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










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38741938

ABSTRACT

Fragile X Syndrome (FXS) is the most prevalent monogenetic form of intellectual disability and autism. Recently, dysregulation of insulin signaling (IS) and aberrations in mitochondrial function have emerged as robust, evolutionarily conserved components of FXS pathophysiology. However, the mechanisms by which altered IS and mitochondrial dysfunction impact behavior in the context of FXS remain elusive. Here, we show that normalization of IS improves mitochondrial volume and function in flies that lack expression of dfmr1, the Drosophila homolog of the causal gene of FXS in humans. Further, we demonstrate that dysregulation of IS underlies diminished expression of the mitochondrial master regulator PGC-1α/Spargel in dfmr1 mutant flies. These results are behaviorally relevant, as we show that pan-neuronal augmentation of PGC-1α/Spargel improves circadian behavior in dfmr1 mutants. Notably, we also show that modulation of PGC-1α/Spargel expression in wild-type flies phenocopies the dfmr1 mutant circadian defect. Taken together, the results presented herein provide a mechanistic link between mitochondrial function and circadian behavior both in FXS pathogenesis as well as more broadly at the interface between metabolism and behavioral output.

2.
Hum Mol Genet ; 31(20): 3422-3438, 2022 10 10.
Article in English | MEDLINE | ID: mdl-35617143

ABSTRACT

Although autism is typically characterized by differences in language, social interaction and restrictive, repetitive behaviors, it is becoming more well known in the field that alterations in energy metabolism and mitochondrial function are comorbid disorders in autism. The synaptic cell adhesion molecule, neurexin-1 (NRXN1), has previously been implicated in autism, and here we show that in Drosophila melanogaster, the homologue of NRXN1, called Nrx-1, regulates energy metabolism and nutrient homeostasis. First, we show that Nrx-1-null flies exhibit decreased resistance to nutrient deprivation and heat stress compared to controls. Additionally, Nrx-1 mutants exhibit a significantly altered metabolic profile characterized by decreased lipid and carbohydrate stores. Nrx-1-null Drosophila also exhibit diminished levels of nicotinamide adenine dinucleotide (NAD+), an important coenzyme in major energy metabolism pathways. Moreover, loss of Nrx-1 resulted in striking abnormalities in mitochondrial morphology in the flight muscle of Nrx-1-null Drosophila and impaired flight ability in these flies. Further, following a mechanical shock Nrx-1-null flies exhibited seizure-like activity, a phenotype previously linked to defects in mitochondrial metabolism and a common symptom of patients with NRXN1 deletions. The current studies indicate a novel role for NRXN1 in the regulation of energy metabolism and uncover a clinically relevant seizure phenotype in Drosophila lacking Nrx-1.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Carbohydrates , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Energy Metabolism/genetics , Lipids , NAD/metabolism , Seizures/genetics
3.
Nat Commun ; 12(1): 4578, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321481

ABSTRACT

Mitochondria are transported along microtubules by opposing kinesin and dynein motors. Kinesin-1 and dynein-dynactin are linked to mitochondria by TRAK proteins, but it is unclear how TRAKs coordinate these motors. We used single-molecule imaging of cell lysates to show that TRAK2 robustly activates kinesin-1 for transport toward the microtubule plus-end. TRAK2 is also a novel dynein activating adaptor that utilizes a conserved coiled-coil motif to interact with dynein to promote motility toward the microtubule minus-end. However, dynein-mediated TRAK2 transport is minimal unless the dynein-binding protein LIS1 is present at a sufficient level. Using co-immunoprecipitation and co-localization experiments, we demonstrate that TRAK2 forms a complex containing both kinesin-1 and dynein-dynactin. These motors are functionally linked by TRAK2 as knockdown of either kinesin-1 or dynein-dynactin reduces the initiation of TRAK2 transport toward either microtubule end. We propose that TRAK2 coordinates kinesin-1 and dynein-dynactin as an interdependent motor complex, providing integrated control of opposing motors for the proper transport of mitochondria.


Subject(s)
Dyneins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kinesins/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Carrier Proteins/metabolism , Dynactin Complex/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Microtubule-Associated Proteins , Microtubules/metabolism , Nerve Tissue Proteins/genetics , Protein Transport/physiology , Transcriptome
4.
Curr Opin Cell Biol ; 68: 28-36, 2021 02.
Article in English | MEDLINE | ID: mdl-32961383

ABSTRACT

Mitochondria form networks that continually remodel and adapt to carry out their cellular function. The mitochondrial network is remodeled through changes in mitochondrial morphology, number, and distribution within the cell. Mitochondrial dynamics depend directly on fission, fusion, shape transition, and transport or tethering along the cytoskeleton. Over the past several years, many of the mechanisms underlying these processes have been uncovered. It has become clear that each process is precisely and contextually regulated within the cell. Here, we discuss the mechanisms regulating each aspect of mitochondrial dynamics, which together shape the network as a whole.


Subject(s)
Mitochondria/physiology , Mitochondrial Dynamics , Animals , Cytoskeleton/chemistry , Cytoskeleton/physiology , Humans , Mitochondria/chemistry , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/physiology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/physiology , Organelle Shape
5.
Neurobiol Learn Mem ; 165: 107000, 2019 11.
Article in English | MEDLINE | ID: mdl-30797034

ABSTRACT

Genome-wide association and whole exome sequencing studies from Autism Spectrum Disorder (ASD) patient populations have implicated numerous risk factor genes whose mutation or deletion results in significantly increased incidence of ASD. Behavioral studies of monogenic mutant mouse models of ASD-associated genes have been useful for identifying aberrant neural circuitry. However, behavioral results often differ from lab to lab, and studies incorporating both males and females are often not performed despite the significant sex-bias of ASD. In this study, we sought to investigate the simple, passive behavior of home-cage activity monitoring across multiple 24-h days in four different monogenic mouse models of ASD: Shank3b-/-, Cntnap2-/-, Pcdh10+/-, and Fmr1 knockout mice. Relative to sex-matched wildtype (WT) littermates, we discovered significant home-cage hypoactivity, particularly in the dark (active) phase of the light/dark cycle, in male mice of all four ASD-associated transgenic models. For Cntnap2-/- and Pcdh10+/- mice, these activity alterations were sex-specific, as female mice did not exhibit home-cage activity differences relative to sex-matched WT controls. These home-cage hypoactivity alterations differ from activity findings previously reported using short-term activity measurements in a novel open field. Despite circadian problems reported in human ASD patients, none of the mouse models studied had alterations in free-running circadian period. Together, these findings highlight a shared phenotype across several monogenic mouse models of ASD, outline the importance of methodology on behavioral interpretation, and in some genetic lines parallel the male-enhanced phenotypic presentation observed in human ASDs.


Subject(s)
Autism Spectrum Disorder/genetics , Disease Models, Animal , Motor Activity/genetics , Animals , Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/psychology , Cadherins/genetics , Cadherins/physiology , Circadian Rhythm , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/physiology , Male , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Knockout , Microfilament Proteins , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Protocadherins , Sex Factors
6.
Hum Mol Genet ; 27(1): 95-106, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29106525

ABSTRACT

Fragile X Syndrome (FXS), the most prevalent form of inherited intellectual disability and the foremost monogenetic cause of autism, is caused by loss of expression of the FMR1 gene . Here, we show that dfmr1 modulates the global metabolome in Drosophila. Despite our previous discovery of increased brain insulin signaling, our results indicate that dfmr1 mutants have reduced carbohydrate and lipid stores and are hypersensitive to starvation stress. The observed metabolic deficits cannot be explained by feeding behavior, as we report that dfmr1 mutants are hyperphagic. Rather, our data identify dfmr1 as a regulator of mitochondrial function. We demonstrate that under supersaturating conditions, dfmr1 mutant mitochondria have significantly increased maximum electron transport system (ETS) capacity. Moreover, electron micrographs of indirect flight muscle reveal striking morphological changes in the dfmr1 mutant mitochondria. Taken together, our results illustrate the importance of dfmr1 for proper maintenance of nutrient homeostasis and mitochondrial function.


Subject(s)
Drosophila Proteins/genetics , Fragile X Mental Retardation Protein/genetics , Animals , Disease Models, Animal , Drosophila , Drosophila Proteins/metabolism , Drosophila melanogaster , Energy Metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Male , Mitochondria/metabolism , Signal Transduction
7.
Front Behav Neurosci ; 10: 136, 2016.
Article in English | MEDLINE | ID: mdl-27445731

ABSTRACT

Fragile X is the most common monogenic disorder associated with intellectual disability (ID) and autism spectrum disorders (ASD). Additionally, many patients are afflicted with executive dysfunction, ADHD, seizure disorder and sleep disturbances. Fragile X is caused by loss of FMRP expression, which is encoded by the FMR1 gene. Both the fly and mouse models of fragile X are also based on having no functional protein expression of their respective FMR1 homologs. The fly model displays well defined cognitive impairments and structural brain defects and the mouse model, although having subtle behavioral defects, has robust electrophysiological phenotypes and provides a tool to do extensive biochemical analysis of select brain regions. Decreased cAMP signaling has been observed in samples from the fly and mouse models of fragile X as well as in samples derived from human patients. Indeed, we have previously demonstrated that strategies that increase cAMP signaling can rescue short term memory in the fly model and restore DHPG induced mGluR mediated long term depression (LTD) in the hippocampus to proper levels in the mouse model (McBride et al., 2005; Choi et al., 2011, 2015). Here, we demonstrate that the same three strategies used previously with the potential to be used clinically, lithium treatment, PDE-4 inhibitor treatment or mGluR antagonist treatment can rescue long term memory in the fly model and alter the cAMP signaling pathway in the hippocampus of the mouse model.

8.
PLoS Genet ; 11(11): e1005655, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26544867

ABSTRACT

Individuals with Cornelia de Lange Syndrome (CdLS) display diverse developmental deficits, including slow growth, multiple limb and organ abnormalities, and intellectual disabilities. Severely-affected individuals most often have dominant loss-of-function mutations in the Nipped-B-Like (NIPBL) gene, and milder cases often have missense or in-frame deletion mutations in genes encoding subunits of the cohesin complex. Cohesin mediates sister chromatid cohesion to facilitate accurate chromosome segregation, and NIPBL is required for cohesin to bind to chromosomes. Individuals with CdLS, however, do not display overt cohesion or segregation defects. Rather, studies in human cells and model organisms indicate that modest decreases in NIPBL and cohesin activity alter the transcription of many genes that regulate growth and development. Sister chromatid cohesion factors, including the Nipped-B ortholog of NIPBL, are also critical for gene expression and development in Drosophila melanogaster. Here we describe how a modest reduction in Nipped-B activity alters growth and neurological function in Drosophila. These studies reveal that Nipped-B heterozygous mutant Drosophila show reduced growth, learning, and memory, and altered circadian rhythms. Importantly, the growth deficits are not caused by changes in systemic growth controls, but reductions in cell number and size attributable in part to reduced expression of myc (diminutive) and other growth control genes. The learning, memory and circadian deficits are accompanied by morphological abnormalities in brain structure. These studies confirm that Drosophila Nipped-B mutants provide a useful model for understanding CdLS, and provide new insights into the origins of birth defects.


Subject(s)
DNA-Binding Proteins/genetics , De Lange Syndrome/genetics , Drosophila Proteins/genetics , Drosophila/growth & development , Drosophila/physiology , Models, Biological , Mutation , Animals , Drosophila/genetics , Heterozygote
9.
Exp Neurol ; 274(Pt A): 14-24, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26026973

ABSTRACT

Fragile X Syndrome (FXS) is the most common heritable form of intellectual impairment as well as the leading monogenetic cause of autism. In addition to its canonical definition as a neurodevelopmental disease, recent findings in the clinic suggest that FXS is a systemic disorder that is characterized by a variety of heterogeneous phenotypes. Efforts to study FXS pathogenesis have been aided by the development and characterization of animal models of the disease. Research efforts in Drosophila melanogaster have revealed key insights into the mechanistic underpinnings of FXS. While much remains unknown, it is increasingly apparent that FXS involves a myriad of spatially and temporally specific alterations in cellular function. Consequently, the literature is filled with numerous discordant findings. Researchers and clinicians alike must be cognizant of this dissonance, as it will likely be important for the design of preclinical studies to assess the efficacy of therapeutic strategies to improve the lives of FXS patients.


Subject(s)
Biomedical Research/methods , Comprehension , Drosophila Proteins/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Animals , Biomedical Research/trends , Drosophila , Fragile X Syndrome/pathology , Humans , Neural Stem Cells/physiology , Time Factors
10.
Cell Rep ; 11(5): 727-36, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25921541

ABSTRACT

The PI3K enhancer PIKE links PI3K catalytic subunits to group 1 metabotropic glutamate receptors (mGlu1/5) and activates PI3K signaling. The roles of PIKE in synaptic plasticity and the etiology of mental disorders are unknown. Here, we show that increased PIKE expression is a key mediator of impaired mGlu1/5-dependent neuronal plasticity in mouse and fly models of the inherited intellectual disability fragile X syndrome (FXS). Normalizing elevated PIKE protein levels in FXS mice reversed deficits in molecular and cellular plasticity and improved behavior. Notably, PIKE reduction rescued PI3K-dependent and -independent neuronal defects in FXS. We further show that PI3K signaling is increased in a fly model of FXS and that genetic reduction of the Drosophila ortholog of PIKE, CenG1A rescued excessive PI3K signaling, mushroom body defects, and impaired short-term memory in these flies. Our results demonstrate a crucial role of increased PIKE expression in exaggerated mGlu1/5 signaling causing neuronal defects in FXS.


Subject(s)
Behavior, Animal/physiology , Fragile X Syndrome/pathology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Animals , Brain/metabolism , Dendritic Spines/metabolism , Disease Models, Animal , Drosophila/metabolism , Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , GTPase-Activating Proteins/metabolism , Mice , Mice, Knockout , Mushroom Bodies/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Biosynthesis , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction
11.
J Neurosci ; 35(1): 396-408, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25568131

ABSTRACT

Fragile X syndrome (FXS) is the leading cause of both intellectual disability and autism resulting from a single gene mutation. Previously, we characterized cognitive impairments and brain structural defects in a Drosophila model of FXS and demonstrated that these impairments were rescued by treatment with metabotropic glutamate receptor (mGluR) antagonists or lithium. A well-documented biochemical defect observed in fly and mouse FXS models and FXS patients is low cAMP levels. cAMP levels can be regulated by mGluR signaling. Herein, we demonstrate PDE-4 inhibition as a therapeutic strategy to ameliorate memory impairments and brain structural defects in the Drosophila model of fragile X. Furthermore, we examine the effects of PDE-4 inhibition by pharmacologic treatment in the fragile X mouse model. We demonstrate that acute inhibition of PDE-4 by pharmacologic treatment in hippocampal slices rescues the enhanced mGluR-dependent LTD phenotype observed in FXS mice. Additionally, we find that chronic treatment of FXS model mice, in adulthood, also restores the level of mGluR-dependent LTD to that observed in wild-type animals. Translating the findings of successful pharmacologic intervention from the Drosophila model into the mouse model of FXS is an important advance, in that this identifies and validates PDE-4 inhibition as potential therapeutic intervention for the treatment of individuals afflicted with FXS.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Disease Models, Animal , Fragile X Syndrome/enzymology , Neuronal Plasticity/physiology , Phosphodiesterase 4 Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Drosophila , Female , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Male , Mice , Mice, Knockout , Neuronal Plasticity/drug effects , Phosphodiesterase 4 Inhibitors/therapeutic use
12.
Cell Rep ; 8(5): 1265-70, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25176649

ABSTRACT

Neurofibromatosis type 1 (NF1) is a common autosomal-dominant disorder associated with attention deficits and learning disabilities. The primary known function of neurofibromin, encoded by the NF1 gene, is to downregulate Ras activity. We show that nf1-deficient zebrafish exhibit learning and memory deficits and that acute pharmacological inhibition of downstream targets of Ras (MAPK and PI3K) restores memory consolidation and recall but not learning. Conversely, acute pharmacological enhancement of cAMP signaling restores learning but not memory. Our data provide compelling evidence that neurofibromin regulates learning and memory by distinct molecular pathways in vertebrates and that deficits produced by genetic loss of function are reversible. These findings support the investigation of cAMP signaling enhancers as a companion therapy to Ras inhibition in the treatment of cognitive dysfunction in NF1.


Subject(s)
Cyclic AMP/metabolism , Learning , Memory , Neurofibromatosis 1/metabolism , Neurofibromin 1/metabolism , Zebrafish Proteins/metabolism , ras Proteins/metabolism , Animals , MAP Kinase Signaling System , Neurofibromatosis 1/physiopathology , Neurofibromin 1/genetics , Zebrafish , Zebrafish Proteins/genetics
13.
Drug Discov Today Technol ; 10(1): e129-36, 2013.
Article in English | MEDLINE | ID: mdl-24050241

ABSTRACT

Despite obvious differences such as the ability to fly, the fruit fly Drosophila melanogaster is similar to humans at many different levels of complexity. Studies of development, cell growth and division, metabolism and even cognition, have borne out these similarities. For example, Drosophila bearing mutations in the fly gene homologue of the known human disease fragile X are affected in fundamentally similar ways as affected humans. The ramification of this degree of similarity is that Drosophila, as a model organism, is a rich resource for learning about human cells, development and even human cognition and behavior. Drosophila has a short generation time of ten days, is cheap to propagate and maintain and has a vast array of genetic tools available to it; making Drosophila an extremely attractive organism for the study of human disease. Here, we summarize research from our lab and others using Drosophila to understand the human neurological disease, called fragile X. We focus on the Drosophila model of fragile X, its characterization, and use as a tool to identify potential drugs for the treatment of fragile X. Several clinical trials are in progress now that were motivated by this research.


Subject(s)
Disease Models, Animal , Drosophila/genetics , Fragile X Syndrome/drug therapy , Animals , Drug Evaluation, Preclinical , Fragile X Syndrome/physiopathology , Humans
14.
Front Pharmacol ; 4: 64, 2013.
Article in English | MEDLINE | ID: mdl-23720628

ABSTRACT

Metabotropic glutamate receptors (mGluRs) have well-established roles in cognition and social behavior in mammals. Whether or not these roles have been conserved throughout evolution from invertebrate species is less clear. Mammals have eight mGluRs whereas Drosophila has a single DmGluRA, which has both Gi and Gq coupled signaling activity. We have utilized Drosophila to examine the role of DmGluRA in social behavior and various phases of memory. We have found that flies that are homozygous or heterozygous for loss of function mutations of DmGluRA have impaired social behavior in male Drosophila. Futhermore, flies that are heterozygous for loss of function mutations of DmGluRA have impaired learning during training, immediate-recall memory, short-term memory, and long-term memory as young adults. This work demonstrates a role for mGluR activity in both social behavior and memory in Drosophila.

15.
PLoS Genet ; 8(11): e1002996, 2012.
Article in English | MEDLINE | ID: mdl-23133398

ABSTRACT

CHTF18 (chromosome transmission fidelity factor 18) is an evolutionarily conserved subunit of the Replication Factor C-like complex, CTF18-RLC. CHTF18 is necessary for the faithful passage of chromosomes from one daughter cell to the next during mitosis in yeast, and it is crucial for germline development in the fruitfly. Previously, we showed that mouse Chtf18 is expressed throughout the germline, suggesting a role for CHTF18 in mammalian gametogenesis. To determine the role of CHTF18 in mammalian germ cell development, we derived mice carrying null and conditional mutations in the Chtf18 gene. Chtf18-null males exhibit 5-fold decreased sperm concentrations compared to wild-type controls, resulting in subfertility. Loss of Chtf18 results in impaired spermatogenesis; spermatogenic cells display abnormal morphology, and the stereotypical arrangement of cells within seminiferous tubules is perturbed. Meiotic recombination is defective and homologous chromosomes separate prematurely during prophase I. Repair of DNA double-strand breaks is delayed and incomplete; both RAD51 and γH2AX persist in prophase I. In addition, MLH1 foci are decreased in pachynema. These findings demonstrate essential roles for CHTF18 in mammalian spermatogenesis and meiosis, and suggest that CHTF18 may function during the double-strand break repair pathway to promote the formation of crossovers.


Subject(s)
Carrier Proteins/genetics , DNA Breaks, Double-Stranded , Meiosis/genetics , Recombination, Genetic/genetics , Spermatogenesis/genetics , ATPases Associated with Diverse Cellular Activities , Animals , Germ Cells/metabolism , Histones/metabolism , Humans , Infertility/genetics , Male , Meiotic Prophase I , Mice , Nuclear Proteins , Rad51 Recombinase/metabolism , Spermatozoa/pathology
16.
Results Probl Cell Differ ; 54: 83-117, 2012.
Article in English | MEDLINE | ID: mdl-22009349

ABSTRACT

This chapter will briefly tie together a captivating string of scientific discoveries that began in the 1800s and catapulted us into the current state of the field where trials are under way in humans that have arisen directly from the discoveries made in model organisms such as Drosophila (fruit flies) and mice. The hope is that research efforts in the field of fragile X currently represent a roadmap that demonstrates the utility of identifying a mutant gene responsible for human disease, tracking down the molecular underpinnings of pathogenic phenotypes, and utilizing model organisms to identify and validate potential pharmacologic targets for testing in afflicted humans. Indeed, in fragile X this roadmap has already yielded successful trials in humans (J. Med. Genetic 46(4) 266-271; Jacquemont et al. Sci Transl Med 3(64):64ra61), although the work in studying these interventions in humans is just getting underway as the work in model organisms continues to generate new potential therapeutic targets.


Subject(s)
Behavior, Animal/physiology , Drosophila/physiology , Fragile X Syndrome/physiopathology , Animals , Circadian Rhythm/physiology , Cognition/physiology , Disease Models, Animal , Drosophila/anatomy & histology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Learning/physiology , Memory/physiology , Phenotype , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Signal Transduction , Social Behavior
17.
Drug Discov Today Technol ; 10(1): e129-e136, 2012 Sep 24.
Article in English | MEDLINE | ID: mdl-23730322

ABSTRACT

Despite obvious differences such as the ability to fly, the fruit fly Drosophila melanogaster is similar to humans at many different levels of complexity. Studies of development, cell growth and division, metabolism, and even cognition, have borne out these similarities. For example, Drosophila bearing mutations in the fly gene homologue of the known human disease Fragile X, are affected in fundamentally similar ways as affected humans. The ramification of this degree of similarity is that Drosophila, as a model organism, is a rich resource for learning about human cells, development and even human cognition and behavior. Drosophila has a short generation time of ten days, is cheap to propagate and maintain and has a vast array of genetic tools available to it; making Drosophila an extremely attractive organism for the study of human disease. Here, we summarize research from our lab and others using Drosophila to understand the human neurological disease, called Fragile X. We focus on the Drosophila model of fragile X, its characterization, and use as a tool to identify potential drugs for the treatment of Fragile X. Several clinical trials are in progress now that were motivated by this research.

18.
Nat Neurosci ; 14(12): 1517-24, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-22037499

ABSTRACT

Loss of FMR1 gene function results in fragile X syndrome, the most common heritable form of intellectual disability. The protein encoded by this locus (FMRP) is an RNA-binding protein that is thought to primarily act as a translational regulator; however, recent studies have implicated FMRP in other mechanisms of gene regulation. We found that the Drosophila fragile X homolog (dFMR1) biochemically interacted with the adenosine-to-inosine RNA-editing enzyme dADAR. Adar and Fmr1 mutant larvae exhibited distinct morphological neuromuscular junction (NMJ) defects. Epistasis experiments based on these phenotypic differences revealed that Adar acts downstream of Fmr1 and that dFMR1 modulates dADAR activity. Furthermore, sequence analyses revealed that a loss or overexpression of dFMR1 affects editing efficiency on certain dADAR targets with defined roles in synaptic transmission. These results link dFMR1 with the RNA-editing pathway and suggest that proper NMJ synaptic architecture requires modulation of dADAR activity by dFMR1.


Subject(s)
Adenosine Deaminase/metabolism , Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein/metabolism , Neuromuscular Junction/metabolism , RNA Editing/genetics , Adenosine Deaminase/genetics , Analysis of Variance , Animals , Animals, Genetically Modified , Cell Line, Transformed , Drosophila , Drosophila Proteins/genetics , Fragile X Mental Retardation Protein/genetics , Gene Expression Regulation, Developmental , Immunoprecipitation , Larva , Mass Spectrometry , Microscopy, Confocal , Mutation , Neuromuscular Junction/cytology , Neuromuscular Junction/genetics , RNA-Binding Proteins , Synaptic Transmission , Transfection
19.
Brain Res ; 1380: 106-19, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21078304

ABSTRACT

Fragile X syndrome is the leading single gene cause of intellectual disabilities. Treatment of a Drosophila model of Fragile X syndrome with metabotropic glutamate receptor (mGluR) antagonists or lithium rescues social and cognitive impairments. A hallmark feature of the Fragile X mouse model is enhanced mGluR-dependent long-term depression (LTD) at Schaffer collateral to CA1 pyramidal synapses of the hippocampus. Here we examine the effects of chronic treatment of Fragile X mice in vivo with lithium or a group II mGluR antagonist on mGluR-LTD at CA1 synapses. We find that long-term lithium treatment initiated during development (5-6 weeks of age) and continued throughout the lifetime of the Fragile X mice until 9-11 months of age restores normal mGluR-LTD. Additionally, chronic short-term treatment beginning in adult Fragile X mice (8 weeks of age) with either lithium or an mGluR antagonist is also able to restore normal mGluR-LTD. Translating the findings of successful pharmacologic intervention from the Drosophila model into the mouse model of Fragile X syndrome is an important advance, in that this identifies and validates these targets as potential therapeutic interventions for the treatment of individuals afflicted with Fragile X syndrome.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Fragile X Syndrome/drug therapy , Lithium Compounds/pharmacology , Neuronal Plasticity/drug effects , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Synaptic Transmission/drug effects , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Disease Models, Animal , Excitatory Amino Acid Antagonists/therapeutic use , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Lithium Compounds/therapeutic use , Male , Mice , Mice, Knockout , Neuronal Plasticity/genetics , Receptors, Metabotropic Glutamate/physiology , Synaptic Transmission/genetics
20.
Dis Model Mech ; 3(11-12): 693-700, 2010.
Article in English | MEDLINE | ID: mdl-20682752

ABSTRACT

Fragile X syndrome (FXS) is a cognitive disorder caused by silencing of the fragile X mental retardation 1 gene (FMR1). Since the discovery of the gene almost two decades ago, most scientific contributions have focused on identifying the molecular function of the fragile X mental retardation protein (FMRP) and understanding how absence of FMR1 gene expression gives rise to the disease phenotypes. The use of model organisms has allowed rapid progression in the FXS field and has given insight into the molecular basis of the disease. The mouse and fly FXS models have enabled studies to identify potential targets and pathways for pharmacological treatment. Here, we briefly review the two primary FXS model systems and describe how studies in these organisms have led us closer to therapeutic treatments for patients afflicted with FXS.


Subject(s)
Disease Models, Animal , Fragile X Syndrome/drug therapy , Molecular Targeted Therapy , Animals , Drosophila melanogaster/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/etiology , Fragile X Syndrome/genetics , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...