Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
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.
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.

3.
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
4.
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
5.
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
6.
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
7.
FEBS Lett ; 590(22): 4034-4051, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27455465

ABSTRACT

The emergence of hematopoietic progenitors and their differentiation into various highly specialized blood cell types constitute a finely tuned process. Unveiling the genetic cascades that control blood cell progenitor fate and understanding how they are modulated in response to environmental changes are two major challenges in the field of hematopoiesis. In the last 20 years, many studies have established important functional analogies between blood cell development in vertebrates and in the fruit fly, Drosophila melanogaster. Thereby, Drosophila has emerged as a powerful genetic model for studying mechanisms that control hematopoiesis during normal development or in pathological situations. Moreover, recent advances in Drosophila have highlighted how intricate cell communication networks and microenvironmental cues regulate blood cell homeostasis. They have also revealed the striking plasticity of Drosophila mature blood cells and the presence of different sites of hematopoiesis in the larva. This review provides an overview of Drosophila hematopoiesis during development and summarizes our current knowledge on the molecular processes controlling larval hematopoiesis, both under normal conditions and in response to an immune challenge, such as wasp parasitism.


Subject(s)
Blood Cells/cytology , Drosophila melanogaster/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells , Animals , Cell Communication , Cell Differentiation/genetics , Cellular Microenvironment/genetics , Drosophila melanogaster/growth & development , Humans , Larva/genetics , Larva/growth & development
8.
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
9.
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
10.
Nat Commun ; 7: 11634, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27193394

ABSTRACT

Self-renewal and differentiation of mammalian haematopoietic stem cells (HSCs) are controlled by a specialized microenvironment called 'the niche'. In the bone marrow, HSCs receive signals from both the endosteal and vascular niches. The posterior signalling centre (PSC) of the larval Drosophila haematopoietic organ, the lymph gland, regulates blood cell differentiation under normal conditions and also plays a key role in controlling haematopoiesis under immune challenge. Here we report that the Drosophila vascular system also contributes to the lymph gland homoeostasis. Vascular cells produce Slit that activates Robo receptors in the PSC. Robo activation controls proliferation and clustering of PSC cells by regulating Myc, and small GTPase and DE-cadherin activity, respectively. These findings reveal that signals from the vascular system contribute to regulating the rate of blood cell differentiation via the regulation of PSC morphology.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/physiology , Hematopoiesis , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Animals , Cadherins/metabolism , Cardiovascular System/metabolism , Drosophila/cytology , Larva/cytology , Larva/physiology , Nerve Tissue Proteins/genetics , Proteoglycans/metabolism , Receptors, Immunologic/genetics , cdc42 GTP-Binding Protein/metabolism , Roundabout Proteins
11.
PLoS One ; 11(2): e0148978, 2016.
Article in English | MEDLINE | ID: mdl-26866694

ABSTRACT

Blood cell production in the Drosophila hematopoietic organ, the lymph gland, is controlled by intrinsic factors and extrinsic signals. Initial analysis of Collier/Early B Cell Factor function in the lymph gland revealed the role of the Posterior Signaling Center (PSC) in mounting a dedicated cellular immune response to wasp parasitism. Further, premature blood cell differentiation when PSC specification or signaling was impaired, led to assigning the PSC a role equivalent to the vertebrate hematopoietic niche. We report here that Collier is expressed in a core population of lymph gland progenitors and cell autonomously maintains this population. The PSC contributes to lymph gland homeostasis by regulating blood cell differentiation, rather than by maintaining core progenitors. In addition to PSC signaling, switching off Collier expression in progenitors is required for efficient immune response to parasitism. Our data show that two independent sites of Collier/Early B Cell Factor expression, hematopoietic progenitors and the PSC, achieve control of hematopoiesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Hemocytes/cytology , Lymph/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Hematopoiesis , Homeostasis , Host-Parasite Interactions , Immune System , In Situ Hybridization , Mitosis , RNA Interference , Signal Transduction , Stem Cell Niche , Stem Cells , Wasps
12.
PLoS One ; 10(7): e0133387, 2015.
Article in English | MEDLINE | ID: mdl-26204530

ABSTRACT

Collier, the single Drosophila COE (Collier/EBF/Olf-1) transcription factor, is required in several developmental processes, including head patterning and specification of muscle and neuron identity during embryogenesis. To identify direct Collier (Col) targets in different cell types, we used ChIP-seq to map Col binding sites throughout the genome, at mid-embryogenesis. In vivo Col binding peaks were associated to 415 potential direct target genes. Gene Ontology analysis revealed a strong enrichment in proteins with DNA binding and/or transcription-regulatory properties. Characterization of a selection of candidates, using transgenic CRM-reporter assays, identified direct Col targets in dorso-lateral somatic muscles and specific neuron types in the central nervous system. These data brought new evidence that Col direct control of the expression of the transcription regulators apterous and eyes-absent (eya) is critical to specifying neuronal identities. They also showed that cross-regulation between col and eya in muscle progenitor cells is required for specification of muscle identity, revealing a new parallel between the myogenic regulatory networks operating in Drosophila and vertebrates. Col regulation of eya, both in specific muscle and neuronal lineages, may illustrate one mechanism behind the evolutionary diversification of Col biological roles.


Subject(s)
Body Patterning/genetics , Chromosome Mapping , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Binding Sites , Embryo, Nonmammalian , Gene Regulatory Networks , Signal Transduction/genetics
13.
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
14.
JAKSTAT ; 2(3): e25700, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24069567

ABSTRACT

Genetic alterations affecting the JAK-STAT signaling pathway are linked to several malignancies and hematological disorders in humans. Despite being one of the most extensively studied pathways, there remain many gaps to fill. JAK-STAT components are widely conserved during evolution. Here, we review the known roles of the JAK-STAT pathway in Drosophila immunity: controlling the different steps of hematopoiesis, both under physiological conditions and in response to immune challenge, and contributing to antiviral responses. We then summarize what is currently known about JAK-STAT signaling in renewal of the adult intestine, under physiological conditions or in response to ingestion of pathogenic bacteria.

15.
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
16.
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
17.
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
18.
Dis Model Mech ; 4(4): 439-45, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21669932

ABSTRACT

Vertebrate haematopoietic stem cells (HSCs) give rise to a hierarchically organised set of progenitors for erythroid, myeloid, lymphoid and megakaryocyte lineages, and are responsible for lifelong maintenance of the blood system. Dysregulation of the haematopoietic differentiation programme is at the origin of numerous pathologies, including leukaemias. With the discoveries that many transcriptional regulators and signalling pathways controlling blood cell development are conserved between humans and Drosophila melanogaster, the fruit fly has become a good model for investigating the mechanisms underlying the generation of blood cell lineages and blood cell homeostasis. In this review article, we discuss how genetic and molecular studies of Drosophila haematopoiesis can contribute to our understanding of the haematopoietic niche, as well as of the origin and/or progression of haematopoietic malignancies in humans.


Subject(s)
Disease Models, Animal , Drosophila melanogaster/genetics , Hematopoiesis/genetics , Leukemia/genetics , Animals , Leukemia/physiopathology , Mammals/physiology , Reactive Oxygen Species/metabolism
19.
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
20.
Int J Dev Biol ; 54(6-7): 1117-25, 2010.
Article in English | MEDLINE | ID: mdl-20711989

ABSTRACT

Over the years, the fruit fly Drosophila melanogaster has become a major invertebrate model to study developmental and evolutionary aspects of both humoral and cellular aspects of innate immunity. Drosophila hematopoiesis which supplies three types of circulating hemocytes, occurs in two spatially and temporally distinct phases during development. The first embryonic phase is described in detail in accompanying reviews in this Int. J. Dev. Biol. Special Issue. The second phase takes place at the end of larval development in a specialised hematopoietic organ, termed the lymph gland. We review here recent studies on the ontogeny of the lymph gland, focusing on the formation and role of the Posterior Signalling Center which acts as a niche for hematopoietic progenitors. We then report recent progress in understanding the dedicated cellular immune response of Drosophila larvae against parasitization by Hymenopterae, a common threat for many Dipterae. This response involves the differentiation of lamellocytes, a cryptic cell fate, revealing the high degree of plasticity of Drosophila hematopoiesis. We end up by integrating studies in Drosophila within a more general picture of insect hematopoiesis and hemocyte homeostasis.


Subject(s)
Drosophila melanogaster/immunology , Hemocytes/immunology , Homeostasis/immunology , Animals , Drosophila melanogaster/embryology , Drosophila melanogaster/parasitology , Hematopoiesis/immunology , Hemocytes/cytology , Hymenoptera/immunology , Immune System/blood supply , Immune System/immunology , Larva/immunology , Larva/parasitology , Models, Immunological
SELECTION OF CITATIONS
SEARCH DETAIL
...