteins is executed. It will also be important to determine how sarcomeres are maintained throughout the life of the organism when defective actin filaments are replaced, and which proteins are responsible for carrying out this process. DOI:http://dx.doi.org/10.7554/eLife.16540.002
Collapse
Affiliation(s)
- Arkadi Shwartz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nagaraju Dhanyasi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
37
|
Zappia MP, Frolov MV. E2F function in muscle growth is necessary and sufficient for viability in Drosophila. Nat Commun 2016; 7:10509. [PMID: 26823289 PMCID: PMC4740182 DOI: 10.1038/ncomms10509] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/22/2015] [Indexed: 01/08/2023] Open
Abstract
The E2F transcription factor is a key cell cycle regulator. However, the inactivation of the entire E2F family in Drosophila is permissive throughout most of animal development until pupation when lethality occurs. Here we show that E2F function in the adult skeletal muscle is essential for animal viability since providing E2F function in muscles rescues the lethality of the whole-body E2F-deficient animals. Muscle-specific loss of E2F results in a significant reduction in muscle mass and thinner myofibrils. We demonstrate that E2F is dispensable for proliferation of muscle progenitor cells, but is required during late myogenesis to directly control the expression of a set of muscle-specific genes. Interestingly, E2f1 provides a major contribution to the regulation of myogenic function, while E2f2 appears to be less important. These findings identify a key function of E2F in skeletal muscle required for animal viability, and illustrate how the cell cycle regulator is repurposed in post-mitotic cells.
Collapse
Affiliation(s)
- Maria Paula Zappia
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Avenue, Chicago, Illinois 60607, USA
| | - Maxim V. Frolov
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland Avenue, Chicago, Illinois 60607, USA
| |
Collapse
|
38
|
Katzemich A, West RJH, Fukuzawa A, Sweeney ST, Gautel M, Sparrow J, Bullard B. Binding partners of the kinase domains in Drosophila obscurin and their effect on the structure of the flight muscle. J Cell Sci 2015; 128:3386-97. [PMID: 26251439 DOI: 10.1242/jcs.170639] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/27/2015] [Indexed: 01/15/2023] Open
Abstract
Drosophila obscurin (Unc-89) is a titin-like protein in the M-line of the muscle sarcomere. Obscurin has two kinase domains near the C-terminus, both of which are predicted to be inactive. We have identified proteins binding to the kinase domains. Kinase domain 1 bound Bällchen (Ball, an active kinase), and both kinase domains 1 and 2 bound MASK (a 400-kDa protein with ankyrin repeats). Ball was present in the Z-disc and M-line of the indirect flight muscle (IFM) and was diffusely distributed in the sarcomere. MASK was present in both the M-line and the Z-disc. Reducing expression of Ball or MASK by siRNA resulted in abnormalities in the IFM, including missing M-lines and multiple Z-discs. Obscurin was still present, suggesting that the kinase domains act as a scaffold binding Ball and MASK. Unlike obscurin in vertebrate skeletal muscle, Drosophila obscurin is necessary for the correct assembly of the IFM sarcomere. We show that Ball and MASK act downstream of obscurin, and both are needed for development of a well defined M-line and Z-disc. The proteins have not previously been identified in Drosophila muscle.
Collapse
Affiliation(s)
- Anja Katzemich
- Department of Biology, University of York, York YO10 5DD, UK
| | - Ryan J H West
- Department of Biology, University of York, York YO10 5DD, UK
| | - Atsushi Fukuzawa
- King's College BHF Centre, Cardiovascular Division, London SE1 1UL, UK
| | - Sean T Sweeney
- Department of Biology, University of York, York YO10 5DD, UK
| | - Mathias Gautel
- King's College BHF Centre, Cardiovascular Division, London SE1 1UL, UK
| | - John Sparrow
- Department of Biology, University of York, York YO10 5DD, UK
| | - Belinda Bullard
- Department of Biology, University of York, York YO10 5DD, UK
| |
Collapse
|
39
|
Wang S, Reuveny A, Volk T. Nesprin provides elastic properties to muscle nuclei by cooperating with spectraplakin and EB1. ACTA ACUST UNITED AC 2015; 209:529-38. [PMID: 26008743 PMCID: PMC4442817 DOI: 10.1083/jcb.201408098] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The myonuclear scaffold in Drosophila larval muscles exhibits both elastic features, contributed by the stretching capacity of MSP300/nesprin, and rigidity, provided by a perinuclear network of microtubules stabilized by Shot/spectraplakin and EB1. Muscle nuclei are exposed to variable cytoplasmic strain produced by muscle contraction and relaxation, but their morphology remains stable. Still, the mechanism responsible for maintaining myonuclear architecture, and its importance, is currently elusive. Herein, we uncovered a unique myonuclear scaffold in Drosophila melanogaster larval muscles, exhibiting both elastic features contributed by the stretching capacity of MSP300 (nesprin) and rigidity provided by a perinuclear network of microtubules stabilized by Shot (spectraplakin) and EB1. Together, they form a flexible perinuclear shield that protects myonuclei from intrinsic or extrinsic forces. The loss of this scaffold resulted in significantly aberrant nuclear morphology and subsequently reduced levels of essential nuclear factors such as lamin A/C, lamin B, and HP1. Overall, we propose a novel mechanism for maintaining myonuclear morphology and reveal its critical link to correct levels of nuclear factors in differentiated muscle fibers. These findings may shed light on the underlying mechanism of various muscular dystrophies.
Collapse
Affiliation(s)
- Shuoshuo Wang
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Adriana Reuveny
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
40
|
Sallimus and the Dynamics of Sarcomere Assembly in Drosophila Flight Muscles. J Mol Biol 2015; 427:2151-8. [DOI: 10.1016/j.jmb.2015.04.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 04/07/2015] [Accepted: 04/07/2015] [Indexed: 11/19/2022]
|
41
|
Myhre JL, Pilgrim D. A Titan but not necessarily a ruler: assessing the role of titin during thick filament patterning and assembly. Anat Rec (Hoboken) 2015; 297:1604-14. [PMID: 25125174 DOI: 10.1002/ar.22987] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/14/2014] [Indexed: 11/12/2022]
Abstract
The sarcomeres of striated muscle are among the most elaborate and dynamic eukaryotic cellular protein machinery, and the mechanisms by which these semicrystalline filament networks are initially patterned and assembled remain contentious. In addition to the acto-myosin filaments that provide motor function, the sarcomere contains titin filaments, comprised of individual molecules of the giant Ig- and fibronectin domain-rich protein titin. Titin is the largest known protein, containing many structurally distinct domains with a variety of proposed functions, including sarcomere stabilization, the prevention of over-stretching, and returning to resting length after contraction. One molecule of titin, which binds to both the Z-disk and the M-line, spans a half-sarcomere, and is proposed to serve as a "molecular ruler" that dictates the spacing of sarcomeres. The semirigid rod-like A-band region of titin has also been proposed to act as a scaffold for thick filament formation during muscle development, but despite decades of research, this hypothesis has not been rigorously tested. Recent studies in zebrafish have brought into question the necessity for the A-band region of titin during the early stages of sarcomere patterning. In this review, we give an overview of the many different roles of titin in the development and function of striated muscle, and address the validity of the "molecular ruler" model of myofibrillogenesis in light of the current literature.
Collapse
Affiliation(s)
- J Layne Myhre
- Faculty of Dentistry, Department of Oral Health Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
42
|
A cis-regulatory mutation in troponin-I of Drosophila reveals the importance of proper stoichiometry of structural proteins during muscle assembly. Genetics 2015; 200:149-65. [PMID: 25747460 DOI: 10.1534/genetics.115.175604] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 03/01/2015] [Indexed: 02/02/2023] Open
Abstract
Rapid and high wing-beat frequencies achieved during insect flight are powered by the indirect flight muscles, the largest group of muscles present in the thorax. Any anomaly during the assembly and/or structural impairment of the indirect flight muscles gives rise to a flightless phenotype. Multiple mutagenesis screens in Drosophila melanogaster for defective flight behavior have led to the isolation and characterization of mutations that have been instrumental in the identification of many proteins and residues that are important for muscle assembly, function, and disease. In this article, we present a molecular-genetic characterization of a flightless mutation, flightless-H (fliH), originally designated as heldup-a (hdp-a). We show that fliH is a cis-regulatory mutation of the wings up A (wupA) gene, which codes for the troponin-I protein, one of the troponin complex proteins, involved in regulation of muscle contraction. The mutation leads to reduced levels of troponin-I transcript and protein. In addition to this, there is also coordinated reduction in transcript and protein levels of other structural protein isoforms that are part of the troponin complex. The altered transcript and protein stoichiometry ultimately culminates in unregulated acto-myosin interactions and a hypercontraction muscle phenotype. Our results shed new insights into the importance of maintaining the stoichiometry of structural proteins during muscle assembly for proper function with implications for the identification of mutations and disease phenotypes in other species, including humans.
Collapse
|
43
|
Yuan CC, Ma W, Schemmel P, Cheng YS, Liu J, Tsaprailis G, Feldman S, Ayme Southgate A, Irving TC. Elastic proteins in the flight muscle of Manduca sexta. Arch Biochem Biophys 2015; 568:16-27. [PMID: 25602701 PMCID: PMC4684177 DOI: 10.1016/j.abb.2014.12.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 12/29/2014] [Accepted: 12/31/2014] [Indexed: 11/20/2022]
Abstract
The flight muscles (DLM1) of the Hawkmoth, Manduca sexta are synchronous, requiring a neural spike for each contraction. Stress/strain curves of skinned DLM1 showed hysteresis indicating the presence of titin-like elastic proteins. Projectin and kettin are titin-like proteins previously identified in Lethocerus and Drosophila flight muscles. Analysis of Manduca muscles with 1% SDS-agarose gels and western blots showed two bands near 1 MDa that cross-reacted with antibodies to Drosophila projectin. Antibodies to Drosophila kettin cross-reacted to bands at ∼500 and ∼700 kDa, but also to bands at ∼1.6 and ∼2.1 MDa, that had not been previously observed in insect flight muscles. Mass spectrometry identified the 2.1 MDa protein as a product of the Sallimus (sls) gene. Analysis of the gene sequence showed that all 4 putative Sallimus and kettin isoforms could be explained as products of alternative splicing of the single sls gene. Both projectin and sallimus isoforms were expressed to higher levels in ventrally located DLM1 subunits, primarily responsible for active work production, as compared to dorsally located subunits, which may act as damped springs. The different expression levels of the 2 projectin isoforms and 4 sallimus/kettin isoforms may be adaptations to the specific requirements of individual muscle subunits.
Collapse
Affiliation(s)
- Chen-Ching Yuan
- Dept. of Biological and Chemical Sciences, Illinois Institute of Technology, 3101 S. Dearborn St., Chicago, IL 60616, USA
| | - Weikang Ma
- Dept. of Biological and Chemical Sciences, Illinois Institute of Technology, 3101 S. Dearborn St., Chicago, IL 60616, USA
| | - Peter Schemmel
- Dept. of Biological and Chemical Sciences, Illinois Institute of Technology, 3101 S. Dearborn St., Chicago, IL 60616, USA
| | - Yu-Shu Cheng
- Dept. of Biological and Chemical Sciences, Illinois Institute of Technology, 3101 S. Dearborn St., Chicago, IL 60616, USA
| | - Jiangmin Liu
- Dept. of Biological and Chemical Sciences, Illinois Institute of Technology, 3101 S. Dearborn St., Chicago, IL 60616, USA
| | | | - Samuel Feldman
- Dept. of Biology, College of Charleston, Charleston, SC, USA
| | | | - Thomas C Irving
- Dept. of Biological and Chemical Sciences, Illinois Institute of Technology, 3101 S. Dearborn St., Chicago, IL 60616, USA.
| |
Collapse
|
44
|
Pézeron G, Millen K, Boukhatmi H, Bray S. Notch directly regulates the cell morphogenesis genes Reck, talin and trio in adult muscle progenitors. J Cell Sci 2014; 127:4634-44. [PMID: 25217625 DOI: 10.1242/jcs.151787] [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: 01/07/2023] Open
Abstract
There is growing evidence that activation of the Notch pathway can result in consequences on cell morphogenesis and behaviour, both during embryonic development and cancer progression. In general, Notch is proposed to coordinate these processes by regulating expression of key transcription factors. However, many Notch-regulated genes identified in genome-wide studies are involved in fundamental aspects of cell behaviour, suggesting a more direct influence on cellular properties. By testing the functions of 25 such genes we confirmed that 12 are required in developing adult muscles, consistent with roles downstream of Notch. Focusing on three, Reck, rhea/talin and trio, we verify their expression in adult muscle progenitors and identify Notch-regulated enhancers in each. Full activity of these enhancers requires functional binding sites for Su(H), the DNA-binding transcription factor in the Notch pathway, validating their direct regulation. Thus, besides its well-known roles in regulating the expression of cell-fate-determining transcription factors, Notch signalling also has the potential to directly affect cell morphology and behaviour by modulating expression of genes such as Reck, rhea/talin and trio. This sheds new light on the functional outputs of Notch activation in morphogenetic processes.
Collapse
Affiliation(s)
- Guillaume Pézeron
- Department of Physiology Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Kat Millen
- Department of Physiology Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Hadi Boukhatmi
- Department of Physiology Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Sarah Bray
- Department of Physiology Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
45
|
The roles of troponin C isoforms in the mechanical function of Drosophila indirect flight muscle. J Muscle Res Cell Motil 2014; 35:211-23. [PMID: 25134799 DOI: 10.1007/s10974-014-9387-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/29/2014] [Indexed: 10/24/2022]
Abstract
Stretch activation (SA) is a fundamental property of all muscle types that increases power output and efficiency, yet its mechanism is unknown. Recently, studies have implicated troponin isoforms as important in the SA mechanism. The highly stretch-activated Drosophila IFMs express two isoforms of the Ca(2+)-binding subunit of troponin (TnC). TnC1 (TnC-F2 in Lethocerus IFM) has two calcium binding sites, while an unusual isoform, TnC4 (TnC-F1 in Lethocerus IFM), has only one binding site. We investigated the roles of these two TnC isoforms in Drosophila IFM by targeting RNAi to each isoform. IFMs with TnC4 expression (normally ~90% of total TnC) replaced by TnC1 did not generate isometric tension, power or display SA. However, TnC4 knockdown resulted in sarcomere ultrastructure disarray, which could explain the lack of mechanical function and thus make interpretation of the influence of TnC4 on SA difficult. Elimination of TnC1 expression (normally ~10% of total TnC) by RNAi resulted in normal muscle structure. In these IFMs, fiber power generation, isometric tension, stretch-activated force and calcium sensitivity were statistically identical to wild type. When TnC1 RNAi was driven by an IFM specific driver, there was no decrease in flight ability or wing beat frequency, which supports our mechanical findings suggesting that TnC1 is not essential for the mechanical function of Drosophila IFM. This finding contrasts with previous work in Lethocerus IFM showing TnC1 is essential for maximum isometric force generation. We propose that differences in TnC1 function in Lethocerus and Drosophila contribute to the ~40-fold difference in IFM isometric tension generated between these species.
Collapse
|
46
|
Fernandes I, Schöck F. The nebulin repeat protein Lasp regulates I-band architecture and filament spacing in myofibrils. ACTA ACUST UNITED AC 2014; 206:559-72. [PMID: 25113030 PMCID: PMC4137052 DOI: 10.1083/jcb.201401094] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
With just two nebulin repeats, the Drosophila protein Lasp controls muscle thin filament length and filament spacing. Mutations in nebulin, a giant muscle protein with 185 actin-binding nebulin repeats, are the major cause of nemaline myopathy in humans. Nebulin sets actin thin filament length in sarcomeres, potentially by stabilizing thin filaments in the I-band, where nebulin and thin filaments coalign. However, the precise role of nebulin in setting thin filament length and its other functions in regulating power output are unknown. Here, we show that Lasp, the only member of the nebulin family in Drosophila melanogaster, acts at two distinct sites in the sarcomere and controls thin filament length with just two nebulin repeats. We found that Lasp localizes to the Z-disc edges to control I-band architecture and also localizes at the A-band, where it interacts with both actin and myosin to set proper filament spacing. Furthermore, introducing a single amino acid change into the two nebulin repeats of Lasp demonstrated different roles for each domain and established Lasp as a suitable system for studying nebulin repeat function.
Collapse
Affiliation(s)
- Isabelle Fernandes
- Department of Biology, McGill University, Montreal, Quebec, Canada H3A 1B1
| | - Frieder Schöck
- Department of Biology, McGill University, Montreal, Quebec, Canada H3A 1B1
| |
Collapse
|
47
|
Perkins AD, Tanentzapf G. An ongoing role for structural sarcomeric components in maintaining Drosophila melanogaster muscle function and structure. PLoS One 2014; 9:e99362. [PMID: 24915196 PMCID: PMC4051695 DOI: 10.1371/journal.pone.0099362] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 05/14/2014] [Indexed: 11/29/2022] Open
Abstract
Animal muscles must maintain their function while bearing substantial mechanical loads. How muscles withstand persistent mechanical strain is presently not well understood. The basic unit of muscle is the sarcomere, which is primarily composed of cytoskeletal proteins. We hypothesized that cytoskeletal protein turnover is required to maintain muscle function. Using the flight muscles of Drosophila melanogaster, we confirmed that the sarcomeric cytoskeleton undergoes turnover throughout adult life. To uncover which cytoskeletal components are required to maintain adult muscle function, we performed an RNAi-mediated knockdown screen targeting the entire fly cytoskeleton and associated proteins. Gene knockdown was restricted to adult flies and muscle function was analyzed with behavioural assays. Here we analyze the results of that screen and characterize the specific muscle maintenance role for several hits. The screen identified 46 genes required for muscle maintenance: 40 of which had no previously known role in this process. Bioinformatic analysis highlighted the structural sarcomeric proteins as a candidate group for further analysis. Detailed confocal and electron microscopic analysis showed that while muscle architecture was maintained after candidate gene knockdown, sarcomere length was disrupted. Specifically, we found that ongoing synthesis and turnover of the key sarcomere structural components Projectin, Myosin and Actin are required to maintain correct sarcomere length and thin filament length. Our results provide in vivo evidence of adult muscle protein turnover and uncover specific functional defects associated with reduced expression of a subset of cytoskeletal proteins in the adult animal.
Collapse
Affiliation(s)
- Alexander D. Perkins
- Department of Cellular and Physiological Sciences, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
48
|
Koppes RA, Swank DM, Corr DT. A new experimental model to study force depression: the Drosophila jump muscle. J Appl Physiol (1985) 2014; 116:1543-50. [PMID: 24790016 DOI: 10.1152/japplphysiol.01029.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Force depression (FD) is a decrease in isometric force following active muscle shortening. Despite being well characterized experimentally, its underlying mechanism remains unknown. To develop a new, genetically manipulatable experimental model that would greatly improve our ability to study the underlying mechanism(s) of FD, we tested the Drosophila jump muscle for classical FD behavior. Steady-state force generation following active shortening decreased by 2, 8, and 11% of maximum isometric force with increasing shortening amplitudes of 5, 10, and 20% of optimal fiber length, and decreased by 11, 8, and 5% with increasing shortening velocities of 4, 20, and 200% of optimal fiber length per second. These steady-state FD (FDSS) characteristics of Drosophila jump muscle mimic those observed in mammalian skeletal muscle. A double exponential fit of transient force recovery following shortening identified two separate phases of force recovery: a rapid initial force redevelopment, and a slower recovery toward steady state. This analysis showed the slower rate of force redevelopment to be inversely proportional to the amount of FDSS, while the faster rate did not correlate with FDSS. This suggests that the mechanism behind the slower, most likely cross-bridge cycling rate, influences the amount of FDSS. Thus the jump muscle, when coupled with the genetic mutability of its sarcomere proteins, offers a unique and powerful experimental model to explore the underlying mechanism behind FD.
Collapse
Affiliation(s)
- Ryan A Koppes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York; and
| | - Douglas M Swank
- Department of Biology & Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York
| | - David T Corr
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York; and
| |
Collapse
|
49
|
Weitkunat M, Kaya-Çopur A, Grill SW, Schnorrer F. Tension and force-resistant attachment are essential for myofibrillogenesis in Drosophila flight muscle. Curr Biol 2014; 24:705-16. [PMID: 24631244 DOI: 10.1016/j.cub.2014.02.032] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/12/2014] [Accepted: 02/13/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Higher animals generate an elaborate muscle-tendon network to perform their movements. To build a functional network, developing muscles must establish stable connections with tendons and assemble their contractile apparatuses. Current myofibril assembly models do not consider the impact of muscle-tendon attachment on myofibrillogenesis. However, if attachment and myofibrillogenesis are not properly coordinated, premature muscle contractions can destroy an unstable myotendinous system, leading to severe myopathies. RESULTS Here, we use Drosophila indirect flight muscles to investigate how muscle-tendon attachment and myofibrillogenesis are coordinated. We find that flight muscles first stably attach to tendons and then assemble their myofibrils. Interestingly, this myofibril assembly is triggered simultaneously throughout the entire muscle, suggesting a self-assembly mechanism. By applying laser-cutting experiments, we show that muscle attachment coincides with an increase in mechanical tension before periodic myofibrils can be detected. We manipulated tension buildup within the myotendinous system either by genetically compromising attachment initiation and integrin recruitment to the myotendinous junction or by optically severing tendons from muscle. Both treatments cause strong myofibrillogenesis defects. We find that myosin motor activity is required for both tension formation and myofibril assembly, suggesting that myofibril assembly itself contributes to tension buildup. CONCLUSIONS Our results demonstrate that force-resistant attachment enables a stark tension increase in the myotendinous system. Subsequently, this tension increase triggers simultaneous myofibril self-assembly throughout the entire muscle fiber. As myofibril and sarcomeric architecture as well as their molecular components are evolutionarily conserved, we propose a similar tension-based mechanism to regulate myofibrillogenesis in vertebrates.
Collapse
Affiliation(s)
- Manuela Weitkunat
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Aynur Kaya-Çopur
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Stephan W Grill
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187 Dresden, Germany; Max Planck Institute for Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Frank Schnorrer
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| |
Collapse
|
50
|
Weitkunat M, Schnorrer F. A guide to study Drosophila muscle biology. Methods 2014; 68:2-14. [PMID: 24625467 DOI: 10.1016/j.ymeth.2014.02.037] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/25/2014] [Accepted: 02/28/2014] [Indexed: 10/25/2022] Open
Abstract
The development and molecular composition of muscle tissue is evolutionarily conserved. Drosophila is a powerful in vivo model system to investigate muscle morphogenesis and function. Here, we provide a short and comprehensive overview of the important developmental steps to build Drosophila body muscle in embryos, larvae and pupae. We describe key methods, including muscle histology, live imaging and genetics, to study these steps at various developmental stages and include simple behavioural assays to assess muscle function in larvae and adults. We list valuable antibodies and fly strains that can be used for these different methods. This overview should guide the reader to choose the best marker or the appropriate method to obtain high quality muscle morphogenesis data in Drosophila.
Collapse
Affiliation(s)
- Manuela Weitkunat
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Frank Schnorrer
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| |
Collapse
|