Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nat Commun ; 15(1): 4327, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773088

RÉSUMÉ

The antitumor efficacy of adoptively transferred T cells is limited by their poor persistence, in part due to exhaustion, but the underlying mechanisms and potential interventions remain underexplored. Here, we show that targeting histone demethylase LSD1 by chemical inhibitors reshapes the epigenome of in vitro activated and expanded CD8+ T cells, and potentiates their antitumor efficacy. Upon T cell receptor activation and IL-2 signaling, a timely and transient inhibition of LSD1 suffices to improve the memory phenotype of mouse CD8+ T cells, associated with a better ability to produce multiple cytokines, resist exhaustion, and persist in both antigen-dependent and -independent manners after adoptive transfer. Consequently, OT1 cells primed with LSD1 inhibitors demonstrate an enhanced antitumor effect in OVA-expressing solid tumor models implanted in female mice, both as a standalone treatment and in combination with PD-1 blockade. Moreover, priming with LSD1 inhibitors promotes polyfunctionality of human CD8+ T cells, and increases the persistence and antitumor efficacy of human CD19-CAR T cells in both leukemia and solid tumor models. Thus, pharmacological inhibition of LSD1 could be exploited to improve adoptive T cell therapy.


Sujet(s)
Lymphocytes T CD8+ , Histone Demethylases , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Souris , Humains , Femelle , Souris de lignée C57BL , Immunothérapie adoptive/méthodes , Lignée cellulaire tumorale , Activation des lymphocytes/effets des médicaments et des substances chimiques , Transfert adoptif , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Interleukine-2/métabolisme , Antigènes CD19/métabolisme , Antigènes CD19/immunologie , Mémoire immunologique/effets des médicaments et des substances chimiques
2.
Mol Cell ; 83(23): 4370-4385.e9, 2023 Dec 07.
Article de Anglais | MEDLINE | ID: mdl-38016475

RÉSUMÉ

Targeting epigenetic regulators to potentiate anti-PD-1 immunotherapy converges on the activation of type I interferon (IFN-I) response, mimicking cellular response to viral infection, but how its strength and duration are regulated to impact combination therapy efficacy remains largely unknown. Here, we show that mitochondrial CPT1A downregulation following viral infection restrains, while its induction by epigenetic perturbations sustains, a double-stranded RNA-activated IFN-I response. Mechanistically, CPT1A recruits the endoplasmic reticulum-localized ZDHHC4 to catalyze MAVS Cys79-palmitoylation, which promotes MAVS stabilization and activation by inhibiting K48- but facilitating K63-linked ubiquitination. Further elevation of CPT1A incrementally increases MAVS palmitoylation and amplifies the IFN-I response, which enhances control of viral infection and epigenetic perturbation-induced antitumor immunity. Moreover, CPT1A chemical inducers augment the therapeutic effect of combined epigenetic treatment with PD-1 blockade in refractory tumors. Our study identifies CPT1A as a stabilizer of MAVS activation, and its link to epigenetic perturbation can be exploited for cancer immunotherapy.


Sujet(s)
Interféron de type I , Maladies virales , Humains , Transduction du signal , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Lipoylation , Épigenèse génétique , Immunité innée
4.
Nat Commun ; 12(1): 6831, 2021 11 24.
Article de Anglais | MEDLINE | ID: mdl-34819502

RÉSUMÉ

Exhausted CD8+ T cells are key targets of immune checkpoint blockade therapy and their ineffective reinvigoration limits the durable benefit in some cancer patients. Here, we demonstrate that histone demethylase LSD1 acts to enforce an epigenetic program in progenitor exhausted CD8+ T cells to antagonize the TCF1-mediated progenitor maintenance and to promote terminal differentiation. Consequently, genetic perturbation or small molecules targeting LSD1 increases the persistence of the progenitor exhausted CD8+ T cells, which provide a sustained source for the proliferative conversion to numerically larger terminally exhausted T cells with tumor-killing cytotoxicity, thereby leading to effective and durable responses to anti-PD1 therapy. Collectively, our findings provide important insights into epigenetic mechanisms that regulate T cell exhaustion and have important implications for durable immunotherapy.


Sujet(s)
Lymphocytes T CD8+/immunologie , Régulation de l'expression des gènes tumoraux/immunologie , Histone Demethylases/métabolisme , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs/immunologie , Animaux , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale/transplantation , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Déméthylation de l'ADN/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/génétique , Épigenèse génétique/immunologie , Femelle , Cellules HEK293 , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/génétique , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lymphocytes TIL , Mâle , Souris , Souris transgéniques , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Culture de cellules primaires , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Protéines recombinantes
5.
Cancer Discov ; 11(8): 1970-1981, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33687985

RÉSUMÉ

Epigenetic regulators are a class of promising targets in combination with immune checkpoint inhibitors for cancer treatment, but the impact of the broad effects of perturbing epigenetic regulators on tumor immunotherapy remains to be fully explored. Here we show that ablation of the histone demethylase LSD1 in multiple tumor cells induces TGFß expression, which exerts an inhibitory effect on T-cell immunity through suppressing the cytotoxicity of intratumoral CD8+ T cells and consequently dampens the antitumor effect of LSD1 ablation-induced T-cell infiltration. Importantly, concurrent depletion of LSD1 and TGFß in combination with PD-1 blockade significantly increases both CD8+ T-cell infiltration and cytotoxicity, leading to eradication of poorly immunogenic tumors and a long-term protection from tumor rechallenge. Thus, combining LSD1 inhibition with blockade of TGFß and PD-1 may represent a promising triple combination therapy for treating certain refractory tumors. SIGNIFICANCE: Cotargeting LSD1 and TGFß cooperatively elevates intratumoral CD8+ T-cell infiltration and unleashes their cytotoxicity, leading to tumor eradication upon anti-PD-1 treatment. Our findings illustrate a duality of epigenetic perturbations in immunotherapy and implicate the combination of LSD1 inhibition with dual PD-1/TGFß blockade in treating certain poorly immunogenic tumors.This article is highlighted in the In This Issue feature, p. 1861.


Sujet(s)
Histone Demethylases/métabolisme , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Facteur de croissance transformant bêta/métabolisme , Animaux , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Femelle , Humains , Immunothérapie , Souris , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs
6.
Bull Math Biol ; 83(2): 15, 2021 01 12.
Article de Anglais | MEDLINE | ID: mdl-33433736

RÉSUMÉ

LSD1 (KDMA1) has gained attention in the last decade as a cancer biomarker and drug target. In particular, recent work suggests that LSD1 inhibition alone reduces tumor growth, increases T cell tumor infiltration, and complements PD1/PDL1 checkpoint inhibitor therapy. In order to elucidate the immunogenic effects of LSD1 inhibition, we develop a mathematical model of tumor growth under the influence of the adaptive immune response. In particular, we investigate the anti-tumor cytotoxicity of LSD1-mediated T cell dynamics, in order to better understand the synergistic potential of LSD1 inhibition in combination immunotherapies, including checkpoint inhibitors. To that end, we formulate a non-spatial delay differential equation model and fit to the B16 mouse model data from Sheng et al. (Cell 174(3):549-563, 2018. https://doi.org/10.1016/j.cell.2018.05.052 ). Our results suggest that the immunogenic effect of LSD1 inhibition accelerates anti-tumor cytotoxicity. However, cytotoxicity does not seem to account for the slower growth observed in LSD1-inhibited tumors, despite evidence suggesting immune-mediation of this effect.


Sujet(s)
Histone Demethylases , Modèles biologiques , Tumeurs , Animaux , Modèles animaux de maladie humaine , Histone Demethylases/métabolisme , Souris , Tumeurs/physiopathologie , Lymphocytes T/immunologie
7.
Mol Cell ; 75(3): 620-630.e9, 2019 08 08.
Article de Anglais | MEDLINE | ID: mdl-31279659

RÉSUMÉ

mRNA modifications play important roles in regulating gene expression. One of the most abundant mRNA modifications is N6,2-O-dimethyladenosine (m6Am). Here, we demonstrate that m6Am is an evolutionarily conserved mRNA modification mediated by the Phosphorylated CTD Interacting Factor 1 (PCIF1), which catalyzes m6A methylation on 2-O-methylated adenine located at the 5' ends of mRNAs. Furthermore, PCIF1 catalyzes only 5' m6Am methylation of capped mRNAs but not internal m6A methylation in vitro and in vivo. To study the biological role of m6Am, we developed a robust methodology (m6Am-Exo-Seq) to map its transcriptome-wide distribution, which revealed no global crosstalk between m6Am and m6A under assayed conditions, suggesting that m6Am is functionally distinct from m6A. Importantly, we find that m6Am does not alter mRNA transcription or stability but negatively impacts cap-dependent translation of methylated mRNAs. Together, we identify the only human mRNA m6Am methyltransferase and demonstrate a mechanism of gene expression regulation through PCIF1-mediated m6Am mRNA methylation.


Sujet(s)
Protéines adaptatrices de la transduction du signal/génétique , Protéines nucléaires/génétique , Maturation post-transcriptionnelle des ARN/génétique , ARN messager/génétique , Transcription génétique , Adénosine/génétique , Régulation de l'expression des gènes/génétique , Humains , Méthylation , Methyltransferases/génétique , Phosphorylation , Transcriptome/génétique
8.
Cell ; 174(3): 549-563.e19, 2018 07 26.
Article de Anglais | MEDLINE | ID: mdl-29937226

RÉSUMÉ

Chromatin regulators play a broad role in regulating gene expression and, when gone awry, can lead to cancer. Here, we demonstrate that ablation of the histone demethylase LSD1 in cancer cells increases repetitive element expression, including endogenous retroviral elements (ERVs), and decreases expression of RNA-induced silencing complex (RISC) components. Significantly, this leads to double-stranded RNA (dsRNA) stress and activation of type 1 interferon, which stimulates anti-tumor T cell immunity and restrains tumor growth. Furthermore, LSD1 depletion enhances tumor immunogenicity and T cell infiltration in poorly immunogenic tumors and elicits significant responses of checkpoint blockade-refractory mouse melanoma to anti-PD-1 therapy. Consistently, TCGA data analysis shows an inverse correlation between LSD1 expression and CD8+ T cell infiltration in various human cancers. Our study identifies LSD1 as a potent inhibitor of anti-tumor immunity and responsiveness to immunotherapy and suggests LSD1 inhibition combined with PD-(L)1 blockade as a novel cancer treatment strategy.


Sujet(s)
Rétrovirus endogènes/génétique , Histone Demethylases/métabolisme , Complexe réprimant l'expression de l'ARN/génétique , Animaux , Lignée cellulaire tumorale , Chromatine , Association thérapeutique , Régulation de l'expression des gènes/génétique , Histone Demethylases/génétique , Humains , Immunité cellulaire , Immunothérapie , Interféron de type I , Cellules MCF-7 , Souris , Récepteur-1 de mort cellulaire programmée/génétique , Récepteur-1 de mort cellulaire programmée/métabolisme , ARN double brin/génétique , Lymphocytes T
10.
Nature ; 543(7646): 573-576, 2017 03 23.
Article de Anglais | MEDLINE | ID: mdl-28297716

RÉSUMÉ

Cell proliferation and survival require the faithful maintenance and propagation of genetic information, which are threatened by the ubiquitous sources of DNA damage present intracellularly and in the external environment. A system of DNA repair, called the DNA damage response, detects and repairs damaged DNA and prevents cell division until the repair is complete. Here we report that methylation at the 6 position of adenosine (m6A) in RNA is rapidly (within 2 min) and transiently induced at DNA damage sites in response to ultraviolet irradiation. This modification occurs on numerous poly(A)+ transcripts and is regulated by the methyltransferase METTL3 (methyltransferase-like 3) and the demethylase FTO (fat mass and obesity-associated protein). In the absence of METTL3 catalytic activity, cells showed delayed repair of ultraviolet-induced cyclobutane pyrimidine adducts and elevated sensitivity to ultraviolet, demonstrating the importance of m6A in the ultraviolet-responsive DNA damage response. Multiple DNA polymerases are involved in the ultraviolet response, some of which resynthesize DNA after the lesion has been excised by the nucleotide excision repair pathway, while others participate in trans-lesion synthesis to allow replication past damaged lesions in S phase. DNA polymerase κ (Pol κ), which has been implicated in both nucleotide excision repair and trans-lesion synthesis, required the catalytic activity of METTL3 for immediate localization to ultraviolet-induced DNA damage sites. Importantly, Pol κ overexpression qualitatively suppressed the cyclobutane pyrimidine removal defect associated with METTL3 loss. Thus, we have uncovered a novel function for RNA m6A modification in the ultraviolet-induced DNA damage response, and our findings collectively support a model in which m6A RNA serves as a beacon for the selective, rapid recruitment of Pol κ to damage sites to facilitate repair and cell survival.


Sujet(s)
Altération de l'ADN/effets des radiations , Méthylation , ARN/composition chimique , ARN/métabolisme , Rayons ultraviolets , Alpha-ketoglutarate-dependent dioxygenase FTO/métabolisme , Animaux , Biocatalyse/effets des radiations , Lignée cellulaire , Survie cellulaire/effets des radiations , Réparation de l'ADN/effets des radiations , Réplication de l'ADN/effets des radiations , DNA-directed DNA polymerase/métabolisme , Humains , Méthylation/effets des radiations , Methyltransferases/déficit , Methyltransferases/métabolisme , Souris , Poly A/métabolisme , ARN/effets des radiations , Phase S/effets des radiations
11.
Cell Res ; 24(12): 1387-402, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25412660

RÉSUMÉ

T helper (TH)-cell subsets, such as TH1 and TH17, mediate inflammation in both peripheral tissues and central nervous system. Here we show that STAT5 is required for T helper-cell pathogenicity in autoimmune neuroinflammation but not in experimental colitis. Although STAT5 promotes regulatory T cell generation and immune suppression, loss of STAT5 in CD4+ T cells resulted in diminished development of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Our results showed that loss of encephalitogenic activity of STAT5-deficient autoreactive CD4+ T cells was independent of IFN-γ or interleukin 17 (IL-17) production, but was due to the impaired expression of granulocyte-macrophage colony-stimulating factor (GM-CSF), a crucial mediator of T-cell pathogenicity. We further showed that IL-7-activated STAT5 promotes the generation of GM-CSF-producing CD4+ T cells, which were preferentially able to induce more severe EAE than TH17 or TH1 cells. Consistent with GM-CSF-producing cells being a distinct subset of TH cells, the differentiation program of these cells was distinct from that of TH17 or TH1 cells. We further found that IL-3 was secreted in a similar pattern as GM-CSF in this subset of TH cells. In conclusion, the IL-7-STAT5 axis promotes the generation of GM-CSF/IL-3-producing TH cells. These cells display a distinct transcriptional profile and may represent a novel subset of T helper cells which we designate as TH-GM.


Sujet(s)
Encéphalomyélite auto-immune expérimentale/immunologie , Facteur de stimulation des colonies de granulocytes et de macrophages/immunologie , Facteur de transcription STAT-5/immunologie , Lymphocytes T auxiliaires/immunologie , Animaux , Colite/génétique , Colite/immunologie , Colite/anatomopathologie , Encéphalomyélite auto-immune expérimentale/génétique , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Délétion de gène , Facteur de stimulation des colonies de granulocytes et de macrophages/analyse , Interleukine-7/analyse , Interleukine-7/immunologie , Souris , Souris de lignée C57BL , Souris knockout , Facteur de transcription STAT-5/analyse , Facteur de transcription STAT-5/génétique , Lymphocytes T auxiliaires/métabolisme , Lymphocytes T auxiliaires/anatomopathologie
12.
Mol Reprod Dev ; 80(9): 744-52, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23836407

RÉSUMÉ

Vasa has been extensively used as a germ-line marker to trace the origin and migration pathway of primordial germ cells (PGCs) in many organisms, but little work has been reported on vasa genes and the origin of PGCs in holothurians. Using in situ hybridization and immunohistochemistry, vasa mRNA and protein of the sea cucumber Apostichopus japonicus (Aj-vasa) was detected in the cytoplasm of the unfertilized egg and was equally distributed in the cytoplasm of early embryos, from the two-cell embryo to the blastula, indicating that Aj-vasa mRNA is maternally supplied. Later, expression of both Aj-vasa mRNA and protein centralizes gradually in newly organized structures from blastula to five-tentacle larva, and then is restricted to PGC-like cells of the original gonad in juveniles with 0.1-cm body length. The structure of the gonad develops further from a simple tubular gonad in 0.5-cm-length juveniles to a branched gonad in 3-cm-length juveniles. Our findings showed that the maternal supply of the vasa gene products in A. japonicus is different from that in sea urchin Strongylocentrotus purpuratus, of echinoderm, and suggested that the specialization of PGCs is an epigenesis mechanism in A. japonicus.


Sujet(s)
DEAD-box RNA helicases/métabolisme , Régulation de l'expression des gènes au cours du développement/physiologie , Gonades/embryologie , ARN messager/métabolisme , Stichopus/embryologie , Stichopus/enzymologie , Animaux , Cytoplasme/métabolisme , Épigenèse génétique/physiologie , Régulation de l'expression des gènes au cours du développement/génétique , Immunohistochimie , Hybridation in situ , Spécificité d'espèce
13.
Gene Expr Patterns ; 13(5-6): 171-6, 2013.
Article de Anglais | MEDLINE | ID: mdl-23542063

RÉSUMÉ

The vasa gene is a reliable germline marker to study the origin and development of germ cells and gonads, although the gene product (mRNA or protein) varies between different species. However, there has been little study on vasa genes in holothuroids to date. Here we determined the expression characteristics of the Apostichopus japonicus vasa gene (Aj-vasa) during gametogenesis in the ovary and testis using in situ hybridization and immunohistochemistry. During oogenesis, the expression pattern of Aj-vasa coincided at the mRNA and protein levels. Intensive signals in oogonia decreased gradually with the development of oocytes. Interestingly, the pattern was different during spermatogenesis. The Aj-vasa mRNA level was the highest in spermatogonia, reduced in spermatocytes, low in spermatids and absent in spermatozoa, but the Aj-VASA protein was restricted to spermatogonia and early spermatocytes. These expression characteristics of Aj-vasa persisted in both male and female gonads throughout the reproductive cycle. Our findings show that Aj-vasa mRNA is a good marker for studying the origin and migration of germline cells; moreover, Aj-VASA is a useful tool to identify spermatogonia in A. japonicus. Our findings indicate that Aj-vasa is vital in the development and differentiation of germ cells.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Ovogenèse/génétique , ARN messager/biosynthèse , Spermatogenèse/génétique , Stichopus/croissance et développement , Animaux , Femelle , Analyse de profil d'expression de gènes , Cellules germinales , Mâle , Ovaire/cytologie , Ovaire/croissance et développement , Spermatogonies/cytologie , Spermatogonies/croissance et développement
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE