Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 21.
Article in English | MEDLINE | ID: mdl-38260379

ABSTRACT

Dihydroceramide desaturases convert dihydroceramides to ceramides, the precursors of all complex sphingolipids. Reduction of DEGS1 dihydroceramide desaturase function causes pediatric neurodegenerative disorder hypomyelinating leukodystrophy-18 (HLD-18). We discovered that infertile crescent (ifc), the Drosophila DEGS1 homolog, is expressed primarily in glial cells to promote CNS development by guarding against neurodegeneration. Loss of ifc causes massive dihydroceramide accumulation and severe morphological defects in cortex glia, including endoplasmic reticulum (ER) expansion, failure of neuronal ensheathment, and lipid droplet depletion. RNAi knockdown of the upstream ceramide synthase schlank in glia of ifc mutants rescues ER expansion, suggesting dihydroceramide accumulation in the ER drives this phenotype. RNAi knockdown of ifc in glia but not neurons drives neuronal cell death, suggesting that ifc function in glia promotes neuronal survival. Our work identifies glia as the primary site of disease progression in HLD-18 and may inform on juvenile forms of ALS, which also feature elevated dihydroceramide levels.

3.
G3 (Bethesda) ; 11(8)2021 08 07.
Article in English | MEDLINE | ID: mdl-33993253

ABSTRACT

The mechanisms that determine the final topology of skeletal muscles remain largely unknown. We have been developing Drosophila body wall musculature as a model to identify and characterize the pathways that control muscle size, shape, and orientation during embryogenesis. Our working model argues muscle morphogenesis is regulated by (1) extracellular guidance cues that direct muscle cells toward muscle attachment sites, and (2) contact-dependent interactions between muscles and tendon cells. While we have identified several pathways that regulate muscle morphogenesis, our understanding is far from complete. Here, we report the results of a recent EMS-based forward genetic screen that identified a myriad of loci not previously associated with muscle morphogenesis. We recovered new alleles of known muscle morphogenesis genes, including back seat driver, kon-tiki, thisbe, and tumbleweed, arguing our screen had the depth and precision to uncover myogenic genes. We also identified new alleles of spalt-major, barren, and patched that presumably disrupt independent muscle morphogenesis pathways. Equally as important, our screen shows that at least 11 morphogenetic loci remain to be mapped and characterized. Our screen has developed exciting new tools to study muscle morphogenesis, which may provide future insights into the mechanisms that regulate skeletal muscle topology.


Subject(s)
Drosophila Proteins , Drosophila , Muscle Development , Animals , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Morphogenesis/genetics , Muscle Development/genetics
4.
Elife ; 92020 03 27.
Article in English | MEDLINE | ID: mdl-32216875

ABSTRACT

The Drosophila ventral nerve cord (VNC) is composed of thousands of neurons born from a set of individually identifiable stem cells. The VNC harbors neuronal circuits required to execute key behaviors, such as flying and walking. Leveraging the lineage-based functional organization of the VNC, we investigated the developmental and molecular basis of behavior by focusing on lineage-specific functions of the homeodomain transcription factor, Unc-4. We found that Unc-4 functions in lineage 11A to promote cholinergic neurotransmitter identity and suppress the GABA fate. In lineage 7B, Unc-4 promotes proper neuronal projections to the leg neuropil and a specific flight-related take-off behavior. We also uncovered that Unc-4 acts peripherally to promote proprioceptive sensory organ development and the execution of specific leg-related behaviors. Through time-dependent conditional knock-out of Unc-4, we found that its function is required during development, but not in the adult, to regulate the above events.


Subject(s)
Brain/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Homeodomain Proteins/physiology , Neurons/physiology , Animals , Behavior, Animal , Cell Lineage , Drosophila Proteins/genetics , Flight, Animal , Homeodomain Proteins/genetics , Neurotransmitter Agents/analysis
5.
Pigment Cell Melanoma Res ; 33(3): 416-425, 2020 05.
Article in English | MEDLINE | ID: mdl-31642595

ABSTRACT

Tissue regeneration and homeostasis often require recruitment of undifferentiated precursors (adult stem cells; ASCs). While many ASCs continuously proliferate throughout the lifetime of an organism, others are recruited from a quiescent state to replenish their target tissue. A long-standing question in stem cell biology concerns how long-lived, non-dividing ASCs regulate the transition between quiescence and proliferation. We study the melanocyte stem cell (MSC) to investigate the molecular pathways that regulate ASC quiescence. Our prior work indicated that GABA-A receptor activation promotes MSC quiescence in larval zebrafish. Here, through pharmacological and genetic approaches we show that GABA-A acts through calcium signaling to maintain MSC quiescence. Unexpectedly, we identified translocator protein (TSPO), a mitochondrial membrane-associated protein that regulates mitochondrial function and metabolic homeostasis, as a parallel regulator of MSC quiescence. We found that both TSPO-specific ligands and induction of gluconeogenesis likely act in the same pathway to promote MSC activation and melanocyte production in larval zebrafish. In contrast, TSPO and gluconeogenesis appear to act in parallel to GABA-A receptor signaling to regulate MSC quiescence and vertebrate pigment patterning.


Subject(s)
Cell Cycle , Melanocytes/cytology , Mitochondria/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA/metabolism , Signal Transduction , Stem Cells/cytology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cell Cycle/drug effects , GABA-A Receptor Antagonists/pharmacology , Gluconeogenesis/drug effects , Larva/drug effects , Ligands , Melanocytes/drug effects , Mitochondria/drug effects , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism
6.
Genetics ; 213(2): 555-566, 2019 10.
Article in English | MEDLINE | ID: mdl-31444245

ABSTRACT

In larval zebrafish, melanocyte stem cells (MSCs) are quiescent, but can be recruited to regenerate the larval pigment pattern following melanocyte ablation. Through pharmacological experiments, we found that inhibition of γ-aminobutyric acid (GABA)-A receptor function, specifically the GABA-A ρ subtype, induces excessive melanocyte production in larval zebrafish. Conversely, pharmacological activation of GABA-A inhibited melanocyte regeneration. We used clustered regularly interspaced short palindromic repeats/Cas9 to generate two mutant alleles of gabrr1, a subtype of GABA-A receptors. Both alleles exhibited robust melanocyte overproduction, while conditional overexpression of gabrr1 inhibited larval melanocyte regeneration. Our data suggest that gabrr1 signaling is necessary to maintain MSC quiescence and sufficient to reduce, but not eliminate, melanocyte regeneration in larval zebrafish.


Subject(s)
Melanocytes/metabolism , Receptors, GABA-A/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Cell Differentiation/genetics , Cell Division/genetics , Larva/genetics , Larva/growth & development , Pigmentation/genetics , Regeneration/genetics , Signal Transduction/genetics , Stem Cells/metabolism , Zebrafish/growth & development , gamma-Aminobutyric Acid/genetics
7.
Development ; 144(17): 3102-3113, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28760813

ABSTRACT

The extracellular matrix (ECM) regulates cell migration and sculpts organ shape. AdamTS proteins are extracellular metalloproteases known to modify ECM proteins and promote cell migration, but demonstrated roles for AdamTS proteins in regulating CNS structure and ensuring cell lineages remain fixed in place have not been uncovered. Using forward genetic approaches in Drosophila, we find that reduction of AdamTS-A function induces both the mass exodus of neural lineages out of the CNS and drastic perturbations to CNS structure. Expressed and active in surface glia, AdamTS-A acts in parallel to perlecan and in opposition to viking/collagen IV and ßPS-integrin to keep CNS lineages rooted in place and to preserve the structural integrity of the CNS. viking/collagen IV and ßPS-integrin are known to promote tissue stiffness and oppose the function of perlecan, which reduces tissue stiffness. Our work supports a model in which AdamTS-A anchors cells in place and preserves CNS architecture by reducing tissue stiffness.


Subject(s)
Cell Lineage , Central Nervous System/cytology , Drosophila Proteins/metabolism , Extracellular Matrix/metabolism , Neurons/cytology , Neurons/metabolism , Alleles , Animals , Basement Membrane/metabolism , Collagen Type IV/metabolism , Drosophila melanogaster/metabolism , Green Fluorescent Proteins/metabolism , Integrin alpha Chains/metabolism , Mutation/genetics , Neuroglia/cytology , Neuroglia/metabolism , Phenotype , Subcellular Fractions/metabolism , Survival Analysis
9.
Nat Commun ; 8: 14112, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28106166

ABSTRACT

Hypomorphic mutations are a valuable tool for both genetic analysis of gene function and for synthetic biology applications. However, current methods to generate hypomorphic mutations are limited to a specific organism, change gene expression unpredictably, or depend on changes in spatial-temporal expression of the targeted gene. Here we present a simple and predictable method to generate hypomorphic mutations in model organisms by targeting translation elongation. Adding consecutive adenosine nucleotides, so-called polyA tracks, to the gene coding sequence of interest will decrease translation elongation efficiency, and in all tested cell cultures and model organisms, this decreases mRNA stability and protein expression. We show that protein expression is adjustable independent of promoter strength and can be further modulated by changing sequence features of the polyA tracks. These characteristics make this method highly predictable and tractable for generation of programmable allelic series with a range of expression levels.


Subject(s)
Genetic Techniques , Mutation , Proteins/genetics , Poly A/genetics , Poly A/metabolism , Promoter Regions, Genetic , Protein Biosynthesis , Proteins/metabolism , RNA Stability
10.
Genetics ; 203(2): 749-62, 2016 06.
Article in English | MEDLINE | ID: mdl-27029730

ABSTRACT

Dis3 encodes a conserved RNase that degrades or processes all RNA species via an N-terminal PilT N terminus (PIN) domain and C-terminal RNB domain that harbor, respectively, endonuclease activity and 3'-5' exonuclease activity. In Schizosaccharomyces pombe, dis3 mutations cause chromosome missegregation and failure in mitosis, suggesting dis3 promotes cell division. In humans, apparently hypomorphic dis3 mutations are found recurrently in multiple myeloma, suggesting dis3 opposes cell division. Except for the observation that RNAi-mediated depletion of dis3 function drives larval arrest and reduces tissue growth in Drosophila, the role of dis3 has not been rigorously explored in higher eukaryotic systems. Using the Drosophila system and newly generated dis3 null alleles, we find that absence of dis3 activity inhibits cell division. We uncover a conserved CDK1 phosphorylation site that when phosphorylated inhibits Dis3's exonuclease, but not endonuclease, activity. Leveraging this information, we show that Dis3's exonuclease function is required for mitotic cell division: in its absence, cells are delayed in mitosis and exhibit aneuploidy and overcondensed chromosomes. In contrast, we find that modest reduction of dis3 function enhances cell proliferation in the presence of elevated Ras activity, apparently by accelerating cells through G2/M even though each insult by itself delays G2/M. Additionally, we find that dis3 and ras genetically interact in worms and that dis3 can enhance cell proliferation under growth stimulatory conditions in murine B cells. Thus, reduction, but not absence, of dis3 activity can enhance cell proliferation in higher organisms.


Subject(s)
Cell Cycle/genetics , Evolution, Molecular , Exosome Multienzyme Ribonuclease Complex/genetics , ras Proteins/genetics , Animals , Caenorhabditis elegans/genetics , Cells, Cultured , Drosophila/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , Mice , Mice, Inbred C57BL , Schizosaccharomyces/genetics , ras Proteins/metabolism
11.
J Nucl Med ; 57(4): 640-5, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26742713

ABSTRACT

UNLABELLED: Very-late-antigen-4 (VLA-4, α4ß1 integrin, CD49d/CD29) is a transmembrane adhesion receptor that plays an important role in cancer and immune responses. Enhanced VLA-4 expression has been observed in multiple myeloma (MM) cells and surrounding stroma. VLA-4 conformational activation has been associated with MM pathogenesis. VLA-4 is a promising MM imaging and therapeutic biomarker. METHODS: Specificity of (64)Cu-LLP2A ((64)Cu-CB-TE1A1P-PEG4-LLP2A), a high-affinity VLA-4 peptidomimetic-based radiopharmaceutical, was evaluated in α4 knock-out mice and by competitive blocking in wild-type tumor-bearing mice. (64)Cu-LLP2A PET/CT (static and dynamic) imaging was conducted in C57BL6/KaLwRij mice bearing murine 5TGM1-GFP syngeneic tumors generated after intravenous injection via the tail. Blood samples were collected for serum protein electrophoresis. Bone marrow and splenic cells extracted from tumor-bearing and control mice (n= 3/group) were coincubated with the optical analog LLP2A-Cy5 and mouse B220, CD4, Gr1, and Mac1 antibodies and analyzed by fluorescence-activated cell sorting. Human radiation dose estimates for (64)Cu-LLP2A were extrapolated from mouse biodistribution data (6 time points, 0.78 MBq/animal, n= 4/group). Ten formalin-fixed paraffin-embedded bone marrow samples from deceased MM patients were stained with LLP2A-Cy5. RESULTS: (64)Cu-LLP2A and LLP2A-Cy5 demonstrated high specificity for VLA-4-positive mouse 5TGM1-GFP myeloma and nonmalignant inflammatory host cells such as T cells and myeloid/monocytic cells. Ex vivo flow cytometric analysis supported a direct effect of myeloma on increased VLA-4 expression in host hematopoietic microenvironmental elements. SUVs and the number of medullar lesions detected by (64)Cu-LLP2A PET corresponded with increased monoclonal (M) protein (g/dL) in tumor-bearing mice over time (3.29 ± 0.58 at week 0 and 9.97 ± 1.52 at week 3). Dynamic PET with (64)Cu-LLP2A and (18)F-FDG demonstrated comparable SUV in the prominent lesions in the femur. Human radiation dose estimates indicated urinary bladder wall as the dose-limiting organ (0.200 mGy/MBq), whereas the dose to the red marrow was 0.006 mGy/MBq. The effective dose was estimated to be 0.017 mSv/MBq. Seven of the ten human samples displayed a high proportion of cells intensely labeled with LLP2A-Cy5 probe. CONCLUSION: (64)Cu-LLP2A and LLP2A-Cy5 demonstrated binding specificity for VLA-4 in an immune-competent murine MM model. (64)Cu-LLP2A displayed favorable dosimetry for human studies and is a potential imaging candidate for overexpressed VLA-4.


Subject(s)
Dipeptides/pharmacokinetics , Integrin alpha4beta1/metabolism , Multiple Myeloma/diagnostic imaging , Multiple Myeloma/metabolism , Phenylurea Compounds/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Animals , Biomarkers, Tumor , Cell Line, Tumor , Copper Radioisotopes , Dipeptides/chemical synthesis , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenylurea Compounds/chemical synthesis , Positron-Emission Tomography , Protein Conformation , Radiopharmaceuticals/chemical synthesis , Tissue Distribution , Whole Body Imaging
12.
Exp Hematol ; 44(3): 161-5.e4, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26607597

ABSTRACT

The retinoblastoma gene (RB1) has been implicated as a tumor suppressor in multiple myeloma (MM), yet its role remains unclear because in the majority of cases with 13q14 deletions, un-mutated RB1 remains expressed from the retained allele. To explore the role of Rb1 in MM, we examined the functional consequences of single- and double-copy Rb1 loss in germinal center B cells, the cells of origin of MM. We generated mice without Rb1 function in germinal center B cells by crossing Rb1(Flox/Flox) with C-γ-1-Cre (Cγ1) mice expressing the Cre recombinase in class-switched B cells in a p107(-/-) background to prevent p107 from compensating for Rb1 loss (Cγ1-Rb1(F/F)-p107(-/-)). All mice developed normally, but B cells with two copies of Rb1 deleted (Cγ1-Rb1(F/F)-p107(-/-)) exhibited increased proliferation and cell death compared with Cγ1-Rb1(+/+)-p107(-/-) controls ex vivo. In vivo, Cγ1-Rb1(F/F)-p107(-/-) mice had a lower percentage of splenic B220+ cells and reduced numbers of bone marrow antigen-specific secreting cells compared with control mice. Our data indicate that Rb1 loss induces both cell proliferation and death in germinal center B cells. Because no B-cell malignancies developed after 1 year of observation, our data also suggest that Rb1 loss is not sufficient to transform post-germinal center B cells and that additional, specific mutations are likely required to cooperate with Rb1 loss to induce malignant transformation.


Subject(s)
B-Lymphocytes/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Germinal Center/metabolism , Mutation , Retinoblastoma Protein/deficiency , Animals , B-Lymphocytes/pathology , Cell Death , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Germinal Center/pathology , Mice , Mice, Knockout
13.
Dev Cell ; 31(5): 614-28, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25490268

ABSTRACT

Signaling pathways and small RNAs direct diverse cellular events, but few examples are known of defined signaling pathways directly regulating small RNA biogenesis. We show that ERK phosphorylates Dicer on two conserved residues in its RNase IIIb and double-stranded RNA (dsRNA)-binding domains and that phosphorylation of these residues is necessary and sufficient to trigger Dicer's nuclear translocation in worms, mice, and human cells. Phosphorylation of Dicer on either site inhibits Dicer function in the female germline and dampens small RNA repertoire. Our data demonstrate that ERK phosphorylates and inhibits Dicer during meiosis I for oogenesis to proceed normally in Caenorhabditis elegans and that this inhibition is released before fertilization for embryogenesis to proceed normally. The conserved Dicer residues, their phosphorylation by ERK, and the consequences of the resulting modifications implicate an ERK-Dicer nexus as a fundamental component of the oocyte-to-embryo transition and an underlying mechanism coupling extracellular cues to small RNA production.


Subject(s)
Caenorhabditis elegans/enzymology , MAP Kinase Signaling System/physiology , Oocytes/metabolism , RNA, Double-Stranded/metabolism , Ribonuclease III/metabolism , Animals , Base Sequence/physiology , Caenorhabditis elegans/embryology , Mice , Oogenesis/physiology , Phosphorylation
14.
Mol Biol Cell ; 25(19): 2956-69, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25079692

ABSTRACT

Once adherens junctions (AJs) are formed between polarized epithelial cells they must be maintained because AJs are constantly remodeled in dynamic epithelia. AJ maintenance involves endocytosis and subsequent recycling of E-cadherin to a precise location along the basolateral membrane. In the Drosophila pupal eye epithelium, Rho1 GTPase regulates AJ remodeling through Drosophila E-cadherin (DE-cadherin) endocytosis by limiting Cdc42/Par6/aPKC complex activity. We demonstrate that Rho1 also influences AJ remodeling by regulating the formation of DE-cadherin-containing, Rab11-positive recycling endosomes in Drosophila postmitotic pupal eye epithelia. This effect of Rho1 is mediated through Rok-dependent, but not MLCK-dependent, stimulation of myosin II activity yet independent of its effects upon actin remodeling. Both Rho1 and pMLC localize on endosomal vesicles, suggesting that Rho1 might regulate the formation of recycling endosomes through localized myosin II activation. This work identifies spatially distinct functions for Rho1 in the regulation of DE-cadherin-containing vesicular trafficking during AJ remodeling in live epithelia.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Drosophila Proteins/metabolism , Eye/metabolism , rab GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Drosophila , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Endocytosis , Endosomes/metabolism , Epithelium/metabolism , GTP-Binding Proteins/metabolism , Myosin Type II/metabolism , Peptides , Protein Kinase C/metabolism , Protein Transport , Pupa/metabolism , rab GTP-Binding Proteins/biosynthesis , rho GTP-Binding Proteins/genetics , rho-Associated Kinases
15.
Dev Biol ; 388(1): 117-33, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24512689

ABSTRACT

Hb9 is a homeodomain-containing transcription factor that acts in combination with Nkx6, Lim3, and Tail-up (Islet) to guide the stereotyped differentiation, connectivity, and function of a subset of neurons in Drosophila. The role of Hb9 in directing neuronal differentiation is well documented, but the lineage of Hb9(+) neurons is only partly characterized, its regulation is poorly understood, and most of the downstream genes through which it acts remain at large. Here, we complete the lineage tracing of all embryonic Hb9(+) neurons (to eight neuronal lineages) and provide evidence that hb9, lim3, and tail-up are coordinately regulated by a common set of upstream factors. Through the parallel use of micro-array gene expression profiling and the Dam-ID method, we searched for Hb9-regulated genes, uncovering transcription factors as the most over-represented class of genes regulated by Hb9 (and Nkx6) in the CNS. By a nearly ten-to-one ratio, Hb9 represses rather than activates transcription factors, highlighting transcriptional repression of other transcription factors as a core mechanism by which Hb9 governs neuronal determination. From the small set of genes activated by Hb9, we characterized the expression and function of two - fd59a/foxd, which encodes a transcription factor, and Nitric oxide synthase. Under standard lab conditions, both genes are dispensable for Drosophila development, but Nos appears to inhibit hyper-active behavior and fd59a appears to act in octopaminergic neurons to control egg-laying behavior. Together our data clarify the mechanisms through which Hb9 governs neuronal specification and differentiation and provide an initial characterization of the expression and function of Nos and fd59a in the Drosophila CNS.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Alleles , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cell Differentiation , Cell Lineage , Central Nervous System/embryology , Enhancer Elements, Genetic , Forkhead Transcription Factors/metabolism , Genetic Association Studies , Genotype , In Situ Hybridization , Molecular Sequence Data , Mutagenesis , Neurons/metabolism , Nitric Oxide Synthase/metabolism , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Transcriptome
16.
Development ; 141(5): 1011-21, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24550109

ABSTRACT

Most neurons of the adult Drosophila ventral nerve cord arise from a burst of neurogenesis during the third larval instar stage. Most of this growth occurs in thoracic neuromeres, which contain 25 individually identifiable postembryonic neuronal lineages. Initially, each lineage consists of two hemilineages--'A' (Notch(On)) and 'B' (Notch(Off))--that exhibit distinct axonal trajectories or fates. No reliable method presently exists to identify these lineages or hemilineages unambiguously other than labor-intensive lineage-tracing methods. By combining mosaic analysis with a repressible cell marker (MARCM) analysis with gene expression studies, we constructed a gene expression map that enables the rapid, unambiguous identification of 23 of the 25 postembryonic lineages based on the expression of 15 transcription factors. Pilot genetic studies reveal that these transcription factors regulate the specification and differentiation of postembryonic neurons: for example, Nkx6 is necessary and sufficient to direct axonal pathway selection in lineage 3. The gene expression map thus provides a descriptive foundation for the genetic and molecular dissection of adult-specific neurogenesis and identifies many transcription factors that are likely to regulate the development and differentiation of discrete subsets of postembryonic neurons.


Subject(s)
Central Nervous System/metabolism , Drosophila Proteins/metabolism , Transcription Factors/metabolism , Animals , Central Nervous System/cytology , Drosophila , Drosophila Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , Transcription Factors/genetics
17.
J Neurosci ; 33(45): 17863-73, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24198375

ABSTRACT

The MAPKKK dual leucine zipper-containing kinase (DLK, Wallenda in Drosophila) is an evolutionarily conserved component of the axonal injury response pathway. After nerve injury, DLK promotes degeneration of distal axons and regeneration of proximal axons. This dual role in coordinating degeneration and regeneration suggests that DLK may be a sensor of axon injury, and so understanding how DLK is activated is important. Two mechanisms are known to activate DLK. First, increasing the levels of DLK via overexpression or loss of the PHR ubiquitin ligases that target DLK activate DLK signaling. Second, in Caenorhabditis elegans, a calcium-dependent mechanism, can activate DLK. Here we describe a new mechanism that activates DLK in Drosophila: loss of the spectraplakin short stop (shot). In a genetic screen for mutants with defective neuromuscular junction development, we identify a hypomorphic allele of shot that displays synaptic terminal overgrowth and a precocious regenerative response to nerve injury. We demonstrate that both phenotypes are the result of overactivation of the DLK signaling pathway. We further show that, unlike mutations in the PHR ligase Highwire, loss of function of shot activates DLK without a concomitant increase in the levels of DLK. As a spectraplakin, Shot binds to both actin and microtubules and promotes cytoskeletal stability. The DLK pathway is also activated by downregulation of the TCP1 chaperonin complex, whose normal function is to promote cytoskeletal stability. These findings support the model that DLK is activated by cytoskeletal instability, which is a shared feature of both spectraplakin mutants and injured axons.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , MAP Kinase Kinase Kinases/genetics , Microfilament Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Alleles , Animals , Cytoskeleton/genetics , Cytoskeleton/metabolism , Down-Regulation , Drosophila/metabolism , Drosophila Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/physiology , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Phenotype
18.
Dev Dyn ; 242(7): 874-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23389965

ABSTRACT

BACKGROUND: The scalloped (sd) and vestigial (vg) genes function together in Drosophila wing development. Little is known about sd protein (SD) expression during development, or whether sd and vg interact in other developing tissues. To begin to address these questions, we generated an anti-SD antibody. RESULTS: During embryogenesis, SD is expressed in both central and peripheral nervous systems, and the musculature. SD is also expressed in developing flight appendages. Despite SD expression herein, the peripheral nervous system, musculature, and dorsal limb primordia appeared generally normal in the absence of sd function. SD is also expressed in subsets of ventral nerve cord cells, including neuroblast 1-2 descendants and ventral unpaired median motor neurons (mVUMs). While sd function is not required to specify these neurons, it is necessary for the correct innervation of somatic muscles by the mVUMs. We also show that SD and vg protein (VG) are co-expressed in overlapping and distinctive subsets of cells in embryonic and larval tissues. CONCLUSIONS: We describe the full breadth of SD expression during Drosophila embryogenesis, and identify a requirement for sd function in a subset of motor neurons. This work provides the necessary foundation for functional studies regarding the roles of sd during Drosophila development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Transcription Factors/metabolism , Animals , Central Nervous System/embryology , Central Nervous System/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Imaginal Discs/embryology , Imaginal Discs/metabolism , Muscles/embryology , Muscles/metabolism , Peripheral Nervous System/embryology , Peripheral Nervous System/metabolism , Transcription Factors/genetics
19.
Curr Biol ; 21(18): 1515-24, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21885285

ABSTRACT

BACKGROUND: In Drosophila, the basic-helix-loop-helix protein DIMM coordinates the molecular and cellular properties of all major neuroendocrine cells, irrespective of the secretory peptides they produce. When expressed by nonneuroendocrine neurons, DIMM confers the major properties of the regulated secretory pathway and converts such cells away from fast neurotransmission and toward a neuroendocrine state. RESULTS: We first identified 134 transcripts upregulated by DIMM in embryos and then evaluated them systematically using diverse assays (including embryo in situ hybridization, in vivo chromatin immunoprecipitation, and cell-based transactivation assays). We conclude that of eleven strong candidates, six are strongly and directly controlled by DIMM in vivo. The six targets include several large dense-core vesicle (LDCV) proteins, but also proteins in non-LDCV compartments such as the RNA-associated protein Maelstrom. In addition, a functional in vivo assay, combining transgenic RNA interference with MS-based peptidomics, revealed that three DIMM targets are especially critical for its action. These include two well-established LDCV proteins, the amidation enzyme PHM and the ascorbate-regenerating electron transporter cytochrome b(561-1). The third key DIMM target, CAT-4 (CG13248), has not previously been associated with peptide neurosecretion-it encodes a putative cationic amino acid transporter, closely related to the Slimfast arginine transporter. Finally, we compared transcripts upregulated by DIMM with those normally enriched in DIMM neurons of the adult brain and found an intersection of 18 DIMM-regulated genes, which included all six direct DIMM targets. CONCLUSIONS: The results provide a rigorous molecular framework with which to describe the fundamental regulatory organization of diverse neuroendocrine cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Drosophila Proteins/physiology , Drosophila/cytology , Neuroendocrine Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian/metabolism , Gene Expression Regulation , In Situ Hybridization , Molecular Sequence Data , RNA, Messenger/metabolism
20.
Curr Biol ; 20(7): 657-62, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20303269

ABSTRACT

The mammalian Ajuba LIM proteins (Ajuba, LIMD1, and WTIP) are adaptor proteins that exhibit the potential to communicate cell adhesive events with nuclear responses to remodel epithelia. Determining their role in vivo, however, has been challenging due to overlapping tissue expression and functional redundancy. Thus, we turned to Drosophila, where a single gene, CG11063 or djub, exists. Drosophila lacking the djub gene or depleted of dJub by RNA interference identify djub as an essential gene for development and a novel regulator of epithelial organ size as a component of the conserved Hippo (Hpo) pathway, which has been implicated in both tissue size control and cancer development. djub-deficient tissues were small and had decreased cell numbers as a result of increased apoptosis and decreased proliferation, due to downregulation of DIAP1 and cyclin E. This phenocopies tissues deficient for Yorkie (Yki), the downstream target of the Hippo pathway. djub genetically interacts with the Hippo pathway, and epistasis suggests that djub lies downstream of hpo. In mammalian and Drosophila cells, Ajuba LIM proteins/dJub interact with LATS/Warts (Wts) and WW45/Sav to inhibit phosphorylation of YAP/Yki. This work describes a novel role for the Ajuba LIM proteins as negative regulators of the Hippo signaling pathway.


Subject(s)
Drosophila Proteins/physiology , Homeodomain Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Animals, Genetically Modified , Apoptosis , Cell Proliferation , Drosophila/genetics , Drosophila/growth & development , Drosophila/physiology , Drosophila Proteins/genetics , Epistasis, Genetic , Eye/cytology , Eye/growth & development , Genes, Insect , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mammals , Models, Biological , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Organ Size , Protein Kinases/genetics , Protein Kinases/physiology , Protein Serine-Threonine Kinases/genetics , RNA Interference , Signal Transduction , Species Specificity , Trans-Activators/genetics , Trans-Activators/physiology , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...