Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 502(3): 415-421, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29856996

ABSTRACT

Acute inflammation can cause serious tissue damage and disease in physiologically-challenged organisms. The precise mechanisms leading to these detrimental effects remain to be determined. In this study, we utilize a reproducible means to induce cellular immune activity in Drosophila larvae in response to mechanical stress. That is, forceps squeeze-administered stress induces lamellocytes, a defensive hemocyte type that normally appears in response to wasp infestation of larvae. The posterior signaling center (PSC) is a cellular microenvironment in the larval hematopoietic lymph gland that is vital for lamellocyte induction upon parasitoid attack. However, we found the PSC was not required for mechanical stress-induced lamellocyte production. In addition, we observed that mechanical injury caused a systemic expression of Unpaired3. This cytokine is both necessary and sufficient to activate the cellular immune response to the imposed stress. These findings provide new insights into the communication between injured tissues and immune system induction, using stress-challenged Drosophila larvae as a tractable model system.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/metabolism , Animals , Animals, Genetically Modified , Cellular Microenvironment , Drosophila Proteins/metabolism , Drosophila melanogaster/parasitology , Hemocytes/cytology , Hemocytes/immunology , Immunity, Cellular , Janus Kinases/metabolism , Larva/immunology , Larva/metabolism , Larva/parasitology , STAT Transcription Factors/metabolism , Signal Transduction , Stress, Mechanical , Transcription Factors/metabolism , Wasps/immunology , Wasps/pathogenicity
2.
Genesis ; 56(5): e23210, 2018 05.
Article in English | MEDLINE | ID: mdl-29663653

ABSTRACT

Hedgehog (Hh) pathway signaling is crucial for the maintenance of blood cell progenitors in the lymph gland hematopoietic organ present in Drosophila third instar larvae. Previous studies from our lab have likewise shown the importance of the mir-7 and bag of marbles (bam) genes in maintaining the progenitor state. Thus, we sought to investigate a possible interaction between the Hh pathway and mir-7/bam in the prohemocyte population within this hematopoietic tissue. Gain of function mir-7 was able to rescue a blood cell progenitor depletion phenotype caused by Patched (Ptc) inhibition of Hh pathway signaling in these cells. Similarly, expression of a dominant/negative version of Ptc was able to rescue the severe reduction of prohemocytes due to bam loss of function. Furthermore, we demonstrated that Suppressor of fused [Su(fu)], another known inhibitor of Hh signaling, likely serves as a translational repression target of the mir-7 miRNA. Our results suggest the mir-7/bam combination regulates the Hh signaling network through repression of Su(fu) to maintain hemocyte progenitors in the larval lymph gland.


Subject(s)
Drosophila Proteins/metabolism , MicroRNAs/metabolism , Animals , Blood Cells , Cell Differentiation/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/genetics , Hematopoietic Stem Cells/metabolism , Larva/metabolism , Lymph Nodes/embryology , Lymph Nodes/metabolism , MicroRNAs/genetics , Receptors, Immunologic/metabolism , Repressor Proteins/genetics , Signal Transduction
3.
Biochem Biophys Res Commun ; 486(4): 893-897, 2017 05 13.
Article in English | MEDLINE | ID: mdl-28342875

ABSTRACT

In vertebrates, interaction between the nervous system and immune system is important to protect a challenged host from stress inputs from external sources. In this study, we demonstrate that sensory neurons are involved in the cellular immune response elicited by wasp infestation of Drosophila larvae. Multidendritic class IV neurons sense contacts from external stimuli and induce avoidance behaviors for host defense. Our findings show that inactivation of these sensory neurons impairs the cellular response against wasp parasitization. We also demonstrate that the nociception genes encoding the mechanosensory receptors Painless and Piezo, both expressed in class IV neurons, are essential for the normal cellular immune response to parasite challenge.


Subject(s)
Drosophila Proteins/immunology , Drosophila/immunology , Drosophila/parasitology , Ion Channels/immunology , Nociceptors/physiology , Wasps/pathogenicity , Animals , Larva/immunology , Larva/parasitology , Neuroimmunomodulation/immunology
4.
G3 (Bethesda) ; 7(2): 437-448, 2017 02 09.
Article in English | MEDLINE | ID: mdl-27913635

ABSTRACT

A GFP expression screen has been conducted on >1000 Janelia FlyLight Project enhancer-Gal4 lines to identify transcriptional enhancers active in the larval hematopoietic system. A total of 190 enhancers associated with 87 distinct genes showed activity in cells of the third instar larval lymph gland and hemolymph. That is, gene enhancers were active in cells of the lymph gland posterior signaling center (PSC), medullary zone (MZ), and/or cortical zone (CZ), while certain of the transcriptional control regions were active in circulating hemocytes. Phenotypic analyses were undertaken on 81 of these hematopoietic-expressed genes, with nine genes characterized in detail as to gain- and loss-of-function phenotypes in larval hematopoietic tissues and blood cells. These studies demonstrated the functional requirement of the cut gene for proper PSC niche formation, the hairy, Btk29A, and E2F1 genes for blood cell progenitor production in the MZ domain, and the longitudinals lacking, dFOXO, kayak, cap-n-collar, and delilah genes for lamellocyte induction and/or differentiation in response to parasitic wasp challenge and infestation of larvae. Together, these findings contribute substantial information to our knowledge of genes expressed during the larval stage of Drosophila hematopoiesis and newly identify multiple genes required for this developmental process.


Subject(s)
Drosophila melanogaster/genetics , Enhancer Elements, Genetic , Hematopoiesis/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cell Differentiation/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/parasitology , E2F1 Transcription Factor/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Hemocytes/metabolism , Larva/genetics , Larva/parasitology , Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Wasps/pathogenicity
5.
Development ; 142(13): 2261-7, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26041767

ABSTRACT

Bag of Marbles (Bam) is known to function as a positive regulator of hematopoietic progenitor maintenance in the lymph gland blood cell-forming organ during Drosophila hematopoiesis. Here, we demonstrate a key function for Bam in cells of the lymph gland posterior signaling center (PSC), a cellular domain proven to function as a hematopoietic niche. Bam is expressed in PSC cells, and gene loss-of-function results in PSC overgrowth and disorganization, indicating that Bam plays a crucial role in controlling the proper development of the niche. It was previously shown that Insulin receptor (InR) pathway signaling is essential for proper PSC cell proliferation. We analyzed PSC cell number in lymph glands double-mutant for bam and InR pathway genes, and observed that bam genetically interacts with pathway members in the formation of a normal PSC. The elF4A protein is a translation factor downstream of InR pathway signaling, and functional knockdown of this crucial regulator rescued the bam PSC overgrowth phenotype, further supporting the cooperative function of Bam with InR pathway members. Additionally, we documented that the Retinoblastoma-family protein (Rbf), a proven regulator of cell proliferation, was present in cells of the PSC, with a bam function-dependent expression. By contrast, perturbation of Decapentaplegic or Wingless signaling failed to affect Rbf niche cell expression. Together, these findings indicate that InR pathway-Bam-Rbf functional interactions represent a newly identified means to regulate the correct size and organization of the PSC hematopoietic niche.


Subject(s)
Cell Size , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Hematopoietic Stem Cells/cytology , Retinoblastoma Protein/metabolism , Somatomedins/metabolism , Stem Cell Niche , Transcription Factors/metabolism , Animals , Cell Count , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Epistasis, Genetic , Eukaryotic Initiation Factor-4A/genetics , Genes, Insect , Hematopoietic Stem Cells/metabolism , Lymphoid Tissue/metabolism , Phenotype , Pluripotent Stem Cells/metabolism , Receptor, Insulin/metabolism , Signal Transduction
6.
Biochem Biophys Res Commun ; 453(3): 467-72, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25280996

ABSTRACT

The Drosophila hematopoietic system is utilized in this study to gain novel insights into the process of growth control of the hematopoietic progenitor niche in blood development. The niche microenvironment is an essential component controlling the balance between progenitor populations and differentiated, mature blood cells and has been shown to lead to hematopoietic malignancies in humans when misregulated. MicroRNAs are one class of regulators associated with blood malignancies; however, there remains a relative paucity of information about the role of miRNAs in the niche. Here we demonstrate that bantam miRNA is endogenously active in the Drosophila hematopoietic progenitor niche, the posterior signaling center (PSC), and functions in the primary hematopoietic organ, the lymph gland, as a positive regulator of growth. Loss of bantam leads to a significant reduction in the PSC and overall lymph gland size, as well as a loss of the progenitor population and correlative premature differentiation of mature hemocytes. Interestingly, in addition to being essential for proper lymph gland development, we have determined bantam to be a novel upstream component of the insulin signaling cascade in the PSC and have unveiled dMyc as one factor central to bantam activity. These important findings identify bantam as a new hematopoietic regulator, place it in an evolutionarily conserved signaling pathway, present one way in which it is regulated, and provide a mechanism through which it facilitates cellular proliferation in the hematopoietic niche.


Subject(s)
Drosophila/genetics , Erythrocytes/cytology , Hematopoietic Stem Cells/cytology , Homeostasis/physiology , MicroRNAs/physiology , Animals
7.
PLoS One ; 8(10): e79019, 2013.
Article in English | MEDLINE | ID: mdl-24205363

ABSTRACT

In Drosophila, circulating hemocytes are derived from the cephalic mesoderm during the embryonic wave of hematopoiesis. These cells are contributed to the larva and persist through metamorphosis into the adult. To analyze this population of hemocytes, we considered data from a previously published RNAi screen in the hematopoietic niche, which suggested several members of the SCF complex play a role in lymph gland development. eater-Gal4;UAS-GFP flies were crossed to UAS-RNAi lines to knockdown the function of all known SCF complex members in a plasmatocyte-specific fashion, in order to identify which members are novel regulators of plasmatocytes. This specific SCF complex contains five core members: Lin-19-like, SkpA, Skp2, Roc1a and complex activator Nedd8. The complex was identified by its very distinctive large cell phenotype. Furthermore, these large cells stained for anti-P1, a plasmatocyte-specific antibody. It was also noted that the DNA in these cells appeared to be over-replicated. Gamma-tubulin and DAPI staining suggest the cells are undergoing re-replication as they had multiple centrioles and excessive DNA content. Further experimentation determined enlarged cells were BrdU-positive indicating they have progressed through S-phase. To determine how these cells become enlarged and undergo re-replication, cell cycle proteins were analyzed by immunofluorescence. This analysis identified three proteins that had altered subcellular localization in these enlarged cells: Cyclin E, Geminin and Double-parked. Previous research has shown that Double-parked must be degraded to exit S-phase, otherwise the DNA will undergo re-replication. When Double-parked was titrated from the nucleus by an excess of its inhibitor, geminin, the enlarged cells and aberrant protein localization phenotypes were partially rescued. The data in this report suggests that the SCF(Skp2) complex is necessary to ubiquitinate Double-parked during plasmatocyte cell division, ensuring proper cell cycle progression and the generation of a normal population of this essential blood cell type.


Subject(s)
Calcium-Binding Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , DNA Replication/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Nuclear Proteins/genetics , Animals , Blood Cells/cytology , Blood Cells/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/physiology , Cell Cycle Proteins/genetics , Cell Size , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cullin Proteins/physiology , Cyclin E/genetics , Cyclin E/metabolism , DNA/metabolism , DNA-Binding Proteins/genetics , Drosophila/cytology , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/physiology , Geminin/genetics , Geminin/metabolism , Gene Expression Regulation , Models, Biological , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , RNA Interference
9.
PLoS One ; 7(7): e41604, 2012.
Article in English | MEDLINE | ID: mdl-22911822

ABSTRACT

Hematopoiesis occurs in two phases in Drosophila, with the first completed during embryogenesis and the second accomplished during larval development. The lymph gland serves as the venue for the final hematopoietic program, with this larval tissue well-studied as to its cellular organization and genetic regulation. While the medullary zone contains stem-like hematopoietic progenitors, the posterior signaling center (PSC) functions as a niche microenvironment essential for controlling the decision between progenitor maintenance versus cellular differentiation. In this report, we utilize a PSC-specific GAL4 driver and UAS-gene RNAi strains, to selectively knockdown individual gene functions in PSC cells. We assessed the effect of abrogating the function of 820 genes as to their requirement for niche cell production and differentiation. 100 genes were shown to be essential for normal niche development, with various loci placed into sub-groups based on the functions of their encoded protein products and known genetic interactions. For members of three of these groups, we characterized loss- and gain-of-function phenotypes. Gene function knockdown of members of the BAP chromatin-remodeling complex resulted in niche cells that do not express the hedgehog (hh) gene and fail to differentiate filopodia believed important for Hh signaling from the niche to progenitors. Abrogating gene function of various members of the insulin-like growth factor and TOR signaling pathways resulted in anomalous PSC cell production, leading to a defective niche organization. Further analysis of the Pten, TSC1, and TSC2 tumor suppressor genes demonstrated their loss-of-function condition resulted in severely altered blood cell homeostasis, including the abundant production of lamellocytes, specialized hemocytes involved in innate immune responses. Together, this cell-specific RNAi knockdown survey and mutant phenotype analyses identified multiple genes and their regulatory networks required for the normal organization and function of the hematopoietic progenitor niche within the lymph gland.


Subject(s)
Cell Differentiation/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Gene Regulatory Networks , Hematopoietic Stem Cells/cytology , Lymph Nodes/cytology , Stem Cell Niche/genetics , Animals , Biomarkers/metabolism , Blood Cells/metabolism , Cell Count , Chromatin Assembly and Disassembly , Diet , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Feeding Behavior , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genes, Insect/genetics , Hematopoietic Stem Cells/metabolism , Hemolymph/cytology , Hemolymph/metabolism , Homeostasis/genetics , Larva/anatomy & histology , Larva/cytology , Lymph Nodes/anatomy & histology , Lymph Nodes/metabolism , Mutation/genetics , Organ Size/genetics , Organ Specificity/genetics , RNA Interference , Signal Transduction/genetics
10.
PLoS One ; 7(1): e30565, 2012.
Article in English | MEDLINE | ID: mdl-22276214

ABSTRACT

The Drosophila dorsal vessel is a beneficial model system for studying the regulation of early heart development. Spire (Spir), an actin-nucleation factor, regulates actin dynamics in many developmental processes, such as cell shape determination, intracellular transport, and locomotion. Through protein expression pattern analysis, we demonstrate that the absence of spir function affects cell division in Myocyte enhancer factor 2-, Tinman (Tin)-, Even-skipped- and Seven up (Svp)-positive heart cells. In addition, genetic interaction analysis shows that spir functionally interacts with Dorsocross, tin, and pannier to properly specify the cardiac fate. Furthermore, through visualization of double heterozygous embryos, we determines that spir cooperates with CycA for heart cell specification and division. Finally, when comparing the spir mutant phenotype with that of a CycA mutant, the results suggest that most Svp-positive progenitors in spir mutant embryos cannot undergo full cell division at cell cycle 15, and that Tin-positive progenitors are arrested at cell cycle 16 as double-nucleated cells. We conclude that Spir plays a crucial role in controlling dorsal vessel formation and has a function in cell division during heart tube morphogenesis.


Subject(s)
Cell Division/physiology , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Heart/embryology , Microfilament Proteins/metabolism , Animals , Cell Division/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , MEF2 Transcription Factors , Microfilament Proteins/genetics , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Genesis ; 50(1): 41-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21809435

ABSTRACT

Eater is a transmembrane protein that mediates phagocytosis in Drosophila. eater was identified in a microarray analysis of genes downregulated in S2 cells, in which Serpent had been knocked down by RNAi. The gene was shown to be expressed predominantly in plasmatocytes after embryonic development. We have extensively analyzed the transcriptional enhancer controlling eater expression with the following findings: the enhancer reproduces the plasmatocyte expression pattern of the gene as verified by anti-P1 antibody staining and a 526-basepair DNA region is active in lymph gland and hemolymph plasmatocytes. This DNA contains several GATA elements that serve as putative-binding sites for Serpent. Site-directed mutagenesis of two of these GATA sites abolishes eater expression in both lymph gland and hemolymph plasmatocytes. This suggests that Serpent regulates eater expression by binding these GATA sites, which was confirmed by gel shift analysis. These analyses allowed us to use eater-Gal4 to force plasmatocyte to lamellocyte differentiation.


Subject(s)
Blood Cells/metabolism , Drosophila Proteins/genetics , Drosophila/genetics , Receptors, Cell Surface/genetics , Animals , Binding Sites/genetics , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/cytology , Drosophila/embryology , Drosophila Proteins/metabolism , Electrophoretic Mobility Shift Assay/methods , Enhancer Elements, Genetic , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Hematopoiesis , Hemocytes/cytology , Hemocytes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Multigene Family , Mutagenesis, Site-Directed , RNA Interference , Receptors, Cell Surface/metabolism , Transcription, Genetic
12.
Development ; 138(18): 3879-84, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21813570

ABSTRACT

Bag of Marbles (Bam) is a stem cell differentiation factor in the Drosophila germ line. Here, we demonstrate that Bam has a crucial function in the lymph gland, the tissue that orchestrates the second phase of Drosophila hematopoiesis. In bam mutant larvae, depletion of hematopoietic progenitors is observed, coupled with prodigious production of differentiated hemocytes. Conversely, forced expression of Bam in the lymph gland results in expansion of prohemocytes and substantial reduction of differentiated blood cells. These findings identify Bam as a regulatory protein that promotes blood cell precursor maintenance and prevents hemocyte differentiation during larval hematopoiesis. Cell-specific knockdown of bam function via RNAi expression revealed that Bam activity is required cell-autonomously in hematopoietic progenitors for their maintenance. microRNA-7 (mir-7) mutant lymph glands present with phenotypes identical to those seen in bam-null animals and mutants double-heterozygous for bam and mir-7 reveal that the two cooperate to maintain the hematopoietic progenitor population. By contrast, analysis of yan mutant lymph glands revealed that this transcriptional regulator promotes blood cell differentiation and the loss of prohemocyte maintenance. Expression of Bam or mir-7 in hematopoietic progenitors leads to a reduction of Yan protein. Together, these results demonstrate that Bam and mir-7 antagonize the differentiation-promoting function of Yan to maintain the stem-like hematopoietic progenitor state during hematopoiesis.


Subject(s)
Drosophila Proteins/physiology , Drosophila/genetics , Drosophila/physiology , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Proliferation , Drosophila/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Eye Proteins/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Germ Cells/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , MicroRNAs/physiology , Microarray Analysis , Repressor Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/physiology
13.
Development ; 137(21): 3561-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20876645

ABSTRACT

The lymph gland is a specialized organ for hematopoiesis, utilized during larval development in Drosophila. This tissue is composed of distinct cellular domains populated by blood cell progenitors (the medullary zone), niche cells that regulate the choice between progenitor quiescence and hemocyte differentiation [the posterior signaling center (PSC)], and mature blood cells of distinct lineages (the cortical zone). Cells of the PSC express the Hedgehog (Hh) signaling molecule, which instructs cells within the neighboring medullary zone to maintain a hematopoietic precursor state while preventing hemocyte differentiation. As a means to understand the regulatory mechanisms controlling Hh production, we characterized a PSC-active transcriptional enhancer that drives hh expression in supportive niche cells. Our findings indicate that a combination of positive and negative transcriptional inputs program the precise PSC expression of the instructive Hh signal. The GATA factor Serpent (Srp) is essential for hh activation in niche cells, whereas the Suppressor of Hairless [Su(H)] and U-shaped (Ush) transcriptional regulators prevent hh expression in blood cell progenitors and differentiated hemocytes. Furthermore, Srp function is required for the proper differentiation of niche cells. Phenotypic analyses also indicated that the normal activity of all three transcriptional regulators is essential for maintaining the progenitor population and preventing premature hemocyte differentiation. Together, these studies provide mechanistic insights into hh transcriptional regulation in hematopoietic progenitor niche cells, and demonstrate the requirement of the Srp, Su(H) and Ush proteins in the control of niche cell differentiation and blood cell precursor maintenance.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila , GATA Transcription Factors/physiology , Hedgehog Proteins/genetics , Hematopoiesis/genetics , Repressor Proteins/physiology , Stem Cell Niche/metabolism , Transcription Factors/physiology , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Drosophila/physiology , Drosophila Proteins/metabolism , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Hemocytes/metabolism , Hemocytes/physiology , Larva/genetics , Larva/metabolism , Larva/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Fly (Austin) ; 4(4): 338-43, 2010.
Article in English | MEDLINE | ID: mdl-20798602

ABSTRACT

The expression of toxic viral proteins for the purpose of eliminating distinct populations of cells, while leaving the rest of an organism unaffected, is a valuable method for analyzing development. Using the Gal4-UAS system, we employed the M2(H37A) toxic ion channel of the influenza-A virus to selectively ablate the Drosophila eye-antennal imaginal discs, hemocytes, dorsal vessel and nervous tissue, and comparatively monitored the effects of expressing the apoptosis-promoting protein Reaper in identical cell populations. In this report, we demonstrate the effectiveness of M2(H37A)-mediated ablation as a new means to selectively eliminate cells of interest during Drosophila development.


Subject(s)
Drosophila/genetics , Genetic Engineering/methods , Ion Channels/genetics , Viral Matrix Proteins/genetics , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Apoptosis , Drosophila/cytology , Drosophila/growth & development , Eye/cytology , Hemocytes/cytology , Hemocytes/metabolism , Larva/cytology , Larva/genetics , Larva/growth & development , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Orthomyxoviridae/genetics , Phenotype
15.
PLoS One ; 5(5): e10713, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20520827

ABSTRACT

The zinc finger protein CF2 is a characterized activator of muscle structural genes in the body wall muscles of the Drosophila larva. To investigate the function of CF2 in the indirect flight muscle (IFM), we examined the phenotypes of flies bearing five homozygous viable mutations. The gross structure of the IFM was not affected, but the stronger hypomorphic alleles caused an increase of up to 1.5X in the diameter of the myofibrils. This size increase did not cause any disruption of the hexameric arrangement of thick and thin filaments. RT-PCR analysis revealed an increase in the transcription of several structural genes. Ectopic overexpression of CF2 in the developing IFM disrupts muscle formation. While our results indicate a role for CF2 as a direct negative regulator of the thin filament protein gene Actin 88F (Act88F), effects on levels of transcripts of myosin heavy chain (mhc) appear to be indirect. This role is in direct contrast to that described in the larval muscles, where CF2 activates structural gene expression. The variation in myofibril phenotypes of CF2 mutants suggest the CF2 may have separate functions in fine-tuning expression of structural genes to insure proper filament stoichiometry, and monitoring and/or controlling the final myofibril size.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Flight, Animal , Gene Expression Regulation, Developmental , Muscle Development/genetics , Transcription Factors/metabolism , Actin Cytoskeleton/ultrastructure , Actins/metabolism , Animals , Cell Nucleus , Drosophila melanogaster/ultrastructure , Enhancer Elements, Genetic/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscles/metabolism , Muscles/pathology , Muscles/ultrastructure , Mutation , Myofibrils/metabolism , Myofibrils/ultrastructure , Myosin Heavy Chains/genetics , Phenotype , Protein Isoforms/metabolism , Protein Transport , Pupa/growth & development , Pupa/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic
16.
Genesis ; 47(11): 771-4, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19830816

ABSTRACT

Based on environmental challenges or altered genetic composition, Drosophila larvae can produce up to three types of blood cells that express genetic programs essential for their distinct functions. Using transcriptional enhancers for genes expressed exclusively in plasmatocytes, crystal cells, or lamellocytes, several new hemocyte-specific enhancer-reporter transgenes were generated to facilitate the analysis of Drosophila hematopoiesis. This approach took advantage of fluorescent variants of insulated P-element reporter vectors for multilabeling cell analyses; two additional color variants were generated in these studies. These vectors were successfully used to produce transgenic fly lines that label specific hemocyte lineages with separate colors. Combining three transgene reporters allowed for the unambiguous identification of plasmatocytes, crystal cells, and lamellocytes within a complex hemocyte population. While this work focused on the hematopoietic process, these new vectors can be used to mark multiple cell types or trace complex cell lineages during any chosen aspect of Drosophila development.


Subject(s)
Drosophila/genetics , Enhancer Elements, Genetic , Genes, Reporter , Hematopoiesis/genetics , Hemocytes/metabolism , Transgenes , Animals
17.
PLoS One ; 4(7): e6429, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19641625

ABSTRACT

Drosophila has emerged as an excellent model system in which to study cellular and genetic aspects of hematopoiesis. Under normal developmental conditions and in wild-type genetic backgrounds, Drosophila possesses two types of blood cells, crystal cells and plasmatocytes. Upon infestation by a parasitic wasp or in certain altered genetic backgrounds, a third hemocyte class called the lamellocyte becomes apparent. Herein we describe the characterization of a novel transcriptional regulatory module, a lamellocyte-active enhancer of the misshapen gene. This transcriptional control sequence appears to be inactive in all cell types of the wild-type larva, including crystal cells and plasmatocytes. However, in lamellocytes induced by wasp infestation or by particular genetic conditions, the enhancer is activated and it directs reporter GFP or DsRed expression exclusively in lamellocytes. The lamellocyte control region was delimited to a 140-bp intronic sequence that contains an essential DNA recognition element for the AP-1 transcription factor. Additionally, mutation of the kayak gene encoding the dFos subunit of AP-1 led to a strong suppression of lamellocyte production in tumorous larvae. As misshapen encodes a protein kinase within the Jun N-terminal kinase signaling pathway that functions to form an active AP-1 complex, the lamellocyte-active enhancer likely serves as a transcriptional target within a genetic auto-regulatory circuit that promotes the production of lamellocytes in immune-challenged or genetically-compromised animals.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Enhancer Elements, Genetic , Protein Serine-Threonine Kinases/genetics , Transcription, Genetic , Animals , Base Sequence , DNA/genetics , Electrophoretic Mobility Shift Assay , Introns , Molecular Sequence Data , Wasps
18.
Dev Biol ; 311(2): 311-23, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17936744

ABSTRACT

Drosophila has emerged as an important model system to discover and analyze genes controlling hematopoiesis. One regulatory network known to control hemocyte differentiation is the Janus kinase (JAK)/Signal Transducer and Activator of Transcription (STAT) signal-transduction pathway. A constitutive activation mutation of the Janus kinase Hopscotch (hopscotch(Tumorous-lethal); hop(Tum-l)) results in a leukemia-like over-proliferation of hemocytes and copious differentiation of lamellocytes during larval stages. Here we show that the Friend of GATA (FOG) protein U-shaped (Ush) is expressed in circulating and lymph gland hemocytes, where it plays a critical role in controlling blood cell proliferation and differentiation. Our findings demonstrate that a reduction in ush function results in hematopoietic phenotypes strikingly similar to those observed in hop(Tum-l) animals. These include lymph gland hypertrophy, increased circulating hemocyte concentration, and abundant production of lamellocytes. Forced expression of N-terminal truncated versions of Ush likewise leads to larvae with severe hematopoietic anomalies. In contrast, expression of wild-type Ush results in a strong suppression of hop(Tum-l) phenotypes. Taken together, our findings demonstrate that U-shaped acts to control larval hemocyte proliferation and suppress lamellocyte differentiation, likely regulating hematopoietic events downstream of Hop kinase activity. Such functions appear to be facilitated through Ush interaction with the hematopoietic GATA factor Serpent (Srp).


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Genes, Tumor Suppressor , Hematopoietic System/physiology , Hemocytes/physiology , Janus Kinases/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/physiology , Cell Proliferation , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Hemocytes/cytology , Janus Kinases/genetics , Larva/cytology , Larva/physiology , Phenotype , Transcription Factors/genetics , Transgenes
19.
Genesis ; 45(10): 607-17, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17941041

ABSTRACT

Heart development is an evolutionarily conserved process. The cardiac organ of Drosophila melanogaster is the dorsal vessel, a linear contractile tissue with cellular and morphogenetic similarities to the primitive heart tube formed at an early stage of vertebrate heart formation. Abundant evidence shows comparable intercellular signaling pathways and transcription factor networks are utilized in Drosophila and vertebrates, to specify cardiac progenitor cells and instruct their differentiation and function in forming the mature heart. With this proven conservation in mind, we screened the second chromosome of Drosophila for genetic intervals that harbor additional loci required for normal dorsal vessel morphogenesis. Our studies identified numerous regions, that when deleted, culminated in dorsal vessels with abnormal cell numbers and/or structural properties. Certain of the deficiency intervals were further characterized to identify individual genes essential for proper cardiac organ formation. Our analyses identified eight genes of diverse functions that are needed for dorsal vessel development. Several of these sequences have known vertebrate homologues, further supporting a conserved genetic basis for heart formation in Drosophila and higher eukaryotes.


Subject(s)
Chromosomes/genetics , Drosophila/embryology , Drosophila/genetics , Heart/embryology , Morphogenesis/genetics , Animals , Biomarkers , Blood Vessels/embryology , Chromosome Mapping , Embryo, Nonmammalian , Genes, Insect/physiology , Genetic Testing , Green Fluorescent Proteins/metabolism , Models, Biological , Mutation
20.
Genesis ; 45(4): 200-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17417793

ABSTRACT

In Drosophila, Black cells (Bc) encodes a Prophenoloxidase and is expressed late in the maturation of crystal cells, which are blood cells involved in wound healing and immune encapsulation. Enhancer analysis of Bc revealed a 1,025-bp upstream sequence that regulates gene expression in a crystal cell exclusive pattern. Expression of this fragment is altered by mutations in the GATA family serpent (srp) and RUNX family lozenge (lz) genes; Srp and Lz are required for crystal cell specification. Deletional analysis uncovered a 330-bp crystal cell-specific sequence, which contains two GATA and three Lz binding sites. Mutational analysis revealed that both GATA sites are necessary, but not sufficient for crystal cell expression. However, one of the Lz sites is essential for crystal cell expression. Thus, Srp and Lz do not just specify the crystal cell lineage, but also regulate the later differentiation of these cells. Additionally, we now have a sensitive tool for marking crystal cells in live animals.


Subject(s)
Catechol Oxidase/genetics , Drosophila/embryology , Drosophila/genetics , Enhancer Elements, Genetic , Enzyme Precursors/genetics , Hemocytes , Animals , Animals, Genetically Modified , Binding Sites , Catechol Oxidase/metabolism , Cell Differentiation/genetics , DNA-Binding Proteins/physiology , Drosophila/cytology , Drosophila/metabolism , Drosophila Proteins/physiology , Embryo, Nonmammalian , Enhancer Elements, Genetic/physiology , Enzyme Precursors/metabolism , Female , GATA Transcription Factors/physiology , Gene Expression Regulation, Developmental , Hematopoietic System/embryology , Hematopoietic System/metabolism , Hemocytes/metabolism , Male , Organ Specificity , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...