Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.121
Filtrer
1.
Aging (Albany NY) ; 16(12): 10348-10365, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38874497

RÉSUMÉ

Exploring the molecular mechanisms of PD-1/PDL-1 blockade for non-small cell lung cancer (NSCLC) would facilitate understanding for tumor microenvironment (TME) and development of individualized medicine. To date, biomarkers of response to PD-1 blockade therapy were still limited. In this study, we hypothesize that cell type in the tumor microenvironment can influence the effect of PD-1 blockade immunotherapy through specific genes. Therefore, we re-analyze the single-cell RNA sequencing data and validation in tissue from lung adenocarcinoma patients. Dynamic changes of cellular subpopulation were observed after anti-PD-1 immunotherapy among TMEs between primary/metastasis or good/poor response patients. Non-exhausted CD8 T cells and dysregulated genes were observed in responsing patients from PD-1 blockade therapy. Among all changed genes, JUN, involved in PD-1 blockade immunotherapy pathway, and could be considered as a PD-1 responsing biomarker.


Sujet(s)
Tumeurs du poumon , Récepteur-1 de mort cellulaire programmée , Analyse sur cellule unique , Microenvironnement tumoral , Humains , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/génétique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Analyse de séquence d'ARN , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Régulation de l'expression des gènes tumoraux , Marqueurs biologiques tumoraux/génétique , Protéines proto-oncogènes c-jun/génétique , Protéines proto-oncogènes c-jun/métabolisme , Immunothérapie/méthodes , Mâle , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/anatomopathologie , Femelle
2.
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
4.
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
5.
J Appl Biomed ; 22(2): 107-114, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38912866

RÉSUMÉ

Diffuse large B-cell lymphoma (DLBCL) stands out as the most common type of malignant cancer, representing the majority of cases of non-Hodgkin's lymphoma. Ethyl pyruvate (EP) is a derivative of pyruvic acid and found to have potent anti-tumor properties. Despite its potential benefits, the impact of EP on DLBCL remains ambiguous. Our objective is to elucidate the role of EP in modulating the development of DLBCL. Analysis of cholecystokinin-8 (CCK-8) revealed that treatment with EP significantly diminished the viability of DLBCL cells. Furthermore, EP administration suppressed colony formation and hindered cell adhesion and invasion in DLBCL cells. Examination of cell cycle progression showed that EP treatment induced arrest at the G1 phase and subsequently reduced the S phase population in DLBCL cells. EP treatment consistently exhibited apoptosis-inducing properties in Annexin-V assays, and notably downregulated the expression of Bcl-2 while increasing levels of proapoptotic cleaved caspase 3 and BAX in DLBCL cells. Additionally, EP treatment decreased the overexpression of c-Jun in c-Jun-transfected DLBCL cells. Further, EP demonstrated DNA-damaging effects in TUNEL assays. In vivo, xenograft animal models revealed that EP treatment significantly mitigated DLBCL tumor growth and suppressed DLBCL cell adhesion to bone marrow stromal cells. In summary, these findings suggest that EP mitigates DLBCL progression by inducing apoptosis, inducing cell cycle arrest, and promoting DNA damage.


Sujet(s)
Adhérence cellulaire , Prolifération cellulaire , Lymphome B diffus à grandes cellules , Pyruvates , Pyruvates/pharmacologie , Pyruvates/usage thérapeutique , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/anatomopathologie , Humains , Animaux , Adhérence cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Apoptose/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-jun/métabolisme , Protéines proto-oncogènes c-jun/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Cells ; 13(9)2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38727318

RÉSUMÉ

CXCR4, JUNB and PD-L1 are implicated in cancer progression and metastasis. The current study investigated these biomarkers in CTCs isolated from metastatic prostate cancer (mPCa) patients at the RNA and protein levels. CTCs were isolated from 48 mPCa patients using the Ficoll density gradient and ISET system (17 out of 48). The (CK/PD-L1/CD45) and (CK/CXCR4/JUNB) phenotypes were identified using two triple immunofluorescence stainings followed by VyCAP platform analysis. Molecular analysis was conducted with an EpCAM-dependent method for 25/48 patients. CK-8, CK-18, CK-19, JUNB, CXCR4, PD-L1, and B2M (reference gene) were analyzed with RT-qPCR. The (CK+/PD-L1+/CD45-) and the (CK+/CXCR4+/JUNB+) were the most frequent phenotypes (61.1% and 62.5%, respectively). Furthermore, the (CK+/CXCR4+/JUNB-) phenotype was correlated with poorer progression-free survival [(PFS), HR: 2.5, p = 0.049], while the (CK+/PD-L1+/CD45-) phenotype was linked to decreased overall survival [(OS), HR: 262.7, p = 0.007]. Molecular analysis revealed that 76.0% of the samples were positive for CK-8,18, and 19, while 28.0% were positive for JUNB, 44.0% for CXCR4, and 48.0% for PD-L1. Conclusively, CXCR4, JUNB, and PD-L1 were highly expressed in CTCs from mPCa patients. The CXCR4 protein expression was associated with poorer PFS, while PD-L1 was correlated with decreased OS, providing new biomarkers with potential clinical relevance.


Sujet(s)
Antigène CD274 , Cellules tumorales circulantes , Tumeurs de la prostate , Récepteurs CXCR4 , Sujet âgé , Humains , Mâle , Adulte d'âge moyen , Antigène CD274/métabolisme , Antigène CD274/génétique , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Régulation de l'expression des gènes tumoraux , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Protéines proto-oncogènes c-jun/métabolisme , Récepteurs CXCR4/métabolisme , Récepteurs CXCR4/génétique
7.
Toxicology ; 505: 153843, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38801936

RÉSUMÉ

Benzene, a widely used industrial chemical, has been clarified to cause hematotoxicity. Our previous study suggested that miR-451a may play a role in benzene-induced impairment of erythroid differentiation. However, the mechanism underlying remains unclear. In this study, we explored the role of miR-451a and its underlying mechanisms in hydroquinone (HQ)-induced suppression of erythroid differentiation in K562 cells. 0, 1.0, 2.5, 5.0, 10.0, and 50 µM HQ treatment of K562 cells resulted in a dose-dependent inhibition of erythroid differentiation, as well as the expression of miR-451a. Bioinformatics analysis was conducted to predict potential target genes of miR-451a and dual-luciferase reporter assays confirmed that miR-451a can directly bind to the 3'-UTR regions of BATF, SETD5, and ARHGEF3 mRNAs. We further demonstrated that over-expression or down-regulation of miR-451a altered the expression of BATF, SETD5, and ARHGEF3, and also modified erythroid differentiation. In addition, BATF, SETD5, and ARHGEF3 were verified to play a role in HQ-induced inhibition of erythroid differentiation in this study. Knockdown of SETD5 and ARHGEF3 reversed HQ-induced suppression of erythroid differentiation while knockdown of BATF had the opposite effect. On the other hand, we also identified c-Jun as a potential transcriptional regulator of miR-451a. Forced expression of c-Jun increased miR-451a expression and reversed the inhibition of erythroid differentiation induced by HQ, whereas knockdown of c-Jun had the opposite effect. And the binding site of c-Jun and miR-451a was verified by dual-luciferase reporter assay. Collectively, our findings indicate that miR-451a and its downstream targets BATF, SETD5, and ARHGEF3 are involved in HQ-induced erythroid differentiation disorder, and c-Jun regulates miR-451a as a transcriptional regulator in this process.


Sujet(s)
Facteurs de transcription à motif basique et à glissière à leucines , Différenciation cellulaire , microARN , Rho guanine nucleotide exchange factors , Humains , microARN/génétique , microARN/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Facteurs de transcription à motif basique et à glissière à leucines/génétique , Facteurs de transcription à motif basique et à glissière à leucines/métabolisme , Cellules K562 , Rho guanine nucleotide exchange factors/génétique , Rho guanine nucleotide exchange factors/métabolisme , Cellules érythroïdes/effets des médicaments et des substances chimiques , Cellules érythroïdes/métabolisme , Protéines proto-oncogènes c-jun/métabolisme , Protéines proto-oncogènes c-jun/génétique , Methyltransferases/génétique , Methyltransferases/métabolisme
8.
Biochem Biophys Res Commun ; 719: 150042, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38761633

RÉSUMÉ

BACKGROUND & AIMS: Psychosocial stress has become an unavoidable part of life, which was reported to promote tumor development. Chronic stress significantly promotes the norepinephrine (NE) secretion and the expression of leptin receptor (LEPR), leading to tumor invasion, metastasis, and proliferation. However, the mechanism of chronic stress-induced tumor proliferation remains unclear. METHODS: To reveal the effect of chronic stress on tumor proliferation, subcutaneous tumor models combined with chronic restraint stress (CRS) were established. Combined with the transcript omics database of liver cancer patients, the target pathways were screened and further verified by in vitro experiments. RESULTS: The results showed that the CRS with subcutaneous tumor transplantation (CRS + tumor) group exhibited significantly larger tumor sizes than the subcutaneous tumor transplantation (tumor) group. Compared with the tumor group, CRS obviously increased the mRNA levels of LEPR, FOS, and JUNB of tumor tissues in the CRS + tumor group. Furthermore, the treatment with norepinephrine (NE) significantly elevated the survival rate of H22 cells and enhanced the expression of LEPR, FOS, and JUNB in vitro. Silencing LEPR significantly reduced the expression of FOS and JUNB, accompanied by a decrease in H22 cell viability. CONCLUSIONS: Our study demonstrated that CRS activates the LEPR-FOS-JUNB signaling pathway by NE, aggravating tumor development. These findings might provide a scientific foundation for investigating the underlying pathological mechanisms of tumors in response to chronic stress.


Sujet(s)
Prolifération cellulaire , Protéines proto-oncogènes c-fos , Récepteurs à la leptine , Transduction du signal , Récepteurs à la leptine/métabolisme , Récepteurs à la leptine/génétique , Animaux , Lignée cellulaire tumorale , Humains , Souris , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-fos/génétique , Mâle , Protéines proto-oncogènes c-jun/métabolisme , Stress psychologique/métabolisme , Contention physique , Norépinéphrine/métabolisme , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Souris de lignée BALB C
9.
Mol Cancer ; 23(1): 114, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38811984

RÉSUMÉ

BACKGROUND: Prostate cancer develops through malignant transformation of the prostate epithelium in a stepwise, mutation-driven process. Although activator protein-1 transcription factors such as JUN have been implicated as potential oncogenic drivers, the molecular programs contributing to prostate cancer progression are not fully understood. METHODS: We analyzed JUN expression in clinical prostate cancer samples across different stages and investigated its functional role in a Pten-deficient mouse model. We performed histopathological examinations, transcriptomic analyses and explored the senescence-associated secretory phenotype in the tumor microenvironment. RESULTS: Elevated JUN levels characterized early-stage prostate cancer and predicted improved survival in human and murine samples. Immune-phenotyping of Pten-deficient prostates revealed high accumulation of tumor-infiltrating leukocytes, particularly innate immune cells, neutrophils and macrophages as well as high levels of STAT3 activation and IL-1ß production. Jun depletion in a Pten-deficient background prevented immune cell attraction which was accompanied by significant reduction of active STAT3 and IL-1ß and accelerated prostate tumor growth. Comparative transcriptome profiling of prostate epithelial cells revealed a senescence-associated gene signature, upregulation of pro-inflammatory processes involved in immune cell attraction and of chemokines such as IL-1ß, TNF-α, CCL3 and CCL8 in Pten-deficient prostates. Strikingly, JUN depletion reversed both the senescence-associated secretory phenotype and senescence-associated immune cell infiltration but had no impact on cell cycle arrest. As a result, JUN depletion in Pten-deficient prostates interfered with the senescence-associated immune clearance and accelerated tumor growth. CONCLUSIONS: Our results suggest that JUN acts as tumor-suppressor and decelerates the progression of prostate cancer by transcriptional regulation of senescence- and inflammation-associated genes. This study opens avenues for novel treatment strategies that could impede disease progression and improve patient outcomes.


Sujet(s)
Évolution de la maladie , Phosphohydrolase PTEN , Tumeurs de la prostate , Microenvironnement tumoral , Mâle , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Animaux , Souris , Humains , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Microenvironnement tumoral/immunologie , Phénotype sécrétoire associé à la sénescence , Protéines proto-oncogènes c-jun/métabolisme , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes , Vieillissement de la cellule/génétique , Modèles animaux de maladie humaine
10.
Cell Signal ; 120: 111179, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38640980

RÉSUMÉ

S100P, a member of the S100 calcium-binding protein family, is closely associated with abnormal proliferation, invasion, and metastasis of various cancers. However, its role in the lung adenocarcinoma (LUAD) tumor microenvironment (TME) remains unclear. In this study, we observed specific expression of S100P on tumor cells in LUAD patients through tissue immunofluorescence analysis. Furthermore, this expression was strongly correlated with the recruitment and polarization of tumor-associated macrophages (TAMs). Bioinformatics analysis revealed that high S100P expression is associated with poorer overall survival in LUAD patients. Subsequently, a subcutaneous mouse model demonstrated that S100P promotes recruitment and polarization of TAMs towards the M2 type. Finally, in vitro studies on LUAD cells revealed that S100P enhances the secretion of chemokines and polarizing factors by activating the PKA/c-Jun pathway, which is implicated in TAM recruitment and polarization towards the M2 phenotype. Moreover, inhibition of c-Jun expression impedes the ability of TAMs to infiltrate and polarize towards the M2 phenotype. In conclusion, our study demonstrates that S100P facilitates LUAD cells growth by recruiting M2 TAMs through PKA/c-Jun signaling, resulting in the production of various cytokines. Considering these findings, S100P holds promise as an important diagnostic marker and potential therapeutic target for LUAD.


Sujet(s)
Protéines de liaison au calcium , Macrophages associés aux tumeurs , Humains , Animaux , Macrophages associés aux tumeurs/métabolisme , Souris , Protéines de liaison au calcium/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Protéines tumorales/métabolisme , Protéines tumorales/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Lignée cellulaire tumorale , Microenvironnement tumoral , Transduction du signal , Femelle , Mâle , Évolution de la maladie , Protéines proto-oncogènes c-jun/métabolisme , Prolifération cellulaire , Polarité de la cellule
11.
Oncogene ; 43(21): 1608-1619, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38565943

RÉSUMÉ

Cancer cells employ adaptive mechanisms to survive various stressors, including genotoxic drugs. Understanding the factors promoting survival is crucial for developing effective treatments. In this study, we unveil a previously unexplored long non-coding RNA, JUNI (JUN-DT, LINC01135), which is upregulated by genotoxic drugs through the activation of stress-activated MAPKs, JNK, and p38 and consequently exerts positive control over the expression of its adjacent gene product c-Jun, a well-known oncoprotein, which transduces signals to multiple transcriptional outputs. JUNI regulates cellular migration and has a crucial role in conferring cellular resistance to chemotherapeutic drugs or UV radiation. Depletion of JUNI markedly increases the sensitivity of cultured cells and spheroids to chemotherapeutic agents. We identified 57 proteins interacting with JUNI. The activity of one of them the MAPK phosphatase and inhibitor, DUSP14, is counteracted by JUNI, thereby, facilitating efficient JNK phosphorylation and c-Jun induction when cells are exposed to UV radiation. The antagonistic interplay with DUSP14 contributes not only to c-Jun induction but also augments the survival of UV-exposed cells. In summary, we introduce JUNI as a novel stress-inducible regulator of c-Jun, positioning it as a potential target for enhancing the sensitivity of cancer cells to chemotherapy.


Sujet(s)
Mouvement cellulaire , Survie cellulaire , Dual-specificity phosphatases , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Dual-specificity phosphatases/métabolisme , Dual-specificity phosphatases/génétique , Mouvement cellulaire/génétique , Survie cellulaire/effets des radiations , Survie cellulaire/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-jun/métabolisme , Protéines proto-oncogènes c-jun/génétique , Lignée cellulaire tumorale , Rayons ultraviolets/effets indésirables , Système de signalisation des MAP kinases/génétique , Régulation de l'expression des gènes tumoraux , JNK Mitogen-Activated Protein Kinases/métabolisme
12.
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
13.
JCI Insight ; 9(5)2024 Mar 08.
Article de Anglais | MEDLINE | ID: mdl-38456508

RÉSUMÉ

IL-33 is a cytokine central to type 2 immune pathology in chronic airway disease. This cytokine is abundantly expressed in the respiratory epithelium and increased in disease, but how expression is regulated is undefined. Here we show that increased IL33 expression occurs from multiple noncanonical promoters in human chronic obstructive pulmonary disease (COPD), and it facilitates production of alternatively spliced isoforms in airway cells. We found that phorbol 12-myristate 13-acetate (PMA) can activate IL33 promoters through protein kinase C in primary airway cells and lines. Transcription factor (TF) binding arrays combined with RNA interference identified activator protein (AP) TFs as regulators of baseline and induced IL33 promoter activity. ATAC-Seq and ChIP-PCR identified chromatin accessibility and differential TF binding as additional control points for transcription from noncanonical promoters. In support of a role for these TFs in COPD pathogenesis, we found that AP-2 (TFAP2A, TFAP2C) and AP-1 (FOS and JUN) family members are upregulated in human COPD specimens. This study implicates integrative and pioneer TFs in regulating IL33 promoters and alternative splicing in human airway basal cells. Our work reveals a potentially novel approach for targeting IL-33 in development of therapeutics for COPD.


Sujet(s)
Interleukine-33 , Broncho-pneumopathie chronique obstructive , Humains , Interleukine-33/génétique , Protéines proto-oncogènes c-fos/génétique , Protéines proto-oncogènes c-jun/métabolisme , Broncho-pneumopathie chronique obstructive/génétique , Facteur de transcription AP-1/génétique , Facteur de transcription AP-1/métabolisme
14.
Cell Death Dis ; 15(3): 180, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38429278

RÉSUMÉ

Gasdermin E (GSDME) has recently been identified as a critical executioner to mediate pyroptosis. While epidermal keratinocytes can initiate GSDME-mediated pyroptosis, the role of keratinocyte GSDME in psoriatic dermatitis remains poorly characterized. Through analysis of GEO datasets, we found elevated GSDME levels in psoriatic lesional skin. Additionally, GSDME levels correlated with both psoriasis severity and response to biologics treatments. Single-cell RNA sequencing (scRNA-seq) from a GEO dataset revealed GSDME upregulation in keratinocytes of psoriasis patients. In the imiquimod (IMQ)-induced psoriasis-like dermatitis mouse model, both full-length and cleaved forms of caspase-3 and GSDME were elevated in the epidermis. Abnormal proliferation and differentiation of keratinocytes and dermatitis were attenuated in Gsdme-/- mice and keratinocyte-specific Gsdme conditional knockout mice after IMQ stimulation. Exposure of keratinocytes to mixed cytokines (M5), mimicking psoriatic conditions, led to GSDME cleavage. Moreover, the interaction between GSDME-FL and p65 or c-jun was significantly increased after M5 stimulation. GSDME knockdown inhibited nuclear translocation of p65 and c-jun and decreased upregulation of psoriatic inflammatory mediators such as IL1ß, CCL20, CXCL1, CXCL8, S100A8, and S100A9 in M5-challenged keratinocytes. In conclusion, GSDME in keratinocytes contributes to the pathogenesis and progression of psoriasis, potentially in a pyroptosis-independent manner by interacting and promoting translocation of p65 and c-jun. These findings suggest that keratinocyte GSDME could serve as a potential therapeutic target for psoriasis treatment.


Sujet(s)
Dermatite , Gasdermines , Psoriasis , Animaux , Humains , Souris , Dermatite/métabolisme , Dermatite/anatomopathologie , Gasdermines/métabolisme , Imiquimod/effets indésirables , Inflammation/anatomopathologie , Kératinocytes/anatomopathologie , Psoriasis/métabolisme , Psoriasis/anatomopathologie , Facteur de transcription RelA/métabolisme , Protéines proto-oncogènes c-jun/métabolisme
15.
Cancer Lett ; 587: 216731, 2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38369005

RÉSUMÉ

Therapy resistance and metastatic progression jointly determine the fatal outcome of cancer, therefore, elucidating their crosstalk may provide new opportunities to improve therapeutic efficacy and prevent recurrence and metastasis in esophageal squamous cell carcinoma (ESCC). Here, we have established radioresistant ESCC cells with the remarkable metastatic capacity, and identified miR-494-3p (miR494) as a radioresistant activator. Mechanistically, we demonstrated that cullin 3 (CUL3) is a direct target of miR494, which is transcriptionally regulated by JunD, and highlighted that JunD-miR494-CUL3 axis promotes radioresistance and metastasis by facilitating epithelial-mesenchymal transition (EMT) and restraining programmed cell death 1 ligand 1 (PD-L1) degradation. In clinical specimens, miR494 is significantly up-regulated and positively associated with T stage and lymph node metastasis in ESCC tissues and serum. Notably, patients with higher serum miR494 expression have poor prognosis, and patients with higher CUL3 expression have more conventional dendritic cells (cDCs) and plasmacytoid DCs (pDCs), less cancer-associated fibroblasts (CAF2/4), and tumor endothelial cells (TEC2/3) infiltration than patients with lower CUL3 expression, suggesting that CUL3 may be involved in tumor microenvironment (TME). Overall, miR494 may serve as a potential prognostic predictor and therapeutic target, providing a promising strategy for ESCC treatment.


Sujet(s)
Carcinome épidermoïde , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , microARN , Humains , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/radiothérapie , Antigène CD274/génétique , Antigène CD274/métabolisme , Carcinome épidermoïde/génétique , Carcinome épidermoïde/radiothérapie , Carcinome épidermoïde/métabolisme , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/radiothérapie , Tumeurs de l'oesophage/métabolisme , Cellules endothéliales/métabolisme , Pronostic , Transition épithélio-mésenchymateuse , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Mouvement cellulaire , Microenvironnement tumoral , Protéines proto-oncogènes c-jun/métabolisme , Cullines/génétique
16.
Cell Death Differ ; 31(2): 136-149, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38104183

RÉSUMÉ

Fos-related antigen-2 (Fra-2) is the most recently discovered member of the Fos family and, by dimerizing with Jun proteins, forms the activator protein 1 (AP-1) transcription factor. By inducing or repressing the transcription of several target genes, Fra-2 is critically involved in the modulation of cell response to a variety of extracellular stimuli, stressors and intracellular changes. In physiological conditions, Fra-2 has been found to be ubiquitously expressed in human cells, regulating differentiation and homeostasis of bone, muscle, nervous, lymphoid and other tissues. While other AP-1 members, like Jun and Fos, are well characterized, studies of Fra-2 functions in cancer are still at an early stage. Due to the lack of a trans-activating domain, which is present in other Fos proteins, it has been suggested that Fra-2 might inhibit cell transformation, eventually exerting an anti-tumor effect. In human malignancies, however, Fra-2 activity is enhanced (or induced) by dysregulation of microRNAs, oncogenes and extracellular signaling, suggesting a multifaceted role. Therefore, Fra-2 can promote or prevent transformation, proliferation, migration, epithelial-mesenchymal transition, drug resistance and metastasis formation in a tumor- and context-dependent manner. Intriguingly, recent data reports that Fra-2 is also expressed in cancer associated cells, contributing to the intricate crosstalk between neoplastic and non-neoplastic cells, that leads to the evolution and remodeling of the tumor microenvironment. In this review we summarize three decades of research on Fra-2, focusing on its oncogenic and anti-oncogenic effects in tumor progression and dissemination.


Sujet(s)
Tumeurs , Facteur de transcription AP-1 , Humains , Transformation cellulaire néoplasique/génétique , Antigène-2 apparenté à fos/génétique , Antigène-2 apparenté à fos/métabolisme , Régulation de l'expression des gènes , Tumeurs/génétique , Protéines proto-oncogènes c-fos/génétique , Protéines proto-oncogènes c-jun/métabolisme , Facteur de transcription AP-1/métabolisme , Microenvironnement tumoral
17.
Stem Cell Res Ther ; 14(1): 371, 2023 12 18.
Article de Anglais | MEDLINE | ID: mdl-38110996

RÉSUMÉ

BACKGROUND: Morbidity and mortality associated with cardiovascular diseases, such as myocardial infarction, stem from the inability of terminally differentiated cardiomyocytes to regenerate, and thus repair the damaged myocardial tissue structure. The molecular biological mechanisms behind the lack of regenerative capacity for those cardiomyocytes remains to be fully elucidated. Recent studies have shown that c-Jun serves as a cell cycle regulator for somatic cell fates, playing a key role in multiple molecular pathways, including the inhibition of cellular reprogramming, promoting angiogenesis, and aggravation of cardiac hypertrophy, but its role in cardiac development is largely unknown. This study aims to delineate the role of c-Jun in promoting early-stage cardiac differentiation. METHODS: The c-Jun gene in mouse embryonic stem cells (mESCs) was knocked out with CRISPR-Cas9, and the hanging drop method used to prepare the resulting embryoid bodies. Cardiac differentiation was evaluated up to 9 days after c-Jun knockout (ko) via immunofluorescence, flow cytometric, and qPCR analyses. RESULTS: Compared to the wild-type control group, obvious beating was observed among the c-Jun-ko mESCs after 6 days, which was also associated with significant increases in myocardial marker expression. Additionally, markers associated with mesoderm and endoderm cell layer development, essential for further differentiation of ESCs into cardiomyocytes, were also up-regulated in the c-Jun-ko cell group. CONCLUSIONS: Knocking out c-Jun directs ESCs toward a meso-endodermal cell lineage fate, in turn leading to generation of beating myocardial cells. Thus, c-Jun plays an important role in regulating early cardiac cell development.


Sujet(s)
Corps embryoïdes , Myocytes cardiaques , Protéines proto-oncogènes c-jun , Animaux , Souris , Différenciation cellulaire , Lignage cellulaire , Cellules souches embryonnaires de souris , Myocarde , Myocytes cardiaques/métabolisme , Protéines proto-oncogènes c-jun/métabolisme
18.
Genome Biol ; 24(1): 268, 2023 Nov 27.
Article de Anglais | MEDLINE | ID: mdl-38012744

RÉSUMÉ

BACKGROUND: Enhancer dysregulation is one of the important features for cancer cells. Enhancers enriched with H3K4me3 have been implicated to play important roles in cancer. However, their detailed features and regulatory mechanisms have not been well characterized. RESULTS: Here, we profile the landscape of H3K4me3-enriched enhancers (m3Es) in 43 pairs of colorectal cancer (CRC) samples. M3Es are widely distributed in CRC and averagely possess around 10% of total active enhancers. We identify 1322 gain variant m3Es and 367 lost variant m3Es in CRC. The target genes of the gain m3Es are enriched in immune response pathways. We experimentally prove that repression of CBX8 and RPS6KA5 m3Es inhibits target gene expression in CRC. Furthermore, we find histone methyltransferase MLL1 is responsible for depositing H3K4me3 on the identified Vm3Es. We demonstrate that the transcription factor AP1/JUN interacts with MLL1 and regulates m3E activity. Application of a small chemical inhibitor for MLL1 activity, OICR-9429, represses target gene expression of the identified Vm3Es, enhances anti-tumor immunity and inhibits CRC growth in an animal model. CONCLUSIONS: Taken together, our study illustrates the genome-wide landscape and the regulatory mechanisms of m3Es in CRC, and reveals potential novel strategies for cancer treatment.


Sujet(s)
Tumeurs colorectales , Histone , Protéine de la leucémie myéloïde-lymphoïde , Protéines proto-oncogènes c-jun , Animaux , Tumeurs colorectales/génétique , Éléments activateurs (génétique) , Histone/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/génétique , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Facteur de transcription AP-1/métabolisme , Humains , Protéines proto-oncogènes c-jun/génétique , Protéines proto-oncogènes c-jun/métabolisme
19.
Life Sci Alliance ; 6(11)2023 11.
Article de Anglais | MEDLINE | ID: mdl-37604584

RÉSUMÉ

Loss of c-JUN leads to early mouse embryonic death, possibly because of a failure to develop a normal cardiac system. How c-JUN regulates human cardiomyocyte cell fate remains unknown. Here, we used the in vitro differentiation of human pluripotent stem cells into cardiomyocytes to study the role of c-JUN. Surprisingly, the knockout of c-JUN improved cardiomyocyte generation, as determined by the number of TNNT2+ cells. ATAC-seq data showed that the c-JUN defect led to increased chromatin accessibility on critical regulatory elements related to cardiomyocyte development. ChIP-seq data showed that the knockout c-JUN increased RBBP5 and SETD1B expression, leading to improved H3K4me3 deposition on key genes that regulate cardiogenesis. The c-JUN KO phenotype could be copied using the histone demethylase inhibitor CPI-455, which also up-regulated H3K4me3 levels and increased cardiomyocyte generation. Single-cell RNA-seq data defined three cell branches, and knockout c-JUN activated more regulons that are related to cardiogenesis. In summary, our data demonstrated that c-JUN could regulate cardiomyocyte cell fate by modulating H3K4me3 modification and chromatin accessibility and shed light on how c-JUN regulates heart development in humans.


Sujet(s)
Cellules souches embryonnaires humaines , Protéines proto-oncogènes c-jun , Animaux , Humains , Souris , Différenciation cellulaire , Chromatine/génétique , Gènes jun , Myocytes cardiaques , Protéines proto-oncogènes c-jun/métabolisme
20.
Front Immunol ; 14: 1224892, 2023.
Article de Anglais | MEDLINE | ID: mdl-37483616

RÉSUMÉ

Activator protein-1 (AP-1) is a transcription factor that consists of a diverse group of members including Jun, Fos, Maf, and ATF. AP-1 involves a number of processes such as proliferation, migration, and invasion in cells. Dysfunctional AP-1 activity is associated with cancer initiation, development, invasion, migration and drug resistance. Therefore, AP-1 is a potential target for cancer targeted therapy. Currently, some small molecule inhibitors targeting AP-1 have been developed and tested, showing some anticancer effects. However, AP-1 is complex and diverse in its structure and function, and different dimers may play different roles in different type of cancers. Therefore, more research is needed to reveal the specific mechanisms of AP-1 in cancer, and how to select appropriate inhibitors and treatment strategies. Ultimately, this review summarizes the potential of combination therapy for cancer.


Sujet(s)
Tumeurs , Facteur de transcription AP-1 , Humains , Facteur de transcription AP-1/métabolisme , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-jun/métabolisme , Régulation de l'expression des gènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...