Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Dev Biol ; 470: 95-107, 2021 02.
Article En | MEDLINE | ID: mdl-33245870

During heart formation, the heart grows and undergoes dramatic morphogenesis to achieve efficient embryonic function. Both in fish and amniotes, much of the growth occurring after initial heart tube formation arises from second heart field (SHF)-derived progenitor cell addition to the arterial pole, allowing chamber formation. In zebrafish, this process has been extensively studied during embryonic life, but it is unclear how larval cardiac growth occurs beyond 3 days post-fertilisation (dpf). By quantifying zebrafish myocardial growth using live imaging of GFP-labelled myocardium we show that the heart grows extensively between 3 and 5 dpf. Using methods to assess cell division, cellular development timing assay and Kaede photoconversion, we demonstrate that proliferation, CM addition, and hypertrophy contribute to ventricle growth. Mechanistically, we show that reduction in Mef2c activity (mef2ca+/-;mef2cb-/-), downstream or in parallel with Nkx2.5 and upstream of Ltbp3, prevents some CM addition and differentiation, resulting in a significantly smaller ventricle by 3 dpf. After 3 dpf, however, CM addition in mef2ca+/-;mef2cb-/- mutants recovers to a normal pace, and the heart size gap between mutants and their siblings diminishes into adulthood. Thus, as in mice, there is an early time window when SHF contribution to the myocardium is particularly sensitive to loss of Mef2c activity.


Heart Ventricles/embryology , Heart/embryology , MEF2 Transcription Factors/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/physiology , Zebrafish Proteins/metabolism , Animals , Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , MEF2 Transcription Factors/genetics , Muscle Proteins/genetics , Mutation , Organ Size , Organogenesis , Zebrafish , Zebrafish Proteins/genetics
2.
Development ; 147(8)2020 04 28.
Article En | MEDLINE | ID: mdl-32345657

Skeletal muscle derives from dorsal mesoderm formed during vertebrate gastrulation. Fibroblast growth factor (Fgf) signalling cooperates with Tbx transcription factors to promote dorsal mesoderm formation, but their role in myogenesis has been unclear. Using zebrafish, we show that dorsally derived Fgf signals act through Tbx16 and Tbxta to induce slow and fast trunk muscle precursors at distinct dorsoventral positions. Tbx16 binds to and directly activates the myf5 and myod genes, which are required for commitment to myogenesis. Tbx16 activity depends on Fgf signalling from the organiser. In contrast, Tbxta is not required for myf5 expression, but binds a specific site upstream of myod that is not bound by Tbx16 and drives (dependent on Fgf signals) myod expression in adaxial slow precursors, thereby initiating trunk myogenesis. After gastrulation, when similar muscle cell populations in the post-anal tail are generated from tailbud, declining Fgf signalling is less effective at initiating adaxial myogenesis, which is instead initiated by Hedgehog signalling from the notochord. Our findings suggest a hypothesis for ancestral vertebrate trunk myogenic patterning and how it was co-opted during tail evolution to generate similar muscle by new mechanisms.This article has an associated 'The people behind the papers' interview.


Fibroblast Growth Factors/metabolism , Muscle Development , MyoD Protein/metabolism , T-Box Domain Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Body Patterning/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Muscle Development/genetics , MyoD Protein/genetics , Signal Transduction , T-Box Domain Proteins/genetics , Transcription, Genetic , Up-Regulation/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics
3.
FASEB J ; 33(8): 9116-9130, 2019 08.
Article En | MEDLINE | ID: mdl-31100023

Tendons are an essential part of the musculoskeletal system, connecting muscle and skeletal elements to enable force generation. The transcription factor scleraxis marks vertebrate tendons from early specification. Scleraxis-null mice are viable and have a range of tendon and bone defects in the trunk and limbs but no described cranial phenotype. We report the expression of zebrafish scleraxis orthologs: scleraxis homolog (scx)-a and scxb in cranial and intramuscular tendons and in other skeletal elements. Single mutants for either scxa or scxb, generated by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), are viable and fertile as adult fish. Although scxb mutants show no obvious phenotype, scxa mutant embryos have defects in cranial tendon maturation and muscle misalignment. Mutation of both scleraxis genes results in more severe defects in cranial tendon differentiation, muscle and cartilage dysmorphogenesis and paralysis, and lethality by 2-5 wk, which indicates an essential function of scleraxis for craniofacial development. At juvenile and adult stages, ribs in scxa mutants fail to mineralize and/or are small and heavily fractured. Scxa mutants also have smaller muscle volume, abnormal swim movement, and defects in bone growth and composition. Scleraxis function is therefore essential for normal craniofacial form and function and vital for fish development.-Kague, E., Hughes, S. M., Lawrence, E. A., Cross, S., Martin-Silverstone, E., Hammond, C. L., Hinits, Y. Scleraxis genes are required for normal musculoskeletal development and for rib growth and mineralization in zebrafish.


Basic Helix-Loop-Helix Transcription Factors/genetics , Musculoskeletal Development/genetics , Zebrafish Proteins/genetics , Zebrafish/growth & development , Zebrafish/genetics , Animals , Animals, Genetically Modified , Bone Development/genetics , Calcification, Physiologic/genetics , Gene Expression Regulation, Developmental , Mutation , Ribs/abnormalities , Ribs/growth & development , Ribs/metabolism , Tendons/abnormalities , Tendons/growth & development , Tendons/metabolism , Zebrafish/metabolism
4.
Nat Commun ; 9(1): 4232, 2018 10 12.
Article En | MEDLINE | ID: mdl-30315160

Each skeletal muscle acquires its unique size before birth, when terminally differentiating myocytes fuse to form a defined number of multinucleated myofibres. Although mice in which the transcription factor Myogenin is mutated lack most myogenesis and die perinatally, a specific cell biological role for Myogenin has remained elusive. Here we report that loss of function of zebrafish myog prevents formation of almost all multinucleated muscle fibres. A second, Myogenin-independent, fusion pathway in the deep myotome requires Hedgehog signalling. Lack of Myogenin does not prevent terminal differentiation; the smaller myotome has a normal number of myocytes forming more mononuclear, thin, albeit functional, fast muscle fibres. Mechanistically, Myogenin binds to the myomaker promoter and is required for expression of myomaker and other genes essential for myocyte fusion. Adult myog mutants display reduced muscle mass, decreased fibre size and nucleation. Adult-derived myog mutant myocytes show persistent defective fusion ex vivo. Myogenin is therefore essential for muscle homeostasis, regulating myocyte fusion to determine both muscle fibre number and size.


RNA, Messenger/metabolism , Zebrafish/metabolism , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Female , Male , Muscle Cells/cytology , Muscle Cells/metabolism , Myogenin/metabolism , NADH Tetrazolium Reductase/metabolism , Reverse Transcriptase Polymerase Chain Reaction
5.
PLoS Biol ; 11(10): e1001679, 2013 Oct.
Article En | MEDLINE | ID: mdl-24143132

Muscle fiber size is activity-dependent and clinically important in ageing, bed-rest, and cachexia, where muscle weakening leads to disability, prolonged recovery times, and increased costs. Inactivity causes muscle wasting by triggering protein degradation and may simultaneously prevent protein synthesis. During development, muscle tissue grows by several mechanisms, including hypertrophy of existing fibers. As in other tissues, the TOR pathway plays a key role in promoting muscle protein synthesis by inhibition of eIF4EBPs (eukaryotic Initiation Factor 4E Binding Proteins), regulators of the translational initiation. Here, we tested the role of TOR-eIF4EBP in a novel zebrafish muscle inactivity model. Inactivity triggered up-regulation of eIF4EBP3L (a zebrafish homolog of eIF4EBP3) and diminished myosin and actin content, myofibrilogenesis, and fiber growth. The changes were accompanied by preferential reduction of the muscle transcription factor Mef2c, relative to Myod and Vinculin. Polysomal fractionation showed that Mef2c decrease was due to reduced translation of mef2ca mRNA. Loss of Mef2ca function reduced normal muscle growth and diminished the reduction in growth caused by inactivity. We identify eIF4EBP3L as a key regulator of Mef2c translation and protein level following inactivity; blocking eIF4EBP3L function increased Mef2ca translation. Such blockade also prevented the decline in mef2ca translation and level of Mef2c and slow myosin heavy chain proteins caused by inactivity. Conversely, overexpression of active eIF4EBP3L mimicked inactivity by decreasing the proportion of mef2ca mRNA in polysomes, the levels of Mef2c and slow myosin heavy chain, and myofibril content. Inhibiting the TOR pathway without the increase in eIF4EBP3L had a lesser effect on myofibrilogenesis and muscle size. These findings identify eIF4EBP3L as a key TOR-dependent regulator of muscle fiber size in response to activity. We suggest that by selectively inhibiting translational initiation of mef2ca and other mRNAs, eIF4EBP3L reprograms the translational profile of muscle, enabling it to adjust to new environmental conditions.


Carrier Proteins/metabolism , MEF2 Transcription Factors/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/genetics , Peptide Chain Initiation, Translational/genetics , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Animals , Carrier Proteins/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , MEF2 Transcription Factors/metabolism , Models, Biological , Myofibrils/metabolism , Myogenic Regulatory Factors/metabolism , Myosin Heavy Chains/metabolism , Up-Regulation , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Development ; 140(14): 2972-84, 2013 Jul.
Article En | MEDLINE | ID: mdl-23760954

Striated muscles that enable mouth opening and swallowing during feeding are essential for efficient energy acquisition, and are likely to have played a fundamental role in the success of early jawed vertebrates. The developmental origins and genetic requirements of these muscles are uncertain. Here, we determine by indelible lineage tracing in mouse that fibres of sternohyoid muscle (SHM), which is essential for mouth opening during feeding, and oesophageal striated muscle (OSM), which is crucial for voluntary swallowing, arise from Pax3-expressing somite cells. In vivo Kaede lineage tracing in zebrafish reveals the migratory route of cells from the anteriormost somites to OSM and SHM destinations. Expression of pax3b, a zebrafish duplicate of Pax3, is restricted to the hypaxial region of anterior somites that generate migratory muscle precursors (MMPs), suggesting that Pax3b plays a role in generating OSM and SHM. Indeed, loss of pax3b function led to defective MMP migration and OSM formation, disorganised SHM differentiation, and inefficient ingestion and swallowing of microspheres. Together, our data demonstrate Pax3-expressing somite cells as a source of OSM and SHM fibres, and highlight a conserved role of Pax3 genes in the genesis of these feeding muscles of vertebrates.


Esophagus/embryology , Jaw/embryology , Muscle Development , Muscle, Striated/embryology , Paired Box Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Movement , Deglutition , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Esophagus/cytology , Fetus/cytology , Fetus/metabolism , Jaw/cytology , Mice , Muscle, Striated/cytology , Muscle, Striated/metabolism , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Somites/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
7.
Dev Biol ; 369(2): 199-210, 2012 Sep 15.
Article En | MEDLINE | ID: mdl-22750409

Mef2 transcription factors have been strongly linked with early heart development. D-mef2 is required for heart formation in Drosophila, but whether Mef2 is essential for vertebrate cardiomyocyte (CM) differentiation is unclear. In mice, although Mef2c is expressed in all CMs, targeted deletion of Mef2c causes lethal loss of second heart field (SHF) derivatives and failure of cardiac looping, but first heart field CMs can differentiate. Here we examine Mef2 function in early heart development in zebrafish. Two Mef2c genes exist in zebrafish, mef2ca and mef2cb. Both are expressed similarly in the bilateral heart fields but mef2cb is strongly expressed in the heart poles at the primitive heart tube stage. By using fish mutants for mef2ca and mef2cb and antisense morpholinos to knock down either or both Mef2cs, we show that Mef2ca and Mef2cb have essential but redundant roles in myocardial differentiation. Loss of both Mef2ca and Mef2cb function does not interfere with early cardiogenic markers such as nkx2.5, gata4 and hand2 but results in a dramatic loss of expression of sarcomeric genes and myocardial markers such as bmp4, nppa, smyd1b and late nkx2.5 mRNA. Rare residual CMs observed in mef2ca;mef2cb double mutants are ablated by a morpholino capable of knocking down other Mef2s. Mef2cb over-expression activates bmp4 within the cardiogenic region, but no ectopic CMs are formed. Surprisingly, anterior mesoderm and other tissues become skeletal muscle. Mef2ca single mutants have delayed heart development, but form an apparently normal heart. Mef2cb single mutants have a functional heart and are viable adults. Our results show that the key role of Mef2c in myocardial differentiation is conserved throughout the vertebrate heart.


Muscle Proteins/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myogenic Regulatory Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Base Sequence , Cell Differentiation/genetics , DNA Primers/genetics , Gene Expression Regulation, Developmental , Heart/embryology , Muscle Development/genetics , Mutation , Myocardium/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Mol Cell Endocrinol ; 361(1-2): 51-68, 2012 Sep 25.
Article En | MEDLINE | ID: mdl-22483947

Myostatin (MSTN) is a negative regulator of skeletal muscle growth. In contrast to mammals, fish possess at least two paralogs of MSTN: MSTN-1 and MSTN-2. In this study, we analyzed the structural-functional features of the four variants of Sparus aurata MSTN-2 5'-flanking region: saMSTN-2a, saMSTN-2as, saMSTN-2b and saMSTN-2c. In silico analysis revealed numerous putative cis regulatory elements including several E-boxes known as binding sites to myogenic transcription factors. Transient transfection experiments using non-muscle and muscle cell lines showed surprisingly high transcriptional activity in muscle cells, suggesting the presence of regulatory elements unique to differentiated myotubes. These observations were confirmed by in situ intramuscular injections of promoter DNA followed by reporter gene assays. Moreover, high promoter activity was found in differentiated neural cell, in agreement with MSTN-2 expression in brain. Progressive 5'-deletion analysis, using reporter gene assays, showed that the core promoter is located within the first -127 bp upstream of the ATG, and suggested the presence of regulatory elements that either repress or induce transcriptional activity. Transient transgenic zebrafish provided evidence for saMSTN-2 promoter ability to direct GFP expression to myofibers. Finally, our data shows that although no mature saMSTN-2 mRNA is observed in muscle; unspliced forms accumulate, confirming high level of transcription. In conclusion, our study shows for the first time that MSTN-2 promoter is a very robust promoter, especially in muscle cells.


Alleles , Gene Expression Regulation , Muscles/metabolism , Myostatin/genetics , Organ Specificity/genetics , Promoter Regions, Genetic , Sea Bream/genetics , Alternative Splicing/genetics , Animals , Animals, Genetically Modified , Aquatic Organisms/genetics , Base Sequence , Binding Sites , Cell Line , Conserved Sequence/genetics , Green Fluorescent Proteins/metabolism , Mice , Molecular Sequence Data , Phylogeny , Rats , Response Elements/genetics , Sequence Deletion/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transgenes/genetics , Zebrafish
9.
Dev Biol ; 357(1): 108-16, 2011 Sep 01.
Article En | MEDLINE | ID: mdl-21741963

The forelimbs of higher vertebrates are composed of two portions: the appendicular region (stylopod, zeugopod and autopod) and the less prominent proximal girdle elements (scapula and clavicle) that brace the limb to the main trunk axis. We show that the formation of the muscles of the proximal limb occurs through two distinct mechanisms. The more superficial girdle muscles (pectoral and latissimus dorsi) develop by the "In-Out" mechanism whereby migration of myogenic cells from the somites into the limb bud is followed by their extension from the proximal limb bud out onto the thorax. In contrast, the deeper girdle muscles (e.g. rhomboideus profundus and serratus anterior) are induced by the forelimb field which promotes myotomal extension directly from the somites. Tbx5 inactivation demonstrated its requirement for the development of all forelimb elements which include the skeletal elements, proximal and distal muscles as well as the sternum in mammals and the cleithrum of fish. Intriguingly, the formation of the diaphragm musculature is also dependent on the Tbx5 programme. These observations challenge our classical views of the boundary between limb and trunk tissues. We suggest that significant structures located in the body should be considered as components of the forelimb.


Body Patterning , Forelimb/embryology , Muscle, Skeletal/embryology , Animals , Chick Embryo , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Forelimb/cytology , Mice , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/cytology , Somites/cytology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics
10.
Dev Biol ; 358(1): 102-12, 2011 Oct 01.
Article En | MEDLINE | ID: mdl-21798255

Myogenic regulatory factors of the myod family (MRFs) are transcription factors essential for mammalian skeletal myogenesis. Here we show that a mutation in the zebrafish myod gene delays and reduces early somitic and pectoral fin myogenesis, reduces miR-206 expression, and leads to a persistent reduction in somite size until at least the independent feeding stage. A mutation in myog, encoding a second MRF, has little obvious phenotype at early stages, but exacerbates the loss of somitic muscle caused by lack of Myod. Mutation of both myod and myf5 ablates all skeletal muscle. Haploinsufficiency of myod leads to reduced embryonic somite muscle bulk. Lack of Myod causes a severe reduction in cranial musculature, ablating most muscles including the protractor pectoralis, a putative cucullaris homologue. This phenotype is accompanied by a severe dysmorphology of the cartilaginous skeleton and failure of maturation of several cranial bones, including the opercle. As myod expression is restricted to myogenic cells, the data show that myogenesis is essential for proper skeletogenesis in the head.


Bone and Bones/embryology , Gene Expression Regulation, Developmental/physiology , Haploinsufficiency/genetics , Muscle Development/physiology , MyoD Protein/genetics , Skull/embryology , Zebrafish/embryology , Animals , Cartilage/embryology , Haploinsufficiency/physiology , Immunohistochemistry , In Situ Hybridization , Larva/physiology , Muscle, Skeletal/embryology , Mutation/genetics , MyoD Protein/metabolism , Upper Extremity/embryology , Zebrafish/genetics
11.
Dev Biol ; 350(2): 464-75, 2011 Feb 15.
Article En | MEDLINE | ID: mdl-21147088

Differentiation often requires conversion of analogue signals to a stable binary output through positive feedback. Hedgehog (Hh) signalling promotes myogenesis in the vertebrate somite, in part by raising the activity of muscle regulatory factors (MRFs) of the Myod family above a threshold. Hh is known to enhance MRF expression. Here we show that Hh is also essential at a second step that increases Myod protein activity, permitting it to promote Myogenin expression. Hh acts by inducing expression of cdkn1c (p57(Kip2)) in slow muscle precursor cells, but neither Hh nor Cdkn1c is required for their cell cycle exit. Cdkn1c co-operates with Myod to drive differentiation of several early zebrafish muscle fibre types. Myod in turn up-regulates cdkn1c, thereby providing a positive feedback loop that switches myogenic cells to terminal differentiation.


Cyclin-Dependent Kinase Inhibitor p57/physiology , Muscle Development , MyoD Protein/physiology , Animals , Cell Differentiation , Feedback, Physiological , Hedgehog Proteins/physiology , Myogenic Regulatory Factor 5/physiology , Myogenin/genetics , Signal Transduction , Zebrafish
12.
Development ; 136(3): 403-14, 2009 Feb.
Article En | MEDLINE | ID: mdl-19141670

Myogenic regulatory factors of the Myod family (MRFs) are transcription factors essential for mammalian skeletal myogenesis. However, the roles of each gene in myogenesis remain unclear, owing partly to genetic linkage at the Myf5/Mrf4 locus and to rapid morphogenetic movements in the amniote somite. In mice, Myf5 is essential for the earliest epaxial myogenesis, whereas Myod is required for timely differentiation of hypaxially derived muscle. A second major subdivision of the somite is between primaxial muscle of the somite proper and abaxial somite-derived migratory muscle precursors. Here, we use a combination of mutant and morphant analysis to ablate the function of each of the four conserved MRF genes in zebrafish, an organism that has retained a more ancestral bodyplan. We show that a fundamental distinction in somite myogenesis is into medial versus lateral compartments, which correspond to neither epaxial/hypaxial nor primaxial/abaxial subdivisions. In the medial compartment, Myf5 and/or Myod drive adaxial slow fibre and medial fast fibre differentiation. Myod-driven Myogenin activity alone is sufficient for lateral fast somitic and pectoral fin fibre formation from the lateral compartment, as well as for cranial myogenesis. Myogenin activity is a significant contributor to fast fibre differentiation. Mrf4 does not contribute to early myogenesis in zebrafish. We suggest that the differential use of duplicated MRF paralogues in this novel two-component myogenic system facilitated the diversification of vertebrates.


Extremities/physiology , Muscle Development/physiology , Muscle Fibers, Skeletal/physiology , Somites/physiology , Zebrafish/physiology , Animals , Animals, Genetically Modified , Body Patterning/physiology , Extremities/embryology , Extremities/growth & development , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Larva , Mutation , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myogenin/genetics , Myogenin/metabolism , Organ Specificity , Somites/embryology , Zebrafish/embryology , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
BMC Dev Biol ; 8: 24, 2008 Feb 28.
Article En | MEDLINE | ID: mdl-18307809

BACKGROUND: During vertebrate head evolution, muscle changes accompanied radical modification of the skeleton. Recent studies have suggested that muscles and their innervation evolve less rapidly than cartilage. The freshwater teleostean zebrafish (Danio rerio) is the most studied actinopterygian model organism, and is sometimes taken to represent osteichthyans as a whole, which include bony fishes and tetrapods. Most work concerning zebrafish cranial muscles has focused on larval stages. We set out to describe the later development of zebrafish head muscles and compare muscle homologies across the Osteichthyes. RESULTS: We describe one new muscle and show that the number of mandibular, hyoid and hypobranchial muscles found in four day-old zebrafish larvae is similar to that found in the adult. However, the overall configuration and/or the number of divisions of these muscles change during development. For example, the undivided adductor mandibulae of early larvae gives rise to the adductor mandibulae sections A0, A1-OST, A2 and Aomega, and the protractor hyoideus becomes divided into dorsal and ventral portions in adults. There is not always a correspondence between the ontogeny of these muscles in the zebrafish and their evolution within the Osteichthyes. All of the 13 mandibular, hyoid and hypobranchial muscles present in the adult zebrafish are found in at least some other living teleosts, and all except the protractor hyoideus are found in at least some extant non-teleost actinopterygians. Of these muscles, about a quarter (intermandibularis anterior, adductor mandibulae, sternohyoideus) are found in at least some living tetrapods, and a further quarter (levator arcus palatini, adductor arcus palatini, adductor operculi) in at least some extant sarcopterygian fish. CONCLUSION: Although the zebrafish occupies a rather derived phylogenetic position within actinopterygians and even within teleosts, with respect to the mandibular, hyoid and hypobranchial muscles it seems justified to consider it an appropriate representative of these two groups. Among these muscles, the three with clear homologues in tetrapods and the further three identified in sarcopterygian fish are particularly appropriate for comparisons of results between the actinopterygian zebrafish and the sarcopterygians.


Biological Evolution , Fishes/embryology , Hyoid Bone/anatomy & histology , Mandible/anatomy & histology , Muscles/anatomy & histology , Muscles/embryology , Zebrafish/embryology , Animals , Extinction, Biological , Fishes/anatomy & histology , Larva/anatomy & histology , Phylogeny , Skull/anatomy & histology , Zebrafish/anatomy & histology
14.
Gene Expr Patterns ; 8(2): 71-8, 2008 Jan.
Article En | MEDLINE | ID: mdl-18042440

Dystrophin/dystrobrevin superfamily proteins play structural and signalling roles at the plasma membrane of many cell types. Defects in them or the associated multiprotein complex cause a range of neuromuscular disorders. Members of the dystrophin branch of the family form heterodimers with members of the dystrobrevin branch, mediated by their coiled-coil domains. To determine which combinations of these proteins might interact during embryonic development, we set out to characterise the gene expression pattern of dystrophin and dystrobrevin family members in zebrafish. gamma-dystrobrevin (dtng), a novel dystrobrevin recently identified in fish, is the predominant form of dystrobrevin in embryonic development. Dtng and dmd (dystrophin) have similar spatial and temporal expression patterns in muscle, where transcripts are localized to the ends of differentiated fibres at the somite borders. Dtng is expressed in the notochord while dmd is expressed in the chordo-neural hinge and then in floor plate and hypochord. In addition, dtng is dynamically expressed in rhombomeres 2 and 4-6 of the hindbrain and in the ventral midbrain. alpha-dystrobrevin (dtna) is expressed widely in the brain with particularly strong expression in the hypothalamus and the telencephalon; drp2 is also expressed widely in the brain. Utrophin expression is found in early pronephros and lateral line development and utrophin and dystrophin are both expressed later in the gut. beta-dystrobrevin (dtnb) is expressed in the pronephric duct and widely at low levels. In summary, we find clear instances of co-expression of dystrophin and dystrobrevin family members in muscle, brain and pronephric duct development and many examples of strong and specific expression of members of one family but not the other, an intriguing finding given the presumed heterodimeric state of these molecules.


Dystrophin-Associated Proteins/metabolism , Dystrophin/metabolism , Gene Expression , Multigene Family , Zebrafish/genetics , Animals , Dystrophin/genetics , Dystrophin-Associated Proteins/genetics , Embryo, Nonmammalian , Expressed Sequence Tags , Immunohistochemistry , RNA, Messenger/metabolism , Tissue Distribution , Zebrafish/embryology
15.
Gene Expr Patterns ; 7(7): 738-45, 2007 Aug.
Article En | MEDLINE | ID: mdl-17638597

Mrf4 (Myf6) is a member of the basic helix-loop-helix (bHLH) myogenic regulatory transcription factor (MRF) family, which also contains Myod, Myf5 and myogenin. Mrf4 is implicated in commitment of amniote cells to skeletal myogenesis and is also abundantly expressed in many adult muscle fibres. The specific role of Mrf4 is unclear both because mrf4 null mice are viable, suggesting redundancy with other MRFs, and because of genetic interactions at the complex mrf4/myf5 locus. We report the cloning and expression of an mrf4 gene from zebrafish, Danio rerio, which shows conservation of linkage to myf5. Mrf4 mRNA accumulates in a subset of terminally differentiated muscle fibres in parallel with myosin protein in the trunk and fin. Although most, possibly all, trunk muscle expresses mrf4, the level of mRNA is dynamically regulated. No expression is detected in muscle precursor cell populations prior to myosin accumulation. Moreover, mrf4 expression is not detected in head muscles, at least at early stages. As fish mature, mrf4 expression is pronounced in the region of slow muscle fibres.


Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/physiology , Amino Acid Sequence , Amnion/cytology , Animals , Cardiac Myosins/biosynthesis , Cardiac Myosins/physiology , Gene Expression Regulation , Models, Biological , Molecular Sequence Data , Muscles/metabolism , Myogenic Regulatory Factor 5/metabolism , Myogenin/metabolism , Myosin Light Chains/biosynthesis , Myosin Light Chains/physiology , Phylogeny , Sequence Homology, Amino Acid , Tissue Distribution , Zebrafish
16.
Development ; 134(13): 2511-9, 2007 Jul.
Article En | MEDLINE | ID: mdl-17537787

During skeletal muscle differentiation, the actomyosin motor is assembled into myofibrils, multiprotein machines that generate and transmit force to cell ends. How expression of muscle proteins is coordinated to build the myofibril is unknown. Here we show that zebrafish Mef2d and Mef2c proteins are required redundantly for assembly of myosin-containing thick filaments in nascent muscle fibres, but not for the earlier steps of skeletal muscle fibre differentiation, elongation, fusion or thin filament gene expression. mef2d mRNA and protein is present in myoblasts, whereas mef2c expression commences in muscle fibres. Knockdown of both Mef2s with antisense morpholino oligonucleotides or in mutant fish blocks muscle function and prevents sarcomere assembly. Cell transplantation and heat-shock-driven rescue reveal a cell-autonomous requirement for Mef2 within fibres. In nascent fibres, Mef2 drives expression of genes encoding thick, but not thin, filament proteins. Among genes analysed, myosin heavy and light chains and myosin-binding protein C require Mef2 for normal expression, whereas actin, tropomyosin and troponin do not. Our findings show that Mef2 controls skeletal muscle formation after terminal differentiation and define a new maturation step in vertebrate skeletal muscle development at which thick filament gene expression is controlled.


Gene Expression Regulation, Developmental , Muscle Fibers, Skeletal/metabolism , Myogenic Regulatory Factors/metabolism , Zebrafish Proteins/metabolism , Animals , Base Sequence , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Heart/embryology , Molecular Sequence Data , Mutation/genetics , Myocardium/metabolism , Myogenic Regulatory Factors/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid , Transcription, Genetic/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
17.
BMC Genomics ; 8: 19, 2007 Jan 17.
Article En | MEDLINE | ID: mdl-17233888

BACKGROUND: Dystrophins and dystrobrevins are distantly related proteins with important but poorly understood roles in the function of metazoan muscular and neuronal tissues. Defects in them and their associated proteins cause a range of neuromuscular disorders. Members of this superfamily have been discovered in a relatively serendipitous way; we set out to compile a comprehensive description of dystrophin- and dystrobrevin-related sequences from available metazoan genome sequences, validated in representative organisms by RT-PCR, or acquired de novo from key species. RESULTS: Features of the superfamily revealed by our survey include: a) Dystrotelin, an entirely novel branch of the superfamily, present in most vertebrates examined. Dystrotelin is expressed in the central nervous system, and is a possible orthologue of Drosophila DAH. We describe the preliminary characterisation of its function, evolution and expression. b) A novel vertebrate member of the dystrobrevin family, gamma-dystrobrevin, an ancient branch now extant only in fish, but probably present in our own ancestors. Like dystrophin, zebrafish gamma-dystrobrevin mRNA is localised to myosepta. c) The extent of conservation of alternative splicing and alternative promoter use in the dystrophin and dystrobrevin genes; alternative splicing of dystrophin exons 73 and 78 and alpha-dystrobrevin exon 13 are conserved across vertebrates, as are the use of the Dp116, Dp71 and G-utrophin promoters; the Dp260 and Dp140 promoters are tetrapod innovations. d) The evolution of the unique N-terminus of DRP2 and its relationship to Dp116 and G-utrophin. e) A C-terminally truncated common ancestor of dystrophin and utrophin in cyclostomes. f) A severely restricted repertoire of dystrophin complex components in ascidians. CONCLUSION: We have refined our understanding of the evolutionary history and isoform diversity of the five previously reported vertebrate superfamily members and describe two novel members, dystrotelin and gamma-dystrobrevin. Dystrotelins, dystrophins and dystrobrevins are roughly equally related to each other. Vertebrates therefore have a repertoire of seven superfamily members (three dystrophins, three dystrobevins, and one dystrotelin), with one lost in tetrapods. Most invertebrates studied have one member from each branch. Although the basic shared function which is implied by the common architecture of these distantly related proteins remains unclear, it clearly permeates metazoan biology.


Drosophila Proteins/chemistry , Dystrophin-Associated Proteins/chemistry , Dystrophin/chemistry , Membrane Proteins/chemistry , Protein Isoforms , Amino Acid Sequence , Animals , Drosophila , Dystrophin/genetics , Expressed Sequence Tags , Humans , Mice , Molecular Sequence Data , Multigene Family , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Zebrafish
18.
Dev Biol ; 302(2): 504-21, 2007 Feb 15.
Article En | MEDLINE | ID: mdl-17094960

Pax3/7 paired homeodomain transcription factors are important markers of muscle stem cells. Pax3 is required upstream of myod for lateral dermomyotomal cells in the amniote somite to form particular muscle cells. Later Pax3/7-dependent cells generate satellite cells and most body muscle. Here we analyse early myogenesis from, and regulation of, a population of Pax3-expressing dermomyotome-like cells in the zebrafish. Zebrafish pax3 is widely expressed in the lateral somite and, along with pax7, becomes restricted anteriorly and then to the external cells on the lateral somite surface. Midline-derived Hedgehog signals appear to act directly on lateral somite cells to repress Pax3/7. Both Hedgehog and Fgf8, signals that induce muscle formation within the somite, suppress Pax3/7 and promote expression of myogenic regulatory factors (MRFs) myf5 and myod in specific muscle precursor cell populations. Loss of MRF function leads to loss of myogenesis by specific populations of muscle fibres, with parallel up-regulation of Pax3/7. Myod is required for lateral fast muscle differentiation from pax3-expressing cells. In contrast, either Myf5 or Myod is sufficient to promote slow muscle formation from adaxial cells. Thus, myogenic signals act to drive somite cells to a myogenic fate through up-regulation of distinct combinations of MRFs. Our data show that the relationship between Pax3/7 genes and myogenesis is evolutionarily ancient, but that changes in the MRF targets for particular signals contribute to myogenic differences between species.


Fibroblast Growth Factors/physiology , Hedgehog Proteins/physiology , MyoD Protein/physiology , Myogenic Regulatory Factor 5/physiology , PAX7 Transcription Factor/biosynthesis , Paired Box Transcription Factors/biosynthesis , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/physiology , Zebrafish/physiology , Animals , Cell Proliferation , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Muscle Development , Muscle Fibers, Skeletal/physiology , Mutation , Neural Crest/embryology , Neural Crest/metabolism , PAX3 Transcription Factor , PAX7 Transcription Factor/genetics , Paired Box Transcription Factors/genetics , Signal Transduction , Somites/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
19.
Gene Expr Patterns ; 6(8): 952-63, 2006 Oct.
Article En | MEDLINE | ID: mdl-16750657

Many immunoglobulin (Ig)-superfamily cell adhesion molecules influence skeletal muscle formation. In Drosophila, dumbfounded (duf/kirre), irreC, sticks and stones and hibris encode related Ig-family proteins expressed in subsets of neurons and muscle precursor cells. The family mediates cell migration, axon guidance and fusion of myoblasts. Despite the importance of these genes in invertebrate myogenesis, no obvious functional parallels are known in vertebrate myogenesis. Here we investigate the gene expression pattern and phylogenetic and protein-structural relationships of the duf-related molecules neurolin and neurolin-like cell adhesion molecule (NLCAM), members of the activated leukocyte cell adhesion molecule (ALCAM) sub-family of Ig-molecules. These proteins are among the closest to Duf/Kirre by sequence. During zebrafish development, neurolin is expressed in subsets of somite and muscle cells, heart and numerous sites of neuronal maturation. The new ALCAM-family member, NLCAM, appears to have arisen by duplication of neurolin/ALCAM. NLCAM is expressed widely during gastrulation, particularly in the nascent neural plate, but later becomes predominantly expressed in sites of muscle and nerve maturation and in the fin fold. The expression of each gene is often in groups of cells in similar parts of the embryo; for example, in the region of Rohon Beard neurons, trigeminal ganglion and fusing fast and migrating slow muscle fibres. However, expression can also be distinct and dynamic; for example, muscle pioneer fibres express neurolin but not NLCAM at high level. Both molecules are expressed in subsets of muscle precursors at times prior to fusion.


Activated-Leukocyte Cell Adhesion Molecule/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Activated-Leukocyte Cell Adhesion Molecule/genetics , Amino Acid Sequence , Animals , Cell Adhesion Molecules, Neuronal/genetics , Central Nervous System/embryology , Central Nervous System/metabolism , Embryo, Nonmammalian , Gastrula/metabolism , Gene Duplication , Gene Expression Regulation, Developmental , Molecular Sequence Data , Phylogeny , Sequence Homology, Amino Acid , Somites/metabolism , Tissue Distribution , Zebrafish/genetics
...