Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.137
Filtrer
1.
Oncol Rep ; 52(4)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39092576

RÉSUMÉ

Senescent cells are known to secrete proteins, including inflammatory cytokines and damage­associated molecular patterns. This phenomenon is known as the senescence­associated secretory phenotype (SASP). SASP in cancer stromal fibroblasts is involved in cancer growth and progression. Conversely, metformin, an antidiabetic drug, has been reported to inhibit SASP induction by inhibiting the activation of NF­κB, a regulator of SASP. To date, at least to the best of our knowledge, there have been no reports regarding cellular senescence in fibroblasts and tumor progression via the SASP­mediated paracrine pathway. The present study thus aimed to elucidate the induction mechanisms of SASP in radiation­induced fibroblasts and to determine its effects on cancer progression via the paracrine pathway. Furthermore, the present study aimed to determine whether controlling SASP using metformin suppresses cancer progression. A well­differentiated esophageal cancer cell line established by the authors' department and fibroblasts isolated and cultured from the non­cancerous esophageal mucosa of resected esophageal cancer cases were used for the experiments. Fibroblasts were irradiated with 8 Gy radiation, and the changes in the expression of the senescence markers, SA­ß­gal, p21, p16 and NF­κB were evaluated using immunofluorescent staining and western blot analysis in the presence or absence of metformin treatment. The culture supernatants of irradiated fibroblasts treated with metformin and those treated without metformin were collected and added to the cancer cells to evaluate their proliferative, invasive and migratory abilities. Vimentin and E­cadherin expression levels were also evaluated using immunofluorescent staining and western blot analysis. The expression levels of p16, p21 and NF­κB in irradiated fibroblasts were attenuated by treatment with metformin. Supernatants collected from irradiated fibroblasts exhibited the proliferative activity of esophageal cancer cells, and the promotion of migratory and invasion abilities, which may be due to epithelial­mesenchymal transition and changes in cell morphology. These reactions were confirmed to be suppressed by the addition of the supernatant of cultured fibroblasts pre­treated with metformin. On the whole, the present study demonstrates that fibroblasts in the cancer stroma may be involved in tumor progression through cellular senescence.


Sujet(s)
Fibroblastes associés au cancer , Prolifération cellulaire , Vieillissement de la cellule , Tumeurs de l'oesophage , Metformine , Metformine/pharmacologie , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des radiations , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/traitement médicamenteux , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des radiations , Fibroblastes associés au cancer/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Évolution de la maladie , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire tumorale , Phénotype sécrétoire associé à la sénescence , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des radiations , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des radiations , Hypoglycémiants/pharmacologie , Fibroblastes/métabolisme , Fibroblastes/effets des radiations , Fibroblastes/effets des médicaments et des substances chimiques
2.
FASEB J ; 38(15): e23846, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39093041

RÉSUMÉ

Epithelial cells play a crucial role in asthma, contributing to chronic inflammation and airway hyperresponsiveness. m6A modification, which involves key proteins such as the demethylase fat mass and obesity-associated protein (FTO), is crucial in the regulation of various diseases, including asthma. However, the role of FTO in epithelial cells and the development of asthma remains unclear. In this study, we investigated the demethylase activity of FTO using a small-molecule inhibitor FB23 in epithelial cells and allergic inflammation in vivo and in vitro. We examined the FTO-regulated transcriptome-wide m6A profiling by methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA-seq under FB23 treatment and allergic inflammation conditions. Immunofluorescence staining was performed to assess the tissue-specific expression of FTO in asthmatic bronchial mucosa. We demonstrated that FB23 alleviated allergic inflammation in IL-4/IL-13-treated epithelial cells and house dust mite (HDM)-induced allergic airway inflammation mouse model. The demethylase activity of FTO contributed to the regulation of TNF-α signaling via NF-κB and epithelial-mesenchymal transition-related pathways under allergic inflammation conditions in epithelial cells. FTO was expressed in epithelial, submucosal gland, and smooth muscle cells in human bronchial mucosa. In conclusion, FB23-induced inhibition of FTO alleviates allergic inflammation in epithelial cells and HDM-induced mice, potentially through diverse cellular processes and epithelial-mesenchymal transition signaling pathways, suggesting that FTO is a potential therapeutic target in asthma management.


Sujet(s)
Alpha-ketoglutarate-dependent dioxygenase FTO , Asthme , Inflammation , Animaux , Alpha-ketoglutarate-dependent dioxygenase FTO/métabolisme , Alpha-ketoglutarate-dependent dioxygenase FTO/génétique , Souris , Asthme/métabolisme , Asthme/génétique , Inflammation/métabolisme , Humains , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , Cellules épithéliales/métabolisme , Souris de lignée BALB C , Femelle , Hypersensibilité/métabolisme , Hypersensibilité/traitement médicamenteux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Souris de lignée C57BL
3.
Sci Rep ; 14(1): 17812, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090292

RÉSUMÉ

Chemotherapy, particularly with oxaliplatin, is a key treatment for advanced gastric cancer (GC), and exosomes derived from human bone marrow mesenchymal stem cells (hBM-MSCs) play a vital role in the tumor microenvironment. The study aims to elucidate the previously unexplored role of exosomes derived from hBM-MSCs in GC tumorigenesis, especially under the influence of chemotherapy. We conducted an experimental study, utilizing miRNA sequencing and biological experiments, to analyze the tumorigenicity of exosomal miR-424-3p secreted by hBM-MSCs and its target gene RHOXF2 in GC cell lines. The results were confirmed through experimentation using a xenograft mouse model. This study demonstrated the role of hBM-MSCs in the GC microenvironment, focusing on their epithelial-mesenchymal transition (EMT) facilitation through exosomes, which led to enhanced tumorigenicity in GC cells. Intriguingly, this pro-tumor effect was abrogated when hBM-MSCs were treated with oxaliplatin. Exosomal miRNA sequencing revealed that oxaliplatin can upregulate the levels of miR-424-3p in exosomes secreted by hBM-MSCs, thereby inhibiting the EMT process in GC cells. Furthermore, miR-424-3p was identified to target and downregulate RHOXF2 expression, impeding the malignant behavior of GC cells both in vitro and in the mouse model. These findings uncover a potential hidden mechanism of oxaliplatin's anti-tumor action and propose the delivery of miR-424-3p via exosomes as a promising avenue for anti-tumor therapy.


Sujet(s)
Transition épithélio-mésenchymateuse , Exosomes , Cellules souches mésenchymateuses , microARN , Oxaliplatine , Tumeurs de l'estomac , microARN/génétique , microARN/métabolisme , Humains , Oxaliplatine/pharmacologie , Cellules souches mésenchymateuses/métabolisme , Exosomes/métabolisme , Exosomes/génétique , Animaux , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/traitement médicamenteux , Souris , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Régulation positive , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques/pharmacologie , Microenvironnement tumoral , Souris nude , Évolution de la maladie
4.
Oncol Res ; 32(8): 1359-1368, 2024.
Article de Anglais | MEDLINE | ID: mdl-39055888

RÉSUMÉ

Multiple myeloma (MM) is a plasma cell malignancy and remains incurable as it lacks effective curative approaches; thus, novel therapeutic strategies are desperately needed. The study aimed to explore the therapeutic role of dihydromyricetin (DHM) in MM and explore its mechanisms. Human MM and normal plasma samples, human MM cell lines, and normal plasma cells were used for in vitro experiments. Cell counting kit-8 (CCK-8), flow cytometry, and trans-well assays were performed for the assessment of cell viability, apoptosis, migration, and invasion, respectively. Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to assess the mRNA expression of signal transducer and activator of transcription 1 (STAT1) and retinoic acid-inducible gene I (RIG-I). Western blotting was employed to assess E-cadherin, N-cadherin, signal transducer, STAT1, p-STAT1, and RIG-I protein expression. A tumor xenograft model was used for in vivo experiments. Here, dihydromyricetin (DHM) dose-dependently restrained viability, apoptosis, migration, and invasion, and facilitated apoptosis of U266 cells. After DHM treatment, the E-cadherin level was increased and the N-cadherin level was decreased in U266 and RPMI-8226 cells, suggesting the inhibitory effects of DHM on epithelial-mesenchymal transition (EMT) in MM. Besides, the levels of p-STAT1/STAT1 and RIG-I were down-regulated in MM. However, the STAT1 inhibitor fludarabine undid the suppressive effect of DMH on the malignant characteristics of U266 cells. Also, DHM inhibited MM tumor growth and EMT, and activated STAT1/RIG-I pathway in vivo. Collectively, this study first revealed that DHM can restrain EMT and tumor growth in MM by activating STAT1/RIG-I signaling, which provides a novel drug for the treatment of MM.


Sujet(s)
Apoptose , Flavonols , Myélome multiple , Facteur de transcription STAT-1 , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Facteur de transcription STAT-1/métabolisme , Flavonols/pharmacologie , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Animaux , Souris , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Ampelopsis/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Mouvement cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Adulte d'âge moyen
5.
Bioorg Chem ; 150: 107604, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38981209

RÉSUMÉ

Nineteen flavonoids were isolated from the fruits of Psoralea corylifolia L., including a novel flavanol (3) and three novel isoflavones (12-14). Their chemical structures were unequivocally determined through comprehensive spectral data analysis. The anti-proliferative effect of the isolated flavonoids was assessed in vitro using the MTT assay. Molecular docking and ELISA were employed to determine the inhibitory effects of the active compounds on ALK5. Isobavachalcone was found to inhibit TGF-ß1 induced EMT in A549 cells by Wound healing assay and Transwell chamber assay. Immunofluorescence assay and Western blot assay showed that IBC could inhibit cytoskeleton rearrangement, reduce the phosphorylation of ALK5, ERK, and Smad, down-regulate Snail expression, and up-regulate E-cadherin expression in TGF-ß1 induced A549 cells, thereby exerting the potential inhibitory effects on epithelial-mesenchymal transition (EMT) process in A549 cells. The findings presented herein establish a fundamental basis for investigating the anti-proliferative and anti-metastatic properties of psoralen flavonoids in human non-small cell lung cancer.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Transition épithélio-mésenchymateuse , Flavonoïdes , Fruit , Tumeurs du poumon , Simulation de docking moléculaire , Psoralea , Humains , Cellules A549 , Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques d'origine végétale/composition chimique , Antinéoplasiques d'origine végétale/isolement et purification , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Flavonoïdes/isolement et purification , Fruit/composition chimique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Structure moléculaire , Psoralea/composition chimique , Récepteur de type I du facteur de croissance transformant bêta , Relation structure-activité , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/antagonistes et inhibiteurs , Isoflavones/composition chimique , Isoflavones/isolement et purification , Isoflavones/pharmacologie
6.
J Nanobiotechnology ; 22(1): 434, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39044233

RÉSUMÉ

Pulmonary Fibrosis (PF) is a fatal disease in the interstitial lung associated with high mortality, morbidity, and poor prognosis. Transforming growth factor-ß1 (TGF-ß1) is a fibroblast-activating protein that promotes fibrous diseases. Herein, an inhalable system was first developed using milk exosomes (M-Exos) encapsulating siRNA against TGF-ß1 (MsiTGF-ß1), and their therapeutic potential for bleomycin (BLM)-induced PF was investigated. M-siTGF-ß1 was introduced into the lungs of mice with PF through nebulization. The collagen penetration effect and lysosomal escape ability were verified in vitro. Inhaled MsiTGF-ß1 notably alleviated inflammatory infiltration, attenuated extracellular matrix (ECM) deposition, and increased the survival rate of PF mice by 4.7-fold. M-siTGF-ß1 protected lung tissue from BLM toxicity by efficiently delivering specific siRNA to the lungs, leading to TGF-ß1 mRNA silencing and epithelial mesenchymal transition pathway inhibition. Therefore, M-siTGF-ß1 offers a promising avenue for therapeutic intervention in fibrosis-related disorders.


Sujet(s)
Bléomycine , Collagène , Transition épithélio-mésenchymateuse , Exosomes , Poumon , Lait , Fibrose pulmonaire , Petit ARN interférent , Facteur de croissance transformant bêta-1 , Animaux , Exosomes/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Souris , Collagène/métabolisme , Bléomycine/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Lait/composition chimique , Souris de lignée C57BL , Humains , Perméabilité , Mâle , Nébuliseurs et vaporisateurs
7.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000270

RÉSUMÉ

The combination of a polyphenol, quercetin, with dasatinib initiated clinical trials to evaluate the safety and efficacy of senolytics in idiopathic pulmonary fibrosis, a lung disease associated with the presence of senescent cells. Another approach to senotherapeutics consists of controlling inflammation related to cellular senescence or "inflammaging", which participates, among other processes, in establishing pulmonary fibrosis. We evaluate whether polyphenols such as caffeic acid, chlorogenic acid, epicatechin, gallic acid, quercetin, or resveratrol combined with different senotherapeutics such as metformin or rapamycin, and antifibrotic drugs such as nintedanib or pirfenidone, could present beneficial actions in an in vitro model of senescent MRC-5 lung fibroblasts. A senescent-associated secretory phenotype (SASP) was evaluated by the measurement of interleukin (IL)-6, IL-8, and IL-1ß. The senescent-associated ß-galactosidase (SA-ß-gal) activity and cellular proliferation were assessed. Fibrosis was evaluated using a Picrosirius red assay and the gene expression of fibrosis-related genes. Epithelial-mesenchymal transition (EMT) was assayed in the A549 cell line exposed to Transforming Growth Factor (TGF)-ß in vitro. The combination that demonstrated the best results was metformin and caffeic acid, by inhibiting IL-6 and IL-8 in senescent MRC-5 cells. Metformin and caffeic acid also restore cellular proliferation and reduce SA-ß-gal activity during senescence induction. The collagen production by senescent MRC-5 cells was inhibited by epicatechin alone or combined with drugs. Epicatechin and nintedanib were able to control EMT in A549 cells. In conclusion, caffeic acid and epicatechin can potentially increase the effectiveness of senotherapeutic drugs in controlling lung diseases whose pathophysiological component is the presence of senescent cells and fibrosis.


Sujet(s)
Vieillissement de la cellule , Fibroblastes , Poumon , Polyphénols , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Polyphénols/pharmacologie , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Cellules A549 , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Metformine/pharmacologie , Acides caféiques/pharmacologie , Indoles/pharmacologie , Sénothérapie/pharmacologie , Lignée cellulaire , Phénotype sécrétoire associé à la sénescence/effets des médicaments et des substances chimiques , Sirolimus/pharmacologie , Interleukine-8/métabolisme , Interleukine-8/génétique , Facteur de croissance transformant bêta/métabolisme , Pyridones
8.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39000339

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) refers to the transformation of polar epithelial cells into motile mesenchymal cells under specific physiological or pathological conditions, thus promoting the metastasis of cancer cells. Epithelial cadherin (E-cadherin) is a protein that plays an important role in the acquisition of tumor cell motility and serves as a key EMT epithelial marker. In the present study, AW01178, a small-molecule compound with potential therapeutic efficacy, was identified via in-cell Western high-throughput screening technology using E-cadherin as the target. The compound induced the upregulation of E-cadherin at both mRNA and protein levels and inhibited the EMT of breast cancer cells in vitro as well as metastasis in vivo. Mechanistically, AW01178 is a novel benzacetamide histone deacetylase inhibitor (HDACi) mainly targeting class I histone deacetylases. AW01178 promoted the transcription and expression of E-cadherin through enhancing the acetylation level of histone H3 in the E-cadherin promoter region, thereby inhibiting the metastasis of breast cancer cells. The collective findings support the potential utility of the novel HDACi compound identified in this study, AW01178, as a therapeutic drug for breast cancer and highlight its value for the future development of HDACi structures as anticancer drugs.


Sujet(s)
Tumeurs du sein , Cadhérines , Transition épithélio-mésenchymateuse , Inhibiteurs de désacétylase d'histone , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Femelle , Animaux , Cadhérines/métabolisme , Cadhérines/génétique , Lignée cellulaire tumorale , Métastase tumorale , Souris , Mouvement cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Souris nude , Histone/métabolisme
9.
PLoS One ; 19(7): e0305612, 2024.
Article de Anglais | MEDLINE | ID: mdl-38990915

RÉSUMÉ

Breast cancer (BC) cells have a high risk of metastasis due to epithelial-mesenchymal transition (EMT). Palbociclib (CDK4/6 inhibitor) is an approved drug for BC treatment. However, the drug resistance and metastasis can impair the treatment outcome of Palbociclib. Understanding the mechanisms of EMT and Palbociclib drug resistance in BC is conducive to the formulation of novel therapeutic strategy. Here, we investigated the role of circHIAT1/miR-19a-3p/CADM2 axis in modulating EMT and Palbociclib resistance in BC. circHIAT1 and CADM2 were down-regulated in BC tissues and cell lines, and miR-19a-3p showed an up-regulation. circHIAT1 could interact with miR-19a-3p and suppress its activity, while miR-19a-3p functioned to negatively regulate CADM2. Forced over-expression of circHIAT1 could impaired the EMT status and migratory ability of BC cells, and this effect was inhibited by miR-19a-3p mimic. In addition, we also generated Palbociclib resistant BC cells, and showed that circHIAT1 and CADM2 were down-regulated in the resistant BC cells while miR-19a-3p showed an up-regulation. Forced circHIAT1 over-expression re-sensitized BC cells to Palbociclib treatment. Quercetin, a bioactive flavonoid, could suppressed the migration and invasion of BC cells, and re-sensitized BC cells to Palbociclib. The anti-cancer effect of quercetin could be attributed to its regulatory effect on circHIAT1/miR-19a-3p/CADM2 axis. In vivo tumorigenesis experiment further revealed that quercetin administration enhanced the anti-cancer effect of Palbociclib, an effect was dependent on the up-regulation of circHIAT1 by quercetin. In summary, this study identified quercetin as a potential anti-cancer compound to reverse Palbociclib resistance and impair EMT in BC cells by targeting circHIAT1/miR-19a-3p/CADM2 axis.


Sujet(s)
Tumeurs du sein , Kinase-6 cycline-dépendante , Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse , microARN , Pipérazines , Pyridines , Quercétine , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Humains , microARN/génétique , microARN/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pipérazines/pharmacologie , Lignée cellulaire tumorale , Quercétine/pharmacologie , Animaux , Souris , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Mouvement cellulaire/effets des médicaments et des substances chimiques , ARN long non codant/génétique , ARN long non codant/métabolisme , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Cells ; 13(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38994997

RÉSUMÉ

Extracellular HSP90α (eHSP90α) is a promoter of tumor development and malignant progression. Patients with malignancies, including pancreatic ductal adenocarcinoma (PDAC), have generally shown 5~10-fold increases in serum/plasma eHSP90α levels. In this study, we developed a humanized antibody HH01 to target eHSP90α and evaluated its anticancer efficacy. HH01, with novel complementarity-determining regions, exhibits high binding affinity toward HSP90α. It recognizes HSP90α epitope sites 235AEEKEDKEEE244 and 251ESEDKPEIED260, with critical amino acid residues E237, E239, D240, K241, E253, and K255. HH01 effectively suppressed eHSP90α-induced invasive and spheroid-forming activities of colorectal cancer and PDAC cell lines by blocking eHSP90α's ligation with the cell-surface receptor CD91. In mouse models, HH01 potently inhibited the tumor growth of PDAC cell grafts/xenografts promoted by endothelial-mesenchymal transition-derived cancer-associated fibroblasts while also reducing serum eHSP90α levels, reflecting its anticancer efficacy. HH01 also modulated tumor immunity by reducing M2 macrophages and reinvigorating immune T-cells. Additionally, HH01 showed low aggregation propensity, high water solubility, and a half-life time of >18 days in mouse blood. It was not cytotoxic to retinal pigmented epithelial cells and showed no obvious toxicity in mouse organs. Our data suggest that targeting eHSP90α with HH01 antibody can be a promising novel strategy for PDAC therapy.


Sujet(s)
Anticorps monoclonaux humanisés , Protéines du choc thermique HSP90 , Tumeurs du pancréas , Humains , Protéines du choc thermique HSP90/métabolisme , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Animaux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Souris , Lignée cellulaire tumorale , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Adénocarcinome/anatomopathologie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/métabolisme ,
11.
PLoS One ; 19(7): e0298353, 2024.
Article de Anglais | MEDLINE | ID: mdl-38995910

RÉSUMÉ

CONTEXT: Nephrotic syndrome(NS) has emerged as a worldwide public health problem. Renal fibrosis is the most common pathological change from NS to end-stage renal failure, seriously affecting the prognosis of renal disease. Although tremendous efforts have been made to treat NS, specific drug therapies to delay the progression of NS toward end-stage renal failure are limited. Epimedium is generally used to treat kidney disease in traditional Chinese medicine. Icariin is a principal active component of Epimedium. METHODS: We used Sprague Dawley rats to establish NS models by injecting doxorubicin through the tail vein. Then icariin and prednisone were intragastric administration. Renal function was examined by an automatic biochemical analyzer. Pathology of the kidney was detected by Hematoxylin-Eosin and Masson staining respectively. Furthermore, RT-PCR, Enzyme-Linked Immunosorbent Assay, Immunohistochemistry, Western Blot and Terminal-deoxynucleotidyl Transferase Mediated Nick End Labeling staining were employed to detect the proteins related to pyroptosis and EMT. HK-2 cells exposed to doxorubicin were treated with icariin, and cell viability was assessed using the MTT. EMT was assessed using Enzyme-Linked Immunosorbent Assay and Western Blot. RESULTS: The study showed that icariin significantly improved renal function and renal fibrosis in rats. In addition, icariin effectively decreased NOD-like receptor thermal protein domain associated protein 3,Caspase-1, Gasdermin D, Ly6C, and interleukin (IL)-1ß. Notably, treatment with icariin also inhibited the levels of TGF-ß, α-SMA and E-cadherin. DISCUSSION AND CONCLUSIONS: It is confirmed that icariin can improve renal function and alleviate renal fibrosis by inhibiting pyroptosis and the mechanism may be related to epithelial-to-mesenchymal transition. Icariin treatment might be recommended as a new approach for NS.


Sujet(s)
Doxorubicine , Transition épithélio-mésenchymateuse , Flavonoïdes , Syndrome néphrotique , Pyroptose , Rat Sprague-Dawley , Animaux , Flavonoïdes/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Pyroptose/effets des médicaments et des substances chimiques , Rats , Syndrome néphrotique/traitement médicamenteux , Syndrome néphrotique/anatomopathologie , Syndrome néphrotique/métabolisme , Mâle , Doxorubicine/pharmacologie , Humains , Fibrose/traitement médicamenteux , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Lignée cellulaire , Modèles animaux de maladie humaine
12.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38999963

RÉSUMÉ

Triple-negative breast cancer (TNBC) is a particularly aggressive mammary neoplasia with a high fatality rate, mainly because of the development of resistance to administered chemotherapy, the standard treatment for this disease. In this study, we employ both bulk RNA-sequencing and single-cell RNA-sequencing (scRNA-seq) to investigate the transcriptional landscape of TNBC cells cultured in two-dimensional monolayers or three-dimensional spheroids, before and after developing resistance to the chemotherapeutic agents paclitaxel and doxorubicin. Our findings reveal significant transcriptional heterogeneity within the TNBC cell populations, with the scRNA-seq identifying rare subsets of cells that express resistance-associated genes not detected by the bulk RNA-seq. Furthermore, we observe a partial shift towards a highly mesenchymal phenotype in chemoresistant cells, suggesting the epithelial-to-mesenchymal transition (EMT) as a prevalent mechanism of resistance in subgroups of these cells. These insights highlight potential therapeutic targets, such as the PDGF signaling pathway mediating EMT, which could be exploited in this setting. Our study underscores the importance of single-cell approaches in understanding tumor heterogeneity and developing more effective, personalized treatment strategies to overcome chemoresistance in TNBC.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Analyse sur cellule unique , Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Humains , Résistance aux médicaments antinéoplasiques/génétique , Analyse sur cellule unique/méthodes , Femelle , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes/méthodes , Paclitaxel/pharmacologie , Transcriptome , Doxorubicine/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
13.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-39000029

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge in terms of diagnosis and treatment, with limited therapeutic options and a poor prognosis. This study explored the potential therapeutic role of NPS-1034, a kinase inhibitor targeting MET and AXL, in PDAC. The investigation included monotherapy with NPS-1034 and its combination with the commonly prescribed chemotherapy agents, fluorouracil and oxaliplatin. Our study revealed that NPS-1034 induces cell death and reduces the viability and clonogenicity of PDAC cells in a dose-dependent manner. Furthermore, NPS-1034 inhibits the migration of PDAC cells by suppressing MET/PI3K/AKT axis-induced epithelial-to-mesenchymal transition (EMT). The combination of NPS-1034 with fluorouracil or oxaliplatin demonstrated a synergistic effect, significantly reducing cell viability and inducing tumor cell apoptosis compared to monotherapies. Mechanistic insights provided by next-generation sequencing indicated that NPS-1034 modulates immune responses by inducing type I interferon and tumor necrosis factor production in PDAC cells. This suggests a broader role for NPS-1034 beyond MET and AXL inhibition, positioning it as a potential immunity modulator. Overall, these findings highlight the anticancer potential of NPS-1034 in PDAC treatment in vitro, both as a monotherapy and in combination with traditional chemotherapy, offering a promising avenue for further in vivo investigation before clinical exploration.


