Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27.760
Filtrer
1.
J Clin Invest ; 134(19)2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39352379

RÉSUMÉ

Clonal hematopoiesis of indeterminate potential (CHIP) has emerged as a previously unrecognized, potent, age-related, and common risk factor for atherosclerosis. Somatic mutations in certain known leukemia driver genes give rise to clones of mutant cells in peripheral blood. The increased risk of developing hematologic malignancy does not, on its own, explain excess mortality in individuals with CHIP. Cardiovascular disease accounts for much of this gap. Experimental evidence supports the causality of certain CHIP mutations in accelerated atherosclerosis. CHIP due to mutations in different driver genes varies in their promotion of atherosclerotic events and in the region of augmented atherosclerotic involvement. For example, CHIP due to mutations in DNMT3a appears less atherogenic than CHIP that arises from TET2 or JAK2, forms of CHIP that incite inflammation. The recognition of certain CHIP mutations as promoters of atherosclerotic risk has opened new insights into understanding of the pathophysiology of this disease. The accentuated cardiovascular risk and involvement of distinct pathways of various forms of CHIP also inform novel approaches to allocation of targeted therapies, affording a step toward personalized medicine.


Sujet(s)
Athérosclérose , Hématopoïèse clonale , DNA methyltransferase 3A , Dioxygenases , Mutation , Humains , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Hématopoïèse clonale/génétique , DNA methyltransferase 3A/génétique , DNA (cytosine-5-)-methyltransferase/génétique , DNA (cytosine-5-)-methyltransferase/métabolisme , Animaux , Kinase Janus-2/génétique , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Facteurs de risque
2.
Cell Death Dis ; 15(10): 720, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39353904

RÉSUMÉ

Nucleotide-binding oligomerization domain 2 (NOD2) is an immune sensor crucial for eliciting the innate immune responses. Nevertheless, discrepancies exist regarding the effect of NOD2 on different types of cancer. This study aimed to investigate these function of NOD2 in melanoma and its underlying mechanisms. We have validated the tumor suppressor effect of NOD2 in melanoma. NOD2 inhibited the proliferation of melanoma cells, hindering their migration and invasion while promoting the onset of apoptosis. Our study showed that NOD2 expression is closely related to pyrimidine and folate metabolism. NOD2 inhibits thymidylate synthase (TYMS) expression by promoting K48-type ubiquitination modification of TYMS, thereby decreasing the resistance of melanoma cells to 5-fluorouracil (5-FU) and capecitabine (CAP). TYMS was identified to form a complex with Polo-like Kinase 1 (PLK1) and activate the PLK1 signaling pathway. Furthermore, we revealed that the combination of the PLK1 inhibitor volasertib (BI6727) with 5-FU or CAP had a synergistic effect repressing the proliferation, migration, and autophagy of melanoma cells. Overall, our research highlights the protective role of NOD2 in melanoma and suggests that targeting NOD2 and the TYMS/PLK1 signaling axis is a high-profile therapy that could be a prospect for melanoma treatment.


Sujet(s)
Protéines du cycle cellulaire , Prolifération cellulaire , Résistance aux médicaments antinéoplasiques , Mélanome , Protéine adaptatrice de signalisation NOD2 , , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Transduction du signal , Thymidylate synthase , Humains , Protein-Serine-Threonine Kinases/métabolisme , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Mélanome/métabolisme , Mélanome/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Thymidylate synthase/métabolisme , Thymidylate synthase/génétique , Protéine adaptatrice de signalisation NOD2/métabolisme , Protéine adaptatrice de signalisation NOD2/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Fluorouracil/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Ptéridines/pharmacologie , Animaux , Souris , Ubiquitination/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques
3.
Mol Biol (Mosk) ; 58(2): 270-281, 2024.
Article de Russe | MEDLINE | ID: mdl-39355884

RÉSUMÉ

The complement inhibitor CD55/DAF is expressed on many cell types. Dysregulation of CD55 expression is associated with increased disease severity in influenza A infection and vascular complications in pathologies that involve excessive activation of the complement system. A luciferase reporter system was used to functionally analyze the single nucleotide polymorphism rs2564978 in the U937 human promonocytic cell line. The polymorphism is in the promoter of the CD55 gene, and its minor allele T is associated with a severe course of influenza A(H1N1)pdm09. A decreased activity of the CD55 promoter carrying the minor rs2564978(T) allele was observed in activated U937 cells, which provide a cell model of human macrophages. Using bioinformatics resources, PU.1 was identified as a potential transcription factor that may bind to the CD55 promoter at the rs2564978 site in an allele-specific manner. The involvement of PU.1 in modulating CD55 promoter activity was verified by a PU.1 genetic knockdown with small interfering RNAs under specific monocyte activation conditions.


Sujet(s)
Allèles , Grippe humaine , Macrophages , Polymorphisme de nucléotide simple , Régions promotrices (génétique) , Protéines proto-oncogènes , Transactivateurs , Humains , Transactivateurs/génétique , Transactivateurs/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Macrophages/métabolisme , Cellules U937 , Grippe humaine/génétique , Sites de fixation , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/pathogénicité , Régulation de l'expression des gènes
4.
Sci Rep ; 14(1): 20713, 2024 09 05.
Article de Anglais | MEDLINE | ID: mdl-39237684

RÉSUMÉ

Lidamycin (LDM) has been confirmed to have a strong anti-pancreatic cancer effect and can affect the mitochondrial function of pancreatic cancer cells. Mitofusin-2 (Mfn2) is located in the outer membrane of mitochondria, and Mfn2 is currently believed to play a role in cancer inhibition in pancreatic cancer. In order to explore whether the anti-pancreatic cancer effect of LDM is related to Mfn2-mediated mitophagy, Bioinformatics and in vitro cell experiments are used for experimental research. The experimental results demonstrated that Mfn2 is correlated with mitochondrial autophagy in pancreatic cancer. Lidamycin can increase the expression of Mfn2 in pancreatic cancer and affect the process of EMT, affect the level of reactive oxygen species and mitochondrial membrane potential, and increase the expression of mitochondrial autophagy marker proteins BNIP3L and Beclin1. These results demonstrate that Mfn2 affects mitophagy in pancreatic cancer cells by regulating the expression of Mfn2.


Sujet(s)
dGTPases , Protéines membranaires , Protéines mitochondriales , Mitophagie , Tumeurs du pancréas , Humains , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Mitophagie/effets des médicaments et des substances chimiques , dGTPases/métabolisme , dGTPases/génétique , Lignée cellulaire tumorale , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Aminosides/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Bécline-1/métabolisme , Bécline-1/génétique , Protéines proto-oncogènes/métabolisme , Protéines suppresseurs de tumeurs
5.
Science ; 385(6713): 1098-1104, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39236175

RÉSUMÉ

Accurate chromosome segregation requires the attachment of microtubules to centromeres, epigenetically defined by the enrichment of CENP-A nucleosomes. During DNA replication, CENP-A nucleosomes undergo dilution. To preserve centromere identity, correct amounts of CENP-A must be restored in a cell cycle-controlled manner orchestrated by the Mis18 complex (Mis18α-Mis18ß-Mis18BP1). We demonstrate here that PLK1 interacts with the Mis18 complex by recognizing self-primed phosphorylations of Mis18α (Ser54) and Mis18BP1 (Thr78 and Ser93) through its Polo-box domain. Disrupting these phosphorylations perturbed both centromere recruitment of the CENP-A chaperone HJURP and new CENP-A loading. Biochemical and functional analyses showed that phosphorylation of Mis18α and PLK1 binding were required to activate Mis18α-Mis18ß and promote Mis18 complex-HJURP interaction. Thus, our study reveals key molecular events underpinning the licensing role of PLK1 in ensuring accurate centromere inheritance.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéines du cycle cellulaire , Protéine A du centromère , Centromère , Protéines chromosomiques nonhistones , , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Humains , Protéines du cycle cellulaire/métabolisme , Centromère/métabolisme , Protéine A du centromère/métabolisme , Protéines chromosomiques nonhistones/métabolisme , Ségrégation des chromosomes , Protéines de liaison à l'ADN/métabolisme , Cellules HeLa , Phosphorylation , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéines adaptatrices de la transduction du signal/métabolisme
6.
Science ; 385(6713): 1091-1097, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39236163

RÉSUMÉ

The centromere, a chromosome locus defined by the histone H3-like protein centromeric protein A (CENP-A), promotes assembly of the kinetochore to bind microtubules during cell division. Centromere maintenance requires CENP-A to be actively replenished by dedicated protein machinery in the early G1 phase of the cell cycle to compensate for its dilution after DNA replication. Cyclin-dependent kinases (CDKs) limit CENP-A deposition to once per cell cycle and function as negative regulators outside of early G1. Antithetically, Polo-like kinase 1 (PLK1) promotes CENP-A deposition in early G1, but the molecular details of this process are still unknown. We reveal here a phosphorylation network that recruits PLK1 to the deposition machinery to control a conformational switch required for licensing the CENP-A deposition reaction. Our findings clarify how PLK1 contributes to the epigenetic maintenance of centromeres.


Sujet(s)
Protéines du cycle cellulaire , Protéine A du centromère , Centromère , Protéines chromosomiques nonhistones , Épigenèse génétique , , Humains , Protéines du cycle cellulaire/métabolisme , Centromère/métabolisme , Protéine A du centromère/métabolisme , Protéines chromosomiques nonhistones/métabolisme , Phase G1 , Cellules HeLa , Kinétochores/métabolisme , Phosphorylation , /génétique , /métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique
7.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273363

RÉSUMÉ

MDM4 is upregulated in the majority of melanoma cases and has been described as a "key therapeutic target in cutaneous melanoma". Numerous isoforms of MDM4 exist, with few studies examining their specific expression in human tissues. The changes in splicing of MDM4 during human melanomagenesis are critical to p53 activity and represent potential therapeutic targets. Compounding this, studies relying on short reads lose "connectivity" data, so full transcripts are frequently only inferred from the presence of splice junction reads. To address this problem, long-read nanopore sequencing was utilized to read the entire length of transcripts. Here, MDM4 transcripts, both alternative and canonical, are characterized in a pilot cohort of human melanoma specimens. RT-PCR was first used to identify the presence of novel splice junctions in these specimens. RT-qPCR then quantified the expression of major MDM4 isoforms observed during sequencing. The current study both identifies and quantifies MDM4 isoforms present in melanoma tumor samples. In the current study, we observed high expression levels of MDM4-S, MDM4-FL, MDM4-A, and the previously undescribed Ensembl transcript MDM4-209. A novel transcript lacking both exons 6 and 9 is observed and named MDM4-A/S for its resemblance to both MDM4-A and MDM4-S isoforms.


Sujet(s)
Mélanome , Isoformes de protéines , Humains , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/métabolisme , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/métabolisme , Épissage alternatif , Régulation de l'expression des gènes tumoraux , Séquençage par nanopores/méthodes
8.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273420

RÉSUMÉ

Radiation therapy continues to be the cornerstone treatment for malignant brain tumors, the majority of which express wild-type p53. Therefore, the identification of drugs that promote the ionizing radiation (IR)-induced activation of p53 is expected to increase the efficacy of radiation therapy for these tumors. The growth inhibitory effects of CEP-1347, a known inhibitor of MDM4 expression, on malignant brain tumor cell lines expressing wild-type p53 were examined, alone or in combination with IR, by dye exclusion and/or colony formation assays. The effects of CEP-1347 on the p53 pathway, alone or in combination with IR, were examined by RT-PCR and Western blot analyses. The combination of CEP-1347 and IR activated p53 in malignant brain tumor cells and inhibited their growth more effectively than either alone. Mechanistically, CEP-1347 and IR each reduced MDM4 expression, while their combination did not result in further decreases. CEP-1347 promoted IR-induced Chk2 phosphorylation and increased p53 expression in concert with IR in a Chk2-dependent manner. The present results show, for the first time, that CEP-1347 is capable of promoting Chk2-mediated p53 activation by IR in addition to inhibiting the expression of MDM4 and, thus, CEP-1347 has potential as a radiosensitizer for malignant brain tumors expressing wild-type p53.


Sujet(s)
Tumeurs du cerveau , Checkpoint kinase 2 , Rayonnement ionisant , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Checkpoint kinase 2/métabolisme , Checkpoint kinase 2/génétique , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Phosphorylation/effets des médicaments et des substances chimiques , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des radiations
9.
Commun Biol ; 7(1): 1112, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39256555

RÉSUMÉ

Recruitment of non-canonical BCOR-PRC1.1 to non-methylated CpG islands via KDM2B plays a fundamental role in transcription control during developmental processes and cancer progression. However, the mechanism is still largely unknown on how this recruitment is regulated. Here, we unveiled the importance of the Poly-D/E regions within the linker of BCOR for its binding to KDM2B. Interestingly, we also demonstrated that these negatively charged Poly-D/E regions on BCOR play autoinhibitory roles in liquid-liquid phase separation (LLPS) of BCORANK-linker-PUFD/PCGF1RAWUL. Through neutralizing negative charges of these Poly-D/E regions, Ca2+ not only weakens the interaction between BCOR/PCGF1 and KDM2B, but also promotes co-condensation of the enzymatic core of BCOR-PRC1.1 with KDM2B into liquid-like droplet. Accordingly, we propose that Ca2+ could modulate the compartmentation and recruitment of the enzymatic core of BCOR-PRC1.1 on KDM2B target loci. Thus, our finding advances the mechanistic understanding on how the tethering of BCOR-PRC1.1 enzymatic core to KDM2B is regulated.


Sujet(s)
Calcium , Jumonji Domain-Containing Histone Demethylases , Complexe répresseur Polycomb-1 , Protéines proto-oncogènes , Protéines de répression , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Calcium/métabolisme , Humains , Jumonji Domain-Containing Histone Demethylases/métabolisme , Jumonji Domain-Containing Histone Demethylases/génétique , Jumonji Domain-Containing Histone Demethylases/composition chimique , Complexe répresseur Polycomb-1/métabolisme , Complexe répresseur Polycomb-1/génétique , Liaison aux protéines , , Protéines F-box
10.
Folia Biol (Praha) ; 70(2): 113-122, 2024.
Article de Anglais | MEDLINE | ID: mdl-39231319

RÉSUMÉ

Recent studies have highlighted the significant role of 5-hydroxymethylcytosine (5hmC) in carcinogenesis. However, the specific role of 5hmC in osteosarcoma (OS) remains largely unexplored. The-re-fore, this study aimed to investigate the function of 5hmC and TET3 in OS. In this study, we found a decreased total level of 5hmC in OS tissues. The expression of the TET3 protein was also decreased in OS. Importantly, the decreased levels of TET3 were associated with a decreased disease-free survival (DFS) rate in patients. To investigate the role of TET3 and 5hmC in OS, we manipulated the levels of TET3 in MG-63 cells. Silencing TET3 in these cells resulted in a twofold increase in proliferation. Additio-nally, the level of 5hmC decreased in these cells. Con-versely, over-expression of TET3 in MG-63 cells led to the expected inhibition of proliferation and invasion, accompanied by an increase in 5hmC levels. In conclusion, both 5hmC and TET3 protein levels were decreased in OS. Additionally, the over-expression of TET3 inhibited the proliferation of MG-63 cells, while the suppression of TET3 had the opposite effect. These findings suggest that decreased levels of 5hmC and TET3 may serve as potential markers for OS.


Sujet(s)
5-Méthyl-cytosine , Prolifération cellulaire , Déméthylation de l'ADN , Dioxygenases , Épigenèse génétique , Femelle , Humains , Mâle , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Lignée cellulaire tumorale , Dioxygenases/métabolisme , Régulation de l'expression des gènes tumoraux , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Ostéosarcome/anatomopathologie , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique
11.
Clin Epigenetics ; 16(1): 125, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39261937

RÉSUMÉ

BACKGROUND: Breast tumorigenesis is a complex and multistep process accompanied by both genetic and epigenetic dysregulation. In contrast to the extensive studies on DNA epigenetic modifications 5-hydroxymethylcytosine (5hmC) and 5-methylcytosine (5mC) in malignant breast tumors, their roles in the early phases of breast tumorigenesis remain ambiguous. RESULTS: DNA 5hmC and 5mC exhibited a consistent and significant decrease from usual ductal hyperplasia to atypical ductal hyperplasia and subsequently to ductal carcinoma in situ (DCIS). However, 5hmC showed a modest increase in invasive ductal breast cancer compared to DCIS. Genomic analyses showed that the changes in 5hmC and 5mC levels occurred around the transcription start sites (TSSs), and the modification levels were strongly correlated with gene expression levels. Meanwhile, it was found that differentially hydroxymethylated regions (DhMRs) and differentially methylated regions (DMRs) were overlapped in the early phases and accompanied by the enrichment of active histone marks. In addition, TET2-related DNA demethylation was found to be involved in breast tumorigenesis, and four transcription factor binding sites (TFs: ESR1, FOXA1, GATA3, FOS) were enriched in TET2-related DhMRs/DMRs. Intriguingly, we also identified a certain number of common DhMRs between tumor samples and cell-free DNA (cfDNA). CONCLUSIONS: Our study reveals that dynamic changes in DNA 5hmC and 5mC play a vital role in propelling breast tumorigenesis. Both TFs and active histone marks are involved in TET2-related DNA demethylation. Concurrent changes in 5hmC signals in primary breast tumors and cfDNA may play a promising role in breast cancer screening.


Sujet(s)
5-Méthyl-cytosine , Tumeurs du sein , Protéines de liaison à l'ADN , Dioxygenases , Protéines proto-oncogènes , Humains , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Femelle , Tumeurs du sein/génétique , Protéines de liaison à l'ADN/génétique , Dioxygenases/génétique , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Carcinogenèse/génétique , Méthylation de l'ADN/génétique , Épigenèse génétique/génétique , Régulation de l'expression des gènes tumoraux , Déméthylation de l'ADN
12.
Cells ; 13(17)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39273014

RÉSUMÉ

Overcoming temozolomide (TMZ)-resistance is a major challenge in glioblastoma therapy. Therefore, identifying the key molecular player in chemo-resistance becomes urgent. We previously reported the downregulation of PDCD10 in primary glioblastoma patients and its tumor suppressor-like function in glioblastoma cells. Here, we demonstrate that the loss of PDCD10 causes a significant TMZ-resistance during treatment and promotes a rapid regrowth of tumor cells after treatment. PDCD10 knockdown upregulated MGMT, a key enzyme mediating chemo-resistance in glioblastoma, accompanied by increased expression of DNA mismatch repair genes, and enabled tumor cells to evade TMZ-induced cell-cycle arrest. These findings were confirmed in independent models of PDCD10 overexpressing cells. Furthermore, PDCD10 downregulation led to the dedifferentiation of glioblastoma cells, as evidenced by increased clonogenic growth, the upregulation of glioblastoma stem cell (GSC) markers, and enhanced neurosphere formation capacity. GSCs derived from PDCD10 knockdown cells displayed stronger TMZ-resistance and regrowth potency, compared to their parental counterparts, indicating that PDCD10-induced stemness may independently contribute to tumor malignancy. These data provide evidence for a dual role of PDCD10 in tumor suppression by controlling both chemo-resistance and dedifferentiation, and highlight PDCD10 as a potential prognostic marker and target for combination therapy with TMZ in glioblastoma.


Sujet(s)
Protéines régulatrices de l'apoptose , Résistance aux médicaments antinéoplasiques , Glioblastome , Témozolomide , Humains , Glioblastome/anatomopathologie , Glioblastome/génétique , Glioblastome/métabolisme , Glioblastome/traitement médicamenteux , Témozolomide/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , DNA modification methylases/métabolisme , DNA modification methylases/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Enzymes de réparation de l'ADN/métabolisme , Enzymes de réparation de l'ADN/génétique
13.
Methods Enzymol ; 703: 87-120, 2024.
Article de Anglais | MEDLINE | ID: mdl-39261005

RÉSUMÉ

In DNA, methylation at the fifth position of cytosine (5mC) by DNA methyltransferases is essential for eukaryotic gene regulation. Methylation patterns are dynamically controlled by epigenetic machinery. Erasure of 5mC by Fe2+ and 2-ketoglutarate (2KG) dependent dioxygenases in the ten-eleven translocation family (TET1-3), plays a key role in nuclear processes. Through the event of active demethylation, TET proteins iteratively oxidize 5mC to 5-hydroxymethyl cytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxycytosine (5caC), each of which has been implicated in numerous diseases when aberrantly generated. A wide range of biochemical assays have been developed to characterize TET activity, many of which require multi-step processing to detect and quantify the 5mC oxidized products. Herein, we describe the development and optimization of a sensitive MALDI mass spectrometry-based technique that directly measures TET activity and eliminates tedious processing steps. Employing optimized assay conditions, we report the steady-state activity of wild type TET2 enzymes to furnish 5hmC, 5fC and 5caC. We next determine IC50 values of several small-molecule inhibitors of TETs. The utility of this assay is further demonstrated by analyzing the activity of V1395A which is an activating mutant of TET2 that primarily generates 5caC. Lastly, we describe the development of a secondary assay that utilizes bisulfite chemistry to further examine the activity of wildtype TET2 and V1395A in a base-resolution manner. The combined results demonstrate that the activity of TET proteins can be gauged, and their products accurately quantified using our methods.


Sujet(s)
5-Méthyl-cytosine , Protéines de liaison à l'ADN , Dioxygenases , Protéines proto-oncogènes , Dioxygenases/métabolisme , Dioxygenases/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Humains , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analyse , 5-Méthyl-cytosine/composition chimique , Spectrométrie de masse MALDI/méthodes , Dosages enzymatiques/méthodes , Mixed function oxygenases/métabolisme , Mixed function oxygenases/génétique , Mixed function oxygenases/composition chimique , Méthylation de l'ADN , Cytosine/analogues et dérivés , Cytosine/analyse , Cytosine/métabolisme , Cytosine/composition chimique , Oxydoréduction
14.
Front Biosci (Landmark Ed) ; 29(9): 334, 2024 Sep 24.
Article de Anglais | MEDLINE | ID: mdl-39344337

RÉSUMÉ

BACKGROUND: Ventilator-induced lung injury (VILI) is a consequence of inflammation and increased alveolar-capillary membrane permeability due to alveolar hyperdistention or elevated intrapulmonary pressure, but the precise mechanisms remain unclear. The aim of the study was to analyze the mechanism by which angiotensin converting enzyme 2 (ACE2) alleviates endoplasmic reticulum stress (ERS) and protects alveolar cells from pyroptosis in VILI by regulating angiotensin (Ang)1-7/Mas. METHODS: VILI was induced in mice by mechanical ventilation by regulating the tidal volume. The alveolar cell line, A549, mimics VILI in vitro by cyclic stretch (CS). Ang (1-7) (100 nmol/L) was added to the medium. ERS was induced in cells by stimulating with tunicamycin (TM, 2 µg/mL). ERS was inhibited by tracheal instillation of 4-phenylbutyric acid (4-PBA) (1 mg/kg). ACE2's enzymatic function was activated or inhibited by subcutaneous injection of resorcinolnaphthalein (RES, 20 µg/kg) or MLN-4760 (20 µg/kg). pGLV-EF1a-GFP-ACE2 was instilled into the trachea to increase the protein expression of ACE2. The Ang (1-7) receptor, Mas, was antagonized by injecting A779 subcutaneously (80 µg/kg). RESULTS: ACE2 protein levels decreased after modeling. Ang (1-7) level was decreased and Ang II was accumulated. ERS was significantly induced in VILI mice, and pyroptosis was observed in cells. When ERS was inhibited, pyroptosis under the VILI condition was significantly inhibited. Ang (1-7) alleviated ERS and pyroptosis under CS. When ERS was continuously activated, the function of Ang (1-7) in inhibiting pyroptosis was blocked. Resorcinolnaphthalein (RES) effectively promoted Ang II conversion, alleviated the Ang (1-7) level in VILI, ameliorated lung injury, and inhibited ERS and cell pyroptosis. Inhibiting ACE2's function in VILI hindered the production of Ang (1-7), promoted the accumulation of Ang II, and exacerbated ERS and pyroptosis, along with lung injury. The Mas antagonist significantly blocked the inhibitory effects of ACE2 on ERS and pyroptosis in VILI. CONCLUSIONS: Reduced ACE2 expression in VILI is involved in ERS and pyroptosis-related injury. ACE2 can alleviate ERS in alveolar cells by catalyzing the production of Ang (1-7), thus inhibiting pyroptosis in VILI.


Sujet(s)
Angiotensine-I , Angiotensin-converting enzyme 2 , Stress du réticulum endoplasmique , Souris de lignée C57BL , Fragments peptidiques , Proto-oncogène Mas , Pyroptose , Lésion pulmonaire induite par la ventilation mécanique , Animaux , Pyroptose/effets des médicaments et des substances chimiques , Lésion pulmonaire induite par la ventilation mécanique/métabolisme , Lésion pulmonaire induite par la ventilation mécanique/prévention et contrôle , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Angiotensine-I/métabolisme , Angiotensine-I/pharmacologie , Fragments peptidiques/métabolisme , Fragments peptidiques/pharmacologie , Mâle , Souris , Peptidyl-Dipeptidase A/métabolisme , Peptidyl-Dipeptidase A/génétique , Protéines proto-oncogènes/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Humains
15.
Sci Rep ; 14(1): 22191, 2024 09 27.
Article de Anglais | MEDLINE | ID: mdl-39333184

RÉSUMÉ

The ROS1 receptor tyrosine kinase (RTK) possesses the largest extracellular amino-terminal domain (ECD) among the human RTK family, yet the mechanisms regulating its activation are not fully understood. While chimeric ROS1 fusion proteins, resulting from chromosomal rearrangements, are well-known oncogenic drivers, their activation mechanisms also remain underexplored. To elucidate the role of the ROS1 ECD in catalytic regulation, we engineered a series of amino-terminal deletion mutants. Our functional studies compared the full-length ROS1 receptor, the CD74-ROS1 oncogenic fusion, and ECD-deleted ROS1 constructs, identifying the ECD regions that inhibit ROS1 tyrosine kinase activity. Notably, we found that deletion of the ROS1 ECD alone significantly increases constitutive catalytic activation and neoplastic transformation in the absence of an amino-terminal fusion partner, challenging the presumed necessity for a dimerization domain in the activation mechanism of kinase fusions in cancer. Our data suggest that inter-genic deletions resulting in the loss of the ECD may be underappreciated oncogenic drivers in cancer. Furthermore, our studies demonstrate that RNASE7 is not a ligand for the ROS1 receptor as previously reported, confirming that ROS1 remains an orphan receptor. Thus, the discovery of a ROS1 ligand remains an important future priority. These findings highlight the potential for disease-associated somatic aberrations or splice variants that modify the ROS1 ECD to promote constitutive receptor activation, warranting further investigation.


Sujet(s)
Domaines protéiques , Protein-tyrosine kinases , Protéines proto-oncogènes , Humains , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/composition chimique , Animaux , Souris , Protéines de fusion oncogènes/métabolisme , Protéines de fusion oncogènes/génétique , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Cellules HEK293 , Activation enzymatique
16.
Genes (Basel) ; 15(9)2024 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-39336822

RÉSUMÉ

The protein menin is encoded by the MEN1 gene and primarily serves as a nuclear scaffold protein, regulating gene expression through its interaction with and regulation of chromatin modifiers and transcription factors. While the scope of menin's functions continues to expand, one area of growing investigation is the role of menin in cancer. Menin is increasingly recognized for its dual function as either a tumor suppressor or a tumor promoter in a highly tumor-dependent and context-specific manner. While menin serves as a suppressor of neuroendocrine tumor growth, as seen in the cancer risk syndrome multiple endocrine neoplasia type 1 (MEN1) syndrome caused by pathogenic germline variants in MEN1, recent data demonstrate that menin also suppresses cholangiocarcinoma, pancreatic ductal adenocarcinoma, gastric adenocarcinoma, lung adenocarcinoma, and melanoma. On the other hand, menin can also serve as a tumor promoter in leukemia, colorectal cancer, ovarian and endometrial cancers, Ewing sarcoma, and gliomas. Moreover, menin can either suppress or promote tumorigenesis in the breast and prostate depending on hormone receptor status and may also have mixed roles in hepatocellular carcinoma. Here, we review the rapidly expanding literature on the role and function of menin across a broad array of different cancer types, outlining tumor-specific differences in menin's function and mechanism of action, as well as identifying its therapeutic potential and highlighting areas for future investigation.


Sujet(s)
Tumeurs , Protéines proto-oncogènes , Humains , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Animaux , Néoplasie endocrinienne multiple de type 1/génétique , Néoplasie endocrinienne multiple de type 1/métabolisme , Régulation de l'expression des gènes tumoraux
17.
Funct Integr Genomics ; 24(5): 161, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39285026

RÉSUMÉ

Epigenetic regulation plays a role in Parkinson's disease (PD), and ten-eleven translocation methylcytosine dioxygenase 1 (TET1) catalyzes the first step in DNA demethylation by converting 5-methylcytosine to 5-hydroxymethylcytosine. We investigated whether TET1 binds to the promoter of the transient receptor potential cation channel subfamily V member 1 (TRPV1) and regulates its expression, thereby controlling oxidative stress in PD. TRPV1 was identified as an oxidative stress-associated gene in the GSE20186 dataset including substantia nigra from 14 patients with PD and 14 healthy controls and the Genecards database. Lentiviral vectors were used to manipulate Trpv1 expression in rats, followed by 6-hydroxydopamine hydrochloride (6-OHDA) injection for modeling. Behavioral tests, immunofluorescence, Nissl staining, western blot assays, DHE fluorescent probe, biochemical analysis, and ELISA were conducted to assess oxidative stress and neurotoxicity. Trpv1 expression was significantly reduced in the brain tissues of 6-OHDA-treated Parkinsonian rats. Trpv1 alleviated behavioral dysfunction, oxidative stress, and dopamine neuron loss in rats. TET1 mediated TRPV1 hydroxymethylation to promote its expression, and Trpv1 inhibition reversed the mitigating effect of Tet1 on oxidative stress and behavioral dysfunction in PD. TRPV1 activated the AMPK signaling by promoting AMPK phosphorylation to alleviate neurotoxicity and oxidative stress in SH-SY5Y cells. Tet1-mediated Trpv1 hydroxymethylation modification promotes the Ampk signaling activation, thereby eliciting neuroprotection in 6-OHDA-treated Parkinsonian rats. These findings provide experimental evidence that targeting the TET1/TRPV1 axis may be neuroprotective for PD by acting on the AMPK signaling.


Sujet(s)
Méthylation de l'ADN , Maladie de Parkinson , Transduction du signal , Canaux cationiques TRPV , Animaux , Humains , Mâle , Rats , AMP-Activated Protein Kinases/métabolisme , AMP-Activated Protein Kinases/génétique , Dioxygenases , Modèles animaux de maladie humaine , Épigenèse génétique , Mixed function oxygenases/métabolisme , Mixed function oxygenases/génétique , Neuroprotecteurs/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Oxidopamine , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Rat Sprague-Dawley , Canaux cationiques TRPV/métabolisme , Canaux cationiques TRPV/génétique
18.
Sci Adv ; 10(38): eadq5226, 2024 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-39292770

RÉSUMÉ

Macrophages orchestrate tissue homeostasis and immunity. In the tumor microenvironment (TME), macrophage presence is largely associated with poor prognosis because of their reprogramming into immunosuppressive cells. We investigated the effects of hypoxia, a TME-associated feature, on the functional, epigenetic, and transcriptional reprogramming of macrophages and found that hypoxia boosts their immunogenicity. Hypoxic inflammatory macrophages are characterized by a cluster of proinflammatory genes undergoing ten-eleven translocation-mediated DNA demethylation and overexpression. These genes are regulated by NF-κB, while HIF1α dominates the transcriptional reprogramming, demonstrated through ChIP-seq and pharmacological inhibition. In bladder and ovarian carcinomas, hypoxic inflammatory macrophages are enriched in immune-infiltrated tumors, correlating with better patient prognoses. Coculture assays and cell-cell communication analyses support that hypoxic-activated macrophages enhance T cell-mediated responses. The NF-κB-associated hypomethylation signature is displayed by a subset of hypoxic inflammatory macrophages, isolated from ovarian tumors. Our results challenge paradigms regarding the effects of hypoxia on macrophages and highlight actionable target cells to modulate anticancer immune responses.


Sujet(s)
Reprogrammation cellulaire , Protéines de liaison à l'ADN , Dioxygenases , Macrophages , Facteur de transcription NF-kappa B , Protéines proto-oncogènes , Microenvironnement tumoral , Humains , Hypoxie cellulaire , Lignée cellulaire tumorale , Méthylation de l'ADN , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Régulation de l'expression des gènes tumoraux , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Macrophages/métabolisme , Macrophages/immunologie , Facteur de transcription NF-kappa B/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Microenvironnement tumoral/immunologie , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/immunologie , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/génétique
19.
Int J Mol Sci ; 25(18)2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39337534

RÉSUMÉ

Recent advancements highlight the intricate interplay between the extracellular matrix (ECM) and immune responses, notably in respiratory diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD). The ECM, a dynamic structural framework within tissues, orches-trates a plethora of cellular processes, including immune cell behavior and tissue repair mecha-nisms. WNT1-inducible-signaling pathway protein 1 (WISP1), a key ECM regulator, controls immune cell behavior, cytokine production, and tissue repair by modulating integrins, PI3K, Akt, ß-catenin, and mTOR signaling pathways. WISP1 also induces macrophage migration inhibitory factor (MIF) expression via Src kinases and epidermal growth factor receptor (EGFR) activation. MIF, through its wide range of activities, enhances inflammation and tissue restructuring. Rec-ognized for its versatile roles in regulating the immune system, MIF interacts with multiple immune components, such as the NLRP3 inflammasome, thereby sustaining inflammatory pro-cesses. The WISP1-MIF axis potentially unveils complex molecular mechanisms governing im-mune responses and inflammation. Understanding the intricate roles of WISP1 and MIF in the pathogenesis of chronic respiratory diseases such as asthma and COPD could lead to the identi-fication of novel targets for therapeutic intervention to alleviate disease severity and enhance patient outcomes.


Sujet(s)
Asthme , Protéines CCN de signalisation intercellulaire , Facteurs inhibiteurs de la migration des macrophages , Protéines proto-oncogènes , Broncho-pneumopathie chronique obstructive , Humains , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/immunologie , Protéines CCN de signalisation intercellulaire/métabolisme , Asthme/métabolisme , Asthme/immunologie , Asthme/traitement médicamenteux , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Protéines proto-oncogènes/métabolisme , Animaux , Inflammation/métabolisme , Transduction du signal , Intramolecular oxidoreductases
20.
BMC Cancer ; 24(1): 1200, 2024 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-39342179

RÉSUMÉ

BACKGROUND: Acute promyelocytic leukemia (APL) is the sub-type of Acute myeloid leukemia (AML) which is described by differentiation block at promyelocytic stage and t(15; 17) translocation with All trans retinoic acid (ATRA) and arsenic trioxide (ATO) as standard treatments. Chronic myeloid leukemia (CML) translocation t (19; 22) causes a rise in granulocytes and their immature precursors in the blood. Different mutations cause resistance to first-line tyrosine kinase therapies in CML. Beside drug resistance, leukemia stem cells (LSC) are critical resources for relapse and resistance in APL and CML. The drug toxicity and resistant profile associated with LSC and current therapeutics of APL and CML necessitate the development of new therapies. Imidazoles are heterocyclic nitrogen compounds with diverse cellular actions. The purpose of this research was to assess the anti-leukemic properties of four novel imidazole derivatives including L-4, L-7, R-35, and R-NIM04. METHODS AND RESULTS: Pharmacological and biochemical approaches were used which showed that all four imidazole derivatives interfere with the NB4 cells proliferation, an APL cell line, while only L-7 exhibit anti-proliferative activity against K562 cells, a CML cell line. The anti-proliferative effect of imidazole derivatives was linked to apoptosis induction. Further real-time polymerase chain reaction (RT-PCR) analysis revealed downregulation of AXL-Receptor Tyrosine Kinase (AXL-RTK) and target genes of Wnt/beta-catenin pathway like c-Myc, Axin2 and EYA3. An additive effect was observed after combinatorial treatment of L-7 with standard drugs ATRA or Imatinib on the proliferation of NB4 and K562 cells respectively which was related to further downregulation of target genes of Wnt/beta catenin pathway. CONCLUSION: Imidazole derivatives significantly reduce proliferation of NB4 and K562 cells by inducing apoptosis, down regulating of AXL-RTK and Wnt/ß-catenin target genes.


Sujet(s)
Apoptose , Prolifération cellulaire , Imidazoles , Humains , Imidazoles/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Cellules K562 , Axl Receptor Tyrosine Kinase , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme , Leucémie aiguë promyélocytaire/traitement médicamenteux , Leucémie aiguë promyélocytaire/anatomopathologie , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Leucémie myéloïde/traitement médicamenteux , Leucémie myéloïde/anatomopathologie , Leucémie myéloïde/métabolisme , Leucémie myéloïde/génétique , Voie de signalisation Wnt/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE