Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.706
Filter
Add more filters

Publication year range
1.
Cell ; 187(14): 3638-3651.e18, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38838667

ABSTRACT

Telomere maintenance requires the extension of the G-rich telomeric repeat strand by telomerase and the fill-in synthesis of the C-rich strand by Polα/primase. At telomeres, Polα/primase is bound to Ctc1/Stn1/Ten1 (CST), a single-stranded DNA-binding complex. Like mutations in telomerase, mutations affecting CST-Polα/primase result in pathological telomere shortening and cause a telomere biology disorder, Coats plus (CP). We determined cryogenic electron microscopy structures of human CST bound to the shelterin heterodimer POT1/TPP1 that reveal how CST is recruited to telomeres by POT1. Our findings suggest that POT1 hinge phosphorylation is required for CST recruitment, and the complex is formed through conserved interactions involving several residues mutated in CP. Our structural and biochemical data suggest that phosphorylated POT1 holds CST-Polα/primase in an inactive, autoinhibited state until telomerase has extended the telomere ends. We propose that dephosphorylation of POT1 releases CST-Polα/primase into an active state that completes telomere replication through fill-in synthesis.


Subject(s)
DNA Polymerase I , Shelterin Complex , Telomere-Binding Proteins , Telomere , Humans , Cryoelectron Microscopy , DNA Polymerase I/metabolism , DNA Primase/metabolism , DNA Primase/genetics , Models, Molecular , Phosphorylation , Shelterin Complex/metabolism , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/metabolism
2.
Cell ; 187(10): 2393-2410.e14, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38653235

ABSTRACT

SARS-CoV-2 and other sarbecoviruses continue to threaten humanity, highlighting the need to characterize common mechanisms of viral immune evasion for pandemic preparedness. Cytotoxic lymphocytes are vital for antiviral immunity and express NKG2D, an activating receptor conserved among mammals that recognizes infection-induced stress ligands (e.g., MIC-A/B). We found that SARS-CoV-2 evades NKG2D recognition by surface downregulation of MIC-A/B via shedding, observed in human lung tissue and COVID-19 patient serum. Systematic testing of SARS-CoV-2 proteins revealed that ORF6, an accessory protein uniquely conserved among sarbecoviruses, was responsible for MIC-A/B downregulation via shedding. Further investigation demonstrated that natural killer (NK) cells efficiently killed SARS-CoV-2-infected cells and limited viral spread. However, inhibition of MIC-A/B shedding with a monoclonal antibody, 7C6, further enhanced NK-cell activity toward SARS-CoV-2-infected cells. Our findings unveil a strategy employed by SARS-CoV-2 to evade cytotoxic immunity, identify the culprit immunevasin shared among sarbecoviruses, and suggest a potential novel antiviral immunotherapy.


Subject(s)
COVID-19 , Immune Evasion , Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily K , SARS-CoV-2 , Humans , SARS-CoV-2/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , COVID-19/immunology , COVID-19/virology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Animals , Cytotoxicity, Immunologic , Down-Regulation , Lung/immunology , Lung/virology , Lung/pathology
3.
Cell ; 185(16): 2936-2951.e19, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35931021

ABSTRACT

We studied the prevalent cytotoxic CD8 T cell response mounted against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike glycoprotein269-277 epitope (sequence YLQPRTFLL) via the most frequent human leukocyte antigen (HLA) class I worldwide, HLA A∗02. The Spike P272L mutation that has arisen in at least 112 different SARS-CoV-2 lineages to date, including in lineages classified as "variants of concern," was not recognized by the large CD8 T cell response seen across cohorts of HLA A∗02+ convalescent patients and individuals vaccinated against SARS-CoV-2, despite these responses comprising of over 175 different individual T cell receptors. Viral escape at prevalent T cell epitopes restricted by high frequency HLAs may be particularly problematic when vaccine immunity is focused on a single protein such as SARS-CoV-2 Spike, providing a strong argument for inclusion of multiple viral proteins in next generation vaccines and highlighting the need for monitoring T cell escape in new SARS-CoV-2 variants.


Subject(s)
COVID-19 , SARS-CoV-2 , CD8-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , HLA-A Antigens , Histocompatibility Antigens Class I , Humans
4.
Cell ; 185(18): 3408-3425.e29, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35985322

ABSTRACT

Genetically encoded voltage indicators are emerging tools for monitoring voltage dynamics with cell-type specificity. However, current indicators enable a narrow range of applications due to poor performance under two-photon microscopy, a method of choice for deep-tissue recording. To improve indicators, we developed a multiparameter high-throughput platform to optimize voltage indicators for two-photon microscopy. Using this system, we identified JEDI-2P, an indicator that is faster, brighter, and more sensitive and photostable than its predecessors. We demonstrate that JEDI-2P can report light-evoked responses in axonal termini of Drosophila interneurons and the dendrites and somata of amacrine cells of isolated mouse retina. JEDI-2P can also optically record the voltage dynamics of individual cortical neurons in awake behaving mice for more than 30 min using both resonant-scanning and ULoVE random-access microscopy. Finally, ULoVE recording of JEDI-2P can robustly detect spikes at depths exceeding 400 µm and report voltage correlations in pairs of neurons.


Subject(s)
Microscopy , Neurons , Animals , Interneurons , Mice , Microscopy/methods , Neurons/physiology , Photons , Wakefulness
5.
Nat Immunol ; 25(8): 1411-1421, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38997431

ABSTRACT

A subset of individuals exposed to Mycobacterium tuberculosis (Mtb) that we refer to as 'resisters' (RSTR) show evidence of IFN-γ- T cell responses to Mtb-specific antigens despite serially negative results on clinical testing. Here we found that Mtb-specific T cells in RSTR were clonally expanded, confirming the priming of adaptive immune responses following Mtb exposure. RSTR CD4+ T cells showed enrichment of TH17 and regulatory T cell-like functional programs compared to Mtb-specific T cells from individuals with latent Mtb infection. Using public datasets, we showed that these TH17 cell-like functional programs were associated with lack of progression to active tuberculosis among South African adolescents with latent Mtb infection and with bacterial control in nonhuman primates. Our findings suggested that RSTR may successfully control Mtb following exposure and immune priming and established a set of T cell biomarkers to facilitate further study of this clinical phenotype.


Subject(s)
CD4-Positive T-Lymphocytes , Mycobacterium tuberculosis , Tuberculosis , Mycobacterium tuberculosis/immunology , Humans , Animals , Adolescent , Tuberculosis/immunology , Tuberculosis/microbiology , CD4-Positive T-Lymphocytes/immunology , Th17 Cells/immunology , Female , Macaca mulatta , Male , Phenotype , Interferon-gamma/metabolism , Interferon-gamma/immunology , Antigens, Bacterial/immunology , Latent Tuberculosis/immunology , Latent Tuberculosis/microbiology , South Africa , Young Adult , T-Lymphocytes, Regulatory/immunology , Adult
6.
Cell ; 184(20): 5179-5188.e8, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34499854

ABSTRACT

We present evidence for multiple independent origins of recombinant SARS-CoV-2 viruses sampled from late 2020 and early 2021 in the United Kingdom. Their genomes carry single-nucleotide polymorphisms and deletions that are characteristic of the B.1.1.7 variant of concern but lack the full complement of lineage-defining mutations. Instead, the remainder of their genomes share contiguous genetic variation with non-B.1.1.7 viruses circulating in the same geographic area at the same time as the recombinants. In four instances, there was evidence for onward transmission of a recombinant-origin virus, including one transmission cluster of 45 sequenced cases over the course of 2 months. The inferred genomic locations of recombination breakpoints suggest that every community-transmitted recombinant virus inherited its spike region from a B.1.1.7 parental virus, consistent with a transmission advantage for B.1.1.7's set of mutations.


Subject(s)
COVID-19/epidemiology , COVID-19/transmission , Pandemics , Recombination, Genetic , SARS-CoV-2/genetics , Base Sequence/genetics , COVID-19/virology , Computational Biology/methods , Gene Frequency , Genome, Viral , Genotype , Humans , Mutation , Phylogeny , Polymorphism, Single Nucleotide , United Kingdom/epidemiology , Whole Genome Sequencing/methods
7.
Cell ; 184(1): 64-75.e11, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33275900

ABSTRACT

Global dispersal and increasing frequency of the SARS-CoV-2 spike protein variant D614G are suggestive of a selective advantage but may also be due to a random founder effect. We investigate the hypothesis for positive selection of spike D614G in the United Kingdom using more than 25,000 whole genome SARS-CoV-2 sequences. Despite the availability of a large dataset, well represented by both spike 614 variants, not all approaches showed a conclusive signal of positive selection. Population genetic analysis indicates that 614G increases in frequency relative to 614D in a manner consistent with a selective advantage. We do not find any indication that patients infected with the spike 614G variant have higher COVID-19 mortality or clinical severity, but 614G is associated with higher viral load and younger age of patients. Significant differences in growth and size of 614G phylogenetic clusters indicate a need for continued study of this variant.


Subject(s)
Amino Acid Substitution , COVID-19/transmission , COVID-19/virology , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/genetics , Aspartic Acid/analysis , Aspartic Acid/genetics , COVID-19/epidemiology , Genome, Viral , Glycine/analysis , Glycine/genetics , Humans , Mutation , SARS-CoV-2/growth & development , United Kingdom/epidemiology , Virulence , Whole Genome Sequencing
8.
Nat Rev Mol Cell Biol ; 24(6): 430-447, 2023 06.
Article in English | MEDLINE | ID: mdl-36596869

ABSTRACT

Genes specifying long non-coding RNAs (lncRNAs) occupy a large fraction of the genomes of complex organisms. The term 'lncRNAs' encompasses RNA polymerase I (Pol I), Pol II and Pol III transcribed RNAs, and RNAs from processed introns. The various functions of lncRNAs and their many isoforms and interleaved relationships with other genes make lncRNA classification and annotation difficult. Most lncRNAs evolve more rapidly than protein-coding sequences, are cell type specific and regulate many aspects of cell differentiation and development and other physiological processes. Many lncRNAs associate with chromatin-modifying complexes, are transcribed from enhancers and nucleate phase separation of nuclear condensates and domains, indicating an intimate link between lncRNA expression and the spatial control of gene expression during development. lncRNAs also have important roles in the cytoplasm and beyond, including in the regulation of translation, metabolism and signalling. lncRNAs often have a modular structure and are rich in repeats, which are increasingly being shown to be relevant to their function. In this Consensus Statement, we address the definition and nomenclature of lncRNAs and their conservation, expression, phenotypic visibility, structure and functions. We also discuss research challenges and provide recommendations to advance the understanding of the roles of lncRNAs in development, cell biology and disease.


Subject(s)
RNA, Long Noncoding , RNA, Long Noncoding/genetics , Cell Nucleus/genetics , Chromatin/genetics , Regulatory Sequences, Nucleic Acid , RNA Polymerase II/genetics
9.
Nat Immunol ; 23(5): 802-813, 2022 05.
Article in English | MEDLINE | ID: mdl-35449416

ABSTRACT

Regulatory T (Treg) cells require (interleukin-2) IL-2 for their homeostasis by affecting their proliferation, survival and activation. Here we investigated transcriptional and epigenetic changes after acute, periodic and persistent IL-2 receptor (IL-2R) signaling in mouse peripheral Treg cells in vivo using IL-2 or the long-acting IL-2-based biologic mouse IL-2-CD25. We show that initially IL-2R-dependent STAT5 transcription factor-dependent pathways enhanced gene activation, chromatin accessibility and metabolic reprogramming to support Treg cell proliferation. Unexpectedly, at peak proliferation, less accessible chromatin prevailed and was associated with Treg cell contraction. Restimulation of IL-2R signaling after contraction activated signature IL-2-dependent genes and others associated with effector Treg cells, whereas genes associated with signal transduction were downregulated to somewhat temper expansion. Thus, IL-2R-dependent Treg cell homeostasis depends in part on a shift from more accessible chromatin and expansion to less accessible chromatin and contraction. Mouse IL-2-CD25 supported greater expansion and a more extensive transcriptional state than IL-2 in Treg cells, consistent with greater efficacy to control autoimmunity.


Subject(s)
Chromatin Assembly and Disassembly , Interleukin-2 , T-Lymphocytes, Regulatory , Animals , Chromatin/metabolism , Interleukin-2/metabolism , Mice , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Signal Transduction
10.
Immunity ; 57(3): 414-428, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38479359

ABSTRACT

Interleukin-2 (IL-2) is a critical cytokine for T cell peripheral tolerance and immunity. Here, we review how IL-2 interaction with the high-affinity IL-2 receptor (IL-2R) supports the development and homeostasis of regulatory T cells and contributes to the differentiation of helper, cytotoxic, and memory T cells. A critical element for each T cell population is the expression of CD25 (Il2rα), which heightens the receptor affinity for IL-2. Signaling through the high-affinity IL-2R also reinvigorates CD8+ exhausted T (Tex) cells in response to checkpoint blockade. We consider the molecular underpinnings reflecting how IL-2R signaling impacts these various T cell subsets and the implications for enhancing IL-2-dependent immunotherapy of autoimmunity, other inflammatory disorders, and cancer.


Subject(s)
Interleukin-2 , Neoplasms , Humans , Interleukin-2/metabolism , Autoimmunity , Receptors, Interleukin-2 , T-Lymphocyte Subsets
11.
Immunity ; 57(8): 1864-1877.e9, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39111315

ABSTRACT

Tumor-infiltrating lymphocyte (TIL) hypofunction contributes to the progression of advanced cancers and is a frequent target of immunotherapy. Emerging evidence indicates that metabolic insufficiency drives T cell hypofunction during tonic stimulation, but the signals that initiate metabolic reprogramming in this context are largely unknown. Here, we found that Meteorin-like (METRNL), a metabolically active cytokine secreted by immune cells in the tumor microenvironment (TME), induced bioenergetic failure of CD8+ T cells. METRNL was secreted by CD8+ T cells during repeated stimulation and acted via both autocrine and paracrine signaling. Mechanistically, METRNL increased E2F-peroxisome proliferator-activated receptor delta (PPARδ) activity, causing mitochondrial depolarization and decreased oxidative phosphorylation, which triggered a compensatory bioenergetic shift to glycolysis. Metrnl ablation or downregulation improved the metabolic fitness of CD8+ T cells and enhanced tumor control in several tumor models, demonstrating the translational potential of targeting the METRNL-E2F-PPARδ pathway to support bioenergetic fitness of CD8+ TILs.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Mitochondria , Tumor Microenvironment , CD8-Positive T-Lymphocytes/immunology , Animals , Mitochondria/metabolism , Mitochondria/immunology , Mice , Tumor Microenvironment/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Humans , Mice, Inbred C57BL , Cytokines/metabolism , Signal Transduction , Energy Metabolism , PPAR delta/metabolism , Cell Line, Tumor , Neoplasms/immunology , Glycolysis , Mice, Knockout , Oxidative Phosphorylation
12.
Nat Rev Mol Cell Biol ; 22(4): 283-298, 2021 04.
Article in English | MEDLINE | ID: mdl-33564154

ABSTRACT

The regulation of telomere length in mammals is crucial for chromosome end-capping and thus for maintaining genome stability and cellular lifespan. This process requires coordination between telomeric protein complexes and the ribonucleoprotein telomerase, which extends the telomeric DNA. Telomeric proteins modulate telomere architecture, recruit telomerase to accessible telomeres and orchestrate the conversion of the newly synthesized telomeric single-stranded DNA tail into double-stranded DNA. Dysfunctional telomere maintenance leads to telomere shortening, which causes human diseases including bone marrow failure, premature ageing and cancer. Recent studies provide new insights into telomerase-related interactions (the 'telomere replisome') and reveal new challenges for future telomere structural biology endeavours owing to the dynamic nature of telomere architecture and the great number of structures that telomeres form. In this Review, we discuss recently determined structures of the shelterin and CTC1-STN1-TEN1 (CST) complexes, how they may participate in the regulation of telomere replication and chromosome end-capping, and how disease-causing mutations in their encoding genes may affect specific functions. Major outstanding questions in the field include how all of the telomere components assemble relative to each other and how the switching between different telomere structures is achieved.


Subject(s)
Chromatin/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Animals , Chromosomes/metabolism , DNA/metabolism , Humans , Telomerase/metabolism
13.
Cell ; 175(1): 14-17, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30217358

ABSTRACT

This year's Lasker-Koshland Special Achievement Award is given to Joan Argetsinger Steitz for her RNA research discoveries and her exemplary international leadership.


Subject(s)
RNA/metabolism , RNA/physiology , Awards and Prizes , Biomedical Research , History, 21st Century , Humans , RNA/history , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/physiology
14.
Nat Immunol ; 21(4): 434-441, 2020 04.
Article in English | MEDLINE | ID: mdl-32205883

ABSTRACT

Adaptive evolution is a key feature of T cell immunity. During acute immune responses, T cells harboring high-affinity T cell antigen receptors (TCRs) are preferentially expanded, but whether affinity maturation by clonal selection continues through the course of chronic infections remains unresolved. Here we investigated the evolution of the TCR repertoire and its affinity during the course of infection with cytomegalovirus, which elicits large T cell populations in humans and mice. Using single-cell and bulk TCR sequencing and structural affinity analyses of cytomegalovirus-specific T cells, and through the generation and in vivo monitoring of defined TCR repertoires, we found that the immunodominance of high-affinity T cell clones declined during the chronic infection phase, likely due to cellular senescence. These data showed that under conditions of chronic antigen exposure, low-affinity TCRs preferentially expanded within the TCR repertoire, with implications for immunotherapeutic strategies.


Subject(s)
Cytomegalovirus Infections/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Cellular Senescence/immunology , Cytomegalovirus/immunology , Female , Humans , Mice , Mice, Inbred C57BL
15.
Cell ; 169(5): 775-776, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28525749

ABSTRACT

Classifying sensory experiences as either novel or familiar represents a fundamental challenge to neural processing. In this issue of Cell, Hattori et al. describe a circuit mechanism by which a novel stimulus that initially interests a fruit fly turns into a familiar one.


Subject(s)
Recognition, Psychology , Humans
16.
Cell ; 170(2): 249-259.e25, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28669536

ABSTRACT

Widespread resistance to first-line TB drugs is a major problem that will likely only be resolved through the development of new drugs with novel mechanisms of action. We have used structure-guided methods to develop a lead molecule that targets the thioesterase activity of polyketide synthase Pks13, an essential enzyme that forms mycolic acids, required for the cell wall of Mycobacterium tuberculosis. Our lead, TAM16, is a benzofuran class inhibitor of Pks13 with highly potent in vitro bactericidal activity against drug-susceptible and drug-resistant clinical isolates of M. tuberculosis. In multiple mouse models of TB infection, TAM16 showed in vivo efficacy equal to the first-line TB drug isoniazid, both as a monotherapy and in combination therapy with rifampicin. TAM16 has excellent pharmacological and safety profiles, and the frequency of resistance for TAM16 is ∼100-fold lower than INH, suggesting that it can be developed as a new antitubercular aimed at the acute infection. PAPERCLIP.


Subject(s)
Antitubercular Agents/pharmacology , Benzofurans/pharmacology , Drug Design , Drug Resistance, Bacterial , Mycobacterium tuberculosis/drug effects , Piperidines/pharmacology , Tuberculosis/microbiology , Animals , Antitubercular Agents/chemistry , Benzofurans/chemistry , Benzofurans/pharmacokinetics , Cell Line , Female , Mice , Mice, Inbred BALB C , Models, Molecular , Piperidines/chemistry , Piperidines/pharmacokinetics , Specific Pathogen-Free Organisms
18.
Immunity ; 55(9): 1732-1746.e5, 2022 09 13.
Article in English | MEDLINE | ID: mdl-35961317

ABSTRACT

Many immunocompromised patients mount suboptimal humoral immunity after SARS-CoV-2 mRNA vaccination. Here, we assessed the single-cell profile of SARS-CoV-2-specific T cells post-mRNA vaccination in healthy individuals and patients with various forms of immunodeficiencies. Impaired vaccine-induced cell-mediated immunity was observed in many immunocompromised patients, particularly in solid-organ transplant and chronic lymphocytic leukemia patients. Notably, individuals with an inherited lack of mature B cells, i.e., X-linked agammaglobulinemia (XLA) displayed highly functional spike-specific T cell responses. Single-cell RNA-sequencing further revealed that mRNA vaccination induced a broad functional spectrum of spike-specific CD4+ and CD8+ T cells in healthy individuals and patients with XLA. These responses were founded on polyclonal repertoires of CD4+ T cells and robust expansions of oligoclonal effector-memory CD45RA+ CD8+ T cells with stem-like characteristics. Collectively, our data provide the functional continuum of SARS-CoV-2-specific T cell responses post-mRNA vaccination, highlighting that cell-mediated immunity is of variable functional quality across immunodeficiency syndromes.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Viral , CD8-Positive T-Lymphocytes , COVID-19/prevention & control , Humans , Immunity, Humoral , RNA, Messenger/genetics , Syndrome , Vaccination , Viral Envelope Proteins
19.
Cell ; 166(5): 1188-1197.e9, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27523609

ABSTRACT

Telomerase maintains genome integrity by adding repetitive DNA sequences to the chromosome ends in actively dividing cells, including 90% of all cancer cells. Recruitment of human telomerase to telomeres occurs during S-phase of the cell cycle, but the molecular mechanism of the process is only partially understood. Here, we use CRISPR genome editing and single-molecule imaging to track telomerase trafficking in nuclei of living human cells. We demonstrate that telomerase uses three-dimensional diffusion to search for telomeres, probing each telomere thousands of times each S-phase but only rarely forming a stable association. Both the transient and stable association events depend on the direct interaction of the telomerase protein TERT with the telomeric protein TPP1. Our results reveal that telomerase recruitment to telomeres is driven by dynamic interactions between the rapidly diffusing telomerase and the chromosome end.


Subject(s)
Telomerase/metabolism , Telomere/enzymology , Active Transport, Cell Nucleus , Bacterial Proteins , CRISPR-Associated Protein 9 , Cell Line , Cell Nucleus/enzymology , Clustered Regularly Interspaced Short Palindromic Repeats , Coiled Bodies/enzymology , Endonucleases , Gene Editing , Genome, Human , HeLa Cells , Humans , Imaging, Three-Dimensional , Protein Domains , S Phase , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Shelterin Complex , Telomerase/chemistry , Telomere/chemistry , Telomere Homeostasis , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/metabolism
20.
Cell ; 166(1): 245-57, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27264607

ABSTRACT

A mechanistic understanding of neural computation requires determining how information is processed as it passes through neurons and across synapses. However, it has been challenging to measure membrane potential changes in axons and dendrites in vivo. We use in vivo, two-photon imaging of novel genetically encoded voltage indicators, as well as calcium imaging, to measure sensory stimulus-evoked signals in the Drosophila visual system with subcellular resolution. Across synapses, we find major transformations in the kinetics, amplitude, and sign of voltage responses to light. We also describe distinct relationships between voltage and calcium signals in different neuronal compartments, a substrate for local computation. Finally, we demonstrate that ON and OFF selectivity, a key feature of visual processing across species, emerges through the transformation of membrane potential into intracellular calcium concentration. By imaging voltage and calcium signals to map information flow with subcellular resolution, we illuminate where and how critical computations arise.


Subject(s)
Drosophila/physiology , Neurons/metabolism , Visual Pathways , Animals , Calcium/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Kinetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurites/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL