Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(6): eadg8816, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38335295

ABSTRACT

To achieve a highly differentiated state, cells undergo multiple transcriptional processes whose coordination and timing are not well understood. In Drosophila embryonic epidermal cells, polished-rice (Pri) smORF peptides act as temporal mediators of ecdysone to activate a transcriptional program leading to cell shape remodeling. Here, we show that the ecdysone/Pri axis concomitantly represses the transcription of a large subset of cuticle genes to ensure proper differentiation of the insect exoskeleton. The repression relies on the transcription factor Ken and persists for several days throughout early larval stages, during which a soft cuticle allows larval crawling. The onset of these cuticle genes normally awaits the end of larval stages when the rigid pupal case assembles, and their premature expression triggers abnormal sclerotization of the larval cuticle. These results uncovered a temporal switch to set up distinct structures of cuticles adapted to the animal lifestyle and which might be involved in the evolutionary history of insects.


Subject(s)
Drosophila Proteins , Ecdysone , Animals , Ecdysone/metabolism , Drosophila/genetics , Drosophila/metabolism , Cell Differentiation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Peptides/metabolism , Larva/genetics , Insecta/genetics , Gene Expression Regulation, Developmental , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
2.
Front Genet ; 12: 714152, 2021.
Article in English | MEDLINE | ID: mdl-34527021

ABSTRACT

There is growing evidence that peptides encoded by small open-reading frames (sORF or smORF) can fulfill various cellular functions and define a novel class regulatory molecules. To which extend transcripts encoding only smORF peptides compare with canonical protein-coding genes, yet remain poorly understood. In particular, little is known on whether and how smORF-encoding RNAs might need tightly regulated expression within a given tissue, at a given time during development. We addressed these questions through the analysis of Drosophila polished rice (pri, a.k.a. tarsal less or mille pattes), which encodes four smORF peptides (11-32 amino acids in length) required at several stages of development. Previous work has shown that the expression of pri during epidermal development is regulated in the response to ecdysone, the major steroid hormone in insects. Here, we show that pri transcription is strongly upregulated by ecdysone across a large panel of cell types, suggesting that pri is a core component of ecdysone response. Although pri is produced as an intron-less short transcript (1.5 kb), genetic assays reveal that the developmental functions of pri require an unexpectedly large array of enhancers (spanning over 50 kb), driving a variety of spatiotemporal patterns of pri expression across developing tissues. Furthermore, we found that separate pri enhancers are directly activated by the ecdysone nuclear receptor (EcR) and display distinct regulatory modes between developmental tissues and/or stages. Alike major developmental genes, the expression of pri in a given tissue often involves several enhancers driving apparently redundant (or shadow) expression, while individual pri enhancers can harbor pleiotropic functions across tissues. Taken together, these data reveal the broad role of Pri smORF peptides in ecdysone signaling and show that the cis-regulatory architecture of the pri gene contributes to shape distinct spatial and temporal patterns of ecdysone response throughout development.

3.
Elife ; 82019 03 21.
Article in English | MEDLINE | ID: mdl-30896406

ABSTRACT

Small open reading frames (smORFs) encoding 'micropeptides' exhibit remarkable evolutionary complexity. Conserved peptides encoded by mille-pattes (mlpt)/polished rice (pri)/tarsal less (tal) are essential for embryo segmentation in Tribolium but, in Drosophila, function in terminal epidermal differentiation and patterning of adult legs. Here, we show that a molecular complex identified in Drosophila epidermal differentiation, comprising Mlpt peptides, ubiquitin-ligase Ubr3 and transcription factor Shavenbaby (Svb), represents an ancient developmental module required for early insect embryo patterning. We find that loss of segmentation function for this module in flies evolved concomitantly with restriction of Svb expression in early Drosophila embryos. Consistent with this observation, artificially restoring early Svb expression in flies causes segmentation defects that depend on mlpt function, demonstrating enduring potency of an ancestral developmental switch despite evolving embryonic patterning modes. These results highlight the evolutionary plasticity of conserved molecular complexes under the constraints of essential genetic networks. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Subject(s)
Body Patterning , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/embryology , Epidermis/embryology , Gene Expression Regulation, Developmental , Organogenesis , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals
4.
Nat Commun ; 9(1): 5123, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504772

ABSTRACT

To compensate for accumulating damages and cell death, adult homeostasis (e.g., body fluids and secretion) requires organ regeneration, operated by long-lived stem cells. How stem cells can survive throughout the animal life remains poorly understood. Here we show that the transcription factor Shavenbaby (Svb, OvoL in vertebrates) is expressed in renal/nephric stem cells (RNSCs) of Drosophila and required for their maintenance during adulthood. As recently shown in embryos, Svb function in adult RNSCs further needs a post-translational processing mediated by the Polished rice (Pri) smORF peptides and impairing Svb function leads to RNSC apoptosis. We show that Svb interacts both genetically and physically with Yorkie (YAP/TAZ in vertebrates), a nuclear effector of the Hippo pathway, to activate the expression of the inhibitor of apoptosis DIAP1. These data therefore identify Svb as a nuclear effector in the Hippo pathway, critical for the survival of adult somatic stem cells.


Subject(s)
Adult Stem Cells/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , DNA-Binding Proteins/genetics , Drosophila , Drosophila Proteins/genetics , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Trans-Activators/genetics , Transcription Factors/genetics , YAP-Signaling Proteins
5.
Elife ; 52016 07 20.
Article in English | MEDLINE | ID: mdl-27438571

ABSTRACT

Each Drosophila muscle is seeded by one Founder Cell issued from terminal division of a Progenitor Cell (PC). Muscle identity reflects the expression by each PC of a specific combination of identity Transcription Factors (iTFs). Sequential emergence of several PCs at the same position raised the question of how developmental time controlled muscle identity. Here, we identified roles of Anterior Open and ETS domain lacking in controlling PC birth time and Eyes absent, No Ocelli, and Sine oculis in specifying PC identity. The windows of transcription of these and other TFs in wild type and mutant embryos, revealed a cascade of regulation integrating time and space, feed-forward loops and use of alternative transcription start sites. These data provide a dynamic view of the transcriptional control of muscle identity in Drosophila and an extended framework for studying interactions between general myogenic factors and iTFs in evolutionary diversification of muscle shapes.


Subject(s)
Drosophila/embryology , Drosophila/genetics , Gene Expression Regulation, Developmental , Muscles/embryology , Stem Cells/physiology , Transcription Factors/metabolism , Transcription, Genetic , Animals , Time Factors
6.
Nat Cell Biol ; 16(11): 1035-44, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25344753

ABSTRACT

Animal development fundamentally relies on the precise control, in space and time, of genome expression. Whereas we have a wealth of information about spatial patterning, the mechanisms underlying temporal control remain poorly understood. Here we show that Pri peptides, encoded by small open reading frames, are direct mediators of the steroid hormone ecdysone for the timing of developmental programs in Drosophila. We identify a previously uncharacterized enzyme of ecdysone biosynthesis, GstE14, and find that ecdysone triggers pri expression to define the onset of epidermal trichome development, through post-translational control of the Shavenbaby transcription factor. We show that manipulating pri expression is sufficient to either put on hold or induce premature differentiation of trichomes. Furthermore, we find that ecdysone-dependent regulation of pri is not restricted to epidermis and occurs over various tissues and times. Together, these findings provide a molecular framework to explain how systemic hormonal control coordinates specific programs of differentiation with developmental timing.


Subject(s)
Arrestins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Ecdysone/metabolism , Gene Expression Regulation, Developmental/physiology , Glutathione Transferase/metabolism , Receptors, Steroid/metabolism , Animals , Arrestins/genetics , Cell Differentiation/genetics , Drosophila Proteins/genetics , Ecdysone/genetics , Glutathione Transferase/genetics , Mutation/genetics , Receptors, Steroid/genetics , Signal Transduction/physiology , Transaldolase/genetics , Transaldolase/metabolism
7.
Semin Cell Dev Biol ; 23(3): 341-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22406682

ABSTRACT

One of the most challenging problems in biology resides in unraveling the molecular mechanisms, hardwired in the genome, that define and regulate the multiscale tridimensional organization of organs, tissues and individual cells. While works in cultured cells have revealed the importance of cytoskeletal networks for cell architecture, in vivo models are now required to explore how such a variety in cell shape is produced during development, in interaction with neighboring cells and tissues. The genetic analysis of epidermis development in Drosophila has provided an unbiased way to identify mechanisms remodeling the shape of epidermal cells, to form apical trichomes during terminal differentiation. Since hearing in vertebrates relies on apical cell extensions in sensory cells of the cochlea, called stereocilia, the mapping of human genes causing hereditary deafness has independently identified several factors required for this peculiar tridimensional organization. In this review, we summarized recent results obtained toward the identification of genes involved in these localized changes in cell shape and discuss their evolution throughout developmental processes and species.


Subject(s)
Biological Evolution , Cell Shape/physiology , Cochlea/cytology , Epidermal Cells , Animals , Cell Communication , Cell Differentiation , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Epidermis/embryology , Humans
8.
Trends Cell Biol ; 20(9): 524-32, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20598543

ABSTRACT

The terminal differentiation of epithelial cells involves changes in the apical compartment, including remodeling of the cytoskeleton and junctions to modify its three-dimensional organization. It also often triggers the building of specialized extracellular matrices, the function of which remains poorly understood. Hundreds of extracellular matrix proteins expressed in a variety of epithelia possess a conserved region called the zona pellucida-domain (ZP domain). There is evidence to suggest that ZP-domains mediate the polymerization of proteins into fibrils or matrices and that mutation of ZP-domains can result in severe pathologies, such as infertility, deafness, and cancer. Recent work in worms and flies demonstrates that ZP-domain proteins play a crucial role in organizing and shaping highly specialized apical structures in epithelial cells.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Intercellular Junctions , Protein Structure, Tertiary , Zona Pellucida/chemistry , Zona Pellucida/metabolism
9.
J Biomed Biotechnol ; 2010: 376927, 2010.
Article in English | MEDLINE | ID: mdl-20625483

ABSTRACT

Collagen V, a fibrillar collagen with important functions in tissues, assembles into distinct chain associations. The most abundant and ubiquitous molecular form is the heterotrimer [alpha1(V)](2)alpha2(V). In the attempt to produce high levels of recombinant collagen V heterotrimer for biomedical device uses, and to identify key factors that drive heterotrimeric chain association, several cell expression systems (yeast, insect, and mammalian cells) have been assayed by cotransfecting the human proalpha1(V) and proalpha2(V) chain cDNAs. Suprisingly, in all recombinant expression systems, the formation of [alpha1(V)](3) homotrimers was considerably favored over the heterotrimer. In addition, pepsin-sensitive proalpha2(V) chains were found in HEK-293 cell media indicating that these cells lack quality control proteins preventing collagen monomer secretion. Additional transfection with Hsp47 cDNA, encoding the collagen-specific chaperone Hsp47, did not increase heterotrimer production. Double immunofluorescence with antibodies against collagen V alpha-chains showed that, contrary to fibroblasts, collagen V alpha-chains did not colocalized intracellularly in transfected cells. Monensin treatment had no effect on the heterotrimer production. The heterotrimer production seems to require specific machinery proteins, which are not endogenously expressed in the expression systems. The different constructs and transfected cells we have generated represent useful tools to further investigate the mechanisms of collagen trimer assembly.


Subject(s)
Collagen Type V/biosynthesis , Protein Multimerization , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Animals , Cell Line , Clone Cells , Collagen Type V/chemistry , Fluorescent Antibody Technique , HSP47 Heat-Shock Proteins/metabolism , Humans , Insecta/cytology , Pichia/metabolism
10.
PLoS One ; 5(4): e10245, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20422006

ABSTRACT

BACKGROUND: Endocytosis is a key regulatory step of diverse signalling pathways, including receptor tyrosine kinase (RTK) signalling. Hrs and Stam constitute the ESCRT-0 complex that controls the initial selection of ubiquitinated proteins, which will subsequently be degraded in lysosomes. It has been well established ex vivo and during Drosophila embryogenesis that Hrs promotes EGFR down regulation. We have recently isolated the first mutations of stam in flies and shown that Stam is required for air sac morphogenesis, a larval respiratory structure whose formation critically depends on finely tuned levels of FGFR activity. This suggest that Stam, putatively within the ESCRT-0 complex, modulates FGF signalling, a possibility that has not been examined in Drosophila yet. PRINCIPAL FINDINGS: Here, we assessed the role of the Hrs/Stam complex in the regulation of signalling activity during Drosophila development. We show that stam and hrs are required for efficient FGFR signalling in the tracheal system, both during cell migration in the air sac primordium and during the formation of fine cytoplasmic extensions in terminal cells. We find that stam and hrs mutant cells display altered FGFR/Btl localisation, likely contributing to impaired signalling levels. Electron microscopy analyses indicate that endosome maturation is impaired at distinct steps by hrs and stam mutations. These somewhat unexpected results prompted us to further explore the function of stam and hrs in EGFR signalling. We show that while stam and hrs together downregulate EGFR signalling in the embryo, they are required for full activation of EGFR signalling during wing development. CONCLUSIONS/SIGNIFICANCE: Our study shows that the ESCRT-0 complex differentially regulates RTK signalling, either positively or negatively depending on tissues and developmental stages, further highlighting the importance of endocytosis in modulating signalling pathways during development.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Drosophila Proteins/physiology , Embryonic Development , Endosomal Sorting Complexes Required for Transport/physiology , Phosphoproteins/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Down-Regulation , Drosophila/growth & development , Drosophila Proteins/genetics , Endocytosis , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes , Mutation , Phosphoproteins/genetics , Protein Transport , Receptors, Fibroblast Growth Factor/metabolism
11.
Dev Cell ; 18(1): 64-76, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20152178

ABSTRACT

The zona pellucida domain (ZPD) defines a conserved family of membrane-anchored matrix proteins that are, as yet, poorly characterized with respect to their functions during development. Using genetic approaches in flies, we show here that a set of eight ZPD proteins is required for the localized reorganization of embryonic epidermal cells during morphogenesis. Despite varying degrees of sequence conservation, these ZPD proteins exert specific and nonredundant functions in the remodeling of epidermal cell shape. Each one accumulates in a restricted subregion of the apical compartment, where it organizes local interactions between the membrane and the extracellular matrix. In addition, ZPD proteins are required to sculpture the actin-rich cell extensions and maintain appropriate organization of the apical compartment. These results on ZPD proteins therefore reveal a functional subcompartmentalization of the apical membrane and its role in the polarized control of epithelial cell shape during development.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Egg Proteins/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Epithelial Cells/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Compartmentation/physiology , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Polarity/physiology , Cell Shape/physiology , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Egg Proteins/genetics , Embryo, Nonmammalian/cytology , Embryonic Development/physiology , Epidermal Cells , Epidermis/metabolism , Epithelial Cells/cytology , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental/physiology , Protein Structure, Tertiary/physiology
12.
Curr Top Dev Biol ; 89: 137-62, 2009.
Article in English | MEDLINE | ID: mdl-19737645

ABSTRACT

Biological tubes are integral components of many organs. Based on their cellular organization, tubes can be divided into three types: multicellular, unicellular, and intracellular. The mechanisms by which these tubes form during development vary significantly, in many cases even for those sharing a similar final architecture. Here, we present recent advances in studying cellular and molecular aspects of tubulogenesis in different organisms.


Subject(s)
Organogenesis , Animals , Cell Physiological Phenomena , Humans
13.
Genetics ; 176(4): 2177-87, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17603108

ABSTRACT

Branching morphogenesis of the Drosophila tracheal system relies on the fibroblast growth factor receptor (FGFR) signaling pathway. The Drosophila FGF ligand Branchless (Bnl) and the FGFR Breathless (Btl/FGFR) are required for cell migration during the establishment of the interconnected network of tracheal tubes. However, due to an important maternal contribution of members of the FGFR pathway in the oocyte, a thorough genetic dissection of the role of components of the FGFR signaling cascade in tracheal cell migration is impossible in the embryo. To bypass this shortcoming, we studied tracheal cell migration in the dorsal air sac primordium, a structure that forms during late larval development. Using a mosaic analysis with a repressible cell marker (MARCM) clone approach in mosaic animals, combined with an ethyl methanesulfonate (EMS)-mutagenesis screen of the left arm of the second chromosome, we identified novel genes implicated in cell migration. We screened 1123 mutagenized lines and identified 47 lines displaying tracheal cell migration defects in the air sac primordium. Using complementation analyses based on lethality, mutations in 20 of these lines were genetically mapped to specific genomic areas. Three of the mutants were mapped to either the Mhc or the stam complementation groups. Further experiments confirmed that these genes are required for cell migration in the tracheal air sac primordium.


Subject(s)
Air Sacs/growth & development , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Genes, Insect , Trachea/growth & development , Air Sacs/cytology , Animals , Base Sequence , Cell Movement/genetics , Crosses, Genetic , DNA Primers/genetics , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , Genetic Complementation Test , Genetic Markers , Larva/cytology , Larva/growth & development , Male , Morphogenesis , Mosaicism , Mutagenesis , Phenotype , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/physiology , Signal Transduction , Trachea/cytology
14.
PLoS Biol ; 4(9): e290, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16933974

ABSTRACT

It is well established that developmental programs act during embryogenesis to determine animal morphogenesis. How these developmental cues produce specific cell shape during morphogenesis, however, has remained elusive. We addressed this question by studying the morphological differentiation of the Drosophila epidermis, governed by a well-known circuit of regulators leading to a stereotyped pattern of smooth cells and cells forming actin-rich extensions (trichomes). It was shown that the transcription factor Shavenbaby plays a pivotal role in the formation of trichomes and underlies all examined cases of the evolutionary diversification of their pattern. To gain insight into the mechanisms of morphological differentiation, we sought to identify shavenbaby's downstream targets. We show here that Shavenbaby controls epidermal cell shape, through the transcriptional activation of different classes of cellular effectors, directly contributing to the organization of actin filaments, regulation of the extracellular matrix, and modification of the cuticle. Individual inactivation of shavenbaby's targets produces distinct trichome defects and only their simultaneous inactivation prevent trichome formation. Our data show that shavenbaby governs an evolutionarily conserved developmental module consisting of a set of genes collectively responsible for trichome formation, shedding new light on molecular mechanisms acting during morphogenesis and the way they can influence evolution of animal forms.


Subject(s)
Body Patterning , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/cytology , Drosophila/embryology , Epidermal Cells , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Cell Shape , DNA-Binding Proteins/genetics , Drosophila/anatomy & histology , Drosophila/genetics , Drosophila Proteins/genetics , Epidermis/embryology , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Membrane Proteins/metabolism , Pigmentation/genetics , Transcription Factors/genetics , Transcription, Genetic
15.
Mol Cell Biol ; 24(13): 6049-57, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15199158

ABSTRACT

Collagen V is a minor component of the heterotypic I/III/V collagen fibrils and the defective product in most cases of classical Ehlers Danlos syndrome (EDS). The present study was undertaken to elucidate the impact of collagen V mutations on skin development, the most severely affected EDS tissues, using mice harboring a targeted deletion of the alpha2(V) collagen gene (Col5a2). Contrary to the original report, our studies indicate that the Col5a2 deletion (a.k.a. the pN allele) represents a functionally null mutation that affects matrix assembly through a complex sequence of events. First the mutation impairs assembly and/or secretion of the alpha1(V)(2)alpha2(V) heterotrimer with the result that the alpha1(V) homotrimer is the predominant species deposited into the matrix. Second, the alpha1(V) homotrimer is excluded from incorporation into the heterotypic collagen fibrils and this in turn severely impairs matrix organization. Third, the mutant matrix stimulates a compensatory loop by the alpha1(V) collagen gene that leads to additional deposition of alpha1(V) homotrimers. These data therefore underscore the importance of the collagen V heterotrimer in dermal fibrillogenesis. Furthermore, reduced thickness of the basement membranes underlying the epidermis and increased apoptosis of the stromal fibroblasts in pN/pN skin strongly indicate additional roles of collagen V in the development of a functional skin matrix.


Subject(s)
Collagen Type V/metabolism , Skin/growth & development , Animals , Apoptosis , Basement Membrane/chemistry , Collagen Type V/deficiency , Collagen Type V/genetics , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/chemistry , Fibroblasts/ultrastructure , Mice , Mice, Knockout , Mutation , Skin/chemistry , Skin/ultrastructure
16.
Mech Dev ; 120(7): 747-58, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12915226

ABSTRACT

In Drosophila, differentiation of the epidermis results in a stereotyped array of cells with F-actin-based extensions at their apical face. We identified Ovo/Shavenbaby (Svb) as a transcription factor that governs changes in epidermal cell shape. Svb is required for the formation of apical extensions and cells deficient in svb differentiate a smooth surface. In both the embryo and the adult, we show that Svb is necessary and sufficient for the cells to grow extensions and that the tight regulation of ovo/svb activity is critical for morphogenesis to occur correctly. We establish that Svb triggers early F-actin redistribution and is able to initiate the entire process of cytoskeletal remodelling, thereby defining it as a major regulator of epidermal differentiation.


Subject(s)
Actins/metabolism , Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Drosophila Proteins , Epidermis/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Drosophila/embryology , Epidermis/embryology , Eye/embryology , Molecular Sequence Data , Wings, Animal/embryology
17.
Med Sci (Paris) ; 19(4): 443-52, 2003 Apr.
Article in French | MEDLINE | ID: mdl-12836217

ABSTRACT

Ehlers-Danlos syndrome (EDS) is a heterogeneous heritable connective tissue disorder characterized by hyper-extensible skin, hypermobile joints and fragile vessels. The molecular causes of this disorder are often, although not strictly, related to collagens and to the enzymes that process these proteins. The classical form of the syndrome, which will be principally discussed in this review, can be due to mutations on collagen V, a fibrillar collagen present in small amounts in affected tissues. However, collagen I and tenascin have also been demonstrated to be involved in the same type of EDS. Moreover gene disruption of several other matrix molecules (thrombospondin, SPARC, small leucine rich proteoglycans...) in mice, lead to phenotypes that mimic EDS and these molecules have thus emerged as new players. As collagen V remains the prime candidate, we discuss, based on fundamental and clinical observations, its physiological role. We also explore its potential interactions with other matrix molecules to determine tissue properties.


Subject(s)
Collagen/genetics , Ehlers-Danlos Syndrome/classification , Ehlers-Danlos Syndrome/genetics , Extracellular Matrix/ultrastructure , Animals , Ehlers-Danlos Syndrome/diagnosis , Extracellular Matrix/genetics , Humans , Mice , Mutation , Tenascin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...