Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Gene ; 631: 16-28, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28739398

ABSTRACT

Muscle contraction is a highly fine-tuned process that requires the precise and timely construction of large protein sub-assemblies to form sarcomeres. Mutations in many genes encoding constituent proteins of this macromolecular machine result in defective functioning of the muscle tissue. However, the pathways underlying muscle degeneration, and manifestation of myopathy phenotypes are not well understood. In this study, we explored transcriptional alterations that ensue from the absence of the two major muscle proteins - myosin and actin - using the Drosophila indirect flight muscles. Our aim was to understand how the muscle tissue responds as a whole to the absence of either of the major scaffold proteins, whether the responses are generic to the tissue; or unique to the thick versus thin filament systems. Our results indicated that muscles respond by altering gene transcriptional levels in multiple systems active in muscle remodelling, protein degradation and heat shock responses. However, there were some responses that were filament-specific signatures of muscle degeneration, like immune responses, metabolic alterations and alterations in expression of muscle structural genes and mitochondrial ribosomal genes. These general and filament-specific changes in gene expression may be of relevance to human myopathies.


Subject(s)
Actins/genetics , Muscle Contraction/genetics , Myosins/genetics , Actins/metabolism , Animals , Drosophila , Drosophila Proteins/genetics , Gene Expression Profiling , Male , Muscle, Striated/physiology , Mutation , Myosins/metabolism , Oligonucleotide Array Sequence Analysis
2.
J Cell Sci ; 126(Pt 1): 139-48, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23178940

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.


Subject(s)
Drosophila Proteins/metabolism , Muscle, Skeletal/metabolism , Protein Isoforms/metabolism , Alternative Splicing/genetics , Alternative Splicing/physiology , Animals , Drosophila , Drosophila Proteins/genetics , Exons/genetics , Filamins , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myosins/genetics , Myosins/metabolism , Protein Isoforms/genetics , Sarcomeres/metabolism
3.
J Mol Biol ; 419(1-2): 22-40, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22370558

ABSTRACT

The roles of myosin during muscle contraction are well studied, but how different domains of this protein are involved in myofibril assembly in vivo is far less understood. The indirect flight muscles (IFMs) of Drosophila melanogaster provide a good model for understanding muscle development and function in vivo. We show that two missense mutations in the rod region of the myosin heavy-chain gene, Mhc, give rise to IFM defects and abnormal myofibrils. These defects likely result from thick filament abnormalities that manifest during early sarcomere development or later by hypercontraction. The thick filament defects are accompanied by marked reduction in accumulation of flightin, a myosin binding protein, and its phosphorylated forms, which are required to stabilise thick filaments. We investigated with purified rod fragments whether the mutations affect the coiled-coil structure, rod aggregate size or rod stability. No significant changes in these parameters were detected, except for rod thermodynamic stability in one mutation. Molecular dynamics simulations suggest that these mutations may produce localised rod instabilities. We conclude that the aberrant myofibrils are a result of thick filament defects, but that these in vivo effects cannot be detected in vitro using the biophysical techniques employed. The in vivo investigation of these mutant phenotypes in IFM development and function provides a useful platform for studying myosin rod and thick filament formation generically, with application to the aetiology of human myosin rod myopathies.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Mutation, Missense , Myofibrils/metabolism , Myosin Subfragments/genetics , Myosin Subfragments/metabolism , Amino Acid Sequence , Animals , Drosophila Proteins/chemistry , Drosophila melanogaster , Filamins , Flight, Animal/physiology , Molecular Dynamics Simulation , Molecular Sequence Data , Muscle Contraction , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myofibrils/chemistry , Myofibrils/genetics , Myofibrils/ultrastructure , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Subfragments/chemistry , Phenotype , Phosphorylation/genetics
4.
Methods ; 56(1): 78-86, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22037247

ABSTRACT

We describe our methods for analysing muscle function in a whole intact small insect, taking advantage of a simple flexible optical beam to produce an inexpensive transducer with wide application. We review our previous data measuring the response to a single action potential driven muscle twitch to explore jumping behaviour in Drosophila melanogaster. In the fruitfly, where the sophisticated and powerful genetic toolbox is being widely employed to investigate neuromuscular function, we further demonstrate the use of the apparatus to analyse in detail, within whole flies, neuronal and muscle mutations affecting activation of muscle contraction in the jump muscle. We have now extended the use of the apparatus to record the muscle forces during larval and other aspects of adult locomotion. The robustness, simplicity and versatility of the apparatus are key to these measurements.


Subject(s)
Drosophila melanogaster , Muscle Contraction/physiology , Animals , Flight, Animal/physiology , Muscle Contraction/genetics , Mutation , Transducers , Walking/physiology
5.
Neuromuscul Disord ; 20(8): 531-3, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20621480

ABSTRACT

We report a female newborn, diagnosed with fetal akinesia in utero, who died one hour after birth. Post-mortem muscle biopsy demonstrated actin-filament myopathy based on immunolabelling for sarcomeric actin, and large areas of filaments, without rod formation, ultrastructurally. Analysis of DNA extracted from the muscle disclosed a novel de novo heterozygous c.44G>A, GGC>GAC, 'p.Gly15Asp' mutation in the ACTA1 gene. Analysis of the location of the mutated amino-acid in the actin molecule suggests the mutation most likely causes abnormal nucleotide binding, and consequent pathological actin polymerization. This case emphasizes the association of fetal akinesia with actin-filament myopathy.


Subject(s)
Actin Cytoskeleton/genetics , Fetal Diseases/genetics , Microfilament Proteins/genetics , Neuromuscular Diseases/genetics , Actin Cytoskeleton/pathology , Adult , DNA/genetics , Female , Fetal Diseases/pathology , Humans , Infant, Newborn , Muscle, Skeletal/pathology , Mutation/genetics , Mutation/physiology , Neuromuscular Diseases/pathology , Pregnancy , Sarcomeres/genetics
6.
Neuromuscul Disord ; 20(6): 363-74, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20452215

ABSTRACT

Most human ACTA1 skeletal actin gene mutations cause dominant, congenital myopathies often with severely reduced muscle function and neonatal mortality. High sequence conservation of actin means many mutated ACTA1 residues are identical to those in the DrosophilaAct88F, an indirect flight muscle specific sarcomeric actin. Four known Act88F mutations occur at the same actin residues mutated in ten ACTA1 nemaline mutations, A138D/P, R256H/L, G268C/D/R/S and R372C/S. These Act88F mutants were examined for similar muscle phenotypes. Mutant homozygotes show phenotypes ranging from a lack of myofibrils to almost normal sarcomeres at eclosion. Aberrant Z-disc-like structures and serial Z-disc arrays, 'zebra bodies', are observed in homozygotes and heterozygotes of all four Act88F mutants. These electron-dense structures show homologies to human nemaline bodies/rods, but are much smaller than those typically found in the human myopathy. We conclude that the Drosophila indirect flight muscles provide a good model system for studying ACTA1 mutations.


Subject(s)
Actins/genetics , Drosophila/physiology , Flight, Animal/physiology , Muscle, Skeletal/physiology , Muscles/physiology , Muscular Diseases/genetics , Actins/physiology , Animals , Blotting, Western , DNA/genetics , Humans , Microscopy, Confocal , Microscopy, Electron , Models, Molecular , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscles/metabolism , Muscles/pathology , Muscular Diseases/congenital , Muscular Diseases/pathology , Mutation , Myofibrils/pathology , Pupa
7.
Hum Mutat ; 30(9): 1267-77, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19562689

ABSTRACT

The ACTA1 gene encodes skeletal muscle alpha-actin, which is the predominant actin isoform in the sarcomeric thin filaments of adult skeletal muscle, and essential, along with myosin, for muscle contraction. ACTA1 disease-causing mutations were first described in 1999, when a total of 15 mutations were known. In this article we describe 177 different disease-causing ACTA1 mutations, including 85 that have not been described before. ACTA1 mutations result in five overlapping congenital myopathies: nemaline myopathy; intranuclear rod myopathy; actin filament aggregate myopathy; congenital fiber type disproportion; and myopathy with core-like areas. Mixtures of these histopathological phenotypes may be seen in a single biopsy from one patient. Irrespective of the histopathology, the disease is frequently clinically severe, with many patients dying within the first year of life. Most mutations are dominant and most patients have de novo mutations not present in the peripheral blood DNA of either parent. Only 10% of mutations are recessive and they are genetic or functional null mutations. To aid molecular diagnosis and establishing genotype-phenotype correlations, we have developed a locus-specific database for ACTA1 variations (http://waimr.uwa.edu.au).


Subject(s)
Actins/genetics , Muscle, Skeletal/metabolism , Mutation , Polymorphism, Genetic , Actins/metabolism , Alleles , Databases, Genetic , Genetic Variation , Humans , Models, Molecular , Muscular Diseases/genetics , Muscular Diseases/pathology , Phenotype
8.
Nat Rev Mol Cell Biol ; 10(4): 293-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19190670

ABSTRACT

Myofibril assembly results in a regular array of identical sarcomeres in striated muscle. Sarcomere structure is conserved across the animal kingdom, which implies that the mechanisms of myofibril assembly are also likely to be conserved. Recent advances from model genetic systems and insights from stress fibre cell biology have shed light on the mechanisms that set sarcomere spacing and the initial assembly of sarcomere arrays. We propose a model of integrin-dependent cell-matrix adhesion as the starting point for myofibrillogenesis.


Subject(s)
Integrins/metabolism , Muscle Development , Animals , Cell-Matrix Junctions/metabolism , Sarcomeres/metabolism
9.
Invert Neurosci ; 8(2): 63-70, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18443837

ABSTRACT

How do deficits in neuronal growth, aging or synaptic function affect the final, mechanical output of a single muscle twitch? We address this in vivo (indeed in situ) with a novel ergometer that records the output of a large specialised muscle, the Drosophila jump muscle. Here, we describe in detail the ergometer, its construction and use. We evaluated the ergometer by showing that adult fly jump muscle output varies little between 3 h and 7 days; but newly eclosed flies produce only 65%. In a mutant with little octopamine (Tbetah), jump muscle performance is reduced by 28%. The initial responses of synaptic growth mutants (highwire and spinster) do not differ from wild type, as expected on the homeostatic hypothesis. However, responses in highwire mutations gradually decline following repeated stimuli, suggesting physiological as well as anatomical abnormalities. We conclude that the assay is robust, sensitive and reliable with a good throughput.


Subject(s)
Drosophila/physiology , Ergometry , Muscles/physiology , Mutation , Neuromuscular Junction/physiology , Animals , Behavior, Animal , Drosophila/genetics , Ergometry/instrumentation , Ergometry/methods , Octopamine/genetics
10.
Ann Neurol ; 62(6): 597-608, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17705262

ABSTRACT

OBJECTIVE: Mutations in the alpha-skeletal actin gene (ACTA1) result in a variety of inherited muscle disorders characterized by different pathologies and variable clinical phenotypes. Mutations at Val163 in ACTA1 result in pure intranuclear rod myopathy; however, the molecular mechanisms by which mutations at Val163 lead to intranuclear rod formation and muscle weakness are unknown. METHODS AND RESULTS: We investigated the effects of the Val163Met mutation in ACTA1 in tissue culture and Drosophila models, and in patient muscle. In cultured cells, the mutant actin tends to aggregate rather than incorporate into cytoplasmic microfilaments, and it affects the dynamics of wild-type actin, causing it to accumulate with the mutant actin in the nucleus. In Drosophila, the Val163Met mutation severely disrupts the structure of the muscle sarcomere. The intranuclear aggregates in patient muscle biopsies impact on nuclear structure and sequester normal Z-disc-associated proteins within the nucleus; however, the sarcomeric structure is relatively well preserved, with evidence of active regeneration. By mass spectrometry, the levels of mutant protein are markedly reduced in patient muscle compared with control. INTERPRETATION: Data from our tissue culture and Drosophila models show that the Val163Met mutation in alpha-skeletal actin can affect the dynamics of other actin isoforms and severely disrupt sarcomeric structure, processes that can contribute to muscle weakness. However, in human muscle, there is evidence of regeneration, and the mutant protein tends to aggregate rather than incorporate into cytoplasmic microfilaments in cells. These are likely compensatory processes that ameliorate the effects of the mutant actin and contribute to the milder clinical and pathological disease phenotype.


Subject(s)
Actins/genetics , Muscular Diseases/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Adaptation, Physiological , Animals , Animals, Genetically Modified , Cell Line , Cytoplasm/metabolism , Drosophila , Humans , Methionine , Mice , Muscle Weakness/etiology , Muscle, Skeletal/physiopathology , Muscular Diseases/complications , Muscular Diseases/metabolism , Muscular Diseases/pathology , Protein Isoforms/metabolism , Regeneration , Sarcomeres/pathology , Transfection , Valine
11.
Fly (Austin) ; 1(2): 68-74, 2007.
Article in English | MEDLINE | ID: mdl-18820447

ABSTRACT

We have developed a novel apparatus, an ergometer, to simultaneously measure the horizontal and vertical components of the work done during takeoff by the fruitfly, Drosophila. We confirm the anatomical prediction that all the work comes from the middle (mesothoracic) legs. With all six legs on the ergometer platform, displacement is directed roughly 45 degrees forwards or backwards. Both directions are equally likely. This provides for a random, rapid horizontal component to the escape behaviour for flies. When the thoracic stiffness is reduced (due to a mutation in which the indirect flight muscles (IFM) do not form myofibrils), jump output is increased. We conclude that the jump muscle, the tergal depressor of trochanter (TDT), which lacks direct muscle antagonists, performs work during the jump against thoracic stiffness. Both cuticle and IFM contribute to the thoracic stiffness as the TDT still produces repeated contractions in the absence of the IFM. Degeneration of the TDT due to mutants in three sarcomeric proteins results in reduction of the jump output. In one of these, the myosin heavy chain mutant, Mhc5, we show that degeneration occurs with age. The anatomical characteristics of Drosophila mean that we are recording, for the first time in the intact fly, the output of a single muscle that has high homology to vertebrate skeletal muscle. Developing an ergometer for Drosophila offers novel opportunities to assess the functional consequences of mutations in muscle proteins, synaptic physiology, neuromuscular development and aging.


Subject(s)
Drosophila melanogaster/physiology , Muscle Contraction/physiology , Animals , Ergometry/instrumentation , Extremities/physiology , Female , Muscle Proteins/genetics , Mutation , Physical Exertion/physiology
12.
BMC Biol ; 4: 17, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16768790

ABSTRACT

BACKGROUND: The processes by which eggs develop in the insect ovary are well characterized. Despite a large number of Drosophila mutants that cannot lay eggs, the way that the egg is moved along the reproductive tract from ovary to uterus is less well understood. We remedy this with an integrative study on the reproductive tract muscles (anatomy, innervation, contractions, aminergic modulation) in female flies. RESULTS: Each ovary, consisting of 15-20 ovarioles, is surrounded by a contractile meshwork, the peritoneal sheath. Individual ovarioles are contained within a contractile epithelial sheath. Both sheaths contain striated muscle fibres. The oviduct and uterine walls contain a circular striated muscle layer. No longitudinal muscle fibres are seen. Neurons that innervate the peritoneal sheath and lateral oviduct have many varicosities and terminate in swellings just outside the muscles of the peritoneal sheath. They all express tyrosine decarboxylase (required for tyramine and octopamine synthesis) and Drosophila vesicular monoamine transporter (DVMAT). No fibres innervate the ovarioles. The common oviduct and uterus are innervated by two classes of neurons, one with similar morphology to those of the peritoneal sheath and another with repeated branches and axon endings similar to type I neuromuscular junctions. In isolated genital tracts from 3- and 7-day old flies, each ovariole contracts irregularly (12.5 +/- 6.4 contractions/minute; mean +/- 95% confidence interval). Peritoneal sheath contractions (5.7 +/- 1.6 contractions/minute) move over the ovary, from tip to base or vice versa, propagating down the oviduct. Rhythmical spermathecal rotations (1.5 +/- 0.29 contractions/minute) also occur. Each genital tract organ exhibits its own endogenous myogenic rhythm. The amplitude of contractions of the peritoneal sheath increase in octopamine (100 nM, 81% P < 0.02) but 1 microM tyramine has no effect. Neither affects the frequency of peritoneal sheath contractions. CONCLUSION: The muscle fibres of the reproductive tract are circular and have complex bursting myogenic rhythms under octopaminergic neuromodulation. We propose a new model of tissue-specific actions of octopamine, in which strengthening of peritoneal sheath contractions, coupled with relaxation of the oviduct, eases ovulation. This model accounts for reduced ovulation in flies with mutations in the octopaminergic system.


Subject(s)
Drosophila/physiology , Neuromuscular Junction/anatomy & histology , Octopamine/physiology , Oviposition/physiology , Tyramine/physiology , Animals , Drosophila/anatomy & histology , Female , Muscle Contraction , Muscles/anatomy & histology , Muscles/innervation , Neuromuscular Junction/physiology , Ovary/anatomy & histology , Ovary/innervation
13.
J Mol Biol ; 347(5): 889-94, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15784249

ABSTRACT

Contraction in striated muscles is regulated by Ca2+-dependent movement of tropomyosin-troponin on thin filaments. Interactions of charged amino acid residues between the surfaces of tropomyosin and actin are believed to play an integral role in this steric mechanism by influencing the position of tropomyosin on the filaments. To investigate this possibility further, thin filaments were isolated from troponin-regulated, indirect flight muscles of Drosophila mutants that express actin with an amino acid charge reversal at residue 93 located at the interface between actin subdomains 1 and 2, in which a lysine residue is substituted for a glutamic acid. Electron microscopy and 3D helical reconstruction were employed to evaluate the structural effects of the mutation. In the absence of Ca2+, tropomyosin was in a position that blocked the myosin-binding sites on actin, as previously found with wild-type filaments. However, in the presence of Ca2+, tropomyosin position in the mutant filaments was much more variable than in the wild-type ones. In most cases (approximately 60%), tropomyosin remained in the blocking position despite the presence of Ca2+, failing to undergo a normal Ca2+-induced change in position. Thus, switching of a negative to a positive charge at position 93 on actin may stabilize negatively charged tropomyosin in the Ca2+-free state regardless of Ca2+ levels, an alteration that, in turn, is likely to interfere with steric regulation and consequently muscle activation. These results highlight the importance of actin's surface charges in determining the distribution of tropomyosin positions on thin filaments derived from troponin-regulated striated muscles.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/metabolism , Glutamic Acid/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Actins/genetics , Actins/ultrastructure , Animals , Calcium/metabolism , Calcium/pharmacology , Drosophila melanogaster , Glutamic Acid/genetics , Microscopy, Electron , Mutation/genetics
14.
Ann Neurol ; 56(5): 689-94, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15468086

ABSTRACT

We report three heterozygous missense mutations of the skeletal muscle alpha actin gene (ACTA1) in three unrelated cases of congenital fiber type disproportion (CFTD) from Japan and Australia. This represents the first genetic cause of CFTD to be identified and confirms that CFTD is genetically heterogeneous. The three mutations we have identified Leucine221Proline, Aspartate292Valine, and Proline332Serine are novel. They have not been found previously in any cases of nemaline, actin, intranuclear rod, or rod-core myopathy caused by mutations in ACTA1. It remains unclear why these mutations cause type 1 fiber hypotrophy but no nemaline bodies. The three mutations all lie on one face of the actin monomer on the surface swept by tropomyosin during muscle activity, which may suggest a common pathological mechanism. All three CFTD cases with ACTA1 mutations had severe congenital weakness and respiratory failure without ophthalmoplegia. There were no clinical features specific to CFTD cases with ACTA1 mutations, but the presence of normal eye movements in a severe CFTD patient may be an important clue for the presence of a mutation in ACTA1.


Subject(s)
Actins/genetics , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Mutation, Missense , Myopathies, Structural, Congenital/genetics , Adenosine Triphosphatases/metabolism , Aspartic Acid/genetics , Biopsy/methods , Child, Preschool , DNA Mutational Analysis/methods , Female , Heterozygote , Humans , Infant , Infant, Newborn , Leucine/genetics , Male , Models, Molecular , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Proline/genetics , Sequence Analysis, Protein , Serine/genetics , Valine/genetics
15.
J Exp Biol ; 207(Pt 20): 3515-22, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15339947

ABSTRACT

In many insects renowned for their jumping ability, elastic storage is used so that high forces can be developed prior to jumping. We have combined physiological, behavioural and genetic approaches to test whether elastic energy storage makes a major contribution to jumping in Drosophila. We describe a sensitive strain gauge setup, which measures the forces produced by tethered flies through their mesothoracic legs. The peak force produced by the main jumping muscle of female flies from a wild-type (Canton-S) strain is 101+/-4.4 microN [and this is indistinguishable from a second wild-type (Texas) strain]. The force takes 8.2 ms to reach its peak. The peak force is not affected significantly by altering the leg angle (femur-tibia joint angle) in the range of 75-120 degrees, but the peak force declines as the leg is extended further. Measurements of jumping ability (distance jumped) showed that female Drosophila (with their wings removed) of two wild-type strains, Canton-S and Texas, produced jumps of 28.6+/-0.7 and 30.2+/-1.0 mm (mean +/- s.e.m.). For a female wild-type Drosophila, a jump of 30 mm corresponds to a kinetic energy of 200 nJ on take-off (allowing 20% of the energy to overcome air resistance). We develop equations of motion for a linear force-time model of take-off and calculate that the time to take-off is 5.0 ms and the peak force should be 274 microN (137 microN leg(-1)). We predicted, from the role of octopamine in enhancing muscle tension in several locust muscles, that if stored elastic energy plays no part in force development, then genetic manipulation of the octopaminergic system would directly affect force production and jumping in Drosophila. Using two mutants deficient in the octopaminergic system, TbhnM18 (M18) and TyrRhono (hono), we found significantly reduced jumping distances (20.7+/-0.7 and 20.7+/-0.4 mm, respectively) and force production (52% and 55%, respectively) compared with wild type. From the reduced distance and force production in M18, a mutant deficient in octopamine synthesis, and in hono, a tyramine/octopamine receptor mutant, we conclude that in Drosophila, as in locusts, octopamine modulates escape jumping. We conclude that the fly does not need to store large quantities of elastic energy in order to make its jump because (1) the measured and calculated forces agree to within 40% and (2) the reduction in distances jumped by the mutants correlates well with their reduction in measured peak force.


Subject(s)
Drosophila melanogaster/physiology , Locomotion/physiology , Models, Biological , Muscle, Skeletal/physiology , Octopamine/metabolism , Animals , Biomechanical Phenomena , Drosophila melanogaster/genetics , Electric Stimulation , Female , Muscle, Skeletal/metabolism , Octopamine/genetics
16.
J Cell Sci ; 117(Pt 9): 1795-805, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15075240

ABSTRACT

Myofibrillar proteins assemble to form the highly ordered repetitive contractile structural unit known as a sarcomere. Studies of myogenesis in vertebrate cell culture and embryonic developmental systems have identified some of the processes involved during sarcomere formation. However, isoform changes during vertebrate muscle development and a lack of mutants have made it difficult to determine how these proteins assemble to form sarcomeres. The indirect flight muscles (IFMs) of Drosophila provide a unique genetic system with which to study myofibrillogenesis in vivo. We show in this paper that neither sarcomeric myosin nor actin are required for myoblast fusion or the subsequent morphogenesis of muscle fibres, i.e. fibre morphogenesis does not depend on myofibrillogenesis. However, fibre formation and myofibrillogenesis are very sensitive to the interactions between the sarcomeric proteins. A troponin I (TnI) mutation, hdp(3), leads to an absence of TnI in the IFMs and tergal depressor of trochanter (TDT) muscles due to a transcript-splicing defect. Sarcomeres do not form and the muscles degenerate. TnI is part of the thin filament troponin complex which regulates muscle contraction. The effects of the hdp(3) mutation are probably caused by unregulated acto-myosin interactions between the thin and thick filaments as they assemble. We have tested this proposal by using a transgenic myosin construct to remove the force-producing myosin heads. The defects in sarcomeric organisation and fibre degeneration in hdp(3) IFMs are suppressed, although not completely, indicating the need for inhibition of muscle contraction during muscle development. We show that mRNA and translated protein products of all the major thin filament proteins are reduced in hdp(3) muscles and discuss how this and previous studies of thin filament protein mutants indicate a common co-ordinated control mechanism that may be the primary cause of the muscle defects.


Subject(s)
Drosophila melanogaster/metabolism , Muscle Development/physiology , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Troponin I/metabolism , Wings, Animal/growth & development , Actins/genetics , Actomyosin/genetics , Actomyosin/metabolism , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/ultrastructure , Gene Expression , Genes, Insect/genetics , Muscle Contraction/genetics , Mutation/genetics , Myosins/genetics , Tropomyosin/genetics , Tropomyosin/metabolism , Troponin I/deficiency , Troponin I/genetics , Wings, Animal/ultrastructure
17.
Biophys J ; 86(3): 1618-24, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990488

ABSTRACT

Wild-type and mutant thin filaments were isolated directly from "myosinless" Drosophila indirect flight muscles to study the structural basis of muscle regulation genetically. Negatively stained filaments showed tropomyosin with periodically arranged troponin complexes in electron micrographs. Three-dimensional helical reconstruction of wild-type filaments indicated that the positions of tropomyosin on actin in the presence and absence of Ca(2+) were indistinguishable from those in vertebrate striated muscle and consistent with a steric mechanism of regulation by troponin-tropomyosin in Drosophila muscles. Thus, the Drosophila model can be used to study steric regulation. Thin filaments from the Drosophila mutant heldup(2), which possesses a single amino acid conversion in troponin I, were similarly analyzed to assess the Drosophila model genetically. The positions of tropomyosin in the mutant filaments, in both the Ca(2+)-free and the Ca(2+)-induced states, were the same, and identical to that of wild-type filaments in the presence of Ca(2+). Thus, cross-bridge cycling would be expected to proceed uninhibited in these fibers, even in relaxing conditions, and this would account for the dramatic hypercontraction characteristic of these mutant muscles. The interaction of mutant troponin I with Drosophila troponin C is discussed, along with functional differences between troponin C from Drosophila and vertebrates.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/ultrastructure , Gene Expression Regulation/physiology , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Microscopy, Electron/methods , Muscle, Skeletal/physiology , Muscle, Skeletal/ultrastructure , Actin Cytoskeleton/physiology , Animals , Drosophila , Genetic Engineering , Muscle, Skeletal/chemistry , Rabbits , Species Specificity , Structure-Activity Relationship
18.
Structure ; 11(10): 1196-7, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14527387
19.
Mol Biol Evol ; 20(12): 2019-33, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12949144

ABSTRACT

Uniquely, the asynchronous flight muscle myofibrils of many insects contain arthrin, a stable 1:1 conjugate between actin and ubiquitin. The function of arthrin is still unknown. Here we survey for the presence of arthrin in 63 species of insect across nine orders using Western blotting. Analysis of the evolutionary distribution shows that arthrin has evolved a limited number of times but at least once in the Diptera and once in the Hemiptera. However, the presence of arthrin does not correlate with any observed common features of flight mechanism, natural history, or morphology. We also identify the site of the isopeptide bond in arthrin from Drosophila melanogaster (Diptera) and Lethocerus griseus (Hemiptera) using mass spectrometry. In both species, the isopeptide bond is formed between lysine 118 of the actin and the C-terminal glycine 76 of ubiquitin. Thus, not only the ubiquitination of actin but also the site of the isopeptide bond has evolved convergently in Diptera and Hemiptera. In terms of the actin monomer, lysine 118 is near neither the binding sites of the major actin-binding proteins, myosin, tropomyosin, or the troponins, nor the actin polymerization sites. However, molecular modeling supports the idea that ubiquitin bound to an actin in one F-actin strand might be able to interact with tropomyosin bound to the actin monomers of the other strand and thereby interfere with thin filament regulation.


Subject(s)
Diptera/genetics , Evolution, Molecular , Hemiptera/genetics , Microfilament Proteins/genetics , Muscle Proteins/genetics , Actins/chemistry , Actins/metabolism , Amino Acid Sequence , Animals , Diptera/classification , Drosophila melanogaster/chemistry , Drosophila melanogaster/genetics , Hemiptera/classification , Insect Proteins , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Models, Molecular , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Phylogeny , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Ubiquitin/chemistry , Ubiquitin/genetics
20.
Neuromuscul Disord ; 13(7-8): 519-31, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12921789

ABSTRACT

Mutations in the skeletal muscle alpha-actin gene (ACTA1) associated with congenital myopathy with excess of thin myofilaments, nemaline myopathy and intranuclear rod myopathy were first described in 1999. At that time, only 15 different missense mutations were known in ACTA1. More than 60 mutations have now been identified. This review analyses this larger spectrum of mutations in ACTA1. It investigates the molecular consequences of the mutations found to date, provides a framework for genotype-phenotype correlation and suggests future studies in light of results of investigation of normal and mutant actin in other systems, notably the actin specific to the indirect flight muscles of Drosophila. The larger series confirms that the majority of ACTA1 mutations are dominant, a small number are recessive and most isolated cases with no previous family history have de novo dominant mutations. The severity of the disease caused ranges from lack of spontaneous movements at birth requiring immediate mechanical ventilation, to mild disease compatible with life to adulthood. Overall, the mutations within ACTA1 are randomly distributed throughout the protein. However, the larger series of mutations now available indicates that there may be clustering of mutations associated with some phenotypes, e.g. actin myopathy. This would suggest that interference with certain actin functions may be more associated with certain phenotypes, though the exact pathophysiology of the actin mutations remains unknown.


Subject(s)
Actins/genetics , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Mutation , Actins/chemistry , Amino Acid Sequence , Animals , Genetic Variation , Humans , Muscle Proteins/metabolism , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Phenotype , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...