Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22.701
Filtrer
1.
Cancer Med ; 13(1): e6945, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-39102671

RÉSUMÉ

INTRODUCTION: Adaptive mutagenesis observed in colorectal cancer (CRC) cells upon exposure to EGFR inhibitors contributes to the development of resistance and recurrence. Multiple investigations have indicated a parallel between cancer cells and bacteria in terms of exhibiting adaptive mutagenesis. This phenomenon entails a transient and coordinated escalation of error-prone translesion synthesis polymerases (TLS polymerases), resulting in mutagenesis of a magnitude sufficient to drive the selection of resistant phenotypes. METHODS: In this study, we conducted a comprehensive pan-transcriptome analysis of the regulatory framework within CRC cells, with the objective of identifying potential transcriptome modules encompassing certain translesion polymerases and the associated transcription factors (TFs) that govern them. Our sampling strategy involved the collection of transcriptomic data from tumors treated with cetuximab, an EGFR inhibitor, untreated CRC tumors, and colorectal-derived cell lines, resulting in a diverse dataset. Subsequently, we identified co-regulated modules using weighted correlation network analysis with a minKMEtostay threshold set at 0.5 to minimize false-positive module identifications and mapped the modules to STRING annotations. Furthermore, we explored the putative TFs influencing these modules using KBoost, a kernel PCA regression model. RESULTS: Our analysis did not reveal a distinct transcriptional profile specific to cetuximab treatment. Moreover, we elucidated co-expression modules housing genes, for example, POLK, POLI, POLQ, REV1, POLN, and POLM. Specifically, POLK, POLI, and POLQ were assigned to the "blue" module, which also encompassed critical DNA damage response enzymes, for example. BRCA1, BRCA2, MSH6, and MSH2. To delineate the transcriptional control of this module, we investigated associated TFs, highlighting the roles of prominent cancer-associated TFs, such as CENPA, HNF1A, and E2F7. CONCLUSION: We found that translesion polymerases are co-regulated with DNA mismatch repair and cell cycle-associated factors. We did not, however, identified any networks specific to cetuximab treatment indicating that the response to EGFR inhibitors relates to a general stress response mechanism.


Sujet(s)
Cétuximab , Tumeurs colorectales , Régulation de l'expression des gènes tumoraux , Cétuximab/pharmacologie , Cétuximab/usage thérapeutique , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , DNA-directed DNA polymerase/métabolisme , DNA-directed DNA polymerase/génétique , Réseaux de régulation génique , Analyse de profil d'expression de gènes , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Protéines Mad2/génétique , Protéines Mad2/métabolisme , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique
2.
Future Med Chem ; 16(13): 1313-1331, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-39109434

RÉSUMÉ

Aim: The main goal was to create two new groups of indole derivatives, hydrazine-1-carbothioamide (4a and 4b) and oxadiazole (5, and 6a-e) that target EGFR (4a, 4b, 5) or VEGFR-2 (6a-e). Materials & methods: The new derivatives were characterized using various spectroscopic techniques. Docking studies were used to investigate the binding patterns to EGFR/VEGFR-2, and the anti-proliferative properties were tested in vitro. Results: Compounds 4a (targeting EGFR) and 6c (targeting VEGFR-2) were the most effective cytotoxic agents, arresting cancer cells in the G2/M phase and inducing the extrinsic apoptosis pathway. Conclusion: The results of this study show that compounds 4a and 6c are promising cytotoxic compounds that inhibit the tyrosine kinase activity of EGFR and VEGFR-2, respectively.


[Box: see text].


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Récepteurs ErbB , Indoles , Récepteur-2 au facteur croissance endothéliale vasculaire , Humains , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Indoles/composition chimique , Indoles/pharmacologie , Simulation de docking moléculaire , Structure moléculaire , Relation structure-activité , /pharmacologie , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme
3.
BMC Oral Health ; 24(1): 898, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107736

RÉSUMÉ

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) plays an important role in the progression of multiple solid tumors and induces resistance to epidermal growth factor receptor (EGFR) target treatment. However, the expression status and the clinical significance of HER2 in oral squamous cell carcinoma (OSCC) is still controversial. Pyrotinib (PYR) is a promising novel EGFR/HER2 dual inhibitor, whose efficacy in OSCC has not been determined. METHODS: 57 locally advanced de novo OSCC patients were included in this study to investigate the relationship between the HER2 expression levels and the prognosis by the tissue microarray analysis (TMA). In vitro and in vivo experiments were performed to retrieve the efficacy of PYR in OSCC. The main downstream of HER2 was evaluated by western blotting in OSCC cell lines and xenograft tumors to explore the potential mechanism of PYR. RESULTS: This study revealed the primary tumor of OSCC had higher HER2 expression levels. Patients with HER2 overexpression had poor overall survival (P < 0.014) and poor disease free survival (P < 0.042). In vitro, PYR suppressed the proliferation, colony formation and migration of OSCC cells. It also promoted apoptosis of OSCC cells and induced cell cycle arrest. Furthermore, PYR was able to inhibit the occurrence and development of OSCC effectively in vivo. Western blotting revealed that PYR suppressed OSCC by inhibiting the phosphorylation of HER2, AKT and ERK. CONCLUSIONS: This study exhibited the anti-OSCC effects of PYR in vitro and in vivo, and demonstrated PYR inhibited OSCC cells by inducing apoptosis via the HER2/ AKT and ERK pathway. The result of this study also indicated locally advanced OSCC patients might benefit from HER2 assay and EGFR/HER2 dual inhibit treatment.


Sujet(s)
Carcinome épidermoïde , Récepteurs ErbB , Tumeurs de la bouche , Récepteur ErbB-2 , Humains , Tumeurs de la bouche/traitement médicamenteux , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/métabolisme , Récepteur ErbB-2/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme , Animaux , Lignée cellulaire tumorale , Femelle , Souris , Mâle , Apoptose/effets des médicaments et des substances chimiques , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Adulte d'âge moyen , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Aminoquinoléines
4.
Nat Commun ; 15(1): 6947, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138174

RÉSUMÉ

Fluxes in human copper levels recently garnered attention for roles in cellular signaling, including affecting levels of the signaling molecule cyclic adenosine monophosphate. We herein apply an unbiased temporal evaluation of the signaling and whole genome transcriptional activities modulated by copper level fluctuations to identify potential copper sensor proteins responsible for driving these activities. We find that fluctuations in physiologically relevant copper levels modulate EGFR signal transduction and activation of the transcription factor CREB. Both intracellular and extracellular assays support Cu1+ inhibition of the EGFR phosphatase PTPN2 (and potentially PTPN1)-via ligation to the PTPN2 active site cysteine side chain-as the underlying mechanism. We additionally show i) copper supplementation drives weak transcriptional repression of the copper importer CTR1 and ii) CREB activity is inversely correlated with CTR1 expression. In summary, our study reveals PTPN2 as a physiological copper sensor and defines a regulatory mechanism linking feedback control of copper stimulated EGFR/CREB signaling and CTR1 expression.


Sujet(s)
Transporteur-1 du cuivre , Cuivre , Protéine de liaison à l'élément de réponse à l'AMP cyclique , Récepteurs ErbB , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Transduction du signal , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Cuivre/métabolisme , Humains , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Transporteur-1 du cuivre/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 2/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 2/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 1/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 1/génétique , Transcription génétique/effets des médicaments et des substances chimiques
5.
Chem Biol Drug Des ; 104(2): e14602, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39134897

RÉSUMÉ

Catalpol, a natural iridoid glycoside, has potential therapeutic benefits, including anti-inflammatory and neuroprotective effects. Investigating catalpol's role in angiogenesis is critical for understanding its potential therapeutic applications, particularly in diseases where modulating angiogenesis is beneficial. This study investigates catalpol's influence on angiogenesis and its mechanisms, combining network pharmacology and in vitro experiments. The target genes corresponding to the catalpol were analyzed by SwissTargetPrediction. Then angiogenesis-related targets were acquired from databases like GeneCards. Subsequently, the Database for Annotation, Visualization and Integrated Discovery was employed for Gene Ontology and pathway analysis, while Cytoscape visualized protein interactions. The effect of catalpol on viability and angiogenesis of HUVECs was further examined using Cell Counting Kit-8 and angiogenesis assays. RT-qPCR and western blot were applied to check the expression of angiogenesis-related proteins. Totally, 312 target genes of catalpol and 823 angiogenesis-related targets were obtained with 56 common targets leading to PPI network analysis, highlighting hub genes (AKT1, EGFR, STAT3, MAPK3, and CASP3). These hub genes were mainly enriched in lipid and atherosclerosis pathway and EGFR-related pathway. The in vitro experimental results showed that catalpol achieved a concentration-dependent increase in HUVECs viability. Catalpol also promoted the migration and angiogenesis of HUVECs and up-regulated the expression of EGFR. EGFR knockdown inhibited the effect of catalpol on HUVECs. Catalpol promotes angiogenesis in HUVECs by upregulating EGFR and angiogenesis-related proteins, indicating its potential therapeutic application in vascular-related diseases.


Sujet(s)
Cellules endothéliales de la veine ombilicale humaine , Glucosides d'iridoïdes , Pharmacologie des réseaux , Humains , Glucosides d'iridoïdes/pharmacologie , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques ,
6.
Cell Commun Signal ; 22(1): 392, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118068

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) is a cellular process in embryonic development, wound healing, organ fibrosis, and cancer metastasis. Previously, we and others have reported that proinflammatory cytokine interleukin-1ß (IL-1ß) induces EMT. However, the exact mechanisms, especially the signal transduction pathways, underlying IL-1ß-mediated EMT are not yet completely understood. Here, we found that IL-1ß stimulation leads to the partial EMT-like phenotype in human lung epithelial A549 cells, including the gain of mesenchymal marker (vimentin) and high migratory potential, without the complete loss of epithelial marker (E-cadherin). IL-1ß-mediated partial EMT induction was repressed by PI3K inhibitor LY294002, indicating that the PI3K/AKT pathway plays a significant role in the induction. In addition, ERK1/2 inhibitor FR180204 markedly inhibited the IL-1ß-mediated partial EMT induction, demonstrating that the MEK/ERK pathway was also involved in the induction. Furthermore, we found that the activation of the PI3K/AKT and MEK/ERK pathways occurred downstream of the epidermal growth factor receptor (EGFR) pathway and the IL-1 receptor (IL-1R) pathway, respectively. Our findings suggest that the PI3K/AKT and MEK/ERK pathways coordinately promote the IL-1ß-mediated partial EMT induction. The inhibition of not one but both pathways is expected yield clinical benefits by preventing partial EMT-related disorders such as organ fibrosis and cancer metastasis.


Sujet(s)
Transition épithélio-mésenchymateuse , Interleukine-1 bêta , Système de signalisation des MAP kinases , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Humains , Interleukine-1 bêta/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Cellules A549 , Récepteurs ErbB/métabolisme
7.
Signal Transduct Target Ther ; 9(1): 216, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39143065

RÉSUMÉ

Third-generation EGFR tyrosine kinase inhibitors (TKIs), exemplified by osimertinib, have demonstrated promising clinical efficacy in the treatment of non-small cell lung cancer (NSCLC). Our previous work has identified ASK120067 as a novel third-generation EGFR TKI with remarkable antitumor effects that has undergone New Drug Application (NDA) submission in China. Despite substantial progress, acquired resistance to EGFR-TKIs remains a significant challenge, impeding the long-term effectiveness of therapeutic approaches. In this study, we conducted a comprehensive investigation utilizing high-throughput proteomics analysis on established TKI-resistant tumor models, and found a notable upregulation of branched-chain amino acid transaminase 1 (BCAT1) expression in both osimertinib- and ASK120067-resistant tumors compared with the parental TKI-sensitive NSCLC tumors. Genetic depletion or pharmacological inhibition of BCAT1 impaired the growth of resistant cells and partially re-sensitized tumor cells to EGFR TKIs. Mechanistically, upregulated BCAT1 in resistant cells reprogrammed branched-chain amino acid (BCAA) metabolism and promoted alpha ketoglutarate (α-KG)-dependent demethylation of lysine 27 on histone H3 (H3K27) and subsequent transcriptional derepression of glycolysis-related genes, thereby enhancing glycolysis and promoting tumor progression. Moreover, we identified WQQ-345 as a novel BCAT1 inhibitor exhibiting antitumor activity both in vitro and in vivo against TKI-resistant lung cancer with high BCAT1 expression. In summary, our study highlighted the crucial role of BCAT1 in mediating resistance to third-generation EGFR-TKIs through epigenetic activation of glycolysis in NSCLC, thereby supporting BCAT1 as a promising therapeutic target for the treatment of TKI-resistant NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Épigenèse génétique , Récepteurs ErbB , Glycolyse , Tumeurs du poumon , Inhibiteurs de protéines kinases , Transaminases , Humains , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Transaminases/génétique , Transaminases/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Épigenèse génétique/effets des médicaments et des substances chimiques , Épigenèse génétique/génétique , Souris , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Acrylamides/pharmacologie , Animaux , Dérivés de l'aniline/pharmacologie , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Indoles , Pyrimidines
8.
Sci Rep ; 14(1): 18366, 2024 08 07.
Article de Anglais | MEDLINE | ID: mdl-39112565

RÉSUMÉ

EGFR mutations are critical oncogenic drivers in lung adenocarcinoma (LUAD). However, the mechanisms by which they impact the tumor microenvironment (TME) and tumor immunity are unclear. Furthermore, the reasons underlying the poor response of EGFR-mutant (EGFR-MU) LUADs to immunotherapy with PD-1/PD-L1 inhibitors are unknown. Utilizing single-cell RNA (sc-RNA) and bulk RNA sequencing datasets, we conducted high-dimensional weighted gene coexpression network analysis to identify key genes and immune-related pathways contributing to the immunosuppressive TME. EGFR-MU cancer cells downregulated MHC class I genes to evade CD8+ cytotoxic T cells, expressed substantial levels of MHC class II molecules, and engaged with CD4+ regulatory T cells (Tregs). EGFR-MU tumors may recruit Tregs primarily through the CCL17/CCL22/CCR4 axis, leading to a Treg-enriched TME. High levels of MHC class II-positive cancer-associated fibroblasts and tumor endothelial cells were found within EGFR-MU tumors. Owing to the absence of costimulatory factors, they may inhibit rather than activate the tumor antigen-specific CD4+ T-cell response, contributing further to immune suppression. Multiplex immunohistochemistry analyses in a LUAD cohort confirmed increased expression of MHC class II molecules in cancer cells and fibroblasts in EGFR-MU tumors. Our research elucidates the highly immunosuppressive TME in EGFR-MU LUAD and suggests potential targets for effective immunotherapy.


Sujet(s)
Récepteurs ErbB , Analyse de profil d'expression de gènes , Tumeurs du poumon , Mutation , Microenvironnement tumoral , Humains , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Microenvironnement tumoral/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Régulation de l'expression des gènes tumoraux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/anatomopathologie , Transcriptome , Analyse sur cellule unique
9.
Eur J Med Chem ; 276: 116727, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-39094428

RÉSUMÉ

A novel series of substituted thiazolo[5,4-b]pyridine analogues were rationally designed and synthesized via a multi-step synthetic pathway, including Suzuki cross-coupling reaction. The anticancer activity of all forty-five synthesized derivatives was evaluated against HCC827, H1975, and A549 cancer cell lines utilizing the standard MTT assay. A significant number of the thiazolo[5,4-b]pyridine derivatives exhibited potent anticancer activity. Notably, compounds 10b, 10c, 10h, 10i, and 10k emerged as the most promising anticancer agents. The lead compound, N-(3-(6-(2-aminopyrimidin-5-yl)thiazolo[5,4-b]pyridin-2-yl)-2-methylphenyl)-2,5-difluorobenzenesulfonamide (10k), displayed remarkable potency with IC50 values of 0.010 µM, 0.08 µM, and 0.82 µM against the HCC827, NCI-H1975 and A-549 cancer cell lines, respectively, which were comparable to the clinically approved drug Osimertinib. Importantly, the potent derivatives 10b, 10c, 10h, 10i, and 10k exhibited selective cytotoxicity towards cancer cells and showing no toxicity against the normal BEAS-2B cell line at concentrations exceeding 35 µM. Mechanistic studies revealed that the active compound 10k acts as an EGFR-TK autophosphorylation inhibitor in HCC827 cells. Furthermore, apoptosis assays demonstrated that compound 10k induced substantial early apoptosis (31.9 %) and late apoptosis (8.8 %) in cancer cells, in contrast to the control condition exhibiting only 2.0 % early and 1.6 % late apoptosis. Molecular docking simulations of the synthesized compounds revealed that they formed essential hinge interactions and established hydrogen bonding with Cys797, indicating potential target engagement. These findings highlight the potential of the synthesized thiazolo [(Woodburn, 1999; Zigrossi et al., 2022) 5,45,4-b]pyridine derivatives as promising anticancer agents, warranting further investigation for the development of novel targeted therapies against non-small cell lung cancer.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Conception de médicament , Tests de criblage d'agents antitumoraux , Récepteurs ErbB , Tumeurs du poumon , Mutation , Inhibiteurs de protéines kinases , Pyridines , Humains , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Relation structure-activité , Pyridines/pharmacologie , Pyridines/composition chimique , Pyridines/synthèse chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Structure moléculaire , Thiazoles/pharmacologie , Thiazoles/composition chimique , Thiazoles/synthèse chimique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Simulation de docking moléculaire
10.
Cell Death Dis ; 15(8): 555, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090096

RÉSUMÉ

Resistance to epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) is a significant cause of treatment failure and cancer recurrence in non-small cell lung cancer (NSCLC). Approximately 30% of patients with EGFR-activating mutations exhibit primary resistance to EGFR-TKIs. However, the potential mechanisms of primary resistance to EGFR-TKIs remain poorly understood. Recent studies have shown that increased expression of programmed death ligand-1 (PD-L1) is associated with EGFR-TKIs resistance. Therefore, the present study aimed to investigate the mechanism of PD-L1 in primary resistance to EGFR-TKIs in EGFR-mutant lung adenocarcinoma (LUAD) cells. We found that PD-L1 was associated with poor prognosis in patients with EGFR-mutant LUAD, while the combination of EGFR-TKIs with chemotherapy could improve its therapeutic efficacy. In vitro and in vivo experiments revealed that PD-L1 promoted the proliferation and autophagy and inhibited the apoptosis of LUAD cells. Mechanistic studies demonstrated that upregulation of PD-L1 was critical in inducing autophagy through the mitogen-activated protein kinase (MAPK) signaling pathway, which was beneficial for tumor progression and the development of gefitinib resistance. Furthermore, we found that gefitinib combined with pemetrexed could synergistically enhance antitumor efficacy in PD-L1-overexpression LUAD cells. Overall, our study demonstrated that PD-L1 contributed to primary resistance to EGFR-TKIs in EGFR-mutant LUAD cells, which may be mediated by inducing autophagy via the MAPK signaling pathway. These findings not only help improve the prognosis of patients with EGFR-mutant LUAD but also provide a reference for the research of other cancer types.


Sujet(s)
Adénocarcinome pulmonaire , Autophagie , Antigène CD274 , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Tumeurs du poumon , Système de signalisation des MAP kinases , Mutation , Inhibiteurs de protéines kinases , Humains , Autophagie/effets des médicaments et des substances chimiques , Autophagie/génétique , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Antigène CD274/métabolisme , Antigène CD274/génétique , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Animaux , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mutation/génétique , Souris , Souris nude , Femelle , Mâle , Géfitinib/pharmacologie , Géfitinib/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Souris de lignée BALB C
11.
PLoS Pathog ; 20(8): e1012437, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39102432

RÉSUMÉ

The ability of Staphylococcus aureus (S. aureus) to survive within macrophages is a critical strategy for immune evasion, contributing to the pathogenesis and progression of osteomyelitis. However, the underlying mechanisms remain poorly characterized. This study discovered that inhibiting the MEK1/2 pathway reduced bacterial load and mitigated bone destruction in a mouse model of S. aureus osteomyelitis. Histological staining revealed increased phosphorylated MEK1/2 levels in bone marrow macrophages surrounding abscess in the mouse model of S. aureus osteomyelitis. Activation of MEK1/2 pathway and its roles in impairing macrophage bactericidal function were confirmed in primary mouse bone marrow-derived macrophages (BMDMs). Transcriptome analysis and in vitro experiments demonstrated that S. aureus activates the MEK1/2 pathway through EGFR signaling. Moreover, we found that excessive activation of EGFR-MEK1/2 cascade downregulates mitochondrial reactive oxygen species (mtROS) levels by suppressing Chek2 expression, thereby impairing macrophage bactericidal function. Furthermore, pharmacological inhibition of EGFR signaling prevented upregulation of phosphorylated MEK1/2 and restored Chek2 expression in macrophages, significantly enhancing S. aureus clearance and improving bone microstructure in vivo. These findings highlight the critical role of the EGFR-MEK1/2 cascade in host immune defense against S. aureus, suggesting that S. aureus may reduce mtROS levels by overactivating the EGFR-MEK1/2 cascade, thereby suppressing macrophage bactericidal function. Therefore, combining EGFR-MEK1/2 pathway blockade with antibiotics could represent an effective therapeutic approach for the treatment of S. aureus osteomyelitis.


Sujet(s)
Récepteurs ErbB , MAP Kinase Kinase 1 , Macrophages , Ostéomyélite , Infections à staphylocoques , Staphylococcus aureus , Animaux , Ostéomyélite/microbiologie , Ostéomyélite/immunologie , Ostéomyélite/métabolisme , Infections à staphylocoques/immunologie , Infections à staphylocoques/métabolisme , Infections à staphylocoques/microbiologie , Souris , Staphylococcus aureus/immunologie , Récepteurs ErbB/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/microbiologie , MAP Kinase Kinase 1/métabolisme , MAP Kinase Kinase 2/métabolisme , Système de signalisation des MAP kinases/physiologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Transduction du signal
12.
Cell Death Dis ; 15(8): 580, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122703

RÉSUMÉ

In EGFR-mutated lung cancer, the duration of response to tyrosine kinase inhibitors (TKIs) is limited by the development of acquired drug resistance. Despite the crucial role played by apoptosis-related genes in tumor cell survival, how their expression changes as resistance to EGFR-TKIs emerges remains unclear. Here, we conduct a comprehensive analysis of apoptosis-related genes, including BCL-2 and IAP family members, using single-cell RNA sequence (scRNA-seq) and spatial transcriptomics (ST). scRNA-seq of EGFR-mutated lung cancer cell lines captures changes in apoptosis-related gene expression following EGFR-TKI treatment, most notably BCL2L1 upregulation. scRNA-seq of EGFR-mutated lung cancer patient samples also reveals high BCL2L1 expression, specifically in tumor cells, while MCL1 expression is lower in tumors compared to non-tumor cells. ST analysis of specimens from transgenic mice with EGFR-driven lung cancer indicates spatial heterogeneity of tumors and corroborates scRNA-seq findings. Genetic ablation and pharmacological inhibition of BCL2L1/BCL-XL overcome or delay EGFR-TKI resistance. Overall, our findings indicate that BCL2L1/BCL-XL expression is important for tumor cell survival as EGFR-TKI resistance emerges.


Sujet(s)
Apoptose , Récepteurs ErbB , Tumeurs du poumon , Mutation , Analyse sur cellule unique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/traitement médicamenteux , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Humains , Apoptose/génétique , Apoptose/effets des médicaments et des substances chimiques , Animaux , Souris , Mutation/génétique , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , RNA-Seq , Transcriptome/génétique , Résistance aux médicaments antinéoplasiques/génétique , Inhibiteurs de protéines kinases/pharmacologie , Souris transgéniques , Analyse de profil d'expression de gènes , Protéine bcl-X/métabolisme , Protéine bcl-X/génétique , Analyse de l'expression du gène de la cellule unique
13.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39126006

RÉSUMÉ

The use of tyrosine kinase inhibitors (TKI) has been growing in veterinary oncology and in the past few years several TKI have been tested in dogs. However, different from human medicine, we lack strategies to select patients to be treated with each TKI. Therefore, this study aimed to screen different tumor subtypes regarding TKI target immunoexpression as a predictor strategy to personalize the canine cancer treatment. It included 18 prostatic carcinomas, 36 soft tissue sarcomas, 20 mammary gland tumors, 6 urothelial bladder carcinomas, and 7 tumors from the endocrine system. A total of 87 patients with paraffin blocks were used to perform immunohistochemistry (IHC) of human epidermal growth factor receptor 2 (HER-2), epidermal growth factor receptors 1 (EGFR1), vascular endothelial growth factor receptor 2 (VEGFR-2), platelet derived growth factor receptor beta (PDGFR-ß), c-KIT, and extracellular signal-regulated kinase 1/2 (ERK1/ERK2). The immunohistochemical screening revealed a heterogeneous protein expression among histological types with mesenchymal tumors showing the lowest expression level and carcinomas the highest expression. We have demonstrated by IHC screening that HER2, EGFR1, VEGFR-2, PDGFR-ß and ERK1/ERK2 are commonly overexpressed in dogs with different carcinomas, and KIT expression is considered relatively low in the analyzed samples.


Sujet(s)
Maladies des chiens , Immunohistochimie , Chiens , Animaux , Maladies des chiens/métabolisme , Maladies des chiens/traitement médicamenteux , Maladies des chiens/anatomopathologie , Mâle , Femelle , Tumeurs/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/médecine vétérinaire , Tumeurs/anatomopathologie , Récepteur au PDGF bêta/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Marqueurs biologiques tumoraux/métabolisme , Récepteur ErbB-2/métabolisme , Protéines proto-oncogènes c-kit/métabolisme , Récepteurs ErbB/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Humains
14.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39126121

RÉSUMÉ

The burgeoning field of cancer theranostics has witnessed advancements through the development of targeted molecular agents, particularly peptides. These agents exploit the overexpression or mutations of specific receptors, such as the Epidermal Growth Factor receptor (EGFR) and αVß3 integrin, which are pivotal in tumor growth, angiogenesis, and metastasis. Despite the extensive research into and promising outcomes associated with antibody-based therapies, peptides offer a compelling alternative due to their smaller size, ease of modification, and rapid bioavailability, factors which potentially enhance tumor penetration and reduce systemic toxicity. However, the application of peptides in clinical settings has challenges. Their lower binding affinity and rapid clearance from the bloodstream compared to antibodies often limit their therapeutic efficacy and diagnostic accuracy. This overview sets the stage for a comprehensive review of the current research landscape as it relates to EGFR- and integrin αVß3-targeting peptides. We aim to delve into their synthesis, radiolabeling techniques, and preclinical and clinical evaluations, highlighting their potential and limitations in cancer theranostics. This review not only synthesizes the extant literature to outline the advancements in peptide-based agents targeting EGFR and integrin αVß3 but also identifies critical gaps that could inform future research directions. By addressing these gaps, we contribute to the broader discourse on enhancing the diagnostic precision and therapeutic outcomes of cancer treatments.


Sujet(s)
Récepteurs ErbB , Intégrine alphaVbêta3 , Tumeurs , Peptides , Radiopharmaceutiques , Humains , Intégrine alphaVbêta3/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Radiopharmaceutiques/usage thérapeutique , Radiopharmaceutiques/composition chimique , Tumeurs/imagerie diagnostique , Tumeurs/thérapie , Tumeurs/métabolisme , Tumeurs/traitement médicamenteux , Peptides/composition chimique , Peptides/usage thérapeutique , Animaux , Médecine de précision/méthodes , Nanomédecine théranostique/méthodes
15.
Proc Natl Acad Sci U S A ; 121(34): e2405959121, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39141345

RÉSUMÉ

TORC1 (target of rapamycin complex 1) is a highly conserved protein kinase that plays a central role in regulating cell growth. Given the role of mammalian TORC1 (mTORC1) in metabolism and disease, understanding mTORC1 downstream signaling and feedback loops is important. mTORC1 recognizes some of its substrates via a five amino acid binding sequence called the TOR signaling (TOS) motif. mTORC1 binding to a TOS motif facilitates phosphorylation of a distinct, distal site. Here, we show that LST2, also known as ZFYVE28, contains a TOS motif (amino acids 401 to 405) and is directly phosphorylated by mTORC1 at serine 670 (S670). mTORC1-mediated S670 phosphorylation promotes LST2 monoubiquitination on lysine 87 (K87). Monoubiquitinated LST2 is stable and displays a broad reticular distribution. When mTORC1 is inactive, unphosphorylated LST2 is degraded by the proteasome. The absence of LST2 enhances EGFR (epidermal growth factor receptor) signaling. We propose that mTORC1 negatively feeds back on its upstream receptor EGFR via LST2.


Sujet(s)
Récepteurs ErbB , Complexe-1 cible mécanistique de la rapamycine , Transduction du signal , Ubiquitination , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Phosphorylation , Humains , Récepteurs ErbB/métabolisme , Cellules HEK293 , Animaux , Motifs d'acides aminés
16.
Drug Des Devel Ther ; 18: 2571-2591, 2024.
Article de Anglais | MEDLINE | ID: mdl-38947223

RÉSUMÉ

Purpose: Activating mutations in epidermal growth factor receptor (EGFR) have been identified as key predictive biomarkers for the customized treatment with EGFR tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer (NSCLC), aiding in improving patient response rates and survival. However, resistance challenges the efficacy of these treatments, with limited understanding of post-resistance therapeutic strategies. A deep understanding of the biology and resistance mechanisms of EGFR-mutant NSCLC is crucial for developing new treatment approaches. This study, through bibliometric analysis, summarizes the trends in research on resistance to EGFR-TKIs. Methods: Research papers on NSCLC with EGFR inhibitor resistance were collected from the Web of Science Core Collection (WoSCC). The analysis utilized bibliometric tools like CiteSpace, VOSviewer, and other platforms for comprehensive analysis and visualization of the outcomes. Results: The WoSCC database contains a total of 5866 documents on resistance to EGFR-TKIs treatment, including 4727 articles (93.48%) and 1139 reviews (6.52%), spanning 81 countries and regions, 4792 institutions, with the involvement of 23,594 authors. Since 2016, there has been a significant increase in publications in this field. China has the highest publication output, while the United States has the highest citation count for papers. Harvard University leads in terms of the number of publications. Among the top ten journals with the highest output, Clinical Cancer Research has the highest impact factor at 11.5, with 90% of the journals classified in Q1 or Q2. Rafael Rosell is one of the most influential authors in this field, ranking second in publication volume and fourth in citation count. Research on EGFR-TKIs resistance mainly focuses on genetic testing, resistance mechanisms, and post-resistance treatment strategies. Conclusion: This study provides researchers with a reliable basis and guidance for finding authoritative references, understanding research trends, and exploring potential directions.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Tumeurs du poumon , Inhibiteurs de protéines kinases , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Bibliométrie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique
17.
FASEB J ; 38(13): e23759, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38949635

RÉSUMÉ

The epidermal growth factor receptor (EGFR) is an important target for cancer therapies. Many head and neck cancer (HNC) cells have been reported to overexpress EGFR; therefore, anti-EGFR therapies have been attempted in patients with HNC. However, its clinical efficacy is limited owing to the development of drug resistance. In this study, we developed an EGFR-targeting immunotoxin consisting of a clinically proven anti-EGFR IgG (cetuximab; CTX) and a toxin fragment (LR-LO10) derived from Pseudomonas exotoxin A (PE) using a novel site-specific conjugation technology (peptide-directed photo-crosslinking reaction), as an alternative option. The immunotoxin (CTX-LR-LO10) showed specific binding to EGFR and properties of a typical IgG, such as stability, interactions with receptors of immune cells, and pharmacokinetics, and inhibited protein synthesis via modification of elongation factor-2. Treatment of EGFR-positive HNC cells with the immunotoxin resulted in apoptotic cell death and the inhibition of cell migration and invasion. The efficacy of CTX-LR-LO10 was evaluated in xenograft mouse models, and the immunotoxin exhibited much stronger tumor suppression than CTX or LR-LO10. Transcriptome analyses revealed that the immunotoxins elicited immune responses and altered the expression of genes related to its mechanisms of action. These results support the notion that CTX-LR-LO10 may serve as a new therapeutic agent targeting EGFR-positive cancers.


Sujet(s)
ADP ribose transferases , Récepteurs ErbB , Exotoxines , Tumeurs de la tête et du cou , Immunoglobuline G , Immunotoxines , , Facteurs de virulence , Humains , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Récepteurs ErbB/immunologie , Animaux , Immunotoxines/pharmacologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/métabolisme , Souris , Immunoglobuline G/pharmacologie , Lignée cellulaire tumorale , Exotoxines/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Cétuximab/pharmacologie , Souris nude , Toxines bactériennes , Apoptose/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Femelle , Mouvement cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie
18.
Commun Biol ; 7(1): 845, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987622

RÉSUMÉ

Adult Neural Stem Cells (aNSCs) in the ventricular-subventricular zone (V-SVZ) are largely quiescent. Here, we characterize the mechanism underlying the functional role of a cell-signalling inhibitory protein, LRIG1, in the control of aNSCs proliferation. Using Lrig1 knockout models, we show that Lrig1 ablation results in increased aNSCs proliferation with no change in neuronal progeny and that this hyperproliferation likely does not result solely from activation of the epidermal growth factor receptor (EGFR). Loss of LRIG1, however, also leads to impaired activation of transforming growth factor beta (TGFß) and bone morphogenic protein (BMP) signalling. Biochemically, we show that LRIG1 binds TGFß/BMP receptors and the TGFß1 ligand. Finally, we show that the consequences of these interactions are to facilitate SMAD phosphorylation. Collectively, these data suggest that unlike in embryonic NSCs where EGFR may be the primary mechanism of action, in aNSCs, LRIG1 and TGFß pathways function together to fulfill their inhibitory roles.


Sujet(s)
Protéines morphogénétiques osseuses , Prolifération cellulaire , Glycoprotéines membranaires , Cellules souches neurales , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Facteur de croissance transformant bêta/métabolisme , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Souris , Protéines morphogénétiques osseuses/métabolisme , Souris knockout , Cellules souches adultes/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Protéines de tissu nerveux
19.
Science ; 385(6705): 194-200, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38991070

RÉSUMÉ

Millions of hibernating bats across North America have died from white-nose syndrome (WNS), an emerging disease caused by a psychrophilic (cold-loving) fungus, Pseudogymnoascus destructans, that invades their skin. Mechanisms of P. destructans invasion of bat epidermis remain obscure. Guided by our in vivo observations, we modeled hibernation with a newly generated little brown bat (Myotis lucifugus) keratinocyte cell line. We uncovered the stealth intracellular lifestyle of P. destructans, which inhibits apoptosis of keratinocytes and spreads through the cells by two epidermal growth factor receptor (EGFR)-dependent mechanisms: active penetration during torpor and induced endocytosis during arousal. Melanin of endocytosed P. destructans blocks endolysosomal maturation, facilitating P. destructans survival and germination after return to torpor. Blockade of EGFR aborts P. destructans entry into keratinocytes.


Sujet(s)
Éveil , Ascomycota , Chiroptera , Récepteurs ErbB , Hibernation , Kératinocytes , Animaux , Apoptose , Ascomycota/physiologie , Ascomycota/pathogénicité , Lignée cellulaire , Chiroptera/microbiologie , Chiroptera/physiologie , Endocytose , Récepteurs ErbB/métabolisme , Kératinocytes/microbiologie , Mélanines/métabolisme
20.
Biol Pharm Bull ; 47(7): 1288-1295, 2024.
Article de Anglais | MEDLINE | ID: mdl-39010214

RÉSUMÉ

The active form of discoidin domain receptors (DDRs) is expressed in cell surface and regulated post-translationally by glucose. The DDR2 and DDR1 transfected in HEK293 cells were expressed mainly in their active forms with sizes of 130 and 120 kDa, respectively. DDRs were observed predominantly as 100 kDa proteins in glucose-depleted culture conditions. However, transfection of endothelial growth factor receptor (EGFR) in HEK293 cells resulted in the expression of only one form regardless of glucose concentration. Vascular smooth muscle cells, HT1080s, and MDA-MB-231 cancer cells expressed DDRs in their active forms in high glucose concentrations, which did not occur with EGFR. In diabetic rats, DDRs were expressed at high levels in arterial tissue but EGFR was not highly expressed. Taken together, these results suggest that DDRs expression depends on glucose concentration it may cooperate in the development of atherosclerosis and kidney fibroblasts, promoting nephropathy in diabetic rats.


Sujet(s)
Diabète expérimental , Diabète de type 2 , Glucose , Animaux , Humains , Glucose/métabolisme , Diabète de type 2/métabolisme , Diabète de type 2/génétique , Mâle , Diabète expérimental/métabolisme , Cellules HEK293 , Rats , Artères/métabolisme , Artères/anatomopathologie , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Lignée cellulaire tumorale , Récepteur-2 à domaine discoïdine/métabolisme , Récepteur-2 à domaine discoïdine/génétique , Muscles lisses vasculaires/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/génétique , Rat Wistar
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE