Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 28.827
Filtrer
2.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38960622

RÉSUMÉ

A pleiotropic immunoregulatory cytokine, TGF-ß, signals via the receptor-regulated SMADs: SMAD2 and SMAD3, which are constitutively expressed in normal cells. Here, we show that selective repression of SMAD3 induces cDC differentiation from the CD115+ common DC progenitor (CDP). SMAD3 was expressed in haematopoietic cells including the macrophage DC progenitor. However, SMAD3 was specifically down-regulated in CD115+ CDPs, SiglecH- pre-DCs, and cDCs, whereas SMAD2 remained constitutive. SMAD3-deficient mice showed a significant increase in cDCs, SiglecH- pre-DCs, and CD115+ CDPs compared with the littermate control. SMAD3 repressed the mRNA expression of FLT3 and the cDC-related genes: IRF4 and ID2. We found that one of the SMAD transcriptional corepressors, c-SKI, cooperated with phosphorylated STAT3 at Y705 and S727 to repress the transcription of SMAD3 to induce cDC differentiation. These data indicate that STAT3 and c-Ski induce cDC differentiation by repressing SMAD3: the repressor of the cDC-related genes during the developmental stage between the macrophage DC progenitor and CD115+ CDP.


Sujet(s)
Différenciation cellulaire , Cellules dendritiques , Facteurs de régulation d'interféron , Facteur de transcription STAT-3 , Protéine Smad-3 , Animaux , Différenciation cellulaire/génétique , Cellules dendritiques/métabolisme , Cellules dendritiques/cytologie , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Souris , Facteurs de régulation d'interféron/métabolisme , Facteurs de régulation d'interféron/génétique , Protéine d'inhibition de la différenciation-2/génétique , Protéine d'inhibition de la différenciation-2/métabolisme , Souris knockout , Souris de lignée C57BL , Tyrosine kinase-3 de type fms/génétique , Tyrosine kinase-3 de type fms/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Phosphorylation , Transduction du signal
3.
Mol Genet Genomic Med ; 12(7): e2488, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38963008

RÉSUMÉ

BACKGROUND: This study aimed to identify disease-causing variants within a Chinese family affected by Birt-Hogg-Dubé syndrome (BHDS), which arises from an autosomal dominant inheritance pattern attributed to variants in the folliculin (FLCN) gene, recognized as a tumor suppressor gene. METHODS: A Chinese proband diagnosed with BHDS due to renal tumors underwent next-generation sequencing (NGS), revealing a novel variant in the FLCN gene. Sanger sequencing was subsequently performed on blood samples obtained from family members to confirm the presence of this variant. RESULTS: A novel germline frameshift variant (NM_144997.5:c.977dup) was identified in five individuals among the screened family members, marking the first report of this variant. Additionally, a somatic frameshift variant (NM_144997.5:c.1252del) was detected in the renal tumors of the proband. No variant was detected in unaffected family members. CONCLUSIONS: A novel heterozygous variant was identified in exon 9 of the FLCN gene, which broadens the spectrum of FLCN variants. We recommend that molecular analysis of the FLCN gene be performed in patients with suspected BHDS and their families.


Sujet(s)
Syndrome de Birt-Hogg-Dubé , Mutation avec décalage du cadre de lecture , Pedigree , Protéines proto-oncogènes , Protéines suppresseurs de tumeurs , Humains , Syndrome de Birt-Hogg-Dubé/génétique , Syndrome de Birt-Hogg-Dubé/anatomopathologie , Protéines suppresseurs de tumeurs/génétique , Protéines proto-oncogènes/génétique , Mâle , Femelle , Adulte , Adulte d'âge moyen , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Mutation germinale , Hétérozygote , Peuples d'Asie de l'Est
4.
Elife ; 132024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38989862

RÉSUMÉ

Recent findings indicate that the translation elongation rate influences mRNA stability. One of the factors that has been implicated in this link between mRNA decay and translation speed is the yeast DEAD-box helicase Dhh1p. Here, we demonstrated that the human ortholog of Dhh1p, DDX6, triggers the deadenylation-dependent decay of inefficiently translated mRNAs in human cells. DDX6 interacts with the ribosome through the Phe-Asp-Phe (FDF) motif in its RecA2 domain. Furthermore, RecA2-mediated interactions and ATPase activity are both required for DDX6 to destabilize inefficiently translated mRNAs. Using ribosome profiling and RNA sequencing, we identified two classes of endogenous mRNAs that are regulated in a DDX6-dependent manner. The identified targets are either translationally regulated or regulated at the steady-state-level and either exhibit signatures of poor overall translation or of locally reduced ribosome translocation rates. Transferring the identified sequence stretches into a reporter mRNA caused translation- and DDX6-dependent degradation of the reporter mRNA. In summary, these results identify DDX6 as a crucial regulator of mRNA translation and decay triggered by slow ribosome movement and provide insights into the mechanism by which DDX6 destabilizes inefficiently translated mRNAs.


Sujet(s)
DEAD-box RNA helicases , Biosynthèse des protéines , Protéines proto-oncogènes , Stabilité de l'ARN , ARN messager , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Humains , ARN messager/métabolisme , ARN messager/génétique , Stabilité de l'ARN/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Ribosomes/métabolisme , Cellules HEK293
5.
Cell Rep Med ; 5(7): 101645, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39019012

RÉSUMÉ

Fms-like tyrosine kinase 3 (FLT3) mutations, present in over 30% of acute myeloid leukemia (AML) cases and dominated by FLT3-internal tandem duplication (FLT3-ITD), are associated with poor outcomes in patients with AML. While tyrosine kinase inhibitors (TKIs; e.g., gilteritinib) are effective, they face challenges such as drug resistance, relapse, and high costs. Here, we report that metformin, a cheap, safe, and widely used anti-diabetic agent, exhibits a striking synergistic effect with gilteritinib in treating FLT3-ITD AML. Metformin significantly sensitizes FLT3-ITD AML cells (including TKI-resistant ones) to gilteritinib. Metformin plus gilteritinib (low dose) dramatically suppresses leukemia progression and prolongs survival in FLT3-ITD AML mouse models. Mechanistically, the combinational treatment cooperatively suppresses polo-like kinase 1 (PLK1) expression and phosphorylation of FLT3/STAT5/ERK/mTOR. Clinical analysis also shows improved survival rates in patients with FLT3-ITD AML taking metformin. Thus, the metformin/gilteritinib combination represents a promising and cost-effective treatment for patients with FLT3-mutated AML, particularly for those with low income/affordability.


Sujet(s)
Dérivés de l'aniline , Protéines du cycle cellulaire , Synergie des médicaments , Leucémie aigüe myéloïde , Metformine , Mutation , , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Pyrazines , Transduction du signal , Tyrosine kinase-3 de type fms , Metformine/pharmacologie , Metformine/usage thérapeutique , Tyrosine kinase-3 de type fms/génétique , Tyrosine kinase-3 de type fms/métabolisme , Tyrosine kinase-3 de type fms/antagonistes et inhibiteurs , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/antagonistes et inhibiteurs , Humains , Animaux , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Pyrazines/pharmacologie , Pyrazines/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Souris , Mutation/génétique , Lignée cellulaire tumorale , Thiophènes/pharmacologie , Thiophènes/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Facteur de transcription STAT-5/métabolisme , Facteur de transcription STAT-5/génétique , Femelle , Tests d'activité antitumorale sur modèle de xénogreffe , Mâle , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Sérine-thréonine kinases TOR/métabolisme
7.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38991726

RÉSUMÉ

PPTC7 is a mitochondrial-localized phosphatase that suppresses BNIP3- and NIX-mediated mitophagy, but the mechanisms underlying this regulation remain ill-defined. Here, we demonstrate that loss of PPTC7 upregulates BNIP3 and NIX post-transcriptionally and independent of HIF-1α stabilization. Loss of PPTC7 prolongs the half-life of BNIP3 and NIX while blunting their accumulation in response to proteasomal inhibition, suggesting that PPTC7 promotes the ubiquitin-mediated turnover of BNIP3 and NIX. Consistently, overexpression of PPTC7 limits the accumulation of BNIP3 and NIX protein levels, which requires an intact catalytic motif but is surprisingly independent of its targeting to mitochondria. Consistently, we find that PPTC7 is dual-localized to the outer mitochondrial membrane and the matrix. Importantly, anchoring PPTC7 to the outer mitochondrial membrane is sufficient to blunt BNIP3 and NIX accumulation, and proximity labeling and fluorescence co-localization experiments demonstrate that PPTC7 dynamically associates with BNIP3 and NIX within the native cellular environment. Collectively, these data reveal that a fraction of PPTC7 localizes to the outer mitochondrial membrane to promote the proteasomal turnover of BNIP3 and NIX, limiting basal mitophagy.


Sujet(s)
Protéines membranaires , Mitochondries , Membranes mitochondriales , Protéines mitochondriales , Mitophagie , Protéines proto-oncogènes , Mitophagie/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Humains , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Mitochondries/métabolisme , Membranes mitochondriales/métabolisme , Phosphoprotein Phosphatases/métabolisme , Phosphoprotein Phosphatases/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Cellules HeLa , Animaux
8.
Methods Mol Biol ; 2842: 155-165, 2024.
Article de Anglais | MEDLINE | ID: mdl-39012594

RÉSUMÉ

DNA methylation, one of the most studied epigenetic modifications, regulates many biological processes. Dysregulation of DNA methylation is implicated in the etiology of several diseases, such as cancer and imprinting diseases. Accordingly, technologies designed to manipulate DNA methylation at specific loci are considered worthwhile and many epigenome editing technologies have been developed, which were based on ZF, TALE, and CRISPR-dCas9. Here, we describe a protocol for the application of a modified dCas9-SunTag system, which increased the efficiency of targeted demethylation and gene activation at specific DNA loci. The original SunTag system consists of 10 copies of the GCN4 peptide separated by 5-amino-acid linkers. To achieve more efficient recruitment of an anti-GCN4 scFv fused to the ten-eleven (TET) 1 hydroxylase, an enzyme that demethylates DNA, we changed the linker length to 22 amino acids. Moreover, we describe the co-recruitment of TET1 and VP64 for efficient gene activation. Since we showed the manipulation of DNA methylation at specific loci and gene activation, its application could lead to its future use in the clinic.


Sujet(s)
Systèmes CRISPR-Cas , Méthylation de l'ADN , Humains , Édition de gène/méthodes , Régulation de l'expression des gènes , Épigenèse génétique , Mixed function oxygenases/génétique , Mixed function oxygenases/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , /génétique
9.
Nat Commun ; 15(1): 5514, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951492

RÉSUMÉ

HIV-1 Vpr promotes efficient spread of HIV-1 from macrophages to T cells by transcriptionally downmodulating restriction factors that target HIV-1 Envelope protein (Env). Here we find that Vpr induces broad transcriptomic changes by targeting PU.1, a transcription factor necessary for expression of host innate immune response genes, including those that target Env. Consistent with this, we find silencing PU.1 in infected macrophages lacking Vpr rescues Env. Vpr downmodulates PU.1 through a proteasomal degradation pathway that depends on physical interactions with PU.1 and DCAF1, a component of the Cul4A E3 ubiquitin ligase. The capacity for Vpr to target PU.1 is highly conserved across primate lentiviruses. In addition to impacting infected cells, we find that Vpr suppresses expression of innate immune response genes in uninfected bystander cells, and that virion-associated Vpr can degrade PU.1. Together, we demonstrate Vpr counteracts PU.1 in macrophages to blunt antiviral immune responses and promote viral spread.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Immunité innée , Macrophages , Protéines proto-oncogènes , Transactivateurs , Produits du gène vpr du virus de l'immunodéficience humaine , Humains , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/virologie , Produits du gène vpr du virus de l'immunodéficience humaine/métabolisme , Produits du gène vpr du virus de l'immunodéficience humaine/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Transactivateurs/métabolisme , Transactivateurs/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Infections à VIH/immunologie , Infections à VIH/virologie , Infections à VIH/génétique , Cellules HEK293 , Virion/métabolisme , Protein-Serine-Threonine Kinases
10.
BMC Cancer ; 24(1): 842, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009968

RÉSUMÉ

BACKGROUND: Targeted therapy is now the standard of care in driver-oncogene-positive non-small cell lung cancer (NSCLC). Its initial clinical effects are remarkable. However, almost all patients experience treatment resistance to targeted therapy. Hence, chemotherapy is considered a subsequent treatment option. In patients with driver-oncogene-negative NSCLC, combined immune checkpoint inhibitors (ICIs) and chemotherapy as the first-line therapy has been found to be beneficial. However, the efficacy of ICI plus chemotherapy against driver-oncogene-positive NSCLC other than epidermal growth factor receptor mutation and anaplastic lymphoma kinase fusion is unclear. METHODS: Using the hospital medical records, we retrospectively reviewed advanced or recurrent NSCLC patients who were treated with chemotherapy with or without ICIs at Aichi Cancer Center Hospital between January 2014 and January 2023. Patients with druggable rare mutations such as KRAS-G12C, MET exon 14 skipping, HER2 20 insertion, BRAF-V600E mutations, and ROS1 and RET rearrangements were analyzed. RESULTS: In total, 61 patients were included in this analysis. ICI plus chemotherapy was administered in 36 patients (the ICI-chemo group) and chemotherapy in 25 patients (the chemo group). The median progression-free survival (PFS) rates were 14.0 months in the ICI-chemo group and 4.8 months in the chemo group (hazard ratio [HR] = 0.54, 95% confidence interval [CI] = 0.28-1.01). The median overall survival rates were 31.3 and 21.7 months in the ICI-chemo and chemo groups, respectively (HR = 0.70, 95% CI = 0.33-1.50). Multivariate Cox regression analysis of PFS revealed that HER2 exon 20 insertion mutation was significantly associated with a poorer PFS (HR: 2.39, 95% CI: 1.19-4.77, P = 0.014). Further, ICI-chemo treatment was significantly associated with a better PFS (HR: 0.48, 95% CI: 0.25-0.91, P = 0.025). CONCLUSION: ICI plus chemotherapy improves treatment efficacy in rare driver-oncogene-positive NSCLC.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Mutation , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mâle , Femelle , Études rétrospectives , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Adulte d'âge moyen , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte , Sujet âgé de 80 ans ou plus , Protéines proto-oncogènes B-raf/génétique , Récepteur ErbB-2/génétique , Protéines proto-oncogènes c-ret/génétique , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes/génétique , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/génétique , Survie sans progression , Résultat thérapeutique
11.
Adv Exp Med Biol ; 1459: 243-258, 2024.
Article de Anglais | MEDLINE | ID: mdl-39017847

RÉSUMÉ

Lineage-specific transcription factors (TFs) regulate differentiation of hematopoietic stem cells (HSCs). They are decisive for the establishment and maintenance of lineage-specific gene expression programs during hematopoiesis. For this they create a regulatory network between TFs, epigenetic cofactors, and microRNAs. They activate cell-type specific genes and repress competing gene expression programs. Disturbance of this process leads to impaired lineage fidelity and diseases of the blood system. The TF T-cell acute leukemia 1 (TAL1) is central for erythroid differentiation and contributes to the formation of distinct gene regulatory complexes in progenitor cells and erythroid cells. A TAL1/E47 heterodimer binds to DNA with the TFs GATA-binding factor 1 and 2 (GATA1/2), the cofactors LIM domain only 1 and 2 (LMO1/2), and LIM domain-binding protein 1 (LDB1) to form a core TAL1 complex. Furthermore, cell-type-dependent interactions of TAL1 with other TFs such as with runt-related transcription factor 1 (RUNX1) and Kruppel-like factor 1 (KLF1) are established. Moreover, TAL1 activity is regulated by the formation of TAL1 isoforms, posttranslational modifications (PTMs), and microRNAs. Here, we describe the function of TAL1 in normal hematopoiesis with a focus on erythropoiesis.


Sujet(s)
Érythropoïèse , Protéine-1 de la lleucémie lymphoïde aiguë à cellules T , Protéine-1 de la lleucémie lymphoïde aiguë à cellules T/métabolisme , Protéine-1 de la lleucémie lymphoïde aiguë à cellules T/génétique , Érythropoïèse/génétique , Humains , Animaux , Cellules souches hématopoïétiques/métabolisme , Différenciation cellulaire/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique
12.
Hematol Oncol ; 42(4): e3295, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38979860

RÉSUMÉ

The biological role of Ten-11 translocation 2 (TET2) and the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in the development of extra-nodal natural killer/T-cell lymphoma (ENKTL) remains unclear. The level of 5mC and 5hmC was detected in 112 cases of ENKTL tissue specimens by immunohistochemical (IHC) staining. Subsequently, TET2 knockdown and the overexpression cell models were constructed in ENKTL cell lines. Biochemical analyses were used to assess proliferation, apoptosis, cell cycle and monoclonal formation in cells treated or untreated with L-Ascorbic acid sodium salt (LAASS). Dot-Blots were used to detect levels of genome 5mC and 5hmC. Additionally, the ILLUMINA 850k methylation chip was used to analyze the changes of TET2 regulatory genes. RNA-Seq was used to profile differentially expressed genes regulated by TET2. The global level of 5hmC was significantly decreased, while 5mC was highly expressed in ENKTL tissue. TET2 protein expression was negatively correlated with the ratio of 5mC/5hmC (p < 0.0001). The 5mC/5hmC status were related to the site of disease, clinical stage, PINK score and Ki-67 index, as well as the 5-year OS. TET2 knockdown prolonged the DNA synthesis period, increased the cloning ability of tumor cells, increased the level of 5mC and decreased the level of 5hmC in ENKTL cells. While overexpression of TET2 presented the opposite effect. Furthermore, treatment of ENKTL cells with LAASS significantly induced ENKTL cell apoptosis. These results suggest that TET2 plays an important role in ENKTL development via regulation of 5mC and 5hmC and may serve as a novel therapeutic target for ENKTL.


Sujet(s)
Méthylation de l'ADN , Protéines de liaison à l'ADN , Dioxygenases , Lymphome T-NK extraganglionnaire , Protéines proto-oncogènes , Humains , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Femelle , Mâle , Lymphome T-NK extraganglionnaire/métabolisme , Lymphome T-NK extraganglionnaire/anatomopathologie , Lymphome T-NK extraganglionnaire/génétique , Adulte d'âge moyen , Adulte , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Sujet âgé , Lignée cellulaire tumorale , Prolifération cellulaire
13.
Epigenetics ; 19(1): 2374979, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38970823

RÉSUMÉ

TET1/2/3 dioxygenases iteratively demethylate 5-methylcytosine, beginning with the formation of 5-hydroxymethylcytosine (5hmC). The post-mitotic brain maintains higher levels of 5hmC than most peripheral tissues, and TET1 ablation studies have underscored the critical role of TET1 in brain physiology. However, deletion of Tet1 precludes the disentangling of the catalytic and non-catalytic functions of TET1. Here, we dissect these functions of TET1 by comparing adult cortex of Tet1 wildtype (Tet1 WT), a novel Tet1 catalytically dead mutant (Tet1 HxD), and Tet1 knockout (Tet1 KO) mice. Using DNA methylation array, we uncover that Tet1 HxD and KO mutations perturb the methylation status of distinct subsets of CpG sites. Gene ontology (GO) analysis on specific differential 5hmC regions indicates that TET1's catalytic activity is linked to neuronal-specific functions. RNA-Seq further shows that Tet1 mutations predominantly impact the genes that are associated with alternative splicing. Lastly, we performed High-performance Liquid Chromatography Mass-Spectrometry lipidomics on WT and mutant cortices and uncover accumulation of lysophospholipids lysophosphatidylethanolamine and lysophosphatidylcholine in Tet1 HxD cortex. In summary, we show that Tet1 HxD does not completely phenocopy Tet1 KO, providing evidence that TET1 modulates distinct cortical functions through its catalytic and non-catalytic roles.


Sujet(s)
5-Méthyl-cytosine , Cortex cérébral , Méthylation de l'ADN , Protéines proto-oncogènes , Animaux , Souris , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analogues et dérivés , Cortex cérébral/métabolisme , Souris knockout , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Ilots CpG , Mutation
14.
Front Immunol ; 15: 1421012, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979414

RÉSUMÉ

Objective: This study revealed a core regulator and common upstream mechanisms for the multifaceted pathological processes of age-related macular degeneration (AMD) and provided proof-of-concept for this new therapeutic target. Methods: Comprehensive gene expression analysis was performed using RNA sequencing of eye cup from old mice as well as laser-induced choroidal neovascularization (CNV) mouse model. Through integrative analysis and protein-protein interaction (PPI) analysis, common pathways and key transcription factor was identified simultaneously engaged in age-related retinal degeneration and CNV, the two typical pathological process of AMD. Subsequently, the expression changes of Spi1, the key regulator, as well as the alternation of the downstream mechanisms were validated in both models through qRT-PCR, Elisa, flow cytometry and immunofluorescence. Further, we assessed the impact of Spi1 knockdown in vitro and in vivo using gene intervention vectors carried by adeno-associated virus or lentivirus to test its potential as a therapeutic target. Results: Compared to corresponding controls, we found 1,939 and 1,319 genes differentially expressed in eye cups of old and CNV mice respectively. The integrative analysis identified a total of 275 overlapping DEGs, of which 150 genes were co-upregulated. PPI analysis verified a central transcription factor, SPI1. The significant upregulation of Spi1 expression was then validated in both models, accompanied by macrophage polarization towards the M1 phenotype. Finally, SPI1 suppression significantly inhibited M1 polarization of BMDMs and attenuated neovascularization in CNV mice. Conclusion: This study demonstrates that SPI1 exerts a pivotal role in AMD by regulation of macrophage polarization and innate immune response, offering promise as an innovative target for treating AMD.


Sujet(s)
Néovascularisation choroïdienne , Modèles animaux de maladie humaine , Macrophages , Dégénérescence maculaire , Transactivateurs , Animaux , Dégénérescence maculaire/immunologie , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/génétique , Dégénérescence maculaire/anatomopathologie , Souris , Macrophages/immunologie , Macrophages/métabolisme , Néovascularisation choroïdienne/immunologie , Néovascularisation choroïdienne/génétique , Néovascularisation choroïdienne/métabolisme , Transactivateurs/génétique , Transactivateurs/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Souris de lignée C57BL , Activation des macrophages/génétique , Humains , Analyse de profil d'expression de gènes , Mâle
16.
Cell Mol Life Sci ; 81(1): 284, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38967794

RÉSUMÉ

Hepatocellular carcinoma (HCC) is a malignancy that occurs worldwide and is generally associated with poor prognosis. The development of resistance to targeted therapies such as sorafenib is a major challenge in clinical cancer treatment. In the present study, Ten-eleven translocation protein 1 (TET1) was found to be highly expressed in sorafenib-resistant HCC cells and knockdown of TET1 can substantially improve the therapeutic effect of sorafenib on HCC, indicating the potential important roles of TET1 in sorafenib resistance in HCC. Mechanistic studies determined that TET1 and Yes-associated protein 1 (YAP1) synergistically regulate the promoter methylation and gene expression of DNA repair-related genes in sorafenib-resistant HCC cells. RNA sequencing indicated the activation of DNA damage repair signaling was extensively suppressed by the TET1 inhibitor Bobcat339. We also identified TET1 as a direct transcriptional target of YAP1 by promoter analysis and chromatin-immunoprecipitation assays in sorafenib-resistant HCC cells. Furthermore, we showed that Bobcat339 can overcome sorafenib resistance and synergized with sorafenib to induce tumor eradication in HCC cells and mouse models. Finally, immunostaining showed a positive correlation between TET1 and YAP1 in clinical samples. Our findings have identified a previously unrecognized molecular pathway underlying HCC sorafenib resistance, thus revealing a promising strategy for cancer therapy.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Carcinome hépatocellulaire , Réparation de l'ADN , Résistance aux médicaments antinéoplasiques , Épigenèse génétique , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Protéines proto-oncogènes , Sorafénib , Facteurs de transcription , Protéines de signalisation YAP , Humains , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Épigenèse génétique/effets des médicaments et des substances chimiques , Animaux , Réparation de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN/génétique , Protéines de signalisation YAP/métabolisme , Souris , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Souris nude , Mixed function oxygenases/génétique , Mixed function oxygenases/métabolisme , Voie de signalisation Hippo , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Souris de lignée BALB C , Méthylation de l'ADN/effets des médicaments et des substances chimiques
18.
Front Biosci (Schol Ed) ; 16(2): 10, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38939973

RÉSUMÉ

The ETS transcription factor PU.1 plays an essential role in blood cell development. Its precise expression pattern is governed by cis-regulatory elements (CRE) acting at the chromatin level. CREs mediate the fine-tuning of graded levels of PU.1, deviations of which can cause acute myeloid leukemia. In this review, we perform an in-depth analysis of the regulation of PU.1 expression in normal and malignant hematopoiesis. We elaborate on the role of trans-acting factors and the biomolecular interplays in mediating local chromatin dynamics. Moreover, we discuss the current understanding of CRE bifunctionality exhibiting enhancer or silencer activities in different blood cell lineages and future directions toward gene-specific chromatin-targeted therapeutic development.


Sujet(s)
Hématopoïèse , Protéines proto-oncogènes , Transactivateurs , Humains , Hématopoïèse/génétique , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Transactivateurs/génétique , Transactivateurs/métabolisme , Lignage cellulaire , Animaux , Transcription génétique , Régulation de l'expression des gènes , Leucémie aigüe myéloïde/génétique , Chromatine/métabolisme , Chromatine/génétique
20.
Int J Biol Sci ; 20(8): 3140-3155, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904029

RÉSUMÉ

Cysteine-rich angiogenic inducer 61 (CYR61), also called CCN1, has long been characterized as a secretory protein. Nevertheless, the intracellular function of CYR61 remains unclear. Here, we found that CYR61 is important for proper cell cycle progression. Specifically, CYR61 interacts with microtubules and promotes microtubule polymerization to ensure mitotic entry. Moreover, CYR61 interacts with PLK1 and accumulates during the mitotic process, followed by degradation as mitosis concludes. The proteolysis of CYR61 requires the PLK1 kinase activity, which directly phosphorylates two conserved motifs on CYR61, enhancing its interaction with the SCF E3 complex subunit FBW7 and mediating its degradation by the proteasome. Mutations of phosphorylation sites of Ser167 and Ser188 greatly increase CYR61's stability, while deletion of CYR61 extends prophase and metaphase and delays anaphase onset. In summary, our findings highlight the precise control of the intracellular CYR61 by the PLK1-FBW7 pathway, accentuating its significance as a microtubule-associated protein during mitotic progression.


Sujet(s)
Protéines du cycle cellulaire , Protéine-61 riche en cystéine , Microtubules , Mitose , , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Humains , Mitose/physiologie , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéine-61 riche en cystéine/métabolisme , Protéine-61 riche en cystéine/génétique , Microtubules/métabolisme , Protéine-7 contenant une boite F et des répétitions WD/métabolisme , Protéine-7 contenant une boite F et des répétitions WD/génétique , Cellules HeLa , Phosphorylation , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéines associées aux microtubules/métabolisme , Protéines associées aux microtubules/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE