Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.152
Filtrer
1.
J Cell Mol Med ; 28(12): e18458, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39031798

RÉSUMÉ

Schistosomiasis is a parasitic disease characterized by liver fibrosis, a process driven by the activation of hepatic stellate cells (HSCs) and subsequent collagen production. Previous studies from our laboratory have demonstrated the ability of Schistosoma japonicum protein P40 (SjP40) to inhibit HSCs activation and exert an antifibrotic effect. In this study, we aimed to elucidate the molecular mechanism underlying the inhibitory effect of recombinant SjP40 (rSjP40) on HSCs activation. Using a cell model in which rSjP40 inhibited LX-2 cell activation, we performed RNA-seq analyses and identified ATF3 as the most significantly altered gene. Further investigation revealed that rSjP40 inhibited HSCs activation partly by suppressing ATF3 activation. Knockdown of ATF3 in mouse liver significantly alleviated S. japonicum-induced liver fibrosis. Moreover, our results indicate that ATF3 is a direct target of microRNA-494-3p, a microRNA associated with anti-liver fibrosis effects. rSjP40 was found to downregulate ATF3 expression by upregulating microRNA-494-3p in LX-2 cells. This downregulation led to the inhibition of the expression of liver fibrosis proteins α-SMA and COL1A1, ultimately alleviating liver fibrosis caused by S. japonicum.


Sujet(s)
Facteur de transcription ATF-3 , Protéines d'helminthes , Cellules étoilées du foie , Cirrhose du foie , microARN , Schistosoma japonicum , Schistosomiase artérioveineuse , Animaux , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/parasitologie , Schistosomiase artérioveineuse/parasitologie , Schistosomiase artérioveineuse/métabolisme , Schistosomiase artérioveineuse/génétique , Cirrhose du foie/parasitologie , Cirrhose du foie/génétique , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Souris , Humains , microARN/génétique , microARN/métabolisme , Protéines d'helminthes/génétique , Protéines d'helminthes/métabolisme , Actines/métabolisme , Actines/génétique , Lignée cellulaire , Régulation de l'expression des gènes , Foie/métabolisme , Foie/parasitologie , Foie/anatomopathologie , Modèles animaux de maladie humaine , Antigènes d'helminthe
2.
Sci Adv ; 10(30): eado3141, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39047111

RÉSUMÉ

Metabolic dysfunction-associated steatohepatitis (MASH) is regulated by complex interplay between the macrophages and surrounding cells in the liver. Here, we show that Atf3 regulates glucose-fatty acid cycle in macrophages attenuates hepatocyte steatosis, and fibrogenesis in hepatic stellate cells (HSCs). Overexpression of Atf3 in macrophages protects against the development of MASH in Western diet-fed mice, whereas Atf3 ablation has the opposite effect. Mechanistically, Atf3 improves the reduction of fatty acid oxidation induced by glucose via forkhead box O1 (FoxO1) and Cd36. Atf3 inhibits FoxO1 activity via blocking Hdac1-mediated FoxO1 deacetylation at K242, K245, and K262 and increases Zdhhc4/5-mediated CD36 palmitoylation at C3, C7, C464, and C466; furthermore, macrophage Atf3 decreases hepatocytes lipogenesis and HSCs activation via retinol binding protein 4 (Rbp4). Anti-Rbp4 can prevent MASH progression that is induced by Atf3 deficiency in macrophages. This study identifies Atf3 as a regulator of glucose-fatty acid cycle. Targeting macrophage Atf3 or Rbp4 may be a plausible therapeutic strategy for MASH.


Sujet(s)
Facteur de transcription ATF-3 , Macrophages , Animaux , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Souris , Macrophages/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/étiologie , Cellules étoilées du foie/métabolisme , Acides gras/métabolisme , Glucose/métabolisme , Foie/métabolisme , Foie/anatomopathologie , Hépatocytes/métabolisme , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Lipogenèse , Humains , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Reprogrammation cellulaire , Souris de lignée C57BL ,
3.
Tissue Cell ; 89: 102447, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38991270

RÉSUMÉ

Activating transcription factor 3 (ATF3) has been identified as a regulator associated with osteoblast differentiation. However, the effects of ATF3 on the osteogenic differentiation and proliferation of human periodontal stem cells (hPDLSCs) in periodontitis have not been reported. With the purpose of establishing an in vitro model of periodontitis, hPDLSCs were challenged with lipopolysaccharide (LPS). The Cell Counting Kit-8 assay was applied to assess cell viability, while reverse transcription-quantitative PCR and western blotting were employed to detect ATF3 expression. Inflammatory release was assessed using ELISA, together with western blotting. Lipid peroxidation was explored using the C11 BODIPY 581/591 probe, biochemical kits, thiobarbituric acid reactive substances (TBARS) assay and DCFH-DA staining. Iron and Fe2+ levels, and the expression levels of ferroptosis-related proteins were measured using corresponding kits and western blotting. Osteogenic differentiative capability was evaluated using alkaline phosphatase staining, Alizarin red staining and western blotting. The expression levels of proteins associated with Nrf2/HO-1 signaling were identified using western blotting. The results indicated that ATF3 expression was upregulated in LPS-induced hPDLSCs. The knockdown of ATF3 alleviated the LPS-induced inflammatory response in hPDLSCs, together with increased levels of TNF-α, IL-6, IL-1ß, Cox-2 and iNOS, and decreased levels of IL-10. ATF3 silencing also led to lower TBARS production rate, and reduced levels of reactive oxygen species, iron, Fe2+, ACSL4 and TFR1, whereas it elevated the levels of SLC7A11 and GPX4. In addition, ATF3 silencing promoted hPDLSC mineralization and cell differentiation, and elevated the levels of OCN2, RUNX2 and BMP2. Additionally, ATF3 depletion upregulated the expression levels of proteins related with Nrf2/HO-1 signaling. The Nrf2 inhibitor ML385 partially counteracted the effects of ATF3 interference on the LPS-challenged inflammatory response, lipid peroxidation, ferroptosis as well as osteogenic differentiative capability in hPDLSCs. In summary, the results revealed that ATF3 silencing suppressed inflammation and ferroptosis, while it improved osteogenic differentiation in LPS-induced hPDLSCs by regulating Nrf2/HO-1 signaling, which may provide promising therapeutic targets for the treatment of periodontitis.


Sujet(s)
Facteur de transcription ATF-3 , Différenciation cellulaire , Ferroptose , Heme oxygenase-1 , Inflammation , Facteur-2 apparenté à NF-E2 , Ostéogenèse , Transduction du signal , Humains , Facteur-2 apparenté à NF-E2/métabolisme , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Heme oxygenase-1/métabolisme , Heme oxygenase-1/génétique , Lipopolysaccharides/pharmacologie , Cellules souches/métabolisme
4.
Chem Biol Drug Des ; 103(6): e14565, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38862254

RÉSUMÉ

Ferroptosis is a novel form of programmed cell death that is triggered by iron-dependent lipid peroxidation. Brusatol (BRU), a natural nuclear factor erythroid 2-related factor 2 inhibitor, exhibits potent anticancer effects in various types of cancer. However, the exact mechanism of BRU in the treatment of hepatocellular carcinoma (HCC) remains unknown. The anticancer effects of BRU in HCC were detected using cell counting kit-8 and colony formation assays and a xenograft model. RNA sequencing (RNA-seq) and bioinformatics analyses of HCC cells were utilized to elucidate the mechanism underlying the effects of BRU in HCC. The levels of reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and Fe2+ were measured using assay kits. The expression of activating transcription factor 3 (ATF3) was tested using RT-qPCR, western blotting, and immunofluorescence staining. The role of ATF3 in BRU-induced ferroptosis was examined using siATF3. BRU significantly inhibited HCC cell proliferation, both in vitro and in vivo. BRU activated the ferroptosis signaling pathway and increased ATF3 expression. Furthermore, ATF3 knockdown impeded BRU-induced ferroptosis. BRU suppressed HCC growth through ATF3-mediated ferroptosis, supporting BRU as a promising therapeutic agent for HCC.


Sujet(s)
Facteur de transcription ATF-3 , Carcinome hépatocellulaire , Ferroptose , Tumeurs du foie , Quassinoïdes , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Ferroptose/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Animaux , Quassinoïdes/pharmacologie , Quassinoïdes/composition chimique , Quassinoïdes/usage thérapeutique , Lignée cellulaire tumorale , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Souris de lignée BALB C , Transduction du signal/effets des médicaments et des substances chimiques
5.
Sci Rep ; 14(1): 14669, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918575

RÉSUMÉ

Non-obese diabetes (NOD) mice are an established, spontaneous model of type 1 diabetes in which diabetes develops through insulitis. Using next-generation sequencing, coupled with pathway analysis, the molecular fingerprint of early insulitis was mapped in a cohort of mice ranging from 4 to 12 weeks of age. The resulting dynamic timeline revealed an initial decrease in proliferative capacity followed by the emergence of an inflammatory signature between 6 and 8 weeks that increased to a regulatory plateau between 10 and 12 weeks. The inflammatory signature is identified by the activation of central immunogenic factors such as Infg, Il1b, and Tnfa, and activation of canonical inflammatory signaling. Analysis of the regulatory landscape revealed the transcription factor Atf3 as a potential novel modulator of inflammatory signaling in the NOD islets. Furthermore, the Hedgehog signaling pathway correlated with Atf3 regulation, suggesting that the two play a role in regulating islet inflammation; however, further studies are needed to establish the nature of this connection.


Sujet(s)
Facteur de transcription ATF-3 , Diabète de type 1 , Ilots pancréatiques , Souris de lignée NOD , Transduction du signal , Animaux , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Souris , Diabète de type 1/génétique , Diabète de type 1/métabolisme , Diabète de type 1/anatomopathologie , Femelle , Inflammation/génétique , Inflammation/anatomopathologie , Inflammation/métabolisme , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Analyse de profil d'expression de gènes , Modèles animaux de maladie humaine
6.
J Cell Mol Med ; 28(12): e18492, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38890795

RÉSUMÉ

Intervertebral disc degeneration (IVDD) severely affects the work and the quality of life of people. We previously demonstrated that silencing activation transcription factor 3 (ATF3) blocked the IVDD pathological process by regulating nucleus pulposus cell (NPC) ferroptosis, apoptosis, inflammation, and extracellular matrix (ECM) metabolism. Nevertheless, whether miR-874-3p mediated the IVDD pathological process by targeting ATF3 remains unclear. We performed single-cell RNA sequencing (scRNA-seq) and bioinformatics analysis to identify ATF3 as a key ferroptosis gene in IVDD. Then, Western blotting, flow cytometry, ELISA, and animal experiments were performed to validate the roles and regulatory mechanisms of miR-874-3p/ATF3 signalling axis in IVDD. ATF3 was highly expressed in IVDD patients and multiple cell types of IVDD rat, as revealed by scRNA-seq and bioinformatics analysis. GO analysis unveiled the involvement of ATF3 in regulating cell apoptosis and ECM metabolism. Furthermore, we verified that miR-874-3p might protect against IVDD by inhibiting NPC ferroptosis, apoptosis, ECM degradation, and inflammatory response by targeting ATF3. In vivo experiments displayed the protective effect of miR-874-3p/ATF3 axis on IVDD. These findings propose the potential of miR-874-3p and ATF3 as biomarkers of IVDD and suggest that targeting the miR-874-3p/ATF3 axis may be a therapeutic target for IVDD.


Sujet(s)
Facteur de transcription ATF-3 , Ferroptose , Dégénérescence de disque intervertébral , microARN , Nucleus pulposus , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Dégénérescence de disque intervertébral/génétique , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/anatomopathologie , microARN/génétique , microARN/métabolisme , Animaux , Humains , Rats , Ferroptose/génétique , Mâle , Nucleus pulposus/métabolisme , Nucleus pulposus/anatomopathologie , Analyse sur cellule unique/méthodes , Apoptose/génétique , Transduction du signal , Femelle , Adulte d'âge moyen , Rat Sprague-Dawley , Analyse de séquence d'ARN/méthodes , Matrice extracellulaire/métabolisme , Adulte , Régulation de l'expression des gènes , Modèles animaux de maladie humaine , Biologie informatique/méthodes
7.
Gene ; 927: 148670, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-38857714

RÉSUMÉ

LINC00894 may be associated with synaptic function, but its biology function in neural cells is still unknown. In this study, LINC00894 knockdown decreased the EdU incorporated into newly synthesized DNA and cell viability in MTT or CCK-8 assay in HEK-293T and BE(2)-M17 (M17) neuroblastoma cells. And LINC00894 knockdown increased cellular apoptosis in Annexin V-FITC staining, the expression of activated Caspase3 and the level of reactive oxygen species (ROS) both in HEK-293T and M17 cells. Moreover, LINC00894 also protected cells from hydrogen peroxide induced apoptosis in in vitro models. Utilizing RNA sequencing (RNA-seq) integrated with quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunoblot, we identified that LINC00894 affected activating transcription factor 3 (ATF3) expression in HEK-293T, M17, and SH-SY5Y neuroblastoma cells. Finally, we found that ectopic expression of ATF3 restored cell proliferation and inhibited cell apoptosis in LINC00894 downregulated M17 cells. While knockdown of ATF3 also significantly increased the cell viability inhibition and apoptosis promotion induced by LINC00894 knockdown in M17 cells. Our results from in vitro models revealed that LINC00894 could promote neuronal cell proliferation and inhibit cellular apoptosis by affecting ATF3 expression.


Sujet(s)
Facteur de transcription ATF-3 , Apoptose , Prolifération cellulaire , Neurones , ARN long non codant , Humains , Facteur de transcription ATF-3/génétique , Facteur de transcription ATF-3/métabolisme , Cellules HEK293 , ARN long non codant/génétique , ARN long non codant/métabolisme , Neurones/métabolisme , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Survie cellulaire , Neuroblastome/génétique , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Techniques de knock-down de gènes , Caspase-3/métabolisme , Caspase-3/génétique , Peroxyde d'hydrogène/pharmacologie
8.
Environ Toxicol Pharmacol ; 108: 104469, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38759848

RÉSUMÉ

We analyzed gene expression in THP-1 cells exposed to metal-based nanomaterials (NMs) [TiO2 (NM-100), ZnO (NM-110), SiO2 (NM-200), Ag (NM-300 K)]. A functional enrichment analysis of the significant differentially expressed genes (DEGs) identified the key modulated biological processes and pathways. DEGs were used to construct protein-protein interaction networks. NM-110 and NM-300 K induced changes in the expression of genes involved in oxidative and genotoxic stress, immune response, alterations of cell cycle, detoxification of metal ions and regulation of redox-sensitive pathways. Both NMs shared a number of highly connected protein nodes (hubs) including CXCL8, ATF3, HMOX1, and IL1B. NM-200 induced limited transcriptional changes, mostly related to the immune response; however, several hubs (CXCL8, ATF3) were identical with NM-110 and NM-300 K. No effects of NM-100 were observed. Overall, soluble nanomaterials NM-110 and NM-300 K exerted a wide variety of toxic effects, while insoluble NM-200 induced immunotoxicity; NM-100 caused no detectable changes on the gene expression level.


Sujet(s)
Cartes d'interactions protéiques , Argent , Titane , Humains , Titane/toxicité , Cellules THP-1 , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Argent/toxicité , Nanostructures/toxicité , Nanoparticules métalliques/toxicité , Oxyde de zinc/toxicité , Oxyde de zinc/composition chimique , Facteur de transcription ATF-3/génétique , Facteur de transcription ATF-3/métabolisme , Transcriptome/effets des médicaments et des substances chimiques , Silice/toxicité , Interleukine-8/métabolisme , Interleukine-8/génétique , Heme oxygenase-1
9.
Neurosci Lett ; 832: 137806, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38714229

RÉSUMÉ

BACKGROUND: Trigeminal neuralgia (TN) is a common and difficult-to-treat neuropathic pain disorder in clinical practice. Previous studies have shown that Toll-like receptor 4 (TLR4) modulates the activation of the NF-κB pathway to affect neuropathic pain in rats. Voltage-gated sodium channels (VGSCs) are known to play an important role in neuropathic pain electrical activity. OBJECTIVE: To investigate whether TLR4 can regulate Nav1.3 through the TRAF6/NF-κB p65 pathway after infraorbital nerve chronic constriction injury (ION-CCI). STUDY DESIGN: ION-CCI modeling was performed on SD (Sprague Dawley) rats. To verify the success of the modeling, we need to detect the mechanical pain threshold and ATF3. Then, detecting the expression of TLR4, TRAF6, NF-κB p65, p-p65, and Nav1.3 in rat TG. Subsequently, investigate the role of TLR4/TRAF6/NF-κB pathway in ION-CCI model by intrathecal injections of LPS-rs (TLR4 antagonist), C25-140 (TRAF6 inhibitor), and PDTC (NF-κB p65 inhibitor). RESULTS: ION-CCI surgery decreased the mechanical pain threshold of rats and increased the expression of ATF3, TLR4, TRAF6, NF-κB p-p65 and Nav1.3, but there was no difference in NF-κB p65 expression. After inject antagonist or inhibitor of the TLR4/TRAF6/NF-κB pathway, the expression of Nav1.3 was decreased and mechanical pain threshold was increased. CONCLUSION: In the rat model of ION-CCI, TLR4 in the rat trigeminal ganglion regulates Nav1.3 through the TRAF6/NF-κB p65 pathway, and TLR4 antagonist alleviates neuropathic pain in ION-CCI rats.


Sujet(s)
Canal sodique voltage-dépendant NAV1.3 , Rat Sprague-Dawley , Transduction du signal , Facteur-6 associé aux récepteurs de TNF , Récepteur de type Toll-4 , Animaux , Récepteur de type Toll-4/métabolisme , Facteur-6 associé aux récepteurs de TNF/métabolisme , Mâle , Canal sodique voltage-dépendant NAV1.3/métabolisme , Transduction du signal/physiologie , Facteur de transcription NF-kappa B/métabolisme , Névralgie essentielle du trijumeau/métabolisme , Rats , Modèles animaux de maladie humaine , Facteur de transcription RelA/métabolisme , Facteur de transcription ATF-3/métabolisme , Seuil nociceptif/physiologie
10.
Article de Anglais | MEDLINE | ID: mdl-38777778

RÉSUMÉ

BACKGROUND: Aristolochic acid nephropathy (AAN) is a rapidly progressive interstitial nephropathy caused by Aristolochic acid (AA). AAN is associated with the development of nephropathy and urothelial carcinoma. It is estimated that more than 100 million people worldwide are at risk of developing AAN. However, the underlying mechanisms driving renal deterioration in AAN remain poorly understood, and the treatment options are limited. METHODS: We obtained GSE27168 and GSE136276 series matrix data from the Gene Expression Omnibus (GEO) related to AAN. Using the R Studio environment, we applied the limma package and WGCNA package to identify co-differently expressed genes (co-DEGs). By GO/KEGG/GSVA analysis, we revealed common biological pathways. Subsequently, co-DEGs were subjected to the String database to construct a protein-protein interaction (PPI) network. The MCC algorithms implemented in the Cytohubba plugin were employed to identify hub genes. The hub genes were cross-referenced with the transcription factor (TF) database to identify hub TFs. Immune infiltration analysis was performed to identify key immune cell groups by utilizing CIBERSORT. The expressions of AAN-associated hub TFs were verified in vivo and in vitro. Finally, siRNA intervention was performed on the two TFs to verify their regulatory effect in AAN. RESULTS: Our analysis identified 88 co-DEGs through the "limma" and "WGCNA" R packages. A PPI network comprising 53 nodes and 34 edges was constructed with a confidence level >0.4. ATF3 and c-JUN were identified as hub TFs potentially linked to AAN. Additionally, expressions of ATF3 and c-JUN positively correlated with monocytes, basophils, and vessels, and negatively correlated with eosinophils and endothelial cells. We observed a significant increase in protein and mRNA levels of these two hub TFs. Furthermore, it was found that siRNA intervention targeting ATF3, but not c-JUN, alleviated cell damage induced by AA. The knockdown of ATF3 protects against oxidative stress and inflammation in the AAN cell model. CONCLUSION: This study provides novel insights into the role of ATF3 in AAN. The comprehensive analysis sheds light on the molecular mechanisms and identifies potential biomarkers and drug targets for AAN treatment.


Sujet(s)
Acides aristolochiques , Maladies du rein , Facteurs de transcription , Acides aristolochiques/toxicité , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Maladies du rein/induit chimiquement , Maladies du rein/génétique , Animaux , Souris , Humains , Facteur de transcription ATF-3/génétique , Facteur de transcription ATF-3/métabolisme , Cartes d'interactions protéiques
11.
Cell Death Dis ; 15(5): 318, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38710703

RÉSUMÉ

Glioblastoma stem cells (GSCs) play a key role in glioblastoma (GBM) resistance to temozolomide (TMZ) chemotherapy. With the increase in research on the tumour microenvironment, exosomes secreted by GSCs have become a new focus in GBM research. However, the molecular mechanism by which GSCs affect drug resistance in GBM cells via exosomes remains unclear. Using bioinformatics analysis, we identified the specific expression of ABCB4 in GSCs. Subsequently, we established GSC cell lines and used ultracentrifugation to extract secreted exosomes. We conducted in vitro and in vivo investigations to validate the promoting effect of ABCB4 and ABCB4-containing exosomes on TMZ resistance. Finally, to identify the transcription factors regulating the transcription of ABCB4, we performed luciferase assays and chromatin immunoprecipitation-quantitative PCR. Our results indicated that ABCB4 is highly expressed in GSCs. Moreover, high expression of ABCB4 promoted the resistance of GSCs to TMZ. Our study found that GSCs can also transmit their highly expressed ABCB4 to differentiated glioma cells (DGCs) through exosomes, leading to high expression of ABCB4 in these cells and promoting their resistance to TMZ. Mechanistic studies have shown that the overexpression of ABCB4 in GSCs is mediated by the transcription factor ATF3. In conclusion, our results indicate that GSCs can confer resistance to TMZ in GBM by transmitting ABCB4, which is transcribed by ATF3, through exosomes. This mechanism may lead to drug resistance and recurrence of GBM. These findings contribute to a deeper understanding of the mechanisms underlying drug resistance in GBM and provide novel insights into its treatment.


Sujet(s)
Sous-famille B de transporteurs à cassette liant l'ATP , Facteur de transcription ATF-3 , Tumeurs du cerveau , Résistance aux médicaments antinéoplasiques , Exosomes , Glioblastome , Cellules souches tumorales , Témozolomide , Témozolomide/pharmacologie , Témozolomide/usage thérapeutique , Glioblastome/génétique , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Humains , Exosomes/métabolisme , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Sous-famille B de transporteurs à cassette liant l'ATP/génétique , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Lignée cellulaire tumorale , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Animaux , Antinéoplasiques alcoylants/pharmacologie , Antinéoplasiques alcoylants/usage thérapeutique , Souris , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris nude
12.
BMC Musculoskelet Disord ; 25(1): 331, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38725009

RÉSUMÉ

BACKGROUND: The development of neuropathic pain (NP) is one of the reasons why the pain is difficult to treat, and microglial activation plays an important role in NP. Recently, platelet-rich plasma (PRP) has emerged as a novel therapeutic method for knee osteoarthritis (KOA). However, it's unclarified whether PRP has analgesic effects on NP induced by KOA and the underlying mechanisms unknown. PURPOSE: To observe the analgesic effects of PRP on NP induced by KOA and explore the potential mechanisms of PRP in alleviating NP. METHODS: KOA was induced in male rats with intra-articular injections of monosodium iodoacetate (MIA) on day 0. The rats received PRP or NS (normal saline) treatment at days 15, 17, and 19 after modeling. The Von Frey and Hargreaves tests were applied to assess the pain-related behaviors at different time points. After euthanizing the rats with deep anesthesia at days 28 and 42, the corresponding tissues were taken for subsequent experiments. The expression of activating transcription factor 3 (ATF3) in dorsal root ganglia (DRG) and ionized-calcium-binding adapter molecule-1(Iba-1) in the spinal dorsal horn (SDH) was detected by immunohistochemical staining. In addition, the knee histological assessment was performed by hematoxylin-eosin (HE) staining. RESULTS: The results indicated that injection of MIA induced mechanical allodynia and thermal hyperalgesia, which could be reversed by PRP treatment. PRP downregulated the expression of ATF3 within the DRG and Iba-1 within the SDH. Furthermore, an inhibitory effect on cartilage degeneration was observed in the MIA + PRP group only on day 28. CONCLUSION: These results indicate that PRP intra-articular injection therapy may be a potential therapeutic agent for relieving NP induced by KOA. This effect could be attributed to downregulation of microglial activation and reduction in nerve injury.


Sujet(s)
Régulation négative , Microglie , Névralgie , Gonarthrose , Plasma riche en plaquettes , Rat Sprague-Dawley , Animaux , Mâle , Névralgie/thérapie , Névralgie/métabolisme , Microglie/métabolisme , Rats , Gonarthrose/thérapie , Facteur de transcription ATF-3/métabolisme , Ganglions sensitifs des nerfs spinaux/métabolisme , Modèles animaux de maladie humaine , Injections articulaires , Protéines de liaison au calcium/métabolisme , Acide iodo-acétique/toxicité , Protéines des microfilaments
13.
Cancer Res ; 84(15): 2450-2467, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38718305

RÉSUMÉ

Peripheral T-cell lymphoma (PTCL) is a heterogeneous and aggressive disease with a poor prognosis. Histone deacetylase (HDAC) inhibitors have shown inhibitory effects on PTCL. A better understanding of the therapeutic mechanism underlying the effects of HDAC inhibitors could help improve treatment strategies. Herein, we found that high expression of HDAC3 is associated with poor prognosis in PTCL. HDAC3 inhibition suppressed lymphoma growth in immunocompetent mice but not in immunodeficient mice. HDAC3 deletion delayed the progression of lymphoma, reduced the lymphoma burden in the thymus, spleen, and lymph nodes, and prolonged the survival of mice bearing N-methyl-N-nitrosourea-induced lymphoma. Furthermore, inhibiting HDAC3 promoted the infiltration and enhanced the function of natural killer (NK) cells. Mechanistically, HDAC3 mediated ATF3 deacetylation, enhancing its transcriptional inhibitory activity. Targeting HDAC3 enhanced CXCL12 secretion through an ATF3-dependent pathway to stimulate NK-cell recruitment and activation. Finally, HDAC3 suppression improved the response of PTCL to conventional chemotherapy. Collectively, this study provides insights into the mechanism by which HDAC3 regulates ATF3 activity and CXCL12 secretion, leading to immune infiltration and lymphoma suppression. Combining HDAC3 inhibitors with chemotherapy may be a promising strategy for treating PTCL. Significance: Targeting HDAC3 suppresses progression of T-cell lymphoma by activating ATF3 to induce secretion of CXCL12 and promote infiltration of NK cells, providing an immunostimulatory approach for treating T-cell lymphoma patients.


Sujet(s)
Facteur de transcription ATF-3 , Chimiokine CXCL12 , Inhibiteurs de désacétylase d'histone , Histone deacetylases , Cellules tueuses naturelles , Lymphome T périphérique , Animaux , Inhibiteurs de désacétylase d'histone/pharmacologie , Souris , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Lymphome T périphérique/anatomopathologie , Lymphome T périphérique/immunologie , Lymphome T périphérique/métabolisme , Lymphome T périphérique/traitement médicamenteux , Lymphome T périphérique/génétique , Humains , Chimiokine CXCL12/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Lignée cellulaire tumorale , Femelle , Mâle , Souris de lignée C57BL , Pronostic
14.
Stem Cells ; 42(8): 763-776, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38733123

RÉSUMÉ

Endometrium fibrosis is the leading cause of uterine infertility. Macrophages participated in the occurrence and development of endometrial fibrosis. We previously reported that human umbilical cord multipotent stromal cells (hUC-MSCs) exerted their therapeutic effect in a macrophage-dependent manner in endometrial fibrosis. However precise mechanisms by which hUC-MSCs may influence macrophages in endometrial fibrosis remain largely unexplored. Here, we demonstrated that abnormal iron and lipid metabolism occurred in patients with intrauterine adhesions (IUA) and murine models. Ferroptosis has been proven to contribute to the progression of fibrotic diseases. Our results revealed that pharmacological activation of ferroptosis by Erastin aggravated endometrial fibrosis, while inhibition of ferroptosis by Ferrostatin-1 ameliorated endometrial fibrosis in vivo. Moreover, ferroptosis of macrophages was significantly upregulated in endometria of IUA murine models. Of note, transcriptome profiles revealed that CD36 gene expression was significantly increased in patients with IUA and immunofluorescence analysis showed CD36 protein was mainly located in macrophages. Silencing CD36 in macrophages could reverse cell ferroptosis. Dual luciferase reporter assay revealed that CD36 was the direct target of activation transcription factor 3 (ATF3). Furthermore, through establishing coculture system and IUA murine models, we found that hUC-MSCs had a protective role against macrophage ferroptosis and alleviated endometrial fibrosis related to decreased CD36 and ATF3. The effect of hUC-MSCs on macrophage ferroptosis was attributed to the upregulation of amphiregulin (AREG). Our data highlighted that macrophage ferroptosis occurred in endometrial fibrosis via the ATF3-CD36 pathway and hUC-MSCs protected against macrophage ferroptosis to alleviate endometrial fibrosis via secreting AREG. These findings provided a potential target for therapeutic implications of endometrial fibrosis.


Sujet(s)
Amphiréguline , Antigènes CD36 , Endomètre , Ferroptose , Fibrose , Macrophages , Cordon ombilical , Femelle , Humains , Cordon ombilical/cytologie , Cordon ombilical/métabolisme , Animaux , Macrophages/métabolisme , Souris , Amphiréguline/métabolisme , Amphiréguline/génétique , Endomètre/métabolisme , Endomètre/anatomopathologie , Antigènes CD36/métabolisme , Antigènes CD36/génétique , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Cellules souches multipotentes/métabolisme
15.
Arch Toxicol ; 98(7): 2065-2084, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38630284

RÉSUMÉ

Arsenic is highly toxic to the human bladder. In the present study, we established a human bladder epithelial cell line that closely mimics normal human bladder epithelial cells by immortalizing primary uroplakin 1B-positive human bladder epithelial cells with human telomerase reverse transcriptase (HBladEC-T). The uroplakin 1B-positive human bladder epithelial cell line was then used to evaluate the toxicity of seven arsenicals (iAsV, iAsIII, MMAV, MMAIII, DMAV, DMAIII, and DMMTAV). The cellular uptake and metabolism of each arsenical was different. Trivalent arsenicals and DMMTAV exhibited higher cellular uptake than pentavalent arsenicals. Except for MMAV, arsenicals were transported into cells by aquaglyceroporin 9 (AQP9). In addition to AQP9, DMAIII and DMMTAV were also taken up by glucose transporter 5. Microarray analysis demonstrated that arsenical treatment commonly activated the NRF2-mediated oxidative stress response pathway. ROS production increased with all arsenicals, except for MMAV. The activating transcription factor 3 (ATF3) was commonly upregulated in response to oxidative stress in HBladEC-T cells: ATF3 is an important regulator of necroptosis, which is crucial in arsenical-induced bladder carcinogenesis. Inorganic arsenics induced apoptosis while MMAV and DMAIII induced necroptosis. MMAIII, DMAV, and DMMTAV induced both cell death pathways. In summary, MMAIII exhibited the strongest cytotoxicity, followed by DMMTAV, iAsIII, DMAIII, iAsV, DMAV, and MMAV. The cytotoxicity of the tested arsenicals on HBladEC-T cells correlated with their cellular uptake and ROS generation. The ROS/NRF2/ATF3/CHOP signaling pathway emerged as a common mechanism mediating the cytotoxicity and carcinogenicity of arsenicals in HBladEC-T cells.


Sujet(s)
Facteur de transcription ATF-3 , Composés de l'arsenic , Cellules épithéliales , Stress oxydatif , Espèces réactives de l'oxygène , Vessie urinaire , Humains , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Vessie urinaire/effets des médicaments et des substances chimiques , Vessie urinaire/métabolisme , Vessie urinaire/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Facteur de transcription ATF-3/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Lignée cellulaire , Apoptose/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques
16.
Clin Transl Med ; 14(4): e1650, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38649772

RÉSUMÉ

BACKGROUND: Although many molecules have been investigated as biomarkers for spinal cord injury (SCI) or ischemic stroke, none of them are specifically induced in central nervous system (CNS) neurons following injuries with low baseline expression. However, neuronal injury constitutes a major pathology associated with SCI or stroke and strongly correlates with neurological outcomes. Biomarkers characterized by low baseline expression and specific induction in neurons post-injury are likely to better correlate with injury severity and recovery, demonstrating higher sensitivity and specificity for CNS injuries compared to non-neuronal markers or pan-neuronal markers with constitutive expressions. METHODS: In animal studies, young adult wildtype and global Atf3 knockout mice underwent unilateral cervical 5 (C5) SCI or permanent distal middle cerebral artery occlusion (pMCAO). Gene expression was assessed using RNA-sequencing and qRT-PCR, while protein expression was detected through immunostaining. Serum ATF3 levels in animal models and clinical human samples were measured using commercially available enzyme-linked immune-sorbent assay (ELISA) kits. RESULTS: Activating transcription factor 3 (ATF3), a molecular marker for injured dorsal root ganglion sensory neurons in the peripheral nervous system, was not expressed in spinal cord or cortex of naïve mice but was induced specifically in neurons of the spinal cord or cortex within 1 day after SCI or ischemic stroke, respectively. Additionally, ATF3 protein levels in mouse blood significantly increased 1 day after SCI or ischemic stroke. Importantly, ATF3 protein levels in human serum were elevated in clinical patients within 24 hours after SCI or ischemic stroke. Moreover, Atf3 knockout mice, compared to the wildtype mice, exhibited worse neurological outcomes and larger damage regions after SCI or ischemic stroke, indicating that ATF3 has a neuroprotective function. CONCLUSIONS: ATF3 is an easily measurable, neuron-specific biomarker for clinical SCI and ischemic stroke, with neuroprotective properties. HIGHLIGHTS: ATF3 was induced specifically in neurons of the spinal cord or cortex within 1 day after SCI or ischemic stroke, respectively. Serum ATF3 protein levels are elevated in clinical patients within 24 hours after SCI or ischemic stroke. ATF3 exhibits neuroprotective properties, as evidenced by the worse neurological outcomes and larger damage regions observed in Atf3 knockout mice compared to wildtype mice following SCI or ischemic stroke.


Sujet(s)
Facteur de transcription ATF-3 , Marqueurs biologiques , Accident vasculaire cérébral ischémique , Neurones , Traumatismes de la moelle épinière , Animaux , Femelle , Humains , Mâle , Souris , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Marqueurs biologiques/métabolisme , Marqueurs biologiques/sang , Modèles animaux de maladie humaine , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/génétique , Accident vasculaire cérébral ischémique/sang , Souris knockout , Neurones/métabolisme , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/génétique , Traumatismes de la moelle épinière/complications
17.
Cell Commun Signal ; 22(1): 240, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38664711

RÉSUMÉ

BACKGROUND: The repair of peripheral nerve injury poses a clinical challenge, necessitating further investigation into novel therapeutic approaches. In recent years, bone marrow mesenchymal stromal cell (MSC)-derived mitochondrial transfer has emerged as a promising therapy for cellular injury, with reported applications in central nerve injury. However, its potential therapeutic effect on peripheral nerve injury remains unclear. METHODS: We established a mouse sciatic nerve crush injury model. Mitochondria extracted from MSCs were intraneurally injected into the injured sciatic nerves. Axonal regeneration was observed through whole-mount nerve imaging. The dorsal root ganglions (DRGs) corresponding to the injured nerve were harvested to test the gene expression, reactive oxygen species (ROS) levels, as well as the degree and location of DNA double strand breaks (DSBs). RESULTS: The in vivo experiments showed that the mitochondrial injection therapy effectively promoted axon regeneration in injured sciatic nerves. Four days after injection of fluorescently labeled mitochondria into the injured nerves, fluorescently labeled mitochondria were detected in the corresponding DRGs. RNA-seq and qPCR results showed that the mitochondrial injection therapy enhanced the expression of Atf3 and other regeneration-associated genes in DRG neurons. Knocking down of Atf3 in DRGs by siRNA could diminish the therapeutic effect of mitochondrial injection. Subsequent experiments showed that mitochondrial injection therapy could increase the levels of ROS and DSBs in injury-associated DRG neurons, with this increase being correlated with Atf3 expression. ChIP and Co-IP experiments revealed an elevation of DSB levels within the transcription initiation region of the Atf3 gene following mitochondrial injection therapy, while also demonstrating a spatial proximity between mitochondria-induced DSBs and CTCF binding sites. CONCLUSION: These findings suggest that MSC-derived mitochondria injected into the injured nerves can be retrogradely transferred to DRG neuron somas via axoplasmic transport, and increase the DSBs at the transcription initiation regions of the Atf3 gene through ROS accumulation, which rapidly release the CTCF-mediated topological constraints on chromatin interactions. This process may enhance spatial interactions between the Atf3 promoter and enhancer, ultimately promoting Atf3 expression. The up-regulation of Atf3 induced by mitochondria further promotes the expression of downstream regeneration-associated genes and facilitates axon regeneration.


Sujet(s)
Facteur de transcription ATF-3 , Axones , Cassures double-brin de l'ADN , Ganglions sensitifs des nerfs spinaux , Cellules souches mésenchymateuses , Mitochondries , Régénération nerveuse , Espèces réactives de l'oxygène , Nerf ischiatique , Régulation positive , Animaux , Facteur de transcription ATF-3/génétique , Facteur de transcription ATF-3/métabolisme , Mitochondries/métabolisme , Mitochondries/génétique , Espèces réactives de l'oxygène/métabolisme , Axones/métabolisme , Régénération nerveuse/génétique , Régulation positive/génétique , Souris , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Nerf ischiatique/traumatismes , Nerf ischiatique/anatomopathologie , Ganglions sensitifs des nerfs spinaux/métabolisme , Souris de lignée C57BL , Mâle
18.
J Med Chem ; 67(8): 6810-6821, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38613772

RÉSUMÉ

Anti-PD-L1 immunotherapy, a new lung cancer treatment, is limited to a few patients due to low PD-L1 expression and tumor immunosuppression. To address these challenges, the upregulation of PD-L1 has the potential to elevate the response rate and efficiency of anti-PD-L1 and alleviate the immunosuppression of the tumor microenvironment. Herein, we developed a novel usnic acid-derived Iridium(III) complex, Ir-UA, that boosts PD-L1 expression and converts "cold tumors" to "hot". Subsequently, we administered Ir-UA combined with anti-PD-L1 in mice, which effectively inhibited tumor growth and promoted CD4+ and CD8+ T cell infiltration. To our knowledge, Ir-UA is the first iridium-based complex to stimulate the expression of PD-L1 by explicitly regulating its transcription factors, which not only provides a promising platform for immune checkpoint blockade but, more importantly, provides an effective treatment strategy for patients with low PD-L1 expression.


Sujet(s)
Antigène CD274 , Immunothérapie , Iridium , Animaux , Iridium/composition chimique , Iridium/pharmacologie , Antigène CD274/métabolisme , Souris , Humains , Immunothérapie/méthodes , Facteur de transcription ATF-3/métabolisme , Lignée cellulaire tumorale , Souris de lignée C57BL , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Femelle , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/synthèse chimique
19.
Circ Res ; 134(11): 1495-1511, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38686580

RÉSUMÉ

BACKGROUND: Abdominal aortic aneurysm (AAA) is a catastrophic disease with little effective therapy, likely due to the limited understanding of the mechanisms underlying AAA development and progression. ATF3 (activating transcription factor 3) has been increasingly recognized as a key regulator of cardiovascular diseases. However, the role of ATF3 in AAA development and progression remains elusive. METHODS: Genome-wide RNA sequencing analysis was performed on the aorta isolated from saline or Ang II (angiotensin II)-induced AAA mice, and ATF3 was identified as the potential key gene for AAA development. To examine the role of ATF3 in AAA development, vascular smooth muscle cell-specific ATF3 knockdown or overexpressed mice by recombinant adeno-associated virus serotype 9 vectors carrying ATF3, or shRNA-ATF3 with SM22α (smooth muscle protein 22-α) promoter were used in Ang II-induced AAA mice. In human and murine vascular smooth muscle cells, gain or loss of function experiments were performed to investigate the role of ATF3 in vascular smooth muscle cell proliferation and apoptosis. RESULTS: In both Ang II-induced AAA mice and patients with AAA, the expression of ATF3 was reduced in aneurysm tissues but increased in aortic lesion tissues. The deficiency of ATF3 in vascular smooth muscle cell promoted AAA formation in Ang II-induced AAA mice. PDGFRB (platelet-derived growth factor receptor ß) was identified as the target of ATF3, which mediated vascular smooth muscle cell proliferation in response to TNF-alpha (tumor necrosis factor-α) at the early stage of AAA. ATF3 suppressed the mitochondria-dependent apoptosis at the advanced stage by upregulating its direct target BCL2. Our chromatin immunoprecipitation results also demonstrated that the recruitment of NFκB1 and P300/BAF/H3K27ac complex to the ATF3 promoter induces ATF3 transcription via enhancer activation. NFKB1 inhibitor (andrographolide) inhibits the expression of ATF3 by blocking the recruiters NFKB1 and ATF3-enhancer to the ATF3-promoter region, ultimately leading to AAA development. CONCLUSIONS: Our results demonstrate a previously unrecognized role of ATF3 in AAA development and progression, and ATF3 may serve as a novel therapeutic and prognostic marker for AAA.


Sujet(s)
Facteur de transcription ATF-3 , Anévrysme de l'aorte abdominale , Muscles lisses vasculaires , Myocytes du muscle lisse , Facteur de transcription ATF-3/génétique , Facteur de transcription ATF-3/métabolisme , Animaux , Anévrysme de l'aorte abdominale/métabolisme , Anévrysme de l'aorte abdominale/anatomopathologie , Anévrysme de l'aorte abdominale/génétique , Anévrysme de l'aorte abdominale/induit chimiquement , Humains , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Souris , Mâle , Souris de lignée C57BL , Apoptose , Cellules cultivées , Angiotensine-II , Prolifération cellulaire , Aorte abdominale/anatomopathologie , Aorte abdominale/métabolisme , Modèles animaux de maladie humaine
20.
J Ethnopharmacol ; 330: 118228, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38643863

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Prostate cancer (PCa) is the most common malignancy of the male genitourinary system and currently lacks effective treatment. Semen Impatientis, the dried ripe seed of Impatiens balsamina L., is described by the Chinese Pharmacopoeia as a traditional Chinese medicine (TCM) and is used in clinical practice to treat tumors, abdominal masses, etc. In our previous study, the ethyl acetate extracts of Semen Impatientis (EAESI) was demonstrated to be the most effective extract against PCa among various extracts. However, the biological effects of EAESI against PCa in vivo and the specific antitumor mechanisms involved remain unknown. AIM OF THE STUDY: In this study, we aimed to investigate the antitumor effect of EAESI on PCa in vitro and in vivo by performing network pharmacology analysis, transcriptomic analysis, and experiments to explore and verify the underlying mechanisms involved. MATERIALS AND METHODS: The antitumor effect of EAESI on PCa in vitro and in vivo was investigated via CCK-8, EdU, flow cytometry, and wound healing assays and xenograft tumor models. Network pharmacology analysis and transcriptomic analysis were employed to explore the underlying mechanism of EAESI against PCa. Activating transcription factor 3 (ATF3) and androgen receptor (AR) were confirmed to be the targets of EAESI against PCa by RT‒qPCR, western blotting, and rescue assays. In addition, the interaction between ATF3 and AR was assessed by coimmunoprecipitation, immunofluorescence, and nuclear-cytoplasmic separation assays. RESULTS: EAESI decreased cell viability, inhibited cell proliferation and migration, and induced apoptosis in AR+ and AR- PCa cells. Moreover, EAESI suppressed the growth of xenograft tumors in vivo. Network pharmacology analysis revealed that the hub targets of EAESI against PCa included AR, AKT1, TP53, and CCND1. Transcriptomic analysis indicated that activating transcription factor 3 (ATF3) was the most likely critical target of EAESI. EAESI downregulated AR expression and decreased the transcriptional activity of AR through ATF3 in AR+ PCa cells; and EAESI promoted the expression of ATF3 and exerted its antitumor effect via ATF3 in AR+ and AR- PCa cells. CONCLUSIONS: EAESI exerts good antitumor effects on PCa both in vitro and in vivo, and ATF3 and AR are the critical targets through which EAESI exerts antitumor effects on AR+ and AR- PCa cells.


Sujet(s)
Acétates , Facteur de transcription ATF-3 , Souris nude , Pharmacologie des réseaux , Tumeurs de la prostate , Récepteurs aux androgènes , Tests d'activité antitumorale sur modèle de xénogreffe , Mâle , Animaux , Humains , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Acétates/composition chimique , Lignée cellulaire tumorale , Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques d'origine végétale/isolement et purification , Souris , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Transcriptome/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Mouvement cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE