Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biomedicines ; 12(2)2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38397909

ABSTRACT

Alzheimer's disease (AD) is a multi-stage neurodegenerative disorder characterized by beta-amyloid accumulation, hyperphosphorylated Tau deposits, neurodegeneration, neuroinflammation, and cognitive impairment. Recent studies implicate CD8 T cells as neuroimmune responders to the accumulation of AD pathology in the brain and potential contributors to toxic neuroinflammation. However, more evidence is needed to understand lymphocytes in disease, including their functional states, molecular mediators, and interacting cell types in diseased brain tissue. The scarcity of lymphocytes in brain tissue samples has limited the unbiased profiling of disease-associated cell types, cell states, drug targets, and relationships to common AD genetic risk variants based on transcriptomic analyses. However, using recent large-scale, high-quality single-nuclear sequencing datasets from over 84 Alzheimer's disease and control cases, we leverage single-nuclear RNAseq data from 800 lymphocytes collected from 70 individuals to complete unbiased molecular profiling. We demonstrate that effector memory CD8 T cells are the major lymphocyte subclass enriched in the brain tissues of individuals with AD dementia. We define disease-enriched interactions involving CD8 T cells and multiple brain cell subclasses including two distinct microglial disease states that correlate, respectively, to beta-amyloid and tau pathology. We find that beta-amyloid-associated microglia are a major hub of multicellular cross-talk gained in disease, including interactions involving both vulnerable neuronal subtypes and CD8 T cells. We reproduce prior reports that amyloid-response microglia are depleted in APOE4 carriers. Overall, these human-based studies provide additional support for the potential relevance of effector memory CD8 T cells as a lymphocyte population of interest in AD dementia and provide new candidate interacting partners and drug targets for further functional study.

2.
Alzheimers Dement ; 20(3): 2240-2261, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38170841

ABSTRACT

INTRODUCTION: The pace of innovation has accelerated in virtually every area of tau research in just the past few years. METHODS: In February 2022, leading international tau experts convened to share selected highlights of this work during Tau 2022, the second international tau conference co-organized and co-sponsored by the Alzheimer's Association, CurePSP, and the Rainwater Charitable Foundation. RESULTS: Representing academia, industry, and the philanthropic sector, presenters joined more than 1700 registered attendees from 59 countries, spanning six continents, to share recent advances and exciting new directions in tau research. DISCUSSION: The virtual meeting provided an opportunity to foster cross-sector collaboration and partnerships as well as a forum for updating colleagues on research-advancing tools and programs that are steadily moving the field forward.


Subject(s)
Alzheimer Disease , Tauopathies , Humans , tau Proteins
3.
bioRxiv ; 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37808727

ABSTRACT

The development of successful therapeutics for dementias requires an understanding of their shared and distinct molecular features in the human brain. We performed single-nuclear RNAseq and ATACseq in Alzheimer disease (AD), Frontotemporal degeneration (FTD), and Progressive Supranuclear Palsy (PSP), analyzing 40 participants, yielding over 1.4M cells from three brain regions ranging in vulnerability and pathological burden. We identify 35 shared disease-associated cell types and 14 that are disease-specific, replicating those previously identified in AD. Disease - specific cell states represent molecular features of disease-specific glial-immune mechanisms and neuronal vulnerability in each disorder, layer 4/5 intra-telencephalic neurons in AD, layer 2/3 intra-telencephalic neurons in FTD, and layer 5/6 near-projection neurons in PSP. We infer intrinsic disease-associated gene regulatory networks, which we empirically validate by chromatin footprinting. We find that causal genetic risk acts in specific neuronal and glial cells that differ across disorders, primarily non-neuronal cells in AD and specific neuronal subtypes in FTD and PSP. These data illustrate the heterogeneous spectrum of glial and neuronal composition and gene expression alterations in different dementias and identify new therapeutic targets by revealing shared and disease-specific cell states.

4.
Alzheimers Dement ; 19(8): 3448-3457, 2023 08.
Article in English | MEDLINE | ID: mdl-36807763

ABSTRACT

INTRODUCTION: We tested sex-dependent associations of variation in the SNAP-25 gene, which encodes a presynaptic protein involved in hippocampal plasticity and memory, on cognitive and Alzheimer's disease (AD) neuroimaging outcomes in clinically normal adults. METHODS: Participants were genotyped for SNAP-25 rs1051312 (T > C; SNAP-25 expression: C-allele > T/T). In a discovery cohort (N = 311), we tested the sex by SNAP-25 variant interaction on cognition, Aß-PET positivity, and temporal lobe volumes. Cognitive models were replicated in an independent cohort (N = 82). RESULTS: In the discovery cohort, C-allele carriers exhibited better verbal memory and language, lower Aß-PET positivity rates, and larger temporal volumes than T/T homozygotes among females, but not males. Larger temporal volumes related to better verbal memory only in C-carrier females. The female-specific C-allele verbal memory advantage was evidenced in the replication cohort. CONCLUSIONS: In females, genetic variation in SNAP-25 is associated with resistance to amyloid plaque formation and may support verbal memory through fortification of temporal lobe architecture. HIGHLIGHTS: The SNAP-25 rs1051312 (T > C) C-allele results in higher basal SNAP-25 expression. C-allele carriers had better verbal memory in clinically normal women, but not men. Female C-carriers had higher temporal lobe volumes, which predicted verbal memory. Female C-carriers also exhibited the lowest rates of amyloid-beta PET positivity. The SNAP-25 gene may influence female-specific resistance to Alzheimer's disease (AD).


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Aged , Female , Humans , Male , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Biomarkers/metabolism , Genotype , Memory , Positron-Emission Tomography
5.
Front Immunol ; 13: 1056417, 2022.
Article in English | MEDLINE | ID: mdl-36618392

ABSTRACT

Introduction: Progranulin (PGRN) is a secreted glycoprotein, the expression of which is linked to several neurodegenerative diseases. Although its specific function is still unclear, several studies have linked it with lysosomal functions and immune system regulation. Here, we have explored the role of PGRN in peripheral and central immune system homeostasis by investigating the consequences of PGRN deficiency on adaptive and innate immune cell populations. Methods: First, we used gene co-expression network analysis of published data to test the hypothesis that Grn has a critical role in regulating the activation status of immune cell populations in both central and peripheral compartments. To investigate the extent to which PGRN-deficiency resulted in immune dysregulation, we performed deep immunophenotyping by flow cytometry of 19-24-month old male and female Grn-deficient mice (PGRN KO) and littermate Grn-sufficient controls (WT). Results: Male PGRN KO mice exhibited a lower abundance of microglial cells with higher MHC-II expression, increased CD44 expression on monocytes in the brain, and more CNS-associated CD8+ T cells compared to WT mice. Furthermore, we observed an increase in CD44 on CD8+ T cells in the peripheral blood. Female PGRN KO mice also had fewer microglia compared to WT mice, and we also observed reduced expression of MHC-II on brain monocytes. Additionally, we found an increase in Ly-6Chigh monocyte frequency and decreased CD44 expression on CD8+ and CD4+ T cells in PGRN KO female blood. Given that Gpnmb, which encodes for the lysosomal protein Glycoprotein non-metastatic melanoma protein B, has been reported to be upregulated in PGRN KO mice, we investigated changes in GPNMB protein expression associated with PGRN deficits and found that GPNMB is modulated in myeloid cells in a sex-specific manner. Discussion: Our data suggest that PGRN and GPNMB jointly regulate the peripheral and the central immune system in a sex-specific manner; thus, understanding their associated mechanisms could pave the way for developing new neuroprotective strategies to modulate central and peripheral inflammation to lower risk for neurodegenerative diseases and possibly delay or halt progression.


Subject(s)
CD8-Positive T-Lymphocytes , Intercellular Signaling Peptides and Proteins , Male , Female , Animals , Mice , Progranulins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Granulins , Mice, Knockout , Immune System
6.
Neuron ; 109(14): 2275-2291.e8, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34133945

ABSTRACT

C9orf72 repeat expansions cause inherited amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD) and result in both loss of C9orf72 protein expression and production of potentially toxic RNA and dipeptide repeat proteins. In addition to ALS/FTD, C9orf72 repeat expansions have been reported in a broad array of neurodegenerative syndromes, including Alzheimer's disease. Here we show that C9orf72 deficiency promotes a change in the homeostatic signature in microglia and a transition to an inflammatory state characterized by an enhanced type I IFN signature. Furthermore, C9orf72-depleted microglia trigger age-dependent neuronal defects, in particular enhanced cortical synaptic pruning, leading to altered learning and memory behaviors in mice. Interestingly, C9orf72-deficient microglia promote enhanced synapse loss and neuronal deficits in a mouse model of amyloid accumulation while paradoxically improving plaque clearance. These findings suggest that altered microglial function due to decreased C9orf72 expression directly contributes to neurodegeneration in repeat expansion carriers independent of gain-of-function toxicities.


Subject(s)
Aging/metabolism , Amyloid/metabolism , C9orf72 Protein/metabolism , Microglia/metabolism , Synapses/metabolism , Aging/genetics , Aging/pathology , Amyloid/genetics , Animals , C9orf72 Protein/genetics , DNA Repeat Expansion , Disease Models, Animal , Lysosomes/metabolism , Mice , Mice, Knockout , Synapses/pathology
7.
Nat Commun ; 12(1): 3958, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172753

ABSTRACT

Astrocytes play important roles in neurological disorders such as stroke, injury, and neurodegeneration. Most knowledge on astrocyte biology is based on studies of mouse models and the similarities and differences between human and mouse astrocytes are insufficiently characterized, presenting a barrier in translational research. Based on analyses of acutely purified astrocytes, serum-free cultures of primary astrocytes, and xenografted chimeric mice, we find extensive conservation in astrocytic gene expression between human and mouse samples. However, the genes involved in defense response and metabolism show species-specific differences. Human astrocytes exhibit greater susceptibility to oxidative stress than mouse astrocytes, due to differences in mitochondrial physiology and detoxification pathways. In addition, we find that mouse but not human astrocytes activate a molecular program for neural repair under hypoxia, whereas human but not mouse astrocytes activate the antigen presentation pathway under inflammatory conditions. Here, we show species-dependent properties of astrocytes, which can be informative for improving translation from mouse models to humans.


Subject(s)
Astrocytes/physiology , Animals , Antigen Presentation , Astrocytes/drug effects , Cells, Cultured , Gene Expression/drug effects , Humans , Inactivation, Metabolic , Inflammation , Mice , Mitochondria/metabolism , Nervous System Diseases/drug therapy , Nervous System Diseases/pathology , Oxidative Stress , Poly I-C/pharmacology , Poly I-C/therapeutic use , Species Specificity , Transcriptome/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/therapeutic use
8.
Cell Rep ; 33(7): 108398, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33207193

ABSTRACT

To understand how neural-immune-associated genes and pathways contribute to neurodegenerative disease pathophysiology, we performed a systematic functional genomic analysis in purified microglia and bulk tissue from mouse and human AD, FTD, and PSP. We uncover a complex temporal trajectory of microglial-immune pathways involving the type 1 interferon response associated with tau pathology in the early stages, followed by later signatures of partial immune suppression and, subsequently, the type 2 interferon response. We find that genetic risk for dementias shows disease-specific patterns of pathway enrichment. We identify drivers of two gene co-expression modules conserved from mouse to human, representing competing arms of microglial-immune activation (NAct) and suppression (NSupp) in neurodegeneration. We validate our findings by using chemogenetics, experimental perturbation data, and single-cell sequencing in post-mortem brains. Our results refine the understanding of stage- and disease-specific microglial responses, implicate microglial viral defense pathways in dementia pathophysiology, and highlight therapeutic windows.


Subject(s)
Dementia/genetics , Tauopathies/genetics , tau Proteins/metabolism , Aged , Animals , Brain/metabolism , Female , Frontotemporal Dementia/genetics , Gene Regulatory Networks/genetics , Genetic Predisposition to Disease , Genomics/methods , Humans , Immunosuppression Therapy , Inflammation/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Primary Cell Culture , Risk Factors , Tauopathies/metabolism , Tauopathies/physiopathology , tau Proteins/genetics , tau Proteins/physiology
9.
Neuropsychology ; 34(8): 835-850, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33030915

ABSTRACT

Objective: We aimed to test the hypothesis that elevated neocortical ß-amyloid (Aß), a hallmark feature of Alzheimer's disease (AD), predicts sex-specific cognitive trajectories in clinically normal older adults, with women showing greater risk of decline than men. Method: Florbetapir Aß positron emission tomography (PET) was acquired in 149 clinically normal older adults (52% female, Mage = 74). Participants underwent cognitive testing at baseline and during annual follow-up visits over a timespan of up to 5.14 years. Mixed-effects regression models evaluated whether relations between baseline neocortical Standardized Uptake Value Ratio (SUVR) and composite scores of episodic memory, executive functioning, and processing speed were moderated by sex (male/female) and apolipoprotein E (APOE) status (ε4 carrier/noncarrier). Results: Higher baseline SUVR was associated with longitudinal decline in episodic memory in women (b = -1.32, p < .001) but not men (b = -0.30, p = .28). Female APOE ε4 carriers with elevated SUVR showed particularly precipitous declines in episodic memory (b = -4.33, p < .001) whereas other cognitive domains were spared. SUVR did not predict changes in executive functioning or processing speed, regardless of sex (ps >.63), though there was a main effect of SUVR on processing speed (b = 2.50, p = .003). Conclusions: Clinically normal women with elevated Aß are more vulnerable to episodic memory decline than men. Understanding sex-related differences in AD, particularly in preclinical stages, is crucial for guiding precision medicine approaches to early detection and intervention. (PsycInfo Database Record (c) 2020 APA, all rights reserved).


Subject(s)
Aging/psychology , Amyloid beta-Peptides/metabolism , Cognition/physiology , Aged , Aged, 80 and over , Aniline Compounds , Apolipoprotein E4/genetics , Ethylene Glycols , Executive Function , Female , Genotype , Heterozygote , Humans , Male , Memory, Episodic , Middle Aged , Neuropsychological Tests , Positron-Emission Tomography , Radiopharmaceuticals , Reaction Time/physiology , Sex Characteristics
10.
Neurocase ; 26(5): 299-304, 2020 10.
Article in English | MEDLINE | ID: mdl-32893728

ABSTRACT

We report a patient with autism-like deficits in emotional connectedness, executive dysfunction, and ataxia beginning at age 39. He had compound heterozygous variants in SPG7 (A510V and 1552+1 G>T substitutions), mutation of which is classically associated with spastic paraparesis. Diffusion MRI demonstrated abnormalities in the cerebellar outflow tracts. Transcranial magnetic stimulation showed a prolonged cortical silent period representing exaggerated cortical inhibition, as previously described with pure cerebellar degeneration. The acquired cerebellar cognitive affective syndrome in association with specific anatomic and neurophysiological abnormalities in the cerebellum expand the spectrum of SPG7-related neurodegeneration and support a role for cerebellar output in socio-emotional behavior.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Affective Symptoms , Cerebellar Diseases , Cognitive Dysfunction , Metalloendopeptidases/genetics , Neurodegenerative Diseases , Social Interaction , Affective Symptoms/diagnosis , Affective Symptoms/etiology , Affective Symptoms/physiopathology , Cerebellar Ataxia/complications , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/genetics , Cerebellar Diseases/complications , Cerebellar Diseases/diagnosis , Cerebellar Diseases/genetics , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Diffusion Magnetic Resonance Imaging , Gait Disorders, Neurologic/diagnosis , Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/physiopathology , Humans , Male , Middle Aged , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/genetics , Transcranial Magnetic Stimulation
11.
J Child Neurol ; 35(14): 953-962, 2020 12.
Article in English | MEDLINE | ID: mdl-32705938

ABSTRACT

Spinocerebellar ataxia type 21 (SCA21/ATX-TMEM240) is a rare form of cerebellar ataxia that commonly presents with motor, cognitive, and behavioral impairments. Although these features have been identified as part of the clinical manifestations of SCA21, the neurodevelopmental disorders associated with SCA21 have not been well studied or described. Here we present extensive phenotypic data for 3 subjects from an SCA21 family in the United States. Genetic testing demonstrated the c.196 G>A (p.Gly66Arg) variant to be a second recurrent mutation associated with the disorder. Standardized developmental assessment revealed significant deficits in cognition, adaptive function, motor skills, and social communication with 2 of the subjects having diagnoses of autism spectrum disorder, which has never been described in SCA21. Quantitative gait analysis showed markedly abnormal spatiotemporal gait variables indicative of poor gait control and cerebellar as well as noncerebellar dysfunction. Clinical evaluation also highlighted a striking variability in clinical symptoms, with greater ataxia correlating with greater severity of neurodevelopmental disorder diagnoses. Notably, neurodevelopmental outcomes have improved with intervention over time. Taken together, this case series identifies that the manifestation of neurodevelopmental disorders is a key feature of SCA21 and may precede the presence of motor abnormalities. Furthermore, the coexistence of ataxia and neurodevelopmental disorders in these subjects suggests a role for spinocerebellar pathways in both outcomes. The findings in this study highlight the importance of evaluation of neurodevelopmental concerns in the context of progressive motor abnormalities and the need for timely intervention to ultimately improve quality of life for individuals with SCA21.


Subject(s)
Gait/physiology , Intellectual Disability/diagnosis , Membrane Proteins/genetics , Motor Skills/physiology , Spinocerebellar Degenerations/diagnosis , Adolescent , Brain/diagnostic imaging , Child , Cognition , Communication , Female , Humans , Intellectual Disability/genetics , Magnetic Resonance Imaging , Male , Mutation , Phenotype , Spinocerebellar Degenerations/genetics , Symptom Assessment
12.
Hum Mutat ; 41(2): 487-501, 2020 02.
Article in English | MEDLINE | ID: mdl-31692161

ABSTRACT

Genetic ataxias are associated with mutations in hundreds of genes with high phenotypic overlap complicating the clinical diagnosis. Whole-exome sequencing (WES) has increased the overall diagnostic rate considerably. However, the upper limit of this method remains ill-defined, hindering efforts to address the remaining diagnostic gap. To further assess the role of rare coding variation in ataxic disorders, we reanalyzed our previously published exome cohort of 76 predominantly adult and sporadic-onset patients, expanded the total number of cases to 260, and introduced analyses for copy number variation and repeat expansion in a representative subset. For new cases (n = 184), our resulting clinically relevant detection rate remained stable at 47% with 24% classified as pathogenic. Reanalysis of the previously sequenced 76 patients modestly improved the pathogenic rate by 7%. For the combined cohort (n = 260), the total observed clinical detection rate was 52% with 25% classified as pathogenic. Published studies of similar neurological phenotypes report comparable rates. This consistency across multiple cohorts suggests that, despite continued technical and analytical advancements, an approximately 50% diagnostic rate marks a relative ceiling for current WES-based methods and a more comprehensive genome-wide assessment is needed to identify the missing causative genetic etiologies for cerebellar ataxia and related neurodegenerative diseases.


Subject(s)
Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/genetics , Exome Sequencing , Exome , Nervous System Diseases/diagnosis , Nervous System Diseases/genetics , DNA Copy Number Variations , Genetic Association Studies , Genetic Linkage , Genetic Predisposition to Disease , Humans , Microsatellite Repeats
13.
Nat Med ; 25(1): 152-164, 2019 01.
Article in English | MEDLINE | ID: mdl-30510257

ABSTRACT

Identifying the mechanisms through which genetic risk causes dementia is an imperative for new therapeutic development. Here, we apply a multistage, systems biology approach to elucidate the disease mechanisms in frontotemporal dementia. We identify two gene coexpression modules that are preserved in mice harboring mutations in MAPT, GRN and other dementia mutations on diverse genetic backgrounds. We bridge the species divide via integration with proteomic and transcriptomic data from the human brain to identify evolutionarily conserved, disease-relevant networks. We find that overexpression of miR-203, a hub of a putative regulatory microRNA (miRNA) module, recapitulates mRNA coexpression patterns associated with disease state and induces neuronal cell death, establishing this miRNA as a regulator of neurodegeneration. Using a database of drug-mediated gene expression changes, we identify small molecules that can normalize the disease-associated modules and validate this experimentally. Our results highlight the utility of an integrative, cross-species network approach to drug discovery.


Subject(s)
Dementia/genetics , Evolution, Molecular , Gene Regulatory Networks , Neurodegenerative Diseases/genetics , Animals , Cell Death/genetics , Disease Models, Animal , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Vectors/metabolism , Humans , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Transcriptome/genetics , tau Proteins/metabolism
14.
Proc Natl Acad Sci U S A ; 113(52): 15120-15125, 2016 12 27.
Article in English | MEDLINE | ID: mdl-27956640

ABSTRACT

O-GlcNAc glycosylation (or O-GlcNAcylation) is a dynamic, inducible posttranslational modification found on proteins associated with neurodegenerative diseases such as α-synuclein, amyloid precursor protein, and tau. Deletion of the O-GlcNAc transferase (ogt) gene responsible for the modification causes early postnatal lethality in mice, complicating efforts to study O-GlcNAcylation in mature neurons and to understand its roles in disease. Here, we report that forebrain-specific loss of OGT in adult mice leads to progressive neurodegeneration, including widespread neuronal cell death, neuroinflammation, increased production of hyperphosphorylated tau and amyloidogenic Aß-peptides, and memory deficits. Furthermore, we show that human cortical brain tissue from Alzheimer's disease patients has significantly reduced levels of OGT protein expression compared with cortical tissue from control individuals. Together, these studies indicate that O-GlcNAcylation regulates pathways critical for the maintenance of neuronal health and suggest that dysfunctional O-GlcNAc signaling may be an important contributor to neurodegenerative diseases.


Subject(s)
Acetylglucosamine/chemistry , Glycosylation , N-Acetylglucosaminyltransferases/chemistry , Neurons/chemistry , Prosencephalon/cytology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain Mapping , Crosses, Genetic , Female , Hippocampus/metabolism , Humans , Inflammation , Male , Memory Disorders , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases/pathology , Neuroglia/metabolism , Neurons/pathology , Phosphorylation , Prosencephalon/metabolism , Signal Transduction , tau Proteins/metabolism
15.
Curr Protoc Chem Biol ; 5(4): 281-302, 2013.
Article in English | MEDLINE | ID: mdl-24391098

ABSTRACT

O-linked N-acetylglucosamine (O-GlcNAc) glycosylation is a dynamic protein posttranslational modification with roles in processes such as transcription, cell cycle regulation, and metabolism. Detailed mechanistic studies of O-GlcNAc have been hindered by a lack of methods for measuring O-GlcNAc stoichiometries and the interplay of glycosylation with other posttranslational modifications. We recently developed a method for labeling O-GlcNAc-modified proteins with resolvable poly(ethylene glycol) mass tags. This mass-tagging approach enables the direct measurement of glycosylation stoichiometries and the visualization of distinct O-GlcNAc-modified subpopulations. Here, we describe procedures for labeling O-GlcNAc glycoproteins in cell lysates with mass tags.


Subject(s)
Acetylglucosamine/metabolism , Glycosylation/drug effects , Polyethylene Glycols/pharmacology , Animals , Azides/chemistry , Carbohydrate Sequence , Cycloaddition Reaction/methods , Galactosyltransferases/chemistry , Galactosyltransferases/genetics , Humans , Molecular Sequence Data , Oximes/metabolism , Protein Processing, Post-Translational/genetics , Proteins/chemistry
16.
Nat Chem Biol ; 8(3): 253-61, 2012 Jan 22.
Article in English | MEDLINE | ID: mdl-22267118

ABSTRACT

The transcription factor cyclic AMP-response element binding protein (CREB) is a key regulator of many neuronal processes, including brain development, circadian rhythm and long-term memory. Studies of CREB have focused on its phosphorylation, although the diversity of CREB functions in the brain suggests additional forms of regulation. Here we expand on a chemoenzymatic strategy for quantifying glycosylation stoichiometries to characterize the functional roles of CREB glycosylation in neurons. We show that CREB is dynamically modified with an O-linked ß-N-acetyl-D-glucosamine sugar in response to neuronal activity and that glycosylation represses CREB-dependent transcription by impairing its association with CREB-regulated transcription coactivator (CRTC; also known as transducer of regulated CREB activity). Blocking glycosylation of CREB alters cellular function and behavioral plasticity, enhancing both axonal and dendritic growth and long-term memory consolidation. Our findings demonstrate a new role for O-glycosylation in memory formation and provide a mechanistic understanding of how glycosylation contributes to critical neuronal functions. Moreover, we identify a previously unknown mechanism for the regulation of activity-dependent gene expression, neural development and memory.


Subject(s)
Acetylglucosamine/metabolism , CREB-Binding Protein/metabolism , Gene Expression Regulation , Memory, Long-Term , Animals , CREB-Binding Protein/chemistry , Glycosylation , Mice , Neurons/cytology , Neurons/drug effects , Neurons/metabolism
17.
Nat Chem Biol ; 6(9): 645-51, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20657584

ABSTRACT

Mechanistic studies of O-GlcNAc glycosylation have been limited by an inability to monitor the glycosylation stoichiometries of proteins obtained from cells. Here we describe a powerful method to visualize the O-GlcNAc-modified protein subpopulation using resolvable polyethylene glycol mass tags. This approach enables rapid quantification of in vivo glycosylation levels on endogenous proteins without the need for protein purification, advanced instrumentation or expensive radiolabels. In addition, it establishes the glycosylation state (for example, mono-, di-, tri-) of proteins, providing information regarding overall O-GlcNAc site occupancy that cannot be obtained using mass spectrometry. Finally, we apply this strategy to rapidly assess the complex interplay between glycosylation and phosphorylation and discover an unexpected reverse 'yin-yang' relationship on the transcriptional repressor MeCP2 that was undetectable by traditional methods. We anticipate that this mass-tagging strategy will advance our understanding of O-GlcNAc glycosylation, as well as other post-translational modifications and poorly understood glycosylation motifs.


Subject(s)
Acetylglucosamine/analysis , Acetylglucosamine/metabolism , Polyethylene Glycols/chemistry , Protein Processing, Post-Translational , Acetylglucosamine/chemistry , Glycosylation , Kinetics , Mass Spectrometry , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Phosphorylation , Polyethylene Glycols/analysis , Protein Processing, Post-Translational/genetics , Transcription, Genetic
18.
Nat Chem Biol ; 4(2): 97-106, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202679

ABSTRACT

O-GlcNAc glycosylation is a unique, dynamic form of glycosylation found on intracellular proteins of all multicellular organisms. Studies suggest that O-GlcNAc represents a key regulatory modification in the brain, contributing to transcriptional regulation, neuronal communication and neurodegenerative disease. Recently, several new chemical tools have been developed to detect and study the modification, including chemoenzymatic tagging methods, quantitative proteomics strategies and small-molecule inhibitors of O-GlcNAc enzymes. Here we highlight some of the emerging roles for O-GlcNAc in the nervous system and describe how chemical tools have significantly advanced our understanding of the scope, functional significance and cellular dynamics of this modification.


Subject(s)
Acetylglucosamine/metabolism , Brain/metabolism , Acetylglucosamine/chemistry , Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Animals , Glycosylation , Humans , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/metabolism , Proteome/metabolism
19.
Nat Chem Biol ; 3(6): 339-48, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17496889

ABSTRACT

The addition of the monosaccharide beta-N-acetyl-D-glucosamine to proteins (O-GlcNAc glycosylation) is an intracellular, post-translational modification that shares features with phosphorylation. Understanding the cellular mechanisms and signaling pathways that regulate O-GlcNAc glycosylation has been challenging because of the difficulty of detecting and quantifying the modification. Here, we describe a new strategy for monitoring the dynamics of O-GlcNAc glycosylation using quantitative mass spectrometry-based proteomics. Our method, which we have termed quantitative isotopic and chemoenzymatic tagging (QUIC-Tag), combines selective, chemoenzymatic tagging of O-GlcNAc proteins with an efficient isotopic labeling strategy. Using the method, we detect changes in O-GlcNAc glycosylation on several proteins involved in the regulation of transcription and mRNA translocation. We also provide the first evidence that O-GlcNAc glycosylation is dynamically modulated by excitatory stimulation of the brain in vivo. Finally, we use electron-transfer dissociation mass spectrometry to identify exact sites of O-GlcNAc modification. Together, our studies suggest that O-GlcNAc glycosylation occurs reversibly in neurons and, akin to phosphorylation, may have important roles in mediating the communication between neurons.


Subject(s)
Acetylglucosamine/metabolism , Brain/metabolism , Glycoproteins/biosynthesis , Proteome , Acetylglucosamine/chemistry , Animals , Carbohydrate Conformation , Glycoproteins/genetics , Glycosylation , Kinetics , Mammals , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Peptides/chemistry , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...