Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 987
Filtrer
1.
PLoS One ; 19(7): e0305612, 2024.
Article de Anglais | MEDLINE | ID: mdl-38990915

RÉSUMÉ

Breast cancer (BC) cells have a high risk of metastasis due to epithelial-mesenchymal transition (EMT). Palbociclib (CDK4/6 inhibitor) is an approved drug for BC treatment. However, the drug resistance and metastasis can impair the treatment outcome of Palbociclib. Understanding the mechanisms of EMT and Palbociclib drug resistance in BC is conducive to the formulation of novel therapeutic strategy. Here, we investigated the role of circHIAT1/miR-19a-3p/CADM2 axis in modulating EMT and Palbociclib resistance in BC. circHIAT1 and CADM2 were down-regulated in BC tissues and cell lines, and miR-19a-3p showed an up-regulation. circHIAT1 could interact with miR-19a-3p and suppress its activity, while miR-19a-3p functioned to negatively regulate CADM2. Forced over-expression of circHIAT1 could impaired the EMT status and migratory ability of BC cells, and this effect was inhibited by miR-19a-3p mimic. In addition, we also generated Palbociclib resistant BC cells, and showed that circHIAT1 and CADM2 were down-regulated in the resistant BC cells while miR-19a-3p showed an up-regulation. Forced circHIAT1 over-expression re-sensitized BC cells to Palbociclib treatment. Quercetin, a bioactive flavonoid, could suppressed the migration and invasion of BC cells, and re-sensitized BC cells to Palbociclib. The anti-cancer effect of quercetin could be attributed to its regulatory effect on circHIAT1/miR-19a-3p/CADM2 axis. In vivo tumorigenesis experiment further revealed that quercetin administration enhanced the anti-cancer effect of Palbociclib, an effect was dependent on the up-regulation of circHIAT1 by quercetin. In summary, this study identified quercetin as a potential anti-cancer compound to reverse Palbociclib resistance and impair EMT in BC cells by targeting circHIAT1/miR-19a-3p/CADM2 axis.


Sujet(s)
Tumeurs du sein , Kinase-6 cycline-dépendante , Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse , microARN , Pipérazines , Pyridines , Quercétine , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Humains , microARN/génétique , microARN/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pipérazines/pharmacologie , Lignée cellulaire tumorale , Quercétine/pharmacologie , Animaux , Souris , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Mouvement cellulaire/effets des médicaments et des substances chimiques , ARN long non codant/génétique , ARN long non codant/métabolisme , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe
2.
JCO Precis Oncol ; 8: e2400219, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39013131

RÉSUMÉ

PURPOSE: Targeted Agent and Profiling Utilization Registry (TAPUR) is a phase II basket trial evaluating the antitumor activity of commercially available targeted agents in patients with advanced cancer and genomic alterations known to be drug targets. Results of a cohort of patients with soft tissue sarcoma with cyclin-dependent kinase 4 (CDK4) amplification treated with palbociclib are reported. METHODS: Eligible patients had measurable disease, Eastern Cooperative Oncology Group performance status 0 to 2, adequate organ function, and no standard treatment options. The primary end point was disease control (DC), defined as objective response (OR) or stable disease (SD) of at least 16+ weeks duration (SD16+) according to RECIST v1.1. The DC rate was estimated with a 90% CI. Secondary end points included OR, progression-free survival (PFS), overall survival (OS), duration of response, duration of SD, and safety. RESULTS: Forty-two patients with CDK4 amplification were enrolled. One patient was not evaluable for efficacy. One patient with partial response and 18 with SD16+ were observed for DC and OR rates of 46% (90% CI, 36 to 100) and 2% (95% CI, <1 to 13), respectively. Median PFS was 16 weeks (95% CI, 9 to 28) and median OS was 69 weeks (95% CI, 31 to 111) for evaluable patients. Twenty patients had at least one grade 3 to 4 adverse event (AE) at least possibly related to palbociclib, including alanine aminotransferase increase, anemia, fatigue, hypophosphatemia, leukopenia, neutropenia, and thrombocytopenia. No serious AEs were reported. CONCLUSION: Palbociclib met prespecified criteria to declare a signal of antitumor activity in patients with sarcoma and CDK4 amplification.


Sujet(s)
Kinase-4 cycline-dépendante , Pipérazines , Pyridines , Enregistrements , Sarcomes , Humains , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Pyridines/usage thérapeutique , Femelle , Mâle , Adulte d'âge moyen , Pipérazines/usage thérapeutique , Adulte , Sujet âgé , Sarcomes/traitement médicamenteux , Sarcomes/génétique , Amplification de gène , Jeune adulte , Sujet âgé de 80 ans ou plus
3.
Sci Rep ; 14(1): 15786, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982214

RÉSUMÉ

Malignant melanoma (MM) is known for its abundance of genetic alterations and a tendency for rapid metastasizing. Identification of novel plasma biomarkers may enhance non-invasive diagnostics and disease monitoring. Initially, we examined copy number variations (CNV) in CDK genes (CDKN2A, CDKN2B, CDK4) using MLPA (gDNA) and ddPCR (ctDNA) analysis. Subsequently, low-coverage whole genome sequencing (lcWGS) was used to identify the most common CNV in plasma samples, followed by ddPCR verification of chosen biomarkers. CNV alterations in CDK genes were identified in 33.3% of FFPE samples (Clark IV, V only). Detection of the same genes in MM plasma showed no significance, neither compared to healthy plasmas nor between pre- versus post-surgery plasma. Sequencing data showed the most common CNV occurring in 6q27, 4p16.1, 10p15.3, 10q22.3, 13q34, 18q23, 20q11.21-q13.12 and 22q13.33. CNV in four chosen genes (KIF25, E2F1, DIP2C and TFG) were verified by ddPCR using 2 models of interpretation. Model 1 was concordant with lcWGS results in 54% of samples, for model 2 it was 46%. Although CDK genes have not been proven to be suitable CNV liquid biopsy biomarkers, lcWGS defined the most frequently affected chromosomal regions by CNV. Among chosen genes, DIP2C demonstrated a potential for further analysis.


Sujet(s)
Marqueurs biologiques tumoraux , Variations de nombre de copies de segment d'ADN , Mélanome , Humains , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/diagnostic , Biopsie liquide/méthodes , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/sang , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/sang , Inhibiteur p16 de kinase cycline-dépendante/génétique , Kinase-4 cycline-dépendante/génétique , Sujet âgé de 80 ans ou plus , Séquençage du génome entier/méthodes , Inhibiteur p15 de kinase cycline-dépendante/génétique
4.
Proc Natl Acad Sci U S A ; 121(30): e2319574121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39024113

RÉSUMÉ

Regulated cell cycle progression ensures homeostasis and prevents cancer. In proliferating cells, premature S phase entry is avoided by the E3 ubiquitin ligase anaphasepromoting complex/cyclosome (APC/C), although the APC/C substrates whose degradation restrains G1-S progression are not fully known. The APC/C is also active in arrested cells that exited the cell cycle, but it is not clear whether APC/C maintains all types of arrest. Here, by expressing the APC/C inhibitor, EMI1, we show that APC/C activity is essential to prevent S phase entry in cells arrested by pharmacological cyclin-dependent kinases 4 and 6 (CDK4/6) inhibition (Palbociclib). Thus, active protein degradation is required for arrest alongside repressed cell cycle gene expression. The mechanism of rapid and robust arrest bypass from inhibiting APC/C involves CDKs acting in an atypical order to inactivate retinoblastoma-mediated E2F repression. Inactivating APC/C first causes mitotic cyclin B accumulation which then promotes cyclin A expression. We propose that cyclin A is the key substrate for maintaining arrest because APC/C-resistant cyclin A, but not cyclin B, is sufficient to induce S phase entry. Cells bypassing arrest from CDK4/6 inhibition initiate DNA replication with severely reduced origin licensing. The simultaneous accumulation of S phase licensing inhibitors, such as cyclin A and geminin, with G1 licensing activators disrupts the normal order of G1-S progression. As a result, DNA synthesis and cell proliferation are profoundly impaired. Our findings predict that cancers with elevated EMI1 expression will tend to escape CDK4/6 inhibition into a premature, underlicensed S phase and suffer enhanced genome instability.


Sujet(s)
Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Humains , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Complexe promoteur de l'anaphase/métabolisme , Complexe promoteur de l'anaphase/génétique , Lignée cellulaire tumorale , Phase S/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pipérazines/pharmacologie , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Facteurs de transcription E2F/métabolisme , Facteurs de transcription E2F/génétique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Cyclines/métabolisme , Cyclines/génétique , Protéines F-box
6.
EBioMedicine ; 105: 105177, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38924839

RÉSUMÉ

BACKGROUND: The 5-year survival rate of oesophageal squamous cell carcinoma (ESCC) is approximately 20%. The prognosis and drug response exhibit substantial heterogeneity in ESCC, impeding progress in survival outcomes. Our goal is to identify a signature for tumour subtype classification, enabling precise clinical treatments. METHODS: Utilising pre-treatment multi-omics data from an ESCC dataset (n = 310), an enhancer methylation-eRNA-target gene regulation network was constructed and validated by in vitro experiments. Four machine learning methods collectively identified core target genes, establishing an Enhancer Demethylation-Regulated Gene Score (EDRGS) model for classification. The molecular function of EDRGS subtyping was explored in scRNA-seq (n = 60) and bulk-seq (n = 310), and the EDRGS's potential to predict treatment response was assessed in datasets of various cancer types. FINDINGS: EDRGS stratified ESCCs into EDRGS-high/low subtypes, with EDRGS-high signifying a less favourable prognosis in ESCC and nine additional cancer types. EDRGS-high exhibited an immune-hot but immune-suppressive phenotype with elevated immune checkpoint expression, increased T cell infiltration, and IFNγ signalling in ESCC, suggesting a better response to immunotherapy. Notably, EDRGS outperformed PD-L1 in predicting anti-PD-1/L1 therapy effectiveness in ESCC (n = 42), kidney renal clear cell carcinoma (KIRC, n = 181), and bladder urothelial carcinoma (BLCA, n = 348) cohorts. EDRGS-low showed a cell cycle-activated phenotype with higher CDK4 and/or CDK6 expression, demonstrating a superior response to the CDK4/6 inhibitor palbociclib, validated in ESCC (n = 26), melanoma (n = 18), prostate cancer (n = 15) cells, and PDX models derived from patients with pancreatic cancer (n = 30). INTERPRETATION: Identification of EDRGS subtypes enlightens ESCC categorisation, offering clinical insights for patient management in immunotherapy (anti-PD-1/L1) and CDK4/6 inhibitor therapy across cancer types. FUNDING: This study was supported by funding from the National Key R&D Program of China (2021YFC2501000, 2020YFA0803300), the National Natural Science Foundation of China (82030089, 82188102), the CAMS Innovation Fund for Medical Sciences (2021-I2M-1-018, 2022-I2M-2-001, 2021-I2M-1-067), the Fundamental Research Funds for the Central Universities (3332021091).


Sujet(s)
Marqueurs biologiques tumoraux , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Régulation de l'expression des gènes tumoraux , Immunothérapie , Humains , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/génétique , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/traitement médicamenteux , Carcinome épidermoïde de l'oesophage/mortalité , Carcinome épidermoïde de l'oesophage/thérapie , Carcinome épidermoïde de l'oesophage/immunologie , Carcinome épidermoïde de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/anatomopathologie , Immunothérapie/méthodes , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de l'oesophage/thérapie , Tumeurs de l'oesophage/mortalité , Tumeurs de l'oesophage/métabolisme , Pronostic , Méthylation de l'ADN , Éléments activateurs (génétique) , Analyse de profil d'expression de gènes , Biologie informatique/méthodes , Lignée cellulaire tumorale , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Réseaux de régulation génique , Animaux
7.
Sci Rep ; 14(1): 13227, 2024 06 09.
Article de Anglais | MEDLINE | ID: mdl-38851782

RÉSUMÉ

There are hundreds of genes typically overexpressed in breast cancer cells and it's often assumed that their overexpression contributes to cancer progression. However, the precise proportion of these overexpressed genes contributing to tumorigenicity remains unclear. To address this gap, we undertook a comprehensive screening of a diverse set of seventy-two genes overexpressed in breast cancer. This systematic screening evaluated their potential for inducing malignant transformation and, concurrently, assessed their impact on breast cancer cell proliferation and viability. Select genes including ALDH3B1, CEACAM5, IL8, PYGO2, and WWTR1, exhibited pronounced activity in promoting tumor formation and establishing gene dependencies critical for tumorigenicity. Subsequent investigations revealed that CEACAM5 overexpression triggered the activation of signaling pathways involving ß-catenin, Cdk4, and mTOR. Additionally, it conferred a growth advantage independent of exogenous insulin in defined medium and facilitated spheroid expansion by inducing multiple layers of epithelial cells while preserving a hollow lumen. Furthermore, the silencing of CEACAM5 expression synergized with tamoxifen-induced growth inhibition in breast cancer cells. These findings underscore the potential of screening overexpressed genes for both oncogenic drivers and tumor dependencies to expand the repertoire of therapeutic targets for breast cancer treatment.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Transduction du signal , Oncogènes , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Tamoxifène/pharmacologie , Animaux , Molécules d'adhérence cellulaire/génétique , Molécules d'adhérence cellulaire/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Transformation cellulaire néoplasique/génétique
8.
Sci Rep ; 14(1): 13497, 2024 06 12.
Article de Anglais | MEDLINE | ID: mdl-38866982

RÉSUMÉ

Antimicrobial peptides (AMPs) have sparked significant interest as potential anti-cancer agents, thereby becoming a focal point in pursuing novel cancer-fighting strategies. These peptides possess distinctive properties, underscoring the importance of developing more potent and selectively targeted versions with diverse mechanisms of action against human cancer cells. Such advancements would offer notable advantages compared to existing cancer therapies. This research aimed to examine the toxicity and selectivity of the nrCap18 peptide in both cancer and normal cell lines. Furthermore, the rate of cellular death was assessed using apoptosis and acridine orange/ethidium bromide (AO/EB) double staining at three distinct incubation times. Additionally, the impact of this peptide on the cancer cell cycle and migration was evaluated, and ultimately, the expression of cyclin-dependent kinase 4/6 (CDK4/6) genes was investigated. The results obtained from the study demonstrated significant toxicity and selectivity in cancer cells compared to normal cells. Moreover, a strong progressive increase in cell death was observed over time. Furthermore, the peptide exhibited the ability to halt the progression of cancer cells in the G1 phase of the cell cycle and impede their migration by suppressing the expression of CDK4/6 genes.


Sujet(s)
Apoptose , Tumeurs du sein , Cathélicidines , Kinase-4 cycline-dépendante , Humains , Animaux , Lignée cellulaire tumorale , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Femelle , Lapins , Mouvement cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Peptides antimicrobiens cationiques/pharmacologie , Kinase-6 cycline-dépendante/métabolisme , Cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Peptides/pharmacologie , Peptides/composition chimique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
9.
Science ; 384(6695): eadi2421, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38696576

RÉSUMÉ

Cell cycle events are coordinated by cyclin-dependent kinases (CDKs) to ensure robust cell division. CDK4/6 and CDK2 regulate the growth 1 (G1) to synthesis (S) phase transition of the cell cycle by responding to mitogen signaling, promoting E2F transcription and inhibition of the anaphase-promoting complex. We found that this mechanism was still required in G2-arrested cells to prevent cell cycle exit after the S phase. This mechanism revealed a role for CDK4/6 in maintaining the G2 state, challenging the notion that the cell cycle is irreversible and that cells do not require mitogens after passing the restriction point. Exit from G2 occurred during ribotoxic stress and was actively mediated by stress-activated protein kinases. Upon relief of stress, a significant fraction of cells underwent a second round of DNA replication that led to whole-genome doubling.


Sujet(s)
Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Endoréplication , Points de contrôle de la phase G2 du cycle cellulaire , Stress physiologique , Humains , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/génétique , Facteurs de transcription E2F/métabolisme , Facteurs de transcription E2F/génétique , Phase S , Lignée cellulaire
10.
Nucleic Acids Res ; 52(12): 6945-6963, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38783095

RÉSUMÉ

Cellular senescence, a major driver of aging, can be stimulated by DNA damage, and is counteracted by the DNA repair machinery. Here we show that in p16INK4a-deficient cells, senescence induction by the environmental genotoxin B[a]P or ionizing radiation (IR) completely depends on p21CIP1. Immunoprecipitation-based mass spectrometry interactomics data revealed that during senescence induction and maintenance, p21CIP1 specifically inhibits CDK4 and thereby activates the DREAM complex. Genome-wide transcriptomics revealed striking similarities in the response induced by B[a]P and IR. Among the top 100 repressed genes 78 were identical between B[a]P and IR and 76 were DREAM targets. The DREAM complex transcriptionally silences the main proliferation-associated transcription factors E2F1, FOXM1 and B-Myb as well as multiple DNA repair factors. Knockdown of p21CIP1, E2F4 or E2F5 diminished both, repression of these factors and senescence. The transcriptional profiles evoked by B[a]P and IR largely overlapped with the profile induced by pharmacological CDK4 inhibition, further illustrating the role of CDK4 inhibition in genotoxic stress-induced senescence. Moreover, data obtained by live-cell time-lapse microscopy suggest the inhibition of CDK4 by p21CIP1 is especially important for arresting cells which slip through mitosis. Overall, we identified the p21CIP1/CDK4/DREAM axis as a master regulator of genotoxic stress-induced senescence.


Sujet(s)
Vieillissement de la cellule , Kinase-4 cycline-dépendante , Inhibiteur p21 de kinase cycline-dépendante , Altération de l'ADN , Protéines KChIP , Vieillissement de la cellule/effets des radiations , Vieillissement de la cellule/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Humains , Protéines KChIP/métabolisme , Protéines KChIP/génétique , Rayonnement ionisant , Réparation de l'ADN , Régulation de l'expression des gènes/effets des radiations , Protéines de répression
11.
Theriogenology ; 224: 9-18, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38714024

RÉSUMÉ

Sertoli cell (SC) proliferation plays an important role in sperm production and quality; however, the regulatory mechanism of SC proliferation is not well understood. This study investigated the role of adenosine monophosphate-activated protein kinase (AMPK) in the regulation of immature boar SC activity. Cell counting kit-8, Seahorse XFe96, mitochondrial respiratory enzyme-related assay kits, and transmission electron microscopy were used to detect SC proliferative viability, oxygen consumption rate (OCR), mitochondrial respiratory enzyme activity, and the ultrastructure of primary cultured SCs in vitro from the testes of 21-day-old boars. A dual luciferase reporter assay was performed to determine the miRNA-mRNA target interaction. Western blotting was used to analyze cell proliferation-related protein expression of p38, p21, proliferating cell nuclear antigen (PCNA), Cyclin-dependent kinase 4 (CDK4), Cyclin D3, and phosphorylated retinoblastoma protein (Rb). Each experiment had a completely randomized design, with three replicates in each experiment. The results showed that the AMPK inhibitor (Compound C, 20 µM-24 h) increased cell proliferation viability, ATP production, and maximal respiration of SCs by 0.64-, 0.12-, and 0.08-fold (p < 0.05), respectively; increased the SC protein expression of PCNA, CDK4, Cyclin D3, and p-Rb by 0.13-, 0.09-, 0.88-, and 0.12-fold (p < 0.05), respectively; and decreased the SC protein expression of p38 and p21 by 0.36- and 0.27-fold (p < 0.05), respectively. The AMPK agonist AICAR (2 mM-6 h) significantly inhibited SC ultrastructure, OCR, mitochondrial respiratory enzyme activity, and cell proliferation-related protein levels. AMPK was validated to be a target gene of miR-1285 based on the result in which the miR-1285 mimic inhibited the luciferase activity of wild-type AMPK by 0.54-fold (p < 0.001). MiR-1285 mimic promoted the OCR of SCs, with 0.45-, 0.15-, 0.21-, and 0.30-fold (p < 0.01) increases in ATP production, basal and maximal respiration, and spare capacity, respectively. MiR-1285 mimic increased the mitochondrial respiratory enzyme activity of SCs, with 0.63-, 0.70-, and 0.97-fold (p < 0.01) increases in NADH-Q oxidoreductase, cytochrome c oxidase, and ATP synthase, respectively. Moreover, the miR-1285 mimic increased the protein expression of PCNA, CDK4, Cyclin D3, and p-Rb by 0.24-, 0.30-, 0.22-, and 0.13-fold (p < 0.05), respectively, and reduced the protein expression of p38 and p21 by 0.58- and 0.66-fold (p < 0.001). MiR-1285 inhibitor showed opposite effects on the above indicators and induced numerous autophagosomes and large lipid droplets in SCs. A high dose of estradiol (10 µM-6 h, showed a promotion of AMPK activation in a previous study) significantly inhibited SC ultrastructure, mitochondrial function, and proliferation-related pathways, while these adverse effects were weakened by Compound C treatment or miR-1285 mimic transfection. Our findings suggest that the activation and inhibition of AMPK induced by specific drugs or synthesized targeted miRNA fragments could regulate immature boar SC proliferative activity by influencing the CDK4/Cyclin D3 pathway and mitochondrial function; this helps to provide a basis for the prevention and treatment of male sterility in clinical practice.


Sujet(s)
AMP-Activated Protein Kinases , Prolifération cellulaire , Kinase-4 cycline-dépendante , Mitochondries , Cellules de Sertoli , Animaux , Mâle , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Suidae , Mitochondries/métabolisme , AMP-Activated Protein Kinases/métabolisme , AMP-Activated Protein Kinases/génétique , Cellules de Sertoli/métabolisme , Cellules de Sertoli/effets des médicaments et des substances chimiques , Cycline D3/métabolisme , Cycline D3/génétique , Transduction du signal , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cellules cultivées
12.
NEJM Evid ; 3(5): EVIDoa2300231, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38815172

RÉSUMÉ

BACKGROUND: In estrogen receptor-positive metastatic breast cancer, ESR1 mutations (ESR1m) are a common mechanism of acquired resistance to aromatase inhibitors (ArIh). However, the impact ESR1 alterations have on CDK4/6 inhibitor (CDK4/6i) sensitivity has not been established. Analyses of CDK4/6i trials suggest that the endocrine therapy partner and specific ESR1 allele may affect susceptibility. We analyzed a real-world data set to investigate CDK4/6i efficacy in ESR1m metastatic breast cancer and associated clinical factors. METHODS: ESR1m were identified by analysis of circulating-tumor deoxyribonucleic acid. The GuardantINFORM database contains genomic information from tumors linked with claims data. Patients who started a CDK4/6i within 30 days of sequencing were categorized as having ESR1m or non-ESR1-mutant (non-ESR1m) breast cancer. Data were analyzed to determine the real-world time-to-next-treatment, defined as the start of a breast cancer treatment to initiation of the subsequent treatment. RESULTS: One hundred forty-five patients with ESR1m and 612 with non-ESR1m metastatic breast cancer were analyzed. ESR1m and non-ESR1m tumors had similar real-world time-to-next-treatment on CDK4/6i regimens (hazard ratio, 1.02; 95% confidence interval, 0.82 to 1.23). Duration on therapy in the first-line and second-line plus treatment settings were comparable regardless of ESR1 status. We stratified treatment duration by concurrent endocrine therapy, and patients with ESR1m had worse outcomes on ArIh but comparable real-world time-to-next-treatment on fulvestrant. CONCLUSIONS: These data suggest ESR1 variants are not associated with pan-CDK4/6i resistance and are consistent with the hypothesis that CDK4/6 blockade combined with a selective estrogen receptor degrader is potentially an effective option for ESR1m metastatic breast cancer.


Sujet(s)
Tumeurs du sein , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Récepteur alpha des oestrogènes , Mutation , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Femelle , Récepteur alpha des oestrogènes/génétique , Adulte d'âge moyen , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/génétique , Sujet âgé , Adulte , Inhibiteurs de l'aromatase/usage thérapeutique , Pipérazines/usage thérapeutique , Métastase tumorale , Fulvestrant/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique
13.
Int J Biochem Cell Biol ; 172: 106601, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38821314

RÉSUMÉ

Abemaciclib (ABM), a cyclin-dependent kinase 4/6 inhibitor, shows pharmacological effects in cell cycle arrest. Epithelial-mesenchymal transition is an important cellular event associated with pathophysiological states such as organ fibrosis and cancer progression. In the present study, we evaluated the contribution of factors associated with cell cycle arrest to ABM-induced epithelial-mesenchymal transition. Treatment with 0.6 µM ABM induced both cell cycle arrest and epithelial-mesenchymal transition-related phenotypic changes. Interestingly, the knockdown of cyclin-dependent kinase 4/6, pharmacological targets of ABM or cyclin D1, which forms complexes with cyclin-dependent kinase 4/6, resulted in cell cycle arrest at the G1-phase and induction of epithelial-mesenchymal transition, indicating that downregulation of cyclin-dependent kinase 4/6-cyclin D1 complexes would mimic ABM. In contrast, knockdown of the Rb protein, which is phosphorylated by cyclin-dependent kinase 4/6, had no effect on the expression level of α-smooth muscle actin, an epithelial-mesenchymal transition marker. Furthermore, ABM-induced epithelial-mesenchymal transition was not affected by Rb knockdown, suggesting that Rb is not involved in the transition process. Our study is the first to suggest that cyclin-dependent kinase 4/6-cyclin D1 complexes, as pharmacological targets of ABM, may contribute to ABM-induced epithelial-mesenchymal transition, followed by clinical disorders such as organ fibrosis and cancer progression. This study suggests that blocking epithelial-mesenchymal transition might be a promising way to prevent negative side effects caused by a medication (ABM) without affecting its ability to treat the disease.


Sujet(s)
Aminopyridines , Benzimidazoles , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Transition épithélio-mésenchymateuse , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Benzimidazoles/pharmacologie , Humains , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Aminopyridines/pharmacologie , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Cycline D1/métabolisme , Cycline D1/génétique
14.
Cancer Lett ; 593: 216968, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38788968

RÉSUMÉ

In patients with ER + metastatic breast cancer (mBC), the first-line treatment involves the combination of endocrine therapy (ET) and CDK4/6 inhibitors (CDK4/6i). However, a significant group of patients experiences disease progression, emphasizing the urgent clinical need to identify novel anti-tumor therapies. We previously generated breast cancer cells resistant to the combination of fulvestrant (ER downregulator) and abemaciclib (CDK4/6 inhibitor) from MCF7 and T47D (MCF7-FAR and T47D-FAR). RNA-seq-based Gene Set Enrichment Analysis (GSEA) revealed hyper-activation of EGFR, HER2, and AKT signaling in both MCF7-FAR and T47D-FAR. Modulating EGFR or ERBB2 expression through loss- and gain-of-function experiments altered tumor sensitivity to fulvestrant and abemaciclib in parental and FAR spheroids, affecting ERK and AKT/S6 pathways. Cetuximab treatment overcame tumor resistance to fulvestrant and abemaciclib in FAR and EGFR-overexpressing breast cancer spheroids and xenografts. Likewise, patient-derived organoids (PDOs) from individuals with ER + mBC, progressing on palbociclib, exhibited up-regulation of EGFR and HER2 pathways. In conclusion, our findings suggest that inhibiting EGFR and HER2 pathways might overcome resistance to ET + CDK4/6i in selected patients with ER + mBC.


Sujet(s)
Tumeurs du sein , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Récepteur ErbB-2 , Récepteurs des oestrogènes , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Femelle , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Animaux , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/génétique , Récepteurs des oestrogènes/métabolisme , Souris , Fulvestrant/pharmacologie , Fulvestrant/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Benzimidazoles/pharmacologie , Aminopyridines/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Cellules MCF-7 , Lignée cellulaire tumorale , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
16.
Clin Cancer Res ; 30(14): 2996-3005, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38718141

RÉSUMÉ

PURPOSE: Treatment paradigms for isocitrate dehydrogenase (IDH)-mutant gliomas are rapidly evolving. Although typically indolent and responsive to initial treatment, these tumors invariably recur at a higher grade and require salvage treatment. Homozygous deletion of the tumor suppressor gene CDKN2A/B frequently emerges at recurrence in these tumors, driving poor patient outcomes. We investigated the effect of CDK-Rb pathway blockade on IDH-mutant glioma growth in vitro and in vivo using CDK4/6 inhibitors (CDKi). EXPERIMENTAL DESIGN: Cell viability, proliferation assays, and flow cytometry were used to examine the pharmacologic effect of two distinct CDKi, palbociclib and abemaciclib, in multiple patient-derived IDH-mutant glioma lines. Isogenic models were used to directly investigate the influence of CDKN2A/B status on CDKi sensitivity. Orthotopic xenograft tumor models were used to examine the efficacy and tolerability of CDKi in vivo. RESULTS: CDKi treatment leads to decreased cell viability and proliferative capacity in patient-derived IDH-mutant glioma lines, coupled with enrichment of cells in the G1 phase. CDKN2A inactivation sensitizes IDH-mutant glioma to CDKi in both endogenous and isogenic models with engineered CDKN2A deletion. CDK4/6 inhibitor administration improves survival in orthotopically implanted IDH-mutant glioma models. CONCLUSIONS: IDH-mutant gliomas with deletion of CDKN2A/B are sensitized to CDK4/6 inhibitors. These results support the investigation of the use of these agents in a clinical setting.


Sujet(s)
Aminopyridines , Benzimidazoles , Prolifération cellulaire , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Inhibiteur p15 de kinase cycline-dépendante , Inhibiteur p16 de kinase cycline-dépendante , Gliome , Isocitrate dehydrogenases , Mutation , Pipérazines , Inhibiteurs de protéines kinases , Pyridines , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/génétique , Gliome/génétique , Gliome/traitement médicamenteux , Gliome/anatomopathologie , Souris , Inhibiteur p16 de kinase cycline-dépendante/génétique , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/antagonistes et inhibiteurs , Benzimidazoles/pharmacologie , Inhibiteur p15 de kinase cycline-dépendante/génétique , Aminopyridines/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Lignée cellulaire tumorale , Délétion de gène , Tumeurs du cerveau/génétique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Homozygote
17.
Cell Biochem Funct ; 42(4): e4064, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38807466

RÉSUMÉ

Human dental pulp stem cells are a potentially useful resource for cell-based therapies and tissue repair in dental and medical applications. However, the primary culture of isolated dental pulp stem cells has notably been limited. A major requirement of an ideal human dental pulp stem cell culture system is the preservation of efficient proliferation and innate stemness over prolonged passaging, while also ensuring ease of handling through standard, user-friendly culture methods. In this study, we have engineered a novel human dental pulp stem cell line, distinguished by the constitutive expression of telomerase reverse transcriptase (TERT), and the conditional expression of the R24C mutant cyclin-dependent kinase 4 (CDK4R24C) and Cyclin D1. We have named this cell line Tet-off K4DT hDPSCs. Furthermore, we have conducted a comprehensive comparative analysis of their biological attributes in relation to a previously immortalized human dental pulp stem cells, hDPSC-K4DT, which were immortalized by the constitutive expression of CDK4R24C, Cyclin D1 and TERT. In Tet-off K4DT cells, the expression of the K4D genes can be precisely suppressed by the inclusion of doxycycline. Remarkably, Tet-off K4DT cells demonstrated an extended cellular lifespan, increased proliferative capacity, and enhanced osteogenic differentiation potential when compared to K4DT cells. Moreover, Tet-off K4DT cells had no observable genomic aberrations and also displayed a sustained expression of stem cell markers even at relatively advanced passages. Taken together, the establishment of this new cell line holds immense promise as powerful experimental tool for both fundamental and applied research involving dental pulp stem cells.


Sujet(s)
Prolifération cellulaire , Kinase-4 cycline-dépendante , Pulpe dentaire , Doxycycline , Cellules souches , Humains , Pulpe dentaire/cytologie , Pulpe dentaire/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Doxycycline/pharmacologie , Cellules souches/métabolisme , Cellules souches/cytologie , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Telomerase/métabolisme , Telomerase/génétique , Cycline D1/métabolisme , Cycline D1/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées
18.
Int J Mol Sci ; 25(9)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38732173

RÉSUMÉ

Lung adenocarcinoma (LUAD) is the most prevalent and aggressive subtype of lung cancer, exhibiting a dismal prognosis with a five-year survival rate below 5%. DEAD-box RNA helicase 18 (DDX18, gene symbol DDX18), a crucial regulator of RNA metabolism, has been implicated in various cellular processes, including cell cycle control and tumorigenesis. However, its role in LUAD pathogenesis remains elusive. This study demonstrates the significant upregulation of DDX18 in LUAD tissues and its association with poor patient survival (from public databases). Functional in vivo and in vitro assays revealed that DDX18 knockdown potently suppresses LUAD progression. RNA sequencing and chromatin immunoprecipitation experiments identified cyclin-dependent kinase 4 (CDK4), a cell cycle regulator, as a direct transcriptional target of DDX18. Notably, DDX18 depletion induced G1 cell cycle arrest, while its overexpression promoted cell cycle progression even in normal lung cells. Interestingly, while the oncogenic protein c-Myc bound to the DDX18 promoter, it did not influence its expression. Collectively, these findings establish DDX18 as a potential oncogene in LUAD, functioning through the CDK4-mediated cell cycle pathway. DDX18 may represent a promising therapeutic target for LUAD intervention.


Sujet(s)
Adénocarcinome pulmonaire , Kinase-4 cycline-dépendante , DEAD-box RNA helicases , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , Animaux , Humains , Souris , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Cycle cellulaire/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris nude , Régulation positive
19.
Cancer Res ; 84(14): 2247-2264, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38657118

RÉSUMÉ

Alveolar soft part sarcoma (ASPS) is a rare mesenchymal malignancy driven by the ASPSCR1::TFE3 fusion. A better understanding of the mechanisms by which this oncogenic transcriptional regulator drives cancer growth is needed to help identify potential therapeutic targets. In this study, we characterized the transcriptional and chromatin landscapes of ASPS tumors and preclinical models, identifying the essential role of ASPSCR1::TFE3 in tumor cell viability by regulating core transcriptional programs involved in cell proliferation, angiogenesis, and mitochondrial biology. ASPSCR1::TFE3 directly interacted with key epigenetic regulators at enhancers and promoters to support ASPS-associated transcription. Among the effector programs driven by ASPSCR1::TFE3, cell proliferation was driven by high levels of cyclin D1 expression. Disruption of cyclin D1/CDK4 signaling led to a loss of ASPS proliferative capacity, and combined inhibition of CDK4/6 and angiogenesis halted tumor growth in xenografts. These results define the ASPS oncogenic program, reveal mechanisms by which ASPSCR1::TFE3 controls tumor biology, and identify a strategy for therapeutically targeting tumor cell-intrinsic vulnerabilities. Significance: The ASPSCR1::TFE3 fusion propels the growth of alveolar soft part sarcoma  by activating transcriptional programs that regulate proliferation, angiogenesis, mitochondrial biogenesis, and differentiation and can be therapeutically targeted to improve treatment.


Sujet(s)
Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Prolifération cellulaire , Protéines de fusion oncogènes , Sarcome alvéolaire des parties molles , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Sarcome alvéolaire des parties molles/génétique , Sarcome alvéolaire des parties molles/anatomopathologie , Sarcome alvéolaire des parties molles/métabolisme , Humains , Animaux , Souris , Prolifération cellulaire/génétique , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Régulation de l'expression des gènes tumoraux , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Femelle , Néovascularisation pathologique/génétique , Néovascularisation pathologique/anatomopathologie , Néovascularisation pathologique/métabolisme , Protéines et peptides de signalisation intracellulaire
20.
Mod Pathol ; 37(6): 100494, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38621503

RÉSUMÉ

GLI1(12q13.3) amplification is identified in a subset of mesenchymal neoplasms with a distinct nested round cell/epithelioid phenotype. MDM2 and CDK4 genes are situated along the oncogenic 12q13-15 segment, amplification of which defines well-differentiated liposarcoma (WDLPS)/dedifferentiated liposarcoma (DDLPS). The 12q amplicon can occasionally include GLI1, a gene in close proximity to CDK4. We hereby describe the first cohort of GLI1/MDM2/CDK4 coamplified WD/DDLPS. The departmental database was queried retrospectively for all cases of WD/DDLPS having undergone next-generation (MSK-IMPACT) sequencing with confirmed MDM2, CDK4, and GLI1 coamplification. Clinicopathologic data was obtained from a review of the medical chart and available histologic material. Four hundred eighty-six WD/DDLPS cases underwent DNA sequencing, 92 (19%) of which harbored amplification of the GLI1 locus in addition to that of MDM2 and CDK4. These included primary tumors (n = 60), local recurrences (n = 29), and metastases (n = 3). Primary tumors were most frequently retroperitoneal (47/60, 78%), mediastinal (4/60, 7%), and paratesticular (3/60, 5%). Average age was 63 years, with a male:female ratio of 3:2. The cohort was comprised of DDLPS (86/92 [93%], 6 of which were WDLPS with early dedifferentiation) and WDLPS without any longitudinal evidence of dedifferentiation (6/92, 7%). One-fifth (13/86, 17%) of DDLPS cases showed no evidence of a well-differentiated component in any of the primary, recurrent, or metastatic specimens. Dedifferentiated areas mostly showed high-grade undifferentiated pleomorphic sarcoma-like (26/86,30%) and high-grade myxofibrosarcoma-like (13/86,16%) morphologies. A disproportionately increased incidence of meningothelial whorls with/without osseous metaplasia was observed as the predominant pattern in 16/86 (19%) cases, and GLI1-altered morphology as described was identified in a total of 10/86 (12%) tumors. JUN (1p32.1), also implicated in the pathogenesis of WD/DDLPS, was coamplified with all 3 of MDM2, CDK4, and GLI1 in 7/91 (8%) cases. Additional loci along chromosomal arms 1p and 6q, including TNFAIP3, LATS1, and ESR1, were also amplified in a subset of cases. In this large-scale cohort of GLI1 coamplified WD/DDLPS, we elucidate uniquely recurrent features including meningothelial whorl-like and GLI-altered morphology in dedifferentiated areas. Assessment of tumor location (retroperitoneal or mediastinal), identification of a well-differentiated liposarcoma component, and coamplification of other spatially discrete genomic segments (1p and 6q) might aid in distinction from tumors with true driver GLI1 alterations.


Sujet(s)
Kinase-4 cycline-dépendante , Amplification de gène , Liposarcome , Protéine à doigt de zinc GLI1 , Femelle , Humains , Mâle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Kinase-4 cycline-dépendante/génétique , Liposarcome/génétique , Liposarcome/anatomopathologie , Protéines proto-oncogènes c-mdm2/génétique , Études rétrospectives , Protéine à doigt de zinc GLI1/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...