Sujet(s)
Apoptose , Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Axl Receptor Tyrosine Kinase , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Fluorouracil/pharmacologie , Fluorouracil/usage thérapeutique , Oxaliplatine/pharmacologie , Oxaliplatine/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique
14.
Zhongguo Zhong Yao Za Zhi ; 49(13): 3515-3525, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39041123

RÉSUMÉ

Regulating the process of epithelial-mesenchymal transition(EMT) is an essential strategy to inhibit tumor growth and metastasis. This study is based on the EMT process of retinoblastoma and constructs quercetin(QUE) and doxorubicin(DOX) co-loaded liposome(QD Lipo) to investigate the therapeutic effect and mechanisms of combined QUE and DOX treatment on retinoblastoma. Single-factor experiments were conducted to optimize the prescription process of QD Lipo. Eventually, spherical particles with a diameter of(108.87±1.93) nm, a PDI of 0.13±0.02, and a Zeta potential of(-34.83±1.92) mV were obtained. The encapsulation rates of QUE and DOX were 96.20%±4.40% and 91.17%±4.41%, respectively. Y79 human retinoblastoma cells were used as an in vitro cellular model, and confocal microscopy demonstrated that QD Lipo could enhance Y79 uptake efficiency. The CCK-8 assay confirmed that the optimal combination therapy effect of QUE and DOX occurred at a mass ratio of 1∶1 to 1∶2. Flow cytometry showed that QD Lipo enhanced the induction of apoptosis in Y79 cells. Western blot analysis revealed that QD Lipo significantly reduced the expression of EMT pathway-related proteins vimentin and α-SMA. Fluorescence assays detected a significant decrease in ROS levels in Y79 cells after treatment with QD. These results indicated that liposomal co-delivery of QUE and DOX can enhance drug delivery efficiency to retinoblastoma cells, inhibit the EMT process in retinoblastoma by downregulating ROS levels, and enhance the cytotoxicity of DOX against retinoblastoma.


Sujet(s)
Doxorubicine , Transition épithélio-mésenchymateuse , Liposomes , Quercétine , Rétinoblastome , Quercétine/administration et posologie , Quercétine/pharmacologie , Quercétine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/administration et posologie , Rétinoblastome/traitement médicamenteux , Humains , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Liposomes/composition chimique , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments , Espèces réactives de l'oxygène/métabolisme
15.
Zhongguo Zhong Yao Za Zhi ; 49(13): 3657-3667, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39041138

RÉSUMÉ

This study aims to predict the possible targets and related signaling pathways of Modified Huoluo Xiaoling Pills against colorectal cancer(CRC) by both network pharmacology and molecular docking and verify the mechanism of action by experiments. TCMSP was used to obtain the active ingredients and targets of Modified Huoluo Xiaoling Pills, and GeneCards, DrugBank, OMIM, and TTD were employed to acquire CRC-related targets. Cytoscape software was utilized to construct the drug-active ingredient-target network, and the STRING database was applied to establish the protein-protein interaction(PPI) network. DAVID platform was adopted to investigate the targets in terms of GO function and KEGG pathway enrichment analysis. Molecular docking was performed in AutoDock Vina. HCT 116 cells were intervened by different concentrations of Modified Huoluo Xiaoling Pills-containing serum, and CCK-8 was used to detect the proliferation inhibition of HCT 116 cells in each group. Transwell was employed to show the invasive abi-lity of HCT 116 cells, and Western blot was taken to reveal the expression levels of ß-catenin, cyclinD1, c-Myc, as well as epithelial-mesenchymal transition(EMT) marker proteins E-cadherin, N-cadherin, vimentin, MMP2, MMP7, MMP9, and TWIST in HCT 116 cells. The network pharmacological analysis yielded 242 active ingredients of Modified Huoluo Xiaoling Pills, 1 844 CRC targets, and 127 overlapping targets of CRC and Modified Huoluo Xiaoling Pills, and the signaling pathways related to CRC involved PI3K-Akt, TNF, HIF-1, IL-17, Wnt, etc. Molecular docking showed that the key active ingredients had a stable binding conformation with the core proteins. CCK-8 indicated that Modified Huoluo Xiaoling Pills significantly inhibited the proliferation of HCT 116 cells. Transwell assay showed that with increasing concentration of Modified Huoluo Xiaoling Pills containing serum, the invasive ability of HCT 116 cells was more obviously inhibited. The expression of ß-catenin, cyclinD1, c-Myc, N-cadherin, vimentin, MMP2, MMP7, MMP9, and TWIST proteins were suppressed, and the expression of E-cadherin was improved by the intervention of drug-containing serum. Thus, it can be seen that Modified Huoluo Xiaoling Pills restrains the proliferation, invasion, and metastasis of CRC cells through multiple components, multiple targets, and multiple pathways, and the mechanism of action may be related to the inhibition of the activation of the Wnt/ß-catenin signaling pathway, thereby affecting the occurrence of EMT.


Sujet(s)
Prolifération cellulaire , Tumeurs colorectales , Médicaments issus de plantes chinoises , Simulation de docking moléculaire , Pharmacologie des réseaux , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules HCT116 , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
16.
Zhongguo Zhong Yao Za Zhi ; 49(11): 3012-3020, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-39041161

RÉSUMÉ

This study investigated the effects of modified Fangji Huangqi Decoction on the expression of proteins related to epithelial-mesenchymal transition(EMT) in a mouse model of unilateral ureteral obstruction( UUO) and in a rat renal tubular epithelial cell(NRK-52E) model of fibrosis induced by transforming growth factor ß1(TGF-ß1). It aims to decipher the molecular mechanism by which modified Fangji Huangqi Decoction alleviates renal interstitial fibrosis. C57/BL mice were subjected to UUO.After the surgery, the mice were treated with 0. 5-fold and 2-fold concentrations of modified Fangji Huangqi Decoction and fosinopril sodium(positive control) for 7 days. The interstitial collagen deposition in the kidney was assessed by Masson staining. Western blot and RT-qPCR were employed to determine the expression levels of TGF-ß1, phosphorylated Smad2/3(p-Smad2/3), Smad2/3, Snail,epithelial cadherin(E-cadherin), alpha smooth muscle actin(α-SMA), and vimentin. The NRK-52E cell model induced by TGF-ß1was treated with the serum samples collected from SD rats treated with different concentrations of modified Fangji Huangqi Decoction.The CCK-8 assay was employed to examine the effects of the serum samples on NRK-52E cell proliferation. The cell morphology in different groups was observed under a microscope. Furthermore, the modeled cells were treated with the serum containing 1-fold decoction. Western blot and RT-qPCR were then employed to measure the expression levels of p-Smad2/3, Smad2/3, Snail,E-cadherin, α-SMA, and vimentin in the cells. Under the same conditions, sh RNA was used to silence the Snail gene, and measurements were repeated before and after treatment with the serum containing 1-fold decoction. The results indicated that modified Fangji Huangqi Decoction alleviated the fibrotic injury in the mouse model of UUO and the fibrosis in the NRK-52E cell model. The treatment with the decoction down-regulated the protein and m RNA levels of EMT-related indicators including p-Smad2/3, α-SMA,Snail, and vimentin, while it up-regulated the expression of E-cadherin. After sh RNA silencing of the Snail gene, the protein and m RNA levels of E-cadherin, α-SMA, and vimentin showed no significant differences before and after treatment with the serum containing the decoction. The results suggest that modified Fangji Huangqi Decoction may alleviate renal interstitial fibrosis by inhibiting the TGF-ß1/Smad/Snail signaling pathway and regulating the EMT process.


Sujet(s)
Médicaments issus de plantes chinoises , Transition épithélio-mésenchymateuse , Fibrose , Souris de lignée C57BL , Transduction du signal , Protéines Smad , Facteurs de transcription de la famille Snail , Facteur de croissance transformant bêta-1 , Animaux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Souris , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/administration et posologie , Fibrose/traitement médicamenteux , Facteurs de transcription de la famille Snail/métabolisme , Facteurs de transcription de la famille Snail/génétique , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Protéines Smad/métabolisme , Protéines Smad/génétique , Humains , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Maladies du rein/traitement médicamenteux , Maladies du rein/métabolisme , Maladies du rein/génétique
17.
J Agric Food Chem ; 72(28): 15740-15754, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38970822

RÉSUMÉ

Hepatic fibrosis is a compensatory response to chronic liver injury and inflammation, and dietary intervention is recommended as one of the fundamental prevention strategies. Raspberry ketone (RK) is an aromatic compound first isolated from raspberry and widely used to prepare food flavors. The current study investigated the hepatoprotection and potential mechanism of RK against hepatic fibrosis. In vitro, hepatic stellate cell (HSC) activation was stimulated with TGF-ß and cultured with RK, farnesoid X receptor (FXR), or peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) agonist or inhibitor, respectively. In vivo, C57BL/6 mice were injected intraperitoneally with thioacetamide (TAA) at 100/200 mg/kg from the first to the fifth week. Mice were intragastrically administrated with RK or Cur once a day from the second to the fifth week. In activated HSCs, RK inhibited extracellular matrix (ECM) accumulation, inflammation, and epithelial-mesenchymal transition (EMT) process. RK both activated FXR/PGC-1α and regulated their crosstalk, which were verified by their inhibitors and agonists. Deficiency of FXR or PGC-1α also attenuated the effect of RK on the reverse of activated HSCs. RK also decreased serum ALT/AST levels, liver histopathological change, ECM accumulation, inflammation, and EMT in mice caused by TAA. Double activation of FXR/PGC-1α might be the key targets for RK against hepatic fibrosis. Above all, these discoveries supported the potential of RK as a novel candidate for the dietary intervention of hepatic fibrosis.


Sujet(s)
Butanones , Cellules étoilées du foie , Cirrhose du foie , Souris de lignée C57BL , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Récepteurs cytoplasmiques et nucléaires , Transduction du signal , Animaux , Humains , Mâle , Souris , Butanones/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Cirrhose du foie/métabolisme , Cirrhose du foie/génétique , Cirrhose du foie/traitement médicamenteux , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Récepteurs cytoplasmiques et nucléaires/métabolisme , Récepteurs cytoplasmiques et nucléaires/génétique , Rubus/composition chimique , Transduction du signal/effets des médicaments et des substances chimiques , Rats
18.
Cell Death Dis ; 15(7): 493, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987529

RÉSUMÉ

Lung cancer is a leading cause of cancer-related mortality globally, with a dismal 5-year survival rate, particularly for Lung Adenocarcinoma (LUAD). Mechanical changes within the tumor microenvironment, such as extracellular matrix (ECM) remodeling and fibroblast activity, play pivotal roles in cancer progression and metastasis. However, the specific impact of the basement membrane (BM) on the mechanical characteristics of LUAD remains unclear. This study aims to identify BM genes influencing internal mechanical stress in tumors, elucidating their effects on LUAD metastasis and therapy resistance, and exploring strategies to counteract these effects. Using Matrigel overlay and Transwell assays, we found that mechanical stress, mimicked by matrix application, augmented LUAD cell migration and invasion, correlating with ECM alterations and activation of the epithelial-mesenchymal transition (EMT) pathway. Employing machine learning, we developed the SVM_Score model based on relevant BM genes, which accurately predicted LUAD patient prognosis and EMT propensity across multiple datasets. Lower SVM_Scores were associated with worse survival outcomes, elevated cancer-related pathways, increased Tumor Mutation Burden, and higher internal mechanical stress in LUAD tissues. Notably, the SVM_Score was closely linked to COL5A1 expression in myofibroblasts, a key marker of mechanical stress. High COL5A1 expression from myofibroblasts promoted tumor invasiveness and EMT pathway activation in LUAD cells. Additionally, treatment with Sorafenib, which targets COL5A1 secretion, attenuated the tumor-promoting effects of myofibroblast-derived COL5A1, inhibiting LUAD cell proliferation, migration, and enhancing chemosensitivity. In conclusion, this study elucidates the complex interplay between mechanical stress, ECM alterations, and LUAD progression. The SVM_Score emerges as a robust prognostic tool reflecting tumor mechanical characteristics, while Sorafenib intervention targeting COL5A1 secretion presents a promising therapeutic strategy to mitigate LUAD aggressiveness. These findings deepen our understanding of the biomechanical aspects of LUAD and offer insights for future research and clinical applications.


Sujet(s)
Adénocarcinome pulmonaire , Collagène de type V , Transition épithélio-mésenchymateuse , Tumeurs du poumon , Myofibroblastes , Contrainte mécanique , Humains , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/traitement médicamenteux , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/anatomopathologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Collagène de type V/métabolisme , Collagène de type V/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Métastase tumorale , Souris , Microenvironnement tumoral , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Matrice extracellulaire/métabolisme
19.
Mol Biol Rep ; 51(1): 795, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39001907

RÉSUMÉ

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a disease of unknown etiology characterized by a constant incidence rate. Unfortunately, effective pharmacological treatments for this condition are lacking and the identification of novel therapeutic approaches and underlying pathological mechanisms are required. This study investigated the potential of quercetin in alleviating pulmonary fibrosis by promoting autophagy and activation of the SIRT1/AMPK pathway. METHODS: Mouse models of IPF were divided into four treatment groups: control, bleomycin (BLM), quercetin (Q), and quercetin + EX-527 (Q + E) treatment. Pulmonary fibrosis was induced in the mouse models through intratracheal instillation of BLM. Various indexes were identified through histological staining, Western blotting analysis, enzyme-linked immunosorbent assay, immunohistochemistry, and transmission electron microscopy. RESULTS: Quercetin treatment ameliorated the pathology of BLM-induced pulmonary fibrosis of mice by reducing α-smooth muscle actin (α-SMA), collagen I (Col I), and collagen III (Col III) levels, and also improved the level of E-cadherin in lung tissue. Furthermore, Quercetin significantly enhanced LC3II/LC3I levels, decreased P62 expression, and increased the number of autophagosomes in lung tissue. These effects were accompanied by the activation of the SIRT1/AMPK pathway. Treatment with EX-527, an inhibitor for SIRT1, reversed all effects induced by quercetin. CONCLUSION: This study showed that quercetin could alleviate pulmonary fibrosis and improve epithelial-mesenchymal transition by acting on the SIRT1/AMPK signaling pathway, which may be achieved by regulating the level of autophagy.


Sujet(s)
AMP-Activated Protein Kinases , Autophagie , Bléomycine , Fibrose pulmonaire , Quercétine , Transduction du signal , Sirtuine-1 , Animaux , Bléomycine/effets indésirables , Quercétine/pharmacologie , Sirtuine-1/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Souris , AMP-Activated Protein Kinases/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/anatomopathologie , Modèles animaux de maladie humaine , Mâle , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/induit chimiquement , Fibrose pulmonaire idiopathique/anatomopathologie , Souris de lignée C57BL
20.
ACS Appl Bio Mater ; 7(7): 4679-4689, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38963794

RÉSUMÉ

Postoperative peritoneal adhesion (PPA) is a prevalent complication of abdominal surgery, posing a significant hindrance to postsurgical recovery. Although several strategies have been developed to alleviate and prevent adhesions, their efficacy remains unsatisfactory. For the first time, we studied the therapeutic effect and mechanism of our recently developed thermally stable oligonucleotide-based mimetics of hepatocyte growth factor (HGF DNA aptamer) to prevent PPA. The HGF DNA aptamer effectively inhibited canonical TGF-ß1 signaling transduction, partially suppressing mesothelial mesenchymal transition. Additionally, the aptamer, respectively, upregulated and downregulated the expression of tissue plasminogen activator and plasminogen activator inhibitor 1, thereby enhancing fibrinolytic activity. As a pleiotropic factor, the HGF DNA aptamer also enhanced the migratory and proliferative capacities of mesothelial cells. Finally, the aptamer demonstrated a higher level of effectiveness in preventing PPAs than the commercially available antiperitoneal adhesion barrier, Seprafilm. Due to its therapeutic benefits, excellent stability, biosafety, cost-effectiveness, and versatility, the HGF DNA aptamer demonstrates promise for preventing PPA in future clinical settings.


Sujet(s)
Aptamères nucléotidiques , Transition épithélio-mésenchymateuse , Fibrinolyse , Facteur de croissance des hépatocytes , Aptamères nucléotidiques/composition chimique , Aptamères nucléotidiques/pharmacologie , Facteur de croissance des hépatocytes/métabolisme , Adhérences tissulaires/prévention et contrôle , Humains , Fibrinolyse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Test de matériaux , Taille de particule , Complications postopératoires/prévention et contrôle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE