Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 27(5): 822-835, 2024 May.
Article in English | MEDLINE | ID: mdl-38589584

ABSTRACT

Learning and memory require activity-induced changes in dendritic translation, but which mRNAs are involved and how they are regulated are unclear. In this study, to monitor how depolarization impacts local dendritic biology, we employed a dendritically targeted proximity labeling approach followed by crosslinking immunoprecipitation, ribosome profiling and mass spectrometry. Depolarization of primary cortical neurons with KCl or the glutamate agonist DHPG caused rapid reprogramming of dendritic protein expression, where changes in dendritic mRNAs and proteins are weakly correlated. For a subset of pre-localized messages, depolarization increased the translation of upstream open reading frames (uORFs) and their downstream coding sequences, enabling localized production of proteins involved in long-term potentiation, cell signaling and energy metabolism. This activity-dependent translation was accompanied by the phosphorylation and recruitment of the non-canonical translation initiation factor eIF4G2, and the translated uORFs were sufficient to confer depolarization-induced, eIF4G2-dependent translational control. These studies uncovered an unanticipated mechanism by which activity-dependent uORF translational control by eIF4G2 couples activity to local dendritic remodeling.


Subject(s)
Dendrites , Eukaryotic Initiation Factor-4G , Neurons , Open Reading Frames , Protein Biosynthesis , Animals , Dendrites/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Protein Biosynthesis/physiology , Neurons/metabolism , Open Reading Frames/genetics , Rats , Mice , Cells, Cultured , Potassium Chloride/pharmacology
2.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-36993710

ABSTRACT

Attention is required for most higher-order cognitive functions. Prior studies have revealed functional roles for the prefrontal cortex and its extended circuits to enabling attention, but the underlying molecular processes and their impacts on cellular and circuit function remain poorly understood. To develop insights, we here took an unbiased forward genetics approach to identify single genes of large effect on attention. We studied 200 genetically diverse mice on measures of pre-attentive processing and through genetic mapping identified a small locus on chromosome 13 (95%CI: 92.22-94.09 Mb) driving substantial variation (19%) in this trait. Further characterization of the locus revealed a causative gene, Homer1, encoding a synaptic protein, where down-regulation of its short isoforms in prefrontal cortex (PFC) during early postnatal development led to improvements in multiple measures of attention in the adult. Subsequent mechanistic studies revealed that prefrontal Homer1 down-regulation is associated with GABAergic receptor up-regulation in those same cells. This enhanced inhibitory influence, together with dynamic neuromodulatory coupling, led to strikingly low PFC activity at baseline periods of the task but targeted elevations at cue onset, predicting short-latency correct choices. Notably high-Homer1, low-attentional performers, exhibited uniformly elevated PFC activity throughout the task. We thus identify a single gene of large effect on attention - Homer1 - and find that it improves prefrontal inhibitory tone and signal-to-noise (SNR) to enhance attentional performance. A therapeutic strategy focused on reducing prefrontal activity and increasing SNR, rather than uniformly elevating PFC activity, may complement the use of stimulants to improve attention.

3.
Nat Commun ; 14(1): 3956, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37407548

ABSTRACT

The ability to use blood to predict the outcomes of Parkinson's disease, including disease progression and cognitive and motor complications, would be of significant clinical value. We undertook bulk RNA sequencing from the caudate and putamen of postmortem Parkinson's disease (n = 35) and control (n = 40) striatum, and compared molecular profiles with clinical features and bulk RNA sequencing data obtained from antemortem peripheral blood. Cognitive and motor complications of Parkinson's disease were associated with molecular changes in the caudate (stress response) and putamen (endothelial pathways) respectively. Later and earlier-onset Parkinson's disease were molecularly distinct, and disease duration was associated with changes in caudate (oligodendrocyte development) and putamen (cellular senescence), respectively. Transcriptome patterns in the postmortem Parkinson's disease brain were also evident in antemortem peripheral blood, and correlated with clinical features of the disease. Together, these findings identify molecular signatures in Parkinson's disease patients' brain and blood of potential pathophysiologic and prognostic importance.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/metabolism , Transcriptome , Brain/metabolism , Corpus Striatum/metabolism , Putamen
4.
Sci Transl Med ; 15(684): eabq8476, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36812347

ABSTRACT

Periodontal disease is more common in individuals with rheumatoid arthritis (RA) who have detectable anti-citrullinated protein antibodies (ACPAs), implicating oral mucosal inflammation in RA pathogenesis. Here, we performed paired analysis of human and bacterial transcriptomics in longitudinal blood samples from RA patients. We found that patients with RA and periodontal disease experienced repeated oral bacteremias associated with transcriptional signatures of ISG15+HLADRhi and CD48highS100A2pos monocytes, recently identified in inflamed RA synovia and blood of those with RA flares. The oral bacteria observed transiently in blood were broadly citrullinated in the mouth, and their in situ citrullinated epitopes were targeted by extensively somatically hypermutated ACPAs encoded by RA blood plasmablasts. Together, these results suggest that (i) periodontal disease results in repeated breaches of the oral mucosa that release citrullinated oral bacteria into circulation, which (ii) activate inflammatory monocyte subsets that are observed in inflamed RA synovia and blood of RA patients with flares and (iii) activate ACPA B cells, thereby promoting affinity maturation and epitope spreading to citrullinated human antigens.


Subject(s)
Arthritis, Rheumatoid , Periodontal Diseases , Humans , Autoantibodies , Mouth Mucosa , Antibody Formation , Epitopes , Bacteria
5.
J Clin Invest ; 132(18)2022 09 15.
Article in English | MEDLINE | ID: mdl-36106631

ABSTRACT

In addition to playing a major role in tumor cell biology, p53 generates a microenvironment that promotes antitumor immune surveillance via tumor-associated macrophages. We examined whether increasing p53 signaling in the tumor microenvironment influences antitumor T cell immunity. Our findings indicate that increased p53 signaling induced either pharmacologically with APR-246 (eprenetapopt) or in p53-overexpressing transgenic mice can disinhibit antitumor T cell immunity and augment the efficacy of immune checkpoint blockade. We demonstrated that increased p53 expression in tumor-associated macrophages induces canonical p53-associated functions such as senescence and activation of a p53-dependent senescence-associated secretory phenotype. This was linked with decreased expression of proteins associated with M2 polarization by tumor-associated macrophages. Our preclinical data led to the development of a clinical trial in patients with solid tumors combining APR-246 with pembrolizumab. Biospecimens from select patients participating in this ongoing trial showed that there was a suppression of M2-polarized myeloid cells and increase in T cell proliferation with therapy in those who responded to the therapy. Our findings, based on both genetic and a small molecule-based pharmacological approach, suggest that increasing p53 expression in tumor-associated macrophages reprograms the tumor microenvironment to augment the response to immune checkpoint blockade.


Subject(s)
Immune Checkpoint Inhibitors , Tumor-Associated Macrophages , Animals , Immune Checkpoint Inhibitors/pharmacology , Mice , Quinuclidines , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics
6.
Elife ; 102021 12 23.
Article in English | MEDLINE | ID: mdl-34939924

ABSTRACT

Neurons rely on translation of synaptic mRNAs in order to generate activity-dependent changes in plasticity. Here, we develop a strategy combining compartment-specific crosslinking immunoprecipitation (CLIP) and translating ribosome affinity purification (TRAP) in conditionally tagged mice to precisely define the ribosome-bound dendritic transcriptome of CA1 pyramidal neurons. We identify CA1 dendritic transcripts with differentially localized mRNA isoforms generated by alternative polyadenylation and alternative splicing, including many that have altered protein-coding capacity. Among dendritic mRNAs, FMRP targets were found to be overrepresented. Cell-type-specific FMRP-CLIP and TRAP in microdissected CA1 neuropil revealed 383 dendritic FMRP targets and suggests that FMRP differentially regulates functionally distinct modules in CA1 dendrites and cell bodies. FMRP regulates ~15-20% of mRNAs encoding synaptic functions and 10% of chromatin modulators, in the dendrite and cell body, respectively. In the absence of FMRP, dendritic FMRP targets had increased ribosome association, consistent with a function for FMRP in synaptic translational repression. Conversely, downregulation of FMRP targets involved in chromatin regulation in cell bodies suggests a role for FMRP in stabilizing mRNAs containing stalled ribosomes in this compartment. Together, the data support a model in which FMRP regulates the translation and expression of synaptic and nuclear proteins within different compartments of a single neuronal cell type.


The brain has over 100 billion neurons that together form vast networks to relay electrical signals. A neuron receives electrical signals from other neurons via branch-like structures known as dendrites. The signals then travel into the cell body of the neuron. If their sum reaches a threshold, they fire a new signal through a single outgoing projection known as the axon, which is connected to the dendrites of other neurons. A single neuron has thousands of dendrites that each receive inputs from different axons, and it is thought that the strengthening and weakening of these dendritic connections enables us to learn and store memories. Dendrites are filled with molecules known as messenger ribonucleic acids (mRNAs) that act as templates to make proteins. Axonal signals reaching the dendrites can trigger these mRNAs to make new proteins that strengthen or weaken the connections between the two neurons, which is believed to be necessary for generating long-term memories. A protein called FMRP is found in both the cell body and dendrites and is able to bind to and regulate the ability of mRNAs to make proteins. A loss of the gene encoding FMRP is the most common cause of inherited intellectual disability and autism in humans, but it remains unclear precisely what role this protein plays in learning and memory. Hale et al. used genetic and bioinformatics approaches to specifically study mRNAs in the dendrites and the cell body of a specific type of neuron involved in memory in mice. The experiments revealed that FMRP played different roles in the dendrites and cell body. In the dendrites, FMRP interacted with mRNAs encoding proteins that can change how the neuron responds to a signal from a neighboring neuron and may alter how strong the connections between the neurons are. On the other hand, FMRP in the cell body modulated the activities of mRNAs encoding proteins that in turn regulate the activities of genes. These findings change the way we think about how memory may work by suggesting that groups of mRNAs encoding proteins with certain activities are found in distinct parts of a single neuron. These observations offer new ways to approach intellectual disabilities and autism spectrum disorder.


Subject(s)
Cell Body/physiology , Dendrites/physiology , Fragile X Mental Retardation Protein/genetics , Gene Expression Regulation , Pyramidal Cells/physiology , RNA, Messenger/genetics , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Pyramidal Cells/classification , Transcriptome
7.
PLoS One ; 16(6): e0252949, 2021.
Article in English | MEDLINE | ID: mdl-34170927

ABSTRACT

To address the need for simple, safe, sensitive, and scalable SARS-CoV-2 tests, we validated and implemented a PCR test that uses a saliva collection kit use at home. Individuals self-collected 300 µl saliva in vials containing Darnell Rockefeller University Laboratory (DRUL) buffer and extracted RNA was assayed by RT-PCR (the DRUL saliva assay). The limit of detection was confirmed to be 1 viral copy/µl in 20 of 20 replicate extractions. Viral RNA was stable in DRUL buffer at room temperature up to seven days after sample collection, and safety studies demonstrated that DRUL buffer immediately inactivated virus at concentrations up to 2.75x106 PFU/ml. Results from SARS-CoV-2 positive nasopharyngeal (NP) swab samples collected in viral transport media and assayed with a standard FDA Emergency Use Authorization (EUA) test were highly correlated with samples placed in DRUL buffer. Direct comparison of results from 162 individuals tested by FDA EUA oropharyngeal (OP) or NP swabs with co-collected saliva samples identified four otherwise unidentified positive cases in DRUL buffer. Over six months, we collected 3,724 samples from individuals ranging from 3 months to 92 years of age. This included collecting weekly samples over 10 weeks from teachers, children, and parents from a pre-school program, which allowed its safe reopening while at-risk pods were quarantined. In sum, we validated a simple, sensitive, stable, and safe PCR-based test using a self-collected saliva sample as a valuable tool for clinical diagnosis and screening at workplaces and schools.


Subject(s)
COVID-19 Nucleic Acid Testing , COVID-19 , SARS-CoV-2 , Saliva/virology , Schools , Specimen Handling , COVID-19/diagnosis , COVID-19/genetics , Child , Female , Humans , Male
8.
N Engl J Med ; 383(3): 218-228, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32668112

ABSTRACT

BACKGROUND: Rheumatoid arthritis, like many inflammatory diseases, is characterized by episodes of quiescence and exacerbation (flares). The molecular events leading to flares are unknown. METHODS: We established a clinical and technical protocol for repeated home collection of blood in patients with rheumatoid arthritis to allow for longitudinal RNA sequencing (RNA-seq). Specimens were obtained from 364 time points during eight flares over a period of 4 years in our index patient, as well as from 235 time points during flares in three additional patients. We identified transcripts that were differentially expressed before flares and compared these with data from synovial single-cell RNA-seq. Flow cytometry and sorted-blood-cell RNA-seq in additional patients were used to validate the findings. RESULTS: Consistent changes were observed in blood transcriptional profiles 1 to 2 weeks before a rheumatoid arthritis flare. B-cell activation was followed by expansion of circulating CD45-CD31-PDPN+ preinflammatory mesenchymal, or PRIME, cells in the blood from patients with rheumatoid arthritis; these cells shared features of inflammatory synovial fibroblasts. Levels of circulating PRIME cells decreased during flares in all 4 patients, and flow cytometry and sorted-cell RNA-seq confirmed the presence of PRIME cells in 19 additional patients with rheumatoid arthritis. CONCLUSIONS: Longitudinal genomic analysis of rheumatoid arthritis flares revealed PRIME cells in the blood during the period before a flare and suggested a model in which these cells become activated by B cells in the weeks before a flare and subsequently migrate out of the blood into the synovium. (Funded by the National Institutes of Health and others.).


Subject(s)
Arthritis, Rheumatoid/blood , B-Lymphocytes/physiology , Gene Expression , Mesenchymal Stem Cells , Sequence Analysis, RNA/methods , Adult , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Female , Fibroblasts/metabolism , Flow Cytometry , Humans , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Patient Acuity , Surveys and Questionnaires , Symptom Flare Up , Synovial Fluid/cytology
9.
Elife ; 82019 12 20.
Article in English | MEDLINE | ID: mdl-31860442

ABSTRACT

Loss of the RNA binding protein FMRP causes Fragile X Syndrome (FXS), the most common cause of inherited intellectual disability, yet it is unknown how FMRP function varies across brain regions and cell types and how this contributes to disease pathophysiology. Here we use conditional tagging of FMRP and CLIP (FMRP cTag CLIP) to examine FMRP mRNA targets in hippocampal CA1 pyramidal neurons, a critical cell type for learning and memory relevant to FXS phenotypes. Integrating these data with analysis of ribosome-bound transcripts in these neurons revealed CA1-enriched binding of autism-relevant mRNAs, and CA1-specific regulation of transcripts encoding circadian proteins. This contrasted with different targets in cerebellar granule neurons, and was consistent with circadian defects in hippocampus-dependent memory in Fmr1 knockout mice. These findings demonstrate differential FMRP-dependent regulation of mRNAs across neuronal cell types that may contribute to phenotypes such as memory defects and sleep disturbance associated with FXS.


Subject(s)
Autistic Disorder/metabolism , CA1 Region, Hippocampal/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Memory Disorders/genetics , Pyramidal Cells/metabolism , Animals , Autistic Disorder/genetics , Autistic Disorder/physiopathology , CA1 Region, Hippocampal/cytology , Cerebellum/cytology , Cerebellum/metabolism , Circadian Clocks/genetics , Circadian Clocks/physiology , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Gene Expression Regulation , Humans , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism
11.
Neuron ; 104(2): 256-270.e5, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31416668

ABSTRACT

Familial Alzheimer's disease (fAD) results from mutations in the amyloid precursor protein (APP) and presenilin (PSEN1 and PSEN2) genes. Here we leveraged recent advances in induced pluripotent stem cell (iPSC) and CRISPR/Cas9 genome editing technologies to generate a panel of isogenic knockin human iPSC lines carrying APP and/or PSEN1 mutations. Global transcriptomic and translatomic profiling revealed that fAD mutations have overlapping effects on the expression of AD-related and endocytosis-associated genes. Mutant neurons also increased Rab5+ early endosome size. APP and PSEN1 mutations had discordant effects on Aß production but similar effects on APP ß C-terminal fragments (ß-CTFs), which accumulate in all mutant neurons. Importantly, endosomal dysfunction correlated with accumulation of ß-CTFs, not Aß, and could be rescued by pharmacological modulation of ß-secretase (BACE). These data display the utility of our mutant iPSCs in studying AD-related phenotypes in a non-overexpression human-based system and support mounting evidence that ß-CTF may be critical in AD pathogenesis.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Endocytosis/genetics , Endosomes/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Presenilin-1/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , CRISPR-Cas Systems , Cell Line , Endosomes/pathology , Gene Expression Profiling , Gene Knock-In Techniques , Heterozygote , Homozygote , Humans , Induced Pluripotent Stem Cells , Mutation , Organelle Size , Phenotype , Proteomics , rab5 GTP-Binding Proteins/metabolism
12.
Cell Rep ; 28(4): 979-991.e6, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31340158

ABSTRACT

Post-transcriptional regulation by microRNAs (miRNAs) is essential for complex molecular responses to physiological insult and disease. Although many disease-associated miRNAs are known, their global targets and culminating network effects on pathophysiology remain poorly understood. We applied Argonaute (AGO) crosslinking immunoprecipitation (CLIP) to systematically elucidate altered miRNA-target interactions in brain following ischemia and reperfusion (I/R) injury. Among 1,190 interactions identified, the most prominent was the cumulative loss of target regulation by miR-29 family members. Integration of translational and time-course RNA profiles revealed a dynamic mode of miR-29 target de-regulation, led by acute translational activation and a later increase in RNA levels, allowing rapid proteomic changes to take effect. These functional regulatory events rely on canonical and non-canonical miR-29 binding and engage glutamate reuptake signals, such as glial glutamate transporter (GLT-1), to control local glutamate levels. These results uncover a miRNA target network that acts acutely to maintain brain homeostasis after ischemic stroke.


Subject(s)
Argonaute Proteins/metabolism , Brain Ischemia/metabolism , Brain/metabolism , Cross-Linking Reagents/chemistry , Glutamic Acid/metabolism , Homeostasis , Stroke/metabolism , Animals , Base Sequence , Brain Ischemia/complications , Brain Ischemia/genetics , Down-Regulation/genetics , Gene Regulatory Networks , Glucose/deficiency , Humans , Immunoprecipitation , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Neuroglia/metabolism , Oxygen , Polymorphism, Genetic , Signal Transduction , Stroke/complications , Stroke/genetics , Time Factors
13.
Nat Genet ; 51(6): 973-980, 2019 06.
Article in English | MEDLINE | ID: mdl-31133750

ABSTRACT

We address the challenge of detecting the contribution of noncoding mutations to disease with a deep-learning-based framework that predicts the specific regulatory effects and the deleterious impact of genetic variants. Applying this framework to 1,790 autism spectrum disorder (ASD) simplex families reveals a role in disease for noncoding mutations-ASD probands harbor both transcriptional- and post-transcriptional-regulation-disrupting de novo mutations of significantly higher functional impact than those in unaffected siblings. Further analysis suggests involvement of noncoding mutations in synaptic transmission and neuronal development and, taken together with previous studies, reveals a convergent genetic landscape of coding and noncoding mutations in ASD. We demonstrate that sequences carrying prioritized mutations identified in probands possess allele-specific regulatory activity, and we highlight a link between noncoding mutations and heterogeneity in the IQ of ASD probands. Our predictive genomics framework illuminates the role of noncoding mutations in ASD and prioritizes mutations with high impact for further study, and is broadly applicable to complex human diseases.


Subject(s)
Autism Spectrum Disorder/genetics , Deep Learning , Genetic Predisposition to Disease , Genome, Human , Genomics , Mutation , RNA, Untranslated , Algorithms , Alleles , Autism Spectrum Disorder/diagnosis , Computational Biology/methods , Gene Expression , Gene Expression Regulation , Genes, Reporter , Genetic Association Studies , Genomics/methods , Humans , Phenotype , RNA Processing, Post-Transcriptional , Transcription, Genetic
14.
Neuron ; 101(4): 707-720.e5, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30638744

ABSTRACT

RNA-binding proteins (RBPs) regulate genetic diversity, but the degree to which they do so in individual cell types in vivo is unknown. We developed NOVA2 cTag-crosslinking and immunoprecipitation (CLIP) to generate functional RBP-RNA maps from different neuronal populations in the mouse brain. Combining cell type datasets from Nova2-cTag and Nova2 conditional knockout mice revealed differential NOVA2 regulatory actions on alternative splicing (AS) on the same transcripts expressed in different neurons. This includes functional differences in transcripts expressed in cortical and cerebellar excitatory versus inhibitory neurons, where we find NOVA2 is required for, respectively, development of laminar structure, motor coordination, and synapse formation. We also find that NOVA2-regulated AS is coupled to NOVA2 regulation of intron retention in hundreds of transcripts, which can sequester the trans-acting splicing factor PTBP2. In summary, cTag-CLIP complements single-cell RNA sequencing (RNA-seq) studies by providing a means for understanding RNA regulation of functional cell diversity.


Subject(s)
Alternative Splicing , Antigens, Neoplasm/genetics , Cerebellum/embryology , Cerebral Cortex/embryology , Neurogenesis , Neurons/metabolism , RNA-Binding Proteins/genetics , Animals , Antigens, Neoplasm/metabolism , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiology , Cerebral Cortex/cytology , Excitatory Postsynaptic Potentials , Female , Inhibitory Postsynaptic Potentials , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuro-Oncological Ventral Antigen , Neurons/cytology , Neurons/physiology , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA-Binding Proteins/metabolism
15.
Elife ; 72018 05 31.
Article in English | MEDLINE | ID: mdl-29848443

ABSTRACT

Dynamic post-transcriptional control of RNA expression by RNA-binding proteins (RBPs) is critical during immune response. ZFP36 RBPs are prominent inflammatory regulators linked to autoimmunity and cancer, but functions in adaptive immunity are less clear. We used HITS-CLIP to define ZFP36 targets in mouse T cells, revealing unanticipated actions in regulating T-cell activation, proliferation, and effector functions. Transcriptome and ribosome profiling showed that ZFP36 represses mRNA target abundance and translation, notably through novel AU-rich sites in coding sequence. Functional studies revealed that ZFP36 regulates early T-cell activation kinetics cell autonomously, by attenuating activation marker expression, limiting T cell expansion, and promoting apoptosis. Strikingly, loss of ZFP36 in vivo accelerated T cell responses to acute viral infection and enhanced anti-viral immunity. These findings uncover a critical role for ZFP36 RBPs in restraining T cell expansion and effector functions, and suggest ZFP36 inhibition as a strategy to enhance immune-based therapies.


Subject(s)
Antiviral Agents/metabolism , Immunity , Lymphocyte Activation , RNA-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Tristetraprolin/metabolism , Animals , Base Sequence , Bone Marrow/virology , CD4-Positive T-Lymphocytes/metabolism , Cell Lineage , Kinetics , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Ribosomes/metabolism , Transcriptome/genetics , Tristetraprolin/genetics
16.
Elife ; 72018 03 26.
Article in English | MEDLINE | ID: mdl-29578408

ABSTRACT

Alternative polyadenylation (APA) regulates mRNA translation, stability, and protein localization. However, it is unclear to what extent APA regulates these processes uniquely in specific cell types. Using a new technique, cTag-PAPERCLIP, we discovered significant differences in APA between the principal types of mouse cerebellar neurons, the Purkinje and granule cells, as well as between proliferating and differentiated granule cells. Transcripts that differed in APA in these comparisons were enriched in key neuronal functions and many differed in coding sequence in addition to 3'UTR length. We characterize Memo1, a transcript that shifted from expressing a short 3'UTR isoform to a longer one during granule cell differentiation. We show that Memo1 regulates granule cell precursor proliferation and that its long 3'UTR isoform is targeted by miR-124, contributing to its downregulation during development. Our findings provide insight into roles for APA in specific cell types and establish a platform for further functional studies.


Subject(s)
3' Untranslated Regions , Neurons/physiology , Polyadenylation , Protein Biosynthesis , RNA Stability , RNA, Messenger/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cerebellum/cytology , Mice
17.
JCI Insight ; 2(21)2017 11 02.
Article in English | MEDLINE | ID: mdl-29093272

ABSTRACT

In the course of modeling the naturally occurring tumor immunity seen in patients with paraneoplastic cerebellar degeneration (PCD), we discovered an unexpectedly high threshold for breaking CD8+ cytotoxic T cell (CTL) tolerance to the PCD autoantigen, CDR2. While CDR2 expression was previously found to be strictly restricted to immune-privileged cells (cerebellum, testes, and tumors), unexpectedly we have found that T cells also express CDR2. This expression underlies inhibition of CTL activation; CTLs that respond to epithelial cells expressing CDR2 fail to respond to T cells expressing CDR2. This was a general phenomenon, as T cells presenting influenza (flu) antigen also fail to activate otherwise potent flu-specific CTLs either in vitro or in vivo. Moreover, transfer of flu peptide-pulsed T cells into flu-infected mice inhibits endogenous flu-specific CTLs. Our finding that T cells serve as a site of immune privilege, inhibiting effector CTL function, uncovers an autorepressive loop with general biologic and clinical relevance.


Subject(s)
Antigens, Viral/immunology , Autoantigens/immunology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Animals , Autoimmune Diseases/immunology , Epithelial Cells/metabolism , HeLa Cells , Humans , Immunization , Influenza A virus , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nervous System Diseases/immunology , Paraneoplastic Cerebellar Degeneration
18.
Neuron ; 95(6): 1334-1349.e5, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28910620

ABSTRACT

Alternative polyadenylation (APA) is increasingly recognized to regulate gene expression across different cell types, but obtaining APA maps from individual cell types typically requires prior purification, a stressful procedure that can itself alter cellular states. Here, we describe a new platform, cTag-PAPERCLIP, that generates APA profiles from single cell populations in intact tissues; cTag-PAPERCLIP requires no tissue dissociation and preserves transcripts in native states. Applying cTag-PAPERCLIP to profile four major cell types in the mouse brain revealed common APA preferences between excitatory and inhibitory neurons distinct from astrocytes and microglia, regulated in part by neuron-specific RNA-binding proteins NOVA2 and PTBP2. We further identified a role of APA in switching Araf protein isoforms during microglia activation, impacting production of downstream inflammatory cytokines. Our results demonstrate the broad applicability of cTag-PAPERCLIP and a previously undiscovered role of APA in contributing to protein diversity between different cell types and cellular states within the brain.


Subject(s)
Brain/cytology , Microglia/metabolism , Neurons/metabolism , Polyadenylation , Protein Serine-Threonine Kinases/metabolism , Animals , Antigens, Neoplasm/physiology , Astrocytes/metabolism , Brain/metabolism , Cells, Cultured , Female , Humans , Male , Mice , Microglia/cytology , Nerve Tissue Proteins/physiology , Neuro-Oncological Ventral Antigen , Organ Specificity , Polypyrimidine Tract-Binding Protein/physiology , Protein Isoforms/metabolism , RNA-Binding Proteins/physiology
19.
Sci Rep ; 7(1): 8190, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28811507

ABSTRACT

With roles in development, cell proliferation and disease, micro-RNA (miRNA) biology is of great importance and a potential therapeutic target. Here we used cross-linking immunoprecipitation (CLIP) and ligation of miRNA-target chimeras on the Argonaute (AGO) protein to globally map miRNA interactions in the cow. The interactome is the deepest reported to date. miRNA targeting principles are consistent with observations in other species, but with expanded pairing rules. Experimental mapping robustly predicted functional miR-17 regulatory sites. From miRNA-specific targeting for >5000 mRNAs we determined gene ontologies (GO). This confirmed repression of genes important for embryonic development and cell cycle progress by the let-7 family, and repression of those involved in cell cycle arrest by the miR-17 family, but also suggested a number of unappreciated miRNA functions. Our results provide a significant resource for understanding of bovine and species-conserved miRNA regulation, and demonstrate the power of experimental methods for establishing comprehensive interaction maps.


Subject(s)
Gene Expression Profiling , MicroRNAs/genetics , RNA Interference , RNA, Messenger/genetics , Transcriptome , 3' Untranslated Regions , Amino Acid Motifs , Animals , Cattle , Cell Line , Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Ontology , Humans , Molecular Sequence Annotation , Organ Specificity/genetics
20.
Genes Dev ; 31(10): 990-1006, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28637692

ABSTRACT

Understanding the biologic role of N6-methyladenosine (m6A) RNA modifications in mRNA requires an understanding of when and where in the life of a pre-mRNA transcript the modifications are made. We found that HeLa cell chromatin-associated nascent pre-mRNA (CA-RNA) contains many unspliced introns and m6A in exons but very rarely in introns. The m6A methylation is essentially completed upon the release of mRNA into the nucleoplasm. Furthermore, the content and location of each m6A modification in steady-state cytoplasmic mRNA are largely indistinguishable from those in the newly synthesized CA-RNA or nucleoplasmic mRNA. This result suggests that quantitatively little methylation or demethylation occurs in cytoplasmic mRNA. In addition, only ∼10% of m6As in CA-RNA are within 50 nucleotides of 5' or 3' splice sites, and the vast majority of exons harboring m6A in wild-type mouse stem cells is spliced the same in cells lacking the major m6A methyltransferase Mettl3. Both HeLa and mouse embryonic stem cell mRNAs harboring m6As have shorter half-lives, and thousands of these mRNAs have increased half-lives (twofold or more) in Mettl3 knockout cells compared with wild type. In summary, m6A is added to exons before or soon after exon definition in nascent pre-mRNA, and while m6A is not required for most splicing, its addition in the nascent transcript is a determinant of cytoplasmic mRNA stability.


Subject(s)
Cytoplasm/metabolism , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , Animals , Chromatin/metabolism , Embryonic Stem Cells , Exons/genetics , Gene Knockout Techniques , HeLa Cells , Humans , Introns/genetics , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...