Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nature ; 622(7981): 112-119, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37704727

ABSTRACT

The molecular mechanisms and evolutionary changes accompanying synapse development are still poorly understood1,2. Here we generate a cross-species proteomic map of synapse development in the human, macaque and mouse neocortex. By tracking the changes of more than 1,000 postsynaptic density (PSD) proteins from midgestation to young adulthood, we find that PSD maturation in humans separates into three major phases that are dominated by distinct pathways. Cross-species comparisons reveal that human PSDs mature about two to three times slower than those of other species and contain higher levels of Rho guanine nucleotide exchange factors (RhoGEFs) in the perinatal period. Enhancement of RhoGEF signalling in human neurons delays morphological maturation of dendritic spines and functional maturation of synapses, potentially contributing to the neotenic traits of human brain development. In addition, PSD proteins can be divided into four modules that exert stage- and cell-type-specific functions, possibly explaining their differential associations with cognitive functions and diseases. Our proteomic map of synapse development provides a blueprint for studying the molecular basis and evolutionary changes of synapse maturation.


Subject(s)
Proteomics , Synapses , Adolescent , Animals , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Mice , Young Adult , Cognition/physiology , Dendritic Spines , Gestational Age , Macaca , Neurons/metabolism , Post-Synaptic Density/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Species Specificity , Synapses/metabolism , Synapses/physiology
3.
J Neurochem ; 162(2): 190-206, 2022 07.
Article in English | MEDLINE | ID: mdl-35567753

ABSTRACT

The two members of the cytoplasmic FMR1-interacting protein family, CYFIP1 and CYFIP2, are evolutionarily conserved multifunctional proteins whose defects are associated with distinct types of brain disorders. Even with high sequence homology between CYFIP1 and CYFIP2, several lines of evidence indicate their different functions in the brain; however, the underlying mechanisms remain largely unknown. Here, we performed reciprocal immunoprecipitation experiments using CYFIP1-2 × Myc and CYFIP2-3 × Flag knock-in mice and found that CYFIP1 and CYFIP2 are not significantly co-immunoprecipitated with each other in the knock-in brains compared with negative control wild-type (WT) brains. Moreover, CYFIP1 and CYFIP2 showed different size distributions by size-exclusion chromatography of WT mouse brains. Specifically, mass spectrometry-based analysis of CYFIP1-2 × Myc knock-in brains identified 131 proteins in the CYFIP1 interactome. Comparison of the CYFIP1 interactome with the previously identified brain region- and age-matched CYFIP2 interactome, consisting of 140 proteins, revealed only eight common proteins. Investigations using single-cell RNA-sequencing databases suggested non-neuronal cell- and neuron-enriched expression of Cyfip1 and Cyfip2, respectively. At the protein level, CYFIP1 was detected in both neurons and astrocytes, while CYFIP2 was detected only in neurons, suggesting the predominant expression of CYFIP1 in astrocytes. Bioinformatic characterization of the CYFIP1 interactome, and co-expression analysis of Cyfip1 with astrocytic genes, commonly linked CYFIP1 with focal adhesion proteins. Immunocytochemical analysis and proximity ligation assay suggested partial co-localization of CYFIP1 and focal adhesion proteins in cultured astrocytes. Together, these results suggest a CYFIP1-specific association with astrocytic focal adhesion, which may contribute to the different brain functions and dysfunctions of CYFIP1 and CYFIP2. Cover Image for this issue: https://doi.org/10.1111/jnc.15410.


Subject(s)
Adaptor Proteins, Signal Transducing , Astrocytes , Focal Adhesions , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Astrocytes/metabolism , Carrier Proteins/genetics , Focal Adhesions/metabolism , Mice
4.
Clin Exp Hypertens ; 44(5): 397-402, 2022 Jul 04.
Article in English | MEDLINE | ID: mdl-35315303

ABSTRACT

OBJECTIVE: To investigate the effects of sacubitril/valsartan (S/V) on cardiopulmonary function and blood pressure response to exercise during hospitalization in patients with acute myocardial infarction (AMI) based on the cardiopulmonary exercise test (CPET). METHODS: A total of 265 AMI patients were treated with either perindopril or S/V within 24 hours of admission. CPET was completed for all patients before discharge. There were 182 cases in the perindopril group and 83 cases in the S/V group. RESULTS: The proportion of exercise oscillatory ventilation (EOV) was higher in the S/V group than in the perindopril group (10.8% vs 1.6%, X2 = 11.148, P = .001). The resting heart rate (HR), resting diastolic blood pressure (DBP), and warm-up DBP were lower in the S/V group than in the perindopril group (P < .05). The resting systolic blood pressure (SBP) was 9.0 mmHg lower (115.7 ± 17.5 vs 106.7 ± 15.0, P < .001), the SBP during warm-up was 9.5 mmHg lower (124.8 ± 23.7 vs 115.3 ± 22.5,P = .002), the SBP at the anaerobic threshold (AT) was 10.5 mmHg lower (135.3 ± 24.8 vs 127.1 ± 25.1,P = .021),the SBP at max watts was 11.5 mmHg lower (148.9 ± 26.4 vs 137.4 ± 26.4,P = .001), and the SBP during one-minute recovery was 12.3 mmHg lower (146.5 ± 27.1 vs 134.2 ± 24.4, P = .001)in the S/V group than in the perindopril group. The S/V group had a higher oxygen ventilation equivalent and carbon dioxide ventilation equivalent (VE/VCO2) at AT and a lower oxygen uptake-work rate relationship during max watts (P < .05). The differences in the oxygen pulse, stroke volume, peak oxygen uptake (VO2 peak), and VE/VCO2 slope were not statistically significant between the two groups. CONCLUSION: Treatment with S/V was able to reduce the exercise blood pressure in patients with AMI during hospitalization, but did not significantly improve the VO2 peak, VE/VCO2 slope, or exercise tolerance.


Subject(s)
Heart Failure , Myocardial Infarction , Aminobutyrates , Biphenyl Compounds , Blood Pressure , Exercise Test , Exercise Tolerance , Hospitalization , Humans , Myocardial Infarction/drug therapy , Oxygen , Oxygen Consumption , Perindopril , Valsartan/therapeutic use
5.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35074918

ABSTRACT

MeCP2 is associated with Rett syndrome (RTT), MECP2 duplication syndrome, and a number of conditions with isolated features of these diseases, including autism, intellectual disability, and motor dysfunction. MeCP2 is known to broadly bind methylated DNA, but the precise molecular mechanism driving disease pathogenesis remains to be determined. Using proximity-dependent biotinylation (BioID), we identified a transcription factor 20 (TCF20) complex that interacts with MeCP2 at the chromatin interface. Importantly, RTT-causing mutations in MECP2 disrupt this interaction. TCF20 and MeCP2 are highly coexpressed in neurons and coregulate the expression of key neuronal genes. Reducing Tcf20 partially rescued the behavioral deficits caused by MECP2 overexpression, demonstrating a functional relationship between MeCP2 and TCF20 in MECP2 duplication syndrome pathogenesis. We identified a patient exhibiting RTT-like neurological features with a missense mutation in the PHF14 subunit of the TCF20 complex that abolishes the MeCP2-PHF14-TCF20 interaction. Our data demonstrate the critical role of the MeCP2-TCF20 complex for brain function.


Subject(s)
Methyl-CpG-Binding Protein 2/metabolism , Multiprotein Complexes/metabolism , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Transcription Factors/metabolism , Alleles , Animals , Biomarkers , Brain/metabolism , Disease Models, Animal , Disease Susceptibility , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Mutation , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Synapses/metabolism , Transcription Factors/genetics
6.
Am J Hum Genet ; 108(3): 502-516, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33596411

ABSTRACT

Deletion 1p36 (del1p36) syndrome is the most common human disorder resulting from a terminal autosomal deletion. This condition is molecularly and clinically heterogeneous. Deletions involving two non-overlapping regions, known as the distal (telomeric) and proximal (centromeric) critical regions, are sufficient to cause the majority of the recurrent clinical features, although with different facial features and dysmorphisms. SPEN encodes a transcriptional repressor commonly deleted in proximal del1p36 syndrome and is located centromeric to the proximal 1p36 critical region. Here, we used clinical data from 34 individuals with truncating variants in SPEN to define a neurodevelopmental disorder presenting with features that overlap considerably with those of proximal del1p36 syndrome. The clinical profile of this disease includes developmental delay/intellectual disability, autism spectrum disorder, anxiety, aggressive behavior, attention deficit disorder, hypotonia, brain and spine anomalies, congenital heart defects, high/narrow palate, facial dysmorphisms, and obesity/increased BMI, especially in females. SPEN also emerges as a relevant gene for del1p36 syndrome by co-expression analyses. Finally, we show that haploinsufficiency of SPEN is associated with a distinctive DNA methylation episignature of the X chromosome in affected females, providing further evidence of a specific contribution of the protein to the epigenetic control of this chromosome, and a paradigm of an X chromosome-specific episignature that classifies syndromic traits. We conclude that SPEN is required for multiple developmental processes and SPEN haploinsufficiency is a major contributor to a disorder associated with deletions centromeric to the previously established 1p36 critical regions.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, X/genetics , DNA-Binding Proteins/genetics , RNA-Binding Proteins/genetics , Adolescent , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Child , Child, Preschool , Chromosome Deletion , Chromosome Disorders/physiopathology , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Female , Haploinsufficiency/genetics , Humans , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Male , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/physiopathology , Phenotype , Young Adult
7.
Genes Dev ; 34(17-18): 1147-1160, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32763910

ABSTRACT

Identifying modifiers of dosage-sensitive genes involved in neurodegenerative disorders is imperative to discover novel genetic risk factors and potential therapeutic entry points. In this study, we focus on Ataxin-1 (ATXN1), a dosage-sensitive gene involved in the neurodegenerative disease spinocerebellar ataxia type 1 (SCA1). While the precise maintenance of ATXN1 levels is essential to prevent disease, the mechanisms that regulate ATXN1 expression remain largely unknown. We demonstrate that ATXN1's unusually long 5' untranslated region (5' UTR) negatively regulates its expression via posttranscriptional mechanisms. Based on recent reports that microRNAs (miRNAs) can interact with both 3' and 5' UTRs to regulate their target genes, we identify miR760 as a negative regulator that binds to a conserved site in ATXN1's 5' UTR to induce RNA degradation and translational inhibition. We found that delivery of Adeno-associated virus (AAV)-expressing miR760 in the cerebellum reduces ATXN1 levels in vivo and mitigates motor coordination deficits in a mouse model of SCA1. These findings provide new insights into the regulation of ATXN1 levels, present additional evidence for miRNA-mediated gene regulation via 5' UTR binding, and raise the possibility that noncoding mutations in the ATXN1 locus may act as risk factors for yet to be discovered progressive ataxias.


Subject(s)
5' Untranslated Regions/genetics , Ataxin-1/genetics , Gene Expression Regulation/genetics , MicroRNAs/metabolism , Spinocerebellar Ataxias/genetics , Animals , Ataxin-1/metabolism , Cell Line , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Mutation , Risk Factors , Spinocerebellar Ataxias/physiopathology
8.
Ann Neurol ; 88(3): 526-543, 2020 09.
Article in English | MEDLINE | ID: mdl-32562430

ABSTRACT

OBJECTIVE: Genetic variants of the cytoplasmic FMR1-interacting protein 2 (CYFIP2) encoding an actin-regulatory protein are associated with brain disorders, including intellectual disability and epilepsy. However, specific in vivo neuronal defects and potential treatments for CYFIP2-associated brain disorders remain largely unknown. Here, we characterized Cyfip2 heterozygous (Cyfip2+/- ) mice to understand their neurobehavioral phenotypes and the underlying pathological mechanisms. Furthermore, we examined a potential treatment for such phenotypes of the Cyfip2+/- mice and specified a neuronal function mediating its efficacy. METHODS: We performed behavioral analyses of Cyfip2+/- mice. We combined molecular, ultrastructural, and in vitro and in vivo electrophysiological analyses of Cyfip2+/- prefrontal neurons. We also selectively reduced CYFIP2 in the prefrontal cortex (PFC) of mice with virus injections. RESULTS: Adult Cyfip2+/- mice exhibited lithium-responsive abnormal behaviors. We found increased filamentous actin, enlarged dendritic spines, and enhanced excitatory synaptic transmission and excitability in the adult Cyfip2+/- PFC that was restricted to layer 5 (L5) neurons. Consistently, adult Cyfip2+/- mice showed increased seizure susceptibility and auditory steady-state responses from the cortical electroencephalographic recordings. Among the identified prefrontal defects, lithium selectively normalized the hyperexcitability of Cyfip2+/- L5 neurons. RNA sequencing revealed reduced expression of potassium channel genes in the adult Cyfip2+/- PFC. Virus-mediated reduction of CYFIP2 in the PFC was sufficient to induce L5 hyperexcitability and lithium-responsive abnormal behavior. INTERPRETATION: These results suggest that L5-specific prefrontal dysfunction, especially hyperexcitability, underlies both the pathophysiology and the lithium-mediated amelioration of neurobehavioral phenotypes in adult Cyfip2+/- mice, which can be implicated in CYFIP2-associated brain disorders. ANN NEUROL 2020;88:526-543.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Lithium Compounds/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Seizures/genetics , Animals , Behavior, Animal/drug effects , Haploinsufficiency , Mice , Mice, Mutant Strains , Neurons/drug effects , Neurons/pathology , Prefrontal Cortex/pathology , Seizures/physiopathology
9.
Genome Res ; 30(6): 835-848, 2020 06.
Article in English | MEDLINE | ID: mdl-32554779

ABSTRACT

A large number of genes have been implicated in neurodevelopmental disorders (NDDs), but their contributions to NDD pathology are difficult to decipher without understanding their diverse roles in different brain cell types. Here, we integrated NDD genetics with single-cell RNA sequencing data to assess coexpression enrichment patterns of various NDD gene sets. We identified midfetal cortical neural progenitor cell development-more specifically, the ventricular radial glia-to-intermediate progenitor cell transition at gestational week 10-as a key point of convergence in autism spectrum disorder (ASD) and epilepsy. Integrated Gene Ontology-based analysis further revealed that ASD genes activate neural differentiation and inhibit cell cycle during the transition, whereas epilepsy genes function as downstream effectors in the same processes, offering one possible explanation for the high comorbidity rate of the two disorders. This approach provides a framework for investigating the cell-type-specific pathophysiology of NDDs.


Subject(s)
Cell Differentiation/genetics , Disease Susceptibility , Gene Expression Profiling , Neural Stem Cells/metabolism , Neurodevelopmental Disorders/etiology , Single-Cell Analysis , Transcriptome , Biomarkers , Brain/metabolism , Brain/physiopathology , Computational Biology/methods , Epilepsy/etiology , Gene Expression Profiling/methods , Gene Ontology , Humans , Interneurons/cytology , Interneurons/metabolism , Neural Stem Cells/cytology , Neurodevelopmental Disorders/diagnosis , Neurons/cytology , Neurons/metabolism , Single-Cell Analysis/methods
10.
Hum Mutat ; 41(5): 921-925, 2020 05.
Article in English | MEDLINE | ID: mdl-31999386

ABSTRACT

The bromodomain adjacent to zinc finger 2B gene (BAZ2B) encodes a protein involved in chromatin remodeling. Loss of BAZ2B function has been postulated to cause neurodevelopmental disorders. To determine whether BAZ2B deficiency is likely to contribute to the pathogenesis of these disorders, we performed bioinformatics analyses that demonstrated a high level of functional convergence during fetal cortical development between BAZ2B and genes known to cause autism spectrum disorder (ASD) and neurodevelopmental disorder. We also found an excess of de novo BAZ2B loss-of-function variants in exome sequencing data from previously published cohorts of individuals with neurodevelopmental disorders. We subsequently identified seven additional individuals with heterozygous deletions, stop-gain, or de novo missense variants affecting BAZ2B. All of these individuals have developmental delay (DD), intellectual disability (ID), and/or ASD. Taken together, our findings suggest that haploinsufficiency of BAZ2B causes a neurodevelopmental disorder, whose cardinal features include DD, ID, and ASD.


Subject(s)
Autism Spectrum Disorder/genetics , Genetic Predisposition to Disease , Haploinsufficiency , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Transcription Factors, General/genetics , Alleles , Amino Acid Substitution , Autism Spectrum Disorder/diagnosis , Gene Expression , Genetic Association Studies , Genotype , Humans , Intellectual Disability/diagnosis , Neurodevelopmental Disorders/diagnosis , Sequence Deletion
11.
Mol Psychiatry ; 25(10): 2504-2516, 2020 10.
Article in English | MEDLINE | ID: mdl-30696942

ABSTRACT

Neurons are sensitive to changes in the dosage of many genes, especially those regulating synaptic functions. Haploinsufficiency of SHANK3 causes Phelan-McDermid syndrome and autism, whereas duplication of the same gene leads to SHANK3 duplication syndrome, a disorder characterized by neuropsychiatric phenotypes including hyperactivity and bipolar disorder as well as epilepsy. We recently demonstrated the functional modularity of Shank3, which suggests that normalizing levels of Shank3 itself might be more fruitful than correcting pathways that function downstream of it for treatment of disorders caused by alterations in SHANK3 dosage. To identify upstream regulators of Shank3 abundance, we performed a kinome-wide siRNA screen and identified multiple kinases that potentially regulate Shank3 protein stability. Interestingly, we discovered that several kinases in the MEK/ERK2 pathway destabilize Shank3 and that genetic deletion and pharmacological inhibition of ERK2 increases Shank3 abundance in vivo. Mechanistically, we show that ERK2 binds Shank3 and phosphorylates it at three residues to promote its poly-ubiquitination-dependent degradation. Altogether, our findings uncover a druggable pathway as a potential therapeutic target for disorders with reduced SHANK3 dosage, provide a rich resource for studying Shank3 regulation, and demonstrate the feasibility of this approach for identifying regulators of dosage-sensitive genes.


Subject(s)
Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Nerve Tissue Proteins/metabolism , Protein Stability , RNA Interference , Animals , Cell Line, Tumor , Chromosome Disorders/genetics , Female , Gene Deletion , Haploinsufficiency , Humans , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phosphorylation/drug effects , Protein Stability/drug effects
12.
Mol Psychiatry ; 25(10): 2534-2555, 2020 10.
Article in English | MEDLINE | ID: mdl-30610205

ABSTRACT

Genome sequencing has revealed an increasing number of genetic variations that are associated with neuropsychiatric disorders. Frequently, studies limit their focus to likely gene-disrupting mutations because they are relatively easy to interpret. Missense variants, instead, have often been undervalued. However, some missense variants can be informative for developing a more profound understanding of disease pathogenesis and ultimately targeted therapies. Here we present an example of this by studying a missense variant in a well-known autism spectrum disorder (ASD) causing gene SHANK3. We analyzed Shank3's in vivo phosphorylation profile and identified S685 as one phosphorylation site where one ASD-linked variant has been reported. Detailed analysis of this variant revealed a novel function of Shank3 in recruiting Abelson interactor 1 (ABI1) and the WAVE complex to the post-synaptic density (PSD), which is critical for synapse and dendritic spine development. This function was found to be independent of Shank3's other functions such as binding to GKAP and Homer. Introduction of this human ASD mutation into mice resulted in a small subset of phenotypes seen previously in constitutive Shank3 knockout mice, including increased allogrooming, increased social dominance, and reduced pup USV. Together, these findings demonstrate the modularity of Shank3 function in vivo. This modularity further indicates that there is more than one independent pathogenic pathway downstream of Shank3 and correcting a single downstream pathway is unlikely to be sufficient for clear clinical improvement. In addition, this study illustrates the value of deep biological analysis of select missense mutations in elucidating the pathogenesis of neuropsychiatric phenotypes.


Subject(s)
Autism Spectrum Disorder/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation, Missense/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autistic Disorder/genetics , Cytoskeletal Proteins/metabolism , Female , Humans , Male , Mice , Post-Synaptic Density/metabolism , Rats
13.
Cell Syst ; 7(1): 28-40.e4, 2018 07 25.
Article in English | MEDLINE | ID: mdl-29936182

ABSTRACT

Discriminating transcriptional changes that drive disease pathogenesis from nonpathogenic and compensatory responses is a daunting challenge. This is particularly true for neurodegenerative diseases, which affect the expression of thousands of genes in different brain regions at different disease stages. Here we integrate functional testing and network approaches to analyze previously reported transcriptional alterations in the brains of Huntington disease (HD) patients. We selected 312 genes whose expression is dysregulated both in HD patients and in HD mice and then replicated and/or antagonized each alteration in a Drosophila HD model. High-throughput behavioral testing in this model and controls revealed that transcriptional changes in synaptic biology and calcium signaling are compensatory, whereas alterations involving the actin cytoskeleton and inflammation drive disease. Knockdown of disease-driving genes in HD patient-derived cells lowered mutant Huntingtin levels and activated macroautophagy, suggesting a mechanism for mitigating pathogenesis. Our multilayered approach can thus untangle the wealth of information generated by transcriptomics and identify early therapeutic intervention points.


Subject(s)
High-Throughput Screening Assays/methods , Huntington Disease/genetics , Animals , Brain/metabolism , Cell Line , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Fibroblasts/metabolism , Gene Expression Profiling/methods , Humans , Huntington Disease/physiopathology , Induced Pluripotent Stem Cells , Male , Transcriptome/genetics
14.
Am J Hum Genet ; 102(2): 296-308, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395075

ABSTRACT

15q13.3 microdeletion syndrome is characterized by a wide spectrum of neurodevelopmental disorders, including developmental delay, intellectual disability, epilepsy, language impairment, abnormal behaviors, neuropsychiatric disorders, and hypotonia. This syndrome is caused by a deletion on chromosome 15q, which typically encompasses six genes. Here, through studies on OTU deubiquitinase 7A (Otud7a) knockout mice, we identify OTUD7A as a critical gene responsible for many of the cardinal phenotypes associated with 15q13.3 microdeletion syndrome. Otud7a-null mice show reduced body weight, developmental delay, abnormal electroencephalography patterns and seizures, reduced ultrasonic vocalizations, decreased grip strength, impaired motor learning/motor coordination, and reduced acoustic startle. We show that OTUD7A localizes to dendritic spines and that Otud7a-null mice have decreased dendritic spine density compared to their wild-type littermates. Furthermore, frequency of miniature excitatory postsynaptic currents (mEPSCs) is reduced in the frontal cortex of Otud7a-null mice, suggesting a role of Otud7a in regulation of dendritic spine density and glutamatergic synaptic transmission. Taken together, our results suggest decreased OTUD7A dosage as a major contributor to the neurodevelopmental phenotypes associated with 15q13.3 microdeletion syndrome, through the misregulation of dendritic spine density and activity.


Subject(s)
Chromosome Disorders/enzymology , Chromosome Disorders/genetics , Deubiquitinating Enzymes/genetics , Endopeptidases/genetics , Intellectual Disability/enzymology , Intellectual Disability/genetics , Seizures/enzymology , Seizures/genetics , Action Potentials , Animals , Base Sequence , Behavior, Animal , Chromosome Deletion , Chromosomes, Human, Pair 15/enzymology , Chromosomes, Human, Pair 15/genetics , Dendritic Spines/metabolism , Disease Models, Animal , Electroencephalography , Endopeptidases/deficiency , Epilepsy/enzymology , Epilepsy/genetics , Epilepsy/physiopathology , Female , Homozygote , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Synapses/metabolism
15.
Exp Neurobiol ; 25(6): 296-306, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28035180

ABSTRACT

Bipolar disorder (BD), characterized by recurrent mood swings between depression and mania, is a highly heritable and devastating mental illness with poorly defined pathophysiology. Recent genome-wide molecular genetic studies have identified several protein-coding genes and microRNAs (miRNAs) significantly associated with BD. Notably, some of the proteins expressed from BD-associated genes function in neuronal synapses, suggesting that abnormalities in synaptic function could be one of the key pathogenic mechanisms of BD. In contrast, however, the role of BD-associated miRNAs in disease pathogenesis remains largely unknown, mainly because of a lack of understanding about their target mRNAs and pathways in neurons. To address this problem, in this study, we focused on a recently identified BD-associated but uncharacterized miRNA, miR-1908-5p. We identified and validated its novel target genes including DLGAP4, GRIN1, STX1A, CLSTN1 and GRM4, which all function in neuronal glutamatergic synapses. Moreover, bioinformatic analyses of human brain expression profiles revealed that the expression levels of miR-1908-5p and its synaptic target genes show an inverse-correlation in many brain regions. In our preliminary experiments, the expression of miR-1908-5p was increased after chronic treatment with valproate but not lithium in control human neural progenitor cells. In contrast, it was decreased by valproate in neural progenitor cells derived from dermal fibroblasts of a BD subject. Together, our results provide new insights into the potential role of miR-1908-5p in the pathogenesis of BD and also propose a hypothesis that neuronal synapses could be a key converging pathway of some BD-associated protein-coding genes and miRNAs.

16.
Mol Brain ; 8(1): 74, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26572867

ABSTRACT

BACKGROUND: Proper neuronal function requires tight control of gene dosage, and failure of this process underlies the pathogenesis of multiple neuropsychiatric disorders. The SHANK3 gene encoding core scaffolding proteins at glutamatergic postsynapse is a typical dosage-sensitive gene, both deletions and duplications of which are associated with Phelan-McDermid syndrome, autism spectrum disorders, bipolar disorder, intellectual disability, or schizophrenia. However, the regulatory mechanism of SHANK3 expression in neurons itself is poorly understood. RESULTS: Here we show post-transcriptional regulation of SHANK3 expression by three microRNAs (miRNAs), miR-7, miR-34a, and miR-504. Notably, the expression profiles of these miRNAs were previously shown to be altered in some neuropsychiatric disorders which are also associated with SHANK3 dosage changes. These miRNAs regulated the expression of SHANK3 and other genes encoding actin-related proteins that interact with Shank3, through direct binding sites in the 3' untranslated region (UTR). Moreover, overexpression or inhibition of miR-7 and miR-504 affected the dendritic spines of the cultured hippocampal neurons in a Shank3-dependent manner. We further characterized miR-504 as it showed the most significant effect on both SHANK3 expression and dendritic spines among the three miRNAs. Lentivirus-mediated overexpression of miR-504, which mimics its reported expression change in postmortem brain tissues of bipolar disorder, decreased endogenous Shank3 protein in cultured hippocampal neurons. We also revealed that miR-504 is expressed in the cortical and hippocampal regions of human and mouse brains. CONCLUSIONS: Our study provides new insight into the miRNA-mediated regulation of SHANK3 expression, and its potential implication in multiple neuropsychiatric disorders associated with altered SHANK3 and miRNA expression profiles.


Subject(s)
Mental Disorders/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/genetics , Transcription, Genetic , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cells, Cultured , Dendritic Spines/metabolism , HEK293 Cells , Hippocampus/metabolism , Humans , Lentivirus/metabolism , Luciferases/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Microfilament Proteins , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Protein Interaction Mapping , Reproducibility of Results
17.
Hum Pathol ; 46(12): 1859-71, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26433703

ABSTRACT

FOXG1 is a transcription factor that interacts with multiple signaling pathways and modulates neuronal differentiation in the telencephalon. Dysregulation of FOXG1 expression has been previously reported in medulloblastoma. In this study, we demonstrate a regional specific expression of FOXG1 and its colocalization with Nestin expression in the premigratory mitotically active (outer) layer of the external granular layer of the cerebellum. An inverse expression of the granular precursor cell markers, Math1 and Musashi1, in the inner nonmitotic migratory layer of the external granular layer and in the internal granular layer was observed. Furthermore, modulation of FOXG1 in the medulloblastoma cell line, DAOY, was associated with the induction of neuronal differentiation markers and significant changes in multiple signaling pathways regulating cell proliferation, differentiation, survival, and apoptosis. Additionally, we observed enhanced survival in intracerebellar mice xenografts injected with DAOY cells bearing shFOXG1 constructs versus shLuciferase construct. Overall, these findings suggest that down-modulation of FOXG1 is a prerequisite for the onset of neuronal differentiation during cerebellar development and that a decrease of FOXG1 in medulloblastoma cells offers a survival advantage in mice. We propose that the disruption of signaling pathways that promote mature neuronal differentiation by overexpressed FOXG1 is a contributing event in the neoplastic transformation of cerebellar stem cells.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cerebellar Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Medulloblastoma/pathology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/pathology , Animals , Blotting, Western , Cell Differentiation/physiology , Cell Transformation, Neoplastic/pathology , Cerebellum/embryology , Disease Models, Animal , Heterografts , Humans , Immunohistochemistry , Mice , Oligonucleotide Array Sequence Analysis , Phenotype , Polymerase Chain Reaction , RNA, Small Interfering , Transduction, Genetic
18.
Bioinformatics ; 30(10): 1456-63, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24463180

ABSTRACT

MOTIVATION: Combinatorial therapies play increasingly important roles in combating complex diseases. Owing to the huge cost associated with experimental methods in identifying optimal drug combinations, computational approaches can provide a guide to limit the search space and reduce cost. However, few computational approaches have been developed for this purpose, and thus there is a great need of new algorithms for drug combination prediction. RESULTS: Here we proposed to formulate the optimal combinatorial therapy problem into two complementary mathematical algorithms, Balanced Target Set Cover (BTSC) and Minimum Off-Target Set Cover (MOTSC). Given a disease gene set, BTSC seeks a balanced solution that maximizes the coverage on the disease genes and minimizes the off-target hits at the same time. MOTSC seeks a full coverage on the disease gene set while minimizing the off-target set. Through simulation, both BTSC and MOTSC demonstrated a much faster running time over exhaustive search with the same accuracy. When applied to real disease gene sets, our algorithms not only identified known drug combinations, but also predicted novel drug combinations that are worth further testing. In addition, we developed a web-based tool to allow users to iteratively search for optimal drug combinations given a user-defined gene set. AVAILABILITY: Our tool is freely available for noncommercial use at http://www.drug.liuzlab.org/. CONTACT: zhandong.liu@bcm.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Programming, Linear , Algorithms , Databases, Factual , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Drug Combinations , Gene Regulatory Networks , Humans , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , Software Design
19.
BMC Bioinformatics ; 14: 89, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23497278

ABSTRACT

BACKGROUND: Cellular heterogeneity is present in almost all gene expression profiles. However, transcriptome analysis of tissue specimens often ignores the cellular heterogeneity present in these samples. Standard deconvolution algorithms require prior knowledge of the cell type frequencies within a tissue or their in vitro expression profiles. Furthermore, these algorithms tend to report biased estimations. RESULTS: Here, we describe a Digital Sorting Algorithm (DSA) for extracting cell-type specific gene expression profiles from mixed tissue samples that is unbiased and does not require prior knowledge of cell type frequencies. CONCLUSIONS: The results suggest that DSA is a specific and sensitivity algorithm in gene expression profile deconvolution and will be useful in studying individual cell types of complex tissues.


Subject(s)
Algorithms , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis
20.
Genes Dev ; 27(5): 485-90, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23431031

ABSTRACT

Proper neurological function in humans requires precise control of levels of the epigenetic regulator methyl CpG-binding protein 2 (MeCP2). MeCP2 protein levels are low in fetal brains, where the predominant MECP2 transcripts have an unusually long 3' untranslated region (UTR). Here, we show that miR-483-5p, an intragenic microRNA of the imprinted IGF2, regulates MeCP2 levels through a human-specific binding site in the MECP2 long 3' UTR. We demonstrate the inverse correlation of miR-483-5p and MeCP2 levels in developing human brains and fibroblasts from Beckwith-Wiedemann syndrome patients. Importantly, expression of miR-483-5p rescues abnormal dendritic spine phenotype of neurons overexpressing human MeCP2. In addition, miR-483-5p modulates the levels of proteins of the MeCP2-interacting corepressor complexes, including HDAC4 and TBL1X. These data provide insight into the role of miR-483-5p in regulating the levels of MeCP2 and interacting proteins during human fetal development.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , MicroRNAs/metabolism , Neurons/metabolism , Binding Sites , Brain/embryology , Brain/physiopathology , Cell Line , Fetus/embryology , Fetus/metabolism , Fetus/physiopathology , Genomic Imprinting , Humans , Neurons/pathology , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...