Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Proc Natl Acad Sci U S A ; 120(18): e2217862120, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37094122

ABSTRACT

Hematopoietic stem and progenitor cells maintain blood cell homeostasis by integrating various cues provided by specialized microenvironments or niches. Biomechanical forces are emerging as key regulators of hematopoiesis. Here, we report that mechanical stimuli provided by blood flow in the vascular niche control Drosophila hematopoiesis. In vascular niche cells, the mechanosensitive channel Piezo transduces mechanical forces through intracellular calcium upregulation, leading to Notch activation and repression of FGF ligand transcription, known to regulate hematopoietic progenitor maintenance. Our results provide insight into how the vascular niche integrates mechanical stimuli to regulate hematopoiesis.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/metabolism , Drosophila Proteins/metabolism , Hematopoiesis/physiology , Blood Cells , Stem Cells/metabolism , Stem Cell Niche , Ion Channels
2.
J Cell Sci ; 129(13): 2673-83, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27206857

ABSTRACT

Chromatin function is involved in many cellular processes, its visualization or modification being essential in many developmental or cellular studies. Here, we present the characterization of chromatibody, a chromatin-binding single-domain, and explore its use in living cells. This non-intercalating tool specifically binds the heterodimer of H2A-H2B histones and displays a versatile reactivity, specifically labeling chromatin from yeast to mammals. We show that this genetically encoded probe, when fused to fluorescent proteins, allows non-invasive real-time chromatin imaging. Chromatibody is a dynamic chromatin probe that can be modulated. Finally, chromatibody is an efficient tool to target an enzymatic activity to the nucleosome, such as the DNA damage-dependent H2A ubiquitylation, which can modify this epigenetic mark at the scale of the genome and result in DNA damage signaling and repair defects. Taken together, these results identify chromatibody as a universal non-invasive tool for either in vivo chromatin imaging or to manipulate the chromatin landscape.


Subject(s)
Chromatin/genetics , DNA Damage/genetics , Nucleosomes/genetics , Animals , Camelids, New World , Chromatin/isolation & purification , Histones/metabolism , Ubiquitination/genetics
3.
Development ; 139(19): 3572-82, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22949613

ABSTRACT

The LIM-homeodomain transcription factor Tailup/Islet1 (Tup) is a key component of cardiogenesis in Drosophila and vertebrates. We report here an additional major role for Drosophila Tup in specifying dorsal muscles. Tup is expressed in the four dorsal muscle progenitors (PCs) and tup-null embryos display a severely disorganized dorsal musculature, including a transformation of the dorsal DA2 into dorsolateral DA3 muscle. This transformation is reciprocal to the DA3 to DA2 transformation observed in collier (col) mutants. The DA2 PC, which gives rise to the DA2 muscle and to an adult muscle precursor, is selected from a cluster of myoblasts transiently expressing both Tinman (Tin) and Col. The activation of tup by Tin in the DA2 PC is required to repress col transcription and establish DA2 identity. The transient, partial overlap between Tin and Col expression provides a window of opportunity to distinguish between DA2 and DA3 muscle identities. The function of Tup in the DA2 PC illustrates how single cell precision can be reached in cell specification when temporal dynamics are combined with positional information. The contributions of Tin, Tup and Col to patterning Drosophila dorsal muscles bring novel parallels with chordate pharyngeal muscle development.


Subject(s)
Drosophila Proteins/physiology , Drosophila/embryology , Drosophila/genetics , Muscles/embryology , Organogenesis/genetics , Transcription Factors/physiology , Animals , Animals, Genetically Modified , Cell Lineage/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Models, Biological , Muscles/metabolism , Organ Specificity/genetics , Organogenesis/physiology , Time Factors , Tissue Distribution , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Proc Natl Acad Sci U S A ; 109(9): 3389-94, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331866

ABSTRACT

The Drosophila melanogaster larval hematopoietic organ, the lymph gland, is a model to study in vivo the function of the hematopoietic niche. A small cluster of cells in the lymph gland, the posterior signaling center (PSC), maintains the balance between hematopoietic progenitors (prohemocytes) and their differentiation into specialized blood cells (hemocytes). Here, we show that Decapentaplegic/bone morphogenetic protein (Dpp/BMP) signaling activity in PSC cells controls niche size. In the absence of BMP signaling, the number of PSC cells increases. Correlatively, no hemocytes differentiate. Controlling PSC size is, thus, essential for normal blood cell homeostasis. Activation of BMP signaling in the PSC requires expression of the Dally-like heparan-sulfate proteoglycan, under the control of the Collier/early B-cell factor (EBF) transcription factor. A Dpp > dpp autoregulatory loop maintains BMP signaling, which limits PSC cell proliferation by repressing the protooncogene dmyc. Dpp antagonizes activity of wingless (Wg)/Wnt signaling, which positively regulates the number of PSC cells via the control of Dmyc expression. Together, our data show that Collier controls hemocyte homeostasis via coordinate regulation of PSC cell number and PSC signaling to prohemocytes. In mouse, EBF2, BMP, and Wnt signaling in osteoblasts is required for the proper number of niche and hematopoietic stem cells. Our findings bring insights to niche size control and draw parallels between Drosophila and mammalian hematopoiesis.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Hematopoiesis/physiology , Hemocytes/cytology , Stem Cell Niche , Transcription Factors/physiology , Animals , Cell Count , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Genes, myc , Hemocytes/metabolism , Larva , Mice , Mitotic Index , Proteoglycans/genetics , Proteoglycans/physiology , Signal Transduction/physiology , Species Specificity , Transcription Factors/genetics , Vertebrates/physiology , Wnt1 Protein/genetics , Wnt1 Protein/physiology
5.
Med Sci (Paris) ; 30(1): 76-81, 2014 Jan.
Article in French | MEDLINE | ID: mdl-24472463

ABSTRACT

Stem cells are required for both tissue renewal and repair in response to injury. The maintenance and function of stem cells is controlled by their specific cellular microenvironment called "niche". Hematopoietic stem cells (HSC) that give rise to all blood cell types have been extensively studied in mammals. Genetic and molecular analyses performed in mice identified several signaling pathways involved in the cellular communications between HSC and their niche. However, hematopoietic niche plasticity remains poorly understood. The discovery of a Drosophila hematopoietic niche, called PSC, established a new model to decipher the niche function in vivo. Size control of the PSC is essential to maintain hematopoietic tissue homeostasis and a molecular cascade controlling the PSC cell number has been characterized. Novel parallels between Drosophila and mammalian hematopoietic niches open new perspectives for studies of HSC biology in human.


Subject(s)
Drosophila , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Stem Cell Niche/physiology , Animals , Drosophila/cytology , Drosophila/physiology , Hematopoiesis/physiology , Humans , Mammals , Mice , Models, Biological
6.
Dev Biol ; 363(1): 27-39, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22200594

ABSTRACT

The diversity of Drosophila muscles correlates with the expression of combinations of identity transcription factors (iTFs) in muscle progenitors. Here, we address the question of when and how a combinatorial code is translated into muscle specific properties, by studying the roles of the Collier and Nautilus iTFs that are expressed in partly overlapping subsets of muscle progenitors. We show that the three dorso-lateral (DL) progenitors which express Nautilus and Collier are specified in a fixed temporal sequence and that each expresses additionally other, distinct iTFs. Removal of Collier leads to changes in expression of some of these iTFs and mis-orientation of several DL muscles, including the dorsal acute DA3 muscle which adopts a DA2 morphology. Detailed analysis of this transformation revealed the existence of two steps in the attachment of elongating muscles to specific tendon cells: transient attachment to alternate tendon cells, followed by a resolution step selecting the final sites. The multiple cases of triangular-shaped muscles observed in col mutant embryos indicate that transient binding of elongating muscle to exploratory sites could be a general feature of the developing musculature. In nau mutants, the DA3 muscle randomly adopts the attachment sites of the DA3 or DO5 muscles that derive from the same progenitor, resulting in a DA3, DO5-like or bifid DA3-DO5 orientation. In addition, nau mutant embryos display thinner muscle fibres. Together, our data show that the sequence of expression and combinatorial activities of Col and Nau control the pattern and morphology of DL muscles.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Muscle Proteins/metabolism , Muscles/metabolism , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Microscopy, Electron, Scanning , Models, Biological , Muscle Proteins/genetics , Muscles/embryology , Muscles/ultrastructure , Mutation , Time Factors , Transcription Factors/genetics
7.
Development ; 137(3): 457-66, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20056681

ABSTRACT

Hox transcription factors control many aspects of animal morphogenetic diversity. The segmental pattern of Drosophila larval muscles shows stereotyped variations along the anteroposterior body axis. Each muscle is seeded by a founder cell and the properties specific to each muscle reflect the expression by each founder cell of a specific combination of 'identity' transcription factors. Founder cells originate from asymmetric division of progenitor cells specified at fixed positions. Using the dorsal DA3 muscle lineage as a paradigm, we show here that Hox proteins play a decisive role in establishing the pattern of Drosophila muscles by controlling the expression of identity transcription factors, such as Nautilus and Collier (Col), at the progenitor stage. High-resolution analysis, using newly designed intron-containing reporter genes to detect primary transcripts, shows that the progenitor stage is the key step at which segment-specific information carried by Hox proteins is superimposed on intrasegmental positional information. Differential control of col transcription by the Antennapedia and Ultrabithorax/Abdominal-A paralogs is mediated by separate cis-regulatory modules (CRMs). Hox proteins also control the segment-specific number of myoblasts allocated to the DA3 muscle. We conclude that Hox proteins both regulate and contribute to the combinatorial code of transcription factors that specify muscle identity and act at several steps during the muscle-specification process to generate muscle diversity.


Subject(s)
Homeodomain Proteins/physiology , Muscles/embryology , Animals , Body Patterning , Drosophila/embryology , Embryo, Nonmammalian , Embryonic Development , Morphogenesis , Muscles/cytology , Stem Cells/cytology , Transcription Factors/physiology
8.
PLoS Biol ; 8(8): e1000441, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20689801

ABSTRACT

The posterior signalling centre (PSC), a small group of specialised cells, controls hemocyte (blood cell) homeostasis in the Drosophila larval hematopoietic organ, the lymph gland. This role of the PSC is very reminiscent of the "niche," the micro-environment of hematopoietic stem cells in vertebrates. We have recently shown that the PSC acts in a non-cell-autonomous manner to maintain janus tyrosine kinase/signal transducers and activators of transcription (JAK/STAT) signalling in hematopoietic progenitors (prohemocytes), thereby preserving the multipotent character necessary for their differentiation into lamellocytes, a cryptic and dedicated immune cell type required to fight specific immune threats such as wasp parasitism. In this report, on the basis of a knock out generated by homologous recombination, we show that a short type I cytokine-related receptor CG14225/Latran is required for switching off JAK/STAT signalling in prohemocytes. This is a prerequisite to massive differentiation of lamellocytes upon wasp parasitisation. In vivo and cell culture assays indicate that Latran forms heteromers with Domeless, the Drosophila type I cytokine signalling receptor related to mammalian GP130, and antagonises Domeless activity in a dose-dependent manner. Our analysis further shows that a primary immune response to wasp parasitism is a strong decrease in cytokine mRNA levels in the lymph gland, followed by an increase in the latran/domeless ratio. We propose that this sequence of events culminates in the complete inhibition of residual JAK/STAT signalling by Latran. JAK/STAT activity has been associated with several human diseases including leukaemia while knock-out studies in mice point to a central role of this pathway in hematopoiesis and regulation of immune functions. The specific function of Drosophila Latran is, to our knowledge, the first in vivo example of a role for a nonsignalling receptor in controlling a dedicated immune response, and thus raises the question of whether short, nonsignalling receptors also control specific aspects of vertebrate cellular immunity.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Hemocytes/immunology , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Animals , DNA-Binding Proteins/genetics , Down-Regulation , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Hemocytes/metabolism , Homeostasis , Immunity, Cellular , Janus Kinases/genetics , STAT Transcription Factors/genetics , Wasps/physiology
9.
Nature ; 446(7133): 325-8, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17361184

ABSTRACT

Drosophila haemocytes (blood cells) originate from a specialized haematopoietic organ-the lymph gland. Larval haematopoietic progenitors (prohaemocytes) give rise to three types of circulating haemocytes: plasmatocytes, crystal cells and lamellocytes. Lamellocytes, which are devoted to encapsulation of large foreign bodies, only differentiate in response to specific immune threats, such as parasitization by wasps. Here we show that a small cluster of signalling cells, termed the PSC (posterior signalling centre), controls the balance between multipotent prohaemocytes and differentiating haemocytes, and is necessary for the massive differentiation of lamellocytes that follows parasitization. Communication between the PSC and haematopoietic progenitors strictly depends on the PSC-restricted expression of Collier, the Drosophila orthologue of mammalian early B-cell factor. PSC cells act, in a non-cell-autonomous manner, to maintain JAK/STAT signalling activity in prohaemocytes, preventing their premature differentiation. Serrate-mediated Notch signalling from the PSC is required to maintain normal levels of col transcription. The key role of the PSC in controlling blood cell homeostasis is reminiscent of interactions between haematopoietic progenitors and their micro-environment in vertebrates, thus further highlighting the interest of Drosophila as a model system for studying the evolution of haematopoiesis and cellular innate immunity.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Hemocytes/cytology , Hemocytes/metabolism , Homeostasis , Signal Transduction , Animals , Cell Differentiation , Drosophila melanogaster/parasitology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Janus Kinases/metabolism , Larva/cytology , Larva/metabolism , Larva/parasitology , Lymphatic System/cytology , Lymphatic System/metabolism , STAT Transcription Factors/metabolism , Wasps/physiology
10.
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.

11.
Dev Biol ; 346(2): 310-9, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20707995

ABSTRACT

The Drosophila lymph gland (LG) is a model system for studying hematopoiesis and blood cell homeostasis. Here, we investigated the patterns of division and differentiation of pro-hemocytes in normal developmental conditions and response to wasp parasitism, by combining lineage analyses and molecular markers for each of the three hemocyte types. Our results show that the embryonic LG contains primordial hematopoietic cells which actively divide to give rise to a pool of pro-hemocytes. We found no evidence for the existence of bona fide stem cells and rather suggest that Drosophila pro-hemocytes are regulated as a group of cells, rather than individual stem cells. The fate-restriction of plasmatocyte and crystal cell progenitors occurs between the end of embryogenesis and the end of the first larval instar, while Notch activity is required for the differentiation of crystal cells in third instar larvae only. Upon parasitism, lamellocyte differentiation prevents crystal cell differentiation and lowers plasmatocyte production. We also found that a new population of intermediate progenitors appears at the onset of hemocyte differentiation and accounts for the increasing number of differentiated hemocytes in the third larval instar. These findings provide a new framework to identify parameters of developmental plasticity of the Drosophila lymph gland and hemocyte homeostasis in physiological conditions and in response to immunological cues.


Subject(s)
Cell Lineage , Drosophila/embryology , Hematopoietic Stem Cells/cytology , Hemocytes/cytology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Hemocytes/metabolism , Larva/metabolism , Mitosis , Receptors, Notch/genetics , Receptors, Notch/metabolism
12.
Front Immunol ; 12: 719349, 2021.
Article in English | MEDLINE | ID: mdl-34484226

ABSTRACT

In adult mammals, blood cells are formed from hematopoietic stem progenitor cells, which are controlled by a complex cellular microenvironment called "niche". Drosophila melanogaster is a powerful model organism to decipher the mechanisms controlling hematopoiesis, due both to its limited number of blood cell lineages and to the conservation of genes and signaling pathways throughout bilaterian evolution. Insect blood cells or hemocytes are similar to the mammalian myeloid lineage that ensures innate immunity functions. Like in vertebrates, two waves of hematopoiesis occur in Drosophila. The first wave takes place during embryogenesis. The second wave occurs at larval stages, where two distinct hematopoietic sites are identified: subcuticular hematopoietic pockets and a specialized hematopoietic organ called the lymph gland. In both sites, hematopoiesis is regulated by distinct niches. In hematopoietic pockets, sensory neurons of the peripheral nervous system provide a microenvironment that promotes embryonic hemocyte expansion and differentiation. In the lymph gland blood cells are produced from hematopoietic progenitors. A small cluster of cells called Posterior Signaling Centre (PSC) and the vascular system, along which the lymph gland develops, act collectively as a niche, under homeostatic conditions, to control the balance between maintenance and differentiation of lymph gland progenitors. In response to an immune stress such as wasp parasitism, lymph gland hematopoiesis is drastically modified and shifts towards emergency hematopoiesis, leading to increased progenitor proliferation and their differentiation into lamellocyte, a specific blood cell type which will neutralize the parasite. The PSC is essential to control this emergency response. In this review, we summarize Drosophila cellular and molecular mechanisms involved in the communication between the niche and hematopoietic progenitors, both under homeostatic and stress conditions. Finally, we discuss similarities between mechanisms by which niches regulate hematopoietic stem/progenitor cells in Drosophila and mammals.


Subject(s)
Cell Communication , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeostasis , Stem Cell Niche , Stress, Physiological , Animals , Cellular Microenvironment , Drosophila , Hemocytes/cytology , Hemocytes/metabolism , Larva , Models, Biological , Neurons/cytology , Neurons/metabolism , Stem Cell Niche/immunology , Stress, Physiological/immunology
13.
Elife ; 102021 01 04.
Article in English | MEDLINE | ID: mdl-33395389

ABSTRACT

In adult mammals, hematopoiesis, the production of blood cells from hematopoietic stem and progenitor cells (HSPCs), is tightly regulated by extrinsic signals from the microenvironment called 'niche'. Bone marrow HSPCs are heterogeneous and controlled by both endosteal and vascular niches. The Drosophila hematopoietic lymph gland is located along the cardiac tube which corresponds to the vascular system. In the lymph gland, the niche called Posterior Signaling Center controls only a subset of the heterogeneous hematopoietic progenitor population indicating that additional signals are necessary. Here we report that the vascular system acts as a second niche to control lymph gland homeostasis. The FGF ligand Branchless produced by vascular cells activates the FGF pathway in hematopoietic progenitors. By regulating intracellular calcium levels, FGF signaling maintains progenitor pools and prevents blood cell differentiation. This study reveals that two niches contribute to the control ofDrosophila blood cell homeostasis through their differential regulation of progenitors.


Subject(s)
Drosophila/physiology , Fibroblast Growth Factors/metabolism , Hematopoiesis/physiology , Signal Transduction , Animals
14.
Dev Cell ; 8(2): 255-66, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15691766

ABSTRACT

We here identify and characterize an extracellular modulator of Hedgehog signaling in Drosophila, Shifted. Shifted is required for high levels of long-range signaling in the developing wing imaginal disc. Surprisingly, shifted encodes the only Drosophila ortholog of the secreted vertebrate protein Wnt Inhibitory Factor-1 (WIF-1), whose known role is to bind to extracellular Wnts and inhibit their activity. However, Shifted does not regulate Hedgehog signaling by affecting Wingless or Wnt signaling. We show instead that Shifted is a secreted protein that acts over a long distance and is required for the normal accumulation of Hh protein and its movement in the wing. Our data further indicate that Shf interacts with Hh and the heparan sulfate proteoglycans. Therefore, we propose that Shf stabilizes the interaction between Hh and the proteoglycans, an unexpected role for a member of the WIF-1 family.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , Adaptor Proteins, Signal Transducing , Alleles , Amino Acid Sequence , Animals , Animals, Genetically Modified , Carrier Proteins/genetics , Carrier Proteins/physiology , Cholesterol/metabolism , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Female , Hedgehog Proteins , Heparan Sulfate Proteoglycans/metabolism , Humans , Male , Models, Biological , Molecular Sequence Data , Mutation , Phenotype , Repressor Proteins/genetics , Repressor Proteins/physiology , Sequence Homology, Amino Acid , Signal Transduction , Wings, Animal/growth & development , Wings, Animal/metabolism
15.
Elife ; 92020 12 24.
Article in English | MEDLINE | ID: mdl-33357377

ABSTRACT

Organisms rely on inducible and constitutive immune defences to combat infection. Constitutive immunity enables a rapid response to infection but may carry a cost for uninfected individuals, leading to the prediction that it will be favoured when infection rates are high. When we exposed populations of Drosophila melanogaster to intense parasitism by the parasitoid wasp Leptopilina boulardi, they evolved resistance by developing a more reactive cellular immune response. Using single-cell RNA sequencing, we found that immune-inducible genes had become constitutively upregulated. This was the result of resistant larvae differentiating precursors of specialized immune cells called lamellocytes that were previously only produced after infection. Therefore, populations evolved resistance by genetically hard-wiring the first steps of an induced immune response to become constitutive.


Subject(s)
Biological Evolution , Disease Resistance/immunology , Drosophila melanogaster/immunology , Immunity, Cellular/immunology , Infections/immunology , Animals , Disease Resistance/genetics , Drosophila melanogaster/parasitology , Female , Gene Expression Regulation , Hemocytes/immunology , Larva/immunology , Male , Wasps
16.
Dev Biol ; 315(1): 232-42, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18234173

ABSTRACT

Proper sampling of sensory inputs critically depends upon neuron morphogenesis and expression of sensory channels. The highly stereotyped organisation of the Drosophila peripheral nervous system (PNS) provides a model to study neuronal determination and morphogenesis. Here, we report that Collier/Knot (Col/Kn), the Drosophila member of the COE family of transcription factors, is transiently expressed in the subset of multidendritic arborisation (da) sensory neurons that display an highly branched dendritic arborisation, class IV neurons. When lacking Col activity, class IV da neurons are formed but display a reduced dendrite arborisation. Col control on dendrite branching is distinct from that exerted by Cut, another transcription factor expressed in class IV neurons and necessary for proper dendrite morphogenesis. Col is also required for the class IV da-specific expression of pickpocket (ppk), which encodes a degenerin/epithelial sodium channel subunit required for larval locomotion. Characterisation of the col upstream region identified a 9-kb cis-regulatory region driving col expression in all class IV md neurons, even though these originate from two types of sensory precursor cells. Altogether, these findings indicate that col is required in at least two distinct programs that control the morphological and sensory specificity of Drosophila md neurons.


Subject(s)
Cell Differentiation/physiology , Dendrites/physiology , Drosophila Proteins/physiology , Drosophila/cytology , Neurons, Afferent/cytology , Transcription Factors/physiology , Animals , Cell Lineage , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Models, Biological , Neurons, Afferent/classification , Neurons, Afferent/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
17.
BMC Evol Biol ; 8: 131, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18454855

ABSTRACT

BACKGROUND: The increasing number of available genomic sequences makes it now possible to study the evolutionary history of specific genes or gene families. Transcription factors (TFs) involved in regulation of gene-specific expression are key players in the evolution of metazoan development. The low complexity COE (Collier/Olfactory-1/Early B-Cell Factor) family of transcription factors constitutes a well-suited paradigm for studying evolution of TF structure and function, including the specific question of protein modularity. Here, we compare the structure of coe genes within the metazoan kingdom and report on the mechanism behind a vertebrate-specific exon duplication. RESULTS: COE proteins display a modular organisation, with three highly conserved domains : a COE-specific DNA-binding domain (DBD), an Immunoglobulin/Plexin/transcription (IPT) domain and an atypical Helix-Loop-Helix (HLH) motif. Comparison of the splice structure of coe genes between cnidariae and bilateriae shows that the ancestral COE DBD was built from 7 separate exons, with no evidence for exon shuffling with other metazoan gene families. It also confirms the presence of an ancestral H1LH2 motif present in all COE proteins which partly overlaps the repeated H2d-H2a motif first identified in rodent EBF. Electrophoretic Mobility Shift Assays show that formation of COE dimers is mediated by this ancestral motif. The H2d-H2a alpha-helical repetition appears to be a vertebrate characteristic that originated from a tandem exon duplication having taken place prior to the splitting between gnathostomes and cyclostomes. We put-forward a two-step model for the inclusion of this exon in the vertebrate transcripts. CONCLUSION: Three main features in the history of the coe gene family can be inferred from these analyses: (i) each conserved domain of the ancestral coe gene was built from multiple exons and the same scattered structure has been maintained throughout metazoan evolution. (ii) There exists a single coe gene copy per metazoan genome except in vertebrates. The H2a-H2d duplication that is specific to vertebrate proteins provides an example of a novel vertebrate characteristic, which may have been fixed early in the gnathostome lineage. (iii) This duplication provides an interesting example of counter-selection of alternative splicing.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Evolution, Molecular , Helix-Loop-Helix Motifs/genetics , Transcription Factors/genetics , Vertebrates/genetics , Amino Acid Sequence , Animals , Bayes Theorem , Drosophila/genetics , Exons , Phylogeny , Sequence Alignment
18.
Trends Genet ; 20(10): 498-505, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15363904

ABSTRACT

The development of the Drosophila wing is a classical model for studying the genetic control of tissue size, shape and patterning. A detailed picture of how positional information is interpreted by cells in the imaginal disc and translated into the adult wing vein pattern has recently emerged. It highlights the central role of dose-dependent activation of distinct cell transcription programs in response to the Hedgehog (Hh) and Decapentaplegic (Dpp) morphogens, as well as an early role of Notch signalling, in connecting the positioning of vein primordia and vein differentiation proper. The biochemical basis of the cross-talk that operates between these different signalling pathways is less well understood. New strategies made possible by the genome sequencing of several insect models should provide an important complement to the knowledge obtained from >60 years of genetic studies.


Subject(s)
Biological Evolution , Drosophila/anatomy & histology , Drosophila/genetics , Gene Expression Regulation, Developmental/genetics , Signal Transduction/genetics , Wings, Animal/anatomy & histology , Animals , Cell Differentiation/genetics , Drosophila Proteins/metabolism , Genomics , Hedgehog Proteins , Membrane Proteins/metabolism , Morphogenesis , Receptors, Notch , Species Specificity
19.
PLoS Biol ; 2(8): E196, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15314643

ABSTRACT

Drosophila immune response involves three types of hemocytes ('blood cells'). One cell type, the lamellocyte, is induced to differentiate only under particular conditions, such as parasitization by wasps. Here, we have investigated the mechanisms underlying the specification of lamellocytes. We first show that collier (col), the Drosophila orthologue of the vertebrate gene encoding early B-cell factor (EBF), is expressed very early during ontogeny of the lymph gland, the larval hematopoietic organ. In this organ, Col expression prefigures a specific posterior region recently proposed to act as a signalling centre, the posterior signalling centre (PSC). The complete lack of lamellocytes in parasitized col mutant larvae revealed the critical requirement for Col activity in specification of this cell type. In wild-type larvae, Col expression remains restricted to the PSC following parasitization, despite the massive production of lamellocytes. We therefore propose that Col endows PSC cells with the capacity to relay an instructive signal that orients hematopoietic precursors towards the lamellocyte fate in response to parasitization. Considered together with the role of EBF in lymphopoiesis, these findings suggest new parallels in cellular immunity between Drosophila and vertebrates. Further investigations on Col/EBF expression and function in other phyla should provide fresh insight into the evolutionary origin of lymphoid cells.


Subject(s)
Drosophila Proteins/physiology , Drosophila/parasitology , Gene Expression Regulation, Developmental , Hemocytes/immunology , Transcription Factors/physiology , Animals , Cell Differentiation , Crosses, Genetic , Drosophila/embryology , Drosophila Proteins/metabolism , Evolution, Molecular , Hemocytes/parasitology , Immune System/pathology , In Situ Hybridization , Insect Proteins/metabolism , Lymph Nodes/pathology , Models, Biological , Receptors, Notch/metabolism , Signal Transduction , Transcription Factors/metabolism , Transgenes , Wasps
20.
Elife ; 62017 11 01.
Article in English | MEDLINE | ID: mdl-29091025

ABSTRACT

Hematopoietic stem/progenitor cells in the adult mammalian bone marrow ensure blood cell renewal. Their cellular microenvironment, called 'niche', regulates hematopoiesis both under homeostatic and immune stress conditions. In the Drosophila hematopoietic organ, the lymph gland, the posterior signaling center (PSC) acts as a niche to regulate the hematopoietic response to immune stress such as wasp parasitism. This response relies on the differentiation of lamellocytes, a cryptic cell type, dedicated to pathogen encapsulation and killing. Here, we establish that Toll/NF-κB pathway activation in the PSC in response to wasp parasitism non-cell autonomously induces the lymph gland immune response. Our data further establish a regulatory network where co-activation of Toll/NF-κB and EGFR signaling by ROS levels in the PSC/niche controls lymph gland hematopoiesis under parasitism. Whether a similar regulatory network operates in mammals to control emergency hematopoiesis is an open question.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/immunology , ErbB Receptors/metabolism , Hematopoiesis , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Receptors, Invertebrate Peptide/metabolism , Toll-Like Receptors/metabolism , Wasps/immunology , Animals , Drosophila/parasitology , Host-Parasite Interactions , Immunity, Innate
SELECTION OF CITATIONS
SEARCH DETAIL