Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 3 de 3
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cancer Res ; 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38885318

RÉSUMÉ

Increasing evidence supports the interplay between oncogenic mutations and immune escape mechanisms. Strategies to counteract the immune escape mediated by oncogenic signaling could provide improved therapeutic options for patients with various malignancies. As mutant calreticulin (CALR) is a common driver of myeloproliferative neoplasms (MPN), we analyzed the impact of oncogenic CALRdel52 on the bone marrow (BM) microenvironment in MPN. Single-cell RNA-sequencing revealed that CALRdel52 led to the expansion of TGF-ß1-producing erythroid progenitor cells and promoted the expansion of FoxP3+ regulatory T cells (Treg) in a murine MPN model. Treatment with an anti-TGF-ß antibody improved mouse survival and increased the glycolytic activity in CD4+ and CD8+ T cells in vivo, while T cell depletion abrogated the protective effects conferred by neutralizing TGF-ß. TGF-ß1 reduced perforin and TNF-α production by T cells in vitro. TGF-ß1 production by CALRdel52 cells was dependent on JAK1/2, PI3K, and ERK activity, which activated the transcription factor Sp1 to induce TGF-ß1 expression. In four independent patient cohorts, TGF-ß1 expression was increased in the BM of MPN patients compared to healthy individuals, and the BM of MPN patients contained a higher frequency of Treg compared to healthy individuals. Together, this study identified an ERK/Sp1/TGF-ß1 axis in CALRdel52 MPNs as a mechanism of immunosuppression that can be targeted to elicit T-cell-mediated cytotoxicity.

2.
PLoS One ; 15(2): e0228362, 2020.
Article de Anglais | MEDLINE | ID: mdl-32017785

RÉSUMÉ

The histone demethylase JMJD1C is overexpressed in patients with myeloproliferative neoplasms (MPNs) and has been implicated in leukemic stem cell function of MLL-AF9 and HOXA9-driven leukemia. In the emerging field of histone demethylase inhibitors, JMJD1C therefore became a potential target. Depletion of Jmjd1c expression significantly reduced cytokine-independent growth in an MPN cell line, indicating a role for JMJD1C in MPN disease maintenance. Here, we investigated a potential role for the demethylase in MPN disease initiation. We introduced a Cre-inducible JAK2V617F mutation into Jmjd1c knockout mice. We show that Jmjd1c is dispensable, both for healthy hematopoiesis as well as for JAK2V617F-driven MPN disease initiation. Jmjd1c knockout mice did not show any significant changes in peripheral blood composition. Likewise, introduction of JAK2V617F into Jmjd1c-/- mice led to a similar MPN phenotype as JAK2V617F in a Jmjd1c wt background. This indicates that there is a difference between the role of JMJD1C in leukemic stem cells and in MPN. In the latter, JMJC domain-containing family members may serve redundant roles, compensating for the loss of individual proteins.


Sujet(s)
Hématopoïèse , Kinase Janus-2/génétique , Jumonji Domain-Containing Histone Demethylases/génétique , Mutation , Syndromes myéloprolifératifs/génétique , Animaux , Études cas-témoins , Lignée cellulaire tumorale , Développement embryonnaire , Techniques de knock-out de gènes , Humains , Mâle , Souris , Souris knockout
3.
Blood ; 131(18): 2065-2073, 2018 05 03.
Article de Anglais | MEDLINE | ID: mdl-29519804

RÉSUMÉ

The transcription factor "nuclear factor erythroid 2" (NFE2) is overexpressed in the majority of patients with myeloproliferative neoplasms (MPNs). In murine models, elevated NFE2 levels cause an MPN phenotype with spontaneous leukemic transformation. However, both the molecular mechanisms leading to NFE2 overexpression and its downstream targets remain incompletely understood. Here, we show that the histone demethylase JMJD1C constitutes a novel NFE2 target gene. JMJD1C levels are significantly elevated in polycythemia vera (PV) and primary myelofibrosis patients; concomitantly, global H3K9me1 and H3K9me2 levels are significantly decreased. JMJD1C binding to the NFE2 promoter is increased in PV patients, decreasing both H3K9me2 levels and binding of the repressive heterochromatin protein-1α (HP1α). Hence, JMJD1C and NFE2 participate in a novel autoregulatory loop. Depleting JMJD1C expression significantly reduced cytokine-independent growth of an MPN cell line. Independently, NFE2 is regulated through the epigenetic JAK2 pathway by phosphorylation of H3Y41. This likewise inhibits HP1α binding. Treatment with decitabine lowered H3Y41ph and augmented H3K9me2 levels at the NFE2 locus in HEL cells, thereby increasing HP1α binding, which normalized NFE2 expression selectively in JAK2V617F-positive cell lines.


Sujet(s)
Épigenèse génétique , Régulation de l'expression des gènes , Expression des gènes , Syndromes myéloprolifératifs/génétique , Sous-unité p45 du facteur de transcription NF-E2/génétique , Marqueurs biologiques , Homologue-5 de la protéine chromobox , Cytokines/métabolisme , Méthylation de l'ADN , Décitabine/pharmacologie , Histone/métabolisme , Humains , Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Jumonji Domain-Containing Histone Demethylases/génétique , Modèles biologiques , Mutation , Syndromes myéloprolifératifs/métabolisme , Sous-unité p45 du facteur de transcription NF-E2/métabolisme , Oxidoreductases, (N-demethylating)/génétique , Phosphorylation , Polyglobulie primitive essentielle/génétique , Régions promotrices (génétique) , Liaison aux protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...