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.
Cancer Discov ; 13(6): 1478-1497, 2023 06 02.
Article de Anglais | MEDLINE | ID: mdl-36847506

RÉSUMÉ

Oncogenic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 occur in a wide range of cancers, including acute myeloid leukemia (AML) and glioma. Mutant IDH enzymes convert 2-oxoglutarate (2OG) to (R)-2-hydroxyglutarate [(R)-2HG], an oncometabolite that is hypothesized to promote cellular transformation by dysregulating 2OG-dependent enzymes. The only (R)-2HG target that has been convincingly shown to contribute to transformation by mutant IDH is the myeloid tumor suppressor TET2. However, there is ample evidence to suggest that (R)-2HG has other functionally relevant targets in IDH-mutant cancers. Here, we show that (R)-2HG inhibits KDM5 histone lysine demethylases and that this inhibition contributes to cellular transformation in IDH-mutant AML and IDH-mutant glioma. These studies provide the first evidence of a functional link between dysregulation of histone lysine methylation and transformation in IDH-mutant cancers. SIGNIFICANCE: Mutant IDH is known to induce histone hypermethylation. However, it is not known if this hypermethylation is functionally significant or is a bystander effect of (R)-2HG accumulation in IDH-mutant cells. Here, we provide evidence that KDM5 inhibition by (R)-2HG contributes to mutant IDH-mediated transformation in AML and glioma. This article is highlighted in the In This Issue feature, p. 1275.


Sujet(s)
Gliome , Leucémie aigüe myéloïde , Humains , Histone/métabolisme , Histone Demethylases/génétique , Mutation , Glutarates , Transformation cellulaire néoplasique/génétique , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Gliome/génétique , Méthylation de l'ADN , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme
2.
Epigenetics ; 17(13): 2056-2074, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-35942521

RÉSUMÉ

Ten-Eleven-Translocation 5-methylcytosine dioxygenases 1-3 (TET1-3) convert 5-methylcytosine to 5-hydroxymethylcytosine (5-hmC), using oxygen as a co-substrate. Contrary to expectations, hypoxia induces 5-hmC gains in MYCN-amplified neuroblastoma (NB) cells via upregulation of TET1. Here, we show that MYCN directly controls TET1 expression in normoxia, and in hypoxia, HIF-1 augments TET1 expression and TET1 protein stability. Through gene-editing, we identify two MYCN and HIF-1 binding sites within TET1 that regulate gene expression. Bioinformatic analyses of 5-hmC distribution and RNA-sequencing data from hypoxic cells implicate hypoxia-regulated genes important for cell migration, including CXCR4. We show that hypoxic cells lacking the two MYCN/HIF-1 binding sites within TET1 migrate slower than controls. Treatment of MYCN-amplified NB cells with a CXCR4 antagonist results in slower migration under hypoxic conditions, suggesting that inclusion of a CXCR4 antagonist into NB treatment regimens could be beneficial for children with MYCN-amplified NBs.


In MYCN-amplified neuroblastoma cell lines, MYCN directly controls TET1 expression in normoxia.In MYCN-amplified neuroblastoma cell lines exposed to hypoxia, HIF-1 augments TET1 expression and TET1 protein stability.Hypoxic MYCN-amplified neuroblastoma cell lines have increased cell migration, mediated by genes including CXCR4 that gain 5-hydroxymethylcytosine density.Treatment of MYCN-amplified NB cells with a CXCR4 antagonist slows hypoxia-associated migration, suggesting a CXCR4 antagonist could be beneficial in treatment regimens for children with MYCN-amplified neuroblastomas.


Sujet(s)
5-Méthyl-cytosine , Facteur-1 induit par l'hypoxie , Mixed function oxygenases , Protéine du proto-oncogène N-Myc , Neuroblastome , Protéines proto-oncogènes , Humains , 5-Méthyl-cytosine/métabolisme , Hypoxie cellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Hypoxie/génétique , Mixed function oxygenases/génétique , Mixed function oxygenases/métabolisme , Protéine du proto-oncogène N-Myc/génétique , Protéine du proto-oncogène N-Myc/métabolisme , Neuroblastome/génétique , Neuroblastome/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Facteur-1 induit par l'hypoxie/génétique , Facteur-1 induit par l'hypoxie/métabolisme
4.
J Hum Genet ; 66(11): 1101-1112, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-33980986

RÉSUMÉ

RBL2/p130, a member of the retinoblastoma family of proteins, is a key regulator of cell division and propagates irreversible senescence. RBL2/p130 is also involved in neuronal differentiation and survival, and eliminating Rbl2 in certain mouse strains leads to embryonic lethality accompanied by an abnormal central nervous system (CNS) phenotype. Conflicting reports exist regarding a role of RBL2/p130 in transcriptional regulation of DNA methyltransferases (DNMTs), as well as the control of telomere length. Here we describe the phenotype of three patients carrying bi-allelic RBL2-truncating variants. All presented with infantile hypotonia, severe developmental delay and microcephaly. Malignancies were not reported in carriers or patients. Previous studies carried out on mice and human cultured cells, associated RBL2 loss to DNA methylation and telomere length dysregulation. Here, we investigated whether patient cells lacking RBL2 display related abnormalities. The study of primary patient fibroblasts did not detect abnormalities in expression of DNMTs. Furthermore, methylation levels of whole genome DNA, and specifically of pericentromeric repeats and subtelomeric regions, were unperturbed. RBL2-null fibroblasts show no evidence for abnormal elongation by telomeric recombination. Finally, gradual telomere shortening, and normal onset of senescence were observed following continuous culturing of RBL2-mutated fibroblasts. Thus, this study resolves uncertainties regarding a potential non-redundant role for RBL2 in DNA methylation and telomere length regulation, and indicates that loss of function variants in RBL2 cause a severe autosomal recessive neurodevelopmental disorder in humans.


Sujet(s)
Dysfonctionnement cognitif/génétique , Méthylation de l'ADN/génétique , Protéine p130 de type rétinoblastome/génétique , Raccourcissement des télomères/génétique , Adolescent , Adulte , Allèles , Animaux , Enfant , Dysfonctionnement cognitif/complications , Dysfonctionnement cognitif/physiopathologie , Incapacités de développement/complications , Incapacités de développement/génétique , Incapacités de développement/physiopathologie , Femelle , Fibroblastes/métabolisme , Prédisposition génétique à une maladie , Humains , Mâle , Methyltransferases/génétique , Souris , Microcéphalie/complications , Microcéphalie/génétique , Microcéphalie/physiopathologie , Activité motrice/physiologie , Hypotonie musculaire/complications , Hypotonie musculaire/génétique , Hypotonie musculaire/physiopathologie , Télomère/génétique ,
5.
Blood Adv ; 4(13): 3053-3062, 2020 07 14.
Article de Anglais | MEDLINE | ID: mdl-32634239

RÉSUMÉ

In mammalian cells, cytosines found within cytosine guanine dinucleotides can be methylated to 5-methylcytosine (5-mC) by DNA methyltransferases and further oxidized by the Ten-eleven translocation dioxygenase (TET) enzymes to 5-hydroxymethylcytosine (5-hmC). We have previously shown that hematopoietic stem and progenitor cells (HSPCs) with TET2 mutations have aberrant 5-hmC distribution and less erythroid differentiation potential. However, these experiments were performed under standard tissue culture conditions with 21% oxygen (O2), whereas HSPCs in human bone marrow reside in ∼1% O2. Therefore, to model human erythropoiesis more accurately, we compared 5-hmC distribution and gene expression in hypoxic vs normoxic conditions. Despite TET enzymes having limited O2 as a substrate in hypoxia, 5-hmC peaks were more numerous and pronounced than in normoxia. Among the TET genes, TET3 was upregulated specifically in hypoxia. We identified 2 HIF-1 binding sites in TET3 by chromatin immunoprecipitation of HIF-1α followed by sequencing, and TET3 upregulation was abrogated with deletion of both sites, indicating that TET3 is a direct HIF-1 target. Finally, we showed that loss of one or both of these HIF-1 binding sites in K562 cells disrupted erythroid differentiation in hypoxia and lowered cell viability. This work provides a molecular link between O2 availability, epigenetic modification of chromatin, and erythroid differentiation.


Sujet(s)
Dioxygenases , Protéines proto-oncogènes , Animaux , Cytosine , Protéines de liaison à l'ADN/génétique , Dioxygenases/génétique , Expression des gènes , Humains , Hypoxie/génétique , Protéines proto-oncogènes/génétique
6.
Cell Chem Biol ; 27(5): 538-550.e7, 2020 05 21.
Article de Anglais | MEDLINE | ID: mdl-32101699

RÉSUMÉ

Mitochondrial D2HGDH and L2HGDH catalyze the oxidation of D-2-HG and L-2-HG, respectively, into αKG. This contributes to cellular homeostasis in part by modulating the activity of αKG-dependent dioxygenases. Signals that control the expression/activity of D2HGDH/L2HGDH are presumed to broadly influence physiology and pathology. Using cell and mouse models, we discovered that MYC directly induces D2HGDH and L2HGDH transcription. Furthermore, in a manner suggestive of D2HGDH, L2HGDH, and αKG dependency, MYC activates TET enzymes and RNA demethylases, and promotes their nuclear localization. Consistent with these observations, in primary B cell lymphomas MYC expression positively correlated with enhancer hypomethylation and overexpression of lymphomagenic genes. Together, these data provide additional evidence for the role of mitochondria metabolism in influencing the epigenome and epitranscriptome, and imply that in specific contexts wild-type TET enzymes could demethylate and activate oncogenic enhancers.


Sujet(s)
Alcohol oxidoreductases/génétique , Épigénome , Lymphome B/génétique , Protéines proto-oncogènes c-myc/génétique , Activation de la transcription , Animaux , Lignée cellulaire , Femelle , Humains , Mâle , Souris de lignée C57BL , Transcriptome , Cellules cancéreuses en culture
7.
J Biol Chem ; 291(8): 4256-65, 2016 Feb 19.
Article de Anglais | MEDLINE | ID: mdl-26703470

RÉSUMÉ

The TET enzymes are members of the 2-oxoglutarate-dependent dioxygenase family and comprise three isoenzymes in humans: TETs 1-3. These TETs convert 5-methylcytosine to 5-hydroxymethylcytosine (5-hmC) in DNA, and high 5-hmC levels are associated with active transcription. The importance of the balance in these modified cytosines is emphasized by the fact that TET2 is mutated in several human cancers, including myeloid malignancies such as acute myeloid leukemia (AML). We characterize here the kinetic and inhibitory properties of Tets and show that the Km value of Tets 1 and 2 for O2 is 30 µm, indicating that they retain high activity even under hypoxic conditions. The AML-associated mutations in the Fe(2+) and 2-oxoglutarate-binding residues increased the Km values for these factors 30-80-fold and reduced the Vmax values. Fumarate and succinate, which can accumulate to millimolar levels in succinate dehydrogenase and fumarate hydratase-mutant tumors, were identified as potent Tet inhibitors in vitro, with IC50 values ∼400-500 µm. Fumarate and succinate also down-regulated global 5-hmC levels in neuroblastoma cells and the expression levels of some hypoxia-inducible factor (HIF) target genes via TET inhibition, despite simultaneous HIFα stabilization. The combination of fumarate or succinate treatment with TET1 or TET3 silencing caused differential effects on the expression of specific HIF target genes. Altogether these data show that hypoxia-inducible genes are regulated in a multilayered manner that includes epigenetic regulation via TETs and 5-hmC levels in addition to HIF stabilization.


Sujet(s)
Protéines de liaison à l'ADN/biosynthèse , Dioxygenases/biosynthèse , Fumarates/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Leucémie aigüe myéloïde/métabolisme , Neuroblastome/métabolisme , Protéines proto-oncogènes/biosynthèse , Acide succinique/pharmacologie , Animaux , Hypoxie cellulaire , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/génétique , Dioxygenases/génétique , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Souris , Mixed function oxygenases , Mutation , Neuroblastome/génétique , Protéines proto-oncogènes/génétique
8.
J Virol ; 88(18): 10990-4, 2014 Sep.
Article de Anglais | MEDLINE | ID: mdl-25008927

RÉSUMÉ

Regulation of the lectin galectin 9 (Gal-9) was investigated for the first time during human cytomegalovirus (HCMV) infection. Gal-9 transcription was significantly upregulated in transplant recipients with reactivated HCMV in vivo. In vitro, Gal-9 was potently upregulated by HCMV independently of viral gene expression, with interferon beta (IFN-ß) identified as the mediator of this effect. This study defines an immunoregulatory protein potently increased by HCMV infection and a novel mechanism to control Gal-9 through IFN-ß induction.


Sujet(s)
Infections à cytomégalovirus/génétique , Cytomegalovirus/physiologie , Galectines/génétique , Interféron bêta/métabolisme , Régulation positive , Infections à cytomégalovirus/métabolisme , Infections à cytomégalovirus/virologie , Galectines/métabolisme , Interactions hôte-pathogène , Humains , Interféron bêta/génétique
9.
J Virol ; 87(18): 10273-82, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23864618

RÉSUMÉ

Several human cytomegalovirus (HCMV) genes encode products that modulate cellular functions in a manner likely to enhance viral pathogenesis. This includes UL111A, which encodes homologs of human interleukin-10 (hIL-10). Depending upon signals received, monocytes and macrophages become polarized to either classically activated (M1 proinflammatory) or alternatively activated (M2 anti-inflammatory) subsets. Skewing of polarization toward an M2 subset may benefit the virus by limiting the proinflammatory responses to infection, and so we determined whether HCMV-encoded viral IL-10 influenced monocyte polarization. Recombinant viral IL-10 protein polarized CD14(+) monocytes toward an anti-inflammatory M2 subset with an M2c phenotype, as demonstrated by high expression of CD163 and CD14 and suppression of major histocompatibility complex (MHC) class II. Significantly, in the context of productive HCMV infection, viral IL-10 produced by infected cells polarized uninfected monocytes toward an M2c phenotype. We also assessed the impact of viral IL-10 on heme oxygenase 1 (HO-1), which is an enzyme linked with suppression of inflammatory responses. Polarization of monocytes by viral IL-10 resulted in upregulation of HO-1, and inhibition of HO-1 function resulted in a loss of capacity of viral IL-10 to suppress tumor necrosis factor alpha (TNF-α) and IL-1ß, implicating HO-1 in viral IL-10-induced suppression of proinflammatory cytokines by M2c monocytes. In addition, a functional consequence of monocytes polarized with viral IL-10 was a decreased capacity to activate CD4(+) T cells. This study identifies a novel role for viral IL-10 in driving M2c polarization, which may limit virus clearance by restricting proinflammatory and CD4(+) T cell responses at sites of infection.


Sujet(s)
Cytomegalovirus/immunologie , Interactions hôte-pathogène , Échappement immunitaire , Interleukine-10/immunologie , Monocytes/immunologie , Monocytes/virologie , Facteurs de virulence/immunologie , Antigènes CD/analyse , Antigènes de différenciation des myélomonocytes/analyse , Cytomegalovirus/physiologie , Heme oxygenase-1/analyse , Antigènes d'histocompatibilité de classe II/analyse , Humains , Interleukine-10/métabolisme , Antigènes CD14/analyse , Monocytes/composition chimique , Récepteurs de surface cellulaire/analyse , Protéines virales/immunologie , Protéines virales/métabolisme , Facteurs de virulence/métabolisme
10.
J Virol ; 85(14): 7465-71, 2011 Jul.
Article de Anglais | MEDLINE | ID: mdl-21593144

RÉSUMÉ

The human cytomegalovirus UL111A gene is expressed during latent and productive infections, and it codes for homologs of interleukin-10 (IL-10). We examined whether viral IL-10 expressed during latency altered differentiation of latently infected myeloid progenitors. In comparison to infection with parental virus or mock infection, latent infection with a virus in which the gene encoding viral IL-10 has been deleted upregulated cytokines associated with dendritic cell (DC) formation and increased the proportion of myeloid DCs. These data demonstrate that viral IL-10 restricts the ability of latently infected myeloid progenitors to differentiate into DCs and identifies an immunomodulatory role for viral IL-10 which may limit the host's ability to clear latent virus.


Sujet(s)
Différenciation cellulaire , Cytomegalovirus/métabolisme , Interleukine-10/métabolisme , Séquence nucléotidique , Amorces ADN , Humains , Interleukine-10/génétique , ARN messager/génétique , RT-PCR
11.
J Virol ; 85(11): 5696-700, 2011 Jun.
Article de Anglais | MEDLINE | ID: mdl-21450824

RÉSUMÉ

The human cytomegalovirus (HCMV) ORF94 gene product has been reported to be expressed during both productive and latent phases of infection, although its function is unknown. We report that expression of pORF94 leads to decreased 2',5'-oligoadenylate synthetase (OAS) expression in transfected cells with and without interferon stimulation. Furthermore, the functional activity of OAS was inhibited by pORF94. Finally, we present evidence of OAS modulation by pORF94 during productive HCMV infection of human fibroblasts. This study provides the first identification of a function for pORF94 and identifies an additional means by which HCMV may limit a critical host cell antiviral response.


Sujet(s)
2',5'-Oligoadenylate synthetase/antagonistes et inhibiteurs , Cytomegalovirus/pathogénicité , Interactions hôte-pathogène , Protéines virales/métabolisme , Facteurs de virulence/métabolisme , Cellules cultivées , Fibroblastes/virologie , Humains
12.
Future Microbiol ; 5(6): 883-900, 2010 Jun.
Article de Anglais | MEDLINE | ID: mdl-20521934

RÉSUMÉ

Human cytomegalovirus (HCMV) is a clinically important and ubiquitous herpesvirus. Following primary productive infection the virus is not completely eliminated from the host, but instead establishes a lifelong latent infection without detectable virus production, from where it can reactivate at a later stage to generate new infectious virus. Reactivated HCMV often results in life-threatening disease in immunocompromised individuals, particularly allogeneic stem cell and solid organ transplant recipients, where it remains one of the most difficult opportunistic pathogens that complicate the care of these patients. The ability of HCMV to establish and reactivate from latency is central to its success as a human pathogen, yet latency remains very poorly understood. This article will cover several aspects of HCMV latency, with a focus on current understanding of viral gene expression and functions during this phase of infection.


Sujet(s)
Infections à cytomégalovirus/virologie , Cytomegalovirus/physiologie , Régulation de l'expression des gènes viraux , Latence virale , Cytomegalovirus/pathogénicité , Humains , Activation virale
13.
Virology ; 378(2): 214-25, 2008 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-18614194

RÉSUMÉ

Initiation of human cytomegalovirus (HCMV) productive infection is dependent on the major immediate early (MIE) genes ie1 and ie2. Several putative binding sites for CCAAT displacement protein (CDP or CUX1) were identified within the MIE promoter/regulatory region. Binding assays demonstrated binding of CUX1 to MIE-region oligonucleotides containing the CUX1 core binding sequence ATCGAT and mutagenesis of this sequence abrogated CUX1 binding. Furthermore, CUX1 repressed expression of a luciferase reporter construct controlled by the MIE promoter, and mutation of CUX1 binding sites within the promoter diminished this repressive function of CUX1. In the context of virus infection of HEK293 cells transfected with the CUX1 expression vector, CUX1 showed evidence of association with the HCMV MIE regulatory region and inhibited the capacity of the virus to express ie1 and ie2 transcripts, suggesting that this cellular factor regulates MIE gene expression following virus entry. These data identify a role for CUX1 in repressing HCMV gene expression essential for initiation of the replicative cycle.


Sujet(s)
Cytomegalovirus/physiologie , Régulation de l'expression des gènes viraux , Gènes précoces , Protéines à homéodomaine/métabolisme , Protéines nucléaires/métabolisme , Protéines de répression/métabolisme , Fusion artificielle de gènes , Lignée cellulaire , Gènes rapporteurs , Humains , Luciferases/biosynthèse , Luciferases/génétique , Régions promotrices (génétique) , Liaison aux protéines , Facteurs de transcription
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...