Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.444
Filter
1.
Cell ; 186(4): 850-863.e16, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36803605

ABSTRACT

It is unknown whether pangolins, the most trafficked mammals, play a role in the zoonotic transmission of bat coronaviruses. We report the circulation of a novel MERS-like coronavirus in Malayan pangolins, named Manis javanica HKU4-related coronavirus (MjHKU4r-CoV). Among 86 animals, four tested positive by pan-CoV PCR, and seven tested seropositive (11 and 12.8%). Four nearly identical (99.9%) genome sequences were obtained, and one virus was isolated (MjHKU4r-CoV-1). This virus utilizes human dipeptidyl peptidase-4 (hDPP4) as a receptor and host proteases for cell infection, which is enhanced by a furin cleavage site that is absent in all known bat HKU4r-CoVs. The MjHKU4r-CoV-1 spike shows higher binding affinity for hDPP4, and MjHKU4r-CoV-1 has a wider host range than bat HKU4-CoV. MjHKU4r-CoV-1 is infectious and pathogenic in human airways and intestinal organs and in hDPP4-transgenic mice. Our study highlights the importance of pangolins as reservoir hosts of coronaviruses poised for human disease emergence.


Subject(s)
Coronavirus Infections , Coronavirus , Dipeptidyl Peptidase 4 , Pangolins , Animals , Humans , Mice , Chiroptera , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Endopeptidases/metabolism , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/metabolism , Peptide Hydrolases/metabolism , Receptors, Virus/metabolism , Virus Internalization , Coronavirus/physiology
2.
Cell ; 186(17): 3706-3725.e29, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37562402

ABSTRACT

The bone marrow in the skull is important for shaping immune responses in the brain and meninges, but its molecular makeup among bones and relevance in human diseases remain unclear. Here, we show that the mouse skull has the most distinct transcriptomic profile compared with other bones in states of health and injury, characterized by a late-stage neutrophil phenotype. In humans, proteome analysis reveals that the skull marrow is the most distinct, with differentially expressed neutrophil-related pathways and a unique synaptic protein signature. 3D imaging demonstrates the structural and cellular details of human skull-meninges connections (SMCs) compared with veins. Last, using translocator protein positron emission tomography (TSPO-PET) imaging, we show that the skull bone marrow reflects inflammatory brain responses with a disease-specific spatial distribution in patients with various neurological disorders. The unique molecular profile and anatomical and functional connections of the skull show its potential as a site for diagnosing, monitoring, and treating brain diseases.


Subject(s)
Bone Marrow , Nervous System Diseases , Skull , Animals , Humans , Mice , Bone Marrow/metabolism , Brain/diagnostic imaging , Brain/metabolism , Carrier Proteins/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Positron-Emission Tomography/methods , Receptors, GABA/metabolism , Skull/cytology , Skull/diagnostic imaging
3.
Nat Immunol ; 25(7): 1270-1282, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38877178

ABSTRACT

The relative and synergistic contributions of genetics and environment to interindividual immune response variation remain unclear, despite implications in evolutionary biology and medicine. Here we quantify interactive effects of genotype and environment on immune traits by investigating C57BL/6, 129S1 and PWK/PhJ inbred mice, rewilded in an outdoor enclosure and infected with the parasite Trichuris muris. Whereas cellular composition was shaped by interactions between genotype and environment, cytokine response heterogeneity including IFNγ concentrations was primarily driven by genotype with consequence on worm burden. In addition, we show that other traits, such as expression of CD44, were explained mostly by genetics on T cells, whereas expression of CD44 on B cells was explained more by environment across all strains. Notably, genetic differences under laboratory conditions were decreased following rewilding. These results indicate that nonheritable influences interact with genetic factors to shape immune variation and parasite burden.


Subject(s)
Gene-Environment Interaction , Mice, Inbred C57BL , Trichuriasis , Trichuris , Animals , Trichuris/immunology , Trichuriasis/immunology , Trichuriasis/parasitology , Mice , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , B-Lymphocytes/immunology , Genotype , Interferon-gamma/metabolism , T-Lymphocytes/immunology , Female , Male
4.
Cell ; 182(5): 1156-1169.e12, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32795415

ABSTRACT

Dysregulated microglia are intimately involved in neurodegeneration, including Alzheimer's disease (AD) pathogenesis, but the mechanisms controlling pathogenic microglial gene expression remain poorly understood. The transcription factor CCAAT/enhancer binding protein beta (c/EBPß) regulates pro-inflammatory genes in microglia and is upregulated in AD. We show expression of c/EBPß in microglia is regulated post-translationally by the ubiquitin ligase COP1 (also called RFWD2). In the absence of COP1, c/EBPß accumulates rapidly and drives a potent pro-inflammatory and neurodegeneration-related gene program, evidenced by increased neurotoxicity in microglia-neuronal co-cultures. Antibody blocking studies reveal that neurotoxicity is almost entirely attributable to complement. Remarkably, loss of a single allele of Cebpb prevented the pro-inflammatory phenotype. COP1-deficient microglia markedly accelerated tau-mediated neurodegeneration in a mouse model where activated microglia play a deleterious role. Thus, COP1 is an important suppressor of pathogenic c/EBPß-dependent gene expression programs in microglia.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Ligases/metabolism , Microglia/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/genetics , Alzheimer Disease/metabolism , Animals , Cell Line , Coculture Techniques/methods , Female , Gene Expression/physiology , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism
5.
Cell ; 182(1): 50-58.e8, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32516571

ABSTRACT

COVID-19 has spread worldwide since 2019 and is now a severe threat to public health. We previously identified the causative agent as a novel SARS-related coronavirus (SARS-CoV-2) that uses human angiotensin-converting enzyme 2 (hACE2) as the entry receptor. Here, we successfully developed a SARS-CoV-2 hACE2 transgenic mouse (HFH4-hACE2 in C3B6 mice) infection model. The infected mice generated typical interstitial pneumonia and pathology that were similar to those of COVID-19 patients. Viral quantification revealed the lungs as the major site of infection, although viral RNA could also be found in the eye, heart, and brain in some mice. Virus identical to SARS-CoV-2 in full-genome sequences was isolated from the infected lung and brain tissues. Last, we showed that pre-exposure to SARS-CoV-2 could protect mice from severe pneumonia. Our results show that the hACE2 mouse would be a valuable tool for testing potential vaccines and therapeutics.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/pathology , Disease Models, Animal , Mice, Transgenic , Pneumonia, Viral/pathology , Angiotensin-Converting Enzyme 2 , Animals , COVID-19 , Female , Humans , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/virology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic/genetics , Pandemics , Peptidyl-Dipeptidase A/genetics , SARS-CoV-2 , Viral Tropism , Weight Loss
6.
Cell ; 167(1): 122-132.e9, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27641505

ABSTRACT

A major determinant of mRNA half-life is the codon-dependent rate of translational elongation. How the processes of translational elongation and mRNA decay communicate is unclear. Here, we establish that the DEAD-box protein Dhh1p is a sensor of codon optimality that targets an mRNA for decay. First, we find mRNAs whose translation elongation rate is slowed by inclusion of non-optimal codons are specifically degraded in a Dhh1p-dependent manner. Biochemical experiments show Dhh1p is preferentially associated with mRNAs with suboptimal codon choice. We find these effects on mRNA decay are sensitive to the number of slow-moving ribosomes on an mRNA. Moreover, we find Dhh1p overexpression leads to the accumulation of ribosomes specifically on mRNAs (and even codons) of low codon optimality. Lastly, Dhh1p physically interacts with ribosomes in vivo. Together, these data argue that Dhh1p is a sensor for ribosome speed, targeting an mRNA for repression and subsequent decay.


Subject(s)
Codon/metabolism , DEAD-box RNA Helicases/metabolism , Protein Biosynthesis , RNA Stability , RNA, Messenger/metabolism , Ribosomes/metabolism , Codon/genetics , DEAD-box RNA Helicases/genetics , Half-Life
7.
Cell ; 160(6): 1111-24, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25768907

ABSTRACT

mRNA degradation represents a critical regulated step in gene expression. Although the major pathways in turnover have been identified, accounting for disparate half-lives has been elusive. We show that codon optimality is one feature that contributes greatly to mRNA stability. Genome-wide RNA decay analysis revealed that stable mRNAs are enriched in codons designated optimal, whereas unstable mRNAs contain predominately non-optimal codons. Substitution of optimal codons with synonymous, non-optimal codons results in dramatic mRNA destabilization, whereas the converse substitution significantly increases stability. Further, we demonstrate that codon optimality impacts ribosome translocation, connecting the processes of translation elongation and decay through codon optimality. Finally, we show that optimal codon content accounts for the similar stabilities observed in mRNAs encoding proteins with coordinated physiological function. This work demonstrates that codon optimization exists as a mechanism to finely tune levels of mRNAs and, ultimately, proteins.


Subject(s)
Codon , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/metabolism , Protein Biosynthesis , RNA Stability , RNA, Fungal/chemistry , RNA, Messenger/chemistry
8.
Cell ; 160(4): 619-630, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25679758

ABSTRACT

A central paradigm within virology is that each viral particle largely behaves as an independent infectious unit. Here, we demonstrate that clusters of enteroviral particles are packaged within phosphatidylserine (PS) lipid-enriched vesicles that are non-lytically released from cells and provide greater infection efficiency than free single viral particles. We show that vesicular PS lipids are co-factors to the relevant enterovirus receptors in mediating subsequent infectivity and transmission, in particular to primary human macrophages. We demonstrate that clustered packaging of viral particles within vesicles enables multiple viral RNA genomes to be collectively transferred into single cells. This study reveals a novel mode of viral transmission, where enteroviral genomes are transmitted from cell-to-cell en bloc in membrane-bound PS vesicles instead of as single independent genomes. This has implications for facilitating genetic cooperativity among viral quasispecies as well as enhancing viral replication.


Subject(s)
Cytoplasmic Vesicles/virology , Enterovirus Infections/transmission , Enterovirus/physiology , Macrophages/virology , Cytoplasmic Vesicles/chemistry , Humans , Macrophages/cytology , Phosphatidylserines , Poliovirus/physiology , RNA, Viral/metabolism , Rhinovirus/physiology , Virus Replication
9.
Nature ; 627(8004): 620-627, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38448595

ABSTRACT

The fungus Candida albicans frequently colonizes the human gastrointestinal tract, from which it can disseminate to cause systemic disease. This polymorphic species can transition between growing as single-celled yeast and as multicellular hyphae to adapt to its environment. The current dogma of C. albicans commensalism is that the yeast form is optimal for gut colonization, whereas hyphal cells are detrimental to colonization but critical for virulence1-3. Here, we reveal that this paradigm does not apply to multi-kingdom communities in which a complex interplay between fungal morphology and bacteria dictates C. albicans fitness. Thus, whereas yeast-locked cells outcompete wild-type cells when gut bacteria are absent or depleted by antibiotics, hyphae-competent wild-type cells outcompete yeast-locked cells in hosts with replete bacterial populations. This increased fitness of wild-type cells involves the production of hyphal-specific factors including the toxin candidalysin4,5, which promotes the establishment of colonization. At later time points, adaptive immunity is engaged, and intestinal immunoglobulin A preferentially selects against hyphal cells1,6. Hyphal morphotypes are thus under both positive and negative selective pressures in the gut. Our study further shows that candidalysin has a direct inhibitory effect on bacterial species, including limiting their metabolic output. We therefore propose that C. albicans has evolved hyphal-specific factors, including candidalysin, to better compete with bacterial species in the intestinal niche.


Subject(s)
Candida albicans , Fungal Proteins , Gastrointestinal Microbiome , Hyphae , Intestines , Mycotoxins , Symbiosis , Animals , Female , Humans , Male , Mice , Bacteria/growth & development , Bacteria/immunology , Candida albicans/growth & development , Candida albicans/immunology , Candida albicans/metabolism , Candida albicans/pathogenicity , Fungal Proteins/metabolism , Gastrointestinal Microbiome/immunology , Hyphae/growth & development , Hyphae/immunology , Hyphae/metabolism , Immunoglobulin A/immunology , Intestines/immunology , Intestines/microbiology , Mycotoxins/metabolism , Virulence
10.
Mol Cell ; 82(4): 716-727, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35016034

ABSTRACT

Protein acetylation is conserved across phylogeny and has been recognized as one of the most prominent post-translational modifications since its discovery nearly 60 years ago. Histone acetylation is an active mark characteristic of open chromatin, but acetylation on specific lysine residues and histone variants occurs in different biological contexts and can confer various outcomes. The significance of acetylation events is indicated by the associations of lysine acetyltransferases, deacetylases, and acetyl-lysine readers with developmental disorders and pathologies. Recent advances have uncovered new roles of acetylation regulators in chromatin-centric events, which emphasize the complexity of these functional networks. In this review, we discuss mechanisms and dynamics of acetylation in chromatin organization and DNA-templated processes, including gene transcription and DNA repair and replication.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Histones/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Chromatin/genetics , DNA Repair , DNA Replication , Genomic Instability , Histones/genetics , Humans , Lysine , Transcription, Genetic
11.
Mol Cell ; 81(16): 3386-3399.e10, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34265249

ABSTRACT

The super elongation complex (SEC) contains the positive transcription elongation factor b (P-TEFb) and the subcomplex ELL2-EAF1, which stimulates RNA polymerase II (RNA Pol II) elongation. Here, we report the cryoelectron microscopy (cryo-EM) structure of ELL2-EAF1 bound to a RNA Pol II elongation complex at 2.8 Å resolution. The ELL2-EAF1 dimerization module directly binds the RNA Pol II lobe domain, explaining how SEC delivers P-TEFb to RNA Pol II. The same site on the lobe also binds the initiation factor TFIIF, consistent with SEC binding only after the transition from transcription initiation to elongation. Structure-guided functional analysis shows that the stimulation of RNA elongation requires the dimerization module and the ELL2 linker that tethers the module to the RNA Pol II protrusion. Our results show that SEC stimulates elongation allosterically and indicate that this stimulation involves stabilization of a closed conformation of the RNA Pol II active center cleft.


Subject(s)
Positive Transcriptional Elongation Factor B/ultrastructure , RNA Polymerase II/genetics , Transcription Factors/genetics , Transcriptional Elongation Factors/genetics , Allosteric Regulation/genetics , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Cryoelectron Microscopy , Humans , Molecular Structure , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Positive Transcriptional Elongation Factor B/genetics , Protein Binding/genetics , Protein Conformation , RNA Polymerase II/ultrastructure , Transcription Elongation, Genetic , Transcription Factors/ultrastructure , Transcription, Genetic/genetics , Transcriptional Elongation Factors/ultrastructure
12.
Mol Cell ; 81(9): 1920-1934.e9, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33689748

ABSTRACT

Transcription by RNA polymerase II (Pol II) is coupled to pre-mRNA splicing, but the underlying mechanisms remain poorly understood. Co-transcriptional splicing requires assembly of a functional spliceosome on nascent pre-mRNA, but whether and how this influences Pol II transcription remains unclear. Here we show that inhibition of pre-mRNA branch site recognition by the spliceosome component U2 snRNP leads to a widespread and strong decrease in new RNA synthesis from human genes. Multiomics analysis reveals that inhibition of U2 snRNP function increases the duration of Pol II pausing in the promoter-proximal region, impairs recruitment of the pause release factor P-TEFb, and reduces Pol II elongation velocity at the beginning of genes. Our results indicate that efficient release of paused Pol II into active transcription elongation requires the formation of functional spliceosomes and that eukaryotic mRNA biogenesis relies on positive feedback from the splicing machinery to the transcription machinery.


Subject(s)
RNA Polymerase II/metabolism , RNA, Messenger/biosynthesis , Ribonucleoprotein, U2 Small Nuclear/metabolism , Spliceosomes/enzymology , Transcription Elongation, Genetic , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Feedback, Physiological , Gene Expression Regulation , HeLa Cells , Humans , K562 Cells , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/genetics , Time Factors
13.
Cell ; 152(1-2): 248-61, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332759

ABSTRACT

Establishment of oligodendrocyte identity is crucial for subsequent events of myelination in the CNS. Here, we demonstrate that activation of ATP-dependent SWI/SNF chromatin-remodeling enzyme Smarca4/Brg1 at the differentiation onset is necessary and sufficient to initiate and promote oligodendrocyte lineage progression and maturation. Genome-wide multistage studies by ChIP-seq reveal that oligodendrocyte-lineage determination factor Olig2 functions as a prepatterning factor to direct Smarca4/Brg1 to oligodendrocyte-specific enhancers. Recruitment of Smarca4/Brg1 to distinct subsets of myelination regulatory genes is developmentally regulated. Functional analyses of Smarca4/Brg1 and Olig2 co-occupancy relative to chromatin epigenetic marking uncover stage-specific cis-regulatory elements that predict sets of transcriptional regulators controlling oligodendrocyte differentiation. Together, our results demonstrate that regulation of the functional specificity and activity of a Smarca4/Brg1-dependent chromatin-remodeling complex by Olig2, coupled with transcriptionally linked chromatin modifications, is critical to precisely initiate and establish the transcriptional program that promotes oligodendrocyte differentiation and subsequent myelination of the CNS.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Nerve Tissue Proteins/metabolism , Oligodendroglia/cytology , Animals , Brain/cytology , Cells, Cultured , DNA Helicases/metabolism , Gene Expression Regulation , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism , Rats , Spinal Cord/cytology , Transcription Factors/metabolism
14.
Development ; 151(20)2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38572957

ABSTRACT

The ovarian microenvironment plays a crucial role in ensuring the reproductive success of viviparous teleosts. However, the molecular mechanism underlying the interaction between spermatozoa and the ovarian microenvironment has remained elusive. This study aimed to contribute to a better understanding of this process in black rockfish (Sebastes schlegelii) using integrated multi-omics approaches. The results demonstrated significant upregulation of ovarian complement-related proteins and pattern recognition receptors, along with remodeling of glycans on the surface of spermatozoa at the early spermatozoa-storage stage (1 month after mating). As spermatozoa were stored over time, ovarian complement proteins were progressively repressed by tryptophan and hippurate, indicating a remarkable adaptation of spermatozoa to the ovarian microenvironment. Before fertilization, a notable upregulation of cellular junction proteins was observed. The study revealed that spermatozoa bind to ZPB2a protein through GSTM3 and that ZPB2a promotes spermatozoa survival and movement in a GSTM3-dependent manner. These findings shed light on a key mechanism that influences the dynamics of spermatozoa in the female reproductive tract, providing valuable insights into the molecular networks regulating spermatozoa adaptation and survival in species with internal fertilization.


Subject(s)
Ovary , Spermatozoa , Animals , Male , Female , Spermatozoa/metabolism , Ovary/metabolism , Fertilization , Viviparity, Nonmammalian , Proteomics , Fish Proteins/metabolism , Fish Proteins/genetics , Fishes/metabolism , Cellular Microenvironment , Multiomics
15.
Nat Methods ; 21(7): 1349-1363, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849569

ABSTRACT

The Long-read RNA-Seq Genome Annotation Assessment Project Consortium was formed to evaluate the effectiveness of long-read approaches for transcriptome analysis. Using different protocols and sequencing platforms, the consortium generated over 427 million long-read sequences from complementary DNA and direct RNA datasets, encompassing human, mouse and manatee species. Developers utilized these data to address challenges in transcript isoform detection, quantification and de novo transcript detection. The study revealed that libraries with longer, more accurate sequences produce more accurate transcripts than those with increased read depth, whereas greater read depth improved quantification accuracy. In well-annotated genomes, tools based on reference sequences demonstrated the best performance. Incorporating additional orthogonal data and replicate samples is advised when aiming to detect rare and novel transcripts or using reference-free approaches. This collaborative study offers a benchmark for current practices and provides direction for future method development in transcriptome analysis.


Subject(s)
Gene Expression Profiling , RNA-Seq , Humans , Animals , Mice , RNA-Seq/methods , Gene Expression Profiling/methods , Transcriptome , Sequence Analysis, RNA/methods , Molecular Sequence Annotation/methods
16.
Nat Methods ; 20(8): 1187-1195, 2023 08.
Article in English | MEDLINE | ID: mdl-37308696

ABSTRACT

Most approaches to transcript quantification rely on fixed reference annotations; however, the transcriptome is dynamic and depending on the context, such static annotations contain inactive isoforms for some genes, whereas they are incomplete for others. Here we present Bambu, a method that performs machine-learning-based transcript discovery to enable quantification specific to the context of interest using long-read RNA-sequencing. To identify novel transcripts, Bambu estimates the novel discovery rate, which replaces arbitrary per-sample thresholds with a single, interpretable, precision-calibrated parameter. Bambu retains the full-length and unique read counts, enabling accurate quantification in presence of inactive isoforms. Compared to existing methods for transcript discovery, Bambu achieves greater precision without sacrificing sensitivity. We show that context-aware annotations improve quantification for both novel and known transcripts. We apply Bambu to quantify isoforms from repetitive HERVH-LTR7 retrotransposons in human embryonic stem cells, demonstrating the ability for context-specific transcript expression analysis.


Subject(s)
Gene Expression Profiling , Transcriptome , Humans , RNA-Seq , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Protein Isoforms/genetics
17.
Immunity ; 46(4): 675-689, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423341

ABSTRACT

Activated T cells produce reactive oxygen species (ROS), which trigger the antioxidative glutathione (GSH) response necessary to buffer rising ROS and prevent cellular damage. We report that GSH is essential for T cell effector functions through its regulation of metabolic activity. Conditional gene targeting of the catalytic subunit of glutamate cysteine ligase (Gclc) blocked GSH production specifically in murine T cells. Gclc-deficient T cells initially underwent normal activation but could not meet their increased energy and biosynthetic requirements. GSH deficiency compromised the activation of mammalian target of rapamycin-1 (mTOR) and expression of NFAT and Myc transcription factors, abrogating the energy utilization and Myc-dependent metabolic reprogramming that allows activated T cells to switch to glycolysis and glutaminolysis. In vivo, T-cell-specific ablation of murine Gclc prevented autoimmune disease but blocked antiviral defense. The antioxidative GSH pathway thus plays an unexpected role in metabolic integration and reprogramming during inflammatory T cell responses.


Subject(s)
Glutamate-Cysteine Ligase/deficiency , Glutathione/metabolism , Inflammation/metabolism , T-Lymphocytes/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Energy Metabolism/genetics , Glutamate-Cysteine Ligase/genetics , Glutamine/metabolism , Glycolysis , Immunoblotting , Inflammation/genetics , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism
18.
Nature ; 579(7798): 274-278, 2020 03.
Article in English | MEDLINE | ID: mdl-32103181

ABSTRACT

Despite the resounding clinical success in cancer treatment of antibodies that block the interaction of PD1 with its ligand PDL11, the mechanisms involved remain unknown. A major limitation to understanding the origin and fate of T cells in tumour immunity is the lack of quantitative information on the distribution of individual clonotypes of T cells in patients with cancer. Here, by performing deep single-cell sequencing of RNA and T cell receptors in patients with different types of cancer, we survey the profiles of various populations of T cells and T cell receptors in tumours, normal adjacent tissue, and peripheral blood. We find clear evidence of clonotypic expansion of effector-like T cells not only within the tumour but also in normal adjacent tissue. Patients with gene signatures of such clonotypic expansion respond best to anti-PDL1 therapy. Notably, expanded clonotypes found in the tumour and normal adjacent tissue can also typically be detected in peripheral blood, which suggests a convenient approach to patient identification. Analyses of our data together with several external datasets suggest that intratumoural T cells, especially in responsive patients, are replenished with fresh, non-exhausted replacement cells from sites outside the tumour, suggesting continued activity of the cancer immunity cycle in these patients, the acceleration of which may be associated with clinical response.


Subject(s)
Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/pathology , Pharmacogenomic Variants , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Clone Cells , Humans , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes/metabolism , Transcriptome
19.
Nature ; 579(7798): 270-273, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32015507

ABSTRACT

Since the outbreak of severe acute respiratory syndrome (SARS) 18 years ago, a large number of SARS-related coronaviruses (SARSr-CoVs) have been discovered in their natural reservoir host, bats1-4. Previous studies have shown that some bat SARSr-CoVs have the potential to infect humans5-7. Here we report the identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started on 12 December 2019, had caused 2,794 laboratory-confirmed infections including 80 deaths by 26 January 2020. Full-length genome sequences were obtained from five patients at an early stage of the outbreak. The sequences are almost identical and share 79.6% sequence identity to SARS-CoV. Furthermore, we show that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. Pairwise protein sequence analysis of seven conserved non-structural proteins domains show that this virus belongs to the species of SARSr-CoV. In addition, 2019-nCoV virus isolated from the bronchoalveolar lavage fluid of a critically ill patient could be neutralized by sera from several patients. Notably, we confirmed that 2019-nCoV uses the same cell entry receptor-angiotensin converting enzyme II (ACE2)-as SARS-CoV.


Subject(s)
Betacoronavirus/classification , Betacoronavirus/genetics , Chiroptera/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Disease Outbreaks , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , Angiotensin-Converting Enzyme 2 , Animals , Antibodies, Viral/blood , Betacoronavirus/metabolism , Betacoronavirus/ultrastructure , COVID-19 , Cell Line , China/epidemiology , Chlorocebus aethiops , Female , Genome, Viral/genetics , Humans , Male , Peptidyl-Dipeptidase A/metabolism , Phylogeny , Severe acute respiratory syndrome-related coronavirus/classification , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2 , Sequence Homology, Nucleic Acid , Severe Acute Respiratory Syndrome , Vero Cells
20.
Mol Cell ; 70(6): 1089-1100.e8, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29932902

ABSTRACT

Translation and decay of eukaryotic mRNAs is controlled by shortening of the poly(A) tail and release of the poly(A)-binding protein Pab1/PABP. The Ccr4-Not complex contains two exonucleases-Ccr4 and Caf1/Pop2-that mediate mRNA deadenylation. Here, using a fully reconstituted biochemical system with proteins from the fission yeast Schizosaccharomyces pombe, we show that Pab1 interacts with Ccr4-Not, stimulates deadenylation, and differentiates the roles of the nuclease enzymes. Surprisingly, Pab1 release relies on Ccr4 activity. In agreement with this, in vivo experiments in budding yeast show that Ccr4 is a general deadenylase that acts on all mRNAs. In contrast, Caf1 only trims poly(A) not bound by Pab1. As a consequence, Caf1 is a specialized deadenylase required for the selective deadenylation of transcripts with lower rates of translation elongation and reduced Pab1 occupancy. These findings reveal a coupling between the rates of translation and deadenylation that is dependent on Pab1 and Ccr4-Not.


Subject(s)
Exoribonucleases/metabolism , Poly(A)-Binding Protein I/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Cytoplasm/metabolism , Endonucleases/metabolism , Exoribonucleases/genetics , Poly A/metabolism , Polyadenylation , RNA Stability , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Ribonucleases/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL