Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 896
Filtrer
2.
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
3.
Eur J Med Chem ; 271: 116414, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38677061

RÉSUMÉ

Sclerostin is a secreted glycoprotein that expresses predominantly in osteocytes and inhibits bone formation by antagonizing the Wnt/ß-catenin signaling pathway, and the loop3 region of sclerostin has recently discovered as a novel therapeutic target for bone anabolic treatment without increasing cardiovascular risk. Herein, we used a structural based virtual screening to search for small molecular inhibitors selectively targeting sclerostin loop3. A novel natural product hit ZINC4228235 (THFA) was identified as the sclerostin loop3-selective inhibitor with a Kd value of 42.43 nM against sclerostin loop3. The simplification and derivation of THFA using molecular modeling-guided modification allowed the discovery of an effective and loop3-selective small molecular inhibitor, compound (4-(3-acetamidoprop-1-yn-1-yl)benzoyl)glycine (AACA), with improved binding affinity (Kd = 15.4 nM) compared to the hit THFA. Further in-vitro experiment revealed that compound AACA could attenuate the suppressive effect of transfected sclerostin on Wnt signaling and bone formation. These results make AACA as a potential candidate for development of anti-osteoporosis agents without increasing cardiovascular risk.


Sujet(s)
Conception de médicament , Ostéoporose , Ostéoporose/traitement médicamenteux , Humains , Relation structure-activité , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/synthèse chimique , Structure moléculaire , Animaux , Souris , Découverte de médicament , Évaluation préclinique de médicament , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/métabolisme , Relation dose-effet des médicaments , Modèles moléculaires , Ostéogenèse/effets des médicaments et des substances chimiques
4.
J Phys Chem B ; 128(16): 3795-3806, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38606592

RÉSUMÉ

The Hippo signaling pathway is a highly conserved signaling network that plays a central role in regulating cellular growth, proliferation, and organ size. This pathway consists of a kinase cascade that integrates various upstream signals to control the activation or inactivation of YAP/TAZ proteins. Phosphorylated YAP/TAZ is sequestered in the cytoplasm; however, when the Hippo pathway is deactivated, it translocates into the nucleus, where it associates with TEAD transcription factors. This partnership is instrumental in regulating the transcription of progrowth and antiapoptotic genes. Thus, in many cancers, aberrantly hyperactivated YAP/TAZ promotes oncogenesis by contributing to cancer cell proliferation, metastasis, and therapy resistance. Because YAP and TAZ exert their oncogenic effects by binding with TEAD, it is critical to understand this key interaction to develop cancer therapeutics. Previous research has indicated that TEAD undergoes autopalmitoylation at a conserved cysteine, and small molecules that inhibit TEAD palmitoylation disrupt effective YAP/TAZ binding. However, how exactly palmitoylation contributes to YAP/TAZ-TEAD interactions and how the TEAD palmitoylation inhibitors disrupt this interaction remains unknown. Utilizing molecular dynamics simulations, our investigation not only provides detailed atomistic insight into the YAP/TAZ-TEAD dynamics but also unveils that the inhibitor studied influences the binding of YAP and TAZ to TEAD in distinct manners. This discovery has significant implications for the design and deployment of future molecular interventions targeting this interaction.


Sujet(s)
Lipoylation , Simulation de dynamique moléculaire , Facteurs de transcription à domaine TEA , Facteurs de transcription , Transcriptional coactivator with PDZ-binding motif proteins , Protéines de signalisation YAP , Humains , Acyltransferases/métabolisme , Acyltransferases/antagonistes et inhibiteurs , Acyltransferases/composition chimique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/composition chimique , Régulation allostérique/effets des médicaments et des substances chimiques , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/composition chimique , Liaison aux protéines , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Facteurs de transcription à domaine TEA/composition chimique , Facteurs de transcription à domaine TEA/métabolisme , Transactivateurs/métabolisme , Transactivateurs/composition chimique , Transactivateurs/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/composition chimique , Transcriptional coactivator with PDZ-binding motif proteins/composition chimique , Transcriptional coactivator with PDZ-binding motif proteins/métabolisme , Protéines de signalisation YAP/composition chimique , Protéines de signalisation YAP/métabolisme
5.
J Ocul Pharmacol Ther ; 40(4): 246-252, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38517736

RÉSUMÉ

Purpose: To investigate the effect of yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) on connective tissue growth factor (CTGF) expression in adult retinal pigment epithelial (ARPE)-19 cells. We also studied the inhibitory effect of K-975, a new pan-transcriptional enhanced associate domain (TEAD) inhibitor, and luteolin, a plant-derived flavonoid on CTGF expression. Methods: ARPE-19 cells were transfected with either YAP or TAZ overexpression plasmid or treated with transforming growth factor (TGF)-ß2. The cells were cultured either with or without K-975 or luteolin. The expression of YAP, TAZ, and CTGF was examined using real-time PCR. Results: ARPE-19 cells overexpressing YAP or TAZ exhibited significantly increased CTGF expression. This increase was attenuated by K-975 or luteolin alone. TGF-ß2 treatment significantly raised the expression of not just YAP and TAZ, but also CTGF in ARPE-19 cells. TGF-ß2 treatment-enhanced CTGF expression was considerably lowered by the addition of K-975 or luteolin. Conclusions: Overexpression of YAP or TAZ and treatment with TGF-ß2 led to an increase in the expression of CTGF in ARPE-19 cells. These increases were attenuated by treatment with K-975 and luteolin. These findings suggest that YAP and TAZ may be related to the expression of CTGF in ARPE-19 cells and that K-975 and luteolin can be explored as potential therapeutic agents for preventing CTGF production in vitreoretinal fibrosis.


Sujet(s)
Facteur de croissance du tissu conjonctif , Lutéoline , Épithélium pigmentaire de la rétine , Facteurs de transcription , Facteur de croissance du tissu conjonctif/métabolisme , Humains , Épithélium pigmentaire de la rétine/effets des médicaments et des substances chimiques , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/cytologie , Lutéoline/pharmacologie , Facteurs de transcription/métabolisme , Transcriptional coactivator with PDZ-binding motif proteins , Lignée cellulaire , Transactivateurs/métabolisme , Facteur de croissance transformant bêta-2/métabolisme , Facteur de croissance transformant bêta-2/pharmacologie , Facteur de croissance transformant bêta-2/antagonistes et inhibiteurs , Protéines de signalisation YAP/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique
6.
Gastroenterology ; 166(6): 1130-1144.e8, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38262581

RÉSUMÉ

BACKGROUND & AIMS: Despite the increasing number of treatment options available for liver cancer, only a small proportion of patients achieve long-term clinical benefits. Here, we aim to develop new therapeutic approaches for liver cancer. METHODS: A compound screen was conducted to identify inhibitors that could synergistically induce senescence when combined with cyclin-dependent kinase (CDK) 4/6 inhibitor. The combination effects of CDK4/6 inhibitor and exportin 1 (XPO1) inhibitor on cellular senescence were investigated in a panel of human liver cancer cell lines and multiple liver cancer models. A senolytic drug screen was performed to identify drugs that selectively killed senescent liver cancer cells. RESULTS: The combination of CDK4/6 inhibitor and XPO1 inhibitor synergistically induces senescence of liver cancer cells in vitro and in vivo. The XPO1 inhibitor acts by causing accumulation of RB1 in the nucleus, leading to decreased E2F signaling and promoting senescence induction by the CDK4/6 inhibitor. Through a senolytic drug screen, cereblon (CRBN)-based proteolysis targeting chimera (PROTAC) ARV-825 was identified as an agent that can selectively kill senescent liver cancer cells. Up-regulation of CRBN was a vulnerability of senescent liver cancer cells, making them sensitive to CRBN-based PROTAC drugs. Mechanistically, we find that ubiquitin specific peptidase 2 (USP2) directly interacts with CRBN, leading to the deubiquitination and stabilization of CRBN in senescent liver cancer cells. CONCLUSIONS: Our study demonstrates a striking synergy in senescence induction of liver cancer cells through the combination of CDK4/6 inhibitor and XPO1 inhibitor. These findings also shed light on the molecular processes underlying the vulnerability of senescent liver cancer cells to CRBN-based PROTAC therapy.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Vieillissement de la cellule , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , , Caryophérines , Tumeurs du foie , Inhibiteurs de protéines kinases , Récepteurs cytoplasmiques et nucléaires , Ubiquitin-protein ligases , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/métabolisme , Caryophérines/antagonistes et inhibiteurs , Caryophérines/métabolisme , Récepteurs cytoplasmiques et nucléaires/antagonistes et inhibiteurs , Récepteurs cytoplasmiques et nucléaires/métabolisme , Ubiquitin-protein ligases/métabolisme , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Lignée cellulaire tumorale , Inhibiteurs de protéines kinases/pharmacologie , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Animaux , Protéines de liaison à la protéine du rétinoblastome/métabolisme , Protéines de liaison à la protéine du rétinoblastome/génétique , Synergie des médicaments , Sénothérapie/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Transduction du signal/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Hydrazines/pharmacologie , Hydrazines/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Cellules HepG2 , Souris , Pipérazines , Pyridines , Triazoles
7.
Bone ; 179: 116985, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38052372

RÉSUMÉ

Sclerostin is an extracellular inhibitor of canonical Wnt signaling that inhibits bone formation and stimulates bone resorption. Anti-sclerostin antibodies (Scl-Ab) have been developed as bone-building agents. DKK1, another extracellular inhibitor of the pathway, is upregulated in osteocytes in response to sclerostin inhibition. To further enhance bone-forming effects, a bispecific antibody inhibiting both sclerostin and DKK1 was created (AMG 147). In nonclinical safety studies, AMG 147 resulted in novel skull findings. In the rat, there was increased thickness of skull bones of neural crest origin due to increased subperiosteal compact lamellar and intramembranous woven bone. Externally, subperiosteal fibroblastic/osteoblastic stromal cell proliferation with woven bone and hemorrhage was also observed. Scl-Ab alone resulted in increased skull thickness in the rat, like AMG 147, but without the stromal cell proliferation/woven bone formation. In contrast to embryonic flat bone development, intramembranous bone formed similar to plexiform bone. In the monkey, AMG 147 resulted in macroscopic skull thickening due to a diffuse increase in appositional lamellar bone and increased intramembranous bone on both periosteal surfaces of all skull bones. These data demonstrate that dual inhibition of sclerostin and DDK1 results in unique effects on the skull not observed with sclerostin inhibition alone.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Anticorps , Os et tissu osseux , Protéines et peptides de signalisation intercellulaire , Animaux , Rats , Anticorps/pharmacologie , Ostéogenèse , Primates , Crâne , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Os et tissu osseux/effets des médicaments et des substances chimiques , Os et tissu osseux/physiologie
8.
Eur J Med Chem ; 236: 114311, 2022 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-35385803

RÉSUMÉ

TRIM24 (tripartite motif-containing protein 24) and BRPF1 (bromodomain and PHD finger containing protein 1) are epigenetics "readers" and potential therapeutic targets for cancer and other diseases. Here we describe the structure-guided design of 1-(indolin-1-yl)ethan-1-ones as novel TRIM24/BRPF1 bromodomain inhibitors. The representative compound 20l (Y08624) is a new TRIM24/BRPF1 dual inhibitor, with IC50 values of 0.98 and 1.16 µM, respectively. Cellular activity of 20l was validated by viability assay in prostate cancer (PC) cell lines. In PC xenograft models, 20l suppressed tumor growth (50 mg/kg/day, TGI = 53%) without exhibiting noticeable toxicity. Compound 20l represents a versatile starting point for the development of more potent TRIM24/BRPF1 inhibitors.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéines de transport , Protéines de liaison à l'ADN , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Animaux , Protéines de transport/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Humains , Mâle , Tumeurs de la prostate/traitement médicamenteux , Domaines protéiques
9.
Molecules ; 27(3)2022 Jan 20.
Article de Anglais | MEDLINE | ID: mdl-35163936

RÉSUMÉ

The multidomain BAG3 protein is a member of the BAG (Bcl-2-associated athanogene) family of co-chaperones, involved in a wide range of protein-protein interactions crucial for many key cellular pathways, including autophagy, cytoskeletal dynamics, and apoptosis. Basal expression of BAG3 is elevated in several tumor cell lines, where it promotes cell survival signaling and apoptosis resistance through the interaction with many protein partners. In addition, its role as a key player of several hallmarks of cancer, such as metastasis, angiogenesis, autophagy activation, and apoptosis inhibition, has been established. Due to its involvement in malignant transformation, BAG3 has emerged as a potential and effective biological target to control multiple cancer-related signaling pathways. Recently, by using a multidisciplinary approach we reported the first synthetic BAG3 modulator interfering with its BAG domain (BD), based on a 2,4-thiazolidinedione scaffold and endowed with significant anti-proliferative activity. Here, a further in silico-driven selection of a 2,4-thiazolidinedione-based compound was performed. Thanks to a straightforward synthesis, relevant binding affinity for the BAG3BD domain, and attractive biological activities, this novel generation of compounds is of great interest for the development of further BAG3 binders, as well as for the elucidation of the biological roles of this protein in tumors. Specifically, we found compound 6 as a new BAG3 modulator with a relevant antiproliferative effect on two different cancer cell lines (IC50: A375 = 19.36 µM; HeLa = 18.67 µM).


Sujet(s)
Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Protéines régulatrices de l'apoptose/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Thiazolidinediones/pharmacologie , Antinéoplasiques/composition chimique , Apoptose , Autophagie , Prolifération cellulaire , Humains , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Thiazolidinediones/composition chimique , Cellules cancéreuses en culture
10.
Comput Math Methods Med ; 2022: 6881932, 2022.
Article de Anglais | MEDLINE | ID: mdl-35116073

