Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.714
Filtrer
1.
BMC Cancer ; 24(1): 928, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090568

RÉSUMÉ

BACKGROUND: Osteosarcoma (OS) is one of the most common primary malignant tumors of bone in children, which develops from osteoblasts and typically occurs during the rapid growth phase of the bone. Recently, Super-Enhancers(SEs)have been reported to play a crucial role in osteosarcoma growth and metastasis. Therefore, there is an urgent need to identify specific targeted inhibitors of SEs to assist clinical therapy. This study aimed to elucidate the role of BRD4 inhibitor GNE-987 targeting SEs in OS and preliminarily explore its mechanism. METHODS: We evaluated changes in osteosarcoma cells following treatment with a BRD4 inhibitor GNE-987. We assessed the anti-tumor effect of GNE-987 in vitro and in vivo by Western blot, CCK8, flow cytometry detection, clone formation, xenograft tumor size measurements, and Ki67 immunohistochemical staining, and combined ChIP-seq with RNA-seq techniques to find its anti-tumor mechanism. RESULTS: In this study, we found that extremely low concentrations of GNE-987(2-10 nM) significantly reduced the proliferation and survival of OS cells by degrading BRD4. In addition, we found that GNE-987 markedly induced cell cycle arrest and apoptosis in OS cells. Further study indicated that VHL was critical for GNE-987 to exert its antitumor effect in OS cells. Consistent with in vitro results, GNE-987 administration significantly reduced tumor size in xenograft models with minimal toxicity, and partially degraded the BRD4 protein. KRT80 was identified through analysis of the RNA-seq and ChIP-seq data. U2OS HiC analysis suggested a higher frequency of chromatin interactions near the KRT80 binding site. The enrichment of H3K27ac modification at KRT80 was significantly reduced after GNE-987 treatment. KRT80 was identified as playing an important role in OS occurrence and development. CONCLUSIONS: This research revealed that GNE-987 selectively degraded BRD4 and disrupted the transcriptional regulation of oncogenes in OS. GNE-987 has the potential to affect KRT80 against OS.


Sujet(s)
Apoptose , Tumeurs osseuses , Protéines du cycle cellulaire , Prolifération cellulaire , Ostéosarcome , Facteurs de transcription , Animaux , Humains , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Protéines contenant un bromodomaine , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Éléments activateurs (génétique) , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris nude , Ostéosarcome/traitement médicamenteux , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Biochem Pharmacol ; 226: 116412, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38971334

RÉSUMÉ

Increases in de novo lipogenesis that disturbed lipid homeostasis and caused lipid accumulation are a major cause of NAFLD and obesity. SREBP1 is a crucial regulatory factor controlling the expression of rate-limiting enzymes of lipid synthesis. A reduction in SREBP1expression can reduce lipid accumulation. Thus, we utilized an SREBP1-luciferase-KI HEK293 cell line constructed by our lab to screen 200 kinds of epigenetic drugs for their ability to downregulate SREBP1expression. BI-7273, an inhibitor of bromodomain-containing protein 9 (BRD9), was screened and found to decrease SREBP1 expression. What is more, BI-7273 has been confirmed that it could reduce lipid accumulation in HepG2 cells by BODIPY staining, and significantly decrease the protein expression of SREBP1 and FASN. To explore the potential mechanism BI-7273 reducing lipid accumulation, RNA sequencing (RNA-seq) was performed and demonstrated that BI-7273 reduced lipid accumulation by downregulating the AKT/mTOR/SREBP1 pathway in vitro. Finally, these results were verified in NAFLD and obesity mouse model induced by high fat diet (HFD). The results indicated that BI-7273 could decrease mouse body weight and improve insulin sensitivity, but also exhibited a strong negative correlation with serum lipid levels, and also demonstrated that BI-7273 reduced lipid accumulation via AKT/mTOR/SREBP1 pathway in vivo. In conclusion, our results revealed that BI-7273 decreases lipid accumulation by downregulating the AKT/mTOR/SREBP1 pathway in vivo and in vitro. This is the first report demonstrating the protective effect of this BRD9 inhibitor against NAFLD and obesity. BRD9 may be a novel target for the discovery of effective drugs to treat lipid metabolism disorders.


Sujet(s)
Régulation négative , Métabolisme lipidique , Souris de lignée C57BL , Protéines proto-oncogènes c-akt , Transduction du signal , Protéine-1 de liaison à l'élément de régulation des stérols , Sérine-thréonine kinases TOR , Facteurs de transcription , Humains , Animaux , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Souris , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Mâle , Cellules HepG2 , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Cellules HEK293 , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/traitement médicamenteux , Alimentation riche en graisse/effets indésirables , Relation dose-effet des médicaments , Protéines contenant un bromodomaine
3.
Bioorg Med Chem ; 110: 117827, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38964169

RÉSUMÉ

Histone deacetylase inhibitors (HDACis) show beneficial effects on different hematological malignancy subtypes. However, their impacts on treating solid tumors are still limited due to diverse resistance mechanisms. Recent studies have found that the feedback activation of BRD4-LIFR-JAK1-STAT3 pathway after HDACi incubation is a vital mechanism inducing resistance of specific solid tumor cells to HDACis. This review summarizes the recent development of multi-target HDACis that can concurrently block BRD4-LIFR-JAK1-STAT3 pathway. Moreover, our findings hope to shed novel lights on developing novel multi-target HDACis with reduced BRD4-LIFR-JAK1-STAT3-mediated drug resistance in some tumors.


Sujet(s)
Inhibiteurs de désacétylase d'histone , Janus kinase 1 , Tumeurs , Facteur de transcription STAT-3 , Transduction du signal , Facteurs de transcription , Humains , Facteur de transcription STAT-3/antagonistes et inhibiteurs , Facteur de transcription STAT-3/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/synthèse chimique , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Janus kinase 1/antagonistes et inhibiteurs , Janus kinase 1/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Protéines contenant un bromodomaine
5.
Bioorg Med Chem Lett ; 109: 129849, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38876177

RÉSUMÉ

Clinical studies have shown that inhibitors of bromodomain and extra-terminal domain (BET) proteins, particularly BRD4, have antitumor activity and efficacy. The BET protein has two domains, BD1 and BD2, and we previously focused on BD1 and reported orally bioavailable BD1-selective inhibitors. In this study, we obtained a BD1 inhibitor, a more potent and highly selective pyrazolopyridone derivative 13a, and confirmed its in vivo efficacy.


Sujet(s)
Pyridones , Humains , Administration par voie orale , Relation structure-activité , Animaux , Pyridones/composition chimique , Pyridones/pharmacologie , Pyridones/synthèse chimique , Pyridones/pharmacocinétique , Pyrazoles/composition chimique , Pyrazoles/pharmacologie , Pyrazoles/synthèse chimique , Découverte de médicament , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Structure moléculaire , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Souris , Domaines protéiques , Relation dose-effet des médicaments , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Rats , Protéines contenant un bromodomaine
6.
Anticancer Res ; 44(7): 2909-2919, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38925848

RÉSUMÉ

BACKGROUND/AIM: NUAK family kinase 2 (NUAK2) is a promising target for cancer therapeutics due to its reported role in protein phosphorylation, a critical process in cancer cell survival, proliferation, invasion, and senescence. This study aimed to identify novel inhibitors that disrupt NUAK2 activity. We have already identified two KRICT Hippo kinase inhibitor (KHKI) compounds, such as KHKI-01128 and KHKI-01215. Our aim was to evaluate the impact of KHKI-01128 and KHKI-01215 on NUAK2 activity and elucidate its mechanism in colorectal cancer cells. MATERIALS AND METHODS: To evaluate anticancer properties of these inhibitors, four in vitro assays in the SW480 cell line (time-resolved fluorescence resonance energy transfer assay, KINOMEscan kinase profiling, viability, and apoptosis assays) and two pharmacological mechanism analyses (Gene Set Enrichment Analysis and western blotting) were performed. RESULTS: KHKI-01128 and KHKI-01215 exhibited potent inhibitory activity against NUAK2 (half-maximal inhibitory concentration=0.024±0.015 µM and 0.052±0.011 µM, respectively). These inhibitors suppressed cell proliferation, with half-maximal inhibitory concentrations of 1.26±0.17 µM and 3.16±0.30 µM, respectively, and induced apoptosis of SW480 cells. Gene Set Enrichment Analysis revealed negative enrichment scores of -0.84 for KHKI-01128 (false-discovery rate=0.70) and 1.37 for KHKI-01215 (false-discovery rate=0.18), indicating that both effectively suppressed the expression of YES1-associated transcriptional regulator (YAP) target genes. CONCLUSION: These results suggest that KHKI-01128 and KHKI-01215 are potent NUAK2 inhibitors with promising potential for pharmaceutical applications.


Sujet(s)
Antinéoplasiques , Apoptose , Prolifération cellulaire , Tumeurs colorectales , Inhibiteurs de protéines kinases , Protein-Serine-Threonine Kinases , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/métabolisme , Antinéoplasiques/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Protein kinases/métabolisme
7.
J Med Chem ; 67(13): 11326-11353, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38913763

RÉSUMÉ

BRD9 is a pivotal epigenetic factor involved in cancers and inflammatory diseases. Still, the limited selectivity and poor phenotypic activity of targeted agents make it an atypically undruggable target. PROTAC offers an alternative strategy for overcoming the issue. In this study, we explored diverse E3 ligase ligands for the contribution of BRD9 PROTAC degradation. Through molecular docking, binding affinity analysis, and structure-activity relationship study, we identified a highly potent PROTAC E5, with excellent BRD9 degradation (DC50 = 16 pM) and antiproliferation in MV4-11 cells (IC50 = 0.27 nM) and OCI-LY10 cells (IC50 = 1.04 nM). E5 can selectively degrade BRD9 and induce cell cycle arrest and apoptosis. Moreover, the therapeutic efficacy of E5 was confirmed in xenograft tumor models, accompanied by further RNA-seq analysis. Therefore, these results may pave the way and provide the reference for the discovery and investigation of highly effective PROTAC degraders.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Simulation de docking moléculaire , Ubiquitin-protein ligases , Humains , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Relation structure-activité , Prolifération cellulaire/effets des médicaments et des substances chimiques , Ubiquitin-protein ligases/métabolisme , Lignée cellulaire tumorale , Souris , Découverte de médicament , Tumeurs hématologiques/traitement médicamenteux , Tumeurs hématologiques/anatomopathologie , Tumeurs hématologiques/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Apoptose/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Souris nude , Souris de lignée BALB C , Tests d'activité antitumorale sur modèle de xénogreffe , Tests de criblage d'agents antitumoraux , Protéines contenant un bromodomaine
8.
Eur J Pharmacol ; 977: 176714, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38849043

RÉSUMÉ

OBJECTIVE: This study aimed to elucidate the molecular mechanisms by which BRD4 play a role in atrial fibrillation (AF). METHODS AND RESULTS: We used a discovery-driven approach to detect BRD4 expression in the atria of patients with AF and in various murine models of atrial fibrosis. We used a BRD4 inhibitor (JQ1) and atrial fibroblast (aFB)-specific BRD4-knockout mice to elucidate the role of BRD4 in AF. We further examined the underlying mechanisms using RNA-seq and ChIP-seq analyses in vitro, to identify key downstream targets of BRD4. We found that BRD4 expression is significantly increased in patients with AF, with accompanying atrial fibrosis and aFB differentiation. We showed that JQ1 treatment and shRNA-based molecular silencing of BRD4 blocked ANG-II-induced extracellular matrix production and cell-cycle progression in aFBs. BRD4-related RNA-seq and ChIP-seq analyses in aFBs demonstrated enrichment of a subset of promoters related to the expression of profibrotic and proliferation-related genes. The pharmacological inhibition of BRD4 in vivo or in aFB-specific BRD4-knockout in mice limited ANG-II-induced atrial fibrosis, atrial enlargement, and AF susceptibility. CONCLUSION: Our findings suggest that BRD4 plays a key role in pathological AF, at least partially by activating aFB proliferation and ECM synthesis. This study provides mechanistic insights into the development of BRD4 inhibitors as targeted antiarrhythmic therapies.


Sujet(s)
Fibrillation auriculaire , Azépines , Protéines du cycle cellulaire , Fibrose , Atrium du coeur , Souris knockout , Facteurs de transcription , Triazoles , Fibrillation auriculaire/génétique , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/anatomopathologie , Fibrillation auriculaire/traitement médicamenteux , Animaux , Facteurs de transcription/génétique , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Atrium du coeur/anatomopathologie , Atrium du coeur/effets des médicaments et des substances chimiques , Atrium du coeur/métabolisme , Humains , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Souris , Azépines/pharmacologie , Azépines/usage thérapeutique , Mâle , Triazoles/pharmacologie , Triazoles/usage thérapeutique , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/antagonistes et inhibiteurs , Modèles animaux de maladie humaine , Souris de lignée C57BL , Angiotensine-II/pharmacologie , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Thérapie moléculaire ciblée , Protéines contenant un bromodomaine
9.
J Med Chem ; 67(12): 10464-10489, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38866424

RÉSUMÉ

The bromodomain and extra terminal (BET) family of bromodomain-containing proteins are important epigenetic regulators that elicit their effect through binding histone tail N-acetyl lysine (KAc) post-translational modifications. Recognition of such markers has been implicated in a range of oncology and immune diseases and, as such, small-molecule inhibition of the BET family bromodomain-KAc protein-protein interaction has received significant interest as a therapeutic strategy, with several potential medicines under clinical evaluation. This work describes the structure- and property-based optimization of a ligand and lipophilic efficient pan-BET bromodomain inhibitor series to deliver candidate I-BET787 (70) that demonstrates efficacy in a mouse model of inflammation and suitable properties for both oral and intravenous (IV) administration. This focused two-phase explore-exploit medicinal chemistry effort delivered the candidate molecule in 3 months with less than 100 final compounds synthesized.


Sujet(s)
Administration par voie intraveineuse , Animaux , Administration par voie orale , Souris , Relation structure-activité , Humains , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Structure moléculaire
10.
Biomater Sci ; 12(14): 3686-3699, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38873991

RÉSUMÉ

PROteolysis TArgeting Chimeras have received increasing attention due to their capability to induce potent degradation of various disease-related proteins. However, the effective and controlled cytosolic delivery of current small-molecule PROTACs remains a challenge, primarily due to their intrinsic shortcomings, including unfavorable solubility, poor cell permeability, and limited spatiotemporal precision. Here, we develop a near-infrared light-controlled PROTAC delivery device (abbreviated as USDPR) that allows the efficient photoactivation of PROTAC function to achieve enhanced protein degradation. The nanodevice is constructed by encapsulating the commercial BRD4-targeting PROTACs (dBET6) in the hollow cavity of mesoporous silica-coated upconversion nanoparticles, followed by coating a Rose Bengal (RB) photosensitizer conjugated poly-L-lysine (PLL-RB). This composition enables NIR light-activatable generation of cytotoxic reactive oxygen species due to the energy transfer from the UCNPs to PLL-RB, which boosts the endo/lysosomal escape and subsequent cytosolic release of dBET6. We demonstrate that USDPR is capable of effectively degrading BRD4 in a NIR light-controlled manner. This in combination with NIR light-triggered photodynamic therapy enables an enhanced antitumor effect both in vitro and in vivo. This work thus presents a versatile strategy for controlled release of PROTACs and codelivery with photosensitizers using an NIR-responsive nanodevice, providing important insight into the design of effective PROTAC-based combination therapy.


Sujet(s)
Lysosomes , Nanoparticules , Photothérapie dynamique , Photosensibilisants , Protéolyse , Humains , Lysosomes/métabolisme , Nanoparticules/composition chimique , Nanoparticules/administration et posologie , Photosensibilisants/composition chimique , Photosensibilisants/pharmacologie , Photosensibilisants/administration et posologie , Animaux , Protéolyse/effets des médicaments et des substances chimiques , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/composition chimique , Souris , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Rayons infrarouges , Rose de Bengale/composition chimique , Rose de Bengale/pharmacologie , Rose de Bengale/administration et posologie , Silice/composition chimique , Polylysine/composition chimique , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/administration et posologie , Protéines contenant un bromodomaine
11.
Cancer Res ; 84(15): 2501-2517, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38833522

RÉSUMÉ

Recurrent somatic mutations in the BRG1/BRM-associated factor (BAF) chromatin remodeling complex subunit ARID1A occur frequently in advanced urothelial, endometrial, and ovarian clear cell carcinomas, creating an alternative chromatin state that may be exploited therapeutically. The histone methyltransferase EZH2 has been previously identified as targetable vulnerability in the context of ARID1A mutations. In this study, we describe the discovery of tulmimetostat, an orally available, clinical stage EZH2 inhibitor, and it elucidates the aspects of its application potential in ARID1A mutant tumors. Tulmimetostat administration achieved efficacy in multiple ARID1A mutant bladder, ovarian, and endometrial tumor models and improved cisplatin response in chemotherapy-resistant models. Consistent with its comprehensive and durable level of target coverage, tulmimetostat demonstrated greater efficacy than other PRC2-targeted inhibitors at comparable or lower exposures in a bladder cancer xenograft mouse model. Tulmimetostat mediated extensive changes in gene expression, in addition to a profound reduction in global H3K27me3 levels in tumors. Phase I clinical pharmacokinetic and pharmacodynamic data indicated that tulmimetostat exhibits durable exposure and profound target engagement. Importantly, a tulmimetostat controlled gene expression signature identified in whole blood from a cohort of 32 patients with cancer correlated with tulmimetostat exposure, representing a pharmacodynamic marker for the assessment of target coverage for PRC2-targeted agents in the clinic. Collectively, these data suggest that tulmimetostat has the potential to achieve clinical benefit in solid tumors as a monotherapy but also in combination with chemotherapeutic agents, and may be beneficial in various indications with recurrent ARID1A mutations. Significance: The EZH2 inhibitor tulmimetostat achieves comprehensive target inhibition in ARID1A mutant solid tumor models and cancer patients that can be assessed with a pharmacodynamic gene signature in peripheral blood.


Sujet(s)
Protéines de liaison à l'ADN , Protéine-2 homologue de l'activateur de Zeste , Mutation , Facteurs de transcription , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Protéine-2 homologue de l'activateur de Zeste/génétique , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Souris , Facteurs de transcription/génétique , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Femelle , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/métabolisme , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/antagonistes et inhibiteurs , Protéines nucléaires/métabolisme , Lignée cellulaire tumorale , Souris nude , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme
12.
Int J Mol Sci ; 25(10)2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38791396

RÉSUMÉ

The Hippo pathway controls organ size and homeostasis and is linked to numerous diseases, including cancer. The transcriptional enhanced associate domain (TEAD) family of transcription factors acts as a receptor for downstream effectors, namely yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), which binds to various transcription factors and is essential for stimulated gene transcription. YAP/TAZ-TEAD facilitates the upregulation of multiple genes involved in evolutionary cell proliferation and survival. TEAD1-4 overexpression has been observed in different cancers in various tissues, making TEAD an attractive target for drug development. The central drug-accessible pocket of TEAD is crucial because it undergoes a post-translational modification called auto-palmitoylation. Crystal structures of the C-terminal TEAD complex with small molecules are available in the Protein Data Bank, aiding structure-based drug design. In this study, we utilized the fragment molecular orbital (FMO) method, molecular dynamics (MD) simulations, shape-based screening, and molecular mechanics-generalized Born surface area (MM-GBSA) calculations for virtual screening, and we identified a novel non-covalent inhibitor-BC-001-with IC50 = 3.7 µM in a reporter assay. Subsequently, we optimized several analogs of BC-001 and found that the optimized compound BC-011 exhibited an IC50 of 72.43 nM. These findings can be used to design effective TEAD modulators with anticancer therapeutic implications.


Sujet(s)
Simulation de dynamique moléculaire , Facteurs de transcription à domaine TEA , Facteurs de transcription , Humains , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/composition chimique , Sites de fixation , Découverte de médicament/méthodes , Liaison aux protéines , Simulation de docking moléculaire , Conception de médicament
13.
Eur J Med Chem ; 272: 116468, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38718626

RÉSUMÉ

High expression of ubiquitin-specific protease 10 (USP10) promote the proliferation of hepatocellular carcinoma (HCC), thus the development of USP10 inhibitors holds promise as a novel therapeutic approach for HCC treatment. However, the development of selective USP10 inhibitor is still limited. In this study, we developed a novel USP10 inhibitor for investigating the feasibility of targeting USP10 for the treatment of HCC. Due to high USP10 inhibition potency and prominent selectivity, compound D1 bearing quinolin-4(1H)-one scaffold was identified as a lead compound. Subsequent research revealed that D1 significantly inhibits cell proliferation and clone formation in HCC cells. Mechanistic insights indicated that D1 targets the ubiquitin pathway, facilitating the degradation of YAP (Yes-associated protein), thereby triggering the downregulation of p53 and its downstream protein p21. Ultimately, this cascade leads to S-phase arrest in HCC cells, followed by cell apoptosis. Collectively, our findings highlight D1 as a promising starting point for USP10-positive HCC treatment, underscoring its potential as a vital tool for unraveling the functional intricacies of USP10.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Antinéoplasiques , Carcinome hépatocellulaire , Prolifération cellulaire , Découverte de médicament , Tumeurs du foie , Facteurs de transcription , Ubiquitin thiolesterase , Protéines de signalisation YAP , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Ubiquitin thiolesterase/antagonistes et inhibiteurs , Ubiquitin thiolesterase/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Relation structure-activité , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines de signalisation YAP/métabolisme , Structure moléculaire , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Apoptose/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/synthèse chimique , Lignée cellulaire tumorale
14.
Cancer Lett ; 592: 216919, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38704133

RÉSUMÉ

Efforts to develop targetable molecular bases for drug resistance for pancreatic ductal adenocarcinoma (PDAC) have been equivocally successful. Using RNA-seq and ingenuity pathway analysis we identified that the superpathway of cholesterol biosynthesis is upregulated in gemcitabine resistant (gemR) tumors using a unique PDAC PDX model with resistance to gemcitabine acquired in vivo. Analysis of additional in vitro and in vivo gemR PDAC models showed that HMG-CoA synthase 2 (HMGCS2), an enzyme involved in cholesterol biosynthesis and rate limiting in ketogenesis, is overexpressed in these models. Mechanistic data demonstrate the novel findings that HMGCS2 contributes to gemR and confers metastatic properties in PDAC models, and that HMGCS2 is BRD4 dependent. Further, BET inhibitor JQ1 decreases levels of HMGCS2, sensitizes PDAC cells to gemcitabine, and a combination of gemcitabine and JQ1 induced regressions of gemR tumors in vivo. Our data suggest that decreasing HMGCS2 may reverse gemR, and that HMGCS2 represents a useful therapeutic target for treating gemcitabine resistant PDAC.


Sujet(s)
Azépines , Carcinome du canal pancréatique , Désoxycytidine , Résistance aux médicaments antinéoplasiques , , Hydroxymethylglutaryl-coA synthase , Tumeurs du pancréas , Triazoles , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Humains , Souris , Antimétabolites antinéoplasiques/pharmacologie , Azépines/pharmacologie , Protéines contenant un bromodomaine , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Hydroxymethylglutaryl-coA synthase/métabolisme , Hydroxymethylglutaryl-coA synthase/génétique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/antagonistes et inhibiteurs , Triazoles/pharmacologie , Femelle , Souris SCID
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167249, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38768929

RÉSUMÉ

RET fusion is an oncogenic driver in 1-2 % of patients with non-small cell lung cancer (NSCLC). Although RET-positive tumors have been treated with multikinase inhibitors such as vandetanib or RET-selective inhibitors, ultimately resistance to them develops. Here we established vandetanib resistance (VR) clones from LC-2/ad cells harboring CCDC6-RET fusion and explored the molecular mechanism of the resistance. Each VR clone had a distinct phenotype, implying they had acquired resistance via different mechanisms. Consistently, whole exome-seq and RNA-seq revealed that the VR clones had unique mutational signatures and expression profiles, and shared only a few common remarkable events. AXL and IGF-1R were activated as bypass pathway in different VR clones, and sensitive to a combination of RET and AXL inhibitors or IGF-1R inhibitors, respectively. SMARCA4 loss was also found in a particular VR clone and 55 % of post-TKI lung tumor tissues, being correlated with higher sensitivity to SMARCA4/SMARCA2 dual inhibition and shorter PFS after subsequent treatments. Finally, we detected an increased number of damaged mitochondria in one VR clone, which conferred sensitivity to mitochondrial electron transfer chain inhibitors. Increased mitochondria were also observed in post-TKI biopsy specimens in 13/20 cases of NSCLC, suggesting a potential strategy targeting mitochondria to treat resistant tumors. Our data propose new promising therapeutic options to combat resistance to RET inhibitors in NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Tumeurs du poumon , Mitochondries , Pipéridines , Inhibiteurs de protéines kinases , Protéines proto-oncogènes c-ret , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Protéines proto-oncogènes c-ret/antagonistes et inhibiteurs , Protéines proto-oncogènes c-ret/génétique , Protéines proto-oncogènes c-ret/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Lignée cellulaire tumorale , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Protéines de fusion oncogènes/antagonistes et inhibiteurs , Helicase/génétique , Helicase/métabolisme , Helicase/antagonistes et inhibiteurs , Protéines du cytosquelette
16.
J Clin Invest ; 134(13)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38771655

RÉSUMÉ

Diffuse midline glioma (DMG) H3K27-altered is one of the most malignant childhood cancers. Radiation therapy remains the only effective treatment yet provides a 5-year survival rate of only 1%. Several clinical trials have attempted to enhance radiation antitumor activity using radiosensitizing agents, although none have been successful. Given this, there is a critical need for identifying effective therapeutics to enhance radiation sensitivity for the treatment of DMG. Using high-throughput radiosensitivity screening, we identified bromo- and extraterminal domain (BET) protein inhibitors as potent radiosensitizers in DMG cells. Genetic and pharmacologic inhibition of BET bromodomain activity reduced DMG cell proliferation and enhanced radiation-induced DNA damage by inhibiting DNA repair pathways. RNA-Seq and the CUT&RUN (cleavage under targets and release using nuclease) analysis showed that BET bromodomain inhibitors regulated the expression of DNA repair genes mediated by H3K27 acetylation at enhancers. BET bromodomain inhibitors enhanced DMG radiation response in patient-derived xenografts as well as genetically engineered mouse models. Together, our results highlight BET bromodomain inhibitors as potential radiosensitizer and provide a rationale for developing combination therapy with radiation for the treatment of DMG.


Sujet(s)
Histone , Radiotolérance , Humains , Animaux , Souris , Radiotolérance/effets des médicaments et des substances chimiques , Radiotolérance/génétique , Histone/métabolisme , Histone/génétique , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Réparation de l'ADN/effets des médicaments et des substances chimiques , Gliome/radiothérapie , Gliome/anatomopathologie , Gliome/génétique , Gliome/métabolisme , Gliome/traitement médicamenteux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/radiothérapie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Radiosensibilisants/pharmacologie , Facteurs de transcription/génétique , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Altération de l'ADN , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Protéines contenant un bromodomaine , Protéines
17.
Bioorg Med Chem ; 107: 117762, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38759254

RÉSUMÉ

Honokiol, derived from Magnolia officinalis (a traditional Chinese medicine), has been reported to have anticancer activity. Here, a series of novel honokiol thioethers bearing a 1,3,4-oxadiazole moiety were prepared and evaluated for their anticancer activities against three types of digestive system tumor cells. Biological evaluation showed that honokiol derivative 3k exhibited the best antiproliferative activity against HCT116 cells with an IC50 value of 6.1 µmol/L, superior to the reference drug 5-fluorouracil (IC50: 9.63 ± 0.27 µmol/L). The structure-activity relationships (SARs) indicated that the introduction of -(4-NO2)Ph, 3-pyridyl, -(2-F)Ph, -(4-F)Ph, -(3-F)Ph, -(4-Cl)Ph, and -(3-Cl)Ph groups was favorable for enhancing the anticancer activity of the title honokiol thioethers. Further study revealed that honokiol thioether 3k can well inhibit the proliferation of colon cancer cells HCT116, arresting the cells in G1 phase and inducing cell death. Moreover, a preliminary mechanism study indicated that 3k directly inhibits the transcription and expression of YAP protein without activating the Hippo signaling pathway. Thus, honokiol thioether 3k could be deeply developed for the development of honokiol-based anticancer candidates.


Sujet(s)
Dérivés du biphényle , Prolifération cellulaire , Tests de criblage d'agents antitumoraux , Lignanes , Protéines de signalisation YAP , Humains , Lignanes/pharmacologie , Lignanes/composition chimique , Lignanes/synthèse chimique , Dérivés du biphényle/pharmacologie , Dérivés du biphényle/antagonistes et inhibiteurs , Dérivés du biphényle/composition chimique , Relation structure-activité , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules HCT116 , Protéines de signalisation YAP/métabolisme , Structure moléculaire , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/métabolisme , Sulfures/composition chimique , Sulfures/pharmacologie , Sulfures/synthèse chimique , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques d'origine végétale/composition chimique , Antinéoplasiques d'origine végétale/synthèse chimique , Relation dose-effet des médicaments , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Composés allyliques , Phénols
18.
Bioorg Med Chem ; 106: 117752, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38749341

RÉSUMÉ

Bromodomain protein 4 (BRD4) is a member of the BET family, and its overexpression is closely associated with the development of many tumors. Inhibition of BRD4 shows great therapeutic potential in anti-tumor, and pan-BRD4 inhibitors show adverse effects of dose limiting toxicity and thrombocytopenia in clinical trials. To improve clinical effects and reduce side effects, more efforts have focused on seeking selective inhibitors of BD1 or BD2. Herein, a series of indole-2-one derivatives were designed and synthesized through docking-guided optimization to find BRD4-BD1 selective inhibitors, and their BRD4 inhibitory and antiproliferation activities were evaluated. Among them, compound 21r had potent BRD4 inhibitory activity (the IC50 values of 41 nM and 313 nM in BD1 and BD2 domain), excellent anti-proliferation (the IC50 values of 4.64 ± 0.30 µM, 0.78 ± 0.03 µM, 5.57 ± 1.03 µM against HL-60, MV-4-11 and HT-29 cells), and displayed low toxicity against normal cell GES-1 cells. Further studies revealed that 21r inhibited proliferation by decreasing the expression of proto-oncogene c-Myc, blocking cell cycle in G0/G1 phase, and inducing apoptosis in MV-4-11 cells in a dose-dependent manner. All the results showed that compound 21r was a potent BRD4 inhibitor with BD1 selectivity, which had potential in treatment of leukemia.


Sujet(s)
Antinéoplasiques , Protéines du cycle cellulaire , Prolifération cellulaire , Tests de criblage d'agents antitumoraux , Indoles , Facteurs de transcription , Humains , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Indoles/composition chimique , Indoles/pharmacologie , Indoles/synthèse chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation structure-activité , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Structure moléculaire , Découverte de médicament , Relation dose-effet des médicaments , Proto-oncogène Mas , Apoptose/effets des médicaments et des substances chimiques , Simulation de docking moléculaire , Lignée cellulaire tumorale , Protéines contenant un bromodomaine
19.
Eur J Med Chem ; 271: 116444, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38691889

RÉSUMÉ

The NAPRT-induced increase in NAD+ levels was proposed as a mechanism contributing to hepatocellular carcinoma (HCC) resistance to NAMPT inhibitors. Thus, concurrently targeting NAMPT and NAPRT could be considered to overcome drug resistance. A BRD4 inhibitor downregulates the expression of NAPRT in HCC, and the combination of NAMPT inhibitors with BRD4 inhibitors simultaneously blocks NAD+ generation via salvage and the PH synthesis pathway. Moreover, the combination of the two agents significantly downregulated the expression of tumor-promoting genes and strongly promoted apoptosis. The present work identified various NAMPT/BRD4 dual inhibitors based on the multitargeted drug rationale. Among them, compound A2, which demonstrated the strongest effect, exhibited potent inhibition of NAMPT and BRD4 (IC50 = 35 and 58 nM, respectively). It significantly suppressed the growth and migration of HCC cells and facilitated their apoptosis. Furthermore, compound A2 also manifested a robust anticancer effect in HCCLM3 xenograft mouse models, with no apparent toxic effects. Our findings in this study provide an effective approach to target NAD+ metabolism for HCC treatment.


Sujet(s)
Antinéoplasiques , Apoptose , Carcinome hépatocellulaire , Protéines du cycle cellulaire , Prolifération cellulaire , Cytokines , Tumeurs du foie , Nicotinamide phosphoribosyltransferase , Facteurs de transcription , Nicotinamide phosphoribosyltransferase/antagonistes et inhibiteurs , Nicotinamide phosphoribosyltransferase/métabolisme , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris , Apoptose/effets des médicaments et des substances chimiques , Relation structure-activité , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Cytokines/métabolisme , Cytokines/antagonistes et inhibiteurs , Découverte de médicament , Tests de criblage d'agents antitumoraux , Structure moléculaire , Relation dose-effet des médicaments , Souris nude , Lignée cellulaire tumorale , Souris de lignée BALB C , Protéines contenant un bromodomaine
20.
Mol Cancer Ther ; 23(8): 1095-1108, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38691847

RÉSUMÉ

Many tumor types harbor alterations in the Hippo pathway, including mesothelioma, where a high percentage of cases are considered YAP1/TEAD dependent. Identification of autopalmitoylation sites in the hydrophobic palmitate pocket of TEADs, which may be necessary for YAP1 protein interactions, has enabled modern drug discovery platforms to generate compounds that allosterically inhibit YAP1/TEAD complex formation and transcriptional activity. We report the discovery and characterization of a novel YAP1/TEAD inhibitor MRK-A from an aryl ether chemical series demonstrating potent and specific inhibition of YAP1/TEAD activity. In vivo, MRK-A showed a favorable tolerability profile in mice and demonstrated pharmacokinetics suitable for twice daily oral dosing in preclinical efficacy studies. Importantly, monotherapeutic targeting of YAP1/TEAD in preclinical models generated regressions in a mesothelioma CDX model; however, rapid resistance to therapy was observed. RNA-sequencing of resistant tumors revealed mRNA expression changes correlated with the resistance state and a marked increase of hepatocyte growth factor (HGF) expression. In vitro, exogenous HGF was able to fully rescue cytostasis induced by MRK-A in mesothelioma cell lines. In addition, co-administration of small molecule inhibitors of the MET receptor tyrosine kinase suppressed the resistance generating effect of HGF on MRK-A induced growth inhibition. In this work, we report the structure and characterization of MRK-A, demonstrating potent and specific inhibition of YAP1/TAZ-TEAD-mediated transcriptional responses, with potential implications for treating malignancies driven by altered Hippo signaling, including factors resulting in acquired drug resistance.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Résistance aux médicaments antinéoplasiques , Facteur de croissance des hépatocytes , Protéines proto-oncogènes c-met , Transduction du signal , Facteurs de transcription , Protéines de signalisation YAP , Humains , Facteur de croissance des hépatocytes/métabolisme , Animaux , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/métabolisme , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Bibliothèques de petites molécules/pharmacologie , Facteurs de transcription à domaine TEA , Prolifération cellulaire/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE