Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.934
Filtrer
1.
Int J Mol Sci ; 25(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892219

RÉSUMÉ

Anoctamin1 (ANO1), a calcium-activated chloride channel, is overexpressed in a variety of cancer cells, including prostate cancer, and is involved in cancer cell proliferation, migration, and invasion. Inhibition of ANO1 in these cancer cells exhibits anticancer effects. In this study, we conducted a screening to identify novel ANO1 inhibitors with anticancer effects using PC-3 human prostate carcinoma cells. Screening of 2978 approved and investigational drugs revealed that hemin is a novel ANO1 inhibitor with an IC50 value of 0.45 µM. Notably, hemin had no significant effect on intracellular calcium signaling and cystic fibrosis transmembrane conductance regulator (CFTR), a cyclic AMP (cAMP)-regulated chloride channel, and it showed a weak inhibitory effect on ANO2 at 3 µM, a concentration that completely inhibits ANO1. Interestingly, hemin also significantly decreased ANO1 protein levels and strongly inhibited the cell proliferation and migration of PC-3 cells in an ANO1-dependent manner. Furthermore, it strongly induced caspase-3 activation, PARP degradation, and apoptosis in PC-3 cells. These findings suggest that hemin possesses anticancer properties via ANO1 inhibition and could be considered for development as a novel treatment for prostate cancer.


Sujet(s)
Anoctamine-1 , Antinéoplasiques , Apoptose , Mouvement cellulaire , Prolifération cellulaire , Hémine , Protéines tumorales , Tumeurs de la prostate , Humains , Anoctamine-1/métabolisme , Anoctamine-1/antagonistes et inhibiteurs , Mâle , Hémine/pharmacologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Mouvement cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Cellules PC-3
2.
Molecules ; 29(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38893358

RÉSUMÉ

Pseudoginsenoside DQ (PDQ), an ocotillol-type ginsenoside, is synthesized with protopanaxadiol through oxidative cyclization. PDQ exhibits good anti-arrhythmia activity. However, the inhibitory effect of PDQ on the cytochrome 450 (CYP450) enzymes and major drug transporters is still unclear. Inhibition of CYP450 and drug transporters may affect the efficacy of the drugs being used together with PDQ. These potential drug-drug interactions (DDIs) are essential for the clinical usage of drugs. In this study, we investigated the inhibitory effect of PDQ on seven CYP450 enzymes and seven drug transporters with in vitro models. PDQ has a significant inhibitory effect on CYP2C19 and P-glycoprotein (P-gp) with a half-inhibitory concentration (IC50) of 0.698 and 0.41 µM, respectively. The inhibition of CYP3A4 and breast cancer-resistant protein (BCRP) is less potent, with IC50 equal to 2.02-6.79 and 1.08 µM, respectively.


Sujet(s)
Cytochrome P-450 enzyme system , Interactions médicamenteuses , Ginsénosides , Humains , Ginsénosides/pharmacologie , Ginsénosides/composition chimique , Cytochrome P-450 enzyme system/métabolisme , Inhibiteurs des enzymes du cytochrome P-450/pharmacologie , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Cytochrome P-450 CYP3A/métabolisme , Cytochrome P-450 CYP2C19/métabolisme , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs
3.
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
4.
Clin Pharmacol Drug Dev ; 13(7): 755-769, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38752475

RÉSUMÉ

Pritelivir is a novel viral helicase-primase inhibitor active against herpes simplex virus. In vitro drug-drug interaction studies indicated that pritelivir has the potential for clinically relevant interactions on the cytochrome P450 (CYP) enzymes 2C8, 2C9, 3A4, and 2B6, and intestinal uptake transporter organic anion transporting polypeptide (OATP) 2B1 and efflux transporter breast cancer resistance protein (BCRP). This was evaluated in 2 clinical trials. In 1 trial the substrates flurbiprofen (CYP2C9), bupropion (CYP2B6), and midazolam (CYP3A4) were administered simultaneously as part of the Geneva cocktail, while the substrate celiprolol (OAPT2B1) was administered separately. In another trial, the substrates repaglinide (CYP2C8) and rosuvastatin (BCRP) were administered separately. Exposure parameters of the substrates and their metabolites (flurbiprofen and bupropion only) were compared after administration with or without pritelivir under therapeutic concentrations. The results of these trials indicated that pritelivir has no clinically relevant effect on the exposure of substrates for the intestinal uptake transporter OATP2B1 and the CYP enzymes 3A4, 2B6, 2C9, and 2C8, and has a weak inhibitory effect on the intestinal efflux transporter BCRP. In summary, the results suggest that pritelivir has a low drug-drug interaction potential.


Sujet(s)
Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP , Cytochrome P-450 enzyme system , Interactions médicamenteuses , Humains , Cytochrome P-450 enzyme system/métabolisme , Cytochrome P-450 enzyme system/effets des médicaments et des substances chimiques , Femelle , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Mâle , Adulte , Bupropion/pharmacologie , Bupropion/pharmacocinétique , Sulfonamides/pharmacologie , Adulte d'âge moyen , Rosuvastatine de calcium/pharmacologie , Rosuvastatine de calcium/pharmacocinétique , Flurbiprofène/pharmacologie , Flurbiprofène/pharmacocinétique , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Transporteurs d'anions organiques/métabolisme , Transporteurs d'anions organiques/antagonistes et inhibiteurs , Carbamates/pharmacologie , Midazolam/pharmacocinétique , Midazolam/pharmacologie , Jeune adulte , Pipéridines/pharmacologie , Pipéridines/pharmacocinétique
5.
Cell Calcium ; 121: 102905, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38788257

RÉSUMÉ

TMEM16 proteins, also known as anoctamins, are a family of ten membrane proteins with various tissue expression and subcellular localization. TMEM16A (anoctamin 1) is a plasma membrane protein that acts as a calcium-activated chloride channel. It is expressed in many types of epithelial cells, smooth muscle cells and some neurons. In airway epithelial cells, TMEM16A expression is particularly enhanced by inflammatory stimuli that also promote goblet cell metaplasia and mucus hypersecretion. Therefore, pharmacological modulation of TMEM16A could be beneficial to improve mucociliary clearance in chronic obstructive respiratory diseases. However, the correct approach to modulate TMEM16A activity (activation or inhibition) is still debated. Pharmacological inhibitors of TMEM16A could also be useful as anti-hypertensive agents given the TMEM16A role in smooth muscle contraction. In contrast to TMEM16A, TMEM16F (anoctamin 6) behaves as a calcium-activated phospholipid scramblase, responsible for the externalization of phosphatidylserine on cell surface. Inhibitors of TMEM16F could be useful as anti-coagulants and anti-viral agents. The role of other anoctamins as therapeutic targets is still unclear since their physiological role is still to be defined.


Sujet(s)
Anoctamine-1 , Humains , Animaux , Anoctamine-1/métabolisme , Anoctamine-1/antagonistes et inhibiteurs , Anoctamines/métabolisme , Canaux chlorure/métabolisme , Canaux chlorure/antagonistes et inhibiteurs , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Protéines de transfert des phospholipides/métabolisme , Protéines de transfert des phospholipides/antagonistes et inhibiteurs
6.
Int J Mol Sci ; 25(10)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38791198

RÉSUMÉ

MTX-211 is a first-in-class dual inhibitor of epidermal growth factor receptor (EGFR) and phosphoinositide-3 kinase (PI3K) signaling pathways with a compelling pharmaceutical profile and could enhance the effectiveness of mitogen-activated protein kinase kinase (MEK) inhibitor therapy in colorectal tumors with KRAS mutations. However, the specific mechanisms contributing to the acquired resistance to MTX-211 in human cancers remain elusive. Here, we discovered that the overexpression of the ATP-binding cassette (ABC) drug transporter ABCG2, a prevalent mechanism associated with multidrug resistance (MDR), could diminish the effectiveness of MTX-211 in human cancer cells. We showed that the drug efflux activity of ABCG2 substantially decreased the intracellular accumulation of MTX-211 in cancer cells. As a result, the cytotoxicity and effectiveness of MTX-211 in suppressing the activation of the EGFR and PI3K pathways were significantly attenuated in cancer cells overexpressing ABCG2. Moreover, the enhancement of the MTX-211-stimulated ATPase activity of ABCG2 and the computational molecular docking analysis illustrating the binding of MTX-211 to the substrate-binding sites of ABCG2 offered a further indication for the interaction between MTX-211 and ABCG2. In summary, our findings indicate that MTX-211 acts as a substrate for ABCG2, underscoring the involvement of ABCG2 in the emergence of resistance to MTX-211. This finding carries clinical implications and merits further exploration.


Sujet(s)
Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Protéines tumorales , Humains , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Protéines tumorales/métabolisme , Protéines tumorales/génétique , Protéines tumorales/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Phosphatidylinositol 3-kinases/métabolisme , Simulation de docking moléculaire , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Antinéoplasiques/pharmacologie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie
7.
Photochem Photobiol Sci ; 23(6): 1067-1075, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38625651

RÉSUMÉ

Photodynamic Therapy (PDT) is an emerging method to treat colorectal cancers (CRC). Hypericin (HYP) is an effective mediator of PDT and the ABCG2 inhibitor, Febuxostat (FBX) could augment PDT. HT29 and HEK293 cells showed light dependant cytotoxic response to PDT in both 2D and 3D cell models. FBX co-treatment was not found to improve PDT cytotoxicity. Next, ABCG2 protein expression was observed in HT29 but not in HEK293 cells. However, ABCG2 gene expression analysis did not support protein expression results as ABCG2 gene expression results were found to be higher in HEK293 cells. Although HYP treatment was found to significantly reduce ABCG2 gene expression levels in both cell lines, FBX treatment partially restored ABCG2 gene expression. Our findings indicate that FBX co-treatment may not be suitable for augmenting HYP-mediated PDT in CRC but could potentially be useful for other applications.


Sujet(s)
Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP , Anthracènes , Tumeurs colorectales , Fébuxostat , Protéines tumorales , Pérylène , Photothérapie dynamique , Photosensibilisants , Humains , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/antagonistes et inhibiteurs , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Anthracènes/pharmacologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Pérylène/analogues et dérivés , Pérylène/pharmacologie , Fébuxostat/pharmacologie , Fébuxostat/usage thérapeutique , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Cellules HEK293 , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules HT29 , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique
8.
Expert Opin Ther Targets ; 28(4): 259-271, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38653737

RÉSUMÉ

INTRODUCTION: Phosphatase of regenerating liver (PRL) family proteins, also known as protein tyrosine phosphatase 4A (PTP4A), have been implicated in many types of cancers. The PRL family of phosphatases consists of three members, PRL1, PRL2, and PRL3. PRLs have been shown to harbor oncogenic potentials and are highly expressed in a variety of cancers. Given their roles in cancer progression and metastasis, PRLs are potential targets for anticancer therapies. However, additional studies are needed to be performed to fully understand the roles of PRLs in blood cancers. AREAS COVERED: In this review, we will summarize recent studies of PRLs in normal and malignant hematopoiesis, the role of PRLs in regulating various signaling pathways, and the therapeutic potentials of targeting PRLs in hematological malignancies. We will also discuss how to improve current PRL inhibitors for cancer treatment. EXPERT OPINION: Although PRL inhibitors show promising therapeutic effects in preclinical studies of different types of cancers, moving PRL inhibitors from bench to bedside is still challenging. More potent and selective PRL inhibitors are needed to target PRLs in hematological malignancies and improve treatment outcomes.


Sujet(s)
Antinéoplasiques , Tumeurs hématologiques , Thérapie moléculaire ciblée , Protein Tyrosine Phosphatases , Transduction du signal , Humains , Protein Tyrosine Phosphatases/antagonistes et inhibiteurs , Protein Tyrosine Phosphatases/métabolisme , Tumeurs hématologiques/traitement médicamenteux , Animaux , Antinéoplasiques/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/métabolisme , Développement de médicament , Protéines membranaires , Protéines du cycle cellulaire
9.
Ann Hematol ; 103(7): 2405-2417, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38538975

RÉSUMÉ

Multiple myeloma (MM) is a common malignant hematologic neoplasm, and the involvement of epigenetic modifications in its development and drug resistance has received widespread attention. Ferroptosis, a new ferroptosis-dependent programmed death mode, is closely associated with the development of MM. The novel methyltransferase inhibitor DCG066 has higher cell activity, but its mechanism of action in MM has not been clarified. Here, we found that DCG066 (5µM) inhibited the proliferation and induced ferroptosis in MM cells; the intracellular levels of ROS, iron, and MDA were significantly elevated, and the level of GSH was reduced after the treatment of DCG066; The protein expression levels of SLC7A11, GPX4, Nrf2 and HO-1 were significantly reduced, and these phenomena could be reversed by ferroptosis inhibitor Ferrostatin-1 (Fer-1) and Nrf2 activator Tert-butyl hydroquinone (TBHQ). Meanwhile, the protein expression levels of Keap1 was increased, and heat shock proteins (HSP70, HSP90 and HSPB1) were reduced after DCG066 treatment. In conclusion, this study confirmed that DCG066 inhibits MM proliferation and induces ferroptosis via the Nrf2/HO-1 pathway.


Sujet(s)
Ferroptose , Heme oxygenase-1 , Myélome multiple , Facteur-2 apparenté à NF-E2 , Transduction du signal , Myélome multiple/traitement médicamenteux , Myélome multiple/anatomopathologie , Myélome multiple/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Humains , Facteur-2 apparenté à NF-E2/métabolisme , Heme oxygenase-1/métabolisme , Heme oxygenase-1/génétique , Lignée cellulaire tumorale , Transduction du signal/effets des médicaments et des substances chimiques , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/antagonistes et inhibiteurs , Histone-lysine N-methyltransferase/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , Antigènes d'histocompatibilité
10.
Comput Biol Chem ; 110: 108039, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38471352

RÉSUMÉ

Hepatocellular carcinoma (HCC) persists to be one of the most devastating and deadliest malignancies globally. Recent research into the molecular signaling networks entailed in many malignancies has given some prominent insights that can be leveraged to create molecular therapeutics for combating HCC. Therefore, in the current communication, an in-silico drug repurposing approach has been employed to target the function of PTP4A3/PRL-3 protein in HCC using antidepressants: Fluoxetine hydrochloride, Citalopram, Amitriptyline, Imipramine, and Escitalopram oxalate as the desired ligands. The density function theory (DFT) and chemical absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters for the chosen ligands were evaluated to comprehend the pharmacokinetics, drug-likeness properties, and bioreactivity of the ligands. The precise interaction mechanism was explored using computational methods such as molecular docking and molecular dynamics (MD) simulation studies to assess the inhibitory effect and the stability of the interactions against the protein of interest. Escitalopram oxalate exhibited a comparatively significant docking score (-7.4 kcal/mol) compared to the control JMS-053 (-6.8 kcal/mol) against the PRL-3 protein. The 2D interaction plots exhibited an array of hydrophobic and hydrogen bond interactions. The findings of the ADMET forecast confirmed that it adheres to Lipinski's rule of five with no violations, and DFT analysis revealed a HOMO-LUMO energy gap of -0.26778 ev, demonstrating better reactivity than the control molecule. The docked complexes were subjected to MD studies (100 ns) showing stable interactions. Considering all the findings, it can be concluded that Escitalopram oxalate and related therapeutics can act as potential pharmacological candidates for targeting the activity of PTP4A3/PRL-3 in HCC.


Sujet(s)
Antidépresseurs , Carcinome hépatocellulaire , Escitalopram , Tumeurs du foie , Simulation de docking moléculaire , Protein Tyrosine Phosphatases , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Protein Tyrosine Phosphatases/antagonistes et inhibiteurs , Protein Tyrosine Phosphatases/métabolisme , Antidépresseurs/pharmacologie , Antidépresseurs/composition chimique , Escitalopram/composition chimique , Escitalopram/pharmacologie , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Simulation de dynamique moléculaire , Oxalates/composition chimique , Oxalates/métabolisme , Théorie de la fonctionnelle de la densité , Structure moléculaire , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique
11.
J Clin Invest ; 134(10)2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38530357

RÉSUMÉ

Despite widespread utilization of immunotherapy, treating immune-cold tumors remains a challenge. Multiomic analyses and experimental validation identified the OTUD4/CD73 proteolytic axis as a promising target in treating immune-suppressive triple negative breast cancer (TNBC). Mechanistically, deubiquitylation of CD73 by OTUD4 counteracted its ubiquitylation by TRIM21, resulting in CD73 stabilization inhibiting tumor immune responses. We further demonstrated the importance of TGF-ß signaling for orchestrating the OTUD4/CD73 proteolytic axis within tumor cells. Spatial transcriptomics profiling discovered spatially resolved features of interacting malignant and immune cells pertaining to expression levels of OTUD4 and CD73. In addition, ST80, a newly developed inhibitor, specifically disrupted proteolytic interaction between CD73 and OTUD4, leading to reinvigoration of cytotoxic CD8+ T cell activities. In preclinical models of TNBC, ST80 treatment sensitized refractory tumors to anti-PD-L1 therapy. Collectively, our findings uncover what we believe to be a novel strategy for targeting the immunosuppressive OTUD4/CD73 proteolytic axis in treating immune-suppressive breast cancers with the inhibitor ST80.


Sujet(s)
5'-Nucleotidase , Protéolyse , Tumeurs du sein triple-négatives , Animaux , Femelle , Humains , Souris , 5'-Nucleotidase/génétique , 5'-Nucleotidase/immunologie , 5'-Nucleotidase/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Protéines liées au GPI/immunologie , Protéines liées au GPI/génétique , Protéines liées au GPI/métabolisme , Protéines liées au GPI/antagonistes et inhibiteurs , Protéines tumorales/immunologie , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Ubiquitination , Ubiquitin-specific proteases
12.
Talanta ; 274: 125987, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38552478

RÉSUMÉ

Multidrug resistance (MDR) is a dominant challenge in cancer chemotherapy failure. The over-expression of breast cancer resistance protein (BCRP) in tumorous cells, along with its extensive substrate profile, is a leading cause of tumor MDR. Herein, on the basis of styrene maleic acid (SMA) polymer membrane protein stabilization strategy and surface plasmon resonance (SPR) biosensor, a novel high-throughput screening (HTS) system for BCRP inhibitors has been established. Firstly, LLC-PK1 and LLC-PK1/BCRP cell membranes were co-incubated with SMA polymers to construct SMA lipid particles (SMALPs). PK1-SMALPs were thus immobilized in channel 1 of the L1 chip as the reference channel, and BCRP-SMALPs were immobilized in channel 2 as the detection channel to establish the BCRP-SMALPs-SPR screening system. The methodological investigation demonstrated that the screening system was highly specific and stable. Three active compounds were screened out from 26 natural products and their affinity constants with BCRP were determined. The KD of xanthotoxin, bergapten, and naringenin were 5.14 µM, 4.57 µM, and 3.72 µM, respectively. The in vitro cell verification experiments demonstrated that xanthotoxin, bergapten, and naringenin all significantly increased the sensitivity of LLC-PK1/BCRP cells to mitoxantrone with possessing reversal BCRP-mediated MDR activity. Collectively, the developed BCRP-SMALPs-SPR screening system in this study has the advantages of rapidity, efficiency, and specificity, providing a novel strategy for the in-depth screening of BCRP inhibitors with less side effects and higher efficacy.


Sujet(s)
Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP , Maléates , Protéines tumorales , Résonance plasmonique de surface , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/antagonistes et inhibiteurs , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Résonance plasmonique de surface/méthodes , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/métabolisme , Protéines tumorales/analyse , Humains , Maléates/composition chimique , Maléates/pharmacologie , Animaux , Tests de criblage à haut débit/méthodes , Suidae , Polystyrènes/composition chimique , Techniques de biocapteur/méthodes
13.
Int J Mol Sci ; 25(2)2024 Jan 20.
Article de Anglais | MEDLINE | ID: mdl-38279296

RÉSUMÉ

Exosomal microRNAs (miRNAs) are novel, non-invasive biomarkers for facilitating communication and diagnosing cancer. However, only a few studies have investigated their function and role in the clinical diagnosis of breast cancer. To address this gap, we established a stable cell line, MDA-MB-231-CD63-RFP, and recruited 112 female participants for serum collection. We screened 88 exosomal miRNAs identified through microarray analysis of 231-CD63 and literature screening using real-time PCR; only exosomal miR-92b-5p was significantly increased in patients with breast cancer. It had a significant correlation with stage and discriminated patients from the control with an AUC of 0.787. Exosomal miR-92b-5p impacted the migration, adhesion, and spreading ability of normal human mammary epithelial recipient cells through the downregulation of the actin dynamics regulator MTSS1L. In clinical breast cancer tissue, the expression of MTSS1L was significantly inversely correlated with tissue miR-92b-5p, and high expression of MTSS1L was associated with better 10-year overall survival rates in patients undergoing hormone therapy. In summary, our studies demonstrated that exosomal miR-92b-5p might function as a non-invasive body fluid biomarker for breast cancer detection and provide a novel therapeutic strategy in the axis of miR-92b-5p to MTSS1L for controlling metastasis and improving patient survival.


Sujet(s)
Marqueurs biologiques , Tumeurs du sein , Exosomes , microARN , Femelle , Humains , Marqueurs biologiques/métabolisme , Tumeurs du sein/diagnostic , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Transformation cellulaire néoplasique/métabolisme , Exosomes/génétique , Exosomes/métabolisme , microARN/métabolisme , Protéines tumorales/antagonistes et inhibiteurs
14.
J Med Chem ; 66(4): 2804-2831, 2023 02 23.
Article de Anglais | MEDLINE | ID: mdl-36780419

RÉSUMÉ

ABCB1 and ABCG2 are the important ATP-binding cassette (ABC) transporters associated with multidrug resistance (MDR). Herein, we designed a series of imidazo[1,2-a]pyridine derivatives as dual-target inhibitors of ABCB1 and ABCG2 through the scaffold hopping strategy. Compound Y22 displayed potential efflux function inhibitory toward both ABCB1 and ABCG2 (reversal fold: ABCB1 = 8.35 and ABCG2 = 2.71) without obvious cytotoxicity. Y22 also enhanced the potency of antiproliferative drugs in vitro. Mechanistic studies demonstrated that Y22 slightly suppressed ATPase activity but did not affect the protein expression of ABCB1 or ABCG2. Notably, Y22 exhibited negligible CYP3A4 inhibition and enhanced the antiproliferative activity of adriamycin in vivo by restoring the sensitivity of resistant cells. Thus, Y22 may be effective clinically in combination with common chemotherapy agents. In summary, Y22 is a potential dual-target inhibitor that reverses MDR by blocking the efflux function of ABCB1 and ABCG2.


Sujet(s)
Membre-2 de la sous-famille B à cassette de liaison à l'ATP , Antinéoplasiques , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Protéines tumorales , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Membre-2 de la sous-famille B à cassette de liaison à l'ATP/antagonistes et inhibiteurs , Protéines tumorales/antagonistes et inhibiteurs , Humains
15.
Proc Natl Acad Sci U S A ; 119(30): e2120339119, 2022 07 26.
Article de Anglais | MEDLINE | ID: mdl-35857873

RÉSUMÉ

During translation initiation, eIF4G1 dynamically interacts with eIF4E and eIF1. While the role of eIF4E-eIF4G1 is well established, the regulatory functions of eIF4G1-eIF1 are poorly understood. Here, we report the identification of the eIF4G1-eIF1 inhibitors i14G1-10 and i14G1-12. i14G1s directly bind eIF4G1 and inhibit translation in vitro and in the cell, and their effects on translation are dependent on eIF4G1 levels. Translatome analyses revealed that i14G1s mimic eIF1 and eIF4G1 perturbations on the stringency of start codon selection and the opposing roles of eIF1-eIF4G1 in scanning-dependent and scanning-independent short 5' untranslated region (UTR) translation. Remarkably, i14G1s activate ER/unfolded protein response (UPR) stress-response genes via enhanced ribosome loading, elevated 5'UTR translation at near-cognate AUGs, and unexpected concomitant up-regulation of coding-region translation. These effects are, at least in part, independent of eIF2α-phosphorylation. Interestingly, eIF4G1-eIF1 interaction itself is negatively regulated by ER stress and mTOR inhibition. Thus, i14G1s uncover an unknown mechanism of ER/UPR translational stress response and are valuable research tools and potential drugs against diseases exhibiting dysregulated translation.


Sujet(s)
Stress du réticulum endoplasmique , Facteur-2 d'initiation eucaryote , Facteur-4G d'initiation eucaryote , Facteurs d'initiation eucaryotes , Protéines tumorales , Protéines de tissu nerveux , Réponse aux protéines mal repliées , Animaux , Codon d'initiation , Stress du réticulum endoplasmique/génétique , Facteur-2 d'initiation eucaryote/métabolisme , Facteur-4G d'initiation eucaryote/antagonistes et inhibiteurs , Facteur-4G d'initiation eucaryote/métabolisme , Facteurs d'initiation eucaryotes/antagonistes et inhibiteurs , Facteurs d'initiation eucaryotes/métabolisme , Humains , Souris , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/métabolisme , Protéines de tissu nerveux/antagonistes et inhibiteurs , Protéines de tissu nerveux/métabolisme , Phosphorylation , Biosynthèse des protéines , Réponse aux protéines mal repliées/génétique
16.
J Med Chem ; 65(10): 7231-7245, 2022 05 26.
Article de Anglais | MEDLINE | ID: mdl-35522528

RÉSUMÉ

MAGE proteins are cancer testis antigens (CTAs) that are characterized by highly conserved MAGE homology domains (MHDs) and are increasingly being found to play pivotal roles in promoting aggressive cancer types. MAGE-A4, in particular, increases DNA damage tolerance and chemoresistance in a variety of cancers by stabilizing the E3-ligase RAD18 and promoting trans-lesion synthesis (TLS). Inhibition of the MAGE-A4:RAD18 axis could sensitize cancer cells to chemotherapeutics like platinating agents. We use an mRNA display of thioether cyclized peptides to identify a series of potent and highly selective macrocyclic inhibitors of the MAGE-A4:RAD18 interaction. Co-crystal structure indicates that these inhibitors bind in a pocket that is conserved across MHDs but take advantage of A4-specific residues to achieve high isoform selectivity. Cumulatively, our data represent the first reported inhibitor of the MAGE-A4:RAD18 interaction and establish biochemical tools and structural insights for the future development of MAGE-A4-targeted cellular probes.


Sujet(s)
Antigènes néoplasiques , Protéines tumorales , Tumeurs , Antigènes néoplasiques/composition chimique , Altération de l'ADN , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Humains , Mâle , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/composition chimique , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Peptides cycliques/composition chimique , Peptides cycliques/pharmacologie , Relation structure-activité , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme
17.
Eur J Med Chem ; 237: 114346, 2022 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-35483322

RÉSUMÉ

The primary source of failure of cancer therapies is multidrug resistance (MDR), which can be caused by different mechanisms, including the overexpression of ABC transporters in cancer cells. Among the 48 human ABC proteins, the breast cancer resistance protein (BCRP/ABCG2) has been described as a pivotal player in cancer resistance. The use of functional inhibitors and expression modulators is a promising strategy to overcome the MDR caused by ABCG2. Despite the lack of clinical trials using ABCG2 inhibitors, many compounds have already been discovered. This review presents an overview about various ABCG2 inhibitors that have been identified, discussing some chemical aspects and the main experimental methods used to identify and characterize the mechanisms of new inhibitors. In addition, some biological requirements to pursue preclinical tests are described. Finally, we discuss the potential use of ABCG2 inhibitors in cancer stem cells (CSC) for improving the objective response rate and the mechanism of ABCG2 modulators at transcriptional and protein expression levels.


Sujet(s)
Antinéoplasiques , Tumeurs du sein , Protéines tumorales , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/antagonistes et inhibiteurs , Membre-2 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Femelle , Humains , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques
18.
Blood ; 139(8): 1160-1176, 2022 02 24.
Article de Anglais | MEDLINE | ID: mdl-35201323

RÉSUMÉ

Anti-CD38 monoclonal antibodies (mAbs) represent a breakthrough in the treatment of multiple myeloma (MM), yet some patients fail to respond or progress quickly with this therapy, highlighting the need for novel approaches. In this study we compared the preclinical efficacy of SAR442085, a next-generation anti-CD38 mAb with enhanced affinity for activating Fcγ receptors (FcγR), with first-generation anti-CD38 mAb daratumumab and isatuximab. In surface plasmon resonance and cellular binding assays, we found that SAR442085 had higher binding affinity than daratumumab and isatuximab for FcγRIIa (CD32a) and FcγRIIIa (CD16a). SAR442085 also exhibited better in vitro antibody-dependent cellular cytotoxicity (ADCC) against a panel of MM cells expressing variable CD38 receptor densities including MM patients' primary plasma cells. The enhanced ADCC of SAR442085 was confirmed using NK-92 cells bearing low and high affinity FcγRIIIa (CD16a)-158F/V variants. Using MM patients' primary bone marrow cells, we confirmed that SAR442085 had an increased ability to engage FcγRIIIa, resulting in higher natural killer (NK) cell activation and degranulation against primary plasma cells than preexisting Fc wild-type anti-CD38 mAbs. Finally, using huFcgR transgenic mice that express human Fcγ receptors under the control of their human regulatory elements, we demonstrated that SAR442085 had higher NK cell-dependent in vivo antitumor efficacy and better survival than daratumumab and isatuximab against EL4 thymoma or VK*MYC myeloma cells overexpressing human CD38. These results highlight the preclinical efficacy of SAR442085 and support the current evaluation of this next-generation anti-CD38 antibody in phase I clinical development in patients with relapsed/refractory MM.


Sujet(s)
Antigènes CD38/antagonistes et inhibiteurs , Antinéoplasiques immunologiques/pharmacologie , Cellules de la moelle osseuse , Glycoprotéines membranaires/antagonistes et inhibiteurs , Myélome multiple , Protéines tumorales/antagonistes et inhibiteurs , Antigènes CD38/métabolisme , Animaux , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Lignée cellulaire tumorale , Cellules HEK293 , Humains , Glycoprotéines membranaires/métabolisme , Souris transgéniques , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Protéines tumorales/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Molecules ; 27(4)2022 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-35208952

RÉSUMÉ

For most researchers, discovering new anticancer drugs to avoid the adverse effects of current ones, to improve therapeutic benefits and to reduce resistance is essential. Because the COX-2 enzyme plays an important role in various types of cancer leading to malignancy enhancement, inhibition of apoptosis, and tumor-cell metastasis, an indispensable objective is to design new scaffolds or drugs that possess combined action or dual effect, such as kinase and COX-2 inhibition. The start compounds A1 to A6 were prepared through the diazo coupling of 3-aminoacetophenone with a corresponding phenol and then condensed with two new chalcone series, C7-18. The newly synthesized compounds were assessed against both COX-2 and epidermal growth factor receptor (EGFR) for their inhibitory effect. All novel compounds were screened for cytotoxicity against five cancer cell lines. Compounds C9 and G10 exhibited potent EGFR inhibition with IC50 values of 0.8 and 1.1 µM, respectively. Additionally, they also displayed great COX-2 inhibition with IC50 values of 1.27 and 1.88 µM, respectively. Furthermore, the target compounds were assessed for their cytotoxicity against pancreatic ductal cancer (Panc-1), lung cancer (H-460), human colon cancer (HT-29), human malignant melanoma (A375) and pancreatic cancer (PaCa-2) cell lines. Interestingly, compounds C10 and G12 exhibited the strongest cytotoxic effect against PaCa-2 with average IC50 values of 0.9 and 0.8 µM, respectively. To understand the possible binding modes of the compounds under investigation with the receptor cites of EGFR and COX-2, a virtual docking study was conducted.


Sujet(s)
Antinéoplasiques , Chalcones , Inhibiteurs de la cyclooxygénase 2 , Protéines tumorales , Tumeurs , Inhibiteurs de protéines kinases , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Chalcones/synthèse chimique , Chalcones/composition chimique , Chalcones/pharmacologie , Inhibiteurs de la cyclooxygénase 2/synthèse chimique , Inhibiteurs de la cyclooxygénase 2/composition chimique , Inhibiteurs de la cyclooxygénase 2/pharmacologie , Tests de criblage d'agents antitumoraux , Récepteurs ErbB/antagonistes et inhibiteurs , Humains , Structure moléculaire , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie
20.
J Adv Res ; 36: 147-161, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35127170

RÉSUMÉ

Introduction: Globally, hepatocellular carcinoma (HCC) is the sixth most common malignancy and it has the fourth highest mortality. MicroRNAs play a significant part in biological processes in cell formation and advancement by targeting genes in many cancers including HCC. Objective: In the present study we examine the involvement of miR-4521 and FAM129A correlations in HCC occurrence and progression. Methods: Expression levels of miR-4521 and FAM129A in HCC tissues and cells were detected. Immunohistochemistry was carried out to detect expression of FAM129A, MMP9 and TIMP-1 in HCC tissues. Western blot assays were used to examine expression levels of different genes involve in signaling pathways. Transwell chamber, MTT and wound healing assays were performed to check cell migration, invasion and proliferation rates. Results: Overexpression of FAM129A positively correlated with upregulation of MMP9 and negatively correlated with TIMP-1 in HCC patient samples, which encouraged progression and metastasis of HCC. An antagonistic relation between miR-4521 and FAM129A was detected in current study, down-regulation of miR-4521 and up-regulation of FAM129A was demonstrated in HCC tissues and cell lines as compare to normal tissue samples and the normal cell line LO2. Overexpressing miR-4521 and silencing FAM129A impaired HCC cell migratory and invasive properties and suppressed cell proliferation. Mutually, miR-4521-FAM129A axial regulation inhibited in vitro proliferation of cells by promoting apoptosis through the p-FAK/p-AKT/MDM2/P53 and p-FAK/p-AKT/BCL-2/BAX/Cytochrome-C/Caspase-3/Caspase-9 pathways, respectively, and suppressed the migration and invasion capabilities of HCCLM3 and HepG2 cells via the TIMP-1/MMP9/MMP2 and p-FAK/p-AKT pathway. Conclusion: Our work found the axial regulation mechanism of miR-4521-FAM129A in HCC. Deficiency of miR-4521 and abundance of FAM129A synergistically enhanced cancer progression by increasing cell proliferation and malignant invasion and by inhibiting apoptosis. These discoveries suggest that miR-4521/FAM129A might play a vital role in hepatic cancer progression and could be a candidate for its therapy.


Sujet(s)
Marqueurs biologiques tumoraux/antagonistes et inhibiteurs , Carcinome hépatocellulaire , Tumeurs du foie , microARN , Protéines tumorales/antagonistes et inhibiteurs , Marqueurs biologiques tumoraux/métabolisme , Carcinome hépatocellulaire/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du foie/métabolisme , microARN/génétique , microARN/métabolisme , Invasion tumorale/génétique , Protéines tumorales/métabolisme , Phosphorylation , Protéines proto-oncogènes c-akt/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...