Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Front Physiol ; 14: 1176148, 2023.
Article in English | MEDLINE | ID: mdl-37143929

ABSTRACT

Myogenesis is a developmental process that is largely conserved in both Drosophila and higher organisms. Consequently, the fruit fly is an excellent in vivo model for identifying the genes and mechanisms involved in muscle development. Moreover, there is growing evidence indicating that specific conserved genes and signaling pathways govern the formation of tissues that connect the muscles to the skeleton. In this review, we present an overview of the different stages of tendon development, from the specification of tendon progenitors to the assembly of a stable myotendinous junction across three different myogenic contexts in Drosophila: larval, flight and leg muscle development. We underline the different aspects of tendon cell specification and differentiation in embryo and during metamorphosis that result into tendon morphological and functional diversity.

2.
EMBO Rep ; 24(4): e56616, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36852954

ABSTRACT

Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy in adults. It is caused by the excessive expansion of noncoding CTG repeats, which when transcribed affects the functions of RNA-binding factors with adverse effects on alternative splicing, processing, and stability of a large set of muscular and cardiac transcripts. Among these effects, inefficient processing and down-regulation of muscle- and heart-specific miRNA, miR-1, have been reported in DM1 patients, but the impact of reduced miR-1 on DM1 pathogenesis has been unknown. Here, we use Drosophila DM1 models to explore the role of miR-1 in cardiac dysfunction in DM1. We show that miR-1 down-regulation in the heart leads to dilated cardiomyopathy (DCM), a DM1-associated phenotype. We combined in silico screening for miR-1 targets with transcriptional profiling of DM1 cardiac cells to identify miR-1 target genes with potential roles in DCM. We identify Multiplexin (Mp) as a new cardiac miR-1 target involved in DM1. Mp encodes a collagen protein involved in cardiac tube formation in Drosophila. Mp and its human ortholog Col15A1 are both highly enriched in cardiac cells of DCM-developing DM1 flies and in heart samples from DM1 patients with DCM, respectively. When overexpressed in the heart, Mp induces DCM, whereas its attenuation rescues the DCM phenotype of aged DM1 flies. Reduced levels of miR-1 and consecutive up-regulation of its target Mp/Col15A1 might be critical in DM1-associated DCM.


Subject(s)
Cardiomyopathy, Dilated , MicroRNAs , Myotonic Dystrophy , Adult , Animals , Humans , Aged , Myotonic Dystrophy/genetics , Myotonic Dystrophy/pathology , Cardiomyopathy, Dilated/genetics , Heart , MicroRNAs/genetics , MicroRNAs/metabolism , Drosophila/genetics , Drosophila/metabolism
3.
Bio Protoc ; 13(3): e4605, 2023 Feb 05.
Article in English | MEDLINE | ID: mdl-36816994

ABSTRACT

Adult muscle stem cells (MuSCs) show remarkable capability in repairing injured tissues. Studying MuSCs in suitable model organisms, which show strong homology with vertebrate counterparts, helps in dissecting the mechanisms regulating their behavior. Additionally, ease of handling and access to technological tools make model organisms well suited for studying biological processes that are conserved across species. MuSCs quiescence, proliferation, and migration are regulated by various input of signals from the surrounding tissues that constitute the MuSCs niche. Observing MuSCs along with their niche in vivo through live imaging provides key information on how MuSCs behave in quiescent and activated states. Drosophila melanogaster is well known for its genetic tool arsenal and the similarity of its different biological processes with vertebrates. Hence, it is widely used to study different types of stem cells. Gained knowledge could then be extrapolated to the vertebrate/mammalian homologous systems to enhance our knowledge in stem cell fields. In this protocol, we discuss how to perform live cell imaging of Drosophila MuSCs, called adult muscle precursors (AMPs) at embryonic stages, using dual-color labelling to visualize both AMPs and the surrounding tissues. This dual-color fluorescent labelling enables the observation of in vivo behavior of two types of cells simultaneously and provides key information on their interactions. The originality of this protocol resides in its biological application to MuSCs and their niche.

4.
Exp Cell Res ; 410(1): 112950, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34838813

ABSTRACT

Drosophila embryonic somatic muscles represent a simple and tractable model system to study the gene regulatory networks that control diversification of cell types. Somatic myogenesis in Drosophila is initiated by intrinsic action of the mesodermal master gene twist, which activates a cascade of transcriptional outputs including myogenic differentiation factor Mef2, which triggers all aspects of the myogenic differentiation program. In parallel, the expression of a combinatorial code of identity transcription factors (iTFs) defines discrete particular features of each muscle fiber, such as number of fusion events, and specific attachment to tendon cells or innervation, thus ensuring diversification of muscle types. Here, we take the example of a subset of lateral transverse (LT) muscles and discuss how the iTF code and downstream effector genes progressively define individual LT properties such as fusion program, attachment and innervation. We discuss new challenges in the field including the contribution of posttranscriptional and epitranscriptomic regulation of gene expression in the diversification of cell types.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Embryonic Development/genetics , Muscle Development/genetics , Myogenic Regulatory Factors/genetics , Animals , Cell Differentiation/genetics , Drosophila melanogaster/growth & development , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/genetics , Mesoderm/growth & development , Mesoderm/metabolism , Muscles/metabolism
5.
Cells ; 10(11)2021 11 08.
Article in English | MEDLINE | ID: mdl-34831301

ABSTRACT

The Drosophila heart, also referred to as the dorsal vessel, pumps the insect blood, the hemolymph. The bilateral heart primordia develop from the most dorsally located mesodermal cells, migrate coordinately, and fuse to form the cardiac tube. Though much simpler, the fruit fly heart displays several developmental and functional similarities to the vertebrate heart and, as we discuss here, represents an attractive model system for dissecting mechanisms of cardiac aging and heart failure and identifying genes causing congenital heart diseases. Fast imaging technologies allow for the characterization of heartbeat parameters in the adult fly and there is growing evidence that cardiac dysfunction in human diseases could be reproduced and analyzed in Drosophila, as discussed here for heart defects associated with the myotonic dystrophy type 1. Overall, the power of genetics and unsuspected conservation of genes and pathways puts Drosophila at the heart of fundamental and applied cardiac research.


Subject(s)
Disease Models, Animal , Drosophila/physiology , Heart Diseases/pathology , Heart/embryology , Aging/genetics , Animals , Gene Expression Regulation, Developmental , Heart Diseases/embryology , Heart Diseases/genetics , Humans
6.
Development ; 148(15)2021 08 01.
Article in English | MEDLINE | ID: mdl-34323270

ABSTRACT

The formation of the cardiac tube is a remarkable example of complex morphogenetic processes conserved from invertebrates to humans. It involves coordinated collective migration of contralateral rows of cardiac cells. The molecular processes underlying the specification of cardioblasts (CBs) prior to migration are well established and significant advances have been made in understanding the process of lumen formation. However, the mechanisms of collective cardiac cells migration remain elusive. Here, we have identified CAP and MSP300 as novel actors involved during CB migration. They both exhibit highly similar temporal and spatial expression patterns in Drosophila migrating cardiac cells, and are necessary for the correct number and alignment of CBs, a prerequisite for the coordination of their collective migration. Our data suggest that CAP and MSP300 are part of a protein complex linking focal adhesion sites to nuclei via the actin cytoskeleton that maintains post-mitotic state and correct alignment of CBs.


Subject(s)
Cell Nucleus/metabolism , Cytoskeleton/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Heart/physiology , Myocardium/metabolism , Organogenesis/physiology , Animals , Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology
7.
Int J Mol Sci ; 22(11)2021 May 26.
Article in English | MEDLINE | ID: mdl-34073503

ABSTRACT

Drug-induced myopathies are classified as acquired myopathies caused by exogenous factors. These pathological conditions develop in patients without muscle disease and are triggered by a variety of medicaments, including lipid-lowering drugs (LLDs) such as statins, fibrates, and ezetimibe. Here we summarise the current knowledge gained via studies conducted using various models, such as cell lines and mammalian models, and compare them with the results obtained in zebrafish (Danio rerio) studies. Zebrafish have proven to be an excellent research tool for studying dyslipidaemias as a model of these pathological conditions. This system enables in-vivo characterization of drug and gene candidates to further the understanding of disease aetiology and develop new therapeutic strategies. Our review also considers important environmental issues arising from the indiscriminate use of LLDs worldwide. The widespread use and importance of drugs such as statins and fibrates justify the need for the meticulous study of their mechanism of action and the side effects they cause.


Subject(s)
Fibric Acids/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscular Diseases , Zebrafish/metabolism , Animals , Disease Models, Animal , Fibric Acids/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Muscular Diseases/chemically induced , Muscular Diseases/metabolism , Muscular Diseases/pathology
8.
Sci Rep ; 11(1): 13197, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162956

ABSTRACT

A combinatorial code of identity transcription factors (iTFs) specifies the diversity of muscle types in Drosophila. We previously showed that two iTFs, Lms and Ap, play critical role in the identity of a subset of larval body wall muscles, the lateral transverse (LT) muscles. Intriguingly, a small portion of ap and lms mutants displays an increased number of LT muscles, a phenotype that recalls pathological split muscle fibers in human. However, genes acting downstream of Ap and Lms to prevent these aberrant muscle feature are not known. Here, we applied a cell type specific translational profiling (TRAP) to identify gene expression signatures underlying identity of muscle subsets including the LT muscles. We found that Gelsolin (Gel) and dCryAB, both encoding actin-interacting proteins, displayed LT muscle prevailing expression positively regulated by, the LT iTFs. Loss of dCryAB function resulted in LTs with irregular shape and occasional branched ends also observed in ap and lms mutant contexts. In contrast, enlarged and then split LTs with a greater number of myonuclei formed in Gel mutants while Gel gain of function resulted in unfused myoblasts, collectively indicating that Gel regulates LTs size and prevents splitting by limiting myoblast fusion. Thus, dCryAB and Gel act downstream of Lms and Ap and contribute to preventing LT muscle branching and splitting. Our findings offer first clues to still unknown mechanisms of pathological muscle splitting commonly detected in human dystrophic muscles and causing muscle weakness.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Gelsolin/physiology , Gene Expression Regulation , Genes, Insect , Muscles/ultrastructure , Muscular Dystrophy, Animal/genetics , alpha-Crystallin B Chain/physiology , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Cell Fusion , Cell Shape , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Gelsolin/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Larva , Loss of Function Mutation , Multigene Family , Muscle Cells/metabolism , Muscles/metabolism , Muscular Dystrophy, Animal/pathology , Myoblasts/metabolism , Myoblasts/ultrastructure , RNA, Messenger/metabolism , Transcription Factors/physiology , Transcription, Genetic , alpha-Crystallin B Chain/genetics
9.
Cell Mol Life Sci ; 78(12): 5043-5049, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33861361

ABSTRACT

Neuromuscular system is constituted of multi-fibrillar muscles, tendons, motor neurons and associated muscle stem cells. Stereotyped pattern of muscle innervation and muscle-specific interactions with tendon cells suggest that neuromuscular system develops in a coordinated way. Remarkably, upon regeneration, coordinated assembly of all neuromuscular components is also critical to rebuild functional muscle. Thus, to ensure muscle function, the neuromuscular system components need to interact both during development and regeneration. Over the last decades, interactions between muscles and tendons, muscles and motor neurons and between muscles and muscle stem cells have been extensively analysed and documented. However, only recent evidence indicates that muscle stem cells interact with motor neurons and that these interactions contribute to building functional muscle both during development and regeneration. From this perspective, we discuss here the relationship between muscle stem cells and motor neurons during Drosophila neuromuscular system development and adverse impact of affected muscle stem cell-motor neuron interactions in regenerating vertebrate muscle.


Subject(s)
Motor Neurons/physiology , Muscles/physiology , Neuromuscular Junction/physiology , Regeneration , Stem Cells/physiology , Animals , Humans , Muscles/cytology , Stem Cells/cytology
10.
Front Cell Dev Biol ; 9: 747563, 2021.
Article in English | MEDLINE | ID: mdl-34977007

ABSTRACT

To ensure locomotion and body stability, the active role of muscle contractions relies on a stereotyped muscle pattern set in place during development. This muscle patterning requires a precise assembly of the muscle fibers with the skeleton via a specialized connective tissue, the tendon. Like in vertebrate limbs, Drosophila leg muscles make connections with specific long tendons that extend through different segments. During the leg disc development, cell precursors of long tendons rearrange and collectively migrate to form a tube-shaped structure. A specific developmental program underlies this unique feature of tendon-like cells in the Drosophila model. We provide for the first time a transcriptomic profile of leg tendon precursors through fluorescence-based cell sorting. From promising candidates, we identified the Krüppel-like factor Dar1 as a critical actor of leg tendon development. Specifically expressed in the leg tendon precursors, loss of dar1 disrupts actin-rich filopodia formation and tendon elongation. Our findings show that Dar1 acts downstream of Stripe and is required to set up the correct number of tendon progenitors.

11.
Cells ; 9(6)2020 06 25.
Article in English | MEDLINE | ID: mdl-32630420

ABSTRACT

In the fruit fly, Drosophila melanogaster, the larval somatic muscles or the adult thoracic flight and leg muscles are the major voluntary locomotory organs. They share several developmental and structural similarities with vertebrate skeletal muscles. To ensure appropriate activity levels for their functions such as hatching in the embryo, crawling in the larva, and jumping and flying in adult flies all muscle components need to be maintained in a functionally stable or homeostatic state despite constant strain. This requires that the muscles develop in a coordinated manner with appropriate connections to other cell types they communicate with. Various signaling pathways as well as extrinsic and intrinsic factors are known to play a role during Drosophila muscle development, diversification, and homeostasis. In this review, we discuss genetic control mechanisms of muscle contraction, development, and homeostasis with particular emphasis on the contractile unit of the muscle, the sarcomere.


Subject(s)
Drosophila melanogaster/genetics , Muscle Development/genetics , Animals , Homeostasis
12.
Adv Exp Med Biol ; 1227: 131-144, 2020.
Article in English | MEDLINE | ID: mdl-32072503

ABSTRACT

Notch plays multiple roles both in development and in adult tissue homeostasis. Notch was first identified in Drosophila in which it has then been extensively studied. Among the flag-ship Notch functions we could mention its capacity to keep precursor and stem cells in a nondifferentiated state but also its ability to activate cell proliferation that in some contexts could led to cancer. In general, both these functions involve, canonical, ligand-dependent Notch activation. However, a ligand-independent Notch activation has also been described in a few cellular contexts. Here, we focus on one of such contexts, Drosophila muscle stem cells, called AMPs, and discuss how insulin-dependent noncanonical activation of Notch pushes quiescent AMPs to proliferation.


Subject(s)
Drosophila Proteins/metabolism , Insulin/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Receptors, Notch/metabolism , Animals , Cell Proliferation , Signal Transduction
13.
Development ; 147(4)2020 02 17.
Article in English | MEDLINE | ID: mdl-32001438

ABSTRACT

Despites several decades of studies on the neuromuscular system, the relationship between muscle stem cells and motor neurons remains elusive. Using the Drosophila model, we provide evidence that adult muscle precursors (AMPs), the Drosophila muscle stem cells, interact with the motor axons during embryogenesis. AMPs not only hold the capacity to attract the navigating intersegmental (ISN) and segmental a (SNa) nerve branches, but are also mandatory to the innervation of muscles in the lateral field. This so-far-ignored AMP role involves their filopodia-based interactions with nerve growth cones. In parallel, we report the previously undetected expression of the guidance molecule-encoding genes sidestep and side IV in AMPs. Altogether, our data support the view that Drosophila muscle stem cells represent spatial landmarks for navigating motor neurons and reveal that their positioning is crucial for the muscles innervation in the lateral region. Furthermore, AMPs and motor axons are interdependent, as the genetic ablation of SNa leads to a specific loss of SNa-associated lateral AMPs.


Subject(s)
Axons/physiology , Motor Neurons/physiology , Muscles/embryology , Muscles/innervation , Myoblasts/physiology , Animals , Apoptosis , Axon Guidance , Cell Movement , Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Genotype , Green Fluorescent Proteins , Growth Cones/physiology , Immunohistochemistry , In Situ Hybridization , Membrane Proteins/physiology , Microscopy, Fluorescence , Pseudopodia/physiology , Signal Transduction , Stem Cells/cytology
14.
Elife ; 82019 12 12.
Article in English | MEDLINE | ID: mdl-31829940

ABSTRACT

Cardiac conduction defects decrease life expectancy in myotonic dystrophy type 1 (DM1), a CTG repeat disorder involving misbalance between two RNA binding factors, MBNL1 and CELF1. However, how DM1 condition translates into conduction disorders remains poorly understood. Here we simulated MBNL1 and CELF1 misbalance in the Drosophila heart and performed TU-tagging-based RNAseq of cardiac cells. We detected deregulations of several genes controlling cellular calcium levels, including increased expression of straightjacket/α2δ3, which encodes a regulatory subunit of a voltage-gated calcium channel. Straightjacket overexpression in the fly heart leads to asynchronous heartbeat, a hallmark of abnormal conduction, whereas cardiac straightjacket knockdown improves these symptoms in DM1 fly models. We also show that ventricular α2δ3 expression is low in healthy mice and humans, but significantly elevated in ventricular muscles from DM1 patients with conduction defects. These findings suggest that reducing ventricular straightjacket/α2δ3 levels could offer a strategy to prevent conduction defects in DM1.


Subject(s)
Calcium Channels/biosynthesis , Cardiac Conduction System Disease/genetics , Cardiac Conduction System Disease/physiopathology , Gene Expression Regulation , Myotonic Dystrophy/complications , Animals , Calcium Channels/genetics , Disease Models, Animal , Drosophila , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Humans , Mice
15.
Biol Open ; 8(3)2019 Mar 18.
Article in English | MEDLINE | ID: mdl-30796048

ABSTRACT

Multiple tissue interactions take place during the development of the limb musculoskeletal system. While appendicular myogenesis has been extensively studied, development of connective tissue associated with muscles has received less attention. In the developing Drosophila leg, tendon-like connective tissue arises from clusters of epithelial cells that invaginate into the leg cavity and then elongate to form internal tube-shape structures along which muscle precursors are distributed. Here we show that stripe-positive appendicular precursors of tendon-like connective tissue are set up among intersegmental leg joint cells expressing odd-skipped genes, and that Notch signaling is necessary and locally sufficient to trigger stripe expression. This study also finds that odd-skipped genes and stripe are both required downstream of Notch to promote morphogenesis of tube-shaped internal tendons of the leg.

16.
Int J Mol Sci ; 19(12)2018 Dec 18.
Article in English | MEDLINE | ID: mdl-30567354

ABSTRACT

Myotonic dystrophy type 1 (DM1), the most common cause of adult-onset muscular dystrophy, is autosomal dominant, multisystemic disease with characteristic symptoms including myotonia, heart defects, cataracts and testicular atrophy. DM1 disease is being successfully modelled in Drosophila allowing to identify and validate new pathogenic mechanisms and potential therapeutic strategies. Here we provide an overview of insights gained from fruit fly DM1 models, either: (i) fundamental with particular focus on newly identified gene deregulations and their link with DM1 symptoms; or (ii) applied via genetic modifiers and drug screens to identify promising therapeutic targets.


Subject(s)
Muscle, Skeletal/physiopathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy , Myotonin-Protein Kinase/genetics , Animals , Disease Models, Animal , Drosophila melanogaster/genetics , Drug Evaluation, Preclinical , Humans , Molecular Targeted Therapy , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase/antagonists & inhibitors , Trinucleotide Repeat Expansion/genetics
17.
Int J Mol Sci ; 19(11)2018 Nov 02.
Article in English | MEDLINE | ID: mdl-30400176

ABSTRACT

Heat shock proteins (Hsps) form a large family of evolutionarily conserved molecular chaperones that help balance protein folding and protect cells from various stress conditions. However, there is growing evidence that Hsps may also play an active role in developmental processes. Here, we take the example of developmental expression and function of one class of Hsps characterized by low molecular weight, the small Hsps (sHsps). We discuss recent reports and genome-wide datasets that support vital sHsps functions in the developing nervous system, reproductive system, and muscles. This tissue- and time-specific sHsp expression is developmentally regulated, so that the enhancer sequence of an sHsp gene expressed in developing muscle, in addition to stress-inducible elements, also carries binding sites for myogenic regulatory factors. One possible reason for sHsp genes to switch on during development and in non-stress conditions is to protect vital developing organs from environmental insults.


Subject(s)
Drosophila Proteins/genetics , Drosophila/growth & development , Drosophila/genetics , Gene Expression Regulation, Developmental , Heat-Shock Proteins, Small/genetics , Animals , Drosophila Proteins/metabolism , Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/metabolism , Organ Specificity/genetics
18.
Cell Mol Life Sci ; 75(23): 4341-4356, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30032358

ABSTRACT

The Drosophila Hsp67Bc gene encodes a protein belonging to the small heat-shock protein (sHSP) family, identified as the nearest functional ortholog of human HSPB8. The most prominent activity of sHSPs is preventing the irreversible aggregation of various non-native polypeptides. Moreover, they are involved in processes such as development, aging, maintenance of the cytoskeletal architecture and autophagy. In larval muscles Hsp67Bc localizes to the Z- and A-bands, which suggests its role as part of the conserved chaperone complex required for Z-disk maintenance. In addition, Hsp67Bc is present at neuromuscular junctions (NMJs), which implies its involvement in the maintenance of NMJ structure. Here, we report the effects of muscle-target overexpression of Drosophila Hsp67Bc hot-spot variants Hsp67BcR126E and Hsp67BcR126N mimicking pathogenic variants of human HSPB8. Depending on the substitutions, we observed a different impact on muscle structure and performance. Expression of Hsp67BcR126E affects larval motility, which may be caused by impairment of mitochondrial respiratory function and/or by NMJ abnormalities manifested by a decrease in the number of synaptic boutons. In contrast, Hsp67BcR126N appears to be an aggregate-prone variant, as reflected in excessive accumulation of mutant proteins and the formation of large aggregates with a lesser impact on muscle structure and performance compared to the Hsp67BcR126E variant.


Subject(s)
Drosophila Proteins/genetics , Heat-Shock Proteins/genetics , Muscles/metabolism , Mutation, Missense , Neuromuscular Junction/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression , Heat-Shock Proteins/metabolism , Larva/genetics , Larva/metabolism , Microscopy, Electron, Transmission , Motor Activity/genetics , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure , Sequence Homology, Amino Acid
19.
Dis Model Mech ; 11(5)2018 05 21.
Article in English | MEDLINE | ID: mdl-29716962

ABSTRACT

Steinert disease, or myotonic dystrophy type 1 (DM1), is a multisystemic disorder caused by toxic noncoding CUG repeat transcripts, leading to altered levels of two RNA binding factors, MBNL1 and CELF1. The contribution of CELF1 to DM1 phenotypes is controversial. Here, we show that the Drosophila CELF1 family member, Bru-3, contributes to pathogenic muscle defects observed in a Drosophila model of DM1. Bru-3 displays predominantly cytoplasmic expression in muscles and its muscle-specific overexpression causes a range of phenotypes also observed in the fly DM1 model, including affected motility, fiber splitting, reduced myofiber length and altered myoblast fusion. Interestingly, comparative genome-wide transcriptomic analyses revealed that Bru-3 negatively regulates levels of mRNAs encoding a set of sarcomere components, including Actn transcripts. Conversely, it acts as a positive regulator of Actn translation. As CELF1 displays predominantly cytoplasmic expression in differentiating C2C12 myotubes and binds to Actn mRNA, we hypothesize that it might exert analogous functions in vertebrate muscles. Altogether, we propose that cytoplasmic Bru-3 contributes to DM1 pathogenesis in a Drosophila model by regulating sarcomeric transcripts and protein levels.


Subject(s)
Drosophila Proteins/metabolism , Muscles/metabolism , Myotonic Dystrophy/metabolism , RNA-Binding Proteins/metabolism , Sarcomeres/metabolism , Animals , Cell Fusion , Cell Line , Drosophila melanogaster/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Larva/metabolism , Mice , Models, Biological , Movement , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscles/pathology , Muscles/physiopathology , Myoblasts/metabolism , Myotonic Dystrophy/pathology , Myotonic Dystrophy/physiopathology , Phenotype , Protein Binding , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Development ; 145(2)2018 01 17.
Article in English | MEDLINE | ID: mdl-29247145

ABSTRACT

The Drosophila heart, composed of discrete subsets of cardioblasts and pericardial cells, undergoes Hox-triggered anterior-posterior morphogenesis, leading to a functional subdivision into heart proper and aorta, with its most anterior part forming a funnel-shaped cardiac outflow. Cardioblasts differentiate into Tin-positive 'working myocytes' and Svp-expressing ostial cells. However, developmental fates and functions of heart-associated pericardial cells remain elusive. Here, we show that the pericardial cells that express the transcription factor Even Skipped adopt distinct fates along the anterior-posterior axis. Among them, the most anterior Antp-Ubx-AbdA-negative cells form a novel cardiac outflow component we call the outflow hanging structure, whereas the Antp-expressing cells differentiate into wing heart precursors. Interestingly, Hox gene expression in the Even Skipped-positive cells not only underlies their antero-posterior diversification, but also influences heart morphogenesis in a non-cell-autonomous way. In brief, we identify a new cardiac outflow component derived from a subset of Even Skipped-expressing cells that stabilises the anterior heart tip, and demonstrate non-cell-autonomous effects of Hox gene expression in the Even Skipped-positive cells on heart morphogenesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Heart/embryology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Genes, Homeobox , Genes, Insect , Homeodomain Proteins/genetics , Organogenesis , Pericardium/cytology , Pericardium/embryology , Pericardium/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...