Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters











Publication year range
1.
Elife ; 132024 Aug 30.
Article in English | MEDLINE | ID: mdl-39213032

ABSTRACT

Environmental insults, including mild head trauma, significantly increase the risk of neurodegeneration. However, it remains challenging to establish a causative connection between early-life exposure to mild head trauma and late-life emergence of neurodegenerative deficits, nor do we know how sex and age compound the outcome. Using a Drosophila model, we demonstrate that exposure to mild head trauma causes neurodegenerative conditions that emerge late in life and disproportionately affect females. Increasing age-at-injury further exacerbates this effect in a sexually dimorphic manner. We further identify sex peptide signaling as a key factor in female susceptibility to post-injury brain deficits. RNA sequencing highlights a reduction in innate immune defense transcripts specifically in mated females during late life. Our findings establish a causal relationship between early head trauma and late-life neurodegeneration, emphasizing sex differences in injury response and the impact of age-at-injury. Finally, our findings reveal that reproductive signaling adversely impacts female response to mild head insults and elevates vulnerability to late-life neurodegeneration.


Subject(s)
Drosophila melanogaster , Animals , Female , Male , Drosophila melanogaster/genetics , Signal Transduction , Neurodegenerative Diseases/genetics , Craniocerebral Trauma , Reproduction , Aging , Disease Models, Animal , Drosophila , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Sex Characteristics , Age Factors , Sex Factors
2.
Mol Biol Cell ; 35(8): ar109, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38985523

ABSTRACT

The Drosophila RNA-binding protein (RBP) Nab2 acts in neurons to regulate neurodevelopment and is orthologous to the human intellectual disability-linked RBP, ZC3H14. Nab2 governs axon projection in mushroom body neurons and limits dendritic arborization of class IV sensory neurons in part by regulating splicing events in ∼150 mRNAs. Analysis of the Sex-lethal (Sxl) mRNA revealed that Nab2 promotes an exon-skipping event and regulates m6A methylation on Sxl pre-mRNA by the Mettl3 methyltransferase. Mettl3 heterozygosity broadly rescues Nab2null phenotypes implying that Nab2 acts through similar mechanisms on other RNAs, including unidentified targets involved in neurodevelopment. Here, we show that Nab2 and Mettl3 regulate the removal of a 5'UTR (untranslated region) intron in the trio pre-mRNA. Trio utilizes two GEF domains to balance Rac and RhoGTPase activity. Intriguingly, an isoform of Trio containing only the RhoGEF domain, GEF2, is depleted in Nab2null nervous tissue. Expression of Trio-GEF2 rescues projection defects in Nab2null axons and dendrites, while the GEF1 Rac1-regulatory domain exacerbates these defects, suggesting Nab2-mediated regulation Trio-GEF activities. Collectively, these data indicate that Nab2-regulated processing of trio is critical for balancing Trio-GEF1 and -GEF2 activity and show that Nab2, Mettl3, and Trio function in a common pathway that shapes axon and dendrite morphology.


Subject(s)
Axons , Dendrites , Drosophila Proteins , Drosophila melanogaster , Guanine Nucleotide Exchange Factors , RNA-Binding Proteins , Animals , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Axons/metabolism , Dendrites/metabolism , Drosophila melanogaster/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , RNA Splicing , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA Precursors/metabolism , RNA Precursors/genetics
3.
Development ; 151(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38775023

ABSTRACT

Regenerative ability often declines as animals mature past embryonic and juvenile stages, suggesting that regeneration requires redirection of growth pathways that promote developmental growth. Intriguingly, the Drosophila larval epithelia require the hormone ecdysone (Ec) for growth but require a drop in circulating Ec levels to regenerate. Examining Ec dynamics more closely, we find that transcriptional activity of the Ec-receptor (EcR) drops in uninjured regions of wing discs, but simultaneously rises in cells around the injury-induced blastema. In parallel, blastema depletion of genes encoding Ec biosynthesis enzymes blocks EcR activity and impairs regeneration but has no effect on uninjured wings. We find that local Ec/EcR signaling is required for injury-induced pupariation delay following injury and that key regeneration regulators upd3 and Ets21c respond to Ec levels. Collectively, these data indicate that injury induces a local source of Ec within the wing blastema that sustains a transcriptional signature necessary for developmental delay and tissue repair.


Subject(s)
Drosophila Proteins , Ecdysone , Regeneration , Wings, Animal , Animals , Ecdysone/metabolism , Wings, Animal/metabolism , Wings, Animal/growth & development , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Epithelium/metabolism , Gene Expression Regulation, Developmental , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Larva/metabolism , Larva/growth & development , Signal Transduction , Drosophila , Receptors, Steroid/metabolism , Receptors, Steroid/genetics
4.
bioRxiv ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38617327

ABSTRACT

Rapidly dividing cells can eliminate slow growing neighbors through the apoptotic process of cell competition. This process ensures that only high fitness cells populate embryonic tissues and is proposed to underlie the ability of oncogene-transformed cells to progressively replace normal cells within a tissue. Patches of cells in the Drosophila wing disc overexpressing the oncogenic Taiman (Tai) transcriptional coactivator kill normal neighbors by secreting Spätzle ligands that trigger pro-apoptotic Toll signaling in receiving cells. However, extracellular signaling mechanisms responsible for elimination of slow growing cells by normal neighbors remain poorly defined. Here we show that slow growing cells with reduced Tai (Tailow) are killed by normal neighbors through a mechanism involving competition for the Wingless (Wg/Wnt) ligand. Elevated Wg signaling significantly rescues elimination of Tailow cells in multiple organs, suggesting that Tai may normally promote Wg activity. Examining distribution of Wg components reveals that Tai promotes extracellular spread of the Wg ligand from source cells across the wing disc, thus ensuring patterned expression of multiple Wg-regulated target genes. Tai controls Wg spread indirectly through the extracellular glypican Dally-like protein (Dlp), which binds Wg and promotes its extracellular diffusion and capture by receptors. Data indicate that Tai likely controls Dlp at two levels: transcription of dlp mRNA and Dlp intracellular trafficking. Overall, these data indicate that the Tai acts through Dlp to enable Wg transport and signaling, and that cell competition in the Tailow model arises due to inequity in the ability of epithelial cells to sequester limiting amounts of the Wg growth factor.

5.
bioRxiv ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38496515

ABSTRACT

Environmental insults, including mild head trauma, significantly increase the risk of neurodegeneration. However, it remains challenging to establish a causative connection between early-life exposure to mild head trauma and late-life emergence of neurodegenerative deficits, nor do we know how sex and age compound the outcome. Using a Drosophila model, we demonstrate that exposure to mild head trauma causes neurodegenerative conditions that emerge late in life and disproportionately affect females. Increasing age-at-injury further exacerbates this effect in a sexually dimorphic manner. We further identify Sex Peptide (SP) signaling as a key factor in female susceptibility to post-injury brain deficits. RNA sequencing highlights a reduction in innate immune defense transcripts specifically in mated females during late life. Our findings establish a causal relationship between early head trauma and late-life neurodegeneration, emphasizing sex differences in injury response and the impact of age-at-injury. Finally, our findings reveal that reproductive signaling adversely impacts female response to mild head insults and elevates vulnerability to late-life neurodegeneration.

6.
bioRxiv ; 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38464192

ABSTRACT

Regenerative ability often declines as animals mature past embryonic and juvenile stages, suggesting that regeneration requires redirection of growth pathways that promote developmental growth. Intriguingly, the Drosophila larval epithelia require the hormone ecdysone (Ec) for growth but require a drop in circulating Ec levels to regenerate. Examining Ec dynamics more closely, we find that transcriptional activity of the Ec-receptor (EcR) drops in uninjured regions of wing discs, but simultaneously rises in cells around the injury-induced blastema. In parallel, blastema depletion of genes encoding Ec biosynthesis enzymes blocks EcR activity and impairs regeneration but has no effect on uninjured wings. We find that local Ec/EcR signaling is required for injury-induced pupariation delay following injury and that key regeneration regulators upd3 and Ets21c respond to Ec levels. Collectively, these data indicate that injury induces a local source of Ec within the wing blastema that sustains a transcriptional signature necessary for developmental delay and tissue repair.

7.
Curr Protoc ; 3(10): e924, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37861353

ABSTRACT

The rapid succession of events during development poses an inherent challenge to achieve precise synchronization required for rigorous, quantitative phenotypic and genotypic analyses in multicellular model organisms. Drosophila melanogaster is an indispensable model for studying the development and function of higher order organisms due to extensive genome homology, tractability, and its relatively short lifespan. Presently, nine Nobel prizes serve as a testament to the utility of this elegant model system. Ongoing advancements in genetic and molecular tools allow for the underlying mechanisms of human disease to be investigated in Drosophila. However, the absence of a method to precisely age-match tissues during larval development prevents further capitalization of this powerful model organism. Drosophila spends nearly half of its life cycle progressing through three morphologically distinct larval instar stages, during which the imaginal discs, precursors of mature adult external structures (e.g., eyes, legs, wings), grow and develop distinct cell fates. Other tissues, such as the central nervous system, undergo massive morphological changes during larval development. While these three larval stages and subsequent pupal stages have historically been identified based on the number of hours post egg-laying under standard laboratory conditions, a reproducible, efficient, and inexpensive method is required to accurately age-match larvae within the third instar. The third instar stage is of particular interest, as this developmental stage spans a 48-hr window during which larval tissues switch from proliferative to differentiation programs. Moreover, some genetic manipulations can lead to developmental delays, further compounding the need for precise age-matching between control and experimental samples. This article provides a protocol optimized for synchronous staging of Drosophila third instar larvae by colorimetric characterization and is useful for age-matching a variety of tissues for numerous downstream applications. We also provide a brief discussion of the technical challenges associated with successful application of this protocol. © 2023 Wiley Periodicals LLC. Basic Protocol: Synchronization of third instar Drosophila larvae.


Subject(s)
Drosophila melanogaster , Drosophila , Animals , Humans , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/genetics , Larva/physiology , Colorimetry , Pupa
8.
Dev Cell ; 58(5): 416-434.e12, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36868234

ABSTRACT

The canonical function of the Hippo signaling pathway is the regulation of organ growth. How this pathway controls cell-fate determination is less well understood. Here, we identify a function of the Hippo pathway in cell-fate decisions in the developing Drosophila eye, exerted through the interaction of Yorkie (Yki) with the transcriptional regulator Bonus (Bon), an ortholog of mammalian transcriptional intermediary factor 1/tripartite motif (TIF1/TRIM) family proteins. Instead of controlling tissue growth, Yki and Bon promote epidermal and antennal fates at the expense of the eye fate. Proteomic, transcriptomic, and genetic analyses reveal that Yki and Bon control these cell-fate decisions by recruiting transcriptional and post-transcriptional co-regulators and by repressing Notch target genes and activating epidermal differentiation genes. Our work expands the range of functions and regulatory mechanisms under Hippo pathway control.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Cell Differentiation , Drosophila/metabolism , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Hippo Signaling Pathway , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mammals/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteomics , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism
9.
G3 (Bethesda) ; 12(6)2022 05 30.
Article in English | MEDLINE | ID: mdl-35471546

ABSTRACT

RNA-binding proteins support neurodevelopment by modulating numerous steps in post-transcriptional regulation, including splicing, export, translation, and turnover of mRNAs that can traffic into axons and dendrites. One such RNA-binding protein is ZC3H14, which is lost in an inherited intellectual disability. The Drosophila melanogaster ZC3H14 ortholog, Nab2, localizes to neuronal nuclei and cytoplasmic ribonucleoprotein granules and is required for olfactory memory and proper axon projection into brain mushroom bodies. Nab2 can act as a translational repressor in conjunction with the Fragile-X mental retardation protein homolog Fmr1 and shares target RNAs with the Fmr1-interacting RNA-binding protein Ataxin-2. However, neuronal signaling pathways regulated by Nab2 and their potential roles outside of mushroom body axons remain undefined. Here, we present an analysis of a brain proteomic dataset that indicates that multiple planar cell polarity proteins are affected by Nab2 loss, and couple this with genetic data that demonstrate that Nab2 has a previously unappreciated role in restricting the growth and branching of dendrites that elaborate from larval body-wall sensory neurons. Further analysis confirms that Nab2 loss sensitizes sensory dendrites to the genetic dose of planar cell polarity components and that Nab2-planar cell polarity genetic interactions are also observed during Nab2-dependent control of axon projection in the central nervous system mushroom bodies. Collectively, these data identify the conserved Nab2 RNA-binding protein as a likely component of post-transcriptional mechanisms that limit dendrite growth and branching in Drosophila sensory neurons and genetically link this role to the planar cell polarity pathway. Given that mammalian ZC3H14 localizes to dendritic spines and controls spine density in hippocampal neurons, these Nab2-planar cell polarity genetic data may highlight a conserved path through which Nab2/ZC3H14 loss affects morphogenesis of both axons and dendrites in diverse species.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Axons/metabolism , Cell Polarity/genetics , Dendrites/metabolism , Drosophila/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Mammals , Proteomics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
10.
Genetics ; 220(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34791182

ABSTRACT

Nab2 encodes the Drosophila melanogaster member of a conserved family of zinc finger polyadenosine RNA-binding proteins (RBPs) linked to multiple steps in post-transcriptional regulation. Mutation of the Nab2 human ortholog ZC3H14 gives rise to an autosomal recessive intellectual disability but understanding of Nab2/ZC3H14 function in metazoan nervous systems is limited, in part because no comprehensive identification of metazoan Nab2/ZC3H14-associated RNA transcripts has yet been conducted. Moreover, many Nab2/ZC3H14 functional protein partnerships remain unidentified. Here, we present evidence that Nab2 genetically interacts with Ataxin-2 (Atx2), which encodes a neuronal translational regulator, and that these factors coordinately regulate neuronal morphology, circadian behavior, and adult viability. We then present the first high-throughput identifications of Nab2- and Atx2-associated RNAs in Drosophila brain neurons using RNA immunoprecipitation-sequencing (RIP-Seq). Critically, the RNA interactomes of each RBP overlap, and Nab2 exhibits high specificity in its RNA associations in neurons in vivo, associating with a small fraction of all polyadenylated RNAs. The identities of shared associated transcripts (e.g., drk, me31B, stai) and of transcripts specific to Nab2 or Atx2 (e.g., Arpc2 and tea) promise insight into neuronal functions of, and genetic interactions between, each RBP. Consistent with prior biochemical studies, Nab2-associated neuronal RNAs are overrepresented for internal A-rich motifs, suggesting these sequences may partially mediate Nab2 target selection. These data support a model where Nab2 functionally opposes Atx2 in neurons, demonstrate Nab2 shares associated neuronal RNAs with Atx2, and reveal Drosophila Nab2 associates with a more specific subset of polyadenylated mRNAs than its polyadenosine affinity alone may suggest.


Subject(s)
Drosophila melanogaster , Animals
11.
STAR Protoc ; 2(3): 100689, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34382016

ABSTRACT

Drosophila melanogaster is an excellent model organism to study neurodegeneration. Assessing evident neurodegeneration within the fly brain involves the laborious preparation of thin-sectioned H&E-stained heads to visualize brain vacuole degeneration. Here, we present an advanced microscopy-based protocol, without the need for sectioning, to detect vacuole degeneration within whole fly brains by applying commonly used stains to reveal the brain parenchyma. This approach preserves the whole-brain architecture and enables rapid, reproducible, and quantitative analyses of vacuole-like degeneration associated with specific brain regions. For complete details on the use and execution of this protocol, please refer to Behnke et al. (2021).


Subject(s)
Brain/diagnostic imaging , Histological Techniques/methods , Neurodegenerative Diseases/diagnostic imaging , Animals , Brain/metabolism , Drosophila Proteins , Drosophila melanogaster , Microscopy/methods , Neurons/metabolism
12.
J Biol Chem ; 297(1): 100877, 2021 07.
Article in English | MEDLINE | ID: mdl-34139237

ABSTRACT

The human ZC3H14 gene, which encodes a ubiquitously expressed polyadenosine zinc finger RNA-binding protein, is mutated in an inherited form of autosomal recessive, nonsyndromic intellectual disability. To gain insight into neurological functions of ZC3H14, we previously developed a Drosophila melanogaster model of ZC3H14 loss by deleting the fly ortholog, Nab2. Studies in this invertebrate model revealed that Nab2 controls final patterns of neuron projection within fully developed adult brains, but the role of Nab2 during development of the Drosophila brain is not known. Here, we identify roles for Nab2 in controlling the dynamic growth of axons in the developing brain mushroom bodies, which support olfactory learning and memory, and regulating abundance of a small fraction of the total brain proteome. The group of Nab2-regulated brain proteins, identified by quantitative proteomic analysis, includes the microtubule-binding protein Futsch, the neuronal Ig-family transmembrane protein turtle, the glial:neuron adhesion protein contactin, the Rac GTPase-activating protein tumbleweed, and the planar cell polarity factor Van Gogh, which collectively link Nab2 to the processes of brain morphogenesis, neuroblast proliferation, circadian sleep/wake cycles, and synaptic development. Overall, these data indicate that Nab2 controls the abundance of a subset of brain proteins during the active process of wiring the pupal brain mushroom body and thus provide a window into potentially conserved functions of the Nab2/ZC3H14 RNA-binding proteins in neurodevelopment.


Subject(s)
Brain/metabolism , Drosophila Proteins/metabolism , Neurogenesis , Proteome/genetics , RNA-Binding Proteins/metabolism , Animals , Brain/growth & development , Contactins/genetics , Contactins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Developmental , Immunoglobulins/genetics , Immunoglobulins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Memory , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proteome/metabolism , RNA-Binding Proteins/genetics
13.
Sci Rep ; 11(1): 9738, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33958652

ABSTRACT

Mild head trauma, including concussion, can lead to chronic brain dysfunction and degeneration but the underlying mechanisms remain poorly understood. Here, we developed a novel head impact system to investigate the long-term effects of mild head trauma on brain structure and function, as well as the underlying mechanisms in Drosophila melanogaster. We find that Drosophila subjected to repetitive head impacts develop long-term deficits, including impaired startle-induced climbing, progressive brain degeneration, and shortened lifespan, all of which are substantially exacerbated in female flies. Interestingly, head impacts elicit an elevation in neuronal activity and its acute suppression abrogates the detrimental effects in female flies. Together, our findings validate Drosophila as a suitable model system for investigating the long-term effects of mild head trauma, suggest an increased vulnerability to brain injury in female flies, and indicate that early altered neuronal excitability may be a key mechanism linking mild brain trauma to chronic degeneration.


Subject(s)
Brain Injuries/physiopathology , Brain/physiopathology , Craniocerebral Trauma/physiopathology , Drosophila melanogaster , Animals , Brain Injuries/etiology , Craniocerebral Trauma/complications , Disease Models, Animal , Drosophila melanogaster/physiology , Female , Longevity , Male , Neurons/pathology
14.
G3 (Bethesda) ; 10(10): 3575-3583, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32817074

ABSTRACT

Mutations in the gene encoding the ubiquitously expressed RNA-binding protein ZC3H14 result in a non-syndromic form of autosomal recessive intellectual disability in humans. Studies in Drosophila have defined roles for the ZC3H14 ortholog, Nab2 (aka Drosophila Nab2 or dNab2), in axon guidance and memory due in part to interaction with a second RNA-binding protein, the fly Fragile X homolog Fmr1, and coregulation of shared Nab2-Fmr1 target mRNAs. Despite these advances, neurodevelopmental mechanisms that underlie defective axonogenesis in Nab2 mutants remain undefined. Nab2 null phenotypes in the brain mushroom bodies (MBs) resemble defects caused by alleles that disrupt the planar cell polarity (PCP) pathway, which regulates planar orientation of static and motile cells via a non-canonical arm of the Wnt/Wg pathway. A kinked bristle phenotype in surviving Nab2 mutant adults additionally suggests a defect in F-actin polymerization and bundling, a PCP-regulated processes. To test for Nab2-PCP genetic interactions, a collection of PCP mutant alleles was screened for modification of a rough-eye phenotype produced by Nab2 overexpression in the eye (GMR>Nab2) and, subsequently, for modification of a viability defect among Nab2 nulls. Multiple PCP alleles dominantly modify GMR>Nab2 eye roughening and a subset rescue low survival and thoracic bristle kinking in Nab2 zygotic nulls. Collectively, these genetic interactions identify the PCP pathway as a potential target of the Nab2 RNA-binding protein in developing eye and wing tissues and suggest that altered PCP signaling could contribute to neurological defects that result from loss of Drosophila Nab2 or its vertebrate ortholog ZC3H14.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Cell Polarity/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Fragile X Mental Retardation Protein/genetics , RNA-Binding Proteins/genetics
15.
PLoS Genet ; 16(7): e1008901, 2020 07.
Article in English | MEDLINE | ID: mdl-32645003

ABSTRACT

The RNA exosome is an evolutionarily-conserved ribonuclease complex critically important for precise processing and/or complete degradation of a variety of cellular RNAs. The recent discovery that mutations in genes encoding structural RNA exosome subunits cause tissue-specific diseases makes defining the role of this complex within specific tissues critically important. Mutations in the RNA exosome component 3 (EXOSC3) gene cause Pontocerebellar Hypoplasia Type 1b (PCH1b), an autosomal recessive neurologic disorder. The majority of disease-linked mutations are missense mutations that alter evolutionarily-conserved regions of EXOSC3. The tissue-specific defects caused by these amino acid changes in EXOSC3 are challenging to understand based on current models of RNA exosome function with only limited analysis of the complex in any multicellular model in vivo. The goal of this study is to provide insight into how mutations in EXOSC3 impact the function of the RNA exosome. To assess the tissue-specific roles and requirements for the Drosophila ortholog of EXOSC3 termed Rrp40, we utilized tissue-specific RNAi drivers. Depletion of Rrp40 in different tissues reveals a general requirement for Rrp40 in the development of many tissues including the brain, but also highlight an age-dependent requirement for Rrp40 in neurons. To assess the functional consequences of the specific amino acid substitutions in EXOSC3 that cause PCH1b, we used CRISPR/Cas9 gene editing technology to generate flies that model this RNA exosome-linked disease. These flies show reduced viability; however, the surviving animals exhibit a spectrum of behavioral and morphological phenotypes. RNA-seq analysis of these Drosophila Rrp40 mutants reveals increases in the steady-state levels of specific mRNAs and ncRNAs, some of which are central to neuronal function. In particular, Arc1 mRNA, which encodes a key regulator of synaptic plasticity, is increased in the Drosophila Rrp40 mutants. Taken together, this study defines a requirement for the RNA exosome in specific tissues/cell types and provides insight into how defects in RNA exosome function caused by specific amino acid substitutions that occur in PCH1b can contribute to neuronal dysfunction.


Subject(s)
Cerebellar Diseases/genetics , Cytoskeletal Proteins/genetics , Drosophila melanogaster/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , RNA-Binding Proteins/genetics , Amino Acid Substitution/genetics , Animals , CRISPR-Cas Systems/genetics , Cerebellar Diseases/pathology , Cerebellum/metabolism , Cerebellum/pathology , Disease Models, Animal , Exosomes/genetics , Humans , Mutation/genetics , Neurons/pathology , RNA/genetics
16.
Elife ; 92020 04 16.
Article in English | MEDLINE | ID: mdl-32297857

ABSTRACT

Notch pathway haploinsufficiency can cause severe developmental syndromes with highly variable penetrance. Currently, we have a limited mechanistic understanding of phenotype variability due to gene dosage. Here, we unexpectedly found that inserting an enhancer containing pioneer transcription factor sites coupled to Notch dimer sites can induce a subset of Notch haploinsufficiency phenotypes in Drosophila with wild type Notch gene dose. Using Drosophila genetics, we show that this enhancer induces Notch phenotypes in a Cdk8-dependent, transcription-independent manner. We further combined mathematical modeling with quantitative trait and expression analysis to build a model that describes how changes in Notch signal production versus degradation differentially impact cellular outcomes that require long versus short signal duration. Altogether, these findings support a 'bind and discard' mechanism in which enhancers with specific binding sites promote rapid Cdk8-dependent Notch turnover, and thereby reduce Notch-dependent transcription at other loci and sensitize tissues to gene dose based upon signal duration.


Subject(s)
Drosophila Proteins/genetics , Enhancer Elements, Genetic/genetics , Haploinsufficiency/genetics , Models, Genetic , Models, Theoretical , Receptors, Notch/genetics , Animals , Drosophila , Phenotype
17.
EMBO Rep ; 21(1): e49647, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31788927

ABSTRACT

The transcriptional co-activators YAP and AIB1 individually promote breast cancer progression, but are not known to be mechanistically linked. A study published in this issue of EMBO Reports [1] now shows that YAP-AIB1 form a physical complex in breast epithelial cells that cooperates in both activation and, unexpectedly, repression of key breast cancer genes. The repressive effect is due to the recruitment of ANCO1, a previously defined AIB1 interactor [2] that binds and inhibits the YAP-AIB1 complex. These data identify ANCO1 as a candidate tumor suppressor through YAP-AIB1 inhibition and could hint at a broader crosstalk between pathways that utilize YAP and AIB1 to control epithelial homeostasis.


Subject(s)
Oncogenes , Disease Progression , Humans , Nuclear Receptor Coactivator 3/genetics
18.
Hum Mol Genet ; 28(14): 2309-2318, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30985904

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by progressive muscular atrophy and respiratory failure. The G4C2 repeat expansion in the C9orf72 gene is the most prevalent genetic risk for ALS. Mutation carriers (C9ALS) display variability in phenotypes such as age-at-onset and duration, suggesting the existence of additional genetic factors. Here we introduce a three-step gene discovery strategy to identify genetic factors modifying the risk of both C9ALS and sporadic ALS (sALS) using limited samples. We first identified 135 candidate genetic modifiers of C9ALS using whole-genome sequencing (WGS) of extreme C9ALS cases diagnosed ~30 years apart. We then performed an unbiased genetic screen using a Drosophila model of the G4C2 repeat expansion with the genes identified from WGS analysis. This genetic screen identified the novel genetic interaction between G4C2 repeat-associated toxicity and 18 genetic factors, suggesting their potential association with C9ALS risk. We went on to test if 14 out of the 18 genes, those which were not known to be risk factors for ALS previously, are also associated with ALS risk in sALS cases. Gene-based-statistical analyses of targeted resequencing and WGS were performed. These analyses together reveal that rare variants in MYH15 represent a likely genetic risk factor for ALS. Furthermore, we show that MYH15 could modulate the toxicity of dipeptides produced from expanded G4C2 repeat. Our study presented here demonstrates the power of combining WGS with fly genetics to facilitate the discovery of fundamental genetic components of complex traits with a limited number of samples.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , DNA Repeat Expansion , Drosophila/genetics , Myosin Heavy Chains/genetics , Adult , Aged , Animals , Animals, Genetically Modified , C9orf72 Protein/metabolism , C9orf72 Protein/toxicity , Dipeptides/metabolism , Dipeptides/toxicity , Disease Models, Animal , Drosophila/cytology , Drosophila/growth & development , Drosophila/ultrastructure , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Humans , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , Myosin Heavy Chains/metabolism , Phenotype , Risk Factors , Whole Genome Sequencing
19.
Mol Cell ; 71(5): 848-857.e6, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30078725

ABSTRACT

A ten-eleven translocation (TET) ortholog exists as a DNA N6-methyladenine (6mA) demethylase (DMAD) in Drosophila. However, the molecular roles of 6mA and DMAD remain unexplored. Through genome-wide 6mA and transcriptome profiling in Drosophila brains and neuronal cells, we found that 6mA may epigenetically regulate a group of genes involved in neurodevelopment and neuronal functions. Mechanistically, DMAD interacts with the Trithorax-related complex protein Wds to maintain active transcription by dynamically demethylating intragenic 6mA. Accumulation of 6mA by depleting DMAD coordinates with Polycomb proteins and contributes to transcriptional repression of these genes. Our findings suggest that active 6mA demethylation by DMAD plays essential roles in fly CNS by orchestrating through added epigenetic mechanisms.


Subject(s)
Adenine/analogs & derivatives , Gene Expression/physiology , Neurons/metabolism , Polycomb-Group Proteins/metabolism , Adenine/metabolism , Animals , DNA Methylation/physiology , Demethylation , Drosophila/metabolism , Drosophila Proteins/metabolism , Epigenesis, Genetic/physiology , Gene Expression Profiling/methods , Genome/physiology
20.
G3 (Bethesda) ; 8(7): 2355-2360, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29752350

ABSTRACT

The Drosophila domino locus encodes DNA-dependent ATPases of the SWI2/SNF2 class. This class of chromatin remodeler is associated with an array of cellular activities encompassing transcription, replication, repair and recombination. Moreover, domino was observed initially to maintain a repressive chromatin state via genetic interaction studies with homeotic genes. Although domino mutations were also characterized with a cell death phenotype, its association with a death pathway has not been investigated. Here we have used targeted RNA interference to depress domino function in the wing. Resultant wing damage phenotypes were found to be enhanced through overexpression of pro-apoptotic loci, and suppressed through loss of function of these loci. Loss of wing margin and blade tissue was correlated with activation of the effector Caspase Dcp-1, a marker for apoptosis. The affected wing regions also exhibited lower levels of the DIAP1 protein, an inhibitor of apoptosis. The lower level of DIAP1 protein was not correlated with an effect on the activity of a DIAP1 gene transgenic reporter (thread-LacZ), suggesting that loss of DIAP1 occurred post transcriptionally. In some cases excessive cell proliferation within the targeted tissue, measured through BrdU incorporation, was also observed. Finally, we used a transgenic reporter construct to monitor the chromatin state upstream of the proapoptotic reaper locus. In genotypes exhibiting targeted domino loss and wing phenotypes, we observed increased reporter activity only in the affected areas. These data support the conclusion that domino normally functions to maintain pro-apoptotic genes in a repressed state.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Animals , Animals, Genetically Modified , Apoptosis/genetics , Biomarkers , Cell Death/genetics , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Epistasis, Genetic , Fluorescent Antibody Technique , Gene Expression Regulation , Genotype , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL