Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
Science ; 383(6685): eadi3808, 2024 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-38386728

RÉSUMÉ

Cancer risk is influenced by inherited mutations, DNA replication errors, and environmental factors. However, the influence of genetic variation in immunosurveillance on cancer risk is not well understood. Leveraging population-level data from the UK Biobank and FinnGen, we show that heterozygosity at the human leukocyte antigen (HLA)-II loci is associated with reduced lung cancer risk in smokers. Fine-mapping implicated amino acid heterozygosity in the HLA-II peptide binding groove in reduced lung cancer risk, and single-cell analyses showed that smoking drives enrichment of proinflammatory lung macrophages and HLA-II+ epithelial cells. In lung cancer, widespread loss of HLA-II heterozygosity (LOH) favored loss of alleles with larger neopeptide repertoires. Thus, our findings nominate genetic variation in immunosurveillance as a critical risk factor for lung cancer.


Sujet(s)
Prédisposition génétique à une maladie , Antigènes d'histocompatibilité de classe II , Surveillance immunologique , Perte d'hétérozygotie , Tumeurs du poumon , Humains , Antigènes d'histocompatibilité de classe II/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Macrophages alvéolaires/immunologie , Facteurs de risque , Fumer/immunologie , Surveillance immunologique/génétique , Adulte d'âge moyen , Sujet âgé , Sujet âgé de 80 ans ou plus , Cartographie chromosomique , Polymorphisme de nucléotide simple
2.
bioRxiv ; 2024 Jan 26.
Article de Anglais | MEDLINE | ID: mdl-38328184

RÉSUMÉ

Generation of functional CD8 + T cell memory typically requires engagement of CD4 + T cells. However, in certain scenarios, such as acutely-resolving viral infections, effector (T E ) and subsequent memory (T M ) CD8 + T cell formation appear impervious to a lack of CD4 + T cell help during priming. Nonetheless, such "helpless" CD8 + T M respond poorly to pathogen rechallenge. At present, the origin and long-term evolution of helpless CD8 + T cell memory remain incompletely understood. Here, we demonstrate that helpless CD8 + T E differentiation is largely normal but a multiplicity of helpless CD8 T M defects, consistent with impaired memory maturation, emerge as a consequence of prolonged yet finite exposure to cognate antigen. Importantly, these defects resolve over time leading to full restoration of CD8 + T M potential and recall capacity. Our findings provide a unified explanation for helpless CD8 + T cell memory and emphasize an unexpected CD8 + T M plasticity with implications for vaccination strategies and beyond.

3.
Nature ; 625(7993): 166-174, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38057662

RÉSUMÉ

Myeloid cells are known to suppress antitumour immunity1. However, the molecular drivers of immunosuppressive myeloid cell states are not well defined. Here we used single-cell RNA sequencing of human and mouse non-small cell lung cancer (NSCLC) lesions, and found that in both species the type 2 cytokine interleukin-4 (IL-4) was predicted to be the primary driver of the tumour-infiltrating monocyte-derived macrophage phenotype. Using a panel of conditional knockout mice, we found that only deletion of the IL-4 receptor IL-4Rα in early myeloid progenitors in bone marrow reduced tumour burden, whereas deletion of IL-4Rα in downstream mature myeloid cells had no effect. Mechanistically, IL-4 derived from bone marrow basophils and eosinophils acted on granulocyte-monocyte progenitors to transcriptionally programme the development of immunosuppressive tumour-promoting myeloid cells. Consequentially, depletion of basophils profoundly reduced tumour burden and normalized myelopoiesis. We subsequently initiated a clinical trial of the IL-4Rα blocking antibody dupilumab2-5 given in conjunction with PD-1/PD-L1 checkpoint blockade in patients with relapsed or refractory NSCLC who had progressed on PD-1/PD-L1 blockade alone (ClinicalTrials.gov identifier NCT05013450 ). Dupilumab supplementation reduced circulating monocytes, expanded tumour-infiltrating CD8 T cells, and in one out of six patients, drove a near-complete clinical response two months after treatment. Our study defines a central role for IL-4 in controlling immunosuppressive myelopoiesis in cancer, identifies a novel combination therapy for immune checkpoint blockade in humans, and highlights cancer as a systemic malady that requires therapeutic strategies beyond the primary disease site.


Sujet(s)
Moelle osseuse , Carcinogenèse , Interleukine-4 , Myélopoïèse , Transduction du signal , Animaux , Humains , Souris , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/métabolisme , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/thérapie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Inhibiteurs de points de contrôle immunitaires/immunologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Interleukine-4/métabolisme , Tumeurs du poumon/immunologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/thérapie , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/immunologie , Monocytes/effets des médicaments et des substances chimiques , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Récidive , Transduction du signal/effets des médicaments et des substances chimiques
4.
Immunity ; 56(7): 1502-1514.e8, 2023 07 11.
Article de Anglais | MEDLINE | ID: mdl-37160117

RÉSUMÉ

Glial cells and central nervous system (CNS)-infiltrating leukocytes contribute to multiple sclerosis (MS). However, the networks that govern crosstalk among these ontologically distinct populations remain unclear. Here, we show that, in mice and humans, CNS-resident astrocytes and infiltrating CD44hiCD4+ T cells generated interleukin-3 (IL-3), while microglia and recruited myeloid cells expressed interleukin-3 receptor-ɑ (IL-3Rɑ). Astrocytic and T cell IL-3 elicited an immune migratory and chemotactic program by IL-3Rɑ+ myeloid cells that enhanced CNS immune cell infiltration, exacerbating MS and its preclinical model. Multiregional snRNA-seq of human CNS tissue revealed the appearance of IL3RA-expressing myeloid cells with chemotactic programming in MS plaques. IL3RA expression by plaque myeloid cells and IL-3 amount in the cerebrospinal fluid predicted myeloid and T cell abundance in the CNS and correlated with MS severity. Our findings establish IL-3:IL-3RA as a glial-peripheral immune network that prompts immune cell recruitment to the CNS and worsens MS.


Sujet(s)
Sclérose en plaques , Animaux , Humains , Souris , Système nerveux central , Interleukine-3 , Microglie , Névroglie/métabolisme
5.
Immunity ; 56(4): 783-796.e7, 2023 04 11.
Article de Anglais | MEDLINE | ID: mdl-36827982

RÉSUMÉ

Diet profoundly influences physiology. Whereas over-nutrition elevates risk for disease via its influence on immunity and metabolism, caloric restriction and fasting appear to be salutogenic. Despite multiple correlations observed between diet and health, the underlying biology remains unclear. Here, we identified a fasting-induced switch in leukocyte migration that prolongs monocyte lifespan and alters susceptibility to disease in mice. We show that fasting during the active phase induced the rapid return of monocytes from the blood to the bone marrow. Monocyte re-entry was orchestrated by hypothalamic-pituitary-adrenal (HPA) axis-dependent release of corticosterone, which augmented the CXCR4 chemokine receptor. Although the marrow is a safe haven for monocytes during nutrient scarcity, re-feeding prompted mobilization culminating in monocytosis of chronologically older and transcriptionally distinct monocytes. These shifts altered response to infection. Our study shows that diet-in particular, a diet's temporal dynamic balance-modulates monocyte lifespan with consequences for adaptation to external stressors.


Sujet(s)
Moelle osseuse , Monocytes , Souris , Animaux , Cellules de la moelle osseuse , Jeûne , Chimiokines/métabolisme
6.
Cell ; 185(7): 1223-1239.e20, 2022 03 31.
Article de Anglais | MEDLINE | ID: mdl-35290801

RÉSUMÉ

While CRISPR screens are helping uncover genes regulating many cell-intrinsic processes, existing approaches are suboptimal for identifying extracellular gene functions, particularly in the tissue context. Here, we developed an approach for spatial functional genomics called Perturb-map. We applied Perturb-map to knock out dozens of genes in parallel in a mouse model of lung cancer and simultaneously assessed how each knockout influenced tumor growth, histopathology, and immune composition. Moreover, we paired Perturb-map and spatial transcriptomics for unbiased analysis of CRISPR-edited tumors. We found that in Tgfbr2 knockout tumors, the tumor microenvironment (TME) was converted to a fibro-mucinous state, and T cells excluded, concomitant with upregulated TGFß and TGFß-mediated fibroblast activation, indicating that TGFß-receptor loss on cancer cells increased TGFß bioavailability and its immunosuppressive effects on the TME. These studies establish Perturb-map for functional genomics within the tissue at single-cell resolution with spatial architecture preserved and provide insight into how TGFß responsiveness of cancer cells can affect the TME.


Sujet(s)
Tumeurs , Microenvironnement tumoral , Animaux , Clustered regularly interspaced short palindromic repeats/génétique , Génomique , Souris , Tumeurs/génétique , Facteur de croissance transformant bêta/génétique
7.
Front Immunol ; 12: 757804, 2021.
Article de Anglais | MEDLINE | ID: mdl-34630437

RÉSUMÉ

Defective DNA mismatch repair (dMMR) is associated with many cancer types including colon, gastric, endometrial, ovarian, hepatobiliary tract, urinary tract, brain and skin cancers. Lynch syndrome - a hereditary cause of dMMR - confers increased lifetime risk of malignancy in different organs and tissues. These Lynch syndrome pathogenic alleles are widely present in humans at a 1:320 population frequency of a single allele and associated with an up to 80% risk of developing microsatellite unstable cancer (microsatellite instability - high, or MSI-H). Advanced MSI-H tumors can be effectively treated with checkpoint inhibitors (CPI), however, that has led to response rates of only 30-60% despite their high tumor mutational burden and favorable immune gene signatures in the tumor microenvironment (TME). We and others have characterized a subset of MSI-H associated highly recurrent frameshift mutations that yield shared immunogenic neoantigens. These frameshifts might serve as targets for off-the-shelf cancer vaccine designs. In this review we discuss the current state of research around MSI-H cancer vaccine development, its application to MSI-H and Lynch syndrome cancer patients and the utility of MSI-H as a biomarker for CPI therapy. We also summarize the tumor intrinsic mechanisms underlying the high occurrence rates of certain frameshifts in MSI-H. Finally, we provide an overview of pivotal clinical trials investigating MSI-H as a biomarker for CPI therapy and MSI-H vaccines. Overall, this review aims to inform the development of novel research paradigms and therapeutics.


Sujet(s)
Vaccins anticancéreux , Tumeurs colorectales héréditaires sans polypose/génétique , Réparation de mésappariement de l'ADN/génétique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Instabilité des microsatellites , Science biomédicale translationnelle/tendances , Marqueurs biologiques tumoraux , Vaccins anticancéreux/usage thérapeutique , Essais cliniques comme sujet , Tumeurs colorectales héréditaires sans polypose/immunologie , Tumeurs colorectales héréditaires sans polypose/prévention et contrôle , Tumeurs colorectales héréditaires sans polypose/thérapie , Prise en charge de la maladie , Repositionnement des médicaments , Résistance aux substances/génétique , Mutation avec décalage du cadre de lecture , Humains , Mutation de type INDEL , Modèles génétiques , Modèles immunologiques
8.
Elife ; 102021 06 02.
Article de Anglais | MEDLINE | ID: mdl-34075878

RÉSUMÉ

High spliceosome activity is a dependency for cancer cells, making them more vulnerable to perturbation of the splicing machinery compared to normal cells. To identify splicing factors important for prostate cancer (PCa) fitness, we performed pooled shRNA screens in vitro and in vivo. Our screens identified heterogeneous nuclear ribonucleoprotein M (HNRNPM) as a regulator of PCa cell growth. RNA- and eCLIP-sequencing identified HNRNPM binding to transcripts of key homeostatic genes. HNRNPM binding to its targets prevents aberrant exon inclusion and backsplicing events. In both linear and circular mis-spliced transcripts, HNRNPM preferentially binds to GU-rich elements in long flanking proximal introns. Mimicry of HNRNPM-dependent linear-splicing events using splice-switching-antisense-oligonucleotides was sufficient to inhibit PCa cell growth. This suggests that PCa dependence on HNRNPM is likely a result of mis-splicing of key homeostatic coding and non-coding genes. Our results have further been confirmed in other solid tumors. Taken together, our data reveal a role for HNRNPM in supporting cancer cell fitness. Inhibition of HNRNPM activity is therefore a potential therapeutic strategy in suppressing growth of PCa and other solid tumors.


Sujet(s)
Adénocarcinome/métabolisme , Prolifération cellulaire , Ribonucléoprotéine nucléaire hétérogène du groupe M/métabolisme , Tumeurs de la prostate/métabolisme , Épissage des ARN , ARN circulaire/biosynthèse , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Animaux , Régulation de l'expression des gènes tumoraux , Cellules HepG2 , Ribonucléoprotéine nucléaire hétérogène du groupe M/génétique , Humains , Mâle , Souris SCID , Cellules PC-3 , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , ARN circulaire/génétique , Charge tumorale , Cellules cancéreuses en culture
9.
Cell ; 183(6): 1634-1649.e17, 2020 12 10.
Article de Anglais | MEDLINE | ID: mdl-33259803

RÉSUMÉ

Microsatellite instability-high (MSI-H) tumors are characterized by high tumor mutation burden and responsiveness to checkpoint blockade. We identified tumor-specific frameshifts encoding multiple epitopes that originated from indel mutations shared among patients with MSI-H endometrial, colorectal, and stomach cancers. Epitopes derived from these shared frameshifts have high population occurrence rates, wide presence in many tumor subclones, and are predicted to bind to the most frequent MHC alleles in MSI-H patient cohorts. Neoantigens arising from these mutations are distinctly unlike self and viral antigens, signifying novel groups of potentially highly immunogenic tumor antigens. We further confirmed the immunogenicity of frameshift peptides in T cell stimulation experiments using blood mononuclear cells isolated from both healthy donors and MSI-H cancer patients. Our study uncovers the widespread occurrence and strong immunogenicity of tumor-specific antigens derived from shared frameshift mutations in MSI-H cancer and Lynch syndrome patients, suitable for the design of common "off-the-shelf" cancer vaccines.


Sujet(s)
Épitopes/génétique , Épitopes/immunologie , Mutation avec décalage du cadre de lecture/génétique , Instabilité des microsatellites , Tumeurs/génétique , Tumeurs/immunologie , Séquence d'acides aminés , Antigènes néoplasiques/immunologie , Antigènes viraux/immunologie , Lignée cellulaire tumorale , Analyse de mutations d'ADN , Régulation de l'expression des gènes tumoraux , Génome humain , Humains , Immunothérapie , Mutation faux-sens/génétique , Tumeurs/thérapie , Peptides/composition chimique , Peptides/immunologie , Analyse de survie , Lymphocytes T/immunologie
10.
Cell ; 181(7): 1502-1517.e23, 2020 06 25.
Article de Anglais | MEDLINE | ID: mdl-32559462

RÉSUMÉ

RNA viruses are a major human health threat. The life cycles of many highly pathogenic RNA viruses like influenza A virus (IAV) and Lassa virus depends on host mRNA, because viral polymerases cleave 5'-m7G-capped host transcripts to prime viral mRNA synthesis ("cap-snatching"). We hypothesized that start codons within cap-snatched host transcripts could generate chimeric human-viral mRNAs with coding potential. We report the existence of this mechanism of gene origination, which we named "start-snatching." Depending on the reading frame, start-snatching allows the translation of host and viral "untranslated regions" (UTRs) to create N-terminally extended viral proteins or entirely novel polypeptides by genetic overprinting. We show that both types of chimeric proteins are made in IAV-infected cells, generate T cell responses, and contribute to virulence. Our results indicate that during infection with IAV, and likely a multitude of other human, animal and plant viruses, a host-dependent mechanism allows the genesis of hybrid genes.


Sujet(s)
Coiffes des ARN/génétique , Infections à virus à ARN/génétique , Protéines de fusion recombinantes/génétique , Régions 5' non traduites/génétique , Animaux , Bovins , Lignée cellulaire , Cricetinae , Chiens , Humains , Virus de la grippe A/métabolisme , Souris , Protéines chimériques mutantes/génétique , Protéines chimériques mutantes/métabolisme , Cadres ouverts de lecture/génétique , Coiffes des ARN/métabolisme , Infections à virus à ARN/métabolisme , Virus à ARN/génétique , ARN messager/génétique , ARN messager/métabolisme , ARN viral/métabolisme , RNA replicase/génétique , RNA replicase/métabolisme , Protéines de fusion recombinantes/métabolisme , Transcription génétique/génétique , Protéines virales/métabolisme , Réplication virale/génétique
11.
Front Immunol ; 11: 27, 2020.
Article de Anglais | MEDLINE | ID: mdl-32117226

RÉSUMÉ

Tumor progression is typically accompanied by an accumulation of driver and passenger somatic mutations. A handful of those mutations occur in protein coding genes which introduce non-synonymous polymorphisms. Certain substitutions may give rise to novel, tumor-associated antigens or neoantigens, presentable by cancer cells to the host adaptive immune system. As antigen recognition is the core of an effective immune response, the identification of patient tumor specific antigens derived from transformed cells is of importance for immunotherapeutic approaches. Recent technological advances in DNA sequencing of tumor genomes, advances in gene expression analysis, algorithm development for antigen predictions and methods for T-cell receptor (TCR) repertoire sequencing have facilitated the selection of candidate immunogenic neoantigens. In this regard, multiple research groups have reported encouraging results of neoantigen-based cancer vaccines that generate tumor antigen specific immune responses, both in mouse models and clinical trials. Additionally, both the quantity and quality of neoantigens has been shown to have predictive value for clinical outcomes in checkpoint-blockade immunotherapy in certain tumor types. Neoantigen recognition by vaccination or through adoptive T cell therapy may have unprecedented potential to advance cancer immunotherapy in combination with other approaches. In our review we discuss three parameters regarding neoantigens: computational methods for epitope prediction, experimental methods for epitope immunogenicity validation and future directions for improvement of those methods. Within each section, we will describe the advantages and limitations of existing methods as well as highlight pressing fundamental problems to be addressed.


Sujet(s)
Antigènes néoplasiques/immunologie , Vaccins anticancéreux/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie adoptive/méthodes , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Vaccination/méthodes , Animaux , Antigènes néoplasiques/génétique , Vaccins anticancéreux/immunologie , Modèles animaux de maladie humaine , Épitopes/immunologie , Humains , Immunogénicité des vaccins , Souris , Mutation , Tumeurs/génétique ,
12.
Cancer Discov ; 9(9): 1192-1207, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31266769

RÉSUMÉ

Somatic frameshift mutations in the calreticulin (CALR) gene are key drivers of cellular transformation in myeloproliferative neoplasms (MPN). All patients carrying these mutations (CALR + MPN) share an identical sequence in the C-terminus of the mutated CALR protein (mut-CALR), with the potential for utility as a shared neoantigen. Here, we demonstrate that although a subset of patients with CALR + MPN develop specific T-cell responses against the mut-CALR C-terminus, PD-1 or CTLA4 expression abrogates the full complement of responses. Significantly, blockade of PD-1 and CLTA4 ex vivo by mAbs and of PD-1 in vivo by pembrolizumab administration restores mut-CALR-specific T-cell immunity in some patients with CALR + MPN. Moreover, mut-CALR elicits antigen-specific responses from both CD4+ and CD8+ T cells, confirming its broad applicability as an immunogen. Collectively, these results establish mut-CALR as a shared, MPN-specific neoantigen and inform the design of novel immunotherapies targeting mut-CALR. SIGNIFICANCE: Current treatment modalities for MPN are not effective in eliminating malignant cells. Here, we show that mutations in the CALR gene, which drive transformation in MPN, elicit T-cell responses that can be further enhanced by checkpoint blockade, suggesting immunotherapies could be employed to eliminate CALR + malignant cells in MPN.This article is highlighted in the In This Issue feature, p. 1143.


Sujet(s)
Anticorps monoclonaux humanisés/administration et posologie , Antinéoplasiques immunologiques/administration et posologie , Calréticuline/génétique , Syndromes myéloprolifératifs/traitement médicamenteux , Lymphocytes T/métabolisme , Anticorps monoclonaux humanisés/pharmacologie , Antigènes néoplasiques/composition chimique , Antigènes néoplasiques/génétique , Antigènes néoplasiques/immunologie , Antinéoplasiques immunologiques/pharmacologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/métabolisme , Calréticuline/composition chimique , Calréticuline/immunologie , Études cas-témoins , Lignée cellulaire tumorale , Mutation avec décalage du cadre de lecture , Humains , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/immunologie , Peptides/immunologie
14.
Clin Cancer Res ; 24(20): 4937-4948, 2018 10 15.
Article de Anglais | MEDLINE | ID: mdl-29950349

RÉSUMÉ

Purpose: Polyinosinic-polycytidylic acid-poly-l-lysine carboxymethylcellulose (poly-ICLC), a synthetic double-stranded RNA complex, is a ligand for toll-like receptor-3 and MDA-5 that can activate immune cells, such as dendritic cells, and trigger natural killer cells to kill tumor cells.Patients and Methods: In this pilot study, eligible patients included those with recurrent metastatic disease in whom prior systemic therapy (head and neck squamous cell cancer and melanoma) failed. Patients received 2 treatment cycles, each cycle consisting of 1 mg poly-ICLC 3× weekly intratumorally (IT) for 2 weeks followed by intramuscular (IM) boosters biweekly for 7 weeks, with a 1-week rest period. Immune response was evaluated by immunohistochemistry (IHC) and RNA sequencing (RNA-seq) in tumor and blood.Results: Two patients completed 2 cycles of IT treatments, and 1 achieved clinical benefit (stable disease, progression-free survival 6 months), whereas the remainder had progressive disease. Poly-ICLC was well tolerated, with principal side effects of fatigue and inflammation at injection site (

Sujet(s)
Carboxyméthylcellulose de sodium/analogues et dérivés , Facteurs immunologiques/administration et posologie , Immunomodulation/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Poly I-C/administration et posologie , Polylysine/analogues et dérivés , Sujet âgé , Biopsie , Carboxyméthylcellulose de sodium/administration et posologie , Carboxyméthylcellulose de sodium/effets indésirables , Calendrier d'administration des médicaments , Femelle , Analyse de profil d'expression de gènes , Séquençage nucléotidique à haut débit , Humains , Immunohistochimie , Facteurs immunologiques/effets indésirables , Injections intralésionnelles , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Tumeurs/diagnostic , Tumeurs/mortalité , Projets pilotes , Poly I-C/effets indésirables , Polylysine/administration et posologie , Polylysine/effets indésirables , Pronostic , Tomodensitométrie , Résultat thérapeutique
15.
Nat Struct Mol Biol ; 20(11): 1281-8, 2013 Nov.
Article de Anglais | MEDLINE | ID: mdl-24121231

RÉSUMÉ

The Ccr4-Not complex is involved in several aspects of gene expression, including mRNA decay, translational repression and transcription. We determined the 2.8-Å-resolution crystal structure of a 120-kDa core complex of the Saccharomyces cerevisiae Not module comprising the C-terminal arm of Not1, Not2 and Not5. Not1 is a HEAT-repeat scaffold. Not2 and Not5 have extended regions that wrap around Not1 and around their globular domains, the Not boxes. The Not boxes resemble Sm folds and interact with each other with a noncanonical dimerization surface. Disruption of the interactions within the ternary complex has severe effects on growth in vivo. The ternary complex forms a composite surface that binds poly(U) RNA in vitro, with a site at the Not5 Not box. The results suggest that the Not module forms a versatile platform for macromolecular interactions.


Sujet(s)
Protéines du cycle cellulaire/composition chimique , Protéines du cycle cellulaire/métabolisme , ARN/métabolisme , Protéines de répression/composition chimique , Protéines de répression/métabolisme , Protéines de Saccharomyces cerevisiae/composition chimique , Protéines de Saccharomyces cerevisiae/métabolisme , Facteurs de transcription/composition chimique , Facteurs de transcription/métabolisme , Séquence d'acides aminés , Cristallographie aux rayons X , Modèles moléculaires , Données de séquences moléculaires , Liaison aux protéines , Conformation des protéines , Protéines de liaison à l'ARN/composition chimique , Protéines de liaison à l'ARN/métabolisme , Saccharomyces cerevisiae/composition chimique , Saccharomyces cerevisiae/enzymologie
16.
Mol Cell ; 48(2): 207-18, 2012 Oct 26.
Article de Anglais | MEDLINE | ID: mdl-22959269

RÉSUMÉ

Shortening eukaryotic poly(A) tails represses mRNA translation and induces mRNA turnover. The major cytoplasmic deadenylase, the Ccr4-Not complex, is a conserved multisubunit assembly. Ccr4-Not is organized around Not1, a large scaffold protein that recruits two 3'-5' exoribonucleases, Caf1 and Ccr4. We report structural studies showing that the N-terminal arm of yeast Not1 has a HEAT-repeat structure with domains related to the MIF4G fold. A MIF4G domain positioned centrally within the Not1 protein recognizes Caf1, which in turn binds the LRR domain of Ccr4 and tethers the Ccr4 nuclease domain. The interactions that form the nuclease core of the Ccr4-Not complex are evolutionarily conserved. Their specific disruption affects cell growth and mRNA deadenylation and decay in vivo in yeast. Thus, the N-terminal arm of Not1 forms an extended platform reminiscent of scaffolding proteins like eIF4G and CBP80, and places the two nucleases in a pivotal position within the Ccr4-Not complex.


Sujet(s)
Protéines du cycle cellulaire , Ribonucléases , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Facteurs de transcription , Sites de fixation , Protéines du cycle cellulaire/composition chimique , Protéines du cycle cellulaire/métabolisme , Cristallographie aux rayons X , Facteur-4G d'initiation eucaryote/métabolisme , Complexes multiprotéiques/génétique , Complexes multiprotéiques/métabolisme , Protéines nucléaires/métabolisme , Liaison aux protéines , Conformation des protéines , Structure tertiaire des protéines , Protéines de liaison à la coiffe de l'ARN/métabolisme , ARN messager/composition chimique , ARN messager/génétique , ARN messager/métabolisme , Ribonucléases/composition chimique , Ribonucléases/métabolisme , Saccharomyces cerevisiae/composition chimique , Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/composition chimique , Protéines de Saccharomyces cerevisiae/métabolisme , Facteurs de transcription/composition chimique , Facteurs de transcription/métabolisme
17.
Proc Natl Acad Sci U S A ; 107(29): 12828-33, 2010 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-20566882

RÉSUMÉ

While many functions of the p53 tumor suppressor affect mitochondrial processes, the role of altered mitochondrial physiology in a modulation of p53 response remains unclear. As mitochondrial respiration is affected in many pathologic conditions such as hypoxia and intoxications, the impaired electron transport chain could emit additional p53-inducing signals and thereby contribute to tissue damage. Here we show that a shutdown of mitochondrial respiration per se does not trigger p53 response, because inhibitors acting in the proximal and distal segments of the respiratory chain do not activate p53. However, strong p53 response is induced specifically after an inhibition of the mitochondrial cytochrome bc1 (the electron transport chain complex III). The p53 response is triggered by the deficiency in pyrimidines that is developed due to a suppression of the functionally coupled mitochondrial pyrimidine biosynthesis enzyme dihydroorotate dehydrogenase (DHODH). In epithelial carcinoma cells the activation of p53 in response to mitochondrial electron transport chain complex III inhibitors does not require phosphorylation of p53 at Serine 15 or up-regulation of p14(ARF). Instead, our data suggest a contribution of NQO1 and NQO2 in stabilization of p53 in the nuclei. The results establish the deficiency in pyrimidine biosynthesis as the cause of p53 response in the cells with impaired mitochondrial respiration.


Sujet(s)
Mitochondries/métabolisme , Pyrimidines/biosynthèse , Transduction du signal , Protéine p53 suppresseur de tumeur/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Respiration cellulaire/effets des médicaments et des substances chimiques , Dihydroorotate dehydrogenase , Complexe III de la chaîne respiratoire/antagonistes et inhibiteurs , Complexe III de la chaîne respiratoire/métabolisme , Humains , Isoxazoles/pharmacologie , Léflunomide , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Méthacrylates/pharmacologie , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/enzymologie , NADPH dehydrogenase (quinone)/métabolisme , Oxidoreductases acting on CH-CH group donors/antagonistes et inhibiteurs , Oxidoreductases acting on CH-CH group donors/déficit , Cyanure de potassium/pharmacologie , Proteasome endopeptidase complex/métabolisme , Maturation post-traductionnelle des protéines/effets des médicaments et des substances chimiques , Quinone reductases/métabolisme , Espèces réactives de l'oxygène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Thiazoles/pharmacologie , Régulation positive/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...