1
|
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
|
2
|
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
|
3
|
Ahmed RE, Tokuyama T, Anzai T, Chanthra N, Uosaki H. Sarcomere maturation: function acquisition, molecular mechanism, and interplay with other organelles. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210325. [PMID: 36189811 PMCID: PMC9527934 DOI: 10.1098/rstb.2021.0325] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 06/15/2022] [Indexed: 12/31/2022] Open
Abstract
During postnatal cardiac development, cardiomyocytes mature and turn into adult ones. Hence, all cellular properties, including morphology, structure, physiology and metabolism, are changed. One of the most important aspects is the contractile apparatus, of which the minimum unit is known as a sarcomere. Sarcomere maturation is evident by enhanced sarcomere alignment, ultrastructural organization and myofibrillar isoform switching. Any maturation process failure may result in cardiomyopathy. Sarcomere function is intricately related to other organelles, and the growing evidence suggests reciprocal regulation of sarcomere and mitochondria on their maturation. Herein, we summarize the molecular mechanism that regulates sarcomere maturation and the interplay between sarcomere and other organelles in cardiomyocyte maturation. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Razan E. Ahmed
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Takeshi Tokuyama
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Tatsuya Anzai
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Nawin Chanthra
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
4
|
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
|
5
|
Pierantozzi E, Szentesi P, Paolini C, Dienes B, Fodor J, Oláh T, Colombini B, Rassier DE, Rubino EM, Lange S, Rossi D, Csernoch L, Bagni MA, Reggiani C, Sorrentino V. Impaired Intracellular Ca 2+ Dynamics, M-Band and Sarcomere Fragility in Skeletal Muscles of Obscurin KO Mice. Int J Mol Sci 2022; 23:1319. [PMID: 35163243 PMCID: PMC8835721 DOI: 10.3390/ijms23031319] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Obscurin is a giant sarcomeric protein expressed in striated muscles known to establish several interactions with other proteins of the sarcomere, but also with proteins of the sarcoplasmic reticulum and costameres. Here, we report experiments aiming to better understand the contribution of obscurin to skeletal muscle fibers, starting with a detailed characterization of the diaphragm muscle function, which we previously reported to be the most affected muscle in obscurin (Obscn) KO mice. Twitch and tetanus tension were not significantly different in the diaphragm of WT and Obscn KO mice, while the time to peak (TTP) and half relaxation time (HRT) were prolonged. Differences in force-frequency and force-velocity relationships and an enhanced fatigability are observed in an Obscn KO diaphragm with respect to WT controls. Voltage clamp experiments show that a sarcoplasmic reticulum's Ca2+ release and SERCA reuptake rates were decreased in muscle fibers from Obscn KO mice, suggesting that an impairment in intracellular Ca2+ dynamics could explain the observed differences in the TTP and HRT in the diaphragm. In partial contrast with previous observations, Obscn KO mice show a normal exercise tolerance, but fiber damage, the altered sarcomere ultrastructure and M-band disarray are still observed after intense exercise.
Collapse
Affiliation(s)
- Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Cecilia Paolini
- Department of Neuroscience, Imaging and Clinical Sciences, University Gabriele d’ Annunzio of Chieti, 66100 Chieti, Italy;
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Tamás Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Barbara Colombini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (B.C.); (M.A.B.)
| | - Dilson E. Rassier
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC H2W 1S4, Canada;
| | - Egidio Maria Rubino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - Stephan Lange
- Biomedical Research Facility 2, School of Medicine, University of California, La Jolla, CA 92093, USA;
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Maria Angela Bagni
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (B.C.); (M.A.B.)
| | - Carlo Reggiani
- Department of Biomedical Science, University of Padova, 35121 Padova, Italy;
- Science and Research Center Koper, Institute for Kinesiology Research, 6000 Koper, Slovenia
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| |
Collapse
|
6
|
Development of the indirect flight muscles of Aedes aegypti, a main arbovirus vector. BMC DEVELOPMENTAL BIOLOGY 2021; 21:11. [PMID: 34445959 PMCID: PMC8394598 DOI: 10.1186/s12861-021-00242-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 08/08/2021] [Indexed: 11/22/2022]
Abstract
Background Flying is an essential function for mosquitoes, required for mating and, in the case of females, to get a blood meal and consequently function as a vector. Flight depends on the action of the indirect flight muscles (IFMs), which power the wings beat. No description of the development of IFMs in mosquitoes, including Aedes aegypti, is available.
Methods A. aegypti thoraces of larvae 3 and larvae 4 (L3 and L4) instars were analyzed using histochemistry and bright field microscopy. IFM primordia from L3 and L4 and IFMs from pupal and adult stages were dissected and processed to detect F-actin labelling with phalloidin-rhodamine or TRITC, or to immunodetection of myosin and tubulin using specific antibodies, these samples were analyzed by confocal microscopy. Other samples were studied using transmission electron microscopy. Results At L3–L4, IFM primordia for dorsal-longitudinal muscles (DLM) and dorsal–ventral muscles (DVM) were identified in the expected locations in the thoracic region: three primordia per hemithorax corresponding to DLM with anterior to posterior orientation were present. Other three primordia per hemithorax, corresponding to DVM, had lateral position and dorsal to ventral orientation. During L3 to L4 myoblast fusion led to syncytial myotubes formation, followed by myotendon junctions (MTJ) creation, myofibrils assembly and sarcomere maturation. The formation of Z-discs and M-line during sarcomere maturation was observed in pupal stage and, the structure reached in teneral insects a classical myosin thick, and actin thin filaments arranged in a hexagonal lattice structure. Conclusions A general description of A. aegypti IFM development is presented, from the myoblast fusion at L3 to form myotubes, to sarcomere maturation at adult stage. Several differences during IFM development were observed between A. aegypti (Nematoceran) and Drosophila melanogaster (Brachyceran) and, similitudes with Chironomus sp. were observed as this insect is a Nematoceran, which is taxonomically closer to A. aegypti and share the same number of larval stages. Supplementary Information The online version contains supplementary material available at 10.1186/s12861-021-00242-8.
Collapse
|
7
|
Transcriptomic analysis of the black tiger shrimp (Penaeus monodon) reveals insights into immune development in their early life stages. Sci Rep 2021; 11:13881. [PMID: 34230553 DOI: 10.1038/s41598-021-93364-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
With the rapid growth in the global demand, the shrimp industry needs integrated approaches for sustainable production. A high-quality shrimp larva is one of the crucial key requirements to maximize shrimp production. Survival and growth rates during larval development are often criteria to evaluate larval quality, however many aspects of gene regulation during shrimp larval development have not yet been identified. To further our understanding of biological processes in their early life, transcriptomic analysis of larval developmental stages (nauplius, zoea, mysis, and postlarva) were determined in the black tiger shrimp, Penaeus monodon using next-generation RNA sequencing. Gene clustering and gene enrichment analyses revealed that most of the transcripts were mainly related to metabolic processes, cell and growth development, and immune system. Interestingly, Spätzle and Toll receptors were found in nauplius stage, providing evidence that Toll pathway was a baseline immune system established in early larval stages. Genes encoding pathogen pattern-recognition proteins (LGBP, PL5-2 and c-type lectin), prophenoloxidase system (PPAE2, PPAF2 and serpin), antimicrobial peptides (crustin and antiviral protein), blood clotting system (hemolymph clottable protein) and heat shock protein (HSP70) were expressed as they developed further, suggesting that these immune defense mechanisms were established in later larval stages.
Collapse
|
8
|
Shenkman BS, Tsaturyan AK, Vikhlyantsev IM, Kozlovskaya IB, Grigoriev AI. Molecular Mechanisms of Muscle Tone Impairment under Conditions of Real and Simulated Space Flight. Acta Naturae 2021; 13:85-97. [PMID: 34377559 PMCID: PMC8327152 DOI: 10.32607/actanaturae.10953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/04/2020] [Indexed: 01/08/2023] Open
Abstract
Kozlovskaya et al. [1] and Grigoriev et al. [2] showed that enormous loss of muscle stiffness (atonia) develops in humans under true (space flight) and simulated microgravity conditions as early as after the first days of exposure. This phenomenon is attributed to the inactivation of slow motor units and called reflectory atonia. However, a lot of evidence indicating that even isolated muscle or a single fiber possesses substantial stiffness was published at the end of the 20th century. This intrinsic stiffness is determined by the active component, i.e. the ability to form actin-myosin cross-bridges during muscle stretch and contraction, as well as by cytoskeletal and extracellular matrix proteins, capable of resisting muscle stretch. The main facts on intrinsic muscle stiffness under conditions of gravitational unloading are considered in this review. The data obtained in studies of humans under dry immersion and rodent hindlimb suspension is analyzed. The results and hypotheses regarding reduced probability of cross-bridge formation in an atrophying muscle due to increased interfilament spacing are described. The evidence of cytoskeletal protein (titin, nebulin, etc.) degradation during gravitational unloading is also discussed. The possible mechanisms underlying structural changes in skeletal muscle collagen and its role in reducing intrinsic muscle stiffness are presented. The molecular mechanisms of changes in intrinsic stiffness during space flight and simulated microgravity are reviewed.
Collapse
Affiliation(s)
- B. S. Shenkman
- State Scientific Center of Russian Federation – Institute of Biomedical Problems, Moscow, 123007 Russia
| | - A. K. Tsaturyan
- Lomonosov Moscow State University Research Institute of Mechanics, Moscow, 119192 Russia
| | - I. M. Vikhlyantsev
- Institute of Experimental and Theoretical Biophysics, Moscow Region, Pushchino, 142290 Russia
| | - I. B. Kozlovskaya
- State Scientific Center of Russian Federation – Institute of Biomedical Problems, Moscow, 123007 Russia
| | - A. I. Grigoriev
- State Scientific Center of Russian Federation – Institute of Biomedical Problems, Moscow, 123007 Russia
| |
Collapse
|
9
|
Czajkowski ER, Cisneros M, Garcia BS, Shen J, Cripps RM. The Drosophila CG1674 gene encodes a synaptopodin 2-like related protein that localizes to the Z-disc and is required for normal flight muscle development and function. Dev Dyn 2021; 250:99-110. [PMID: 32893414 PMCID: PMC7902442 DOI: 10.1002/dvdy.250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/12/2020] [Accepted: 09/01/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND To identify novel myofibrillar components of the Drosophila flight muscles, we carried out a proteomic analysis of chemically demembranated flight muscle myofibrils, and characterized the knockdown phenotype of a novel gene identified in the screen, CG1674. RESULTS The CG1674 protein has some similarity to vertebrate synaptopodin 2-like, and when expressed as a FLAG-tagged fusion protein, it was localized during development to the Z-disc and cytoplasm. Knockdown of CG1674 expression affected the function of multiple muscle types, and defective flight in adults was accompanied by large actin-rich structures in the flight muscles that resembled overgrown Z-discs. Localization of CG1674 to the Z-disc depended predominantly upon presence of the Z-disc component alpha-actinin, but also depended upon other Z-disc components, including Mask, Zasp52, and Sals. We also observed re-localization of FLAG-CG1674 to the nucleus in Alpha-actinin and sals knockdown animals. CONCLUSIONS These studies identify and characterize a previously unreported myofibrillar component of Drosophila muscle that is necessary for proper myofibril assembly during development.
Collapse
Affiliation(s)
| | - Marilyn Cisneros
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Bianca S. Garcia
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Jim Shen
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Richard M. Cripps
- Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
10
|
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
|
11
|
Daneshparvar N, Taylor DW, O'Leary TS, Rahmani H, Abbasiyeganeh F, Previs MJ, Taylor KA. CryoEM structure of Drosophila flight muscle thick filaments at 7 Å resolution. Life Sci Alliance 2020; 3:3/8/e202000823. [PMID: 32718994 PMCID: PMC7391215 DOI: 10.26508/lsa.202000823] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 11/24/2022] Open
Abstract
Striated muscle thick filaments are composed of myosin II and several non-myosin proteins. Myosin II's long α-helical coiled-coil tail forms the dense protein backbone of filaments, whereas its N-terminal globular head containing the catalytic and actin-binding activities extends outward from the backbone. Here, we report the structure of thick filaments of the flight muscle of the fruit fly Drosophila melanogaster at 7 Å resolution. Its myosin tails are arranged in curved molecular crystalline layers identical to flight muscles of the giant water bug Lethocerus indicus Four non-myosin densities are observed, three of which correspond to ones found in Lethocerus; one new density, possibly stretchin-mlck, is found on the backbone outer surface. Surprisingly, the myosin heads are disordered rather than ordered along the filament backbone. Our results show striking myosin tail similarity within flight muscle filaments of two insect orders separated by several hundred million years of evolution.
Collapse
Affiliation(s)
- Nadia Daneshparvar
- Department of Physics, Florida State University, Tallahassee, FL, USA.,Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, USA
| | - Dianne W Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, USA
| | - Thomas S O'Leary
- Department of Molecular Physiology & Biophysics, University of Vermont College of Medicine, Burlington, VT, USA
| | - Hamidreza Rahmani
- Department of Physics, Florida State University, Tallahassee, FL, USA.,Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, USA
| | | | - Michael J Previs
- Department of Molecular Physiology & Biophysics, University of Vermont College of Medicine, Burlington, VT, USA
| | - Kenneth A Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
González-Morales N, Schöck F. Commentary: Nanoscopy reveals the layered organization of the sarcomeric H-zone and I-band complexes. Front Cell Dev Biol 2020; 8:74. [PMID: 32117994 PMCID: PMC7031406 DOI: 10.3389/fcell.2020.00074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/28/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Frieder Schöck
- Department of Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
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
|
16
|
Subramaniam J, Yang P, McCarthy MJ, Cunha SR. Identification and characterization of self-association domains on small ankyrin 1 isoforms. J Mol Cell Cardiol 2020; 139:225-237. [PMID: 32035138 PMCID: PMC11042479 DOI: 10.1016/j.yjmcc.2020.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 01/13/2020] [Accepted: 02/03/2020] [Indexed: 10/25/2022]
Abstract
In striated muscles, the large scaffolding protein obscurin and a small SR-integral membrane protein sAnk1.5 control the retention of longitudinal SR across the sarcomere. How a complex of these proteins facilitates localization of longitudinal SR has yet to be resolved, but we hypothesize that obscurin interacts with a complex of sAnk1.5 proteins. To begin to address this hypothesis, we demonstrate that sAnk1.5 interacts with itself and identify two domains mediating self-association. Specifically, we show by co-precipitation and FLIM-FRET analysis that sAnk1.5 and another small AnkR isoform (sAnk1.6) interact with themselves and each other. We demonstrate that obscurin interacts with a complex of sAnk1.5 proteins and that this complex formation is enhanced by obscurin-binding. Using FLIM-FRET analysis, we show that obscurin interacts with sAnk1.5 alone and with sAnk1.6 in the presence of sAnk1.5. We find that sAnk1.5 self-association is disrupted by mutagenesis of residues Arg64-Arg69, residues previously associated with obscurin-binding. Molecular modeling of two interacting sAnk1.5 monomers facilitated the identification of Gly31-Val36 as an additional site of interaction, which was subsequently corroborated by co-precipitation and FLIM-FRET analysis. In closing, these results support a model in which sAnk1.5 forms large oligomers that interact with obscurin to facilitate the retention of longitudinal SR throughout skeletal and cardiac myocytes.
Collapse
Affiliation(s)
- Janani Subramaniam
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America
| | - Pu Yang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America
| | - Michael J McCarthy
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America
| | - Shane R Cunha
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America.
| |
Collapse
|
17
|
González-Morales N, Marsh TW, Katzemich A, Marescal O, Xiao YS, Schöck F. Different Evolutionary Trajectories of Two Insect-Specific Paralogous Proteins Involved in Stabilizing Muscle Myofibrils. Genetics 2019; 212:743-755. [PMID: 31123042 PMCID: PMC6614898 DOI: 10.1534/genetics.119.302217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/07/2019] [Indexed: 12/17/2022] Open
Abstract
Alp/Enigma family members have a unique PDZ domain followed by zero to four LIM domains, and are essential for myofibril assembly across all species analyzed so far. Drosophila melanogaster has three Alp/Enigma family members, Zasp52, Zasp66, and Zasp67. Ortholog search and phylogenetic tree analysis suggest that Zasp genes have a common ancestor, and that Zasp66 and Zasp67 arose by duplication in insects. While Zasp66 has a conserved domain structure across orthologs, Zasp67 domains and lengths are highly variable. In flies, Zasp67 appears to be expressed only in indirect flight muscles, where it colocalizes with Zasp52 at Z-discs. We generated a CRISPR null mutant of Zasp67, which is viable but flightless. We can rescue all phenotypes by re-expressing a Zasp67 transgene at endogenous levels. Zasp67 mutants show extended and broken Z-discs in adult flies, indicating that the protein helps stabilize the highly regular myofibrils of indirect flight muscles. In contrast, a Zasp66 CRISPR null mutant has limited viability, but only mild indirect flight muscle defects illustrating the diverging evolutionary paths these two paralogous genes have taken since they arose by duplication.
Collapse
Affiliation(s)
| | - Thomas W Marsh
- Department of Biology, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Anja Katzemich
- Department of Biology, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Océane Marescal
- Department of Biology, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Yu Shu Xiao
- Department of Biology, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Frieder Schöck
- Department of Biology, McGill University, Montreal, Quebec H3A 1B1, Canada
| |
Collapse
|
18
|
Blondelle J, Marrocco V, Clark M, Desmond P, Myers S, Nguyen J, Wright M, Bremner S, Pierantozzi E, Ward S, Estève E, Sorrentino V, Ghassemian M, Lange S. Murine obscurin and Obsl1 have functionally redundant roles in sarcolemmal integrity, sarcoplasmic reticulum organization, and muscle metabolism. Commun Biol 2019; 2:178. [PMID: 31098411 PMCID: PMC6509138 DOI: 10.1038/s42003-019-0405-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/28/2019] [Indexed: 12/19/2022] Open
Abstract
Biological roles of obscurin and its close homolog Obsl1 (obscurin-like 1) have been enigmatic. While obscurin is highly expressed in striated muscles, Obsl1 is found ubiquitously. Accordingly, obscurin mutations have been linked to myopathies, whereas mutations in Obsl1 result in 3M-growth syndrome. To further study unique and redundant functions of these closely related proteins, we generated and characterized Obsl1 knockouts. Global Obsl1 knockouts are embryonically lethal. In contrast, skeletal muscle-specific Obsl1 knockouts show a benign phenotype similar to obscurin knockouts. Only deletion of both proteins and removal of their functional redundancy revealed their roles for sarcolemmal stability and sarcoplasmic reticulum organization. To gain unbiased insights into changes to the muscle proteome, we analyzed tibialis anterior and soleus muscles by mass spectrometry, uncovering additional changes to the muscle metabolism. Our analyses suggest that all obscurin protein family members play functions for muscle membrane systems.
Collapse
Affiliation(s)
- Jordan Blondelle
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Valeria Marrocco
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Madison Clark
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Patrick Desmond
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Stephanie Myers
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Jim Nguyen
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Matthew Wright
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Shannon Bremner
- Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100 Italy
| | - Samuel Ward
- Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, 92093 CA USA
| | - Eric Estève
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
- Université Grenoble Alpes, HP2, Grenoble, 38706 France
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, 53100 Italy
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry, University of California, San Diego, 92093 CA USA
| | - Stephan Lange
- Division of Cardiology, School of Medicine, University of California, San Diego, 92093 CA USA
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, 413 45 Sweden
| |
Collapse
|
19
|
Dasbiswas K, Hu S, Schnorrer F, Safran SA, Bershadsky AD. Ordering of myosin II filaments driven by mechanical forces: experiments and theory. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0114. [PMID: 29632266 DOI: 10.1098/rstb.2017.0114] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2017] [Indexed: 12/27/2022] Open
Abstract
Myosin II filaments form ordered superstructures in both cross-striated muscle and non-muscle cells. In cross-striated muscle, myosin II (thick) filaments, actin (thin) filaments and elastic titin filaments comprise the stereotypical contractile units of muscles called sarcomeres. Linear chains of sarcomeres, called myofibrils, are aligned laterally in registry to form cross-striated muscle cells. The experimentally observed dependence of the registered organization of myofibrils on extracellular matrix elasticity has been proposed to arise from the interactions of sarcomeric contractile elements (considered as force dipoles) through the matrix. Non-muscle cells form small bipolar filaments built of less than 30 myosin II molecules. These filaments are associated in registry forming superstructures ('stacks') orthogonal to actin filament bundles. Formation of myosin II filament stacks requires the myosin II ATPase activity and function of the actin filament crosslinking, polymerizing and depolymerizing proteins. We propose that the myosin II filaments embedded into elastic, intervening actin network (IVN) function as force dipoles that interact attractively through the IVN. This is in analogy with the theoretical picture developed for myofibrils where the elastic medium is now the actin cytoskeleton itself. Myosin stack formation in non-muscle cells provides a novel mechanism for the self-organization of the actin cytoskeleton at the level of the entire cell.This article is part of the theme issue 'Self-organization in cell biology'.
Collapse
Affiliation(s)
- Kinjal Dasbiswas
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA
| | - Shiqiong Hu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Samuel A Safran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore .,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
20
|
Lynch JM, Dolman AJ, Guo C, Dolan K, Xiang C, Reda S, Li B, Prasanna G. Mutant myocilin impacts sarcomere ultrastructure in mouse gastrocnemius muscle. PLoS One 2018; 13:e0206801. [PMID: 30395621 PMCID: PMC6218065 DOI: 10.1371/journal.pone.0206801] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 10/19/2018] [Indexed: 11/19/2022] Open
Abstract
Myocilin (MYOC) is the gene with mutations most common in glaucoma. In the eye, MYOC is in trabecular meshwork, ciliary body, and retina. Other tissues with high MYOC transcript levels are skeletal muscle and heart. To date, the function of wild-type MYOC remains unknown and how mutant MYOC causes high intraocular pressure and glaucoma is ambiguous. By investigating mutant MYOC in a non-ocular tissue we hoped to obtain novel insight into mutant MYOC pathology. For this study, we utilized a transgenic mouse expressing human mutant MYOC Y437H protein and we examined its skeletal (gastrocnemius) muscle phenotype. Electron micrographs showed that sarcomeres in the skeletal muscle of mutant CMV-MYOC-Y437H mice had multiple M-bands. Western blots of soluble muscle lysates from transgenics indicated a decrease in two M-band proteins, myomesin 1 (MYOM1) and muscle creatine kinase (CKM). Immunoprecipitation identified CKM as a MYOC binding partner. Our results suggest that binding of mutant MYOC to CKM is changing sarcomere ultrastructure and this may adversely impact muscle function. We speculate that a person carrying the mutant MYOC mutation will likely have a glaucoma phenotype and may also have undiagnosed muscle ailments or vice versa, both of which will have to be monitored and treated.
Collapse
MESH Headings
- Animals
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Disease Models, Animal
- Eye Proteins/genetics
- Eye Proteins/metabolism
- Female
- Gene Expression
- Glaucoma, Open-Angle/genetics
- Glaucoma, Open-Angle/metabolism
- Glaucoma, Open-Angle/pathology
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Humans
- Intraocular Pressure/genetics
- Male
- Mice
- Mice, Mutant Strains
- Mice, Transgenic
- Microscopy, Electron, Transmission
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- Mutant Proteins/genetics
- Mutant Proteins/metabolism
- Mutation
- Myocardium/metabolism
- Phenotype
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- Sarcomeres/genetics
- Sarcomeres/metabolism
- Sarcomeres/ultrastructure
- Trabecular Meshwork/metabolism
- Trabecular Meshwork/ultrastructure
Collapse
Affiliation(s)
- Jeffrey M. Lynch
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
- * E-mail:
| | - Andrew J. Dolman
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Chenying Guo
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Katie Dolan
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Chuanxi Xiang
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Samir Reda
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Bing Li
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| | - Ganesh Prasanna
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States of America
| |
Collapse
|
21
|
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
|
22
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
23
|
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
|
24
|
Manring HR, Carter OA, Ackermann MA. Obscure functions: the location-function relationship of obscurins. Biophys Rev 2017; 9:245-258. [PMID: 28510116 DOI: 10.1007/s12551-017-0254-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/05/2017] [Indexed: 12/18/2022] Open
Abstract
The obscurin family of polypeptides is essential for normal striated muscle function and contributes to the pathogenesis of fatal diseases, including cardiomyopathies and cancers. The single mammalian obscurin gene, OBSCN, gives rise to giant (∼800 kDa) and smaller (∼40-500 kDa) proteins that are composed of tandem adhesion and signaling motifs. Mammalian obscurin proteins are expressed in a variety of cell types, including striated muscles, and localize to distinct subcellular compartments where they contribute to diverse cellular processes. Obscurin homologs in Caenorhabditis elegans and Drosophila possess a similar domain architecture and are also expressed in striated muscles. The long sought after question, "what does obscurin do?" is complex and cannot be addressed without taking into consideration the subcellular distribution of these proteins and local isoform concentration. Herein, we present an overview of the functions of obscurins and begin to define the intricate relationship between their subcellular distributions and functions in striated muscles.
Collapse
Affiliation(s)
- Heather R Manring
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA
| | - Olivia A Carter
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA
| | - Maegen A Ackermann
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA. .,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
25
|
Leclère L, Röttinger E. Diversity of Cnidarian Muscles: Function, Anatomy, Development and Regeneration. Front Cell Dev Biol 2017; 4:157. [PMID: 28168188 PMCID: PMC5253434 DOI: 10.3389/fcell.2016.00157] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/30/2016] [Indexed: 12/12/2022] Open
Abstract
The ability to perform muscle contractions is one of the most important and distinctive features of eumetazoans. As the sister group to bilaterians, cnidarians (sea anemones, corals, jellyfish, and hydroids) hold an informative phylogenetic position for understanding muscle evolution. Here, we review current knowledge on muscle function, diversity, development, regeneration and evolution in cnidarians. Cnidarian muscles are involved in various activities, such as feeding, escape, locomotion and defense, in close association with the nervous system. This variety is reflected in the large diversity of muscle organizations found in Cnidaria. Smooth epithelial muscle is thought to be the most common type, and is inferred to be the ancestral muscle type for Cnidaria, while striated muscle fibers and non-epithelial myocytes would have been convergently acquired within Cnidaria. Current knowledge of cnidarian muscle development and its regeneration is limited. While orthologs of myogenic regulatory factors such as MyoD have yet to be found in cnidarian genomes, striated muscle formation potentially involves well-conserved myogenic genes, such as twist and mef2. Although satellite cells have yet to be identified in cnidarians, muscle plasticity (e.g., de- and re-differentiation, fiber repolarization) in a regenerative context and its potential role during regeneration has started to be addressed in a few cnidarian systems. The development of novel tools to study those organisms has created new opportunities to investigate in depth the development and regeneration of cnidarian muscle cells and how they contribute to the regenerative process.
Collapse
Affiliation(s)
- Lucas Leclère
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV) Villefranche-sur-mer, France
| | - Eric Röttinger
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging (IRCAN) Nice, France
| |
Collapse
|
26
|
Xiao YS, Schöck F, González-Morales N. Rapid IFM Dissection for Visualizing Fluorescently Tagged Sarcomeric Proteins. Bio Protoc 2017; 7:e2606. [PMID: 29423427 DOI: 10.21769/bioprotoc.2606] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Sarcomeres, the smallest contractile unit of muscles, are arguably the most impressive actomyosin structure. Yet a complete understanding of sarcomere formation and maintenance is missing. The Drosophila indirect flight muscle (IFM) has proven to be a very valuable model to study sarcomeres. Here, we present a protocol for the rapid dissection of IFM and analysis of sarcomeres using fluorescently tagged proteins.
Collapse
Affiliation(s)
- Yu Shu Xiao
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Frieder Schöck
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
27
|
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
|
28
|
Pernigo S, Fukuzawa A, Beedle AEM, Holt M, Round A, Pandini A, Garcia-Manyes S, Gautel M, Steiner RA. Binding of Myomesin to Obscurin-Like-1 at the Muscle M-Band Provides a Strategy for Isoform-Specific Mechanical Protection. Structure 2016; 25:107-120. [PMID: 27989621 PMCID: PMC5222588 DOI: 10.1016/j.str.2016.11.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/16/2016] [Accepted: 11/18/2016] [Indexed: 12/03/2022]
Abstract
The sarcomeric cytoskeleton is a network of modular proteins that integrate mechanical and signaling roles. Obscurin, or its homolog obscurin-like-1, bridges the giant ruler titin and the myosin crosslinker myomesin at the M-band. Yet, the molecular mechanisms underlying the physical obscurin(-like-1):myomesin connection, important for mechanical integrity of the M-band, remained elusive. Here, using a combination of structural, cellular, and single-molecule force spectroscopy techniques, we decode the architectural and functional determinants defining the obscurin(-like-1):myomesin complex. The crystal structure reveals a trans-complementation mechanism whereby an incomplete immunoglobulin-like domain assimilates an isoform-specific myomesin interdomain sequence. Crucially, this unconventional architecture provides mechanical stability up to forces of ∼135 pN. A cellular competition assay in neonatal rat cardiomyocytes validates the complex and provides the rationale for the isoform specificity of the interaction. Altogether, our results reveal a novel binding strategy in sarcomere assembly, which might have implications on muscle nanomechanics and overall M-band organization. The structure of the human obscurin-like-1:myomesin complex has been determined A myomesin sequence complements an immunoglobulin fold of obscurin-like-1 This binding mechanism provides mechanical stability up to forces of ∼135 pN Possible implications on muscle nanomechanics and M-band organization are discussed
Collapse
Affiliation(s)
- Stefano Pernigo
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Atsushi Fukuzawa
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Cardiovascular Division, King's College London BHF Centre of Research Excellence, London SE1 1UL, UK
| | - Amy E M Beedle
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Department of Physics, King's College London, London WC2R 2LS, UK
| | - Mark Holt
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Cardiovascular Division, King's College London BHF Centre of Research Excellence, London SE1 1UL, UK
| | - Adam Round
- European Molecular Biology Laboratory, Grenoble Outstation, 38042 Grenoble, France; School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire, UK
| | - Alessandro Pandini
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Department of Computer Science and Synthetic Biology Theme, Brunel University London, London UB8 3PH, UK
| | - Sergi Garcia-Manyes
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Department of Physics, King's College London, London WC2R 2LS, UK.
| | - Mathias Gautel
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Cardiovascular Division, King's College London BHF Centre of Research Excellence, London SE1 1UL, UK.
| | - Roberto A Steiner
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK.
| |
Collapse
|
29
|
Randazzo D, Blaauw B, Paolini C, Pierantozzi E, Spinozzi S, Lange S, Chen J, Protasi F, Reggiani C, Sorrentino V. Exercise-induced alterations and loss of sarcomeric M-line organization in the diaphragm muscle of obscurin knockout mice. Am J Physiol Cell Physiol 2016; 312:C16-C28. [PMID: 27784675 DOI: 10.1152/ajpcell.00098.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/25/2016] [Indexed: 11/22/2022]
Abstract
We recently reported that skeletal muscle fibers of obscurin knockout (KO) mice present altered distribution of ankyrin B (ankB), disorganization of the subsarcolemmal microtubules, and reduced localization of dystrophin at costameres. In addition, these mice have impaired running endurance and increased exercise-induced sarcolemmal damage compared with wild-type animals. Here, we report results from a combined approach of physiological, morphological, and structural studies in which we further characterize the skeletal muscles of obscurin KO mice. A detailed examination of exercise performance, using different running protocols, revealed that the reduced endurance of obscurin KO animals on the treadmill depends on exercise intensity and age. Indeed, a mild running protocol did not evidence significant differences between control and obscurin KO mice, whereas comparison of running abilities of 2-, 6-, and 11-mo-old mice exercised at exhaustion revealed a progressive age-dependent reduction of the exercise tolerance in KO mice. Histological analysis indicated that heavy exercise induced leukocyte infiltration, fibrotic connective tissue deposition, and hypercontractures in the diaphragm of KO mice. On the same line, electron microscopy revealed that, in the diaphragm of exercised obscurin KO mice, but not in the hindlimb muscles, both M-line and H-zone of sarcomeres appeared wavy and less defined. Altogether, these results suggest that obscurin is required for the maintenance of morphological and ultrastructural integrity of skeletal muscle fibers against damage induced by intense mechanical stress and point to the diaphragm as the skeletal muscle most severely affected in obscurin-deficient mice.
Collapse
Affiliation(s)
- D Randazzo
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - B Blaauw
- Venetian Institute of Molecular Medicine, Padua, Italy.,Interuniversity Institute of Myology
| | - C Paolini
- Center for Research on Ageing and Translational Medicine & Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti, Chieti, Italy.,Interuniversity Institute of Myology
| | - E Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - S Spinozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - S Lange
- University of California, San Diego School of Medicine, San Diego, California
| | - J Chen
- University of California, San Diego School of Medicine, San Diego, California
| | - F Protasi
- Center for Research on Ageing and Translational Medicine & Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti, Chieti, Italy.,Interuniversity Institute of Myology
| | - C Reggiani
- Department of Biomedical Sciences, University of Padua, Padua, Italy; and.,Interuniversity Institute of Myology
| | - V Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy; .,Interuniversity Institute of Myology
| |
Collapse
|
30
|
Liao KA, González-Morales N, Schöck F. Zasp52, a Core Z-disc Protein in Drosophila Indirect Flight Muscles, Interacts with α-Actinin via an Extended PDZ Domain. PLoS Genet 2016; 12:e1006400. [PMID: 27783625 PMCID: PMC5081203 DOI: 10.1371/journal.pgen.1006400] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/04/2016] [Indexed: 11/18/2022] Open
Abstract
Z-discs are organizing centers that establish and maintain myofibril structure and function. Important Z-disc proteins are α-actinin, which cross-links actin thin filaments at the Z-disc and Zasp PDZ domain proteins, which directly interact with α-actinin. Here we investigate the biochemical and genetic nature of this interaction in more detail. Zasp52 is the major Drosophila Zasp PDZ domain protein, and is required for myofibril assembly and maintenance. We show by in vitro biochemistry that the PDZ domain plus a C-terminal extension is the only area of Zasp52 involved in the interaction with α-actinin. In addition, site-directed mutagenesis of 5 amino acid residues in the N-terminal part of the PDZ domain, within the PWGFRL motif, abolish binding to α-actinin, demonstrating the importance of this motif for α-actinin binding. Rescue assays of a novel Zasp52 allele demonstrate the crucial importance of the PDZ domain for Zasp52 function. Flight assays also show that a Zasp52 mutant suppresses the α-actinin mutant phenotype, indicating that both proteins are core structural Z-disc proteins required for optimal Z-disc function. Although Zasp PDZ domain proteins are known to bind α-actinin and play a role in muscle assembly and maintenance, the details and importance of this interaction have not been assessed. Here we demonstrate that a conserved motif in the N-terminal part of the Zasp52 PDZ domain is responsible for α-actinin binding and that a C-terminal extension of the PDZ domain is required for optimal α-actinin binding. We show using transgenic animals that in the absence of the PDZ domain no aspect of myofibril assembly can be rescued. Intriguingly, α-actinin/+ heterozygous animals show irregularities in wing beat frequency, which can be suppressed by removing one copy of Zasp52. This suggests that both proteins are required at fixed levels at the Z-disc to support optimal functionality.
Collapse
Affiliation(s)
- Kuo An Liao
- Department of Biology, McGill University, 1205 Dr Penfield Avenue, Montreal, Quebec, CANADA
| | | | - Frieder Schöck
- Department of Biology, McGill University, 1205 Dr Penfield Avenue, Montreal, Quebec, CANADA
- * E-mail:
| |
Collapse
|
31
|
Sarov M, Barz C, Jambor H, Hein MY, Schmied C, Suchold D, Stender B, Janosch S, K J VV, Krishnan RT, Krishnamoorthy A, Ferreira IRS, Ejsmont RK, Finkl K, Hasse S, Kämpfer P, Plewka N, Vinis E, Schloissnig S, Knust E, Hartenstein V, Mann M, Ramaswami M, VijayRaghavan K, Tomancak P, Schnorrer F. A genome-wide resource for the analysis of protein localisation in Drosophila. eLife 2016; 5:e12068. [PMID: 26896675 PMCID: PMC4805545 DOI: 10.7554/elife.12068] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 02/19/2016] [Indexed: 02/07/2023] Open
Abstract
The Drosophila genome contains >13000 protein-coding genes, the majority of which remain poorly investigated. Important reasons include the lack of antibodies or reporter constructs to visualise these proteins. Here, we present a genome-wide fosmid library of 10000 GFP-tagged clones, comprising tagged genes and most of their regulatory information. For 880 tagged proteins, we created transgenic lines, and for a total of 207 lines, we assessed protein expression and localisation in ovaries, embryos, pupae or adults by stainings and live imaging approaches. Importantly, we visualised many proteins at endogenous expression levels and found a large fraction of them localising to subcellular compartments. By applying genetic complementation tests, we estimate that about two-thirds of the tagged proteins are functional. Moreover, these tagged proteins enable interaction proteomics from developing pupae and adult flies. Taken together, this resource will boost systematic analysis of protein expression and localisation in various cellular and developmental contexts. DOI:http://dx.doi.org/10.7554/eLife.12068.001 The fruit fly Drosophila melanogaster is a popular model organism in biological research. Studies using Drosophila have led to important insights into human biology, because related proteins often fulfil similar roles in flies and humans. Thus, studying the role of a protein in Drosophila can teach us about what it might do in a human. To fulfil their biological roles, proteins often occupy particular locations inside cells, such as the cell’s nucleus or surface membrane. Many proteins are also only found in specific types of cell, such as neurons or muscle cells. A protein’s location thus provides clues about what it does, however cells contain many thousands of proteins and identifying the location of each one is a herculean task. Sarov et al. took on this challenge and developed a new resource to study the localisation of all Drosophila proteins during this animal’s development. First, genetic engineering was used to tag thousands of Drosophila proteins with a green fluorescent protein, so that they could be tracked under a microscope. Sarov et al. tagged about 10000 Drosophila proteins in bacteria, and then introduced almost 900 of them into flies to create genetically modified flies. Each fly line contains an extra copy of the tagged gene that codes for one tagged protein. About two-thirds of these tagged proteins appeared to work normally after they were introduced into flies. Sarov et al. then looked at over 200 of these fly lines in more detail and observed that many of the proteins were found in particular cell types and localized to specific parts of the cells. Video imaging of the tagged proteins in living fruit fly embryos and pupae revealed the proteins’ movements, while other techniques showed which proteins bind to the tagged proteins, and may therefore work together in protein complexes. This resource is openly available to the community, and so researchers can use it to study their favourite protein and gain new insights into how proteins work and are regulated during Drosophila development. Following on from this work, the next challenge will be to create more flies carrying tagged proteins, and to swap the green fluorescent tag with other experimentally useful tags. DOI:http://dx.doi.org/10.7554/eLife.12068.002
Collapse
Affiliation(s)
- Mihail Sarov
- Max Planck Institute of Cell Biology and Genetics, Dresden, Germany
| | - Christiane Barz
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Helena Jambor
- Max Planck Institute of Cell Biology and Genetics, Dresden, Germany
| | - Marco Y Hein
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Dana Suchold
- Max Planck Institute of Cell Biology and Genetics, Dresden, Germany
| | - Bettina Stender
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Stephan Janosch
- Max Planck Institute of Cell Biology and Genetics, Dresden, Germany
| | - Vinay Vikas K J
- Centre for Cellular and Molecular Platforms, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - R T Krishnan
- Centre for Cellular and Molecular Platforms, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Aishwarya Krishnamoorthy
- Centre for Cellular and Molecular Platforms, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Irene R S Ferreira
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Katja Finkl
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Susanne Hasse
- Max Planck Institute of Cell Biology and Genetics, Dresden, Germany
| | - Philipp Kämpfer
- Heidelberg Institute of Theoretical Studies, Heidelberg, Germany
| | - Nicole Plewka
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Elisabeth Vinis
- Max Planck Institute of Cell Biology and Genetics, Dresden, Germany
| | | | - Elisabeth Knust
- Max Planck Institute of Cell Biology and Genetics, Dresden, Germany
| | - Volker Hartenstein
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Mani Ramaswami
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - K VijayRaghavan
- Centre for Cellular and Molecular Platforms, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Pavel Tomancak
- Max Planck Institute of Cell Biology and Genetics, Dresden, Germany
| | - Frank Schnorrer
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
32
|
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
|
33
|
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]
|
34
|
Benian GM, Mayans O. Titin and obscurin: giants holding hands and discovery of a new Ig domain subset. J Mol Biol 2014; 427:707-714. [PMID: 25555989 DOI: 10.1016/j.jmb.2014.12.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Guy M Benian
- Department of Pathology, Emory University, Atlanta, GA 30322, USA.
| | - Olga Mayans
- Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK.
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Garcia-Guerra L, Vila-Bedmar R, Carrasco-Rando M, Cruces-Sande M, Martín M, Ruiz-Gómez A, Ruiz-Gómez M, Lorenzo M, Fernández-Veledo S, Mayor F, Murga C, Nieto-Vázquez I. Skeletal muscle myogenesis is regulated by G protein-coupled receptor kinase 2. J Mol Cell Biol 2014; 6:299-311. [PMID: 24927997 DOI: 10.1093/jmcb/mju025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
G protein-coupled receptor kinase 2 (GRK2) is an important serine/threonine-kinase regulating different membrane receptors and intracellular proteins. Attenuation of Drosophila Gprk2 in embryos or adult flies induced a defective differentiation of somatic muscles, loss of fibers, and a flightless phenotype. In vertebrates, GRK2 hemizygous mice contained less but more hypertrophied skeletal muscle fibers than wild-type littermates. In C2C12 myoblasts, overexpression of a GRK2 kinase-deficient mutant (K220R) caused precocious differentiation of cells into immature myotubes, which were wider in size and contained more fused nuclei, while GRK2 overexpression blunted differentiation. Moreover, p38MAPK and Akt pathways were activated at an earlier stage and to a greater extent in K220R-expressing cells or upon kinase downregulation, while the activation of both kinases was impaired in GRK2-overexpressing cells. The impaired differentiation and fewer fusion events promoted by enhanced GRK2 levels were recapitulated by a p38MAPK mutant, which was able to mimic the inhibitory phosphorylation of p38MAPK by GRK2, whereas the blunted differentiation observed in GRK2-expressing clones was rescued in the presence of a constitutively active upstream stimulator of the p38MAPK pathway. These results suggest that balanced GRK2 function is necessary for a timely and complete myogenic process.
Collapse
Affiliation(s)
- Lucia Garcia-Guerra
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Complutense University, 28040 Madrid, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08017 Barcelona, Spain Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain CIBER de enfermedades neurodegenerativas (CIBERNED), 28049 Madrid, Spain
| | - Rocío Vila-Bedmar
- Departament of Molecular Biology and Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain Instituto de Investigación Sanitaria la Princesa, 28006 Madrid, Spain
| | | | - Marta Cruces-Sande
- Departament of Molecular Biology and Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain Instituto de Investigación Sanitaria la Princesa, 28006 Madrid, Spain
| | - Mercedes Martín
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), 28049 Madrid, Spain
| | - Ana Ruiz-Gómez
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), 28049 Madrid, Spain
| | - Mar Ruiz-Gómez
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), 28049 Madrid, Spain
| | - Margarita Lorenzo
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Complutense University, 28040 Madrid, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08017 Barcelona, Spain
| | - Sonia Fernández-Veledo
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08017 Barcelona, Spain Hospital Universitari de Tarragona Joan XXIII. IISPV. Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Federico Mayor
- Departament of Molecular Biology and Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain Instituto de Investigación Sanitaria la Princesa, 28006 Madrid, Spain
| | - Cristina Murga
- Departament of Molecular Biology and Centro de Biología Molecular Severo Ochoa (CSIC-UAM), 28049 Madrid, Spain Instituto de Investigación Sanitaria la Princesa, 28006 Madrid, Spain
| | - Iria Nieto-Vázquez
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Complutense University, 28040 Madrid, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08017 Barcelona, Spain
| |
Collapse
|
38
|
Pérez-Moreno JJ, Bischoff M, Martín-Bermudo MD, Estrada B. The conserved transmembrane proteoglycan Perdido/Kon-tiki is essential for myofibrillogenesis and sarcomeric structure in Drosophila. J Cell Sci 2014; 127:3162-73. [PMID: 24794494 PMCID: PMC4095857 DOI: 10.1242/jcs.150425] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Muscle differentiation requires the assembly of high-order structures called myofibrils, composed of sarcomeres. Even though the molecular organization of sarcomeres is well known, the mechanisms underlying myofibrillogenesis are poorly understood. It has been proposed that integrin-dependent adhesion nucleates myofibrils at the periphery of the muscle cell to sustain sarcomere assembly. Here, we report a role for the gene perdido (perd, also known as kon-tiki, a transmembrane chondroitin proteoglycan) in myofibrillogenesis. Expression of perd RNAi in muscles, prior to adult myogenesis, can induce misorientation and detachment of Drosophila adult abdominal muscles. In comparison to controls, perd-depleted muscles contain fewer myofibrils, which are localized at the cell periphery. These myofibrils are detached from each other and display a defective sarcomeric structure. Our results demonstrate that the extracellular matrix receptor Perd has a specific role in the assembly of myofibrils and in sarcomeric organization. We suggest that Perd acts downstream or in parallel to integrins to enable the connection of nascent myofibrils to the Z-bands. Our work identifies the Drosophila adult abdominal muscles as a model to investigate in vivo the mechanisms behind myofibrillogenesis.
Collapse
Affiliation(s)
- Juan J Pérez-Moreno
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC, 41013 Seville, Spain
| | - Marcus Bischoff
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Maria D Martín-Bermudo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC, 41013 Seville, Spain
| | - Beatriz Estrada
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC, 41013 Seville, Spain
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Molnár I, Migh E, Szikora S, Kalmár T, Végh AG, Deák F, Barkó S, Bugyi B, Orfanos Z, Kovács J, Juhász G, Váró G, Nyitrai M, Sparrow J, Mihály J. DAAM is required for thin filament formation and Sarcomerogenesis during muscle development in Drosophila. PLoS Genet 2014; 10:e1004166. [PMID: 24586196 PMCID: PMC3937221 DOI: 10.1371/journal.pgen.1004166] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 12/23/2013] [Indexed: 11/19/2022] Open
Abstract
During muscle development, myosin and actin containing filaments assemble into the highly organized sarcomeric structure critical for muscle function. Although sarcomerogenesis clearly involves the de novo formation of actin filaments, this process remained poorly understood. Here we show that mouse and Drosophila members of the DAAM formin family are sarcomere-associated actin assembly factors enriched at the Z-disc and M-band. Analysis of dDAAM mutants revealed a pivotal role in myofibrillogenesis of larval somatic muscles, indirect flight muscles and the heart. We found that loss of dDAAM function results in multiple defects in sarcomere development including thin and thick filament disorganization, Z-disc and M-band formation, and a near complete absence of the myofibrillar lattice. Collectively, our data suggest that dDAAM is required for the initial assembly of thin filaments, and subsequently it promotes filament elongation by assembling short actin polymers that anneal to the pointed end of the growing filaments, and by antagonizing the capping protein Tropomodulin.
Collapse
Affiliation(s)
- Imre Molnár
- Institute of Genetics, Biological Research Centre HAS, Szeged, Hungary
| | - Ede Migh
- Institute of Genetics, Biological Research Centre HAS, Szeged, Hungary
| | - Szilárd Szikora
- Institute of Genetics, Biological Research Centre HAS, Szeged, Hungary
| | - Tibor Kalmár
- Institute of Genetics, Biological Research Centre HAS, Szeged, Hungary
| | - Attila G. Végh
- Institute of Biophysics, Biological Research Centre HAS, Szeged, Hungary
| | - Ferenc Deák
- Institute of Biochemistry, Biological Research Centre HAS, Szeged, Hungary
| | - Szilvia Barkó
- University of Pécs, Department of Biophysics, Pécs, Hungary
| | - Beáta Bugyi
- University of Pécs, Department of Biophysics, Pécs, Hungary
| | | | - János Kovács
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Gábor Juhász
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - György Váró
- Institute of Biophysics, Biological Research Centre HAS, Szeged, Hungary
| | - Miklós Nyitrai
- University of Pécs, Department of Biophysics, Pécs, Hungary
- Hungarian Academy of Sciences, Office for Subsidized Research Units, Budapest, Hungary
| | - John Sparrow
- Department of Biology, University of York, York, United Kingdom
| | - József Mihály
- Institute of Genetics, Biological Research Centre HAS, Szeged, Hungary
- * E-mail:
| |
Collapse
|
42
|
Domsch K, Ezzeddine N, Nguyen HT. Abba is an essential TRIM/RBCC protein to maintain the integrity of sarcomeric cytoarchitecture. J Cell Sci 2013; 126:3314-23. [PMID: 23729735 DOI: 10.1242/jcs.122366] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Organized sarcomeric striations are an evolutionarily conserved hallmark of functional skeletal muscles. Here, we demonstrate that the Drosophila Abba protein, a member of the TRIM/RBCC superfamily, has a pivotal regulatory role in maintaining proper sarcomeric cytoarchitecture during development and muscle usage. abba mutant embryos initially form muscles, but F-actin and Myosin striations become progressively disrupted when the muscles undergo growth and endure increased contractile forces during larval development. Abnormal Myosin aggregates and myofiber atrophy are also notable in the abba mutants. The larval defects result in compromised muscle function, and hence important morphogenetic events do not occur properly during pupation, leading to lethality. Abba is localized at larval Z-discs, and genetic evidence indicates that abba interacts with α-actinin, kettin/D-titin and mlp84B, genes that encode important Z-disc proteins for stable myofibrillar organization and optimal muscle function. RNAi experiments and ultrastructural analysis reveal that Abba has an additional crucial role in sarcomere maintenance in adult muscles. Abba is required to ensure the integrity and function of Z-discs and M-lines. Rescue experiments further show that Abba function is dependent upon its B-box/coiled-coil domain, NHL repeats and RING finger domain. The importance of these presumed protein-protein interactions and ubiquitin ligase-associated domains supports our hypothesis that Abba is needed for specific protein complex formation and stabilization at Z-discs and M-lines.
Collapse
Affiliation(s)
- Katrin Domsch
- Department of Biology, Division of Developmental Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Staudtstrasse 5, 91058 Erlangen, Germany
| | | | | |
Collapse
|
43
|
Perry NA, Ackermann MA, Shriver M, Hu LYR, Kontrogianni-Konstantopoulos A. Obscurins: unassuming giants enter the spotlight. IUBMB Life 2013; 65:479-86. [PMID: 23512348 DOI: 10.1002/iub.1157] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 01/31/2013] [Indexed: 02/04/2023]
Abstract
Discovered about a decade ago, obscurin (~720 kDa) is a member of a family of giant proteins expressed in striated muscle that are essential for normal muscle function. Much of what we understand about obscurin stems from its functions in cardiac and skeletal muscle. However, recent evidence has indicated that variants of obscurin ("obscurins") are expressed in diverse cell types, where they contribute to distinct cellular processes. Dysfunction or abrogation of obscurins has also been implicated in the development of several pathological conditions, including cardiac hypertrophy and cancer. Herein, we present an overview of obscurins with an emphasis on novel findings that demonstrate their heretofore-unsuspected importance in cell signaling and disease progression.
Collapse
Affiliation(s)
- Nicole A Perry
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | |
Collapse
|
44
|
Lehmann FO, Skandalis DA, Berthé R. Calcium signalling indicates bilateral power balancing in the Drosophila flight muscle during manoeuvring flight. J R Soc Interface 2013; 10:20121050. [PMID: 23486171 DOI: 10.1098/rsif.2012.1050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Manoeuvring flight in animals requires precise adjustments of mechanical power output produced by the flight musculature. In many insects such as fruit flies, power generation is most likely varied by altering stretch-activated tension, that is set by sarcoplasmic calcium levels. The muscles reside in a thoracic shell that simultaneously drives both wings during wing flapping. Using a genetically expressed muscle calcium indicator, we here demonstrate in vivo the ability of this animal to bilaterally adjust its calcium activation to the mechanical power output required to sustain aerodynamic costs during flight. Motoneuron-specific comparisons of calcium activation during lift modulation and yaw turning behaviour suggest slightly higher calcium activation for dorso-longitudinal than for dorsoventral muscle fibres, which corroborates the elevated need for muscle mechanical power during the wings' downstroke. During turning flight, calcium activation explains only up to 54 per cent of the required changes in mechanical power, suggesting substantial power transmission between both sides of the thoracic shell. The bilateral control of muscle calcium runs counter to the hypothesis that the thorax of flies acts as a single, equally proportional source for mechanical power production for both flapping wings. Collectively, power balancing highlights the precision with which insects adjust their flight motor to changing energetic requirements during aerial steering. This potentially enhances flight efficiency and is thus of interest for the development of technical vehicles that employ bioinspired strategies of power delivery to flapping wings.
Collapse
Affiliation(s)
- Fritz-Olaf Lehmann
- Department of Animal Physiology, University of Rostock, Rostock, Germany.
| | | | | |
Collapse
|
45
|
Alp/Enigma family proteins cooperate in Z-disc formation and myofibril assembly. PLoS Genet 2013; 9:e1003342. [PMID: 23505387 PMCID: PMC3591300 DOI: 10.1371/journal.pgen.1003342] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 01/10/2013] [Indexed: 11/19/2022] Open
Abstract
The Drosophila Alp/Enigma family protein Zasp52 localizes to myotendinous junctions and Z-discs. It is required for terminal muscle differentiation and muscle attachment. Its vertebrate ortholog ZASP/Cypher also localizes to Z-discs, interacts with α-actinin through its PDZ domain, and is involved in Z-disc maintenance. Human mutations in ZASP cause myopathies and cardiomyopathies. Here we show that Drosophila Zasp52 is one of the earliest markers of Z-disc assembly, and we use a Zasp52-GFP fusion to document myofibril assembly by live imaging. We demonstrate that Zasp52 is required for adult Z-disc stability and pupal myofibril assembly. In addition, we show that two closely related proteins, Zasp66 and the newly identified Zasp67, are also required for adult Z-disc stability and are participating with Zasp52 in Z-disc assembly resulting in more severe, synergistic myofibril defects in double mutants. Zasp52 and Zasp66 directly bind to α-actinin, and they can also form a ternary complex. Our results indicate that Alp/Enigma family members cooperate in Z-disc assembly and myofibril formation; and we propose, based on sequence analysis, a novel class of PDZ domain likely involved in α-actinin binding.
Collapse
|
46
|
Gokhin DS, Fowler VM. A two-segment model for thin filament architecture in skeletal muscle. Nat Rev Mol Cell Biol 2013; 14:113-9. [PMID: 23299957 DOI: 10.1038/nrm3510] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Correct specification of myofilament length is essential for efficient skeletal muscle contraction. The length of thin actin filaments can be explained by a novel 'two-segment' model, wherein the thin filaments consist of two concatenated segments, which are of either constant or variable length. This is in contrast to the classic 'nebulin ruler' model, which postulates that thin filaments are uniform structures, the lengths of which are dictated by nebulin. The two-segment model implicates position-specific microregulation of actin dynamics as a general principle underlying actin filament length and stability.
Collapse
Affiliation(s)
- David S Gokhin
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
47
|
Orfanos Z, Sparrow JC. Myosin isoform switching during assembly of the Drosophila flight muscle thick filament lattice. J Cell Sci 2012. [PMID: 23178940 DOI: 10.1242/jcs.110361] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During muscle development myosin molecules form symmetrical thick filaments, which integrate with the thin filaments to produce the regular sarcomeric lattice. In Drosophila indirect flight muscles (IFMs) the details of this process can be studied using genetic approaches. The weeP26 transgenic line has a GFP-encoding exon inserted into the single Drosophila muscle myosin heavy chain gene, Mhc. The weeP26 IFM sarcomeres have a unique MHC-GFP-labelling pattern restricted to the sarcomere core, explained by non-translation of the GFP exon following alternative splicing. Characterisation of wild-type IFM MHC mRNA confirmed the presence of an alternately spliced isoform, expressed earlier than the major IFM-specific isoform. The two wild-type IFM-specific MHC isoforms differ by the presence of a C-terminal 'tailpiece' in the minor isoform. The sequential expression and assembly of these two MHCs into developing thick filaments suggest a role for the tailpiece in initiating A-band formation. The restriction of the MHC-GFP sarcomeric pattern in weeP26 is lifted when the IFM lack the IFM-specific myosin binding protein flightin, suggesting that it limits myosin dissociation from thick filaments. Studies of flightin binding to developing thick filaments reveal a progressive binding at the growing thick filament tips and in a retrograde direction to earlier assembled, proximal filament regions. We propose that this flightin binding restricts myosin molecule incorporation/dissociation during thick filament assembly and explains the location of the early MHC isoform pattern in the IFM A-band.
Collapse
|