Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 9: 795680, 2021.
Article in English | MEDLINE | ID: mdl-35178397

ABSTRACT

GATA transcription factors play crucial roles in various developmental processes in organisms ranging from flies to humans. In mammals, GATA factors are characterized by the presence of two highly conserved domains, the N-terminal (N-ZnF) and the C-terminal (C-ZnF) zinc fingers. The Drosophila GATA factor Serpent (Srp) is produced in different isoforms that contains either both N-ZnF and C-ZnF (SrpNC) or only the C-ZnF (SrpC). Here, we investigated the functional roles ensured by each of these isoforms during Drosophila development. Using the CRISPR/Cas9 technique, we generated new mutant fly lines deleted for one (ΔsrpNC) or the other (ΔsrpC) encoded isoform, and a third one with a single point mutation in the N-ZnF that alters its interaction with its cofactor, the Drosophila FOG homolog U-shaped (Ush). Analysis of these mutants revealed that the Srp zinc fingers are differentially required for Srp to fulfill its functions. While SrpC is essential for embryo to adult viability, SrpNC, which is the closest conserved isoform to that of vertebrates, is not. However, to ensure its specific functions in larval hematopoiesis and fertility, Srp requires the presence of both N- and C-ZnF (SrpNC) and interaction with its cofactor Ush. Our results also reveal that in vivo the presence of N-ZnF restricts rather than extends the ability of GATA factors to regulate the repertoire of C-ZnF bound target genes.

2.
Nucleic Acids Res ; 48(3): 1206-1224, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31799607

ABSTRACT

The histone demethylase LSD1 is a key chromatin regulator that is often deregulated in cancer. Its ortholog, dLsd1 plays a crucial role in Drosophila oogenesis; however, our knowledge of dLsd1 function is insufficient to explain its role in the ovary. Here, we have performed genome-wide analysis of dLsd1 binding in the ovary, and we document that dLsd1 is preferentially associated to the transcription start site of developmental genes. We uncovered an unanticipated interplay between dLsd1 and the GATA transcription factor Serpent and we report an unexpected role for Serpent in oogenesis. Besides, our transcriptomic data show that reducing dLsd1 levels results in ectopic transposable elements (TE) expression correlated with changes in H3K4me2 and H3K9me2 at TE loci. In addition, our results suggest that dLsd1 is required for Piwi dependent TE silencing. Hence, we propose that dLsd1 plays crucial roles in establishing specific gene expression programs and in repressing transposons during oogenesis.


Subject(s)
DNA Transposable Elements/genetics , Drosophila Proteins/genetics , GATA Transcription Factors/genetics , Oogenesis/genetics , Oxidoreductases, N-Demethylating/genetics , Animals , Argonaute Proteins/genetics , Chromatin/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Female , Gene Expression Regulation, Developmental/genetics , Genes, Developmental/genetics , Histones/genetics , Ovary/growth & development , Ovary/metabolism , Transcription Initiation Site
3.
Mol Cell Biol ; 39(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30670567

ABSTRACT

DNA-bound transcription factors (TFs) governing developmental gene regulation have been proposed to recruit polymerase II machinery at gene promoters through specific interactions with dedicated subunits of the evolutionarily conserved Mediator (MED) complex. However, whether such MED subunit-specific functions and partnerships have been conserved during evolution has been poorly investigated. To address this issue, we generated the first Drosophila melanogaster loss-of-function mutants for Med1, known as a specific cofactor for GATA TFs and hormone nuclear receptors in mammals. We show that Med1 is required for cell proliferation and hematopoietic differentiation depending on the GATA TF Serpent (Srp). Med1 physically binds Srp in cultured cells and in vitro through its conserved GATA zinc finger DNA-binding domain and the divergent Med1 C terminus. Interestingly, GATA-Srp interaction occurs through the longest Med1 isoform, suggesting a functional diversity of MED complex populations. Furthermore, we show that Med1 acts as a coactivator for the GATA factor Pannier during thoracic development. In conclusion, the Med1 requirement for GATA-dependent regulatory processes is a common feature in insects and mammals, although binding interfaces have diverged. Further work in Drosophila should bring valuable insights to fully understand GATA-MED functional partnerships, which probably involve other MED subunits depending on the cellular context.


Subject(s)
Mediator Complex Subunit 1/metabolism , Mediator Complex/metabolism , Animals , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , GATA Transcription Factors/metabolism , GATA1 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/genetics , Loss of Function Mutation , Mediator Complex Subunit 1/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism
4.
PLoS Genet ; 13(7): e1006932, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28742844

ABSTRACT

A tight regulation of transcription factor activity is critical for proper development. For instance, modifications of RUNX transcription factors dosage are associated with several diseases, including hematopoietic malignancies. In Drosophila, Myeloid Leukemia Factor (MLF) has been shown to control blood cell development by stabilizing the RUNX transcription factor Lozenge (Lz). However, the mechanism of action of this conserved family of proteins involved in leukemia remains largely unknown. Here we further characterized MLF's mode of action in Drosophila blood cells using proteomic, transcriptomic and genetic approaches. Our results show that MLF and the Hsp40 co-chaperone family member DnaJ-1 interact through conserved domains and we demonstrate that both proteins bind and stabilize Lz in cell culture, suggesting that MLF and DnaJ-1 form a chaperone complex that directly regulates Lz activity. Importantly, dnaj-1 loss causes an increase in Lz+ blood cell number and size similarly as in mlf mutant larvae. Moreover we find that dnaj-1 genetically interacts with mlf to control Lz level and Lz+ blood cell development in vivo. In addition, we show that mlf and dnaj-1 loss alters Lz+ cell differentiation and that the increase in Lz+ blood cell number and size observed in these mutants is caused by an overactivation of the Notch signaling pathway. Finally, using different conditions to manipulate Lz activity, we show that high levels of Lz are required to repress Notch transcription and signaling. All together, our data indicate that the MLF/DnaJ-1-dependent increase in Lz level allows the repression of Notch expression and signaling to prevent aberrant blood cell development. Thus our findings establish a functional link between MLF and the co-chaperone DnaJ-1 to control RUNX transcription factor activity and Notch signaling during blood cell development in vivo.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , HSP40 Heat-Shock Proteins/genetics , Hematopoiesis/genetics , Receptors, Notch/genetics , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Drosophila Proteins/biosynthesis , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Larva/genetics , Larva/growth & development , Proteomics , Receptors, Notch/biosynthesis , Signal Transduction/genetics
8.
Proc Natl Acad Sci U S A ; 112(29): 9052-7, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26150488

ABSTRACT

The maintenance of stem or progenitor cell fate relies on intrinsic factors as well as local cues from the cellular microenvironment and systemic signaling. In the lymph gland, an hematopoietic organ in Drosophila larva, a group of cells called the Posterior Signaling Centre (PSC), whose specification depends on the EBF transcription factor Collier (Col) and the HOX factor Antennapedia (Antp), has been proposed to form a niche required to maintain the pool of hematopoietic progenitors (prohemocytes). In contrast with this model, we show here that genetic ablation of the PSC does not cause an increase in blood cell differentiation or a loss of blood cell progenitors. Furthermore, although both col and Antp mutant larvae are devoid of PSC, the massive prohemocyte differentiation observed in col mutant is not phenocopied in Antp mutant. Interestingly, beside its expression in the PSC, Col is also expressed at low levels in prohemocytes and we show that this expression persists in PSC-ablated and Antp mutant larvae. Moreover, targeted knockdown and rescue experiments indicate that Col expression is required in the prohemocytes to prevent their differentiation. Together, our findings show that the PSC is dispensable for blood cell progenitor maintenance and reveal the key role of the conserved transcription factor Col as an intrinsic regulator of hematopoietic progenitor fate.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Stem Cell Niche , Transcription Factors/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Deletion , Green Fluorescent Proteins/metabolism , Hemocytes/cytology , Hemocytes/metabolism , Larva/cytology , Larva/metabolism , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mutation , Phenotype , RNA Interference , Signal Transduction
9.
Fly (Austin) ; 9(4): 160-4, 2015.
Article in English | MEDLINE | ID: mdl-26925971

ABSTRACT

Drosophila lymph gland, a larval haematopoietic organ, has emerged as a popular model to study regulatory mechanisms controlling blood cell progenitor fate. In this organ, the Posterior Signaling Center (PSC), a small group of cells expressing the EBF transcription factor Collier, has been proposed to act as a niche required for progenitor maintenance. Accordingly, several reports showed that PSC size/activity modulation impacts on blood cell differentiation. Yet our recent results challenge this model. Indeed, we found that PSC ablation does not affect haematopoietic progenitor maintenance. This unexpected result led us to reinvestigate the role of the PSC and collier in hematopoiesis. Consistent with previous findings, the PSC appears required for the production of a specialized blood cell type in response to parasitization. Moreover, our results indicate that the massive blood cell differentiation observed in collier mutant larvae is not due to the lack of PSC but to collier expression within the haematopoietic progenitors. We thus propose a paradigm shift whereby larval blood cell progenitor maintenance is largely independent of the PSC but requires the cell-autonomous function of collier.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Hematopoiesis , Transcription Factors/metabolism , Animals , Drosophila melanogaster/cytology , Larva/cytology , Larva/metabolism , Signal Transduction , Stem Cell Niche
10.
Transcription ; 3(5): 250-4, 2012.
Article in English | MEDLINE | ID: mdl-22885977

ABSTRACT

Even though deregulation of human MLF1, the founding member of the Myeloid Leukemia Factor family, has been associated with acute myeloid leukemia, the function and mode of action of this family of genes have remained rather mysterious. Yet, recent findings in Drosophila shed new light on their biological activity and suggest that they play an important role in hematopoiesis and leukemia, notably by regulating the stability of RUNX transcription factors, another family of conserved proteins with prominent roles in normal and malignant blood cell development.


Subject(s)
Core Binding Factor alpha Subunits/metabolism , Hematopoiesis/physiology , Leukemia, Myeloid/metabolism , Proteins/metabolism , Animals , Cell Cycle Proteins , DNA-Binding Proteins , Drosophila Proteins/metabolism , Humans
11.
Development ; 139(10): 1713-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22510984

ABSTRACT

The interconnected Insulin/IGF signaling (IlS) and Target of Rapamycin (TOR) signaling pathways constitute the main branches of the nutrient-sensing system that couples growth to nutritional conditions in Drosophila. Here, we addressed the influence of these pathways and of diet restriction on the balance between the maintenance of multipotent hematopoietic progenitors and their differentiation in the Drosophila lymph gland. In this larval hematopoietic organ, a pool of stem-like progenitor blood cells (prohemocytes) is kept undifferentiated in response to signaling from a specialized group of cells forming the posterior signaling center (PSC), which serves as a stem cell niche. We show that, reminiscent of the situation in human, loss of the negative regulator of IIS Pten results in lymph gland hyperplasia, aberrant blood cell differentiation and hematopoietic progenitor exhaustion. Using site-directed loss- and gain-of-function analysis, we demonstrate that components of the IIS/TOR pathways control lymph gland homeostasis at two levels. First, they cell-autonomously regulate the size and activity of the hematopoietic niche. Second, they are required within the prohemocytes to control their growth and maintenance. Moreover, we show that diet restriction or genetic alteration mimicking amino acid deprivation triggers progenitor cell differentiation. Hence, our study highlights the role of the IIS/TOR pathways in orchestrating hematopoietic progenitor fate and links blood cell fate to nutritional status.


Subject(s)
Drosophila Proteins/metabolism , Insulin/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Drosophila , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , In Situ Hybridization , Insulin/genetics , Lymphatic System/embryology , Lymphatic System/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
12.
Proc Natl Acad Sci U S A ; 109(13): 4986-91, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22411814

ABSTRACT

Defining the function of the genes that, like RUNX1, are deregulated in blood cell malignancies represents an important challenge. Myeloid leukemia factors (MLFs) constitute a poorly characterized family of conserved proteins whose founding member, MLF1, has been associated with acute myeloid leukemia in humans. To gain insight into the functions of this family, we investigated the role of the Drosophila MLF homolog during blood cell development. Here we report that mlf controls the homeostasis of the Drosophila hematopoietic system. Notably, mlf participates in a positive feedback loop to fine tune the activity of the RUNX transcription factor Lozenge (LZ) during development of the crystal cells, one of the two main blood cell lineages in Drosophila. At the molecular level, our data in cell cultures and in vivo strongly suggest that MLF controls the number of crystal cells by protecting LZ from degradation. Remarkably, it appears that the human MLF1 protein can substitute for MLF in the crystal cell lineage. In addition, MLF stabilizes the human oncogenic fusion protein RUNX1-ETO and is required for RUNX1-ETO-induced blood cell disorders in a Drosophila model of leukemia. Finally, using the human leukemic blood cell line Kasumi-1, we show that MLF1 depletion impairs RUNX1-ETO accumulation and reduces RUNX1-ETO-dependent proliferation. Thus, we propose that the regulation of RUNX protein levels is a conserved feature of MLF family members that could be critical for normal and pathological blood cell development.


Subject(s)
Conserved Sequence/genetics , Core Binding Factor alpha Subunits/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Hematopoiesis , Transcription Factors/metabolism , Animals , Cell Lineage , Core Binding Factor Alpha 2 Subunit/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Humans , Larva/cytology , Larva/metabolism , Oncogene Proteins, Fusion/metabolism , Phenotype , Protein Stability , Proteolysis , Transcriptional Activation/genetics
13.
Prog Mol Biol Transl Sci ; 100: 51-82, 2011.
Article in English | MEDLINE | ID: mdl-21377624

ABSTRACT

The basic cellular processes deregulated during carcinogenesis and the vast majority of the genes implicated in cancer appear conserved from humans to flies. This conservation, together with an ever-expanding fly genetic toolbox, has made of Drosophila melanogaster a remarkably profitable model to study many fundamental aspects of carcinogenesis. In particular, Drosophila has played a major role in the identification of genes and pathways implicated in cancer and in disclosing novel functional relationships between cancer genes. It has also proved to be a genetically tractable system where to mimic cancer-like situations and characterize the mode of action of human oncogenes. Here, we outline some advances in the study of cancer, both at the basic and more translational levels, which have benefited from research carried out in flies.


Subject(s)
Disease Models, Animal , Drosophila melanogaster/physiology , Neoplasms/pathology , Animals , Cell Differentiation , Cell Polarity , Cell Proliferation
14.
PLoS One ; 6(3): e17763, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21423573

ABSTRACT

BACKGROUND: Genetic analysis of the Drosophila septate junctions has greatly contributed to our understanding of the mechanisms controlling the assembly of these adhesion structures, which bear strong similarities with the vertebrate tight junctions and the paranodal septate junctions. These adhesion complexes share conserved molecular components and have a common function: the formation of paracellular barriers restraining the diffusion of solutes through epithelial and glial envelopes. METHODOLOGY/PRINCIPAL FINDINGS: In this work we characterise the function of the Drosophila cold gene, that codes for a protein belonging to the Ly6 superfamily of extracellular ligands. Analysis of cold mutants shows that this gene is specifically required for the organisation of the septate junctions in epithelial tissues and in the nervous system, where its contribution is essential for the maintenance of the blood-brain barrier. We show that cold acts in a cell autonomous way, and we present evidence indicating that this protein could act as a septate junction component. CONCLUSION/SIGNIFICANCE: We discuss the specific roles of cold and three other Drosophila members of the Ly6 superfamily that have been shown to participate in a non-redundant way in the process of septate junction assembly. We propose that vertebrate Ly6 proteins could fulfill analogous roles in tight junctions and/or paranodal septate junctions.


Subject(s)
Blood-Brain Barrier/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Tight Junctions/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Ectoderm/cytology , Ectoderm/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Genes, Insect/genetics , Larva/cytology , Larva/metabolism , Molecular Sequence Data , Morphogenesis , Neuroglia/cytology , Neuroglia/metabolism , Phenotype , Protein Transport , Salivary Glands/cytology , Salivary Glands/metabolism , Subcellular Fractions/metabolism , Trachea/cytology , Trachea/embryology , Trachea/metabolism , Wings, Animal/cytology , Wings, Animal/metabolism
15.
Int J Dev Biol ; 54(6-7): 1107-15, 2010.
Article in English | MEDLINE | ID: mdl-20711988

ABSTRACT

Transcription factors play a key role in regulating blood cell fate choice and differentiation. In this review, we examine current knowledge of the function and mode of action of the transcription factors implicated in haematopoiesis in Drosophila. We particularly emphasize regulation by transcription factors and cofactors, such as GATA, FOG and RUNX, whose homologues in mammals also control blood cell formation and we discuss the cross talks between these transcriptional regulators at the different stages of haematopoietic cell fate decision.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , Hematopoiesis/physiology , Transcription Factors/physiology , Animals , Core Binding Factor alpha Subunits/genetics , Drosophila/embryology , Drosophila/genetics , Drosophila Proteins/genetics , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , GATA Transcription Factors/genetics , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Transcription Factors/genetics
16.
BMC Dev Biol ; 10: 65, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20540764

ABSTRACT

BACKGROUND: In metazoans, the hematopoietic system plays a key role both in normal development and in defense of the organism. In Drosophila, the cellular immune response involves three types of blood cells: plasmatocytes, crystal cells and lamellocytes. This last cell type is barely present in healthy larvae, but its production is strongly induced upon wasp parasitization or in mutant contexts affecting larval blood cell homeostasis. Notably, several zygotic mutations leading to melanotic mass (or "tumor") formation in larvae have been associated to the deregulated differentiation of lamellocytes. To gain further insights into the gene regulatory network and the mechanisms controlling larval blood cell homeostasis, we conducted a tissue-specific loss of function screen using hemocyte-specific Gal4 drivers and UAS-dsRNA transgenic lines. RESULTS: By targeting around 10% of the Drosophila genes, this in vivo RNA interference screen allowed us to recover 59 melanotic tumor suppressor genes. In line with previous studies, we show that melanotic tumor formation is associated with the precocious differentiation of stem-cell like blood progenitors in the larval hematopoietic organ (the lymph gland) and the spurious differentiation of lamellocytes. We also find that melanotic tumor formation can be elicited by defects either in the fat body, the embryo-derived hemocytes or the lymph gland. In addition, we provide a definitive confirmation that lymph gland is not the only source of lamellocytes as embryo-derived plasmatocytes can differentiate into lamellocytes either upon wasp infection or upon loss of function of the Friend of GATA cofactor U-shaped. CONCLUSIONS: In this study, we identify 55 genes whose function had not been linked to blood cell development or function before in Drosophila. Moreover our analyses reveal an unanticipated plasticity of embryo-derived plasmatocytes, thereby shedding new light on blood cell lineage relationship, and pinpoint the Friend of GATA transcription cofactor U-shaped as a key regulator of the plasmatocyte to lamellocyte transformation.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Gene Regulatory Networks , Homeostasis , Animals , Drosophila melanogaster/immunology , Drosophila melanogaster/metabolism , Genes, Tumor Suppressor , Hematopoiesis , Hemocytes/cytology , Hemocytes/immunology , RNA Interference
17.
Mol Cell Biol ; 30(11): 2837-48, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20368357

ABSTRACT

Transcription factors of the RUNX and GATA families play key roles in the control of cell fate choice and differentiation, notably in the hematopoietic system. During Drosophila hematopoiesis, the RUNX factor Lozenge and the GATA factor Serpent cooperate to induce crystal cell differentiation. We used Serpent/Lozenge-activated transcription as a paradigm to identify modulators of GATA/RUNX activity by a genome-wide RNA interference screen in cultured Drosophila blood cells. Among the 129 factors identified, several belong to the Mediator complex. Mediator is organized in three modules plus a regulatory "CDK8 module," composed of Med12, Med13, CycC, and Cdk8, which has long been thought to behave as a single functional entity. Interestingly, our data demonstrate that Med12 and Med13 but not CycC or Cdk8 are essential for Serpent/Lozenge-induced transactivation in cell culture. Furthermore, our in vivo analysis of crystal cell development show that, while the four CDK8 module subunits control the emergence and the proliferation of this lineage, only Med12 and Med13 regulate its differentiation. We thus propose that Med12/Med13 acts as a coactivator for Serpent/Lozenge during crystal cell differentiation independently of CycC/Cdk8. More generally, we suggest that the set of conserved factors identified herein may regulate GATA/RUNX activity in mammals.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , GATA Transcription Factors/metabolism , Mediator Complex/metabolism , Protein Subunits/metabolism , RNA Interference , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Cyclin-Dependent Kinase 8/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , GATA Transcription Factors/genetics , Gene Expression Regulation, Developmental , Genome , Hematopoiesis/physiology , High-Throughput Screening Assays/methods , Mediator Complex/genetics , Protein Interaction Mapping , Protein Subunits/genetics , Transcription Factors/genetics
19.
Proc Natl Acad Sci U S A ; 106(29): 12043-8, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19581587

ABSTRACT

The t(8:21)(q22;q22) translocation is 1 of the most common chromosomal abnormalities linked to acute myeloid leukemia (AML). AML1-ETO, the product of this translocation, fuses the N-terminal portion of the RUNX transcription factor AML1 (also known as RUNX1), including its DNA-binding domain, to the almost entire transcriptional corepressor ETO (also known as MTG8 or RUNX1T1). This fusion protein acts primarily by interfering with endogenous AML1 function during myeloid differentiation, although relatively few genes are known that participate with AML1-ETO during leukemia progression. Here, we assessed the consequences of expressing this chimera in Drosophila blood cells. Reminiscent of what is observed in AML, AML1-ETO specifically inhibited the differentiation of the blood cell lineage whose development depends on the RUNX factor Lozenge (LZ) and induced increased numbers of LZ(+) progenitors. Using an in vivo RNAi-based screen for suppressors of AML1-ETO, we identified calpainB as required for AML1-ETO-induced blood cell disorders in Drosophila. Remarkably, calpain inhibition triggered AML1-ETO degradation and impaired the clonogenic potential of the human t(8;21) leukemic blood cell line Kasumi-1. Therefore Drosophila provides a promising genetically tractable model to investigate the conserved basis of leukemogenesis and to open avenues in AML therapy.


Subject(s)
Calpain/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Blood Cells/cytology , Calpain/antagonists & inhibitors , Cell Count , Cell Differentiation , Cell Line , Cell Lineage , Colony-Forming Units Assay , DNA-Binding Proteins/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Genes, Suppressor , Genetic Testing , Humans , Models, Animal , RUNX1 Translocation Partner 1 Protein , Stem Cells/cytology , Transcription Factors/metabolism
20.
Development ; 136(13): 2199-209, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19502482

ABSTRACT

The Ly6 superfamily, present in most metazoan genomes, codes for different cell-surface proteins and secreted ligands containing an extracellular motif called a Ly6 domain or three-finger domain. We report the identification of 36 novel genes coding for proteins of this family in Drosophila. One of these fly Ly6 proteins, coded by the gene boudin (bou), is essential for tracheal morphogenesis in the fly embryo and contributes to the maintenance of the paracellular barrier and the organisation of the septate junctions in this tissue. Bou, a glycosylphosphatidylinositol anchored membrane protein, is also required for septate junction organisation in epithelial tissues and in the chordotonal organ glial cells, but not in the central nervous system. Our study reveals interesting parallelisms between the Ly6 proteins of flies and vertebrates, such as the CD59 antigen. Similarly to this human protein, Bou travels from cell to cell associated with extracellular particles and, consistently, we show that it is required in a non-cell-autonomous fashion. Our work opens the way for future studies addressing the function of Ly6 proteins using Drosophila as a model system.


Subject(s)
Antigens, Ly/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Intercellular Junctions/metabolism , Membrane Proteins/metabolism , Trachea/embryology , Amino Acid Sequence , Animals , Antigens, Ly/genetics , Blood-Brain Barrier/embryology , Blood-Brain Barrier/ultrastructure , Cell Membrane/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Humans , Membrane Proteins/genetics , Molecular Sequence Data , Morphogenesis/physiology , Nerve Tissue/anatomy & histology , Nerve Tissue/embryology , Sequence Alignment , Trachea/anatomy & histology
SELECTION OF CITATIONS
SEARCH DETAIL