Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 5730, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37714829

ABSTRACT

The re-use of genes in new organs forms the base of many evolutionary novelties. A well-characterised case is the recruitment of the posterior spiracle gene network to the Drosophila male genitalia. Here we find that this network has also been co-opted to the testis mesoderm where is required for sperm liberation, providing an example of sequentially repeated developmental co-options. Associated to this co-option event, an evolutionary expression novelty appeared, the activation of the posterior segment determinant Engrailed to the anterior A8 segment controlled by common testis and spiracle regulatory elements. Enhancer deletion shows that A8 anterior Engrailed activation is not required for spiracle development but only necessary in the testis. Our study presents an example of pre-adaptive developmental novelty: the activation of the Engrailed transcription factor in the anterior compartment of the A8 segment where, despite having no specific function, opens the possibility of this developmental factor acquiring one. We propose that recently co-opted networks become interlocked, so that any change to the network because of its function in one organ, will be mirrored by other organs even if it provides no selective advantage to them.


Subject(s)
Drosophila , Gene Regulatory Networks , Male , Animals , Drosophila/genetics , Semen , Mesoderm , Genes, Developmental
2.
Elife ; 112022 11 03.
Article in English | MEDLINE | ID: mdl-36326091

ABSTRACT

The identification of genes affecting gonad development is essential to understand the mechanisms causing Variations/Differences in Sex Development (DSD). Recently, a DLC3 mutation was associated with male gonadal dysgenesis in 46,XY DSD patients. We have studied the requirement of Cv-c, the Drosophila ortholog of DLC3, in Drosophila gonad development, as well as the functional capacity of DLC3 human variants to rescue cv-c gonad defects. We show that Cv-c is required to maintain testis integrity during fly development. We find that Cv-c and human DLC3 can perform the same function in fly embryos, as flies carrying wild type but not patient DLC3 variations can rescue gonadal dysgenesis, suggesting functional conservation. We also demonstrate that the StART domain mediates Cv-c's function in the male gonad independently from the GAP domain's activity. This work demonstrates a role for DLC3/Cv-c in male gonadogenesis and highlights a novel StART domain mediated function required to organize the gonadal mesoderm and maintain its interaction with the germ cells during testis development.


Subject(s)
Drosophila Proteins , Gonadal Dysgenesis , Animals , Humans , Male , Drosophila , Drosophila Proteins/genetics , Germ Cells , GTPase-Activating Proteins/genetics , Sex Differentiation , Testis
3.
Sci Rep ; 10(1): 11454, 2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32632122

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Curr Biol ; 30(11): R655-R657, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32516617

ABSTRACT

Wings and halteres are homologous flight appendages whose shape differences are controlled by the Ubx transcription factor. Recent research shows how Ubx regulates apical and basal extracellular matrix proteases and their inhibitors to achieve this morphological divergence.


Subject(s)
Genes, Homeobox , Wings, Animal , Animals , Morphogenesis , Transcription Factors/genetics
5.
Nat Commun ; 9(1): 4210, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297704

ABSTRACT

The original version of this Article contained an error in ref. 39, which incorrectly cited 'Fristrom, D. & Fristrom, J. W. in The Development of Drosophila melanogaster (eds. Bate, M. & Martinez-Arias, A.) II, (Cold spring harbor laboratory press, 1993)'. The correct reference is 'Condic, M.L, Fristrom, D. & Fristrom, J.W. Apical cell shape changes during Drosophila imaginal leg disc elongation: a novel morphogenetic mechanism. Development 111: 23-33 (1991)'. Furthermore, the last sentence of the fourth paragraph of the introduction incorrectly omitted citation of work by Rupprecht et al. The correct citation is given below. These errors have now been corrected in both the PDF and HTML versions of the Article. Rupprecht, J.F., Ong, K.H., Yin, J., Huang, A., Dinh, H.H., Singh, A.P., Zhang, S., Yu, W. & Saunders, T.E. Geometric constraints alter cell arrangements within curved epithelial tissues. Mol. Biol. Cell 28, 3582-3594 (2017).

6.
Nat Commun ; 9(1): 2960, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054479

ABSTRACT

As animals develop, tissue bending contributes to shape the organs into complex three-dimensional structures. However, the architecture and packing of curved epithelia remains largely unknown. Here we show by means of mathematical modelling that cells in bent epithelia can undergo intercalations along the apico-basal axis. This phenomenon forces cells to have different neighbours in their basal and apical surfaces. As a consequence, epithelial cells adopt a novel shape that we term "scutoid". The detailed analysis of diverse tissues confirms that generation of apico-basal intercalations between cells is a common feature during morphogenesis. Using biophysical arguments, we propose that scutoids make possible the minimization of the tissue energy and stabilize three-dimensional packing. Hence, we conclude that scutoids are one of nature's solutions to achieve epithelial bending. Our findings pave the way to understand the three-dimensional organization of epithelial organs.


Subject(s)
Cell Shape , Epithelial Cells/cytology , Epithelium/embryology , Epithelium/physiology , Models, Biological , Animals , Biophysical Phenomena , Computational Biology , Drosophila , Female , Morphogenesis , Salivary Glands/cytology , Zebrafish
7.
Sci Rep ; 8(1): 4601, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545526

ABSTRACT

RhoGAP proteins control the precise regulation of the ubiquitous small RhoGTPases. The Drosophila Crossveinless-c (Cv-c) RhoGAP is homologous to the human tumour suppressor proteins Deleted in Liver Cancer 1-3 (DLC1-3) sharing an identical arrangement of SAM, GAP and START protein domains. Here we analyse in Drosophila the requirement of each Cv-c domain to its function and cellular localization. We show that the basolateral membrane association of Cv-c is key for its epithelial function and find that the GAP domain targeted to the membrane can perform its RhoGAP activity independently of the rest of the protein, implying the SAM and START domains perform regulatory roles. We propose the SAM domain has a repressor effect over the GAP domain that is counteracted by the START domain, while the basolateral localization is mediated by a central, non-conserved Cv-c region. We find that DLC3 and Cv-c expression in the Drosophila ectoderm cause identical effects. In contrast, DLC1 is inactive but becomes functional if the central non-conserved DLC1 domain is substituted for that of Cv-c. Thus, these RhoGAP proteins are functionally equivalent, opening up the use of Drosophila as an in vivo model to analyse pharmacologically and genetically the human DLC proteins.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , GTPase-Activating Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Drosophila/growth & development , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Embryo, Nonmammalian/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Malpighian Tubules/metabolism , Protein Domains , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
8.
Small GTPases ; 9(4): 352-359, 2018 07 04.
Article in English | MEDLINE | ID: mdl-27687567

ABSTRACT

A tight relationship between apico-basal polarity and trafficking is essential for epithelial physiology and tissue homeostasis. Recent studies have described how some Rab GTPases, key components of the intracellular traffic machinery, contribute to the establishment of cell polarity in vertebrates. We have demonstrated a novel connection between cell polarity and trafficking: in Drosophila epithelia, the apical determinant aPKC is recycled via Rab11-Nuf-recycling endosomes to maintain cell polarity. Furthermore, the phosphorylation of Nuf by aPKC allows aPKC to control the sub-cellular localization of Nuf and its own membrane accumulation. Here we review these data and show the different contribution of the 2 Drosophila Rab11 adaptor proteins, Nuf and Rip11, to the maintenance of Drosophila embryonic ectoderm polarity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity , Drosophila Proteins/metabolism , Drosophila/cytology , Drosophila/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Animals
9.
BMC Biol ; 14: 32, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27089924

ABSTRACT

BACKGROUND: Cell polarity, essential for cell physiology and tissue coherence, emerges as a consequence of asymmetric localization of protein complexes and directional trafficking of cellular components. Although molecules required in both processes are well known their relationship is still poorly understood. RESULTS: Here we show a molecular link between Nuclear Fallout (Nuf), an adaptor of Rab11-GTPase to the microtubule motor proteins during Recycling Endosome (RE) trafficking, and aPKC, a pivotal kinase in the regulation of cell polarity. We demonstrate that aPKC phosphorylates Nuf modifying its subcellular distribution. Accordingly, in aPKC mutants Nuf and Rab11 accumulate apically indicating altered RE delivery. We show that aPKC localization in the apico-lateral cortex is dynamic. When we block exocytosis, by means of exocyst-sec mutants, aPKC accumulates inside the cells. Moreover, apical aPKC concentration is reduced in nuf mutants, suggesting aPKC levels are maintained by recycling. CONCLUSIONS: We demonstrate that active aPKC interacts with Nuf, phosphorylating it and, as a result, modifying its subcellular distribution. We propose a regulatory loop by which Nuf promotes aPKC apical recycling until sufficient levels of active aPKC are reached. Thus, we provide a novel link between cell polarity regulation and traffic control in epithelia.


Subject(s)
Drosophila Proteins/metabolism , Nuclear Proteins/metabolism , Protein Kinase C/metabolism , Animals , Cell Polarity , Drosophila Proteins/analysis , Nuclear Proteins/analysis , Phosphorylation , Protein Interaction Maps , Protein Kinase C/analysis , Protein Transport
10.
Curr Biol ; 24(1): 76-81, 2014 Jan 06.
Article in English | MEDLINE | ID: mdl-24332544

ABSTRACT

Segmented organisms have serially repeated structures [1] that become specialized in some segments [2]. We show here that the Drosophila corpora allata, prothoracic glands, and trachea have a homologous origin and can convert into each other. The tracheal epithelial tubes develop from ten trunk placodes [3, 4], and homologous ectodermal cells in the maxilla and labium form the corpora allata and the prothoracic glands. The early endocrine and trachea gene networks are similar, with STAT and Hox genes inducing their activation. The initial invagination of the trachea and the endocrine primordia is identical, but activation of Snail in the glands induces an epithelial-mesenchymal transition (EMT), after which the corpora allata and prothoracic gland primordia coalesce and migrate dorsally, joining the corpora cardiaca to form the ring gland. We propose that the arthropod ectodermal endocrine glands and respiratory organs arose through an extreme process of divergent evolution from a metameric repeated structure.


Subject(s)
Corpora Allata/anatomy & histology , Drosophila melanogaster/anatomy & histology , Trachea/anatomy & histology , Animals , Corpora Allata/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Genes, Homeobox , STAT Transcription Factors/metabolism , Signal Transduction , Trachea/metabolism
11.
JAKSTAT ; 2(3): e26089, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24069568

ABSTRACT

One of the main contributions of Drosophila to the JAK-STAT field is the study of morphogenesis. JAK-STAT signaling controls the formation of many different structures through surprisingly different morphogenetic behaviors that include induction of cell rearrangements, invagination, folding of tissues, modulation of cell shape, and migration. This variability may be explained by the many transcription factors and signaling molecules STAT regulates at early stages of development. But is STAT just acting as an upstream inducer of morphogenesis or does it have a more direct role in controlling cell behaviors? Here we review what is known about how the canonical phosphorylation of STAT contributes to shaping the embryonic and imaginal structures.

12.
Development ; 140(7): 1507-16, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23462467

ABSTRACT

Intercellular communication depends on the correct organization of the signal transduction complexes. In many signalling pathways, the mechanisms controlling the overall cell polarity also localize components of these pathways to different domains of the plasma membrane. In the Drosophila ectoderm, the JAK/STAT pathway components are highly polarized with apical localization of the receptor, the associated kinase and the STAT92E protein itself. The apical localization of STAT92E is independent of the receptor complex and is due to its direct association with the apical determining protein Bazooka (Baz). Here, we find that Baz-STAT92E interaction depends on the presence of the Drosophila Src kinases. In the absence of Src, STAT92E cannot bind to Baz in cells or in whole embryos, and this correlates with an impairment of JAK/STAT signalling function. We believe that the requirement of Src proteins for STAT92E apical localization is mediated through Baz, as we can co-precipitate Src with Baz but not with STAT92E. This is the first time that a functional link between cell polarity, the JAK/STAT signalling pathway and the Src kinases has been established in a whole organism.


Subject(s)
Drosophila Proteins/metabolism , Drosophila , Ectoderm/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , STAT Transcription Factors/metabolism , src-Family Kinases/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Polarity/genetics , Drosophila/embryology , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Ectoderm/embryology , Embryo, Nonmammalian , Female , Intracellular Signaling Peptides and Proteins/genetics , Janus Kinases/genetics , Janus Kinases/metabolism , Janus Kinases/physiology , Male , Protein Binding/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , src-Family Kinases/genetics , src-Family Kinases/metabolism
13.
Proc Natl Acad Sci U S A ; 110(11): 4303-8, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23440219

ABSTRACT

The Abdominal-B selector protein induces organogenesis of the posterior spiracles by coordinating an organ-specific gene network. The complexity of this network begs the questions of how it originated and what selective pressures drove its formation. Given that the network likely formed in a piecemeal fashion, with elements recruited sequentially, we studied the consequences of expressing individual effectors of this network in naive epithelial cells. We found that, with exception of the Crossveinless-c (Cv-c) Rho GTPase-activating protein, most effectors exert little morphogenetic effect by themselves. In contrast, Cv-c expression causes cell motility and down-regulates epithelial polarity and cell adhesion proteins. These effects differ in cells endogenously expressing Cv-c, which have acquired compensatory mechanisms. In spiracle cells, the down-regulation of polarity and E-cadherin expression caused by Cv-c-induced Rho1 inactivation are compensated for by the simultaneous spiracle up-regulation of guanine nucleotide exchange factor (GEF) proteins, cell polarity, and adhesion molecules. Other epithelial cells that have coopted Cv-c to their morphogenetic gene networks are also resistant to Cv-c's deleterious effects. We propose that cooption of a novel morphogenetic regulator to a selector cascade causes cellular instability, resulting in strong selective pressure that leads that same cascade to recruit molecules that compensate it. This experimental-based hypothesis proposes how the frequently observed complex organogenetic gene networks are put together.


Subject(s)
Drosophila Proteins/biosynthesis , GTPase-Activating Proteins/biosynthesis , Gene Expression Regulation, Developmental/physiology , Gene Regulatory Networks/physiology , Genes, Homeobox/physiology , Morphogenesis/physiology , rho GTP-Binding Proteins/biosynthesis , Animals , Cell Movement/physiology , Down-Regulation/physiology , Drosophila Proteins/genetics , Drosophila melanogaster , GTPase-Activating Proteins/genetics , rho GTP-Binding Proteins/genetics
14.
J Cell Sci ; 124(Pt 2): 240-51, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21172808

ABSTRACT

Many epithelial tissues undergo extensive remodelling during morphogenesis. How their epithelial features, such as apicobasal polarity or adhesion, are maintained and remodelled and how adhesion and polarity proteins contribute to morphogenesis are two important questions in development. Here, we approach these issues by investigating the role of the apical determinant protein Crumbs (Crb) during the morphogenesis of the embryonic Drosophila tracheal system. Crb accumulates differentially throughout tracheal development and is required for different tracheal events. The earliest requirement for Crb is for tracheal invagination, which is preceded by an enhanced accumulation of Crb in the invagination domain. There, Crb, acting in parallel with the epidermal growth factor receptor (Egfr) pathway, is required for tracheal cell apical constriction and for organising an actomyosin complex, which we propose is mediated by Crb recruitment of moesin (Moe). The ability of a Crb isoform unable to rescue polarity in crb mutants to otherwise rescue their invagination phenotype, and the converse inability of a FERM-binding domain mutant Crb to rescue faulty invagination, support our hypothesis that it is the absence of Crb-dependent Moe enrichment, and not the polarity defect, that mainly underlies the crb invagination phenotype. This hypothesis is supported by the phenotype of lethal giant larvae (lgl); crb double mutants. These results unveil a link between Crb and the organisation of the actin cytoskeleton during morphogenesis.


Subject(s)
Cell Polarity , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Morphogenesis , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Membrane Proteins/genetics , Trachea/cytology , Trachea/embryology , Trachea/metabolism
15.
Dev Biol ; 340(2): 571-82, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20171201

ABSTRACT

The ventral veinless (vvl) and trachealess (trh) genes are determinants of the Drosophila trachea. Early in development both genes are independently activated in the tracheal primordia by signals that are ill defined. Mutants blocking JAK/STAT signaling at any level do not form a tracheal tree suggesting that STAT92E may be an upstream transcriptional activator of the early trachea determinants. To test this hypothesis we have searched for STAT92E responsive enhancers activating the expression of vvl and trh in the tracheal primordia. We show that STAT92E regulated enhancers can be rapidly and efficiently isolated by focusing the analysis on genomic regions with clusters of putative STAT binding sites where at least some of them are phylogenetically conserved. Detailed analysis of a vvl early tracheal enhancer shows that non-conserved sites collaborate with conserved sites for enhancer activation. We find that STAT92E regulated enhancers can be located as far 60 kb from the promoters. Our results indicate that vvl and trh are independently activated by STAT92E which is the most important transcription factor required for trachea specification.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , STAT Transcription Factors/metabolism , Trachea/metabolism , Animals , Drosophila/embryology , Drosophila/genetics , Drosophila Proteins/genetics , Embryo, Nonmammalian/metabolism , Enhancer Elements, Genetic , Fluorescent Antibody Technique, Direct , Gene Expression Regulation, Developmental , Genes, Insect , Genes, Reporter , Immunohistochemistry , In Situ Hybridization , Mutation , STAT Transcription Factors/genetics , Trachea/embryology
16.
Int J Dev Biol ; 53(8-10): 1349-58, 2009.
Article in English | MEDLINE | ID: mdl-19247941

ABSTRACT

The posterior spiracle has become one of the best systems to study how Hox genes control morphogenesis. Interaction of Abdominal-B (ABD-B) with dorso ventral and intrasegmental positional information leads to the local activation of ABD-B primary targets in the dorsal region of the eighth abdominal segment (A8). Primary targets pattern the spiracle subdividing it into two broad areas: external stigmatophore vs. internal spiracular chamber precursor cells. Primary targets then activate secondary targets and modulate the expression of signalling molecules in the spiracle primordium creating unique spiracle positional values. This genetic cascade activates the realisator genes that modulate the cell behaviours causing invagination, elongation and cell rearrangements responsible for spiracle morphogenesis. The spiracle realisators that have been identified to date correspond to cell adhesion proteins, cytoskeleton regulators and cell polarity molecules. Interestingly, these realisators localise to different apico-basal locations in the cell (RhoGEF apical, Crumbs subapical, E-cadherin in the adherens junction, RhoGAP basolateral). Therefore, the Hox anterior-posterior code is converted in the cell into apico-basal information required to implement the posterior spiracle morphogenetic program. We believe this may be a common characteristic for Hox induced organogenesis.


Subject(s)
Body Patterning/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Homeodomain Proteins/physiology , Organogenesis/physiology , Animals , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Models, Biological , Mutation , Organogenesis/genetics
17.
Semin Cell Dev Biol ; 19(4): 370-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18590829

ABSTRACT

The components of many signalling pathways are localised in specific cellular compartments in polarised cells. This is particularly clear in the case of the receptors that localise to the apical or basal membrane in the epithelial cells. In many cases this subcellular localisation is important for the activation of the signalling pathways. In this review we analyse recent developments uncovering an interesting interplay between JAK/STAT signalling and components regulating cell polarity and adhesion during development. Not only the JAK/STAT signalling components are polarised in epithelial cells but many genes controlling cell polarity and adhesion are targets of STAT and in some cases these components act as pathway activators. The fact that in most morphogenetic processes cell adhesion and polarity proteins are regulated downstream of the pathway, hints at a possible unifying mechanistic explanation for the diverse morphogenetic processes controlled by JAK/STAT during development.


Subject(s)
Cell Adhesion/physiology , Cell Polarity , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Animals , Cadherins/metabolism , Cell Movement , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial Cells/physiology , Janus Kinases/genetics , Morphogenesis , STAT Transcription Factors/genetics
18.
Curr Biol ; 18(8): 624-9, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18424141

ABSTRACT

Three protein complexes control polarization of epithelial cells: the apicolateral Crumbs and Par-3 complexes and the basolateral Lethal giant larvae complex. Polarization results in the specific localization of proteins and lipids to different membrane domains. The receptors of the Notch, Hedgehog, and WNT pathways are among the proteins that are polarized, with subcellular receptor localization representing an important aspect of signaling regulation. For example, in the WNT pathway, differential DFz2 receptor localization results in activation of either the canonical or the planar polarity pathway. Despite the large body of research on the vertebrate JAK/STAT pathway, there are no reports indicating polarized signaling. By using the conserved Drosophila JAK/STAT pathway as a system, we find that the receptor and its associated kinase are located in the apical membrane of epithelial cells. Unexpectedly, the transcription factor STAT is enriched in the apicolateral membrane domain of ectoderm epithelial cells in a Par-3-dependent manner. Our results indicate that preassembly of STAT and the Receptor/JAK complex to specific membrane domains is a key aspect for signaling efficiency. Our results also suggest that receptor polarization in the ectoderm cell membrane restricts the cell's response to ligands provided by neighboring cells.


Subject(s)
Cell Polarity/physiology , Drosophila/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Animals , Drosophila/embryology , Ectoderm/metabolism , Epithelial Cells/metabolism , Mesoderm/metabolism
19.
Curr Biol ; 16(22): 2206-16, 2006 Nov 21.
Article in English | MEDLINE | ID: mdl-17113384

ABSTRACT

BACKGROUND: Hox genes control animal body plans by directing the morphogenesis of segment-specific structures. As transcription factors, HOX proteins achieve this through the activation of downstream target genes. Much research has been devoted to the search for these targets and the characterization of their roles in organogenesis. This has shown that the direct targets of Hox activation are often transcription factors or signaling molecules, which form hierarchical genetic networks directing the morphogenesis of particular organs. Importantly, very few of the direct Hox targets known are "realizator" genes involved directly in the cellular processes of organogenesis. RESULTS: Here, we describe for the first time a complete network linking the Hox gene Abdominal-B to the realizator genes it controls during the organogenesis of the external respiratory organ of the larva. In this process, Abdominal-B induces the expression of four intermediate signaling molecules and transcription factors, and this expression results in the mosaic activation of several realizator genes. The ABD-B spiracle realizators include at least five cell-adhesion proteins, cell-polarity proteins, and GAP and GEF cytoskeleton regulators. Simultaneous ectopic expression of the Abd-B downstream targets can induce spiracle-like structure formation in the absence of ABD-B protein. CONCLUSION: Hox realizators include cytoskeletal regulators and molecules required for the apico-basal cell organization. HOX-coordinated activation of these realizators in mosaic patterns confers to the organ primordium its assembling properties. We propose that during animal development, Hox-controlled genetic cascades coordinate the local cell-specific behaviors that result in organogenesis of segment-specific structures.


Subject(s)
Cell Adhesion/physiology , Cell Polarity/physiology , Cytoskeleton/physiology , Drosophila Proteins/metabolism , Drosophila/growth & development , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Organogenesis/physiology , Signal Transduction/physiology , Animals , Binding Sites/genetics , DNA Primers , Larva/growth & development , Mutagenesis, Site-Directed , RNA Interference
20.
Development ; 133(21): 4257-67, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17021037

ABSTRACT

During development, small RhoGTPases control the precise cell shape changes and movements that underlie morphogenesis. Their activity must be tightly regulated in time and space, but little is known about how Rho regulators (RhoGEFs and RhoGAPs) perform this function in the embryo. Taking advantage of a new probe that allows the visualisation of small RhoGTPase activity in Drosophila, we present evidence that Rho1 is apically activated and essential for epithelial cell invagination, a common morphogenetic movement during embryogenesis. In the posterior spiracles of the fly embryo, this asymmetric activation is achieved by at least two mechanisms: the apical enrichment of Rho1; and the opposing distribution of Rho activators and inhibitors to distinct compartments of the cell membrane. At least two Rho1 activators, RhoGEF2 and RhoGEF64C are localised apically, whereas the Rho inhibitor RhoGAP Cv-c localises at the basolateral membrane. Furthermore, the mRNA of RhoGEF64C is also apically enriched, depending on signals present within its open reading frame, suggesting that apical transport of RhoGEF mRNA followed by local translation is a mechanism to spatially restrict Rho1 activity during epithelial cell invagination.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Epithelial Cells/metabolism , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Morphogenesis , rho GTP-Binding Proteins/metabolism , Animals , Cell Polarity , Cell Shape , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Enzyme Activation , Epithelial Cells/cytology , GTPase-Activating Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , In Situ Hybridization , Myosin Type II/genetics , Myosin Type II/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors , rho GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...