Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Development ; 147(8)2020 04 24.
Article in English | MEDLINE | ID: mdl-32188630

ABSTRACT

Alary muscles (AMs) have been described as a component of the cardiac system in various arthropods. Lineage-related thoracic muscles (TARMs), linking the exoskeleton to specific gut regions, have recently been discovered in Drosophila Asymmetrical attachments of AMs and TARMs, to the exoskeleton on one side and internal organs on the other, suggested an architectural function in moving larvae. Here, we analysed the shape and sarcomeric organisation of AMs and TARMs, and imaged their atypical deformability in crawling larvae. We then selectively eliminated AMs and TARMs by targeted apoptosis. Elimination of AMs revealed that AMs are required for suspending the heart in proper intra-haemocelic position and for opening of the heart lumen, and that AMs constrain the curvature of the respiratory tracheal system during crawling; TARMs are required for proper positioning of visceral organs and efficient food transit. AM/TARM cardiac versus visceral attachment depends on Hox control, with visceral attachment being the ground state. TARMs and AMs are the first example of multinucleate striated muscles connecting the skeleton to the cardiac and visceral systems in bilaterians, with multiple physiological functions.


Subject(s)
Drosophila melanogaster/anatomy & histology , Muscle, Striated/physiology , Organ Specificity , Thorax/physiology , Animals , Calcium/metabolism , Digestive System/metabolism , Drosophila melanogaster/genetics , Food , Gastrointestinal Transit , Genes, Homeobox , Heart/physiology , Intracellular Space/metabolism , Larva/physiology , Locomotion , Sarcomeres/metabolism , Trachea/physiology
2.
PLoS Genet ; 14(2): e1007167, 2018 02.
Article in English | MEDLINE | ID: mdl-29420531

ABSTRACT

Several transcription factors have been identified that activate an epithelial-to-mesenchymal transition (EMT), which endows cells with the capacity to break through basement membranes and migrate away from their site of origin. A key program in development, in recent years it has been shown to be a crucial driver of tumour invasion and metastasis. However, several of these EMT-inducing transcription factors are often expressed long before the initiation of the invasion-metastasis cascade as well as in non-invasive tumours. Increasing evidence suggests that they may promote primary tumour growth, but their precise role in this process remains to be elucidated. To investigate this issue we have focused our studies on two Drosophila transcription factors, the classic EMT inducer Snail and the Drosophila orthologue of hGATAs4/6, Serpent, which drives an alternative mechanism of EMT; both Snail and GATA are specifically expressed in a number of human cancers, particularly at the invasive front and in metastasis. Thus, we recreated conditions of Snail and of Serpent high expression in the fly imaginal wing disc and analysed their effect. While either Snail or Serpent induced a profound loss of epithelial polarity and tissue organisation, Serpent but not Snail also induced an increase in the size of wing discs. Furthermore, the Serpent-induced tumour-like tissues were able to grow extensively when transplanted into the abdomen of adult hosts. We found the differences between Snail and Serpent to correlate with the genetic program they elicit; while activation of either results in an increase in the expression of Yorki target genes, Serpent additionally activates the Ras signalling pathway. These results provide insight into how transcription factors that induce EMT can also promote primary tumour growth, and how in some cases such as GATA factors a 'multi hit' effect may be achieved through the aberrant activation of just a single gene.


Subject(s)
Cell Proliferation/genetics , Drosophila Proteins/physiology , Drosophila/genetics , Epithelial-Mesenchymal Transition/genetics , GATA Transcription Factors/physiology , Neoplasms/pathology , Snail Family Transcription Factors/physiology , Animals , Animals, Genetically Modified , Cell Line, Tumor , Drosophila/embryology , Drosophila/growth & development , Drosophila/physiology , Drosophila Proteins/genetics , Embryo, Nonmammalian , Female , GATA Transcription Factors/genetics , Neoplasm Invasiveness , Neoplasms/genetics , Snail Family Transcription Factors/genetics , Transcription Factors/genetics , Transcription Factors/physiology , Tumor Burden/genetics , Wings, Animal/embryology , Wings, Animal/transplantation
3.
Dev Dyn ; 245(3): 372-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26342211

ABSTRACT

BACKGROUND: The migration of individual cells relies on their capacity to evaluate differences across their bodies and to move either toward or against a chemoattractant or a chemorepellent signal respectively. However, the direction of collective migration is believed to depend on the internal organization of the cell cluster while the role of the external signal is limited to single out some cells in the cluster, conferring them with motility properties. RESULTS: Here we analyzed the role of Fibroblast Growth Factor (FGF) signaling in collective migration in the Drosophila trachea. While ligand-binding FGF receptor (FGFR) activity in a single cell can drive migration of a tracheal cluster, we show that activity from a constitutively activated FGFR cannot-an observation that contrasts with previously analyzed cases. CONCLUSIONS: Our results indicate that individual cells in the tracheal cluster can "read" differences in the distribution of FGFR activity and lead migration of the cluster accordingly. Thus, FGF can act as a chemoattractant rather than as a motogen in collective cell migration. This finding has many implications in both development and pathology.


Subject(s)
Animal Structures/embryology , Cell Movement/physiology , Drosophila Proteins/metabolism , Embryo, Nonmammalian/embryology , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/physiology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Receptors, Fibroblast Growth Factor/genetics
4.
Dev Biol ; 405(2): 183-8, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26206615

ABSTRACT

Hox genes are highly conserved selector genes controlling tissue identity and organogenesis. Recent work indicates that Hox genes also controls cell segregation and segmental boundary in various species, however the underlying cellular mechanisms involved in this function are poorly understood. In Drosophila melanogaster, the Hox gene Deformed (Dfd) is required for specification and organogenesis of the adult Maxillary (Mx) palp. Here, we demonstrate that differential Dfd expression control Mx morphogenesis through the formation of a physical boundary separating the Mx field and the Peripodial Epithelium (PE). We show that this boundary relies on DE-cadherin (DE-cad) basal accumulation in Mx cells controlled by differential Dfd expression. Indeed, Dfd controls boundary formation through cell autonomous basal redistribution of DE-cad which leads to subsequent fold at the Dfd expression border. Finally, the loss of Mx DE-cad basal accumulation and hence of Mx-PE folding is sufficient to prevent Mx organogenesis thus revealing the crucial role of boundaries in organ differentiation. Altogether, these results reveal that Hox coordination of tissue morphogenesis relies on boundary fold formation through the modulation of DE-cad positioning.


Subject(s)
Cadherins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Actins/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation , Epithelium/embryology , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Image Processing, Computer-Assisted , Maxilla/embryology , Microscopy, Confocal , Mitosis , Organogenesis , Protein Folding , RNA Interference
5.
J Cell Sci ; 127(Pt 2): 465-74, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24213534

ABSTRACT

The role of tip and rear cells in collective migration is still a matter of debate and their differences at the cytoskeletal level are poorly understood. Here, we analysed these issues in the Drosophila trachea, an organ that develops from the collective migration of clusters of cells that respond to Branchless (Bnl), a fibroblast growth factor (FGF) homologue expressed in surrounding tissues. We track individual cells in the migratory cluster and characterise their features and unveil two prototypical types of cytoskeletal organisation that account for tip and rear cells respectively. Indeed, once the former are specified, they remain as such throughout migration. Furthermore, we show that FGF signalling in a single tip cell can trigger the migration of the cells in the branch. Finally, we found specific Rac activation at the tip cells and analysed how FGF-independent cell features, such as adhesion and motility, act on coupling the behaviour of trailing and tip cells. Thus, the combined effect of FGF promoting leading cell behaviour and the modulation of cell properties in a cluster can account for the wide range of migratory events driven by FGF.


Subject(s)
Cell Movement , Drosophila melanogaster/cytology , Trachea/cytology , Animals , Cell Aggregation , Cell Shape , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Enzyme Activation , Fibroblast Growth Factors/metabolism , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/metabolism , Genes, Dominant , Mutation , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism
6.
Dev Biol ; 362(2): 187-93, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22178247

ABSTRACT

Connection of epithelial tubes to generate a common network is a key step in the formation of tubular organs such as the tracheal respiratory and the vascular systems. However, it is not clear how these connecting tubes arise. Here we address this issue by studying the dorsal fusion branches in the Drosophila trachea, taking into account the morphology and contribution of each cell type on the basis of their individual labeling. Our results explain how a fusion branch forms and also illustrate the different nature of the two seamless tubes in the Drosophila trachea, generated by fusion and terminal cells respectively.


Subject(s)
Drosophila/embryology , Morphogenesis/physiology , Trachea/embryology , Animals , Cell Adhesion , Cell Fusion , Cell Shape , Immunohistochemistry , Microscopy, Fluorescence , Microtubules/physiology , Time-Lapse Imaging , Trachea/cytology
7.
Front Cell Dev Biol ; 11: 1337708, 2023.
Article in English | MEDLINE | ID: mdl-38288343

ABSTRACT

Three types of muscles, cardiac, smooth and skeletal muscles are classically distinguished in eubilaterian animals. The skeletal, striated muscles are innervated multinucleated syncytia, which, together with bones and tendons, carry out voluntary and reflex body movements. Alary muscles (AMs) are another type of striated syncytial muscles, which connect the exoskeleton to the heart in adult arthropods and were proposed to control hemolymph flux. Developmental studies in Drosophila showed that larval AMs are specified in embryos under control of conserved myogenic transcription factors and interact with excretory, respiratory and hematopoietic tissues in addition to the heart. They also revealed the existence of thoracic AMs (TARMs) connecting to specific gut regions. Their asymmetric attachment sites, deformation properties in crawling larvae and ablation-induced phenotypes, suggest that AMs and TARMs could play both architectural and signalling functions. During metamorphosis, and heart remodelling, some AMs trans-differentiate into another type of muscles. Remaining critical questions include the enigmatic modes and roles of AM innervation, mechanical properties of AMs and TARMS and their evolutionary origin. The purpose of this review is to consolidate facts and hypotheses surrounding AMs/TARMs and underscore the need for further detailed investigation into these atypical muscles.

8.
Front Cell Dev Biol ; 10: 834720, 2022.
Article in English | MEDLINE | ID: mdl-35237606

ABSTRACT

The Drosophila lymph gland is the larval hematopoietic organ and is aligned along the anterior part of the cardiovascular system, composed of cardiac cells, that form the cardiac tube and its associated pericardial cells or nephrocytes. By the end of embryogenesis the lymph gland is composed of a single pair of lobes. Two additional pairs of posterior lobes develop during larval development to contribute to the mature lymph gland. In this study we describe the ontogeny of lymph gland posterior lobes during larval development and identify the genetic basis of the process. By lineage tracing we show here that each posterior lobe originates from three embryonic pericardial cells, thus establishing a bivalent blood cell/nephrocyte potential for a subset of embryonic pericardial cells. The posterior lobes of L3 larvae posterior lobes are composed of heterogeneous blood progenitors and their diversity is progressively built during larval development. We further establish that in larvae, homeotic genes and the transcription factor Klf15 regulate the choice between blood cell and nephrocyte fates. Our data underline the sequential production of blood cell progenitors during larval development.

9.
Elife ; 92020 07 24.
Article in English | MEDLINE | ID: mdl-32706334

ABSTRACT

Myogenesis is an evolutionarily conserved process. Little known, however, is how the morphology of each muscle is determined, such that movements relying upon contraction of many muscles are both precise and coordinated. Each Drosophila larval muscle is a single multinucleated fibre whose morphology reflects expression of distinctive identity Transcription Factors (iTFs). By deleting transcription cis-regulatory modules of one iTF, Collier, we generated viable muscle identity mutants, allowing live imaging and locomotion assays. We show that both selection of muscle attachment sites and muscle/muscle matching is intrinsic to muscle identity and requires transcriptional reprogramming of syncytial nuclei. Live-imaging shows that the staggered muscle pattern involves attraction to tendon cells and heterotypic muscle-muscle adhesion. Unbalance leads to formation of branched muscles, and this correlates with locomotor behavior deficit. Thus, engineering Drosophila muscle identity mutants allows to investigate, in vivo, physiological and mechanical properties of abnormal muscles.


Each muscle in the body has a unique size, shape and set of attachment points. Animals need all of their muscles to have the correct identity to help maintain posture and control movement. A specific set of proteins, called transcription factors, co-ordinate and regulate gene activity in cells so that each muscle develops in the right way. To create a muscle, multiple precursor cells fuse together to form a muscle fibre, which then elongates and attaches to specific sites. Correct attachment is critical so that the fibre is properly oriented. When this process goes wrong, for example in disease, muscle fibres sometimes attach to the wrong site; they become branched and cannot work properly. Collier is a transcription factor protein that controls muscle identity in the fruit fly Drosophila melanogaster. However, like many transcription factors, Collier also has several other roles throughout the body. This made it difficult to evaluate the effect of the protein on the formation of specific muscles. Here, Carayon et al. managed to selectively deactivate Collier in just one muscle per body section in the larvae of fruit flies. This showed that the transcription factor is needed throughout muscle development; in particular, it is required for muscle fibres to select the correct attachment sites, and to be properly oriented. Affected muscles showed an altered orientation, with branched fibres attaching to the wrong site. Even minor changes, which only affect a single muscle from each body segment, greatly impaired the movement of the larvae. The work by Carayon et al. offers a new approach to the study of muscular conditions. Branched muscles are seen in severe human illnesses such as Duchenne muscular dystrophy. Studying the impact of these changes in a living animal could help to understand how this disease progress, and how it can be prevented.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Muscle Development/genetics , Transcription Factors/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Larva/genetics , Larva/growth & development , Transcription Factors/metabolism
10.
Dev Cell ; 33(6): 675-89, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26073018

ABSTRACT

Left-right (LR) asymmetry is essential for organ development and function in metazoans, but how initial LR cue is relayed to tissues still remains unclear. Here, we propose a mechanism by which the Drosophila LR determinant Myosin ID (MyoID) transfers LR information to neighboring cells through the planar cell polarity (PCP) atypical cadherin Dachsous (Ds). Molecular interaction between MyoID and Ds in a specific LR organizer controls dextral cell polarity of adjoining hindgut progenitors and is required for organ looping in adults. Loss of Ds blocks hindgut tissue polarization and looping, indicating that Ds is a crucial factor for both LR cue transmission and asymmetric morphogenesis. We further show that the Ds/Fat and Frizzled PCP pathways are required for the spreading of LR asymmetry throughout the hindgut progenitor tissue. These results identify a direct functional coupling between the LR determinant MyoID and PCP, essential for non-autonomous propagation of early LR asymmetry.


Subject(s)
Body Patterning/physiology , Cadherins/physiology , Digestive System/growth & development , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cadherins/genetics , Cell Polarity/genetics , Cell Polarity/physiology , Digestive System/cytology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genes, Insect , Models, Biological , Myosins/genetics , Myosins/physiology
11.
Development ; 135(13): 2301-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18508864

ABSTRACT

The Drosophila adult head mostly derives from the composite eye-antenna imaginal disc. The antennal disc gives rise to two adult olfactory organs: the antennae and maxillary palps. Here, we have analysed the regional specification of the maxillary palp within the antennal disc. We found that a maxillary field, defined by expression of the Hox gene Deformed, is established at about the same time as the eye and antennal fields during the L2 larval stage. The genetic program leading to maxillary regionalisation and identity is very similar to the antennal one, but is distinguished primarily by delayed prepupal expression of the ventral morphogen Wingless (Wg). We find that precociously expressing Wg in the larval maxillary field suffices to transform it towards antennal identity, whereas overexpressing Wg later in prepupae does not. These results thus indicate that temporal regulation of Wg is decisive to distinguishing maxillary and antennal organs. Wg normally acts upstream of the antennal selector spineless (ss) in maxillary development. However, mis-expression of Ss can prematurely activate wg via a positive-feedback loop leading to a maxillary-to-antenna transformation. We characterised: (1) the action of Wg through ss selector function in distinguishing maxillary from antenna; and (2) its direct contribution to identity choice.


Subject(s)
Animal Structures/growth & development , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Optic Nerve/growth & development , Optic Nerve/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Animal Structures/anatomy & histology , Animal Structures/metabolism , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Larva/genetics , Larva/growth & development , Larva/metabolism , Proto-Oncogene Proteins/genetics , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Time Factors , Wnt1 Protein
12.
Development ; 134(24): 4347-55, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18003742

ABSTRACT

Specification of muscle identity in Drosophila is a multistep process: early positional information defines competence groups termed promuscular clusters, from which muscle progenitors are selected, followed by asymmetric division of progenitors into muscle founder cells (FCs). Each FC seeds the formation of an individual muscle with morphological and functional properties that have been proposed to reflect the combination of transcription factors expressed by its founder. However, it is still unclear how early patterning and muscle-specific differentiation are linked. We addressed this question, using Collier (Col; also known as Knot) expression as both a determinant and read-out of DA3 muscle identity. Characterization of the col upstream region driving DA3 muscle specific expression revealed the existence of three separate phases of cis-regulation, correlating with conserved binding sites for different mesodermal transcription factors. Examination of col transcription in col and nautilus (nau) loss-of-function and gain-of-function conditions showed that both factors are required for col activation in the ;naïve' myoblasts that fuse with the DA3 FC, thereby ensuring that all DA3 myofibre nuclei express the same identity programme. Together, these results indicate that separate sets of cis-regulatory elements control the expression of identity factors in muscle progenitors and myofibre nuclei and directly support the concept of combinatorial control of muscle identity.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Muscles/embryology , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Body Patterning/genetics , Drosophila Proteins/physiology , Gene Expression Regulation, Developmental , Genes, Insect , Models, Biological , Muscle Proteins/genetics , Muscle Proteins/physiology , Mutation , Myoblasts/cytology , Transcription Factors/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL