Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
1.
Mol Cell ; 83(8): 1216-1236.e12, 2023 04 20.
Article de Anglais | MEDLINE | ID: mdl-36944333

RÉSUMÉ

Highly coordinated changes in gene expression underlie T cell activation and exhaustion. However, the mechanisms by which such programs are regulated and how these may be targeted for therapeutic benefit remain poorly understood. Here, we comprehensively profile the genomic occupancy of mSWI/SNF chromatin remodeling complexes throughout acute and chronic T cell stimulation, finding that stepwise changes in localization over transcription factor binding sites direct site-specific chromatin accessibility and gene activation leading to distinct phenotypes. Notably, perturbation of mSWI/SNF complexes using genetic and clinically relevant chemical strategies enhances the persistence of T cells with attenuated exhaustion hallmarks and increased memory features in vitro and in vivo. Finally, pharmacologic mSWI/SNF inhibition improves CAR-T expansion and results in improved anti-tumor control in vivo. These findings reveal the central role of mSWI/SNF complexes in the coordination of T cell activation and exhaustion and nominate small-molecule-based strategies for the improvement of current immunotherapy protocols.


Sujet(s)
Assemblage et désassemblage de la chromatine , Protéines chromosomiques nonhistones , Protéines chromosomiques nonhistones/génétique , Protéines chromosomiques nonhistones/métabolisme , Facteurs de transcription/métabolisme , Chromatine/génétique , Activation de la transcription
2.
Nat Immunol ; 22(8): 1020-1029, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34312547

RÉSUMÉ

T cell exhaustion is an induced state of dysfunction that arises in response to chronic infection and cancer. Exhausted CD8+ T cells acquire a distinct epigenetic state, but it is not known whether that chromatin landscape is fixed or plastic following the resolution of a chronic infection. Here we show that the epigenetic state of exhaustion is largely irreversible, even after curative therapy. Analysis of chromatin accessibility in HCV- and HIV-specific responses identifies a core epigenetic program of exhaustion in CD8+ T cells, which undergoes only limited remodeling before and after resolution of infection. Moreover, canonical features of exhaustion, including super-enhancers near the genes TOX and HIF1A, remain 'epigenetically scarred.' T cell exhaustion is therefore a conserved epigenetic state that becomes fixed and persists independent of chronic antigen stimulation and inflammation. Therapeutic efforts to reverse T cell exhaustion may require new approaches that increase the epigenetic plasticity of exhausted T cells.


Sujet(s)
Antigènes viraux/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/anatomopathologie , Hepacivirus/immunologie , Hépatite C chronique/immunologie , Mémoire immunologique/immunologie , 2-Naphtylamine/usage thérapeutique , Anilides/usage thérapeutique , Antiviraux/usage thérapeutique , Chromatine/métabolisme , Cyclopropanes/usage thérapeutique , Épigenèse génétique/génétique , Hepacivirus/effets des médicaments et des substances chimiques , Hépatite C chronique/traitement médicamenteux , Protéines HMG/génétique , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Lactames macrocycliques/usage thérapeutique , Proline/analogues et dérivés , Proline/usage thérapeutique , Ribavirine/usage thérapeutique , Ritonavir/usage thérapeutique , Sulfonamides/usage thérapeutique , Uracile/analogues et dérivés , Uracile/usage thérapeutique , Valine/usage thérapeutique
3.
Cell ; 179(6): 1342-1356.e23, 2019 11 27.
Article de Anglais | MEDLINE | ID: mdl-31759698

RÉSUMÉ

Mammalian switch/sucrose non-fermentable (mSWI/SNF) complexes are multi-component machines that remodel chromatin architecture. Dissection of the subunit- and domain-specific contributions to complex activities is needed to advance mechanistic understanding. Here, we examine the molecular, structural, and genome-wide regulatory consequences of recurrent, single-residue mutations in the putative coiled-coil C-terminal domain (CTD) of the SMARCB1 (BAF47) subunit, which cause the intellectual disability disorder Coffin-Siris syndrome (CSS), and are recurrently found in cancers. We find that the SMARCB1 CTD contains a basic α helix that binds directly to the nucleosome acidic patch and that all CSS-associated mutations disrupt this binding. Furthermore, these mutations abrogate mSWI/SNF-mediated nucleosome remodeling activity and enhancer DNA accessibility without changes in genome-wide complex localization. Finally, heterozygous CSS-associated SMARCB1 mutations result in dominant gene regulatory and morphologic changes during iPSC-neuronal differentiation. These studies unmask an evolutionarily conserved structural role for the SMARCB1 CTD that is perturbed in human disease.


Sujet(s)
Assemblage et désassemblage de la chromatine/génétique , Protéines chromosomiques nonhistones/métabolisme , Mutation/génétique , Nucléosomes/métabolisme , Protéine SMARCB1/génétique , Facteurs de transcription/métabolisme , Séquence d'acides aminés , Éléments activateurs (génétique)/génétique , Femelle , Génome humain , Cellules HEK293 , Cellules HeLa , Hétérozygote , Humains , Mâle , Modèles moléculaires , Protéines mutantes/composition chimique , Protéines mutantes/métabolisme , Liaison aux protéines , Domaines protéiques , Protéine SMARCB1/composition chimique , Protéine SMARCB1/métabolisme
5.
Nat Immunol ; 20(3): 326-336, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30778252

RÉSUMÉ

T cell dysfunction is a hallmark of many cancers, but the basis for T cell dysfunction and the mechanisms by which antibody blockade of the inhibitory receptor PD-1 (anti-PD-1) reinvigorates T cells are not fully understood. Here we show that such therapy acts on a specific subpopulation of exhausted CD8+ tumor-infiltrating lymphocytes (TILs). Dysfunctional CD8+ TILs possess canonical epigenetic and transcriptional features of exhaustion that mirror those seen in chronic viral infection. Exhausted CD8+ TILs include a subpopulation of 'progenitor exhausted' cells that retain polyfunctionality, persist long term and differentiate into 'terminally exhausted' TILs. Consequently, progenitor exhausted CD8+ TILs are better able to control tumor growth than are terminally exhausted T cells. Progenitor exhausted TILs can respond to anti-PD-1 therapy, but terminally exhausted TILs cannot. Patients with melanoma who have a higher percentage of progenitor exhausted cells experience a longer duration of response to checkpoint-blockade therapy. Thus, approaches to expand the population of progenitor exhausted CD8+ T cells might be an important component of improving the response to checkpoint blockade.


Sujet(s)
Anticorps bloquants/pharmacologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes TIL/effets des médicaments et des substances chimiques , Mélanome expérimental/prévention et contrôle , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Animaux , Anticorps bloquants/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/virologie , Lignée cellulaire tumorale , Femelle , Humains , Sous-populations de lymphocytes/effets des médicaments et des substances chimiques , Sous-populations de lymphocytes/immunologie , Sous-populations de lymphocytes/virologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/virologie , Chorioméningite lymphocytaire/immunologie , Chorioméningite lymphocytaire/prévention et contrôle , Chorioméningite lymphocytaire/virologie , Virus de la chorioméningite lymphocytaire/effets des médicaments et des substances chimiques , Virus de la chorioméningite lymphocytaire/immunologie , Virus de la chorioméningite lymphocytaire/physiologie , Mélanome expérimental/immunologie , Mélanome expérimental/virologie , Souris congéniques , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/immunologie , Récepteur-1 de mort cellulaire programmée/métabolisme
6.
Nature ; 565(7737): 43-48, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30559380

RÉSUMÉ

Most patients with cancer either do not respond to immune checkpoint blockade or develop resistance to it, often because of acquired mutations that impair antigen presentation. Here we show that loss of function of the RNA-editing enzyme ADAR1 in tumour cells profoundly sensitizes tumours to immunotherapy and overcomes resistance to checkpoint blockade. In the absence of ADAR1, A-to-I editing of interferon-inducible RNA species is reduced, leading to double-stranded RNA ligand sensing by PKR and MDA5; this results in growth inhibition and tumour inflammation, respectively. Loss of ADAR1 overcomes resistance to PD-1 checkpoint blockade caused by inactivation of antigen presentation by tumour cells. Thus, effective anti-tumour immunity is constrained by inhibitory checkpoints such as ADAR1 that limit the sensing of innate ligands. The induction of sufficient inflammation in tumours that are sensitized to interferon can bypass the therapeutic requirement for CD8+ T cell recognition of cancer cells and may provide a general strategy to overcome immunotherapy resistance.


Sujet(s)
Adenosine deaminase/déficit , Adenosine deaminase/métabolisme , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/génétique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Protéines de liaison à l'ARN/métabolisme , Adenosine deaminase/génétique , Animaux , Systèmes CRISPR-Cas/génétique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Antigènes d'histocompatibilité de classe I/immunologie , Immunothérapie , Inflammation/génétique , Inflammation/immunologie , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Interférons/immunologie , Mélanome expérimental/immunologie , Mélanome expérimental/radiothérapie , Souris , Souris de lignée C57BL , Phénotype , Édition des ARN , ARN double brin/génétique , Protéines de liaison à l'ARN/génétique , Récepteurs couplés aux protéines G/métabolisme
7.
Biochem Biophys Res Commun ; 499(2): 314-320, 2018 05 05.
Article de Anglais | MEDLINE | ID: mdl-29574159

RÉSUMÉ

Malate Dehydrogenase (MDH) 1 has recently been shown to be highly expressed and display prognostic value in non-small cell lung carcinomas (NSCLCs). However, it is not known how MDH1 expression is regulated and there is no current molecular or chemical strategy that specifically targets MDH1. This may be due to structural and enzymatic similarities with its isoenzyme, malate dehydrogenase 2 (MDH2). However, MDH1 and MDH2 are encoded by distinct genes and this opens up the possibility for modulation at the expression level. Here, we screened in silico for microRNAs (miRs) that selectively targets the 3'UTR region of MDH1. These analyses revealed that mir-126-5p has three binding sites in the 3'UTR region of MDH1. Additionally, we show that expression of miR-126-5p suppresses the enzymatic activity of MDH1, mitochondrial respiration and caused cell death in NSCLC cell lines.


Sujet(s)
Carcinome pulmonaire non à petites cellules/enzymologie , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/enzymologie , Tumeurs du poumon/génétique , Malate dehydrogenase/métabolisme , microARN/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Mort cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Respiration cellulaire , Clones cellulaires , Humains , Tumeurs du poumon/anatomopathologie , microARN/génétique , Mitochondries/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...