Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 334
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38996528

ABSTRACT

In developing brains, axons exhibit remarkable precision in selecting synaptic partners among many non-partner cells. Evolutionarily conserved teneurins are transmembrane proteins that instruct synaptic partner matching. However, how intracellular signaling pathways execute teneurins' functions is unclear. Here, we use in situ proximity labeling to obtain the intracellular interactome of a teneurin (Ten-m) in the Drosophila brain. Genetic interaction studies using quantitative partner matching assays in both olfactory receptor neurons (ORNs) and projection neurons (PNs) reveal a common pathway: Ten-m binds to and negatively regulates a RhoGAP, thus activating the Rac1 small GTPases to promote synaptic partner matching. Developmental analyses with single-axon resolution identify the cellular mechanism of synaptic partner matching: Ten-m signaling promotes local F-actin levels and stabilizes ORN axon branches that contact partner PN dendrites. Combining spatial proteomics and high-resolution phenotypic analyses, this study advanced our understanding of both cellular and molecular mechanisms of synaptic partner matching.

2.
Cell ; 186(15): 3307-3324.e30, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37385249

ABSTRACT

The ability to map trafficking for thousands of endogenous proteins at once in living cells would reveal biology currently invisible to both microscopy and mass spectrometry. Here, we report TransitID, a method for unbiased mapping of endogenous proteome trafficking with nanometer spatial resolution in living cells. Two proximity labeling (PL) enzymes, TurboID and APEX, are targeted to source and destination compartments, and PL with each enzyme is performed in tandem via sequential addition of their small-molecule substrates. Mass spectrometry identifies the proteins tagged by both enzymes. Using TransitID, we mapped proteome trafficking between cytosol and mitochondria, cytosol and nucleus, and nucleolus and stress granules (SGs), uncovering a role for SGs in protecting the transcription factor JUN from oxidative stress. TransitID also identifies proteins that signal intercellularly between macrophages and cancer cells. TransitID offers a powerful approach for distinguishing protein populations based on compartment or cell type of origin.


Subject(s)
Mitochondria , Proteome , Proteome/metabolism , Mitochondria/metabolism , Cell Nucleolus/metabolism , Mass Spectrometry/methods , Gene Expression Regulation
3.
Cell ; 186(22): 4834-4850.e23, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37794589

ABSTRACT

Regulation of viral RNA biogenesis is fundamental to productive SARS-CoV-2 infection. To characterize host RNA-binding proteins (RBPs) involved in this process, we biochemically identified proteins bound to genomic and subgenomic SARS-CoV-2 RNAs. We find that the host protein SND1 binds the 5' end of negative-sense viral RNA and is required for SARS-CoV-2 RNA synthesis. SND1-depleted cells form smaller replication organelles and display diminished virus growth kinetics. We discover that NSP9, a viral RBP and direct SND1 interaction partner, is covalently linked to the 5' ends of positive- and negative-sense RNAs produced during infection. These linkages occur at replication-transcription initiation sites, consistent with NSP9 priming viral RNA synthesis. Mechanistically, SND1 remodels NSP9 occupancy and alters the covalent linkage of NSP9 to initiating nucleotides in viral RNA. Our findings implicate NSP9 in the initiation of SARS-CoV-2 RNA synthesis and unravel an unsuspected role of a cellular protein in orchestrating viral RNA production.


Subject(s)
COVID-19 , RNA, Viral , Humans , COVID-19/metabolism , Endonucleases/metabolism , RNA, Viral/metabolism , SARS-CoV-2/genetics , Virus Replication
4.
Cell ; 186(18): 3945-3967.e26, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37582358

ABSTRACT

Post-translational modifications (PTMs) play key roles in regulating cell signaling and physiology in both normal and cancer cells. Advances in mass spectrometry enable high-throughput, accurate, and sensitive measurement of PTM levels to better understand their role, prevalence, and crosstalk. Here, we analyze the largest collection of proteogenomics data from 1,110 patients with PTM profiles across 11 cancer types (10 from the National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium [CPTAC]). Our study reveals pan-cancer patterns of changes in protein acetylation and phosphorylation involved in hallmark cancer processes. These patterns revealed subsets of tumors, from different cancer types, including those with dysregulated DNA repair driven by phosphorylation, altered metabolic regulation associated with immune response driven by acetylation, affected kinase specificity by crosstalk between acetylation and phosphorylation, and modified histone regulation. Overall, this resource highlights the rich biology governed by PTMs and exposes potential new therapeutic avenues.


Subject(s)
Neoplasms , Protein Processing, Post-Translational , Proteomics , Humans , Acetylation , Histones/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Proteomics/methods
5.
Cell ; 185(12): 2035-2056.e33, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35688132

ABSTRACT

Alpha-synuclein (αS) is a conformationally plastic protein that reversibly binds to cellular membranes. It aggregates and is genetically linked to Parkinson's disease (PD). Here, we show that αS directly modulates processing bodies (P-bodies), membraneless organelles that function in mRNA turnover and storage. The N terminus of αS, but not other synucleins, dictates mutually exclusive binding either to cellular membranes or to P-bodies in the cytosol. αS associates with multiple decapping proteins in close proximity on the Edc4 scaffold. As αS pathologically accumulates, aberrant interaction with Edc4 occurs at the expense of physiologic decapping-module interactions. mRNA decay kinetics within PD-relevant pathways are correspondingly disrupted in PD patient neurons and brain. Genetic modulation of P-body components alters αS toxicity, and human genetic analysis lends support to the disease-relevance of these interactions. Beyond revealing an unexpected aspect of αS function and pathology, our data highlight the versatility of conformationally plastic proteins with high intrinsic disorder.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , Parkinson Disease/metabolism , Processing Bodies , RNA Stability , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
6.
Cell ; 184(15): 3962-3980.e17, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34171305

ABSTRACT

T cell-mediated immunity plays an important role in controlling SARS-CoV-2 infection, but the repertoire of naturally processed and presented viral epitopes on class I human leukocyte antigen (HLA-I) remains uncharacterized. Here, we report the first HLA-I immunopeptidome of SARS-CoV-2 in two cell lines at different times post infection using mass spectrometry. We found HLA-I peptides derived not only from canonical open reading frames (ORFs) but also from internal out-of-frame ORFs in spike and nucleocapsid not captured by current vaccines. Some peptides from out-of-frame ORFs elicited T cell responses in a humanized mouse model and individuals with COVID-19 that exceeded responses to canonical peptides, including some of the strongest epitopes reported to date. Whole-proteome analysis of infected cells revealed that early expressed viral proteins contribute more to HLA-I presentation and immunogenicity. These biological insights, as well as the discovery of out-of-frame ORF epitopes, will facilitate selection of peptides for immune monitoring and vaccine development.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Open Reading Frames/genetics , Peptides/immunology , Proteome/immunology , SARS-CoV-2/immunology , A549 Cells , Alleles , Amino Acid Sequence , Animals , Antigen Presentation/immunology , COVID-19/immunology , COVID-19/virology , Female , HEK293 Cells , Humans , Kinetics , Male , Mice , Peptides/chemistry , T-Lymphocytes/immunology
7.
Cell ; 180(2): 373-386.e15, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31955847

ABSTRACT

Molecular interactions at the cellular interface mediate organized assembly of single cells into tissues and, thus, govern the development and physiology of multicellular organisms. Here, we developed a cell-type-specific, spatiotemporally resolved approach to profile cell-surface proteomes in intact tissues. Quantitative profiling of cell-surface proteomes of Drosophila olfactory projection neurons (PNs) in pupae and adults revealed global downregulation of wiring molecules and upregulation of synaptic molecules in the transition from developing to mature PNs. A proteome-instructed in vivo screen identified 20 cell-surface molecules regulating neural circuit assembly, many of which belong to evolutionarily conserved protein families not previously linked to neural development. Genetic analysis further revealed that the lipoprotein receptor LRP1 cell-autonomously controls PN dendrite targeting, contributing to the formation of a precise olfactory map. These findings highlight the power of temporally resolved in situ cell-surface proteomic profiling in discovering regulators of brain wiring.


Subject(s)
Olfactory Pathways/metabolism , Olfactory Receptor Neurons/metabolism , Proteomics/methods , Animals , Axons/metabolism , Brain/metabolism , Dendrites/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/metabolism , Neurogenesis/physiology , Olfactory Nerve/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/physiology , Receptors, Lipoprotein/metabolism , Smell/physiology
8.
Cell ; 183(5): 1436-1456.e31, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33212010

ABSTRACT

The integration of mass spectrometry-based proteomics with next-generation DNA and RNA sequencing profiles tumors more comprehensively. Here this "proteogenomics" approach was applied to 122 treatment-naive primary breast cancers accrued to preserve post-translational modifications, including protein phosphorylation and acetylation. Proteogenomics challenged standard breast cancer diagnoses, provided detailed analysis of the ERBB2 amplicon, defined tumor subsets that could benefit from immune checkpoint therapy, and allowed more accurate assessment of Rb status for prediction of CDK4/6 inhibitor responsiveness. Phosphoproteomics profiles uncovered novel associations between tumor suppressor loss and targetable kinases. Acetylproteome analysis highlighted acetylation on key nuclear proteins involved in the DNA damage response and revealed cross-talk between cytoplasmic and mitochondrial acetylation and metabolism. Our results underscore the potential of proteogenomics for clinical investigation of breast cancer through more accurate annotation of targetable pathways and biological features of this remarkably heterogeneous malignancy.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Molecular Targeted Therapy , Proteogenomics , APOBEC Deaminases/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Cohort Studies , DNA Damage , DNA Repair , Female , Humans , Immunotherapy , Metabolomics , Middle Aged , Mutagenesis/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Receptor, ErbB-2/metabolism , Retinoblastoma Protein/metabolism , Tumor Microenvironment/immunology
9.
Nat Immunol ; 23(1): 109-121, 2022 01.
Article in English | MEDLINE | ID: mdl-34937919

ABSTRACT

Anemia is a major comorbidity in aging, chronic kidney and inflammatory diseases, and hematologic malignancies. However, the transcriptomic networks governing hematopoietic differentiation in blood cell development remain incompletely defined. Here we report that the atypical kinase RIOK2 (right open reading frame kinase 2) is a master transcription factor (TF) that not only drives erythroid differentiation, but also simultaneously suppresses megakaryopoiesis and myelopoiesis in primary human stem and progenitor cells. Our study reveals the previously uncharacterized winged helix-turn-helix DNA-binding domain and two transactivation domains of RIOK2 that are critical to regulate key hematopoietic TFs GATA1, GATA2, SPI1, RUNX3 and KLF1. This establishes RIOK2 as an integral component of the transcriptional regulatory network governing human hematopoietic differentiation. Importantly, RIOK2 mRNA expression significantly correlates with these TFs and other hematopoietic genes in myelodysplastic syndromes, acute myeloid leukemia and chronic kidney disease. Further investigation of RIOK2-mediated transcriptional pathways should yield therapeutic approaches to correct defective hematopoiesis in hematologic disorders.


Subject(s)
Blood Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Erythropoiesis/physiology , Gene Expression Regulation/physiology , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/metabolism , Myelopoiesis/physiology , Transcription Factors/metabolism , Transcription, Genetic/physiology
10.
Nat Immunol ; 22(4): 520-529, 2021 04.
Article in English | MEDLINE | ID: mdl-33753942

ABSTRACT

Patients with myelodysplastic syndromes (MDSs) display severe anemia but the mechanisms underlying this phenotype are incompletely understood. Right open-reading-frame kinase 2 (RIOK2) encodes a protein kinase located at 5q15, a region frequently lost in patients with MDS del(5q). Here we show that hematopoietic cell-specific haploinsufficient deletion of Riok2 (Riok2f/+Vav1cre) led to reduced erythroid precursor frequency leading to anemia. Proteomic analysis of Riok2f/+Vav1cre erythroid precursors suggested immune system activation, and transcriptomic analysis revealed an increase in p53-dependent interleukin (IL)-22 in Riok2f/+Vav1cre CD4+ T cells (TH22). Further, we discovered that the IL-22 receptor, IL-22RA1, was unexpectedly present on erythroid precursors. Blockade of IL-22 signaling alleviated anemia not only in Riok2f/+Vav1cre mice but also in wild-type mice. Serum concentrations of IL-22 were increased in the subset of patients with del(5q) MDS as well as patients with anemia secondary to chronic kidney disease. This work reveals a possible therapeutic opportunity for reversing many stress-induced anemias by targeting IL-22 signaling.


Subject(s)
Anemia/metabolism , Antibodies, Neutralizing/pharmacology , Erythroid Cells/metabolism , Erythropoiesis/drug effects , Interleukins/antagonists & inhibitors , Myelodysplastic Syndromes/drug therapy , Receptors, Interleukin/metabolism , Anemia/blood , Anemia/immunology , Anemia/prevention & control , Animals , Cells, Cultured , Cellular Microenvironment , Disease Models, Animal , Erythroid Cells/immunology , Humans , Interleukins/immunology , Interleukins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , Receptors, Interleukin/genetics , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Interleukin-22
11.
Cell ; 173(1): 90-103.e19, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29551269

ABSTRACT

Blood cell formation is classically thought to occur through a hierarchical differentiation process, although recent studies have shown that lineage commitment may occur earlier in hematopoietic stem and progenitor cells (HSPCs). The relevance to human blood diseases and the underlying regulation of these refined models remain poorly understood. By studying a genetic blood disorder, Diamond-Blackfan anemia (DBA), where the majority of mutations affect ribosomal proteins and the erythroid lineage is selectively perturbed, we are able to gain mechanistic insight into how lineage commitment is programmed normally and disrupted in disease. We show that in DBA, the pool of available ribosomes is limited, while ribosome composition remains constant. Surprisingly, this global reduction in ribosome levels more profoundly alters translation of a select subset of transcripts. We show how the reduced translation of select transcripts in HSPCs can impair erythroid lineage commitment, illuminating a regulatory role for ribosome levels in cellular differentiation.


Subject(s)
Anemia, Diamond-Blackfan/pathology , Ribosomes/metabolism , 5' Untranslated Regions , Anemia, Diamond-Blackfan/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bone Marrow Cells/metabolism , Cells, Cultured , Female , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Male , Mutation, Missense , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Immunity ; 56(7): 1681-1698.e13, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37301199

ABSTRACT

CD4+ T cell responses are exquisitely antigen specific and directed toward peptide epitopes displayed by human leukocyte antigen class II (HLA-II) on antigen-presenting cells. Underrepresentation of diverse alleles in ligand databases and an incomplete understanding of factors affecting antigen presentation in vivo have limited progress in defining principles of peptide immunogenicity. Here, we employed monoallelic immunopeptidomics to identify 358,024 HLA-II binders, with a particular focus on HLA-DQ and HLA-DP. We uncovered peptide-binding patterns across a spectrum of binding affinities and enrichment of structural antigen features. These aspects underpinned the development of context-aware predictor of T cell antigens (CAPTAn), a deep learning model that predicts peptide antigens based on their affinity to HLA-II and full sequence of their source proteins. CAPTAn was instrumental in discovering prevalent T cell epitopes from bacteria in the human microbiome and a pan-variant epitope from SARS-CoV-2. Together CAPTAn and associated datasets present a resource for antigen discovery and the unraveling genetic associations of HLA alleles with immunopathologies.


Subject(s)
COVID-19 , Deep Learning , Humans , Captan , SARS-CoV-2 , HLA Antigens , Epitopes, T-Lymphocyte , Peptides
13.
Cell ; 170(1): 199-212.e20, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28666119

ABSTRACT

Type 2 diabetes (T2D) affects Latinos at twice the rate seen in populations of European descent. We recently identified a risk haplotype spanning SLC16A11 that explains ∼20% of the increased T2D prevalence in Mexico. Here, through genetic fine-mapping, we define a set of tightly linked variants likely to contain the causal allele(s). We show that variants on the T2D-associated haplotype have two distinct effects: (1) decreasing SLC16A11 expression in liver and (2) disrupting a key interaction with basigin, thereby reducing cell-surface localization. Both independent mechanisms reduce SLC16A11 function and suggest SLC16A11 is the causal gene at this locus. To gain insight into how SLC16A11 disruption impacts T2D risk, we demonstrate that SLC16A11 is a proton-coupled monocarboxylate transporter and that genetic perturbation of SLC16A11 induces changes in fatty acid and lipid metabolism that are associated with increased T2D risk. Our findings suggest that increasing SLC16A11 function could be therapeutically beneficial for T2D. VIDEO ABSTRACT.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Basigin/metabolism , Cell Membrane/metabolism , Chromosomes, Human, Pair 17/metabolism , Gene Knockdown Techniques , Haplotypes , Hepatocytes/metabolism , Heterozygote , Histone Code , Humans , Liver/metabolism , Models, Molecular , Monocarboxylic Acid Transporters/chemistry
14.
Mol Cell ; 84(12): 2238-2254.e11, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38870936

ABSTRACT

Transcriptional coregulators and transcription factors (TFs) contain intrinsically disordered regions (IDRs) that are critical for their association and function in gene regulation. More recently, IDRs have been shown to promote multivalent protein-protein interactions between coregulators and TFs to drive their association into condensates. By contrast, here we demonstrate how the IDR of the corepressor LSD1 excludes TF association, acting as a dynamic conformational switch that tunes repression of active cis-regulatory elements. Hydrogen-deuterium exchange shows that the LSD1 IDR interconverts between transient open and closed conformational states, the latter of which inhibits partitioning of the protein's structured domains with TF condensates. This autoinhibitory switch controls leukemic differentiation by modulating repression of active cis-regulatory elements bound by LSD1 and master hematopoietic TFs. Together, these studies unveil alternative mechanisms by which disordered regions and their dynamic crosstalk with structured regions can shape coregulator-TF interactions to control cis-regulatory landscapes and cell fate.


Subject(s)
Enhancer Elements, Genetic , Histone Demethylases , Histone Demethylases/metabolism , Histone Demethylases/genetics , Humans , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/chemistry , Transcription Factors/metabolism , Transcription Factors/genetics , Animals , Protein Binding , Mice , Cell Differentiation , Gene Silencing
15.
Cell ; 166(5): 1295-1307.e21, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565350

ABSTRACT

Cellular compartments that cannot be biochemically isolated are challenging to characterize. Here we demonstrate the proteomic characterization of the synaptic clefts that exist at both excitatory and inhibitory synapses. Normal brain function relies on the careful balance of these opposing neural connections, and understanding how this balance is achieved relies on knowledge of their protein compositions. Using a spatially restricted enzymatic tagging strategy, we mapped the proteomes of two of the most common excitatory and inhibitory synaptic clefts in living neurons. These proteomes reveal dozens of synaptic candidates and assign numerous known synaptic proteins to a specific cleft type. The molecular differentiation of each cleft allowed us to identify Mdga2 as a potential specificity factor influencing Neuroligin-2's recruitment of presynaptic neurotransmitters at inhibitory synapses.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , GABAergic Neurons/metabolism , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Proteome/metabolism , Synaptic Membranes/metabolism , Animals , Antigens, CD/metabolism , Glutamic Acid/metabolism , HEK293 Cells , Humans , Mice , Neural Cell Adhesion Molecules/metabolism , Peroxidase/genetics , Peroxidase/metabolism , Proteomics , Rats , Receptors, GABA/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thalamus/metabolism
16.
Cell ; 162(3): 675-86, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26189680

ABSTRACT

Finding the components of cellular circuits and determining their functions systematically remains a major challenge in mammalian cells. Here, we introduced genome-wide pooled CRISPR-Cas9 libraries into dendritic cells (DCs) to identify genes that control the induction of tumor necrosis factor (Tnf) by bacterial lipopolysaccharide (LPS), a key process in the host response to pathogens, mediated by the Tlr4 pathway. We found many of the known regulators of Tlr4 signaling, as well as dozens of previously unknown candidates that we validated. By measuring protein markers and mRNA profiles in DCs that are deficient in known or candidate genes, we classified the genes into three functional modules with distinct effects on the canonical responses to LPS and highlighted functions for the PAF complex and oligosaccharyltransferase (OST) complex. Our findings uncover new facets of innate immune circuits in primary cells and provide a genetic approach for dissection of mammalian cell circuits.


Subject(s)
CRISPR-Cas Systems , Genetic Techniques , Immunity, Innate , Animals , Bone Marrow Cells/immunology , Cell Differentiation , Cell Survival , Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Knockout Techniques , Gene Regulatory Networks , Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/immunology
18.
Cell ; 155(6): 1409-21, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24269006

ABSTRACT

N(6)-methyladenosine (m(6)A) is the most ubiquitous mRNA base modification, but little is known about its precise location, temporal dynamics, and regulation. Here, we generated genomic maps of m(6)A sites in meiotic yeast transcripts at nearly single-nucleotide resolution, identifying 1,308 putatively methylated sites within 1,183 transcripts. We validated eight out of eight methylation sites in different genes with direct genetic analysis, demonstrated that methylated sites are significantly conserved in a related species, and built a model that predicts methylated sites directly from sequence. Sites vary in their methylation profiles along a dense meiotic time course and are regulated both locally, via predictable methylatability of each site, and globally, through the core meiotic circuitry. The methyltransferase complex components localize to the yeast nucleolus, and this localization is essential for mRNA methylation. Our data illuminate a conserved, dynamically regulated methylation program in yeast meiosis and provide an important resource for studying the function of this epitranscriptomic modification.


Subject(s)
Meiosis , RNA, Fungal/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces/cytology , Saccharomyces/metabolism , Adenosine/analogs & derivatives , Adenosine/analysis , Adenosine/metabolism , Cell Nucleolus/metabolism , Genome, Fungal , Methylation , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , tRNA Methyltransferases/metabolism
19.
Nature ; 605(7910): 532-538, 2022 05.
Article in English | MEDLINE | ID: mdl-35508657

ABSTRACT

Within the tumour microenvironment, CD4+ T cells can promote or suppress antitumour responses through the recognition of antigens presented by human leukocyte antigen (HLA) class II molecules1,2, but how cancers co-opt these physiologic processes to achieve immune evasion remains incompletely understood. Here we performed in-depth analysis of the phenotype and tumour specificity of CD4+ T cells infiltrating human melanoma specimens, finding that exhausted cytotoxic CD4+ T cells could be directly induced by melanoma cells through recognition of HLA class II-restricted neoantigens, and also HLA class I-restricted tumour-associated antigens. CD4+ T regulatory (TReg) cells could be indirectly elicited through presentation of tumour antigens via antigen-presenting cells. Notably, numerous tumour-reactive CD4+ TReg clones were stimulated directly by HLA class II-positive melanoma and demonstrated specificity for melanoma neoantigens. This phenomenon was observed in the presence of an extremely high tumour neoantigen load, which we confirmed to be associated with HLA class II positivity through the analysis of 116 melanoma specimens. Our data reveal the landscape of infiltrating CD4+ T cells in melanoma and point to the presentation of HLA class II-restricted neoantigens and direct engagement of immunosuppressive CD4+ TReg cells as a mechanism of immune evasion that is favoured in HLA class II-positive melanoma.


Subject(s)
Antigens, Neoplasm , CD4-Positive T-Lymphocytes , Melanoma , Skin Neoplasms , Antigen-Presenting Cells , Antigens, Neoplasm/immunology , HLA Antigens , Humans , Melanoma/immunology , Phenotype , Skin Neoplasms/immunology , Tumor Cells, Cultured , Tumor Microenvironment
20.
Immunity ; 48(6): 1183-1194.e5, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29802019

ABSTRACT

HIV-1 infection of CD4+ T cells leads to cytopathic effects and cell demise, which is counter to the observation that certain HIV-1-infected cells possess a remarkable long-term stability and can persist lifelong in infected individuals treated with suppressive antiretroviral therapy (ART). Using quantitative mass spectrometry-based proteomics, we showed that HIV-1 infection activated cellular survival programs that were governed by BIRC5, a molecular inhibitor of cell apoptosis that is frequently overexpressed in malignant cells. BIRC5 and its upstream regulator OX40 were upregulated in productively and latently infected CD4+ T cells and were functionally involved in maintaining their viability. Moreover, OX40-expressing CD4+ T cells from ART-treated patients were enriched for clonally expanded HIV-1 sequences, and pharmacological inhibition of BIRC5 resulted in a selective decrease of HIV-1-infected cells in vitro. Together, these findings suggest that BIRC5 supports long-term survival of HIV-1-infected cells and may lead to clinical strategies to reduce persisting viral reservoirs.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Survivin/metabolism , Virus Latency/physiology , Adult , Aged , Apoptosis , Cell Survival/physiology , Female , HIV Infections/metabolism , HIV Infections/virology , HIV-1 , Humans , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL