Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Publication year range
1.
Brain ; 147(6): 2053-2068, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38739752

ABSTRACT

Aggregation of the RNA-binding protein TAR DNA binding protein (TDP-43) is a hallmark of TDP-proteinopathies including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). As TDP-43 aggregation and dysregulation are causative of neuronal death, there is a special interest in targeting this protein as a therapeutic approach. Previously, we found that TDP-43 extensively co-aggregated with the dual function protein GEF (guanine exchange factor) and RNA-binding protein rho guanine nucleotide exchange factor (RGNEF) in ALS patients. Here, we show that an N-terminal fragment of RGNEF (NF242) interacts directly with the RNA recognition motifs of TDP-43 competing with RNA and that the IPT/TIG domain of NF242 is essential for this interaction. Genetic expression of NF242 in a fruit fly ALS model overexpressing TDP-43 suppressed the neuropathological phenotype increasing lifespan, abolishing motor defects and preventing neurodegeneration. Intracerebroventricular injections of AAV9/NF242 in a severe TDP-43 murine model (rNLS8) improved lifespan and motor phenotype, and decreased neuroinflammation markers. Our results demonstrate an innovative way to target TDP-43 proteinopathies using a protein fragment with a strong affinity for TDP-43 aggregates and a mechanism that includes competition with RNA sequestration, suggesting a promising therapeutic strategy for TDP-43 proteinopathies such as ALS and FTD.


Subject(s)
Amyotrophic Lateral Sclerosis , DNA-Binding Proteins , Disease Models, Animal , Guanine Nucleotide Exchange Factors , Phenotype , Animals , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mice , Humans , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Drosophila , Mice, Transgenic , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Male
2.
Am J Hum Genet ; 104(4): 596-610, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30879640

ABSTRACT

Mutations in several genes encoding components of the SWI/SNF chromatin remodeling complex cause neurodevelopmental disorders (NDDs). Here, we report on five individuals with mutations in SMARCD1; the individuals present with developmental delay, intellectual disability, hypotonia, feeding difficulties, and small hands and feet. Trio exome sequencing proved the mutations to be de novo in four of the five individuals. Mutations in other SWI/SNF components cause Coffin-Siris syndrome, Nicolaides-Baraitser syndrome, or other syndromic and non-syndromic NDDs. Although the individuals presented here have dysmorphisms and some clinical overlap with these syndromes, they lack their typical facial dysmorphisms. To gain insight into the function of SMARCD1 in neurons, we investigated the Drosophila ortholog Bap60 in postmitotic memory-forming neurons of the adult Drosophila mushroom body (MB). Targeted knockdown of Bap60 in the MB of adult flies causes defects in long-term memory. Mushroom-body-specific transcriptome analysis revealed that Bap60 is required for context-dependent expression of genes involved in neuron function and development in juvenile flies when synaptic connections are actively being formed in response to experience. Taken together, we identify an NDD caused by SMARCD1 mutations and establish a role for the SMARCD1 ortholog Bap60 in the regulation of neurodevelopmental genes during a critical time window of juvenile adult brain development when neuronal circuits that are required for learning and memory are formed.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Memory , Neurodevelopmental Disorders/genetics , Neurons/metabolism , Animals , Child , Child, Preschool , Developmental Disabilities/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Gene Expression Regulation , Humans , Intellectual Disability/genetics , Learning , Male , Mitosis , Muscle Hypotonia/genetics , Mushroom Bodies , Mutation , Syndrome , Transcription Factors/genetics
3.
Mol Psychiatry ; 26(6): 2013-2024, 2021 06.
Article in English | MEDLINE | ID: mdl-32346159

ABSTRACT

Defects in histone methyltransferases (HMTs) are major contributing factors in neurodevelopmental disorders (NDDs). Heterozygous variants of SETD1A involved in histone H3 lysine 4 (H3K4) methylation were previously identified in individuals with schizophrenia. Here, we define the clinical features of the Mendelian syndrome associated with haploinsufficiency of SETD1A by investigating 15 predominantly pediatric individuals who all have de novo SETD1A variants. These individuals present with a core set of symptoms comprising global developmental delay and/or intellectual disability, subtle facial dysmorphisms, behavioral and psychiatric problems. We examined cellular phenotypes in three patient-derived lymphoblastoid cell lines with three variants: p.Gly535Alafs*12, c.4582-2_4582delAG, and p.Tyr1499Asp. These patient cell lines displayed DNA damage repair defects that were comparable to previously observed RNAi-mediated depletion of SETD1A. This suggested that these variants, including the p.Tyr1499Asp in the catalytic SET domain, behave as loss-of-function (LoF) alleles. Previous studies demonstrated a role for SETD1A in cell cycle control and differentiation. However, individuals with SETD1A variants do not show major structural brain defects or severe microcephaly, suggesting that defective proliferation and differentiation of neural progenitors is unlikely the single underlying cause of the disorder. We show here that the Drosophila melanogaster SETD1A orthologue is required in postmitotic neurons of the fly brain for normal memory, suggesting a role in post development neuronal function. Together, this study defines a neurodevelopmental disorder caused by dominant de novo LoF variants in SETD1A and further supports a role for H3K4 methyltransferases in the regulation of neuronal processes underlying normal cognitive functioning.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Animals , Child , Drosophila , Drosophila melanogaster , Haploinsufficiency/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics
4.
PLoS Biol ; 17(3): e2006146, 2019 03.
Article in English | MEDLINE | ID: mdl-30860988

ABSTRACT

Stress responses are crucial processes that require activation of genetic programs that protect from the stressor. Stress responses are also energy consuming and can thus be deleterious to the organism. The mechanisms coordinating energy consumption during stress response in multicellular organisms are not well understood. Here, we show that loss of the epigenetic regulator G9a in Drosophila causes a shift in the transcriptional and metabolic responses to oxidative stress (OS) that leads to decreased survival time upon feeding the xenobiotic paraquat. During OS exposure, G9a mutants show overactivation of stress response genes, rapid depletion of glycogen, and inability to access lipid energy stores. The OS survival deficiency of G9a mutants can be rescued by a high-sugar diet. Control flies also show improved OS survival when fed a high-sugar diet, suggesting that energy availability is generally a limiting factor for OS tolerance. Directly limiting access to glycogen stores by knocking down glycogen phosphorylase recapitulates the OS-induced survival defects of G9a mutants. We propose that G9a mutants are sensitive to stress because they experience a net reduction in available energy due to (1) rapid glycogen use, (2) an inability to access lipid energy stores, and (3) an overinduced transcriptional response to stress that further exacerbates energy demands. This suggests that G9a acts as a critical regulatory hub between the transcriptional and metabolic responses to OS. Our findings, together with recent studies that established a role for G9a in hypoxia resistance in cancer cell lines, suggest that G9a is of wide importance in controlling the cellular and organismal response to multiple types of stress.


Subject(s)
Histone Methyltransferases/metabolism , Animals , Antioxidants/metabolism , Energy Metabolism/genetics , Energy Metabolism/physiology , Epigenesis, Genetic/genetics , Glycogen Phosphorylase/genetics , Glycogen Phosphorylase/metabolism , Histone Methyltransferases/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , Oxidative Stress/genetics , Oxidative Stress/physiology , Phylogeny , Sequence Analysis, RNA
5.
BMC Biol ; 19(1): 112, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34030685

ABSTRACT

BACKGROUND: Resistance and tolerance are two coexisting defense strategies for fighting infections. Resistance is mediated by signaling pathways that induce transcriptional activation of resistance factors that directly eliminate the pathogen. Tolerance refers to adaptations that limit the health impact of a given pathogen burden, without targeting the infectious agent. The key players governing immune tolerance are largely unknown. In Drosophila, the histone H3 lysine 9 (H3K9) methyltransferase G9a was shown to mediate tolerance to virus infection and oxidative stress (OS), suggesting that abiotic stresses like OS may also evoke tolerance mechanisms. In response to both virus and OS, stress resistance genes were overinduced in Drosophila G9a mutants, suggesting an intact but overactive stress response. We recently demonstrated that G9a promotes tolerance to OS by maintaining metabolic homeostasis and safeguarding energy availability, but it remained unclear if this mechanism also applies to viral infection, or is conserved in other species and stress responses. To address these questions, we analyzed publicly available datasets from Drosophila, mouse, and human in which global gene expression levels were measured in G9a-depleted conditions and controls at different time points upon stress exposure. RESULTS: In all investigated datasets, G9a attenuates the transcriptional stress responses that confer resistance against the encountered stressor. Comparative analysis of conserved G9a-dependent stress response genes suggests that G9a is an intimate part of the design principles of stress resistance, buffering the induction of promiscuous stress signaling pathways and stress-specific resistance factors. Importantly, we find stress-dependent downregulation of metabolic genes to also be dependent on G9a across all of the tested datasets. CONCLUSIONS: These results suggest that G9a sets the balance between activation of resistance genes and maintaining metabolic homeostasis, thereby ensuring optimal organismal performance during exposure to diverse types of stress across different species. We therefore propose G9a as a potentially conserved master regulator underlying the widely important, yet poorly understood, concept of stress tolerance.


Subject(s)
Epigenesis, Genetic , Animals , Drosophila/genetics , Drosophila/metabolism , Epigenomics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Mice , Oxidative Stress/genetics , Transcription, Genetic
6.
J Neurogenet ; 35(3): 154-167, 2021 09.
Article in English | MEDLINE | ID: mdl-33522326

ABSTRACT

Drosophila melanogaster males reduce courtship behaviour after mating failure. In the lab, such conditioned courtship suppression, aka 'courtship conditioning', serves as a complex learning and memory assay. Interestingly, variations in the courtship conditioning assay can establish different types of memory. Here, we review research investigating the underlying cellular and molecular mechanisms that allow male flies to form memories of previous mating failures.


Subject(s)
Courtship , Drosophila melanogaster/physiology , Memory/physiology , Sexual Behavior, Animal/physiology , Animals , Conditioning, Classical , Male
7.
Proc Natl Acad Sci U S A ; 114(47): 12518-12523, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29078350

ABSTRACT

Little is known about how genetic variation and epigenetic marks interact to shape differences in behavior. The foraging (for) gene regulates behavioral differences between the rover and sitter Drosophila melanogaster strains, but the molecular mechanisms through which it does so have remained elusive. We show that the epigenetic regulator G9a interacts with for to regulate strain-specific adult foraging behavior through allele-specific histone methylation of a for promoter (pr4). Rovers have higher pr4 H3K9me dimethylation, lower pr4 RNA expression, and higher foraging scores than sitters. The rover-sitter differences disappear in the presence of G9a null mutant alleles, showing that G9a is necessary for these differences. Furthermore, rover foraging scores can be phenocopied by transgenically reducing pr4 expression in sitters. This compelling evidence shows that genetic variation can interact with an epigenetic modifier to produce differences in gene expression, establishing a behavioral polymorphism in Drosophila.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Alleles , Animals , Base Sequence , Cyclic GMP-Dependent Protein Kinases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Exploratory Behavior , Gene Deletion , Genetic Variation , Histone-Lysine N-Methyltransferase/deficiency , Histones/metabolism , Methylation , Phenotype , Promoter Regions, Genetic
8.
PLoS Genet ; 13(10): e1006864, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29069077

ABSTRACT

Kleefstra syndrome, caused by haploinsufficiency of euchromatin histone methyltransferase 1 (EHMT1), is characterized by intellectual disability (ID), autism spectrum disorder (ASD), characteristic facial dysmorphisms, and other variable clinical features. In addition to EHMT1 mutations, de novo variants were reported in four additional genes (MBD5, SMARCB1, NR1I3, and KMT2C), in single individuals with clinical characteristics overlapping Kleefstra syndrome. Here, we present a novel cohort of five patients with de novo loss of function mutations affecting the histone methyltransferase KMT2C. Our clinical data delineates the KMT2C phenotypic spectrum and reinforces the phenotypic overlap with Kleefstra syndrome and other related ID disorders. To elucidate the common molecular basis of the neuropathology associated with mutations in KMT2C and EHMT1, we characterized the role of the Drosophila KMT2C ortholog, trithorax related (trr), in the nervous system. Similar to the Drosophila EHMT1 ortholog, G9a, trr is required in the mushroom body for short term memory. Trr ChIP-seq identified 3371 binding sites, mainly in the promoter of genes involved in neuronal processes. Transcriptional profiling of pan-neuronal trr knockdown and G9a null mutant fly heads identified 613 and 1123 misregulated genes, respectively. These gene sets show a significant overlap and are associated with nearly identical gene ontology enrichments. The majority of the observed biological convergence is derived from predicted indirect target genes. However, trr and G9a also have common direct targets, including the Drosophila ortholog of Arc (Arc1), a key regulator of synaptic plasticity. Our data highlight the clinical and molecular convergence between the KMT2 and EHMT protein families, which may contribute to a molecular network underlying a larger group of ID/ASD-related disorders.


Subject(s)
Autism Spectrum Disorder/genetics , Craniofacial Abnormalities/genetics , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Heart Defects, Congenital/genetics , Histone-Lysine N-Methyltransferase/genetics , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Adolescent , Adult , Animals , Autism Spectrum Disorder/physiopathology , Binding Sites/genetics , Child , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Constitutive Androstane Receptor , Craniofacial Abnormalities/physiopathology , Drosophila melanogaster/genetics , Female , Gene Expression Regulation , Haploinsufficiency , Heart Defects, Congenital/physiopathology , Histones/genetics , Humans , Intellectual Disability/physiopathology , Male , Mutation , Neuronal Plasticity/genetics , Promoter Regions, Genetic
9.
Hum Mol Genet ; 26(21): 4278-4289, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28973161

ABSTRACT

Defects in neuronal migration cause brain malformations, which are associated with intellectual disability (ID) and epilepsy. Using exome sequencing, we identified compound heterozygous variants (p.Arg71His and p. Leu729ThrfsTer6) in TMTC3, encoding transmembrane and tetratricopeptide repeat containing 3, in four siblings with nocturnal seizures and ID. Three of the four siblings have periventricular nodular heterotopia (PVNH), a common brain malformation caused by failure of neurons to migrate from the ventricular zone to the cortex. Expression analysis using patient-derived cells confirmed reduced TMTC3 transcript levels and loss of the TMTC3 protein compared to parental and control cells. As TMTC3 function is currently unexplored in the brain, we gathered support for a neurobiological role for TMTC3 by generating flies with post-mitotic neuron-specific knockdown of the highly conserved Drosophila melanogaster TMTC3 ortholog, CG4050/tmtc3. Neuron-specific knockdown of tmtc3 in flies resulted in increased susceptibility to induced seizures. Importantly, this phenotype was rescued by neuron-specific expression of human TMTC3, suggesting a role for TMTC3 in seizure biology. In addition, we observed co-localization of TMTC3 in the rat brain with vesicular GABA transporter (VGAT), a presynaptic marker for inhibitory synapses. TMTC3 is localized at VGAT positive pre-synaptic terminals and boutons in the rat hypothalamus and piriform cortex, suggesting a role for TMTC3 in the regulation of GABAergic inhibitory synapses. TMTC3 did not co-localize with Vglut2, a presynaptic marker for excitatory neurons. Our data identified TMTC3 as a synaptic protein that is involved in PVNH with ID and epilepsy, in addition to its previously described association with cobblestone lissencephaly.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Periventricular Nodular Heterotopia/metabolism , Adult , Animals , Brain/abnormalities , Cerebral Cortex/metabolism , Drosophila melanogaster , Epilepsy/genetics , Epilepsy/metabolism , Female , Gene Knockdown Techniques , Heterozygote , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Nervous System Malformations/metabolism , Neurons/metabolism , Pedigree , Periventricular Nodular Heterotopia/genetics , Presynaptic Terminals , Rats , Seizures/metabolism , Synapses/metabolism , Exome Sequencing
10.
Am J Hum Genet ; 98(1): 149-64, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26748517

ABSTRACT

Intellectual disability (ID) disorders are genetically and phenotypically extremely heterogeneous. Can this complexity be depicted in a comprehensive way as a means of facilitating the understanding of ID disorders and their underlying biology? We provide a curated database of 746 currently known genes, mutations in which cause ID (ID-associated genes [ID-AGs]), classified according to ID manifestation and associated clinical features. Using this integrated resource, we show that ID-AGs are substantially enriched with co-expression, protein-protein interactions, and specific biological functions. Systematic identification of highly enriched functional themes and phenotypes revealed typical phenotype combinations characterizing process-defined groups of ID disorders, such as chromatin-related disorders and deficiencies in DNA repair. Strikingly, phenotype classification efficiently breaks down ID-AGs into subsets with significantly elevated biological coherence and predictive power. Custom-made functional Drosophila datasets revealed further characteristic phenotypes among ID-AGs and specific clinical classes. Our study and resource provide systematic insights into the molecular and clinical landscape of ID disorders, represent a significant step toward overcoming current limitations in ID research, and prove the utility of systematic human and cross-species phenomics analyses in highly heterogeneous genetic disorders.


Subject(s)
Intellectual Disability/genetics , Mutation , Phenotype , Animals , Drosophila/genetics , Humans
11.
Hum Mol Genet ; 24(23): 6736-55, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26376863

ABSTRACT

ATP6AP2, an essential accessory component of the vacuolar H+ ATPase (V-ATPase), has been associated with intellectual disability (ID) and Parkinsonism. ATP6AP2 has been implicated in several signalling pathways; however, little is known regarding its role in the nervous system. To decipher its function in behaviour and cognition, we generated and characterized conditional knockdowns of ATP6AP2 in the nervous system of Drosophila and mouse models. In Drosophila, ATP6AP2 knockdown induced defective phototaxis and vacuolated photoreceptor neurons and pigment cells when depleted in eyes and altered short- and long-term memory when depleted in the mushroom body. In mouse, conditional Atp6ap2 deletion in glutamatergic neurons (Atp6ap2(Camk2aCre/0) mice) caused increased spontaneous locomotor activity and altered fear memory. Both Drosophila ATP6AP2 knockdown and Atp6ap2(Camk2aCre/0) mice presented with presynaptic transmission defects, and with an abnormal number and morphology of synapses. In addition, Atp6ap2(Camk2aCre/0) mice showed autophagy defects that led to axonal and neuronal degeneration in the cortex and hippocampus. Surprisingly, axon myelination was affected in our mutant mice, and axonal transport alterations were observed in Drosophila. In accordance with the identified phenotypes across species, genome-wide transcriptome profiling of Atp6ap2(Camk2aCre/0) mouse hippocampi revealed dysregulation of genes involved in myelination, action potential, membrane-bound vesicles and motor behaviour. In summary, ATP6AP2 disruption in mouse and fly leads to cognitive impairment and neurodegeneration, mimicking aspects of the neuropathology associated with ATP6AP2 mutations in humans. Our results identify ATP6AP2 as an essential gene for the nervous system.


Subject(s)
Cognition Disorders/etiology , Drosophila Proteins/genetics , Membrane Proteins/genetics , Nerve Degeneration/etiology , Proton-Translocating ATPases/genetics , Receptors, Cell Surface/genetics , Animals , Brain/metabolism , Brain/physiopathology , Cognition Disorders/genetics , Cognition Disorders/physiopathology , Disease Models, Animal , Drosophila , Female , Gene Knockdown Techniques , Intellectual Disability/genetics , Male , Mice , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/physiology , Neurons/ultrastructure , Parkinsonian Disorders/genetics , Synapses/metabolism , Synapses/physiology , Synapses/ultrastructure
12.
PLoS Pathog ; 11(4): e1004692, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25880195

ABSTRACT

Little is known about the tolerance mechanisms that reduce the negative effects of microbial infection on host fitness. Here, we demonstrate that the histone H3 lysine 9 methyltransferase G9a regulates tolerance to virus infection by shaping the response of the evolutionary conserved Jak-Stat pathway in Drosophila. G9a-deficient mutants are more sensitive to RNA virus infection and succumb faster to infection than wild-type controls, which was associated with strongly increased Jak-Stat dependent responses, but not with major differences in viral load. Genetic experiments indicate that hyperactivated Jak-Stat responses are associated with early lethality in virus-infected flies. Our results identify an essential epigenetic mechanism underlying tolerance to virus infection.


Subject(s)
Drosophila melanogaster/virology , Epigenesis, Genetic , Gene Expression Regulation/immunology , Histone-Lysine N-Methyltransferase/immunology , Immune Tolerance/immunology , RNA Virus Infections/immunology , Animals , Chromatin Immunoprecipitation , Drosophila melanogaster/enzymology , Drosophila melanogaster/immunology , RNA Viruses , Reverse Transcriptase Polymerase Chain Reaction
13.
Biochem Cell Biol ; 94(1): 26-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26198080

ABSTRACT

The euchromatin histone methyltransferases (EHMTs) are an evolutionarily conserved protein family that are known for their ability to dimethylate histone 3 at lysine 9 in euchromatic regions of the genome. In mammals there are two EHMT proteins, G9a, encoded by EHMT2, and GLP, encoded by EHMT1. EHMTs have diverse roles in the differentiation of different tissues and cell types and are involved in adult-specific processes like memory, drug addiction, and immune response. This review discusses recent findings from rodent and Drosophila models that are beginning to reveal the broad biological role and complex mechanistic functioning of EHMT proteins.


Subject(s)
Cell Differentiation , Euchromatin/enzymology , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Adipogenesis , Animals , Drosophila melanogaster , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunity, Cellular , Lysine/metabolism , Memory , Methylation , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/physiology , Neuronal Plasticity , Rats , Substance-Related Disorders/enzymology
14.
Hum Mol Genet ; 22(10): 1960-70, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23390136

ABSTRACT

AnkyrinG, encoded by the ANK3 gene, is involved in neuronal development and signaling. It has previously been implicated in bipolar disorder and schizophrenia by association studies. Most recently, de novo missense mutations in this gene were identified in autistic patients. However, the causative nature of these mutations remained controversial. Here, we report inactivating mutations in the Ankyrin 3 (ANK3) gene in patients with severe cognitive deficits. In a patient with a borderline intelligence, severe attention deficit hyperactivity disorder (ADHD), autism and sleeping problems, all isoforms of the ANK3 gene, were disrupted by a balanced translocation. Furthermore, in a consanguineous family with moderate intellectual disability (ID), an ADHD-like phenotype and behavioral problems, we identified a homozygous truncating frameshift mutation in the longest isoform of the same gene, which represents the first reported familial mutation in the ANK3 gene. The causality of ANK3 mutations in the two families and the role of the gene in cognitive function were supported by memory defects in a Drosophila knockdown model. Thus we demonstrated that ANK3 plays a role in intellectual functioning. In addition, our findings support the suggested association of ANK3 with various neuropsychiatric disorders and illustrate the genetic and molecular relation between a wide range of neurodevelopmental disorders.


Subject(s)
Ankyrins/genetics , Frameshift Mutation , Heterozygote , Homozygote , Mental Disorders/genetics , Neurogenesis/genetics , Sleep Wake Disorders/genetics , Adult , Animals , Disease Models, Animal , Drosophila melanogaster , Female , Gene Knockdown Techniques , Humans , Infant , Male
15.
Hum Mol Genet ; 22(15): 3138-51, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23575228

ABSTRACT

It is estimated that the human mitochondrial proteome consists of 1000-1500 distinct proteins. The majority of these support the various biochemical pathways that are active in these organelles. Individuals with an oxidative phosphorylation disorder of unknown cause provide a unique opportunity to identify novel genes implicated in mitochondrial biology. We identified a homozygous deletion of CEP89 in a patient with isolated complex IV deficiency, intellectual disability and multisystemic problems. CEP89 is a ubiquitously expressed and highly conserved gene of unknown function. Immunocytochemistry and cellular fractionation experiments showed that CEP89 is present both in the cytosol and in the mitochondrial intermembrane space. Furthermore, we ascertained in vitro that downregulation of CEP89 resulted in a severe decrease in complex IV in-gel activity and altered mobility, suggesting that the complex is aberrantly formed. Two-dimensional BN-SDS gel analysis revealed that CEP89 associates with a high-molecular weight complex. Together, these data confirm a role for CEP89 in mitochondrial metabolism. In addition, we modeled CEP89 loss of function in Drosophila. Ubiquitous knockdown of fly Cep89 decreased complex IV activity and resulted in complete lethality. Furthermore, Cep89 is required for mitochondrial integrity, membrane depolarization and synaptic transmission of photoreceptor neurons, and for (sub)synaptic organization of the larval neuromuscular junction. Finally, we tested neuronal Cep89 knockdown flies in the light-off jump reflex habituation assay, which revealed its role in learning. We conclude that CEP89 proteins play an important role in mitochondrial metabolism, especially complex IV activity, and are required for neuronal and cognitive function across evolution.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Mitochondria/metabolism , Neurons/metabolism , Animals , Cell Cycle Proteins/genetics , Child , Chromosomes, Human, Pair 19 , Cytochrome-c Oxidase Deficiency/genetics , Cytochrome-c Oxidase Deficiency/metabolism , Cytosol , Disease Models, Animal , Drosophila/genetics , Drosophila Proteins/genetics , Female , Gene Deletion , Gene Expression , Gene Knockdown Techniques , Homozygote , Humans , Learning , Microtubule-Associated Proteins , Mitochondria/genetics , Mutation , Organ Specificity/genetics , Polymorphism, Single Nucleotide , Protein Transport , Synapses/genetics , Synapses/metabolism
16.
Am J Hum Genet ; 91(1): 73-82, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22726846

ABSTRACT

Intellectual disability (ID) disorders are genetically and phenotypically highly heterogeneous and present a major challenge in clinical genetics and medicine. Although many genes involved in ID have been identified, the etiology is unknown in most affected individuals. Moreover, the function of most genes associated with ID remains poorly characterized. Evidence is accumulating that the control of gene transcription through epigenetic modification of chromatin structure in neurons has an important role in cognitive processes and in the etiology of ID. However, our understanding of the key molecular players and mechanisms in this process is highly fragmentary. Here, we identify a chromatin-modification module that underlies a recognizable form of ID, the Kleefstra syndrome phenotypic spectrum (KSS). In a cohort of KSS individuals without mutations in EHMT1 (the only gene known to be disrupted in KSS until now), we identified de novo mutations in four genes, MBD5, MLL3, SMARCB1, and NR1I3, all of which encode epigenetic regulators. Using Drosophila, we demonstrate that MBD5, MLL3, and NR1I3 cooperate with EHMT1, whereas SMARCB1 is known to directly interact with MLL3. We propose a highly conserved epigenetic network that underlies cognition in health and disease. This network should allow the design of strategies to treat the growing group of ID pathologies that are caused by epigenetic defects.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Intellectual Disability/genetics , Animals , Chromatin , Chromosomal Proteins, Non-Histone/genetics , Constitutive Androstane Receptor , DNA-Binding Proteins/genetics , Drosophila , Epigenesis, Genetic , Female , Humans , Infant, Newborn , Male , Mutation , SMARCB1 Protein , Syndrome , Transcription Factors/genetics
17.
Hum Mutat ; 35(12): 1495-505, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224183

ABSTRACT

Glycoprotein M6A (GPM6A) is a neuronal transmembrane protein of the PLP/DM20 (proteolipid protein) family that associates with cholesterol-rich lipid rafts and promotes filopodia formation. We identified a de novo duplication of the GPM6A gene in a patient with learning disability and behavioral anomalies. Expression analysis in blood lymphocytes showed increased GPM6A levels. An increase of patient-derived lymphoblastoid cells carrying membrane protrusions supports a functional effect of this duplication. To study the consequences of GPM6A dosage alterations in an intact nervous system, we employed Drosophila melanogaster as a model organism. We found that knockdown of Drosophila M6, the sole member of the PLP family in flies, in the wing, and whole organism causes malformation and lethality, respectively. These phenotypes as well as the protrusions of patient-derived lymphoblastoid cells with increased GPM6A levels can be alleviated by cholesterol supplementation. Notably, overexpression as well as loss of M6 in neurons specifically compromises long-term memory in the courtship conditioning paradigm. Our findings thus indicate a critical role of correct GPM6A/M6 levels for cognitive function and support a role of the GPM6A duplication for the patient's phenotype. Together with other recent findings, this study highlights compromised cholesterol homeostasis as a recurrent feature in cognitive phenotypes.


Subject(s)
Cholesterol/physiology , Cognition Disorders/genetics , Gene Dosage , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Duplication , Humans , In Situ Hybridization, Fluorescence , Locomotion , Phenotype , Real-Time Polymerase Chain Reaction , Sexual Behavior, Animal
18.
PLoS Biol ; 9(1): e1000569, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21245904

ABSTRACT

The epigenetic modification of chromatin structure and its effect on complex neuronal processes like learning and memory is an emerging field in neuroscience. However, little is known about the "writers" of the neuronal epigenome and how they lay down the basis for proper cognition. Here, we have dissected the neuronal function of the Drosophila euchromatin histone methyltransferase (EHMT), a member of a conserved protein family that methylates histone 3 at lysine 9 (H3K9). EHMT is widely expressed in the nervous system and other tissues, yet EHMT mutant flies are viable. Neurodevelopmental and behavioral analyses identified EHMT as a regulator of peripheral dendrite development, larval locomotor behavior, non-associative learning, and courtship memory. The requirement for EHMT in memory was mapped to 7B-Gal4 positive cells, which are, in adult brains, predominantly mushroom body neurons. Moreover, memory was restored by EHMT re-expression during adulthood, indicating that cognitive defects are reversible in EHMT mutants. To uncover the underlying molecular mechanisms, we generated genome-wide H3K9 dimethylation profiles by ChIP-seq. Loss of H3K9 dimethylation in EHMT mutants occurs at 5% of the euchromatic genome and is enriched at the 5' and 3' ends of distinct classes of genes that control neuronal and behavioral processes that are corrupted in EHMT mutants. Our study identifies Drosophila EHMT as a key regulator of cognition that orchestrates an epigenetic program featuring classic learning and memory genes. Our findings are relevant to the pathophysiological mechanisms underlying Kleefstra Syndrome, a severe form of intellectual disability caused by mutations in human EHMT1, and have potential therapeutic implications. Our work thus provides novel insights into the epigenetic control of cognition in health and disease.


Subject(s)
Drosophila/genetics , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Animals , Courtship , DNA/metabolism , Dendrites/metabolism , Drosophila/growth & development , Drosophila/physiology , Euchromatin/chemistry , Euchromatin/metabolism , Gene Expression Profiling , Histone-Lysine N-Methyltransferase/genetics , Humans , Larva , Learning , Locomotion , Memory , Methylation , Nervous System/growth & development , Nervous System/metabolism , Phylogeny , Sequence Deletion
19.
HGG Adv ; 4(1): 100157, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36408368

ABSTRACT

WDR5 is a broadly studied, highly conserved key protein involved in a wide array of biological functions. Among these functions, WDR5 is a part of several protein complexes that affect gene regulation via post-translational modification of histones. We collected data from 11 unrelated individuals with six different rare de novo germline missense variants in WDR5; one identical variant was found in five individuals and another variant in two individuals. All individuals had neurodevelopmental disorders including speech/language delays (n = 11), intellectual disability (n = 9), epilepsy (n = 7), and autism spectrum disorder (n = 4). Additional phenotypic features included abnormal growth parameters (n = 7), heart anomalies (n = 2), and hearing loss (n = 2). Three-dimensional protein structures indicate that all the residues affected by these variants are located at the surface of one side of the WDR5 protein. It is predicted that five out of the six amino acid substitutions disrupt interactions of WDR5 with RbBP5 and/or KMT2A/C, as part of the COMPASS (complex proteins associated with Set1) family complexes. Our experimental approaches in Drosophila melanogaster and human cell lines show normal protein expression, localization, and protein-protein interactions for all tested variants. These results, together with the clustering of variants in a specific region of WDR5 and the absence of truncating variants so far, suggest that dominant-negative or gain-of-function mechanisms might be at play. All in all, we define a neurodevelopmental disorder associated with missense variants in WDR5 and a broad range of features. This finding highlights the important role of genes encoding COMPASS family proteins in neurodevelopmental disorders.


Subject(s)
Autism Spectrum Disorder , Drosophila Proteins , Language Development Disorders , Neurodevelopmental Disorders , Animals , Humans , Autism Spectrum Disorder/genetics , Drosophila melanogaster/genetics , Neurodevelopmental Disorders/genetics , Cluster Analysis , Chromatin , Intracellular Signaling Peptides and Proteins/genetics , Histone-Lysine N-Methyltransferase/genetics , Drosophila Proteins/genetics
20.
Hum Mol Genet ; 18(12): 2257-65, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19336477

ABSTRACT

Pigmented hypertrichotic dermatosis with insulin-dependent diabetes (PHID) syndrome is a recently described autosomal recessive disorder associated with predominantly antibody negative, insulin-dependent diabetes mellitus. In order to identify the genetic basis of PHID and study its relationship with glucose metabolism, we performed homozygosity mapping in five unrelated families followed by candidate gene sequencing. Five loss-of-function mutations were identified in the SLC29A3 gene which encodes a member of a highly conserved protein family that transports nucleosides, nucleobases and nucleoside analogue drugs, hENT3. We show that PHID is allelic with a related syndrome without diabetes mellitus, H syndrome. The interaction of SLC29A3 with insulin signaling pathways was then studied using an established model in Drosophila melanogaster. Ubiquitous knockdown of the Drosophila ortholog of hENT3, dENT1 is lethal under stringent conditions; whereas milder knockdown induced scutellar bristle phenotypes similar to those previously reported in the knockdown of the Drosophila ortholog of the Islet gene. A cellular growth assay showed a reduction of cell size/number which could be rescued or enhanced by manipulation of the Drosophila insulin receptor and its downstream signaling effectors, dPI3K and dAkt. In summary, inactivating mutations in SLC29A3 cause a syndromic form of insulin-dependent diabetes in humans and in Drosophila profoundly affect cell size/number through interactions with the insulin signaling pathway. These data suggest that further investigation of the role of SLC29A3 in glucose metabolism is a priority for diabetes research.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Hypertrichosis/genetics , Insulin/metabolism , Mutation , Nucleoside Transport Proteins/genetics , Signal Transduction , Amino Acid Sequence , Animals , Base Sequence , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Humans , Hypertrichosis/metabolism , Insulin/genetics , Male , Molecular Sequence Data , Nucleoside Transport Proteins/chemistry , Nucleoside Transport Proteins/metabolism , Pedigree , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Sequence Alignment , Skin Pigmentation
SELECTION OF CITATIONS
SEARCH DETAIL