Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Am J Hum Biol ; : e24125, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38940191

ABSTRACT

INTRODUCTION: Black people had the highest prevalence of Alzheimer's disease and related dementias (ADRD) of any racial/ethnic group in the United States (US) as of 2020. As racial disparities in the prevalence of ADRD are being investigated, more evidence is necessary to determine the pathways and mechanisms that either slow ADRD progression or improve quality of life for those affected. Religion/spirituality (R/S) has been shown to affect health outcomes but has rarely been studied as a possible pathway for reducing ADRD risk. Crucially, Black people also report higher levels of R/S than other racial/ethnic groups in the United States. This research asks if R/S affects ADRD risk among Black adults and if any effects persist after controlling for hypertension. METHODS: We conducted a secondary data analysis drawing from the Health and Retirement Study (HRS), a nationally representative longitudinal dataset with an oversampling of Black adults. RESULTS: We used logistic regression analysis to demonstrate how R/S has an ameliorating impact on ADRD risk among Black people, even after controlling for hypertension. Those who never attended religious services had 2.37 higher odds of being diagnosed with ADRD than those who attended more than once a week. Further, as R/S attendance increased, ADRD risk decreased linearly. CONCLUSION: These findings demonstrate the importance that existing cultural networks (e.g., R/S) can have for reducing ADRD burden for Black people and has important implications for the role of R/S in shaping ADRD symptomatology.

2.
bioRxiv ; 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38585858

ABSTRACT

Torpor and hibernation are extreme physiological adaptations of homeotherms associated with pro-longevity effects. Yet the underlying mechanisms of how torpor affects aging, and whether hypothermic and hypometabolic states can be induced to slow aging and increase health span, remain unknown. We demonstrate that the activity of a spatially defined neuronal population in the avMLPA, which has previously been identified as a torpor-regulating brain region, is sufficient to induce a torpor like state (TLS) in mice. Prolonged induction of TLS slows epigenetic aging across multiple tissues and improves health span. We isolate the effects of decreased metabolic rate, long-term caloric restriction, and decreased core body temperature (Tb) on blood epigenetic aging and find that the pro-longevity effect of torpor-like states is mediated by decreased Tb. Taken together, our findings provide novel mechanistic insight into the pro-longevity effects of torpor and hibernation and support the growing body of evidence that Tb is an important mediator of aging processes.

3.
Clin Gerontol ; : 1-14, 2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38367001

ABSTRACT

OBJECTIVES: COVID-19 escalated stress within family/neighborhood (local) and national/cultural (global) levels. However, the impact of socioecological levels of stress on pandemic emotion regulation remains largely unexplored. METHODS: Thirty older adults from the Northeast US (63-92 years) reported on pandemic stress and emotion regulation in semi-structured interviews. Responses were coded into socioecological sources of local and global stress, and associated use of cognitive emotion regulation strategies from the Cognitive Emotion Regulation Questionnaire was explored. RESULTS: Older adults experienced significant distress at global levels, and perception of lacking top-down safety governance may have exacerbated local distress of engaging in daily activities during the COVID-19 pandemic. Participants endorsed coping with local stressors via perspective-taking, acceptance, and other adaptive strategies, while global sources of stress were associated with greater use of maladaptive strategies, including other-blame and rumination. CONCLUSION: Quantitative assessments may underestimate significant older adult distress and maladaptive coping toward global stressors. Findings should be replicated with more diverse populations beyond the COVID-19 context.

4.
Gerontologist ; 64(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37061845

ABSTRACT

As cases of Alzheimer's disease and related dementias (ADRD) increase worldwide, research design has placed additional emphasis on social and behavioral factors that affect ADRD symptomatology and quality of life. Despite this, few studies have incorporated people living with ADRD as research partners. We propose 5 community-engaged recommendations for incorporating people living with ADRD into future research as full collaborators. The proposed recommendations center the experiences of people living with ADRD as crucial contributions to scientific inquiry. The guidelines are based on experiences at a 2-day "Empowering Partnerships" workshop in 2019; post workshop activity continued through 2021 with ongoing collaborations, analysis, and reflective practice. The workshop and subsequent conversations engaged a network of people living with ADRD, informal carepartners, and researchers to collectively build their capacities to partner in all aspects of person-centered research. To empower people living with ADRD as research partners, we recommend that research teams (a) create a flexible schedule of communication and/or meetings to accommodate a wide range of ADRD symptoms, (b) generate team-specific communication strategies/guidelines, (c) incorporate lived experiences of people living with ADRD into research protocols, (d) involve people living with ADRD in all aspects of a project, beginning in the developmental stages, and (e) incorporate skilled facilitators to facilitate communication between stakeholder groups. This multi-vocal approach to research will diversify ADRD research and ensure that projects align with the priorities and capacities of principal stakeholders by incorporating individuals with a wide range of cognitive capabilities that more fully represent the diversity of ADRD experiences.


Subject(s)
Alzheimer Disease , Quality of Life , Humans , Altruism , Cognitive Reflection , Power, Psychological
5.
Arthrosc Tech ; 12(9): e1479-e1485, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37780666

ABSTRACT

The hip capsule has been recognized as a vital structure in the stability and proper function of the hip. Preserving its integrity during arthroscopic surgery is one of the utmost important principles in hip preservation surgery. When capsular deficiency is present, capsular reconstruction may be indicated to restore stability and proper hip mechanics. In this technical note, we introduce a simple and reproducible shuttle method technique for hip capsular reconstruction using a dermal allograft.

6.
Proc Natl Acad Sci U S A ; 120(44): e2310344120, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37871205

ABSTRACT

Mutations in MECP2 give rise to Rett syndrome (RTT), an X-linked neurodevelopmental disorder that results in broad cognitive impairments in females. While the exact etiology of RTT symptoms remains unknown, one possible explanation for its clinical presentation is that loss of MECP2 causes miswiring of neural circuits due to defects in the brain's capacity to respond to changes in neuronal activity and sensory experience. Here, we show that MeCP2 is phosphorylated at four residues in the mouse brain (S86, S274, T308, and S421) in response to neuronal activity, and we generate a quadruple knock-in (QKI) mouse line in which all four activity-dependent sites are mutated to alanines to prevent phosphorylation. QKI mice do not display overt RTT phenotypes or detectable gene expression changes in two brain regions. However, electrophysiological recordings from the retinogeniculate synapse of QKI mice reveal that while synapse elimination is initially normal at P14, it is significantly compromised at P20. Notably, this phenotype is distinct from the synapse refinement defect previously reported for Mecp2 null mice, where synapses initially refine but then regress after the third postnatal week. We thus propose a model in which activity-induced phosphorylation of MeCP2 is critical for the proper timing of retinogeniculate synapse maturation specifically during the early postnatal period.


Subject(s)
Methyl-CpG-Binding Protein 2 , Rett Syndrome , Female , Mice , Animals , Phosphorylation , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Rett Syndrome/genetics , Rett Syndrome/metabolism , Brain/metabolism , Synapses/metabolism , Neurons/metabolism , Mice, Knockout , Disease Models, Animal
7.
bioRxiv ; 2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37461668

ABSTRACT

Mutations in MECP2 give rise to Rett syndrome (RTT), an X-linked neurodevelopmental disorder that results in broad cognitive impairments in females. While the exact etiology of RTT symptoms remains unknown, one possible explanation for its clinical presentation is that loss of MeCP2 causes miswiring of neural circuits due to defects in the brain's capacity to respond to changes in neuronal activity and sensory experience. Here we show that MeCP2 is phosphorylated at four residues in the brain (S86, S274, T308, and S421) in response to neuronal activity, and we generate a quadruple knock-in (QKI) mouse line in which all four activity-dependent sites are mutated to alanines to prevent phosphorylation. QKI mice do not display overt RTT phenotypes or detectable gene expression changes in two brain regions. However, electrophysiological recordings from the retinogeniculate synapse of QKI mice reveal that while synapse elimination is initially normal at P14, it is significantly compromised at P20. Notably, this phenotype is distinct from that previously reported for Mecp2 null mice, where synapses initially refine but then regress after the third postnatal week. We thus propose a model in which activity-induced phosphorylation of MeCP2 is critical for the proper timing of retinogeniculate synapse maturation specifically during the early postnatal period. SIGNIFICANCE STATEMENT: Rett syndrome (RTT) is an X-linked neurodevelopmental disorder that predominantly affects girls. RTT is caused by loss of function mutations in a single gene MeCP2. Girls with RTT develop normally during their first year of life, but then experience neurological abnormalities including breathing and movement difficulties, loss of speech, and seizures. This study investigates the function of the MeCP2 protein in the brain, and how MeCP2 activity is modulated by sensory experience in early life. Evidence is presented that sensory experience affects MeCP2 function, and that this is required for synaptic pruning in the brain. These findings provide insight into MeCP2 function, and clues as to what goes awry in the brain when the function of MeCP2 is disrupted.

8.
Nature ; 614(7949): 732-741, 2023 02.
Article in English | MEDLINE | ID: mdl-36792830

ABSTRACT

Neuronal activity is crucial for adaptive circuit remodelling but poses an inherent risk to the stability of the genome across the long lifespan of postmitotic neurons1-5. Whether neurons have acquired specialized genome protection mechanisms that enable them to withstand decades of potentially damaging stimuli during periods of heightened activity is unknown. Here we identify an activity-dependent DNA repair mechanism in which a new form of the NuA4-TIP60 chromatin modifier assembles in activated neurons around the inducible, neuronal-specific transcription factor NPAS4. We purify this complex from the brain and demonstrate its functions in eliciting activity-dependent changes to neuronal transcriptomes and circuitry. By characterizing the landscape of activity-induced DNA double-strand breaks in the brain, we show that NPAS4-NuA4 binds to recurrently damaged regulatory elements and recruits additional DNA repair machinery to stimulate their repair. Gene regulatory elements bound by NPAS4-NuA4 are partially protected against age-dependent accumulation of somatic mutations. Impaired NPAS4-NuA4 signalling leads to a cascade of cellular defects, including dysregulated activity-dependent transcriptional responses, loss of control over neuronal inhibition and genome instability, which all culminate to reduce organismal lifespan. In addition, mutations in several components of the NuA4 complex are reported to lead to neurodevelopmental and autism spectrum disorders. Together, these findings identify a neuronal-specific complex that couples neuronal activity directly to genome preservation, the disruption of which may contribute to developmental disorders, neurodegeneration and ageing.


Subject(s)
Brain , DNA Repair , Multiprotein Complexes , Neurons , Synapses , Basic Helix-Loop-Helix Transcription Factors , Brain/metabolism , DNA Breaks, Double-Stranded , Gene Expression Regulation , Lysine Acetyltransferase 5/metabolism , Multiprotein Complexes/metabolism , Neurons/metabolism , Synapses/metabolism , Mutation , Longevity/genetics , Genome , Aging/genetics , Neurodegenerative Diseases
9.
Nat Neurosci ; 25(10): 1353-1365, 2022 10.
Article in English | MEDLINE | ID: mdl-36171426

ABSTRACT

The precise regulation of gene expression is fundamental to neurodevelopment, plasticity and cognitive function. Although several studies have profiled transcription in the developing human brain, there is a gap in understanding of accompanying translational regulation. In this study, we performed ribosome profiling on 73 human prenatal and adult cortex samples. We characterized the translational regulation of annotated open reading frames (ORFs) and identified thousands of previously unknown translation events, including small ORFs that give rise to human-specific and/or brain-specific microproteins, many of which we independently verified using proteomics. Ribosome profiling in stem-cell-derived human neuronal cultures corroborated these findings and revealed that several neuronal activity-induced non-coding RNAs encode previously undescribed microproteins. Physicochemical analysis of brain microproteins identified a class of proteins that contain arginine-glycine-glycine (RGG) repeats and, thus, may be regulators of RNA metabolism. This resource expands the known translational landscape of the human brain and illuminates previously unknown brain-specific protein products.


Subject(s)
Gene Expression Regulation , Protein Biosynthesis , Adult , Arginine/genetics , Arginine/metabolism , Brain/metabolism , Glycine , Humans , RNA, Messenger/metabolism
10.
Nat Neurosci ; 24(3): 437-448, 2021 03.
Article in English | MEDLINE | ID: mdl-33542524

ABSTRACT

Neuronal activity-dependent gene expression is essential for brain development. Although transcriptional and epigenetic effects of neuronal activity have been explored in mice, such an investigation is lacking in humans. Because alterations in GABAergic neuronal circuits are implicated in neurological disorders, we conducted a comprehensive activity-dependent transcriptional and epigenetic profiling of human induced pluripotent stem cell-derived GABAergic neurons similar to those of the early developing striatum. We identified genes whose expression is inducible after membrane depolarization, some of which have specifically evolved in primates and/or are associated with neurological diseases, including schizophrenia and autism spectrum disorder (ASD). We define the genome-wide profile of human neuronal activity-dependent enhancers, promoters and the transcription factors CREB and CRTC1. We found significant heritability enrichment for ASD in the inducible promoters. Our results suggest that sequence variation within activity-inducible promoters of developing human forebrain GABAergic neurons contributes to ASD risk.


Subject(s)
Brain/metabolism , Epigenesis, Genetic , GABAergic Neurons/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Humans , Induced Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic
11.
Nat Neurosci ; 24(2): 204-213, 2021 02.
Article in English | MEDLINE | ID: mdl-33361822

ABSTRACT

Maternal infection and inflammation during pregnancy are associated with neurodevelopmental disorders in offspring, but little is understood about the molecular mechanisms underlying this epidemiologic phenomenon. Here, we leveraged single-cell RNA sequencing to profile transcriptional changes in the mouse fetal brain in response to maternal immune activation (MIA) and identified perturbations in cellular pathways associated with mRNA translation, ribosome biogenesis and stress signaling. We found that MIA activates the integrated stress response (ISR) in male, but not female, MIA offspring in an interleukin-17a-dependent manner, which reduced global mRNA translation and altered nascent proteome synthesis. Moreover, blockade of ISR activation prevented the behavioral abnormalities as well as increased cortical neural activity in MIA male offspring. Our data suggest that sex-specific activation of the ISR leads to maternal inflammation-associated neurodevelopmental disorders.


Subject(s)
Brain/immunology , Fetus/immunology , Immunity, Innate/genetics , Proteostasis/genetics , Animals , Behavior, Animal , Developmental Disabilities/genetics , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Pregnancy , Protein Biosynthesis/genetics , Proteome/biosynthesis , RNA/biosynthesis , RNA/genetics , RNA, Small Interfering , Sex Characteristics , Signal Transduction , Stress, Psychological/genetics , Stress, Psychological/psychology
12.
Nature ; 590(7844): 115-121, 2021 02.
Article in English | MEDLINE | ID: mdl-33299180

ABSTRACT

Behavioural experiences activate the FOS transcription factor in sparse populations of neurons that are critical for encoding and recalling specific events1-3. However, there is limited understanding of the mechanisms by which experience drives circuit reorganization to establish a network of Fos-activated cells. It is also not known whether FOS is required in this process beyond serving as a marker of recent neural activity and, if so, which of its many gene targets underlie circuit reorganization. Here we demonstrate that when mice engage in spatial exploration of novel environments, perisomatic inhibition of Fos-activated hippocampal CA1 pyramidal neurons by parvalbumin-expressing interneurons is enhanced, whereas perisomatic inhibition by cholecystokinin-expressing interneurons is weakened. This bidirectional modulation of inhibition is abolished when the function of the FOS transcription factor complex is disrupted. Single-cell RNA-sequencing, ribosome-associated mRNA profiling and chromatin analyses, combined with electrophysiology, reveal that FOS activates the transcription of Scg2, a gene that encodes multiple distinct neuropeptides, to coordinate these changes in inhibition. As parvalbumin- and cholecystokinin-expressing interneurons mediate distinct features of pyramidal cell activity4-6, the SCG2-dependent reorganization of inhibitory synaptic input might be predicted to affect network function in vivo. Consistent with this prediction, hippocampal gamma rhythms and pyramidal cell coupling to theta phase are significantly altered in the absence of Scg2. These findings reveal an instructive role for FOS and SCG2 in establishing a network of Fos-activated neurons via the rewiring of local inhibition to form a selectively modulated state. The opposing plasticity mechanisms acting on distinct inhibitory pathways may support the consolidation of memories over time.


Subject(s)
Nerve Net/cytology , Nerve Net/physiology , Neural Inhibition , Neuronal Plasticity/physiology , Proto-Oncogene Proteins c-fos/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Cholecystokinin/metabolism , Exploratory Behavior/physiology , Female , Gamma Rhythm , Interneurons/metabolism , Male , Memory Consolidation , Mice , Parvalbumins/metabolism , Pyramidal Cells/metabolism , Secretogranin II/genetics , Secretogranin II/metabolism , Spatial Navigation/physiology , Theta Rhythm
13.
Nature ; 583(7814): 115-121, 2020 07.
Article in English | MEDLINE | ID: mdl-32528180

ABSTRACT

The advent of endothermy, which is achieved through the continuous homeostatic regulation of body temperature and metabolism1,2, is a defining feature of mammalian and avian evolution. However, when challenged by food deprivation or harsh environmental conditions, many mammalian species initiate adaptive energy-conserving survival strategies-including torpor and hibernation-during which their body temperature decreases far below its homeostatic set-point3-5. How homeothermic mammals initiate and regulate these hypothermic states remains largely unknown. Here we show that entry into mouse torpor, a fasting-induced state with a greatly decreased metabolic rate and a body temperature as low as 20 °C6, is regulated by neurons in the medial and lateral preoptic area of the hypothalamus. We show that restimulation of neurons that were activated during a previous bout of torpor is sufficient to initiate the key features of torpor, even in mice that are not calorically restricted. Among these neurons we identify a population of glutamatergic Adcyap1-positive cells, the activity of which accurately determines when mice naturally initiate and exit torpor, and the inhibition of which disrupts the natural process of torpor entry, maintenance and arousal. Taken together, our results reveal a specific neuronal population in the mouse hypothalamus that serves as a core regulator of torpor. This work forms a basis for the future exploration of mechanisms and circuitry that regulate extreme hypothermic and hypometabolic states, and enables genetic access to monitor, initiate, manipulate and study these ancient adaptations of homeotherm biology.


Subject(s)
Energy Metabolism/physiology , Hypothalamus/cytology , Neural Pathways/physiology , Neurons/physiology , Torpor/physiology , Animals , Fasting , Female , Food Deprivation , Glutamine/metabolism , Hypothalamus/physiology , Male , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
14.
Mol Cell ; 77(2): 294-309.e9, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31784358

ABSTRACT

Mutations in the methyl-DNA-binding repressor protein MeCP2 cause the devastating neurodevelopmental disorder Rett syndrome. It has been challenging to understand how MeCP2 regulates transcription because MeCP2 binds broadly across the genome and MeCP2 mutations are associated with widespread small-magnitude changes in neuronal gene expression. We demonstrate here that MeCP2 represses nascent RNA transcription of highly methylated long genes in the brain through its interaction with the NCoR co-repressor complex. By measuring the rates of transcriptional initiation and elongation directly in the brain, we find that MeCP2 has no measurable effect on transcriptional elongation, but instead represses the rate at which Pol II initiates transcription of highly methylated long genes. These findings suggest a new model of MeCP2 function in which MeCP2 binds broadly across highly methylated regions of DNA, but acts at transcription start sites to attenuate transcriptional initiation.


Subject(s)
DNA Methylation/genetics , Methyl-CpG-Binding Protein 2/genetics , Repressor Proteins/genetics , Transcription, Genetic/genetics , Animals , Brain/physiology , DNA/genetics , Male , Mice , Mice, Knockout , Mutation/genetics , Neurons/physiology , RNA/genetics , Rett Syndrome/genetics
15.
Elife ; 82019 09 23.
Article in English | MEDLINE | ID: mdl-31545165

ABSTRACT

Enhancers are the primary DNA regulatory elements that confer cell type specificity of gene expression. Recent studies characterizing individual enhancers have revealed their potential to direct heterologous gene expression in a highly cell-type-specific manner. However, it has not yet been possible to systematically identify and test the function of enhancers for each of the many cell types in an organism. We have developed PESCA, a scalable and generalizable method that leverages ATAC- and single-cell RNA-sequencing protocols, to characterize cell-type-specific enhancers that should enable genetic access and perturbation of gene function across mammalian cell types. Focusing on the highly heterogeneous mammalian cerebral cortex, we apply PESCA to find enhancers and generate viral reagents capable of accessing and manipulating a subset of somatostatin-expressing cortical interneurons with high specificity. This study demonstrates the utility of this platform for developing new cell-type-specific viral reagents, with significant implications for both basic and translational research.


Subject(s)
Gene Expression Regulation/drug effects , Molecular Biology/methods , Neurons/drug effects , Neurophysiology/methods , Recombinant Proteins/biosynthesis , Somatostatin/metabolism , Viruses/genetics , Animals , Animals, Genetically Modified , Cerebral Cortex/physiology , Genes, Regulator , Genetic Vectors , Interneurons/physiology , Mice , Recombinant Proteins/genetics
16.
Nat Neurosci ; 21(12): 1670-1679, 2018 12.
Article in English | MEDLINE | ID: mdl-30455458

ABSTRACT

In females with X-linked genetic disorders, wild-type and mutant cells coexist within brain tissue because of X-chromosome inactivation, posing challenges for interpreting the effects of X-linked mutant alleles on gene expression. We present a single-nucleus RNA sequencing approach that resolves mosaicism by using single-nucleotide polymorphisms in genes expressed in cis with the X-linked mutation to determine which nuclei express the mutant allele even when the mutant gene is not detected. This approach enables gene expression comparisons between mutant and wild-type cells within the same individual, eliminating variability introduced by comparisons to controls with different genetic backgrounds. We apply this approach to mosaic female mouse models and humans with Rett syndrome, an X-linked neurodevelopmental disorder caused by mutations in the gene encoding the methyl-DNA-binding protein MECP2, and observe that cell-type-specific DNA methylation predicts the degree of gene upregulation in MECP2-mutant neurons. This approach can be broadly applied to study gene expression in mosaic X-linked disorders.


Subject(s)
Brain/metabolism , Methyl-CpG-Binding Protein 2/genetics , Rett Syndrome/genetics , Alleles , DNA Methylation , Female , Humans , Methyl-CpG-Binding Protein 2/metabolism , Mosaicism , Mutation , Neurons/metabolism , Polymorphism, Single Nucleotide , Rett Syndrome/metabolism , Sequence Analysis, RNA
17.
Nature ; 539(7628): 242-247, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830782

ABSTRACT

Sensory stimuli drive the maturation and function of the mammalian nervous system in part through the activation of gene expression networks that regulate synapse development and plasticity. These networks have primarily been studied in mice, and it is not known whether there are species- or clade-specific activity-regulated genes that control features of brain development and function. Here we use transcriptional profiling of human fetal brain cultures to identify an activity-dependent secreted factor, Osteocrin (OSTN), that is induced by membrane depolarization of human but not mouse neurons. We find that OSTN has been repurposed in primates through the evolutionary acquisition of DNA regulatory elements that bind the activity-regulated transcription factor MEF2. In addition, we demonstrate that OSTN is expressed in primate neocortex and restricts activity-dependent dendritic growth in human neurons. These findings suggest that, in response to sensory input, OSTN regulates features of neuronal structure and function that are unique to primates.


Subject(s)
Evolution, Molecular , Muscle Proteins/metabolism , Neocortex/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Transcriptome , Animals , Base Sequence , Bone and Bones/metabolism , Dendrites/metabolism , Enhancer Elements, Genetic/genetics , Female , Humans , MEF2 Transcription Factors/metabolism , Macaca mulatta , Male , Mice , Molecular Sequence Data , Muscle Proteins/genetics , Muscles/metabolism , Neocortex/cytology , Neurons/cytology , Organ Specificity , Species Specificity , Transcription Factors/genetics
18.
Proc Natl Acad Sci U S A ; 112(22): 6800-6, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25739960

ABSTRACT

DNA methylation at CpG dinucleotides is an important epigenetic regulator common to virtually all mammalian cell types, but recent evidence indicates that during early postnatal development neuronal genomes also accumulate uniquely high levels of two alternative forms of methylation, non-CpG methylation and hydroxymethylation. Here we discuss the distinct landscape of DNA methylation in neurons, how it is established, and how it might affect the binding and function of protein readers of DNA methylation. We review studies of one critical reader of DNA methylation in the brain, the Rett syndrome protein methyl CpG-binding protein 2 (MeCP2), and discuss how differential binding affinity of MeCP2 for non-CpG and hydroxymethylation may affect the function of this methyl-binding protein in the nervous system.


Subject(s)
Brain/metabolism , DNA Methylation/physiology , Gene Expression Regulation/physiology , Methyl-CpG-Binding Protein 2/metabolism , Models, Biological , Neurons/metabolism , Animals , Cytosine/chemistry , Humans , Molecular Structure , Protein Binding
19.
Cell ; 140(5): 704-16, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20211139

ABSTRACT

Angelman Syndrome is a debilitating neurological disorder caused by mutation of the E3 ubiquitin ligase Ube3A, a gene whose mutation has also recently been associated with autism spectrum disorders (ASDs). The function of Ube3A during nervous system development and how Ube3A mutations give rise to cognitive impairment in individuals with Angleman Syndrome and ASDs are not clear. We report here that experience-driven neuronal activity induces Ube3A transcription and that Ube3A then regulates excitatory synapse development by controlling the degradation of Arc, a synaptic protein that promotes the internalization of the AMPA subtype of glutamate receptors. We find that disruption of Ube3A function in neurons leads to an increase in Arc expression and a concomitant decrease in the number of AMPA receptors at excitatory synapses. We propose that this deregulation of AMPA receptor expression at synapses may contribute to the cognitive dysfunction that occurs in Angelman Syndrome and possibly other ASDs.


Subject(s)
Angelman Syndrome/physiopathology , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cells, Cultured , Cognition , Humans , Mice , Mice, Knockout , Receptors, AMPA/metabolism , Synapses/metabolism , Ubiquitination
20.
Neuron ; 53(2): 217-32, 2007 Jan 18.
Article in English | MEDLINE | ID: mdl-17224404

ABSTRACT

We report the results of a genetic screen to identify molecules important for synapse formation and/or maintenance. siRNAs were used to decrease the expression of candidate genes in neurons, and synapse development was assessed. We surveyed 22 cadherin family members and demonstrated distinct roles for cadherin-11 and cadherin-13 in synapse development. Our screen also revealed roles for the class 4 Semaphorins Sema4B and Sema4D in the development of glutamatergic and/or GABAergic synapses. We found that Sema4D affects the formation of GABAergic, but not glutamatergic, synapses. Our screen also identified the activity-regulated small GTPase Rem2 as a regulator of synapse development. A known calcium channel modulator, Rem2 may function as part of a homeostatic mechanism that controls synapse number. These experiments establish the feasibility of RNAi screens to characterize the mechanisms that control mammalian neuronal development and to identify components of the genetic program that regulate synapse formation and/or maintenance.


Subject(s)
Glutamic Acid/metabolism , Nerve Tissue Proteins/physiology , RNA Interference , Synapses/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Cadherins/physiology , Feasibility Studies , Humans , Molecular Biology , Monomeric GTP-Binding Proteins/physiology , RNA, Small Interfering , Semaphorins/classification , Semaphorins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...