RÉSUMÉ

OBJECTIVE: GIT1 is identified as a novel tumor oncogene in breast cancer. In this article, we aimed to explore the role of GIT1 in the progression of head and neck squamous cell carcinoma (HNSCC). METHODS: GIT1 expression in HNSCC was detected by RT-qPCR, immunohistochemistry assay, and Western blot. HNSCC cell proliferation, migration, and invasion were examined by CCK-8 assay, Wound healing assay, and Transwell assay, respectively. Cell apoptosis was detected by flow cytometric analysis. RESULTS: In our study, GIT1 was notably upregulated in HNSCC tissues and cells. Moreover, GIT1 expression level had positive corelation with pathological grade and nodal status of HNSCC. Functional experiments showed that knockdown of GIT1 restrained HNSCC proliferation, invasion, migration, and EMT and facilitated cell apoptosis. Furthermore, GIT1 knockdown was found to restrain HNSCC tumor growth and lung metastasis. Additionally, PI3K/AKT/mTOR signal pathway inhibitors suppressed the effect of GIT1 on HNSCC cell progression. CONCLUSION: GIT1 was upregulated in HNSCC and facilitated HNSCC cell progression by inducing PI3K/AKT/mTOR signal pathway. Therefore, we suggested that GIT1 might be a potential target for HNSCC treatment.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Protéines du cycle cellulaire/métabolisme , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/secondaire , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/génétique , Sujet âgé , Animaux , Apoptose , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Biologie informatique , Femelle , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Tumeurs de la tête et du cou/génétique , Hétérogreffes , Humains , Mâle , Souris , Souris nude , Adulte d'âge moyen , Invasion tumorale , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Carcinome épidermoïde de la tête et du cou/génétique , Sérine-thréonine kinases TOR/métabolisme , Régulation positive
11.
Bioorg Chem ; 119: 105516, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34856444

RÉSUMÉ

Both ruthenium (Ru) and isoquinoline (IQ) compounds are regarded as potential anticancer drug candidates. Here, we report the synthesis and characterization of three novel cyclometalated Ru(II)-isoquinoline complexes: RuIQ-3, RuIQ-4, and RuIQ-5, and evaluation of their in vitro cytotoxicities against a panel of cell lines including A549/DDP, a cisplatin-resistant human lung cancer cell line. A549/DDP 3D multicellular tumor spheroids (MCTSs) were also used to detect the drug resistance reversal effect of Ru(II)-IQ complexes. Our results indicated that the cytotoxic activities against cancer cells of Ru(II)-IQ complexes, especially RuIQ-5, were superior compared with cisplatin. In addition, RuIQ-5 exhibited low toxicity towards both normal HBE cells in vitro and zebrafish embryos in vivo. Further investigation on cellular mechanism of action indicated that after absorption by A549/DDP cells, RuIQ-5 was mainly distributed in the nucleus, which is different from cisplatin. Besides, RuIQ-5 could induce apoptosis through mitochondrial dysfunction, reactive oxygen species (ROS) accumulation, ROS-mediated DNA damage, and cycle arrest at both S and G2/M phases. Moreover, RuIQ-5 could inhibit the overexpression of Nrf2 through regulation of Akt/GSK-3ß/Fyn signaling pathway and hindering the nuclear translocation of Nrf2. Based on these findings, we firmly believe that the studied Ru(II)-IQ complexes hold great promise as anticancer therapeutics with high effectiveness and low toxicity.


Sujet(s)
Antinéoplasiques/pharmacologie , Cisplatine/pharmacologie , Complexes de coordination/pharmacologie , Isoquinoléines/pharmacologie , Ruthénium/pharmacologie , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cisplatine/composition chimique , Complexes de coordination/synthèse chimique , Complexes de coordination/composition chimique , Relation dose-effet des médicaments , Régulation négative/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux , Glycogen synthase kinase 3 beta/antagonistes et inhibiteurs , Glycogen synthase kinase 3 beta/métabolisme , Humains , Isoquinoléines/composition chimique , Structure moléculaire , Facteur-2 apparenté à NF-E2/antagonistes et inhibiteurs , Facteur-2 apparenté à NF-E2/métabolisme , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Protéines proto-oncogènes c-akt/métabolisme , Ruthénium/composition chimique , Relation structure-activité , Cellules cancéreuses en culture , Danio zébré
12.
Oxid Med Cell Longev ; 2021: 2896674, 2021.
Article de Anglais | MEDLINE | ID: mdl-34820053

RÉSUMÉ

Bone resorption diseases, including osteoporosis, are usually caused by excessive osteoclastogenesis. Unc-51-like autophagy activating kinase 1 (ULK1), a mammalian serine/threonine kinase, may participate in the regulation of bone homeostasis and osteolytic metastasis. In this study, ULK1 expression during osteoclastogenesis was detected with RT-PCR. We knocked down or overexpressed ULK1 through siRNA or lentiviral transduction in bone marrow macrophage (BMM). TRAP and phalloidin staining were performed to detect the osteoclastogenesis activity. Ovariectomized (OVX) mouse model of osteoporosis and a mouse of model osteoclast-induced bone resorption were applied to explore the role of ULK1 in bone resorption in vivo. The results showed that ULK1 expression was downregulated during osteoclast differentiation and was clinically associated with osteoporosis. ULK1 inhibited osteoclast differentiation in vitro. Knockdown of ULK1 expression activated phosphorylation of c-Jun N-terminal kinase (JNK) and spleen tyrosine kinase (Syk). Docking protein 3 (DOK3) was coexpressed with ULK1 during osteoclastogenesis. Downregulation of DOK3 offsets the effect of ULK1 on osteoclastogenesis and induced phosphorylation of JNK and Syk. Activation of ULK1 impeded bone loss in OVX mice with osteoporosis. Additionally, upregulation of ULK1 inhibited osteoclast-induced bone resorption in vivo. Therefore, our study reveals a novel ULK1/DOK3/Syk axis that regulates osteoclast differentiation and bone resorption, and targeting ULK1 is a potential therapeutic strategy for osteoporosis.


Sujet(s)
Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Homologue de la protéine-1 associée à l'autophagie/métabolisme , Résorption osseuse/prévention et contrôle , Différenciation cellulaire , Ostéoclastes/métabolisme , Ostéoporose/prévention et contrôle , Syk kinase/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Homologue de la protéine-1 associée à l'autophagie/génétique , Résorption osseuse/étiologie , Résorption osseuse/métabolisme , Résorption osseuse/anatomopathologie , Femelle , Mâle , Souris , Souris de lignée BALB C , Ostéoclastes/anatomopathologie , Ostéogenèse , Ostéoporose/étiologie , Ostéoporose/métabolisme , Ostéoporose/anatomopathologie , Syk kinase/génétique , Syk kinase/métabolisme
13.
Molecules ; 26(20)2021 Oct 14.
Article de Anglais | MEDLINE | ID: mdl-34684786

RÉSUMÉ

Two targeted sets of novel 1,5-diaryl-1H-imidazole-4-carboxylic acids 10 and carbohydrazides 11 were designed and synthesized from their corresponding ester intermediates 17, which were prepared via cycloaddition of ethyl isocyanoacetate 16 and diarylimidoyl chlorides 15. Evaluation of these new target scaffolds in the AlphaScreenTM HIV-1 IN-LEDGF/p75 inhibition assay identified seventeen compounds exceeding the pre-defined 50% inhibitory threshold at 100 µM concentration. Further evaluation of these compounds in the HIV-1 IN strand transfer assay at 100 µM showed that none of the compounds (with the exception of 10a, 10l, and 11k, with marginal inhibitory percentages) were actively bound to the active site, indicating that they are selectively binding to the LEDGF/p75-binding pocket. In a cell-based HIV-1 antiviral assay, compounds 11a, 11b, 11g, and 11h exhibited moderate antiviral percentage inhibition of 33-45% with cytotoxicity (CC50) values of >200 µM, 158.4 µM, >200 µM, and 50.4 µM, respectively. The antiviral inhibitory activity displayed by 11h was attributed to its toxicity. Upon further validation of their ability to induce multimerization in a Western blot gel assay, compounds 11a, 11b, and 11h appeared to increase higher-order forms of IN.


Sujet(s)
Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Inhibiteurs de l'intégrase du VIH/composition chimique , Inhibiteurs de l'intégrase du VIH/synthèse chimique , Intégrase du VIH/effets des médicaments et des substances chimiques , Facteurs de transcription/antagonistes et inhibiteurs , Domaine catalytique , Lignée cellulaire , Simulation numérique , Conception de médicament , Évaluation préclinique de médicament , Intégrase du VIH/composition chimique , Intégrase du VIH/métabolisme , Inhibiteurs de l'intégrase du VIH/pharmacologie , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Humains , Imidazoles/synthèse chimique , Imidazoles/composition chimique , Imidazoles/pharmacologie , Simulation de docking moléculaire , Structure moléculaire , Multimérisation de protéines/effets des médicaments et des substances chimiques
14.
BMC Cancer ; 21(1): 1079, 2021 Oct 06.
Article de Anglais | MEDLINE | ID: mdl-34615513

RÉSUMÉ

BACKGROUND: Activation of the oncogene yes-associated protein (YAP) is frequently detected in intrahepatic cholangiocarcinoma (iCCA); however, the expression pattern and the functional impact of its paralogue WW domain-containing transcription regulator 1 (WWTR1; synonym: TAZ) are not well described in different CCA subtypes. METHODS: Immunohistochemical analysis of YAP and TAZ in iCCA and extrahepatic CCA (eCCA) cohorts was performed. YAP/TAZ shuttling and their functional impact on CCA cell lines were investigated. Target genes expression after combined YAP/TAZ inhibition was analyzed. RESULTS: Immunohistochemical analysis of iCCA and eCCA revealed YAP or TAZ positivity in up to 49.2%; however, oncogene co-expression was less frequent (up to 23%). In contrast, both proteins were jointly detectable in most CCA cell lines and showed nuclear/cytoplasmic shuttling in a cell density-dependent manner. Next to the pro-proliferative function of YAP/TAZ, both transcriptional co-activators cooperated in the regulation of a gene signature that indicated the presence of chromosomal instability (CIN). A correlation between YAP and the CIN marker phospho-H2A histone family member X (pH2AX) was particularly observed in tissues from iCCA and distal CCA (dCCA). The presence of the CIN genes in about 25% of iCCA was statistically associated with worse prognosis. CONCLUSIONS: YAP and TAZ activation is not uncoupled from cell density in CCA cells and both factors cooperatively contribute to proliferation and expression of CIN-associated genes. The corresponding group of CCA patients is characterized by CIN and may benefit from YAP/TAZ-directed therapies.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Tumeurs des canaux biliaires/génétique , Cholangiocarcinome/génétique , Instabilité des chromosomes/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Facteurs de transcription/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Conduits biliaires extrahépatiques , Conduits biliaires intrahépatiques , Numération cellulaire , Lignée cellulaire tumorale , Cholangiocarcinome/métabolisme , Cholangiocarcinome/anatomopathologie , Histone/métabolisme , Humains , Immunohistochimie , Protéines et peptides de signalisation intracellulaire/antagonistes et inhibiteurs , Pronostic , Analyse sur puce à tissus , Facteurs de transcription/antagonistes et inhibiteurs , Transcriptional coactivator with PDZ-binding motif proteins , Protéines de signalisation YAP
15.
Commun Biol ; 4(1): 1165, 2021 10 07.
Article de Anglais | MEDLINE | ID: mdl-34621018

RÉSUMÉ

Hypoxia-inducible factor-1 (HIF-1) plays essential roles in human diseases, though its central role in oxygen homoeostasis hinders the development of direct HIF-1-targeted pharmacological approaches. Here, we surveyed small-molecule compounds that efficiently inhibit the transcriptional activity of HIF-1 without affecting body homoeostasis. We focused on Mint3, which activates HIF-1 transcriptional activity in limited types of cells, such as cancer cells and macrophages, by suppressing the factor inhibiting HIF-1 (FIH-1). We identified naphthofluorescein, which inhibited the Mint3-FIH-1 interaction in vitro and suppressed Mint3-dependent HIF-1 activity and glycolysis in cancer cells and macrophages without evidence of cytotoxicity in vitro. In vivo naphthofluorescein administration suppressed tumour growth and metastasis without adverse effects, similar to the genetic depletion of Mint3. Naphthofluorescein attenuated inflammatory cytokine production and endotoxic shock in mice. Thus, Mint3 inhibitors may present a new targeted therapeutic option for cancer and inflammatory diseases by avoiding severe adverse effects.


Sujet(s)
Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Carcinogenèse/effets des médicaments et des substances chimiques , Métastase tumorale/traitement médicamenteux , Tumeurs/traitement médicamenteux , Choc septique/traitement médicamenteux , Lignée cellulaire tumorale , Fluorescéines/pharmacologie , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Mixed function oxygenases/génétique , Mixed function oxygenases/métabolisme , Métastase tumorale/génétique , Tumeurs/génétique , Protéines de répression/génétique , Protéines de répression/métabolisme
16.
Cell Rep ; 36(11): 109687, 2021 09 14.
Article de Anglais | MEDLINE | ID: mdl-34525355

RÉSUMÉ

Mammalian cell cytoskeletal reorganization for efficient directional movement requires tight coordination of actomyosin and microtubule networks. In this study, we show that LRAP35a potentiates microtubule stabilization by promoting CLASP2/EB1 interaction besides its complex formation with MRCK/MYO18A for retrograde actin flow. The alternate regulation of these two networks by LRAP35a is tightly regulated by a series of phosphorylation events that dictated its specificity. Sequential phosphorylation of LRAP35a by Protein Kinase A (PKA) and Glycogen Synthase Kinase-3ß (GSK3ß) initiates the association of LRAP35a with CLASP2, while subsequent binding and further phosphorylation by Casein Kinase 1δ (CK1δ) induce their dissociation, which facilitates LRAP35a/MRCK association in driving lamellar actomyosin flow. Importantly, microtubule dynamics is directly moderated by CK1δ activity on CLASP2 to regulate GSK3ß phosphorylation of the SxIP motifs that blocks EB1 binding, an event countered by LRAP35a interaction and its competition for CK1δ activity. Overall this study reveals an essential role for LRAP35a in coordinating lamellar contractility and microtubule polarization in cell migration.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Casein Kinase Idelta/métabolisme , Glycogen synthase kinase 3 beta/métabolisme , Protéines associées aux microtubules/métabolisme , Microtubules/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Actines/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/génétique , Casein Kinase Idelta/antagonistes et inhibiteurs , Casein Kinase Idelta/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Cyclic AMP-Dependent Protein Kinases/antagonistes et inhibiteurs , Cyclic AMP-Dependent Protein Kinases/métabolisme , Humains , Protéines associées aux microtubules/composition chimique , Mutagenèse dirigée , Phosphorylation , Liaison aux protéines , Interférence par ARN , Petit ARN interférent/métabolisme , Protéines suppresseurs de tumeurs/antagonistes et inhibiteurs , Protéines suppresseurs de tumeurs/génétique
17.
Nat Commun ; 12(1): 5655, 2021 09 27.
Article de Anglais | MEDLINE | ID: mdl-34580292

RÉSUMÉ

High-throughput sequencing describes multiple alterations in individual tumors, but their functional relevance is often unclear. Clinic-close, individualized molecular model systems are required for functional validation and to identify therapeutic targets of high significance for each patient. Here, we establish a Cre-ERT2-loxP (causes recombination, estrogen receptor mutant T2, locus of X-over P1) based inducible RNAi- (ribonucleic acid interference) mediated gene silencing system in patient-derived xenograft (PDX) models of acute leukemias in vivo. Mimicking anti-cancer therapy in patients, gene inhibition is initiated in mice harboring orthotopic tumors. In fluorochrome guided, competitive in vivo trials, silencing of the apoptosis regulator MCL1 (myeloid cell leukemia sequence 1) correlates to pharmacological MCL1 inhibition in patients´ tumors, demonstrating the ability of the method to detect therapeutic vulnerabilities. The technique identifies a major tumor-maintaining potency of the MLL-AF4 (mixed lineage leukemia, ALL1-fused gene from chromosome 4) fusion, restricted to samples carrying the translocation. DUX4 (double homeobox 4) plays an essential role in patients' leukemias carrying the recently described DUX4-IGH (immunoglobulin heavy chain) translocation, while the downstream mediator DDIT4L (DNA-damage-inducible transcript 4 like) is identified as therapeutic vulnerability. By individualizing functional genomics in established tumors in vivo, our technique decisively complements the value chain of precision oncology. Being broadly applicable to tumors of all kinds, it will considerably reinforce personalizing anti-cancer treatment in the future.


Sujet(s)
Antinéoplasiques/pharmacologie , Marqueurs biologiques tumoraux/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Génétique inverse/méthodes , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/génétique , Adulte , Animaux , Antinéoplasiques/usage thérapeutique , Marqueurs biologiques tumoraux/antagonistes et inhibiteurs , Enfant , Femelle , Extinction de l'expression des gènes , Protéines à homéodomaine/antagonistes et inhibiteurs , Protéines à homéodomaine/génétique , Humains , Leucémie aigüe myéloïde/génétique , Mâle , Souris , Protéine Mcl-1/antagonistes et inhibiteurs , Protéine Mcl-1/génétique , Protéine de la leucémie myéloïde-lymphoïde/antagonistes et inhibiteurs , Protéine de la leucémie myéloïde-lymphoïde/génétique , Protéines de fusion oncogènes/antagonistes et inhibiteurs , Protéines de fusion oncogènes/génétique , Médecine de précision/méthodes , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Biomed Res Int ; 2021: 9957161, 2021.
Article de Anglais | MEDLINE | ID: mdl-34485531

RÉSUMÉ

OBJECTIVE: To investigate the biological role and potential mechanism of miR-25 in nasopharyngeal carcinoma. METHODS: The expression of miR-25 in nasopharyngeal carcinoma cell lines was detected by qRT-PCR. The effect of inhibition of miR-25 expression on the proliferative activity of nasopharyngeal carcinoma cell line HONE-1 was examined by CCK-8 method. Flow cytometry was used to detect the effect of miR-25 expression inhibition on the apoptosis rate of nasopharyngeal carcinoma cell line HONE-1. The miRNA target gene prediction site TargetScan predicts the target protein action site of miR-124 and verifies whether miR-25 interacts with the target by luciferase activity assay, qPCR, and Western experiments. The miR-25 inhibitor and target egg gene expression plasmids were cotransfected into HONE-1 cells for rescue experiments to investigate whether miR-25 inhibits proliferation of nasopharyngeal carcinoma cells by target genes. At the same time, qRT-PCR was used to detect the mRNA expression levels of Wnt/ß-catenin pathway key proteins TCF4, c-Myc, and Cyclin D1 in different transfected cells. RESULTS: miR-25 expression was upregulated in nasopharyngeal carcinoma cell lines. Functional studies showed that inhibition of miR-25 expression significantly inhibited the proliferation of nasopharyngeal carcinoma cell line HONE-1 (p < 0.05). Inhibition of miR-25 expression by flow cytometry significantly promoted apoptosis (p < 0.05). Detection of dual luciferase activity indicated that DKK3 is a direct target site for miR-25. Western blots showed that inhibition of miR-25 significantly upregulated DKK3 mRNA and protein levels. Supplementation with DKK3 significantly attenuated the inhibitory effect of miR-25 on the proliferation of nasopharyngeal carcinoma cell line HONE-1 (p < 0.05). qRT-PCR found that mRNA levels of TCF4, c-Myc, and Cyclin D1 were significantly upregulated in miR-25-transfected cells compared to control transfection. QRT PCR showed that the mRNA and protein levels of Tcf4, c-myc, and Cyclin D1 were significantly upregulated in miR-25 overexpression-transfected cells. CONCLUSION: Inhibition of miR-25 expression promotes DKK3 gene expression, and inactivation of Wnt/ß-catenin signaling pathway inhibits proliferation and promotes apoptosis of nasopharyngeal carcinoma cells.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , microARN/génétique , Cancer du nasopharynx/anatomopathologie , Tumeurs du rhinopharynx/anatomopathologie , Voie de signalisation Wnt , bêta-Caténine/métabolisme , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Apoptose , Lignée cellulaire tumorale , Prolifération cellulaire , Humains , Cancer du nasopharynx/génétique , Cancer du nasopharynx/métabolisme , Tumeurs du rhinopharynx/génétique , Tumeurs du rhinopharynx/métabolisme , Régulation positive
19.
Int J Biol Macromol ; 190: 636-648, 2021 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-34517025

RÉSUMÉ

SARS-CoV-2 nucleocapsid (N) protein undergoes RNA-induced phase separation (LLPS) and sequesters the host key stress granule (SG) proteins, Ras-GTPase-activating protein SH3-domain-binding protein 1 and 2 (G3BP1 and G3BP2) to inhibit SG formation. This will allow viral packaging and propagation in host cells. Based on a genomic-guided meta-analysis, here we identify upstream regulatory elements modulating the expression of G3BP1 and G3BP2 (collectively called G3BP1/2). Using this strategy, we have identified FOXA1, YY1, SYK, E2F-1, and TGFBR2 as activators and SIN3A, SRF, and AKT-1 as repressors of G3BP1/2 genes. Panels of the activators and repressors were then used to identify drugs that change their gene expression signatures. Two drugs, imatinib, and decitabine have been identified as putative modulators of G3BP1/2 genes and their regulators, suggesting their role as COVID-19 mitigation agents. Molecular docking analysis suggests that both drugs bind to G3BP1/2 with a much higher affinity than the SARS-CoV-2 N protein. This study reports imatinib and decitabine as candidate drugs against N protein and G3BP1/2 protein.


Sujet(s)
Protéines adaptatrices de la transduction du signal/composition chimique , Traitements médicamenteux de la COVID-19 , Protéines de la nucléocapside des coronavirus/composition chimique , Helicase/composition chimique , Décitabine/composition chimique , Mésilate d'imatinib/composition chimique , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Protéines liant le poly-adp-ribose/composition chimique , RNA helicases/composition chimique , Protéines à motif de reconnaissance de l'ARN/composition chimique , Protéines de liaison à l'ARN/composition chimique , SARS-CoV-2/composition chimique , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Protéines adaptatrices de la transduction du signal/métabolisme , COVID-19/métabolisme , Protéines de la nucléocapside des coronavirus/métabolisme , Helicase/antagonistes et inhibiteurs , Helicase/métabolisme , Décitabine/pharmacologie , Systèmes de délivrance de médicaments , Génomique , Mésilate d'imatinib/pharmacologie , Phosphoprotéines/composition chimique , Phosphoprotéines/métabolisme , Protéines liant le poly-adp-ribose/antagonistes et inhibiteurs , Protéines liant le poly-adp-ribose/métabolisme , RNA helicases/antagonistes et inhibiteurs , RNA helicases/métabolisme , Protéines à motif de reconnaissance de l'ARN/antagonistes et inhibiteurs , Protéines à motif de reconnaissance de l'ARN/métabolisme , Protéines de liaison à l'ARN/antagonistes et inhibiteurs , Protéines de liaison à l'ARN/métabolisme , SARS-CoV-2/métabolisme
20.
Cancer Lett ; 520: 409-421, 2021 11 01.
Article de Anglais | MEDLINE | ID: mdl-34419501

RÉSUMÉ

Bcl2-associated athanogene 4 (BAG4) has been found to be aberrantly expressed in several types of human cancers. However, little is known about its expression, role, and clinical significance in gastric cancer (GC). In this study, we aimed to address these issues and to explore the underlying mechanisms. The expression level of BAG4, measured by immunohistochemistry, was significantly higher in GC tissues than in paired normal tissues. Elevated BAG4 expression was positively correlated with T stage, lymph node metastasis, and tumor size of GC and was associated with unfavorable outcomes of the patients. The overexpression of BAG4 promoted the in vitro invasion and in vivo metastasis of GC cells, and opposite results were observed after silencing of BAG4. Silencing of BAG4 significantly reduced the phosphorylation of PI3K, AKT, and p65, whereas overexpression of BAG4 markedly enhanced the phosphorylation of these molecules. At the same time, manipulating BAG4 expression resulted in the corresponding changes in p65 nuclear translocation and ZEB1 expression. Luciferase reporter and chromatin immunoprecipitation assays verified that p65 binds to the promoter of ZEB1 to upregulate its transcription. Our results demonstrate that BAG4 plays an oncogenic role in the invasion and metastasis of GC cells by activating the PI3K/AKT/NF-κB/ZEB1 axis to induce epithelial-mesenchymal transition.


Sujet(s)
Protéines adaptatrices de la transduction du signal/génétique , Tumeurs de l'estomac/génétique , Facteur de transcription RelA/génétique , Facteur de transcription Zeb1/génétique , Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux/génétique , Humains , Facteur de transcription NF-kappa B/génétique , Invasion tumorale/génétique , Invasion tumorale/anatomopathologie , Protéine oncogène v-akt/génétique , Phosphatidylinositol 3-kinases/génétique , Transduction du signal/génétique , Tumeurs de l'estomac/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...