Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nat Rev Mol Cell Biol ; 10(4): 293-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19190670

RESUMEN

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.


Asunto(s)
Integrinas/metabolismo , Desarrollo de Músculos , Animales , Uniones Célula-Matriz/metabolismo , Sarcómeros/metabolismo
2.
J Cell Sci ; 126(Pt 1): 139-48, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23178940

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/metabolismo , Músculo Esquelético/metabolismo , Isoformas de Proteínas/metabolismo , Empalme Alternativo/genética , Empalme Alternativo/fisiología , Animales , Drosophila , Proteínas de Drosophila/genética , Exones/genética , Filaminas , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miosinas/genética , Miosinas/metabolismo , Isoformas de Proteínas/genética , Sarcómeros/metabolismo
3.
Methods ; 56(1): 78-86, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22037247

RESUMEN

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.


Asunto(s)
Drosophila melanogaster , Contracción Muscular/fisiología , Animales , Vuelo Animal/fisiología , Contracción Muscular/genética , Mutación , Transductores , Caminata/fisiología
4.
Hum Mutat ; 30(9): 1267-77, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19562689

RESUMEN

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).


Asunto(s)
Actinas/genética , Músculo Esquelético/metabolismo , Mutación , Polimorfismo Genético , Actinas/metabolismo , Alelos , Bases de Datos Genéticas , Variación Genética , Humanos , Modelos Moleculares , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Fenotipo
5.
Ann Neurol ; 62(6): 597-608, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17705262

RESUMEN

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.


Asunto(s)
Actinas/genética , Enfermedades Musculares/genética , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Adaptación Fisiológica , Animales , Animales Modificados Genéticamente , Línea Celular , Citoplasma/metabolismo , Drosophila , Humanos , Metionina , Ratones , Debilidad Muscular/etiología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/complicaciones , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Isoformas de Proteínas/metabolismo , Regeneración , Sarcómeros/patología , Transfección , Valina
6.
Invert Neurosci ; 8(2): 63-70, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18443837

RESUMEN

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.


Asunto(s)
Drosophila/fisiología , Ergometría , Músculos/fisiología , Mutación , Unión Neuromuscular/fisiología , Animales , Conducta Animal , Drosophila/genética , Ergometría/instrumentación , Ergometría/métodos , Octopamina/genética
7.
BMC Biol ; 4: 17, 2006 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-16768790

RESUMEN

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.


Asunto(s)
Drosophila/fisiología , Unión Neuromuscular/anatomía & histología , Octopamina/fisiología , Oviposición/fisiología , Tiramina/fisiología , Animales , Drosophila/anatomía & histología , Femenino , Contracción Muscular , Músculos/anatomía & histología , Músculos/inervación , Unión Neuromuscular/fisiología , Ovario/anatomía & histología , Ovario/inervación
8.
Gene ; 631: 16-28, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28739398

RESUMEN

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.


Asunto(s)
Actinas/genética , Contracción Muscular/genética , Miosinas/genética , Actinas/metabolismo , Animales , Drosophila , Proteínas de Drosophila/genética , Perfilación de la Expresión Génica , Masculino , Músculo Estriado/fisiología , Mutación , Miosinas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos
9.
J Mol Biol ; 347(5): 889-94, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15784249

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/metabolismo , Ácido Glutámico/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/genética , Actinas/genética , Actinas/ultraestructura , Animales , Calcio/metabolismo , Calcio/farmacología , Drosophila melanogaster , Ácido Glutámico/genética , Microscopía Electrónica , Mutación/genética
10.
Genetics ; 164(1): 209-22, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12750333

RESUMEN

The indirect flight muscles (IFM) of Drosophila melanogaster provide a good genetic system with which to investigate muscle function. Flight muscle contraction is regulated by both stretch and Ca(2+)-induced thin filament (actin + tropomyosin + troponin complex) activation. Some mutants in troponin-I (TnI) and troponin-T (TnT) genes cause a "hypercontraction" muscle phenotype, suggesting that this condition arises from defects in Ca(2+) regulation and actomyosin-generated tension. We have tested the hypothesis that missense mutations of the myosin heavy chain gene, Mhc, which suppress the hypercontraction of the TnI mutant held-up(2) (hdp(2)), do so by reducing actomyosin force production. Here we show that a "headless" Mhc transgenic fly construct that reduces the myosin head concentration in the muscle thick filaments acts as a dose-dependent suppressor of hypercontracting alleles of TnI, TnT, Mhc, and flightin genes. The data suggest that most, if not all, mutants causing hypercontraction require actomyosin-produced forces to do so. Whether all Mhc suppressors act simply by reducing the force production of the thick filament is discussed with respect to current models of myosin function and thin filament activation by the binding of calcium to the troponin complex.


Asunto(s)
Drosophila melanogaster/genética , Contracción Muscular/genética , Músculos/metabolismo , Cadenas Pesadas de Miosina/genética , Animales , Drosophila melanogaster/metabolismo , Contracción Muscular/fisiología , Cadenas Pesadas de Miosina/metabolismo
11.
Neuromuscul Disord ; 13(7-8): 519-31, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12921789

RESUMEN

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.


Asunto(s)
Actinas/genética , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Mutación , Actinas/química , Secuencia de Aminoácidos , Animales , Variación Genética , Humanos , Proteínas Musculares/metabolismo , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Fenotipo , Relación Estructura-Actividad
12.
J Mol Biol ; 419(1-2): 22-40, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22370558

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Mutación Missense , Miofibrillas/metabolismo , Subfragmentos de Miosina/genética , Subfragmentos de Miosina/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Drosophila/química , Drosophila melanogaster , Filaminas , Vuelo Animal/fisiología , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Contracción Muscular , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miofibrillas/química , Miofibrillas/genética , Miofibrillas/ultraestructura , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Subfragmentos de Miosina/química , Fenotipo , Fosforilación/genética
13.
14.
Neuromuscul Disord ; 20(6): 363-74, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20452215

RESUMEN

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.


Asunto(s)
Actinas/genética , Drosophila/fisiología , Vuelo Animal/fisiología , Músculo Esquelético/fisiología , Músculos/fisiología , Enfermedades Musculares/genética , Actinas/fisiología , Animales , Western Blotting , ADN/genética , Humanos , Microscopía Confocal , Microscopía Electrónica , Modelos Moleculares , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculos/metabolismo , Músculos/patología , Enfermedades Musculares/congénito , Enfermedades Musculares/patología , Mutación , Miofibrillas/patología , Pupa
15.
Neuromuscul Disord ; 20(8): 531-3, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20621480

RESUMEN

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.


Asunto(s)
Citoesqueleto de Actina/genética , Enfermedades Fetales/genética , Proteínas de Microfilamentos/genética , Enfermedades Neuromusculares/genética , Citoesqueleto de Actina/patología , Adulto , ADN/genética , Femenino , Enfermedades Fetales/patología , Humanos , Recién Nacido , Músculo Esquelético/patología , Mutación/genética , Mutación/fisiología , Enfermedades Neuromusculares/patología , Embarazo , Sarcómeros/genética
16.
Fly (Austin) ; 1(2): 68-74, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18820447

RESUMEN

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.


Asunto(s)
Drosophila melanogaster/fisiología , Contracción Muscular/fisiología , Animales , Ergometría/instrumentación , Extremidades/fisiología , Femenino , Proteínas Musculares/genética , Mutación , Esfuerzo Físico/fisiología
17.
J Muscle Res Cell Motil ; 24(8): 489-98, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14870964

RESUMEN

The potential to explore myosin function through the alternative exons and mutations of the single muscle myosin heavy chain gene, Mhc of Drosophila requires detailed kinetic analysis of the myosins. We have obtained microgram quantities of enzymatically active Drosophila myosin and subfragment 1 (S1) from dissected indirect flight muscles. Using recent developments in stopped-flow and flash-photolysis methods combined with fluorescent/light scattering technologies we have determined some of the key kinetic parameters of actin-myosin and myosin-nucleotide interactions. The rate of ATP-induced dissociation of actin from Drosophila myosin (0.23 microM(-1) s(-1)) and subfragment 1 (S1, 0.82 microM(-1) s(-1)) are both fast and similar to values measured for mammalian skeletal muscle myosins and S1 fragments respectively. The ATP-induced cross bridge dissociation of Drosophila acto.S1 is expected to be fast since, for a rapidly contracting muscle like the Drosophila flight muscle, the post power stroke cross bridge must detach rapidly from actin or become a drag on the contracting filament. ATP-induced detachment is preceded by ADP release and this is proposed as the rate-limiting step that defines muscle shortening velocity. We show that the affinity of ADP for acto.S1 at 400 microM is 2-3 fold weaker than fast vertebrate myosins. This leads to an estimate of the ADP release rate constant of 4000 s(-1). We show that this predicts a maximum shortening velocity very similar to that obtained from in vivo estimates of indirect flight muscle shortening. The data is therefore compatible with ADP dissociation limiting the in vivo shortening velocity.


Asunto(s)
Actomiosina/metabolismo , Drosophila/enzimología , Músculos/enzimología , Subfragmentos de Miosina/metabolismo , Miosinas/metabolismo , Actinas/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Unión Proteica , Conejos
18.
J Cell Sci ; 117(Pt 9): 1795-805, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15075240

RESUMEN

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.


Asunto(s)
Drosophila melanogaster/metabolismo , Desarrollo de Músculos/fisiología , Sarcómeros/metabolismo , Sarcómeros/ultraestructura , Troponina I/metabolismo , Alas de Animales/crecimiento & desarrollo , Actinas/genética , Actomiosina/genética , Actomiosina/metabolismo , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/ultraestructura , Expresión Génica , Genes de Insecto/genética , Contracción Muscular/genética , Mutación/genética , Miosinas/genética , Tropomiosina/genética , Tropomiosina/metabolismo , Troponina I/deficiencia , Troponina I/genética , Alas de Animales/ultraestructura
19.
J Exp Biol ; 207(Pt 20): 3515-22, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15339947

RESUMEN

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.


Asunto(s)
Drosophila melanogaster/fisiología , Locomoción/fisiología , Modelos Biológicos , Músculo Esquelético/fisiología , Octopamina/metabolismo , Animales , Fenómenos Biomecánicos , Drosophila melanogaster/genética , Estimulación Eléctrica , Femenino , Músculo Esquelético/metabolismo , Octopamina/genética
20.
News Physiol Sci ; 17: 213-8, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12270959

RESUMEN

Muscle myosin II is an ATP-driven, actin-based molecular motor. Recent developments in optical tweezers technology have made it possible to study movement and force production on the single-molecule level and to find out how different myosin isoforms may have adapted to their specific physiological roles.


Asunto(s)
Músculos/fisiología , Miosinas/fisiología , Adenosina Trifosfato/fisiología , Animales , Humanos , Contracción Muscular/fisiología , Músculo Liso Vascular/fisiología , Miosinas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA