Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.281
Filtrer
1.
Clinics (Sao Paulo) ; 79: 100415, 2024.
Article de Anglais | MEDLINE | ID: mdl-38897099

RÉSUMÉ

INTRODUCTION: Patients with Human Papillomavirus (HPV+)-associated Laryngeal Squamous Cell Carcinoma (LSCC) exhibit dramatically improved survival relative to those with HPV-Negative (HPV-) tumors. In this study, the authors aimed to investigate the radiosensitivity of all available confirmed HPV+ and HPV-LSCC cells in vitro and in vivo. METHODS: Primary LSCC cells were generated from tumor specimens obtained from patients. Real-time PCR was performed to confirm HPV infection and the expression of HPV-related genes (E6 and E7), p53, and pRB. Clonogenic survival assays, western blotting, and flow cytometry were used to assess radiation sensitivity, apoptosis, and the expression of p53 and pRB. p53 and pRB knockout cells were generated using CRISPR/Cas9 technology. RESULTS: HPV+ LSCC cells displayed enhanced radiation sensitivity compared to HPV- cells. Radiation-induced apoptosis in HPV+ LSCC cells, accompanied by increased levels of p53 and pRB. Knockout of p53 or pRB led to radiation resistance and attenuated radiation-induced apoptosis in HPV+ LSCC cells. In vivo experiments showed similar results, where knockout of p53 or pRB decreased radiosensitivity in tumor-bearing mice. CONCLUSION: The present findings demonstrated that HPV+ LSCC cells displayed obvious inherent radiation sensitivity, corresponding to increased apoptosis following radiation exposure. Mechanism study showed that the expression of p53 and pRB in HPV+ cells are required for radiation sensitivity. These findings highlight a novel mechanism by which p53 and pRB play key roles in the radiation sensitivity of HPV+ LSCC compared to HPV-LSCC.


Sujet(s)
Apoptose , Carcinome épidermoïde , Tumeurs du larynx , Infections à papillomavirus , Radiotolérance , Protéine p53 suppresseur de tumeur , Humains , Tumeurs du larynx/radiothérapie , Tumeurs du larynx/virologie , Carcinome épidermoïde/radiothérapie , Carcinome épidermoïde/virologie , Protéine p53 suppresseur de tumeur/métabolisme , Infections à papillomavirus/radiothérapie , Infections à papillomavirus/virologie , Infections à papillomavirus/complications , Apoptose/effets des radiations , Animaux , Lignée cellulaire tumorale , Réaction de polymérisation en chaine en temps réel , Mâle , Souris , Cytométrie en flux , Technique de Western , Protéine du rétinoblastome/métabolisme
2.
Biochem Pharmacol ; 225: 116310, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38788960

RÉSUMÉ

Targeting the DNA damage response (DDR) is a promising strategy in oncotherapy, as most tumor cells are sensitive to excess damage due to their repair defects. Ataxia telangiectasia mutated and RAD3-related protein (ATR) is a damage response signal transduction sensor, and its therapeutic potential in tumor cells needs to be precisely investigated. Herein, we identified a new axis that could be targeted by ATR inhibitors to decrease the DNA-dependent protein kinase catalytic subunit (DNAPKcs), downregulate the expression of the retinoblastoma (RB), and drive G1/S-phase transition. Four-way DNA Holliday junctions (FJs) assembled in this process could trigger S-phase arrest and induce lethal chromosome damage in RB-positive triple-negative breast cancer (TNBC) cells. Furthermore, these unrepaired junctions also exerted toxic effects to RB-deficient TNBC cells when the homologous recombination repair (HRR) was inhibited. This study proposes a precise strategy for treating TNBC by targeting the DDR and extends our understanding of ATR and HJ in tumor treatment.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie , ADN cruciforme , Tumeurs du sein triple-négatives , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Protéines mutées dans l'ataxie-télangiectasie/génétique , Humains , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/traitement médicamenteux , Lignée cellulaire tumorale , ADN cruciforme/métabolisme , ADN cruciforme/génétique , Protéine du rétinoblastome/métabolisme , Protéine du rétinoblastome/génétique , Femelle , Phase S/effets des médicaments et des substances chimiques , Phase S/physiologie , Animaux , Antinéoplasiques/pharmacologie , Altération de l'ADN/physiologie , Altération de l'ADN/effets des médicaments et des substances chimiques
3.
Cancer Lett ; 593: 216956, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38735381

RÉSUMÉ

Anti-CDK4/6 therapy has been employed for the treatment for head and neck squamous cell carcinoma (HNSCC) with CDK4/6 hyperactivation, but the response rate is relatively low. In this study, we first showed that CDK4 and CDK6 was over-expressed and conferred poor prognosis in HNSCC. Moreover, in RB-positive HNSCC, STAT3 signaling was activated induced by CDK4/6 inhibition and STAT3 promotes RB deficiency by upregulation of MYC. Thirdly, the combination of Stattic and CDK4/6 inhibitor results in striking anti-tumor effect in vitro and in Cal27 derived animal models. Additionally, phospho-STAT3 level negatively correlates with RB expression and predicts poor prognosis in patients with HNSCC. Taken together, our findings suggest an unrecognized function of STAT3 confers to CDK4/6 inhibitors resistance and presenting a promising combination strategy for patients with HNSCC.


Sujet(s)
Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Tumeurs de la tête et du cou , Inhibiteurs de protéines kinases , Facteur de transcription STAT-3 , Carcinome épidermoïde de la tête et du cou , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Facteur de transcription STAT-3/métabolisme , Animaux , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , Lignée cellulaire tumorale , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique , Femelle , Mâle , Souris nude , Souris , Protéine du rétinoblastome/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Phosphorylation
4.
Discov Med ; 36(184): 923-935, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38798252

RÉSUMÉ

BACKGROUND: SGI-1027 is a recognized inhibitor of DNA methyltransferase 1 (DNMT1), and earlier investigations have indicated an inverse correlation between dysregulated DNMT1 expression in gastric cancer (GC) and retinoblastoma 1 (RB1) gene expression. Despite this knowledge, the precise mechanisms underlying the action of SGI-1027 in GC cells remain inadequately comprehended. The primary objective of this study is to elucidate the impact of SGI-1027 on the behavior of GC cells, encompassing aspects such as growth and metastatic potential, by intervening in DNMT1, thereby influencing the regulation of RB1 gene expression. METHOD: The acquisition of the normal gastric mucosal cell line GES-1 and the human gastric cancer cell line MKN45 was followed by employing Western blot (WB) and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) techniques to evaluate the expression levels of RB1 and DNMT1 in these two cell lines. Subsequently, the MKN45 cell line was cultured in medium containing varying concentrations of SGI-1027, and the impact of SGI-1027 on the regulation of RB1 and DNMT1 in GC cells was reassessed using WB and qRT-PCR techniques. To scrutinize the effect of SGI-1027 on GC cells, we utilized the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H tetrazolium bromide (MTT) assay to determine cell proliferation and performed Transwell experiments to assess cell migration and invasion capabilities. Throughout this process, we also employed WB to assess the levels of cell cycle-associated proteins (Cyclin D1, Cyclin E1, and Cyclin B1) and proteins related to apoptosis (BCL-2 associated protein X apoptosis regulator (BAX) and B-cell lymphoma 2 apoptosis regulator (BCL-2)). Furthermore, we injected the MKN45 cell line and MKN45 cell line cultured with the optimal concentration of SGI-1027 for 5 days and 10 days into mice subcutaneously and through the tail vein, dividing them into the Model group, Model+SGI-1027 5d group, and Model+SGI-1027 10d group. We monitored changes in tumor size and volume in mice, and tumor tissues as well as lung tissues were collected for hematoxylin and eosin (HE) staining. Finally, DNMT1 expression levels in GC tissues were detected using both WB and immunohistochemistry (IHC) techniques. Additionally, RB1 expression levels in GC tissues were assessed using WB. RESULT: In contrast to GES-1 cells, MKN45 cells displayed a distinctive profile characterized by increased DNMT1 expression and decreased RB1 expression (p < 0.05). However, upon the introduction of SGI-1027, a notable decrease in DNMT1 levels within GC cells was observed, concomitant with an elevation in RB1 gene expression, with 25 µmol/L SGI-1027 identified as the optimal concentration (p < 0.05). Functional assays demonstrated that SGI-1027-treated GC cells exhibited pronounced features of inhibited proliferation, migration, and invasion when compared to untreated MKN45 cells (p < 0.05). Moreover, in SGI-1027-treated GC cells, the levels of Cyclin D1, Cyclin E1, Cyclin B1, and BCL-2 were significantly reduced, while the expression level of BAX increased (p < 0.05). Notably, the most pronounced impact was observed at 25 µmol/L SGI-1027, further underscoring its regulatory effects on tumor cell behavior (p < 0.05). In animal experiments, the Model group exhibited a substantial increase in tumor volume, with HE staining results indicating extensive necrosis in most gastric tissues and noticeable signs of lung metastasis, accompanied by increased DNMT1 expression and decreased RB1 gene expression. In contrast, the SGI-1027 group displayed a reduction in gastric tumor volume, decreased necrosis, and reduced lung tumor metastasis (p < 0.05). Additionally, the expression of DNMT1 was significantly reduced in SGI-1027-treated GC cells, while RB1 expression increased (p < 0.05), further confirming the inhibitory effects of SGI-1027 on tumor growth and metastasis. CONCLUSIONS: SGI-1027 effectively hinders the proliferation and dissemination of GC cells by downregulating DNMT1 and promoting the expression of RB1.


Sujet(s)
Prolifération cellulaire , DNA (Cytosine-5-)-methyltransferase 1 , Régulation de l'expression des gènes tumoraux , Protéines de liaison à la protéine du rétinoblastome , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/génétique , Lignée cellulaire tumorale , Animaux , Prolifération cellulaire/génétique , Protéines de liaison à la protéine du rétinoblastome/génétique , Protéines de liaison à la protéine du rétinoblastome/métabolisme , Souris , Métastase tumorale , Mouvement cellulaire/génétique , Souris nude , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Protéine du rétinoblastome/métabolisme , Protéine du rétinoblastome/génétique , Souris de lignée BALB C , Protéines de répression
5.
Nat Commun ; 15(1): 4450, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38789411

RÉSUMÉ

Histone deacetylases (HDACs) play a crucial role in transcriptional regulation and are implicated in various diseases, including cancer. They are involved in histone tail deacetylation and canonically linked to transcriptional repression. Previous studies suggested that HDAC recruitment to cell-cycle gene promoters via the retinoblastoma (RB) protein or the DREAM complex through SIN3B is essential for G1/S and G2/M gene repression during cell-cycle arrest and exit. Here we investigate the interplay among DREAM, RB, SIN3 proteins, and HDACs in the context of cell-cycle gene repression. Knockout of SIN3B does not globally derepress cell-cycle genes in non-proliferating HCT116 and C2C12 cells. Loss of SIN3A/B moderately upregulates several cell-cycle genes in HCT116 cells but does so independently of DREAM/RB. HDAC inhibition does not induce general upregulation of RB/DREAM target genes in arrested transformed or non-transformed cells. Our findings suggest that E2F:RB and DREAM complexes can repress cell-cycle genes without relying on HDAC activity.


Sujet(s)
Facteurs de transcription E2F , Histone deacetylases , Protéines de répression , Protéine du rétinoblastome , Humains , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Cellules HCT116 , Protéines de répression/métabolisme , Protéines de répression/génétique , Facteurs de transcription E2F/métabolisme , Facteurs de transcription E2F/génétique , Protéine du rétinoblastome/métabolisme , Protéine du rétinoblastome/génétique , Souris , Animaux , Complexe Sin3-histone désacétylases-corépresseurs/métabolisme , Complexe Sin3-histone désacétylases-corépresseurs/génétique , Protéines KChIP/métabolisme , Protéines KChIP/génétique , Cycle cellulaire/génétique , Régions promotrices (génétique)/génétique , Régulation de l'expression des gènes , Gènes cdc
6.
Mol Biol Rep ; 51(1): 606, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38704498

RÉSUMÉ

BACKGROUND: Recent in vitro studies using RB1+/- fibroblasts and MSCs have shown molecular and functional disruptions without the need for biallelic loss of RB1. However, this was not reflected in the recent in vitro studies employing RB1+/- retinal organoids. To gain further insights into the molecular disruptions in the RB1+/- retinal organoids, we performed a high throughput RNA sequencing analysis. METHODS AND RESULTS: iPSCs were generated from RB1+/+ and RB1+/- OAMSCs derived from retinoblastoma patients. RB1+/+ and RB1+/- iPSCs were subjected to a step-wise retinal differentiation protocol. Retinal differentiation was evaluated by Real-time PCR and flow cytometry analysis of the retinal markers. To gain further insights into the molecular differences in RB1+/- retinal organoids, a high throughput RNA sequencing followed by differential gene expression analysis and gene set enrichment analysis (GSEA) was performed. The analysis revealed a shift from the regular metabolic process of glycolysis to oxidative phosphorylation in the RB1+/- retinal organoids. To investigate further, we performed assays to determine the levels of pyruvate, lactate and ATP in the retinal organoids. The results revealed significant increase in ATP and pyruvate levels in RB1+/- retinal organoids of day 120 compared to that of the RB1+/+. The results thus revealed enhanced ATP production in the RB1+/- retinal organoids. CONCLUSION: The study provides novel insights into the metabolic phenotype of heterozygous RB1 mutant suggesting dysregulation of energy metabolism and glycolytic pathways to be first step even before the changes in cellular proliferation or other phenotypic consequences ensue.


Sujet(s)
Adénosine triphosphate , Différenciation cellulaire , Cellules souches pluripotentes induites , Organoïdes , Rétine , Protéines de liaison à la protéine du rétinoblastome , Rétinoblastome , Ubiquitin-protein ligases , Humains , Adénosine triphosphate/métabolisme , Différenciation cellulaire/génétique , Glycolyse/génétique , Hétérozygote , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Mutation/génétique , Organoïdes/métabolisme , Rétine/métabolisme , Rétine/cytologie , Rétinoblastome/génétique , Rétinoblastome/métabolisme , Protéines de liaison à la protéine du rétinoblastome/génétique , Protéines de liaison à la protéine du rétinoblastome/métabolisme , Protéine du rétinoblastome/génétique , Protéine du rétinoblastome/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme
7.
FEBS J ; 291(10): 2091-2093, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38646863

RÉSUMÉ

Cellular immortalization is a complex process that requires multiple genetic alterations to overcome restricting barriers, including senescence. Not surprisingly, many of these alterations are associated with cancer; two tumor suppressor pathways, the cellular tumor antigen p53 and p16-Retinoblastoma (RB) pathways, are the best-characterized examples, but their mutations alone are known to be insufficient to drive full immortalization. En et al. identified a role for the lamin B receptor (LBR) in promoting cellular proliferation and immortalization in p53- and RB-deficient cells by maintaining their genome integrity and suppressing senescence. Thus, modulation of LBR could be exploited to treat cancer and potentially also to promote cell rejuvenation.


Sujet(s)
Vieillissement de la cellule , Instabilité du génome , , Protéine p53 suppresseur de tumeur , Vieillissement de la cellule/génétique , Humains , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Récepteurs cytoplasmiques et nucléaires/génétique , Récepteurs cytoplasmiques et nucléaires/métabolisme , Animaux , Protéine du rétinoblastome/génétique , Protéine du rétinoblastome/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Prolifération cellulaire/génétique , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/anatomopathologie
8.
Nat Commun ; 15(1): 3580, 2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38678032

RÉSUMÉ

The lethality, chemoresistance and metastatic characteristics of cancers are associated with phenotypically plastic cancer stem cells (CSCs). How the non-cell autonomous signalling pathways and cell-autonomous transcriptional machinery orchestrate the stem cell-like characteristics of CSCs is still poorly understood. Here we use a quantitative proteomic approach for identifying secreted proteins of CSCs in pancreatic cancer. We uncover that the cell-autonomous E2F1/4-pRb/RBL2 axis balances non-cell-autonomous signalling in healthy ductal cells but becomes deregulated upon KRAS mutation. E2F1 and E2F4 induce whereas pRb/RBL2 reduce WNT ligand expression (e.g. WNT7A, WNT7B, WNT10A, WNT4) thereby regulating self-renewal, chemoresistance and invasiveness of CSCs in both PDAC and breast cancer, and fibroblast proliferation. Screening for epigenetic enzymes identifies GCN5 as a regulator of CSCs that deposits H3K9ac onto WNT promoters and enhancers. Collectively, paracrine signalling pathways are controlled by the E2F-GCN5-RB axis in diverse cancers and this could be a therapeutic target for eliminating CSCs.


Sujet(s)
Facteur de transcription E2F1 , Facteur de transcription E2F4 , Cellules souches tumorales , Tumeurs du pancréas , Communication paracrine , Humains , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Facteur de transcription E2F1/métabolisme , Facteur de transcription E2F1/génétique , Lignée cellulaire tumorale , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Facteur de transcription E2F4/métabolisme , Facteur de transcription E2F4/génétique , Animaux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Protéines de type Wingless/métabolisme , Protéines de type Wingless/génétique , Protéine du rétinoblastome/métabolisme , Protéine du rétinoblastome/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Facteurs de transcription CBP-p300/métabolisme , Facteurs de transcription CBP-p300/génétique , Régulation de l'expression des gènes tumoraux , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Femelle , Prolifération cellulaire , Souris , Transduction du signal , Résistance aux médicaments antinéoplasiques/génétique
9.
Cancer Sci ; 115(5): 1576-1586, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38468443

RÉSUMÉ

While loss of function (LOF) of retinoblastoma 1 (RB1) tumor suppressor is known to drive initiation of small-cell lung cancer and retinoblastoma, RB1 mutation is rarely observed in breast cancers at their initiation. In this study, we investigated the impact on untransformed mammary epithelial cells given by RB1 LOF. Depletion of RB1 in anon-tumorigenic MCF10A cells induced reversible growth arrest (quiescence) featured by downregulation of multiple cyclins and MYC, upregulation of p27KIP1, and lack of expression of markers which indicate cellular senescence or epithelial-mesenchymal transition (EMT). We observed a similar phenomenon in human mammary epithelial cells (HMEC) as well. Additionally, we found that RB1 depletion attenuated the activity of RAS and the downstream MAPK pathway in an RBL2/p130-dependent manner. The expression of farnesyltransferase ß, which is essential for RAS maturation, was found to be downregulated following RB1 depletion also in an RBL2/p130-dependent manner. These findings unveiled an unexpected mechanism whereby normal mammary epithelial cells resist to tumor initiation upon RB1 LOF.


Sujet(s)
Régulation négative , Cellules épithéliales , Protéines de liaison à la protéine du rétinoblastome , Transduction du signal , Protéines G ras , Humains , Cellules épithéliales/métabolisme , Femelle , Protéines de liaison à la protéine du rétinoblastome/métabolisme , Protéines de liaison à la protéine du rétinoblastome/génétique , Protéines G ras/métabolisme , Protéines G ras/génétique , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Transition épithélio-mésenchymateuse/génétique , Glandes mammaires humaines/métabolisme , Glandes mammaires humaines/anatomopathologie , Glandes mammaires humaines/cytologie , Lignée cellulaire tumorale , Protéine du rétinoblastome/métabolisme , Protéine du rétinoblastome/génétique , Inhibiteur p27 de kinase cycline-dépendante/métabolisme , Inhibiteur p27 de kinase cycline-dépendante/génétique
10.
Cell Death Differ ; 31(5): 592-604, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38514847

RÉSUMÉ

RB transcriptional corepressor 1 (RB) deletion is the most important genomic factor associated with the prognosis of castration-resistant prostate cancer (CRPC) patients receiving androgen receptor (AR) signaling inhibitor therapy. Loss of RB could support prostate cancer cell growth in a hormone-independent manner, but the underlying mechanism by which RB regulates tumor progression extends far beyond the cell cycle pathway. A previous study indicated that RB inactivates AKT signaling but has no effect on mTOR signaling in cancer cells. Here, we found that the S249/T252 site in RB is key to regulating the transcriptional activity of the tumor-promoting factor TRIM24 in CRPC, as identified through FXXXV mapping. The RB/TRIM24 complex functions through DUSP2, which serves as an intermediate bridge, to activate the mTOR pathway and promote prostate cancer progression. Accordingly, we designed RB-linker-proteolysis-targeting chimera (PROTAC) molecules, which decreased TRIM24 protein levels and inactivated the mTOR signaling pathway, thereby inhibiting prostate cancer. Therefore, this study not only elucidates the novel function of RB but also provides a theoretical basis for the development of new drugs for treating prostate cancer.


Sujet(s)
Transduction du signal , Sérine-thréonine kinases TOR , Mâle , Humains , Sérine-thréonine kinases TOR/métabolisme , Lignée cellulaire tumorale , Animaux , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Protéine du rétinoblastome/métabolisme , Protéines de transport/métabolisme , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/génétique , Souris , Souris nude , Prolifération cellulaire
11.
Histopathology ; 84(7): 1178-1191, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38445509

RÉSUMÉ

AIMS: p16 is a sensitive surrogate marker for transcriptionally active high-risk human papillomavirus (HR-HPV) infection in endocervical adenocarcinoma (ECA); however, its specificity is not perfect. METHODS AND RESULTS: We examined p16 and Rb expressions by immunohistochemistry (IHC) and the transcriptionally active HR-HPV infection by mRNA in-situ hybridisation (ISH) with histological review in 108 ECA cases. Thirteen adenocarcinomas of endometrial or equivocal origin (six endometrioid and seven serous carcinomas) were compared as the control group. HR-HPV was detected in 83 of 108 ECA cases (77%), including five HPV-associated adenocarcinomas in situ and 78 invasive HPV-associated adenocarcinomas. All 83 HPV-positive cases showed consistent morphology, p16 positivity and partial loss pattern of Rb. Among the 25 cases of HPV-independent adenocarcinoma, four (16%) were positive for p16, and of these four cases, three of 14 (21%) were gastric type adenocarcinomas and one of 10 (10%) was a clear cell type adenocarcinoma. All 25 HPV-independent adenocarcinomas showed preserved expression of Rb irrespective of the p16 status. Similarly, all 13 cases of the control group were negative for HR-HPV with preserved expression of Rb, even though six of 13 (46%) cases were positive for p16. Compared with p16 alone, the combination of p16 overexpression and Rb partial loss pattern showed equally excellent sensitivity (each 100%) and improved specificity (100 versus 73.6%) and positive predictive values (100 versus 89.2%) in the ECA and control groups. Furthermore, HR-HPV infection correlated with better prognosis among invasive ECAs. CONCLUSIONS: The results suggest that the combined use of p16 and Rb IHC could be a reliable method to predict HR-HPV infection in primary ECAs and mimics. This finding may contribute to prognostic prediction and therapeutic strategy.


Sujet(s)
Adénocarcinome , Marqueurs biologiques tumoraux , Inhibiteur p16 de kinase cycline-dépendante , Immunohistochimie , Infections à papillomavirus , Tumeurs du col de l'utérus , Humains , Femelle , Infections à papillomavirus/complications , Tumeurs du col de l'utérus/virologie , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/diagnostic , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Adénocarcinome/virologie , Adénocarcinome/anatomopathologie , Adénocarcinome/métabolisme , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/métabolisme , Adulte d'âge moyen , Adulte , Sujet âgé , Protéine du rétinoblastome/métabolisme , Hybridation in situ , Papillomaviridae/génétique
12.
BMC Plant Biol ; 24(1): 157, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38424498

RÉSUMÉ

BACKGROUND: D-type cyclins (CYCD) regulate the cell cycle G1/S transition and are thus closely involved in cell cycle progression. However, little is known about their functions in rice. RESULTS: We identified 14 CYCD genes in the rice genome and confirmed the presence of characteristic cyclin domains in each. The expression of the OsCYCD genes in different tissues was investigated. Most OsCYCD genes were expressed at least in one of the analyzed tissues, with varying degrees of expression. Ten OsCYCD proteins could interact with both retinoblastoma-related protein (RBR) and A-type cyclin-dependent kinases (CDKA) forming holistic complexes, while OsCYCD3;1, OsCYCD6;1, and OsCYCD7;1 bound only one component, and OsCYCD4;2 bound to neither protein. Interestingly, all OsCYCD genes except OsCYCD7;1, were able to induce tobacco pavement cells to re-enter mitosis with different efficiencies. Transgenic rice plants overexpressing OsCYCD2;2, OsCYCD6;1, and OsCYCD7;1 (which induced cell division in tobacco with high-, low-, and zero-efficiency, respectively) were created. Higher levels of cell division were observed in both the stomatal lineage and epidermal cells of the OsCYCD2;2- and OsCYCD6;1-overexpressing plants, with lower levels seen in OsCYCD7;1-overexpressing plants. CONCLUSIONS: The distinct expression patterns and varying effects on the cell cycle suggest different functions for the various OsCYCD proteins. Our findings will enhance understanding of the CYCD family in rice and provide a preliminary foundation for the future functional verification of these genes.


Sujet(s)
Cyclines , Oryza , Cyclines/génétique , Cyclines/métabolisme , Oryza/génétique , Oryza/métabolisme , Phosphorylation , Kinases cyclines-dépendantes/génétique , Kinases cyclines-dépendantes/métabolisme , Cycle cellulaire/génétique , Protéine du rétinoblastome/génétique , Protéine du rétinoblastome/métabolisme , Mitose
13.
Int J Biol Macromol ; 260(Pt 1): 129559, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38242392

RÉSUMÉ

Cancer is a medical condition that is caused by the abnormal growth and division of cells, leading to the formation of tumors. The E2F1 and RB pathways are critical in regulating cell cycle, and their dysregulation can contribute to the development of cancer. In this study, we analyzed experimentally reported SNPs in E2F1 and assessed their effects on the binding affinity with RB. Out of 46, nine mutations were predicted as deleterious, and further analysis revealed four highly destabilizing mutations (L206W, R232C, I254T, A267T) that significantly altered the protein structure. Molecular docking of wild-type and mutant E2F1 with RB revealed a docking score of -242 kcal/mol for wild-type, while the mutant complexes had scores ranging from -217 to -220 kcal/mol. Molecular simulation analysis revealed variations in the dynamics features of both mutant and wild-type complexes due to the acquired mutations. Furthermore, the total binding free energy for the wild-type E2F1-RB complex was -64.89 kcal/mol, while those of the L206W, R232C, I254T, and A267T E2F1-RB mutants were -45.90 kcal/mol, -53.52 kcal/mol, -55.67 kcal/mol, and -61.22 kcal/mol, respectively. Our study is the first to extensively analyze E2F1 gene mutations and identifies candidate mutations for further validation and potential targeting for cancer therapeutics.


Sujet(s)
Tumeurs , Protéine du rétinoblastome , Humains , Protéine du rétinoblastome/génétique , Protéine du rétinoblastome/métabolisme , Polymorphisme de nucléotide simple/génétique , Simulation de docking moléculaire , Cycle cellulaire , Facteur de transcription E2F1/génétique , Facteur de transcription E2F1/métabolisme , Tumeurs/génétique
14.
Trends Cell Biol ; 34(4): 288-298, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-37648594

RÉSUMÉ

The retinoblastoma protein (RB)-mediated regulation of E2F is a component of a highly conserved cell cycle machine. However, RB's tumor suppressor activity, like RB's requirement in animal development, is tissue-specific, context-specific, and sometimes appears uncoupled from cell proliferation. Detailed new information about RB's genomic distribution provides a new perspective on the complexity of RB function, suggesting that some of its functional specificity results from context-specific RB association with chromatin. Here we summarize recent evidence showing that RB targets different types of chromatin regulatory elements at different cell cycle stages. RB controls traditional RB/E2F targets prior to S-phase, but, when cells proliferate, RB redistributes to cell type-specific chromatin loci. We discuss the broad implications of the new data for RB research.


Sujet(s)
Chromatine , Protéine du rétinoblastome , Animaux , Facteurs de transcription E2F/métabolisme , Cycle cellulaire/génétique , Protéine du rétinoblastome/génétique , Protéine du rétinoblastome/métabolisme , Division cellulaire
15.
J Pediatr Surg ; 59(3): 473-482, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37919169

RÉSUMÉ

BACKGROUND: Sarcomas are a heterogenous collection of bone and soft tissue tumors. The heterogeneity of these tumors makes it difficult to standardize treatment. CDK 4/6 inhibitors are a family of targeted agents which limit cell cycle progression and have been shown to be upregulated in sarcomas. In the current preclinical study, we evaluated the effects of lerociclib, a CDK4/6 inhibitor, on pediatric sarcomas in vitro and in 3D bioprinted tumors. METHODS: The effects of lerociclib on viability, proliferation, cell cycle, motility, and stemness were assessed in established sarcoma cell lines, U-2 OS and MG-63, as well as sarcoma patient-derived xenografts (PDXs). 3D printed biotumors of each of the U-2 OS, MG-63, and COA79 cells were utilized to study the effects of lerociclib on tumor growth ex vivo. RESULTS: CDK 4/6, as well as the intermediaries retinoblastoma protein (Rb) and phosphorylated Rb were identified as targets in the four sarcoma cell lines. Lerociclib treatment induced cell cycle arrest, decreased proliferation, motility, and stemness of sarcoma cells. Treatment with lerociclib decreased sarcoma cell viability in both traditional 2D culture as well as 3D bioprinted microtumors. CONCLUSIONS: Inhibition of CDK 4/6 activity with lerociclib was efficacious in traditional 2D sarcoma cell culture as well as in 3D bioprints. Lerociclib holds promise and warrants further investigation as a novel therapeutic strategy for management of these heterogenous groups of tumors.


Sujet(s)
Antinéoplasiques , Sarcomes , Enfant , Humains , Sarcomes/traitement médicamenteux , Sarcomes/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Protéine du rétinoblastome/métabolisme , Protéine du rétinoblastome/pharmacologie , Protéine du rétinoblastome/usage thérapeutique , Phosphorylation , Lignée cellulaire tumorale , Prolifération cellulaire , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/usage thérapeutique
16.
Cell Death Dis ; 14(12): 801, 2023 12 07.
Article de Anglais | MEDLINE | ID: mdl-38062013

RÉSUMÉ

Colorectal cancer (CRC), the third most common cancer worldwide, remains highly lethal as the disease only becomes symptomatic at an advanced stage. Growing evidence suggests that histone deacetylases (HDACs), a group of epigenetic enzymes overexpressed in precancerous lesions of CRC, may represent promising molecular targets for CRC treatment. Histone deacetylase inhibitors (HDACis) have gradually become powerful anti-cancer agents targeting epigenetic modulation and have been widely used in the clinical treatment of hematologic malignancies, while only few studies on the benefit of HDACis in the treatment of CRC. In the present study, we designed a series of small-molecule Thiazole-based HDACis, among which HR488B bound to HDAC1 with a high affinity and exerted effective anti-CRC activity both in vitro and in vivo. Moreover, we revealed that HR488B specifically suppressed the growth of CRC cells by inducing cell cycle G0/G1 arrest and apoptosis via causing mitochondrial dysfunction, reactive oxygen species (ROS) generation, and DNA damage accumulation. Importantly, we noticed that HR488B significantly decreased the expression of the E2F transcription factor 1 (E2F1), which was crucial for the inhibitory effect of HR488B on CRC. Mechanistically, HR488B obviously decreased the phosphorylation level of the retinoblastoma protein (Rb), and subsequently prevented the release of E2F1 from the E2F1/Rb/HDAC1 complex, which ultimately suppressed the growth of CRC cells. Overall, our study suggests that HR488B, a novel and efficient HDAC1 inhibitor, may be a potential candidate for CRC therapy in the future. Furthermore, targeting the E2F1/Rb/HDAC1 axis with HR488B provides a promising therapeutic avenue for CRC.


Sujet(s)
Antinéoplasiques , Tumeurs colorectales , Humains , Facteur de transcription E2F1/génétique , Facteur de transcription E2F1/métabolisme , Protéine du rétinoblastome/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Inhibiteurs de désacétylase d'histone/pharmacologie , Tumeurs colorectales/traitement médicamenteux , Protéines du cycle cellulaire/métabolisme , Histone Deacetylase 1/métabolisme
17.
Nat Commun ; 14(1): 7847, 2023 Nov 29.
Article de Anglais | MEDLINE | ID: mdl-38030655

RÉSUMÉ

Cyclin-dependent kinases 4 and 6 (CDK4/6) are critical for initiating cell proliferation by inactivating the retinoblastoma (Rb) protein. However, mammalian cells can bypass CDK4/6 for Rb inactivation. Here we show a non-canonical pathway for Rb inactivation and its interplay with external signals. We find that the non-phosphorylated Rb protein in quiescent cells is intrinsically unstable, offering an alternative mechanism for initiating E2F activity. Nevertheless, this pathway incompletely induces Rb-protein loss, resulting in minimal E2F activity. To trigger cell proliferation, upregulation of mitogenic signaling is required for stabilizing c-Myc, thereby augmenting E2F activity. Concurrently, stress signaling promotes Cip/Kip levels, competitively regulating cell proliferation with mitogenic signaling. In cancer, driver mutations elevate c-Myc levels, facilitating adaptation to CDK4/6 inhibitors. Differentiated cells, despite Rb-protein loss, maintain quiescence through the modulation of c-Myc and Cip/Kip levels. Our findings provide mechanistic insights into an alternative model of cell-cycle entry and the maintenance of quiescence.


Sujet(s)
Protéines du cycle cellulaire , Transduction du signal , Animaux , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Cycle cellulaire/génétique , Division cellulaire , Phosphorylation , Protéines du cycle cellulaire/métabolisme , Protéine du rétinoblastome/génétique , Protéine du rétinoblastome/métabolisme , Mitogènes , Mammifères/métabolisme
18.
Dev Cell ; 58(21): 2261-2274.e6, 2023 11 06.
Article de Anglais | MEDLINE | ID: mdl-37848027

RÉSUMÉ

The retinoblastoma (RB) and Hippo pathways interact to regulate cell proliferation and differentiation. However, the mechanism of interaction is not fully understood. Drosophila photoreceptors with inactivated RB and Hippo pathways specify normally but fail to maintain their neuronal identity and dedifferentiate. We performed single-cell RNA sequencing to elucidate the cause of dedifferentiation and to determine the fate of these cells. We find that dedifferentiated cells adopt a progenitor-like fate due to inappropriate activation of the retinal differentiation suppressor homothorax (hth) by Yki/Sd. This results in the activation of a distinct Yki/Hth transcriptional program, driving photoreceptor dedifferentiation. We show that Rbf physically interacts with Yki and, together with the GAGA factor, inhibits the hth expression. Thus, RB and Hippo pathways cooperate to maintain photoreceptor differentiation by preventing inappropriate expression of hth in differentiating photoreceptors. Our work highlights the importance of both RB and Hippo pathway activities for maintaining the state of terminal differentiation.


Sujet(s)
Protéines de Drosophila , Tumeurs de la rétine , Rétinoblastome , Animaux , Drosophila/métabolisme , Drosophila melanogaster/métabolisme , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Protéines nucléaires/métabolisme , Protéine du rétinoblastome/métabolisme , Transduction du signal/génétique , Cellules souches/métabolisme , Transactivateurs/métabolisme , Facteurs de transcription/métabolisme
19.
Cell Rep ; 42(11): 113198, 2023 11 28.
Article de Anglais | MEDLINE | ID: mdl-37865915

RÉSUMÉ

Cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) are key therapeutic agents in the management of metastatic hormone-receptor-positive breast cancer. However, the emergence of drug resistance limits their long-term efficacy. Here, we show that breast cancer cells develop CDK4/6i resistance via a sequential two-step process of E2F activation. This process entails retinoblastoma (Rb)-protein degradation, followed by c-Myc-mediated amplification of E2F transcriptional activity. CDK4/6i treatment halts cell proliferation in an Rb-dependent manner but dramatically reduces Rb-protein levels. However, this reduction in Rb levels insufficiently induces E2F activity. To develop CDK4/6i resistance, upregulation or activating mutations in mitogenic or hormone signaling are required to stabilize c-Myc levels, thereby augmenting E2F activity. Our analysis of pre-treatment tumor samples reveals a strong correlation between c-Myc levels, rather than Rb levels, and poor therapeutic outcomes after CDK4/6i treatment. Moreover, we propose that proteasome inhibitors can potentially reverse CDK4/6i resistance by restoring Rb levels.


Sujet(s)
Tumeurs du sein , Tumeurs de la rétine , Rétinoblastome , Humains , Femelle , Kinase-4 cycline-dépendante/métabolisme , Tumeurs du sein/anatomopathologie , Kinase-6 cycline-dépendante/métabolisme , Protéine du rétinoblastome/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique
20.
Cancer Res Commun ; 3(10): 1992-2002, 2023 10 03.
Article de Anglais | MEDLINE | ID: mdl-37728504

RÉSUMÉ

Inactivation of the retinoblastoma (RB) tumor suppressor in lung adenocarcinoma is associated with the rapid acquisition of metastatic ability and the loss of lung cell lineage commitment. We previously showed that restoration of RB in advanced lung adenocarcinomas in the mouse was correlated with a decreased frequency of lineage decommitted tumors and overt metastases. To identify a causal relationship for RB and its role in reprogramming lineage commitment and reducing metastatic competency in lung adenocarcinoma, we developed multiple tumor spheroid forming lines where RB restoration could be achieved after characterization of the degree of each spheroid's lineage commitment and metastatic ability. Surprisingly, we discovered that RB inactivation dramatically promoted tumor spheroid forming potential in tumors that arise in the KrasLSL-G12D/+; p53flox/flox lung adenocarcinoma model. However, RB reactivation had no effect on the maintenance of tumor spheroid lines once established. In addition, we show that RB-deficient tumor spheroid lines are not uniformly metastatically competent but are equally likely to be nonmetastatic. Interestingly, unlike tumor spheroid maintenance, RB restoration could functionally revert metastatic tumor spheroids to a nonmetastatic cell state. Thus, strategies to reinstate RB pathway activity in lung cancer may reverse metastatic ability and have therapeutic potential. Finally, the acquisition of tumor spheroid forming potential reflects underlying cell state plasticity, which is often predictive of, or even conflated with metastatic ability. Our data support that each is a discrete cell state restricted by RB and question the suitability of tumor spheroid models for their predictive potential of advanced metastatic tumor cell states. SIGNIFICANCE: Members of the RB pathway are frequently mutated in lung adenocarcinoma. We show that RB regulates cell state plasticity, tumor spheroid formation, and metastatic competency. Our data indicate that these are independent states where spheroid formation is distinct from metastatic competency. Thus, we caution against conflating spheroid formation and other signs of cell state plasticity with advanced metastatic cell states. Nevertheless, our work supports clinical strategies to reactivate RB pathways.


Sujet(s)
Adénocarcinome pulmonaire , Adénocarcinome , Tumeurs du poumon , Humains , Adénocarcinome/génétique , Protéine du rétinoblastome/métabolisme , Adénocarcinome pulmonaire/génétique , Tumeurs du poumon/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...