Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.628
Filtrer
1.
Postepy Biochem ; 70(1): 33-38, 2024 05 23.
Article de Anglais | MEDLINE | ID: mdl-39016230

RÉSUMÉ

The early stress response by AP-1 (FOS/JUN), supported by upregulation of c-Myc and involved in cell-fate changes and adaptation to hostile environments, is increased in cancer. The review shows the biphasic character of this response with negative feed-back typically lasting a few hours as a feature of the genome regulation by self-organising criticality. It involves  rapid splitting of the pericentromeric heterochromatin clusters, opening of the active chromatin, and a massive transcription acceleration wave. Phylostratigraphic analysis revealed that AP-1 genes evolved in the Cambrian explosion ~500 Mya integrating the protein interaction networks of reproduction including proto-placenta intertwined with cytokine and immunity pathways, sex determination, oocyte maturation, and embryonal stemness. The peak  of this response as part of accelerated cell senescence led by AP-1 and IL-1ß was found in breast cancer cell-line resistant to doxorubicin. Adaptability of aggressive cancer to treatments can be explained by emergent stress response evolutionarily protecting reproduction.


Sujet(s)
Tumeurs , Stress physiologique , Facteur de transcription AP-1 , Humains , Tumeurs/métabolisme , Stress physiologique/physiologie , Facteur de transcription AP-1/métabolisme , Animaux
2.
Nat Commun ; 15(1): 4914, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851846

RÉSUMÉ

FOXA family proteins act as pioneer factors by remodeling compact chromatin structures. FOXA1 is crucial for the chromatin binding of the androgen receptor (AR) in both normal prostate epithelial cells and the luminal subtype of prostate cancer (PCa). Recent studies have highlighted the emergence of FOXA2 as an adaptive response to AR signaling inhibition treatments. However, the role of the FOXA1 to FOXA2 transition in regulating cancer lineage plasticity remains unclear. Our study demonstrates that FOXA2 binds to distinct classes of developmental enhancers in multiple AR-independent PCa subtypes, with its binding depending on LSD1. Moreover, we reveal that FOXA2 collaborates with JUN at chromatin and promotes transcriptional reprogramming of AP-1 in lineage-plastic cancer cells, thereby facilitating cell state transitions to multiple lineages. Overall, our findings underscore the pivotal role of FOXA2 as a pan-plasticity driver that rewires AP-1 to induce the differential transcriptional reprogramming necessary for cancer cell lineage plasticity.


Sujet(s)
Lignage cellulaire , Régulation de l'expression des gènes tumoraux , Facteur nucléaire hépatocytaire HNF-3 bêta , Tumeurs de la prostate , Facteur de transcription AP-1 , Mâle , Humains , Facteur nucléaire hépatocytaire HNF-3 bêta/métabolisme , Facteur nucléaire hépatocytaire HNF-3 bêta/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Facteur de transcription AP-1/métabolisme , Facteur de transcription AP-1/génétique , Lignée cellulaire tumorale , Lignage cellulaire/génétique , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Facteur nucléaire hépatocytaire HNF-3 alpha/métabolisme , Facteur nucléaire hépatocytaire HNF-3 alpha/génétique , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Animaux , Chromatine/métabolisme , Chromatine/génétique , Plasticité cellulaire/génétique , Reprogrammation cellulaire/génétique , Souris , Protéines proto-oncogènes c-jun/métabolisme , Protéines proto-oncogènes c-jun/génétique , Éléments activateurs (génétique)/génétique , Transcription génétique
3.
Front Immunol ; 15: 1347018, 2024.
Article de Anglais | MEDLINE | ID: mdl-38887289

RÉSUMÉ

Purpose: Inflammation is involved in the pathogenesis of diabetes, however the impact of diabetes on organ-specific autoimmune diseases remains unexplored. Experimental autoimmune uveoretinitis (EAU) is a widely accepted animal model of human endogenous uveitis. In this study, we investigated the effects of diabetic conditions on the development of EAU using a mouse diabetes model. Methods: EAU was induced in wild-type C57BL/6 (WT) mice and Ins2Akita (Akita) mice with spontaneous diabetes by immunization with IRBP peptide. Clinical and histopathological examinations, and analysis of T cell activation state were conducted. In addition, alternations in the composition of immune cell types and gene expression profiles of relevant immune functions were identified using single-cell RNA sequencing. Results: The development of EAU was significantly attenuated in immunized Akita (Akita-EAU) mice compared with immunized WT (WT-EAU) mice, although T cells were fully activated in Akita-EAU mice, and the differentiation into Th17 cells and regulatory T (Treg) cells was promoted. However, Th1 cell differentiation was inhibited in Akita-EAU mice, and single-cell analysis indicated that gene expression associated AP-1 signaling pathway (JUN, FOS, and FOSB) was downregulated not only in Th1 cells but also in Th17, and Treg cells in Akita-EAU mice at the onset of EAU. Conclusions: In diabetic mice, EAU was significantly attenuated. This was related to selective inhibition of Th1 cell differentiation and downregulated AP-1 signaling pathway in both Th1 and Th17 cells.


Sujet(s)
Maladies auto-immunes , Différenciation cellulaire , Souris de lignée C57BL , Transduction du signal , Lymphocytes auxiliaires Th1 , Cellules Th17 , Facteur de transcription AP-1 , Uvéite , Animaux , Uvéite/immunologie , Cellules Th17/immunologie , Lymphocytes auxiliaires Th1/immunologie , Souris , Facteur de transcription AP-1/métabolisme , Différenciation cellulaire/immunologie , Maladies auto-immunes/immunologie , Diabète expérimental/immunologie , Modèles animaux de maladie humaine , Femelle
4.
Sci Rep ; 14(1): 14341, 2024 06 21.
Article de Anglais | MEDLINE | ID: mdl-38906916

RÉSUMÉ

Glioblastoma (GBM) is a highly aggressive and deadly brain cancer. Temozolomide (TMZ) is the standard chemotherapeutic agent for GBM, but the majority of patients experience recurrence and invasion of tumor cells. We investigated whether TMZ treatment of GBM cells regulates matrix metalloproteinases (MMPs), which have the main function to promote tumor cell invasion. TMZ effectively killed GL261, U343, and U87MG cells at a concentration of 500 µM, and surviving cells upregulated MMP9 expression and its activity but not those of MMP2. TMZ also elevated levels of MMP9 mRNA and MMP9 promoter activity. Subcutaneous graft tumors survived from TMZ treatment also exhibited increased expression of MMP9 and enhanced gelatinolytic activity. TMZ-mediated MMP9 upregulation was specifically mediated through the phosphorylation of p38 and JNK. This then stimulates AP-1 activity through the upregulation of c-Fos and c-Jun. Inhibition of the p38, JNK, or both pathways counteracted the TMZ-induced upregulation of MMP9 and AP-1. This study proposes a potential adverse effect of TMZ treatment for GBM: upregulation of MMP9 expression potentially associated with increased invasion and poor prognosis. This study also provides valuable insights into the molecular mechanisms by which TMZ treatment leads to increased MMP9 expression in GBM cells.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Glioblastome , Matrix metalloproteinase 9 , Témozolomide , p38 Mitogen-Activated Protein Kinases , Témozolomide/pharmacologie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Glioblastome/génétique , Glioblastome/anatomopathologie , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 9/génétique , Humains , p38 Mitogen-Activated Protein Kinases/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Antinéoplasiques alcoylants/pharmacologie , Animaux , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Facteur de transcription AP-1/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Souris
5.
Nature ; 630(8016): 475-483, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38839958

RÉSUMÉ

Senescence is a cellular state linked to ageing and age-onset disease across many mammalian species1,2. Acutely, senescent cells promote wound healing3,4 and prevent tumour formation5; but they are also pro-inflammatory, thus chronically exacerbate tissue decline. Whereas senescent cells are active targets for anti-ageing therapy6-11, why these cells form in vivo, how they affect tissue ageing and the effect of their elimination remain unclear12,13. Here we identify naturally occurring senescent glia in ageing Drosophila brains and decipher their origin and influence. Using Activator protein 1 (AP1) activity to screen for senescence14,15, we determine that senescent glia can appear in response to neuronal mitochondrial dysfunction. In turn, senescent glia promote lipid accumulation in non-senescent glia; similar effects are seen in senescent human fibroblasts in culture. Targeting AP1 activity in senescent glia mitigates senescence biomarkers, extends fly lifespan and health span, and prevents lipid accumulation. However, these benefits come at the cost of increased oxidative damage in the brain, and neuronal mitochondrial function remains poor. Altogether, our results map the trajectory of naturally occurring senescent glia in vivo and indicate that these cells link key ageing phenomena: mitochondrial dysfunction and lipid accumulation.


Sujet(s)
Vieillissement , Encéphale , Vieillissement de la cellule , Drosophila melanogaster , Métabolisme lipidique , Mitochondries , Névroglie , Animaux , Femelle , Humains , Mâle , Vieillissement/métabolisme , Vieillissement/anatomopathologie , Encéphale/métabolisme , Encéphale/anatomopathologie , Encéphale/cytologie , Drosophila melanogaster/métabolisme , Drosophila melanogaster/cytologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Longévité , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Névroglie/métabolisme , Névroglie/anatomopathologie , Neurones/métabolisme , Neurones/anatomopathologie , Stress oxydatif , Facteur de transcription AP-1/métabolisme , Lipides , Inflammation/métabolisme , Inflammation/anatomopathologie
6.
Molecules ; 29(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38893478

RÉSUMÉ

Transient receptor potential melastatin-8 (TRPM8) is a cation channel that is activated by cold and "cooling agents" such as menthol and icilin, which induce a cold sensation. The stimulation of TRPM8 activates an intracellular signaling cascade that ultimately leads to a change in the gene expression pattern of the cells. Here, we investigate the TRPM8-induced signaling pathway that links TRPM8 channel activation to gene transcription. Using a pharmacological approach, we show that the inhibition of phosphatidylinositol 4-phosphate 5 kinase α (PIP5K), an enzyme essential for the biosynthesis of phosphatidylinositol 4,5-bisphosphate, attenuates TRPM8-induced gene transcription. Analyzing the link between TRPM8 and Gq proteins, we show that the pharmacological inhibition of the ßγ subunits impairs TRPM8 signaling. In addition, genetic studies show that TRPM8 requires an activated Gα subunit for signaling. In the nucleus, the TRPM8-induced signaling cascade triggers the activation of the transcription factor AP-1, a complex consisting of a dimer of basic region leucine zipper (bZIP) transcription factors. Here, we identify the bZIP protein c-Jun as an essential component of AP-1 within the TRPM8-induced signaling cascade. In summary, with PIP5K, Gq subunits, and c-Jun, we identified key molecules in TRPM8-induced signaling from the plasma membrane to the nucleus.


Sujet(s)
Sous-unités alpha Gq-G11 des protéines G , Phosphotransferases (Alcohol Group Acceptor) , Transduction du signal , Canaux cationiques TRPM , Canaux cationiques TRPM/métabolisme , Canaux cationiques TRPM/génétique , Sous-unités alpha Gq-G11 des protéines G/métabolisme , Humains , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Phosphotransferases (Alcohol Group Acceptor)/génétique , Facteur de transcription AP-1/métabolisme , Cellules HEK293 , Protéines proto-oncogènes c-jun/métabolisme , Animaux
7.
Int J Oncol ; 65(1)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38847230

RÉSUMÉ

CD46, a transmembrane protein known for protecting cells from complement­mediated damage, is frequently dysregulated in various types of cancer. Its overexpression in bladder cancers safeguards the cancer cells against both complement and antibody­mediated cytotoxicity. The present study explored a new role of CD46 in facilitating cancer cell invasion and metastasis, examining its regulatory effect on matrix metalloproteases (MMPs) and their effect on the metastatic capability of bladder cancer cells. Specifically, CD46 alteration positively influenced MMP9 expression, but not MMP2, in several bladder cancer cell lines. Furthermore, CD46 overexpression triggered phosphorylation of p38 MAPK and protein kinase B (AKT), leading to enhanced activator protein 1 (AP­1) activity via c­Jun upregulation. The inhibition of p38 or AKT pathways attenuated the CD46­induced MMP9 and AP­1 upregulation, indicating that the promotion of MMP9 by CD46 involved activating both p38 MAPK and AKT. Functionally, the upregulation of MMP9 by CD46 translated to increased migratory and invasive capabilities of bladder cancer cells, as well as enhanced in vivo metastasis. Overall, the present study revealed a novel role for CD46 as a metastasis promoter through MMP9 activation in bladder cancers and highlighted the regulatory mechanism of CD46­mediated MMP9 promotion via p38 MAPK and AKT activation.


Sujet(s)
Mouvement cellulaire , Matrix metalloproteinase 9 , Antigènes CD46 , Protéines proto-oncogènes c-akt , Tumeurs de la vessie urinaire , p38 Mitogen-Activated Protein Kinases , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/génétique , Humains , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 9/génétique , Lignée cellulaire tumorale , p38 Mitogen-Activated Protein Kinases/métabolisme , Souris , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Antigènes CD46/métabolisme , Antigènes CD46/génétique , Régulation de l'expression des gènes tumoraux , Métastase tumorale , Invasion tumorale , Facteur de transcription AP-1/métabolisme , Régulation positive , Transduction du signal
8.
Bratisl Lek Listy ; 125(6): 382-386, 2024.
Article de Anglais | MEDLINE | ID: mdl-38757596

RÉSUMÉ

OBJECTIVES: To distinguish whether idiopathic intracranial hypertension (IIH) is a condition predisposing to multiple sclerosis (MS) or an isolated disease, the current gene transcription factor Activator Protein-1 (AP-1) was evaluated with its potential to differentiate both diseases. BACKGROUND: The aim of this study was to investigate the use of AP-1 as biomarkers for the discrimination of IIH and MS. METHODS: AP-1, TNF-α, and IL-6 protein values in the CSF of the cases were evaluated by the ELISA method. The numerical measures of the groups and the ability of AP-1 to distinguish the groups were analyzed with the ROC curve. RESULTS: There was no difference between the groups in CSF TNF-α, IL-6, CSF, and serum biochemistry analyses. However, it was determined that the AP-1 concentration (pg/ml) was significantly higher in the IIH group, the sensitivity of AP-1 in separating those with IIH was 75%, and the specificity in separating those with MS was 60% in those with an AP-1 concentration of 606.5 and above. CONCLUSION: According to our results, the fact that CSF TNF-α and IL-6 values did not differ in IIH compared to MS revealed that IIH could not methodologically control MS, and AP-1 was a supportive parameter in differentiating both diseases (Tab. 2, Fig. 1, Ref. 31).


Sujet(s)
Marqueurs biologiques , Interleukine-6 , Sclérose en plaques , Facteur de transcription AP-1 , Facteur de nécrose tumorale alpha , Humains , Marqueurs biologiques/liquide cérébrospinal , Interleukine-6/liquide cérébrospinal , Sclérose en plaques/liquide cérébrospinal , Sclérose en plaques/diagnostic , Adulte , Femelle , Diagnostic différentiel , Mâle , Facteur de transcription AP-1/liquide cérébrospinal , Facteur de transcription AP-1/métabolisme , Facteur de nécrose tumorale alpha/liquide cérébrospinal , Syndrome d'hypertension intracrânienne bénigne/liquide cérébrospinal , Syndrome d'hypertension intracrânienne bénigne/diagnostic , Sensibilité et spécificité , Adulte d'âge moyen , Courbe ROC
9.
Chemosphere ; 359: 142299, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38761826

RÉSUMÉ

Sulfur mustard (SM, dichlorodiethyl sulfide) is a potent erosive chemical poison that can cause pulmonary lung, skin and eye disease complications in humans. Currently, there is no designated remedy for SM, and its operation's toxicological process remains unidentified. This work employed zebrafish as a model organism to investigate the toxic manifestations and mechanisms of exposure to SM, aiming to offer novel insights for preventing and treating this condition. The results showed that SM caused a decrease in the survival rate of the zebrafish larvae (LC50 = 2.47 mg/L), a reduction in the hatching rate, an increase in the pericardial area, and small head syndrome. However, T-5224 (a selective inhibitor of c-Fos/activator protein) attenuated the reduction in mortality (LC50 = 2.79 mg/L), the reduction in hatching rate, and the worsening of morphological changes. We discovered that SM causes cartilage developmental disorders in zebrafish larvae. The reverse transcription-quantitative polymerase chain reaction found that SM increased the expression of inflammation-related genes (IL-1ß, IL-6, and TNF-α) and significantly increased cartilage development-related gene expression (fosab, mmp9, and atf3). However, the expression of sox9a, sox9b, and Col2a1a was reduced. The protein level detection also found an increase in c-fos protein expression and a significant decrease in COL2A1 expression. However, T-5224,also and mitigated the changes in gene expression, and protein levels caused by SM exposure. The results of this study indicate that SM-induced cartilage development disorders are closely related to the c-Fos/AP-1 pathway in zebrafish.


Sujet(s)
Chondrogenèse , Larve , Gaz moutarde , Protéines proto-oncogènes c-fos , Facteur de transcription AP-1 , Danio zébré , Animaux , Gaz moutarde/toxicité , Larve/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-fos/génétique , Chondrogenèse/effets des médicaments et des substances chimiques , Facteur de transcription AP-1/métabolisme , Protéines de poisson-zèbre/métabolisme , Protéines de poisson-zèbre/génétique
10.
Nat Commun ; 15(1): 4650, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38821936

RÉSUMÉ

Synovial tissue inflammation is a hallmark of rheumatoid arthritis (RA). Recent work has identified prominent pathogenic cell states in inflamed RA synovial tissue, such as T peripheral helper cells; however, the epigenetic regulation of these states has yet to be defined. Here, we examine genome-wide open chromatin at single-cell resolution in 30 synovial tissue samples, including 12 samples with transcriptional data in multimodal experiments. We identify 24 chromatin classes and predict their associated transcription factors, including a CD8 + GZMK+ class associated with EOMES and a lining fibroblast class associated with AP-1. By integrating with an RA tissue transcriptional atlas, we propose that these chromatin classes represent 'superstates' corresponding to multiple transcriptional cell states. Finally, we demonstrate the utility of this RA tissue chromatin atlas through the associations between disease phenotypes and chromatin class abundance, as well as the nomination of classes mediating the effects of putatively causal RA genetic variants.


Sujet(s)
Polyarthrite rhumatoïde , Chromatine , Membrane synoviale , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/immunologie , Humains , Chromatine/métabolisme , Chromatine/génétique , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Épigenèse génétique , Analyse sur cellule unique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Fibroblastes/métabolisme , Facteur de transcription AP-1/métabolisme , Facteur de transcription AP-1/génétique , Transcription génétique , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme
11.
Sci China Life Sci ; 67(7): 1468-1478, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38703348

RÉSUMÉ

Dietary exposure to aflatoxin B1 (AFB1) is harmful to the health and performance of domestic animals. The hepatic cytochrome P450s (CYPs), CYP1A1 and CYP2A6, are the primary enzymes responsible for the bioactivation of AFB1 to the highly toxic exo-AFB1-8,9-epoxide (AFBO) in chicks. However, the transcriptional regulation mechanism of these CYP genes in the liver of chicks in AFB1 metabolism remains unknown. Dual-luciferase reporter assay, bioinformatics and site-directed mutation results indicated that specificity protein 1 (SP1) and activator protein-1 (AP-1) motifs were located in the core region -1,063/-948, -606/-541 of the CYP1A1 promoter as well as -636/-595, -503/-462, -147/-1 of the CYP2A6 promoter. Furthermore, overexpression and decoy oligodeoxynucleotide technologies demonstrated that SP1 and AP-1 were pivotal transcriptional activators regulating the promoter activity of CYP1A1 and CYP2A6. Moreover, bioactivation of AFB1 to AFBO could be increased by upregulation of CYP1A1 and CYP2A6 expression, which was trans-activated owing to the upregulalion of AP-1, rather than SP1, stimulated by AFB1-induced reactive oxygen species. Additionally, nano-selenium could reduce ROS, downregulate AP-1 expression and then decrease the expression of CYP1A1 and CYP2A6, thus alleviating the toxicity of AFB1. In conclusion, AP-1 and SP1 played important roles in the transactivation of CYP1A1 and CYP2A6 expression and further bioactivated AFB1 to AFBO in chicken liver, which could provide novel targets for the remediation of aflatoxicosis in chicks.


Sujet(s)
Aflatoxine B1 , Poulets , Cytochrome P-450 CYP1A1 , Cytochrome P-450 CYP2A6 , Foie , Régions promotrices (génétique) , Facteur de transcription Sp1 , Facteur de transcription AP-1 , Animaux , Aflatoxine B1/métabolisme , Poulets/métabolisme , Foie/métabolisme , Facteur de transcription Sp1/métabolisme , Facteur de transcription Sp1/génétique , Cytochrome P-450 CYP1A1/génétique , Cytochrome P-450 CYP1A1/métabolisme , Facteur de transcription AP-1/métabolisme , Facteur de transcription AP-1/génétique , Cytochrome P-450 CYP2A6/métabolisme , Cytochrome P-450 CYP2A6/génétique , Activation de la transcription
12.
J Ethnopharmacol ; 331: 118345, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38754645

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Justicia adhatoda L. is used as traditional medicine in Nepal to treat cough, asthma, and inflammatory disorders, and is indicated as "Asuro". Leaves are used worldwide as herbal medicine due to cardiotonic, expectorant, anti-asthmatic, and bronchodilatory properties. The aim of this work was to study the phytochemical composition of leaves of Nepalese J. adhatoda and assess their anti-inflammatory and antioxidant properties in vitro. MATERIALS AND METHODS: Secondary metabolites were extracted from dried leaves using methanol (JAME: J. adhatoda methanol extract). They were analysed by means of liquid chromatography coupled with multiple-stage mass spectrometry (LC-MSn). Anti-inflammatory potential was determined by the NF-κB and AP-1 inhibition assay, and DPPH, ABTS, and ß-carotene bleaching assays were performed to assess its antioxidant properties. RESULTS: JAME is a rich source of secondary metabolites, especially quinazoline alkaloids such as vasicine, vasicinone, vasicoline, and adhatodine. 7-Hydroxy derivatives of peganidine, vasicolinone, and adhatodine were also identified by means of MSn data and are here reported in J. adhatoda for the first time. JAME inhibited NF-κB and AP-1 expression in THP-1 cells to a greater extent than the positive control prednisolone. A moderate radical-quenching property was observed in DPPH and ABTS assays, but the anti-carotene bleaching activity was significantly higher than the reference BHT. CONCLUSIONS: To the best of our knowledge, this is the first insight into the phytochemical composition of Asuro leaves from Nepal and their bioactivity. Our results will contribute to the valorisation of this medicinal species still widely used in the traditional and complementary medicine.


Sujet(s)
Alcaloïdes , Anti-inflammatoires , Antioxydants , Adhatoda , Facteur de transcription NF-kappa B , Extraits de plantes , Feuilles de plante , Quinazolines , Facteur de transcription AP-1 , Feuilles de plante/composition chimique , Facteur de transcription NF-kappa B/métabolisme , Antioxydants/pharmacologie , Antioxydants/isolement et purification , Adhatoda/composition chimique , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/isolement et purification , Alcaloïdes/pharmacologie , Alcaloïdes/isolement et purification , Facteur de transcription AP-1/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Quinazolines/pharmacologie , Quinazolines/isolement et purification , Humains , Médecine traditionnelle
13.
Pathol Res Pract ; 258: 155334, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38718468

RÉSUMÉ

Placental transmogrification of the lung (PTL) is a rare pulmonary condition characterized by the presence of immature placental villous structures. The etiology and molecular mechanisms underlying this disease remain largely unknown. This functional study aimed to identify the molecular signatures in the pathogenesis of PTL via comprehensive transcriptome analysis. Comparative transcriptomic assessment of PTL tissue and stromal cells showed differential expression of 257 genes in PTL tissue and 189 genes in stromal cells. Notably, several transcription factors and regulators, including FOSB, FOS, JUN, and ATF3, were upregulated in PTL tissue. Additionally, genes associated with the extracellular matrix and connective tissue, such as COL1A1, MMP2, and SPARC, were significantly altered, indicating possible fibrotic changes. Gene set enrichment analysis highlighted the role of vascular development and extracellular matrix organization, and the Activator Protein-1 (AP-1) transcription factor was significantly activated in PTL tissue. Furthermore, the analysis highlighted an overlap of 25 genes between PTL tissue and stromal cells, underscoring the importance of shared molecular pathways in the pathogenesis of PTL. Among the shared genes, JUND, COL4A2, COL6A2, IGFBP5, and IGFBP7 were consistently upregulated, highlighting the possible involvement of AP-1-mediated signaling and fibrotic changes in the pathogenesis of PTL. The present findings pave the way for further research into the molecular mechanisms underlying PTL and offer novel insights for therapeutic interventions. Given the rarity of PTL, these molecular findings represent a significant step forward in our understanding this enigmatic disease.


Sujet(s)
Analyse de profil d'expression de gènes , Facteur de transcription AP-1 , Humains , Femelle , Facteur de transcription AP-1/métabolisme , Facteur de transcription AP-1/génétique , Grossesse , Transcriptome , Poumon/anatomopathologie , Poumon/métabolisme , Fibrose/anatomopathologie , Fibrose/génétique , Placenta/anatomopathologie , Placenta/métabolisme , Maladies pulmonaires/génétique , Maladies pulmonaires/anatomopathologie , Maladies pulmonaires/métabolisme
14.
Inflamm Res ; 73(7): 1173-1184, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38739197

RÉSUMÉ

OBJECTIVE AND DESIGN: To elucidate Sirt1's role in gouty arthritis inflammation and its potential mechanisms. MATERIAL: Constructed murine models of gouty arthritis and conducted THP-1 cell experiments. TREATMENT: 1 mg of MSU crystals injected into mice ankle joints for a 72-h intervention. After a 3-h pre-treatment with Sirt1-specific inhibitor (EX527) and agonist (SRT2104), inflammation was induced for 21 h using lipopolysaccharide (LPS) plus MSU crystals. METHODS: We assessed gouty arthritis severity through joint inflammation index, swelling, and hematoxylin and eosin (H&E) staining, and measured CD68 mononuclear macrophages and Sirt1 expression in synovial tissue via immunohistochemistry. ELISA, NO assay, RT-qPCR, Flow cytometry, and Western blot were utilized to examine macrophage inflammatory factors, polarization, reactive oxygen species(ROS), MAPK/NF-κB/AP-1 and Nrf2/HO-1 pathways proteins. RESULTS: Significant joint swelling, synovial tissue edema, and inflammatory cell infiltration were observed. CD68 mononuclear macrophages and Sirt1 expression were elevated in synovium. Sirt1 activation decreased inflammatory factors, M1 polarization, and ROS generation. Sirt1 activation reduced p38/JNK phosphorylation, thereby inhibiting downstream NF-κB p65/AP-1 and enhancing Nrf2/HO-1, thus suppressing inflammation. CONCLUSIONS: Sirt1 alleviates M1 macrophage polarization and inflammation in gouty arthritis by inhibiting the MAPK/NF-κB/AP-1 pathway and activating the Nrf2/HO-1 pathway. Thus, activating Sirt1 may provide a new therapeutic target for gouty arthritis.


Sujet(s)
Goutte articulaire , Heme oxygenase-1 , Macrophages , Facteur-2 apparenté à NF-E2 , Facteur de transcription NF-kappa B , Sirtuine-1 , Facteur de transcription AP-1 , Animaux , Goutte articulaire/traitement médicamenteux , Goutte articulaire/métabolisme , Goutte articulaire/immunologie , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/immunologie , Facteur-2 apparenté à NF-E2/métabolisme , Humains , Mâle , Facteur de transcription NF-kappa B/métabolisme , Heme oxygenase-1/métabolisme , Souris , Facteur de transcription AP-1/métabolisme , Cellules THP-1 , Souris de lignée C57BL , Inflammation , Transduction du signal/effets des médicaments et des substances chimiques , Lipopolysaccharides/pharmacologie , Carbazoles , Protéines membranaires
15.
Genome Res ; 34(5): 680-695, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38777607

RÉSUMÉ

Gastric cancer (GC) is the fifth most common cancer worldwide and is a heterogeneous disease. Among GC subtypes, the mesenchymal phenotype (Mes-like) is more invasive than the epithelial phenotype (Epi-like). Although gene expression of the epithelial-to-mesenchymal transition (EMT) has been studied, the regulatory landscape shaping this process is not fully understood. Here we use ATAC-seq and RNA-seq data from a compendium of GC cell lines and primary tumors to detect drivers of regulatory state changes and their transcriptional responses. Using the ATAC-seq data, we developed a machine learning approach to determine the transcription factors (TFs) regulating the subtypes of GC. We identified TFs driving the mesenchymal (RUNX2, ZEB1, SNAI2, AP-1 dimer) and the epithelial (GATA4, GATA6, KLF5, HNF4A, FOXA2, GRHL2) states in GC. We identified DNA copy number alterations associated with dysregulation of these TFs, specifically deletion of GATA4 and amplification of MAPK9 Comparisons with bulk and single-cell RNA-seq data sets identified activation toward fibroblast-like epigenomic and expression signatures in Mes-like GC. The activation of this mesenchymal fibrotic program is associated with differentially accessible DNA cis-regulatory elements flanking upregulated mesenchymal genes. These findings establish a map of TF activity in GC and highlight the role of copy number driven alterations in shaping epigenomic regulatory programs as potential drivers of GC heterogeneity and progression.


Sujet(s)
Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Apprentissage machine , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Transition épithélio-mésenchymateuse/génétique , Facteur de transcription AP-1/métabolisme , Facteur de transcription AP-1/génétique , Lignée cellulaire tumorale , Fibrose/génétique , Sous-unité alpha 1 du facteur CBF/génétique , Sous-unité alpha 1 du facteur CBF/métabolisme , Variations de nombre de copies de segment d'ADN , Sous-unité alpha 2 du facteur CBF
16.
PLoS One ; 19(4): e0297847, 2024.
Article de Anglais | MEDLINE | ID: mdl-38635533

RÉSUMÉ

The uterine muscular layer, or myometrium, undergoes profound changes in global gene expression during its progression from a quiescent state during pregnancy to a contractile state at the onset of labor. In this study, we investigate the role of SOX family transcription factors in myometrial cells and provide evidence for the role of SOX4 in regulating labor-associated genes. We show that Sox4 has elevated expression in the murine myometrium during a term laboring process and in two mouse models of preterm labor. Additionally, SOX4 differentially affects labor-associated gene promoter activity in cooperation with activator protein 1 (AP-1) dimers. SOX4 exerted no effect on the Gja1 promoter; a JUND-specific activation effect at the Fos promoter; a positive activation effect on the Mmp11 promoter with the AP-1 dimers; and surprisingly, we noted that the reporter expression of the Ptgs2 promoter in the presence of JUND and FOSL2 was repressed by the addition of SOX4. Our data indicate SOX4 may play a diverse role in regulating gene expression in the laboring myometrium in cooperation with AP-1 factors. This study enhances our current understanding of the regulatory network that governs the transcriptional changes associated with the onset of labor and highlights a new molecular player that may contribute to the labor transcriptional program.


Sujet(s)
Travail obstétrical , Myomètre , Animaux , Femelle , Souris , Grossesse , Travail obstétrical/génétique , Travail obstétrical/métabolisme , Myomètre/métabolisme , Régions promotrices (génétique) , Facteur de transcription AP-1/génétique , Facteur de transcription AP-1/métabolisme , Utérus/métabolisme
17.
Proc Natl Acad Sci U S A ; 121(18): e2404188121, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38657045

RÉSUMÉ

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death. HCC incidence is on the rise, while treatment options remain limited. Thus, a better understanding of the molecular pathways involved in HCC development has become a priority to guide future therapies. While previous studies implicated the Activator Protein-1 (AP-1) (Fos/Jun) transcription factor family members c-Fos and c-Jun in HCC formation, the contribution of Fos-related antigens (Fra-) 1 and 2 is unknown. Here, we show that hepatocyte-restricted expression of a single chain c-Jun~Fra-2 protein, which functionally mimics the c-Jun/Fra-2 AP-1 dimer, results in spontaneous HCC formation in c-Jun~Fra-2hep mice. Several hallmarks of human HCC, such as cell cycle dysregulation and the expression of HCC markers are observed in liver tumors arising in c-Jun~Fra-2hep mice. Tumorigenesis occurs in the context of mild inflammation, low-grade fibrosis, and Pparγ-driven dyslipidemia. Subsequent analyses revealed increased expression of c-Myc, evidently under direct regulation by AP-1 through a conserved distal 3' enhancer. Importantly, c-Jun~Fra-2-induced tumors revert upon switching off transgene expression, suggesting oncogene addiction to the c-Jun~Fra-2 transgene. Tumors escaping reversion maintained c-Myc and c-Myc target gene expression, likely due to increased c-Fos. Interfering with c-Myc in established tumors using the Bromodomain and Extra-Terminal motif inhibitor JQ-1 diminished liver tumor growth in c-Jun~Fra-2 mutant mice. Thus, our data establish c-Jun~Fra-2hep mice as a model to study liver tumorigenesis and identify the c-Jun/Fra-2-Myc interaction as a potential target to improve HCC patient stratification and/or therapy.


Sujet(s)
Carcinome hépatocellulaire , Antigène-2 apparenté à fos , Tumeurs du foie , Protéines proto-oncogènes c-fos , Protéines proto-oncogènes c-jun , Protéines proto-oncogènes c-myc , Facteur de transcription AP-1 , Animaux , Facteur de transcription AP-1/métabolisme , Facteur de transcription AP-1/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Souris , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-fos/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Protéines proto-oncogènes c-jun/métabolisme , Antigène-2 apparenté à fos/métabolisme , Antigène-2 apparenté à fos/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Humains , Hépatocytes/métabolisme , Multimérisation de protéines , Régulation de l'expression des gènes tumoraux , Souris transgéniques
18.
Biomed Pharmacother ; 175: 116633, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38670049

RÉSUMÉ

Sepsis is a severe inflammatory disorder that can lead to life-threatening multiple organ injury. Lipopolysaccharide (LPS)-induced inflammation is the leading cause of multiple organ failure in sepsis. This study aimed to explore the effect of a novel agent, 2-(4-hydroxy-3-methoxyphenyl)-benzothiazole (YL-109), on LPS-induced multiple organ injury and the molecular mechanisms underlying these processes. The results showed that YL-109 protected against LPS-induced high mortality, cardiac dysfunction, pulmonary and intestinal injury through inhibiting the proinflammatory response, NLRP3 expression and pyroptosis-associated indicators in mouse tissues. YL-109 suppressed LPS-initiated cytokine release, pyroptosis and pyroptosis-related protein expression in HL-1, IEC-6 and MLE-12 cells, which was consistent with the results of the in vivo experiments. Mechanistically, YL-109 reduces phosphorylated ERK (extracellular signal-regulated kinase) levels and NF-κB activation, which are achieved through upregulating CHIP (carboxy terminus of Hsc70-interacting protein) expression, thereby inhibiting c-Jun and c-Fos activation as well as NLRP3 expression. As an E3 ligase, CHIP overexpression obviously promoted the degradation of phosphorylated ERK and inhibited the expression of NF-κB-mediated NLRP3 in cells stimulated with LPS. The protective effects of YL-109 against cardiac, pulmonary and intestinal damage, inflammation and pyroptosis caused by LPS were eliminated in CHIP knockout mice. Our results not only reveal the protective effect and molecular mechanism of YL-109 against LPS-mediated organs damage but also provide additional insights into the effect of CHIP on negatively regulating pyroptosis and inflammatory pathways.


Sujet(s)
Lipopolysaccharides , Souris de lignée C57BL , Défaillance multiviscérale , Pyroptose , Sepsie , Facteur de transcription AP-1 , Ubiquitin-protein ligases , Régulation positive , Animaux , Pyroptose/effets des médicaments et des substances chimiques , Sepsie/complications , Sepsie/traitement médicamenteux , Sepsie/métabolisme , Souris , Régulation positive/effets des médicaments et des substances chimiques , Défaillance multiviscérale/métabolisme , Défaillance multiviscérale/prévention et contrôle , Défaillance multiviscérale/traitement médicamenteux , Mâle , Facteur de transcription AP-1/métabolisme , Ubiquitin-protein ligases/métabolisme , Lignée cellulaire , Benzothiazoles/pharmacologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
19.
Sci Adv ; 10(14): eadl5012, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38569033

RÉSUMÉ

The ß-coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the global COVID-19 pandemic. Coronaviral Envelope (E) proteins are pentameric viroporins that play essential roles in assembly, release, and pathogenesis. We developed a nondisruptive tagging strategy for SARS-CoV-2 E and find that, at steady state, it localizes to the Golgi and to lysosomes. We identify sequences in E, conserved across Coronaviridae, responsible for endoplasmic reticulum-to-Golgi export, and relate this activity to interaction with COP-II via SEC24. Using proximity biotinylation, we identify an ADP ribosylation factor 1/adaptor protein-1 (ARFRP1/AP-1)-dependent pathway allowing Golgi-to-lysosome trafficking of E. We identify sequences in E that bind AP-1, are conserved across ß-coronaviruses, and allow E to be trafficked from Golgi to lysosomes. We show that E acts to deacidify lysosomes and, by developing a trans-complementation assay for SARS-CoV-2 structural proteins, that lysosomal delivery of E and its viroporin activity is necessary for efficient viral replication and release.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , SARS-CoV-2/métabolisme , Protéines de l'enveloppe virale/métabolisme , Facteur de transcription AP-1/métabolisme , Pandémies , Réplication virale , Lysosomes/métabolisme , Facteurs d'ADP-ribosylation/métabolisme
20.
J Cell Biol ; 223(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38578286

RÉSUMÉ

The AP-1 adaptor complex is found in all eukaryotes, but it has been implicated in different pathways in different organisms. To look directly at AP-1 function, we generated stably transduced HeLa cells coexpressing tagged AP-1 and various tagged membrane proteins. Live cell imaging showed that AP-1 is recruited onto tubular carriers trafficking from the Golgi apparatus to the plasma membrane, as well as onto transferrin-containing early/recycling endosomes. Analysis of single AP-1 vesicles showed that they are a heterogeneous population, which starts to sequester cargo 30 min after exit from the ER. Vesicle capture showed that AP-1 vesicles contain transmembrane proteins found at the TGN and early/recycling endosomes, as well as lysosomal hydrolases, but very little of the anterograde adaptor GGA2. Together, our results support a model in which AP-1 retrieves proteins from post-Golgi compartments back to the TGN, analogous to COPI's role in the early secretory pathway. We propose that this is the function of AP-1 in all eukaryotes.


Sujet(s)
Appareil de Golgi , Protéines membranaires , Transport des protéines , Facteur de transcription AP-1 , Humains , Protéines adaptatrices du transport vésiculaire/métabolisme , Membrane cellulaire/métabolisme , Endosomes/génétique , Endosomes/métabolisme , Appareil de Golgi/génétique , Appareil de Golgi/métabolisme , Cellules HeLa , Protéines membranaires/métabolisme , Réseau trans-golgien/métabolisme , Facteur de transcription AP-1/génétique , Facteur de transcription AP-1/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...