Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.506
Filtrer
1.
Mol Biol Rep ; 51(1): 711, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824245

RÉSUMÉ

BACKGROUND: Diabetes is a chronic metabolic disease that affects many parts of the body. Considering diabetes as a beta cells' defect and loss, the focus is on finding mechanisms and compounds involved in stimulating the function and regeneration of pancreatic ß-cells. DNA methylation as an epigenetic mechanism plays a pivotal role in the ß-cells' function and development. Considering the regenerative and anti-diabetic effects of Rosa canina extract, this study aimed to assess the methylation levels of Pdx-1, Pax-4, and Ins-1 genes in diabetic rats treated with Rosa Canina extract. METHODS AND RESULTS: Streptozotocin-induced diabetic rats were used to evaluate the frequency of Pdx-1, Pax-4, and Ins-1 gene methylation. Treatment groups were exposed to Rosa canina as spray-dried and decoction extracts. Following blood glucose measurement, pancreatic DNA was extracted and bisulfited. Genes' methylation was measured using MSP-PCR and qRT-PCR techniques. Oral administration of Rosa canina extracts significantly reduced blood sugar levels in diabetic rats compared to the control group. The methylation levels of the Pdx-1, Pax-4, and Ins-1 genes promoter in streptozotocin-induced diabetic rats increased compared to the control rats while, the treatment of diabetic rats with Rosa canina extracts, spray-dried samples especially, led to a decreased methylation in these genes. CONCLUSION: The results of this study showed that Rosa canina extract as a spray-dried sample could be effective in treating diabetes by regulating the methylation of genes including Pdx-1, Pax-4, and Ins-1 involved in the activity and regeneration of pancreatic islet cells.


Sujet(s)
Glycémie , Méthylation de l'ADN , Diabète expérimental , Extraits de plantes , Rosa , Transactivateurs , Animaux , Diabète expérimental/génétique , Diabète expérimental/traitement médicamenteux , Rosa/composition chimique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Méthylation de l'ADN/génétique , Rats , Extraits de plantes/pharmacologie , Mâle , Transactivateurs/génétique , Transactivateurs/métabolisme , Glycémie/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Pancréas/effets des médicaments et des substances chimiques , Pancréas/métabolisme , Pancréas/anatomopathologie , Streptozocine , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Régions promotrices (génétique)/génétique , Facteurs de transcription PAX/génétique , Facteurs de transcription PAX/métabolisme , Insuline/métabolisme
2.
Chin J Nat Med ; 22(6): 554-567, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38906602

RÉSUMÉ

Diffuse large B-cell lymphoma (DLBCL) is characterized by significant treatment resistance. Palmitic acid (PA) has shown promising antitumor properties. This study aims to elucidate the molecular mechanisms by which PA influences DLBCL progression. We quantified the expression levels of microRNAs (miRNAs), Forkhead box protein O1 (FOXO1), and DNA methyltransferase 3A (DNMT3A) in both untreated and PA-treated DLBCL tumors and cell lines. Assessments were made of cell viability, apoptosis, and autophagy-related protein expression following PA administration. Interaction analyses among miR-429, DNMT3A, and FOXO1 were conducted using luciferase reporter assays and methylation-specific (MSP) Polymerase chain reaction (PCR). After transfecting the miR-429 inhibitor, negative control (NC) inhibitor, shRNA against DNMT3A (sh-DNMT3A), shRNA negative control (sh-NC), overexpression vector for DNMT3A (oe-DNMT3A), or overexpression negative control (oe-NC), we evaluated the effects of miR-429 and DNMT3A on cell viability, mortality, and autophagy-related protein expression in PA-treated DLBCL cell lines. The efficacy of PA was also tested in vivo using DLBCL tumor-bearing mouse models. MiR-429 and FOXO1 expression levels were downregulated, whereas DNMT3A was upregulated in DLBCL compared to the control group. PA treatment was associated with enhanced autophagy, mediated by the upregulation of miR-429 and downregulation of DNMT3A. The luciferase reporter assay and MSP confirmed that miR-429 directly inhibits DNMT3A, thereby reducing FOXO1 methylation. Subsequent experiments demonstrated that PA promotes autophagy and inhibits DLBCL progression by upregulating miR-429 and modulating the DNMT3A/FOXO1 axis. In vivo PA significantly reduced the growth of xenografted tumors through its regulatory impact on the miR-429/DNMT3A/FOXO1 axis. Palmitic acid may modulate autophagy and inhibit DLBCL progression by targeting the miR-429/DNMT3A/FOXO1 signaling pathway, suggesting a novel therapeutic target for DLBCL management.


Sujet(s)
DNA methyltransferase 3A , Protéine O1 à motif en tête de fourche , Lymphome B diffus à grandes cellules , microARN , Acide palmitique , microARN/génétique , microARN/métabolisme , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/métabolisme , Humains , Protéine O1 à motif en tête de fourche/génétique , Protéine O1 à motif en tête de fourche/métabolisme , Animaux , Souris , Acide palmitique/pharmacologie , Lignée cellulaire tumorale , Méthylation de l'ADN/effets des médicaments et des substances chimiques , DNA (cytosine-5-)-methyltransferase/génétique , DNA (cytosine-5-)-methyltransferase/métabolisme , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Souris nude , Mâle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Femelle , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Souris de lignée BALB C
3.
PeerJ ; 12: e17363, 2024.
Article de Anglais | MEDLINE | ID: mdl-38766487

RÉSUMÉ

Background: Promoter hypermethylation of the tumor suppressor gene is one of the well-studied causes of cancer development. The drugs that reverse the process by driving demethylation could be a candidate for anticancer therapy. This study was designed to investigate the effects of arsenic disulfide on PTPL1 methylation in diffuse large B cell lymphoma (DLBCL). Methods: We knocked down the expression of PTPL1 in two DLBCL cell lines (i.e., DB and SU-DHL-4 cells) using siRNA. Then the DLBCL proliferation was determined in the presence of PTPL1 knockdown. The methylation of PTPL1 in DLBCL cells was analyzed by methylation specific PCR (MSPCR). The effect of arsenic disulfide on the PTPL1 methylation was determined in DLBCL cell lines in the presence of different concentrations of arsenic disulfide (5 µM, 10 µM and 20 µM), respectively. To investigate the potential mechanism on the arsenic disulfide-mediated methylation, the mRNA expression of DNMT1, DNMT3B and MBD2 was determined. Results: PTPL1 functioned as a tumor suppressor gene in DLBCL cells, which was featured by the fact that PTPL1 knockdown promoted the proliferation of DLBCL cells. PTPL1 was found hypermethylated in DLBCL cells. Arsenic disulfide promoted the PTPL1 demethylation in a dose-dependent manner, which was related to the inhibition of DNMTs and the increase of MBD2. Conclusion: Experimental evidence shows that PTPL1 functions as a tumor suppressor gene in DLBCL progression. PTPL1 hyper-methylation could be reversed by arsenic disulfide in a dose-dependent manner.


Sujet(s)
Prolifération cellulaire , Méthylation de l'ADN , Lymphome B diffus à grandes cellules , Humains , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/anatomopathologie , Lymphome B diffus à grandes cellules/métabolisme , Lignée cellulaire tumorale , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Composés de l'arsenic/pharmacologie , , Disulfures/pharmacologie , DNA (cytosine-5-)-methyltransferase/génétique , DNA (cytosine-5-)-methyltransferase/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/génétique , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Techniques de knock-down de gènes , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
4.
Clin Transl Med ; 14(5): e1703, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38769666

RÉSUMÉ

BACKGROUND: Hotspot mutations in the promoter of telomerase reverse transcriptase (TERT) gene are the most common genetic variants in hepatocellular carcinoma (HCC) and associated with poor prognosis of the disease. However, no drug was currently approved for treating TERT promoter mutation positive HCC patients. Here, we aim to explore the potential therapeutic strategy for targeting TERT promoter mutation in HCC. METHODS: The Liver Cancer Model Repository database was used for screening potential drugs to selectively suppress the growth of TERT promoter mutant HCC cells. RNA-seq, CRISPR-Cas9 technology and siRNA transfection were performed for mechanistic studies. Cell counting kit-8 (CCK8) assay and the xenograft tumour models were used for cell growth detection in vitro and in vivo, respectively. Cell apoptosis and cell cycle arrest were analysed by Annexin V-FITC staining and/or propidium iodide staining. RESULTS: PLK1 inhibitors were remarkably more sensitive to HCC cells harbouring TERT promoter mutation than wild-type cells in vitro and in vivo, which were diminished after TERT promoter mutation was edited to the wild-type nucleotide. Comparing the HCC cells with wild-type promoter of TERT, PLK1 inhibitors specifically downregulated Smad3 to regulate TERT for inducing apoptosis and G2/M arrest in TERT mutant HCC cells. Moreover, knockout of Smad3 counteracted the effects of PLK1 inhibitors in TERT mutant HCC cells. Finally, a cooperative effect of PLK1 and Smad3 inhibition was observed in TERT mutant cells. CONCLUSIONS: PLK1 inhibition selectively suppressed the growth of TERT mutant HCC cells through Smad3, thus contributed to discover a novel therapeutic strategy to treat HCC patients harbouring TERT promoter mutations. KEY POINTS: TERT promoter mutation confers sensitivity to PLK1 inhibitors in HCC. The selective growth inhibition of TERT mutant HCC cells induced by PLK1 inhibitor was mediated by Smad3. Combined inhibition of PLK1 and Smad3 showed a cooperative anti-tumor effect in TERT mutant HCC cells.


Sujet(s)
Carcinome hépatocellulaire , Protéines du cycle cellulaire , Tumeurs du foie , , Régions promotrices (génétique) , Protein-Serine-Threonine Kinases , Protéines proto-oncogènes , Telomerase , Telomerase/génétique , Telomerase/antagonistes et inhibiteurs , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Régions promotrices (génétique)/génétique , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Animaux , Mutation , Souris , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique
5.
Biochim Biophys Acta Gen Subj ; 1868(7): 130631, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38685534

RÉSUMÉ

BACKGROUND: Vascular endothelial growth factor (VEGF) is overexpressed in most malignant tumors, which has important impact on tumor angiogenesis and development. Its gene promoter i-motif structure formed by C-rich sequence can regulate gene expression, which is a promising new target for anti-tumor therapy. METHODS: We screened various compounds and studied their effects on VEGF through extensive experiments, including SPR, MST, TO displacement, FRET, CD, ESI-MS, NMR, MTT, clone formation, qPCR, Western blot, dual-luciferase reporter assay, immunofluorescence, cell scrape, apoptosis, transwell assay, and animal model. RESULTS: After extensive screening, bisacridine derivative B09 was found to have selective binding and stabilization to VEGF promoter i-motif, which could down-regulate VEGF gene expression. B09 showed potent inhibition on MCF-7 and HGC-27 cell proliferation and metastasis. B09 significantly inhibited tumor growth in xenograft mice model with HGC-27 cells, showing decreased VEGF expression analyzed through immunohistochemistry. CONCLUSION: B09 could specifically regulate VEGF gene expression, possibly through interacting with promoter i-motif structure. As a lead compound, B09 could be further developed for innovative anti-cancer agent targeting VEGF.


Sujet(s)
Acridines , Régulation de l'expression des gènes tumoraux , Régions promotrices (génétique) , Facteur de croissance endothéliale vasculaire de type A , Humains , Animaux , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Souris , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Acridines/pharmacologie , Acridines/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Cellules MCF-7 , Souris nude , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Femelle , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique
6.
Drug Metab Dispos ; 52(6): 555-564, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38565301

RÉSUMÉ

Cytochrome P450 1A2 (CYP1A2) is a known tumor suppressor in hepatocellular carcinoma (HCC), but its expression is repressed in HCC and the underlying mechanism is unclear. In this study, we investigated the epigenetic mechanisms of CYP1A2 repression and potential therapeutic implications. In HCC tumor tissues, the methylation rates of CYP1A2 CpG island (CGI) and DNA methyltransferase (DNMT) 3A protein levels were significantly higher, and there was a clear negative correlation between DNMT3A and CYP1A2 protein expression. Knockdown of DNMT3A by siRNA significantly increased CYP1A2 expression in HCC cells. Additionally, treating HCC cells with decitabine (DAC) resulted in a dose-dependent upregulation of CYP1A2 expression by reducing the methylation level of CYP1A2 CGI. Furthermore, we observed a decreased enrichment of H3K27Ac in the promoter region of CYP1A2 in HCC tissues. Treatment with the trichostatin A (TSA) restored CYP1A2 expression in HCC cells by increasing H3K27Ac levels in the CYP1A2 promoter region. Importantly, combination treatment of sorafenib with DAC or TSA resulted in a leftward shift of the dose-response curve, lower IC50 values, and reduced colony numbers in HCC cells. Our findings suggest that hypermethylation of the CGI at the promoter, mediated by the high expression of DNMT3A, and hypoacetylation of H3K27 in the CYP1A2 promoter region, leads to CYP1A2 repression in HCC. Epigenetic drugs DAC and TSA increase HCC cell sensitivity to sorafenib by restoring CYP1A2 expression. Our study provides new insights into the epigenetic regulation of CYP1A2 in HCC and highlights the potential of epigenetic drugs as a therapeutic approach for HCC. SIGNIFICANCE STATEMENT: This study marks the first exploration of the epigenetic mechanisms underlying cytochrome P450 (CYP) 1A2 suppression in hepatocellular carcinoma (HCC). Our findings reveal that heightened DNA methyltransferase expression induces hypermethylation of the CpG island at the promoter, coupled with diminished H3K27Ac levels, resulting in the repression of CYP1A2 in HCC. The use of epigenetic drugs such as decitabine and trichostatin A emerges as a novel therapeutic avenue, demonstrating their potential to restore CYP1A2 expression and enhance sorafenib sensitivity in HCC cells.


Sujet(s)
Carcinome hépatocellulaire , Cytochrome P-450 CYP1A2 , Méthylation de l'ADN , Épigenèse génétique , Tumeurs du foie , Sorafénib , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Humains , Sorafénib/pharmacologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Épigenèse génétique/effets des médicaments et des substances chimiques , Épigenèse génétique/génétique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cytochrome P-450 CYP1A2/génétique , Cytochrome P-450 CYP1A2/métabolisme , DNA methyltransferase 3A , Antinéoplasiques/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , DNA (cytosine-5-)-methyltransferase/génétique , DNA (cytosine-5-)-methyltransferase/métabolisme , Décitabine/pharmacologie , Ilots CpG/génétique , Acides hydroxamiques/pharmacologie , Régions promotrices (génétique)/génétique , Régions promotrices (génétique)/effets des médicaments et des substances chimiques
7.
Cancer Res Commun ; 4(5): 1307-1320, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38669046

RÉSUMÉ

Anticancer drug-tolerant persister (DTP) cells at an early phase of chemotherapy reshape refractory tumors. Aldehyde dehydrogenase 1 family member A3 (ALDH1A3) is commonly upregulated by various anticancer drugs in gastric cancer patient-derived cells (PDC) and promotes tumor growth. However, the mechanism underlying the generation of ALDH1A3-positive DTP cells remains elusive. Here, we investigated the mechanism of ALDH1A3 expression and a combination therapy targeting gastric cancer DTP cells. We found that gastric cancer tissues treated with neoadjuvant chemotherapy showed high ALDH1A3 expression. Chromatin immunoprecipitation (ChIP)-PCR and ChIP sequencing analyses revealed that histone H3 lysine 27 acetylation was enriched in the ALDH1A3 promoter in 5-fluorouracil (5-FU)-tolerant persister PDCs. By chemical library screening, we found that the bromodomain and extraterminal (BET) inhibitors OTX015/birabresib and I-BET-762/molibresib suppressed DTP-related ALDH1A3 expression and preferentially inhibited DTP cell growth. In DTP cells, BRD4, but not BRD2/3, was recruited to the ALDH1A3 promoter and BRD4 knockdown decreased drug-induced ALDH1A3 upregulation. Combination therapy with 5-FU and OTX015 significantly suppressed in vivo tumor growth. These observations suggest that BET inhibitors are efficient DTP cell-targeting agents for gastric cancer treatment. SIGNIFICANCE: Drug resistance hampers the cure of patients with cancer. To prevent stable drug resistance, DTP cancer cells are rational therapeutic targets that emerge during the early phase of chemotherapy. This study proposes that the epigenetic regulation by BET inhibitors may be a rational therapeutic strategy to eliminate DTP cells.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Fluorouracil , Histone , Tumeurs de l'estomac , Facteurs de transcription , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Humains , Animaux , Histone/métabolisme , Souris , Acétylation/effets des médicaments et des substances chimiques , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Fluorouracil/pharmacologie , Fluorouracil/usage thérapeutique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris nude , Aldehyde oxidoreductases/métabolisme , Aldehyde oxidoreductases/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mâle , Femelle , Antinéoplasiques/pharmacologie , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Protéines contenant un bromodomaine
8.
Molecules ; 29(8)2024 Apr 21.
Article de Anglais | MEDLINE | ID: mdl-38675711

RÉSUMÉ

Although much less common than anthocyanins, 3-Deoxyanthocyanidins (3-DAs) and their glucosides can be found in cereals such as red sorghum. It is speculated that their bioavailability is higher than that of anthocyanins. Thus far, little is known regarding the therapeutic effects of 3-DAs and their O-ß-D-glucosides on cancer, including prostate cancer. Thus, we evaluated their potential to decrease cell viability, to modulate the activity of transcription factors such as NFκB, CREB, and SOX, and to regulate the expression of the gene CDH1, encoding E-Cadherin. We found that 4',7-dihydroxyflavylium chloride (P7) and the natural apigeninidin can reduce cell viability, whereas 4',7-dihydroxyflavylium chloride (P7) and 4'-hydroxy-7-O-ß-D-glucopyranosyloxyflavylium chloride (P3) increase the activities of NFkB, CREB, and SOX transcription factors, leading to the upregulation of CDH1 promoter activity in PC-3 prostate cancer cells. Thus, these compounds may contribute to the inhibition of the epithelial-to-mesenchymal transition in cancer cells and prevent the metastatic activity of more aggressive forms of androgen-resistant prostate cancer.


Sujet(s)
Anthocyanes , Cadhérines , Glucosides , Régions promotrices (génétique) , Tumeurs de la prostate , Sorghum , Humains , Mâle , Anthocyanes/pharmacologie , Anthocyanes/composition chimique , Antigènes CD/métabolisme , Antigènes CD/génétique , Cadhérines/effets des médicaments et des substances chimiques , Cadhérines/génétique , Cadhérines/métabolisme , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Glucosides/pharmacologie , Glucosides/composition chimique , Facteur de transcription NF-kappa B/métabolisme , Cellules PC-3 , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Sorghum/composition chimique
9.
Int J Mol Sci ; 25(4)2024 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-38396982

RÉSUMÉ

Cystic fibrosis (CF) is a genetic disorder caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR), a selective anion channel expressed in the epithelium of various organs. The most frequent mutation is F508del. This mutation leads to a misfolded CFTR protein quickly degraded via ubiquitination in the endoplasmic reticulum. Although preventing ubiquitination stabilizes the protein, functionality is not restored due to impaired plasma membrane transport. However, inhibiting the ubiquitination process can improve the effectiveness of correctors which act as chemical chaperones, facilitating F508del CFTR trafficking to the plasma membrane. Previous studies indicate a crosstalk between SUMOylation and ubiquitination in the regulation of CFTR. In this study, we investigated the potential of inhibiting SUMOylation to increase the effects of correctors and enhance the rescue of the F508del mutant across various cell models. In the widely used CFBE41o-cell line expressing F508del-CFTR, inhibiting SUMOylation substantially boosted F508del expression, thereby increasing the efficacy of correctors. Interestingly, this outcome did not result from enhanced stability of the mutant channel, but rather from augmented cytomegalovirus (CMV) promoter-mediated gene expression of F508del-CFTR. Notably, CFTR regulated by endogenous promoters in multiple cell lines or patient cells was not influenced by SUMOylation inhibitors.


Sujet(s)
Protéine CFTR , Mucoviscidose , Sumoylation , Humains , Mucoviscidose/traitement médicamenteux , Mucoviscidose/génétique , Mucoviscidose/métabolisme , Protéine CFTR/génétique , Protéine CFTR/métabolisme , Cytomegalovirus , Mutation , Sumoylation/effets des médicaments et des substances chimiques , Régions promotrices (génétique)/effets des médicaments et des substances chimiques
10.
Steroids ; 187: 109094, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35905833

RÉSUMÉ

Hormonal contraceptives (HCs) and hormone replacement therapy (HRT) are therapies designed to target the progesterone receptor (PR) to prevent unwanted pregnancy and to alleviate the symptoms of menopause, respectively, in women. Although these therapies are widely used globally, few studies have investigated in parallel how the transcriptional responses of the progestogens used in these therapies compare to each other via the PR isoforms (PR-A and PR-B). Using dose-response promoter-reporter and endogenous gene expression assays, we compared the transcriptional responses of six widely-used progestogens via each PR isoform. The present study shows that progestogens exhibit progestogen-specific potencies and efficacies via both PR isoforms. In addition, the endogenous gene expression data reveals that progestogens exhibit promoter-specific effects. Furthermore, this study reveals that progestogen responses via PR-A are significantly more potent and less efficacious than those observed via PR-B, and that this is unlikely due to differences in PR protein levels. Correlation analysis revealed that there is no detectable correlation between potency or efficacy of progestogens for PR-B or PR-A versus reported relative binding affinity (RBA) of progestogens for the PR, consistent with complex mechanisms of PR regulation. Taken together, our data show that it cannot be assumed that all progestogens have similar transcriptional responses on all genes. Since the PR plays a role in cognition, regulation of inflammation, mitochondrial function, neurogenesis, female reproduction and disease, the data suggest that these important physiological functions could be differentially affected depending on progestogen, promoter, and ratios of PR isoforms.


Sujet(s)
Progestines , Récepteurs à la progestérone , Femelle , Humains , Grossesse , Contraceptifs , Ménopause , Progestérone/pharmacologie , Progestines/pharmacologie , Isoformes de protéines/génétique , Récepteurs à la progestérone/génétique , Récepteurs à la progestérone/métabolisme , Régions promotrices (génétique)/effets des médicaments et des substances chimiques
11.
Bioengineered ; 13(6): 14595-14604, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35758042

RÉSUMÉ

It is acknowledged that nonsteroidal anti-inflammatory drugs (NSAIDs) can participate in various signaling pathways, while information about their epigenetic effects are limited. p75NTR (p75 neurotrophin receptor) can inhibit tumor growth by inducing cell cycle arrest and regulating cell cycle arrest and apoptotic cell death. The expression of p75NTR is influenced by epigenetic roles. We explored the effects of ibuprofen on p75NTR expression and investigated whether promoter methylation and N6-methyladenosine (m6A) RNA methylation regulates this process in human gastric cancer cells (SGC7901 and MKN45). Cell lines were treated with ibuprofen 0, 2.5, 5, 10, 20 µM, and then DNA, RNA, and protein were isolated 24 h later. Expression and promoter methylation of p75NTR were detected by RT-qPCR and Western blot. The levels of m6A-p75NTR were measured by RNA immunoprecipitation. We also used RT-qPCR to determine the levels of m6A-related regulators, METTL3, METTL14, ALKBH5, FTO, YTHDC2, and YTHDF1-3. Ibuprofen attenuated p75NTR promoter methylation (p < 0.01) and increased p75NTR level (p < 0.001). Ibuprofen increased m6A-p53 expression (p < 0.01) by promoting the expression of METTL3 (p < 0.01) and METTL14 (p < 0.05); and increased levels of YTHDF1 (p < 0.001), YTHDF3 (p < 0.001), and YTHDC2 (p < 0.01) that finally reinforced p53 translation (p < 0.01). Therefore, our results present that ibuprofen epigenetically increased p75NTR expression by downregulating promoter methylation and upregulating m6A-RNA-methylation in SGC7901 and MKN45 cells. Our study unveils a novel mechanism for p75NTR regulation by NSAIDs and helps the design of treatment targets.


Sujet(s)
Adénosine , Méthylation de l'ADN , Ibuprofène , Methyltransferases , Protéines de tissu nerveux , Récepteurs facteur croissance nerf , Tumeurs de l'estomac , Adénosine/analogues et dérivés , Adénosine/métabolisme , Adénosine/pharmacologie , Alpha-ketoglutarate-dependent dioxygenase FTO/génétique , Alpha-ketoglutarate-dependent dioxygenase FTO/métabolisme , Anti-inflammatoires non stéroïdiens/pharmacologie , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Humains , Ibuprofène/pharmacologie , Methyltransferases/génétique , Methyltransferases/métabolisme , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , ARN/génétique , ARN/métabolisme , ARN messager/génétique , Récepteur facteur croissance nerf/métabolisme , Récepteurs facteur croissance nerf/génétique , Récepteurs facteur croissance nerf/métabolisme , Tumeurs de l'estomac/anatomopathologie , Protéine p53 suppresseur de tumeur/métabolisme
12.
EMBO Mol Med ; 14(4): e14841, 2022 04 07.
Article de Anglais | MEDLINE | ID: mdl-35263037

RÉSUMÉ

Small-Cell Lung Cancer (SCLC) is an aggressive neuroendocrine malignancy with a poor prognosis. Here, we focus on the neuroendocrine SCLC subtypes, SCLC-A and SCLC-N, whose transcription addiction was driven by ASCL1 and NEUROD1 transcription factors which target E-box motifs to activate up to 40% of total genes, the promoters of which are maintained in a steadily open chromatin environment according to ATAC and H3K27Ac signatures. This leverage is used by the marine agent lurbinectedin, which preferentially targets the CpG islands located downstream of the transcription start site, thus arresting elongating RNAPII and promoting its degradation. This abrogates the expression of ASCL1 and NEUROD1 and of their dependent genes, such as BCL2, INSM1, MYC, and AURKA, which are responsible for relevant SCLC tumorigenic properties such as inhibition of apoptosis and cell survival, as well as for a part of its neuroendocrine features. In summary, we show how the transcription addiction of these cells becomes their Achilles's heel, and how this is effectively exploited by lurbinectedin as a novel SCLC therapeutic endeavor.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice , Carbolines , Composés hétérocycliques avec 4 noyaux ou plus , Tumeurs du poumon , Protéines de répression , Carcinome pulmonaire à petites cellules , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Carbolines/pharmacologie , Lignée cellulaire tumorale , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Protéines de répression/métabolisme , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/métabolisme
13.
PLoS One ; 17(2): e0263430, 2022.
Article de Anglais | MEDLINE | ID: mdl-35139106

RÉSUMÉ

BMP7 is a morphogen capable of counteracting the OA chondrocyte hypertrophic phenotype via NKX3-2. NKX3-2 represses expression of RUNX2, an important transcription factor for chondrocyte hypertrophy. Since RUNX2 has previously been described as an inhibitor for 47S pre-rRNA transcription, we hypothesized that BMP7 positively influences 47S pre-rRNA transcription through NKX3-2, resulting in increased protein translational capacity. Therefor SW1353 cells and human primary chondrocytes were exposed to BMP7 and rRNA (18S, 5.8S, 28S) expression was determined by RT-qPCR. NKX3-2 knockdown was achieved via transfection of a NKX3-2-specific siRNA duplex. Translational capacity was assessed by the SUNsET assay, and 47S pre-rRNA transcription was determined by transfection of a 47S gene promoter-reporter plasmid. BMP7 treatment increased protein translational capacity. This was associated by increased 18S and 5.8S rRNA and NKX3-2 mRNA expression, as well as increased 47S gene promotor activity. Knockdown of NKX3-2 led to increased expression of RUNX2, accompanied by decreased 47S gene promotor activity and rRNA expression, an effect BMP7 was unable to restore. Our data demonstrate that BMP7 positively influences protein translation capacity of SW1353 cells and chondrocytes. This is likely caused by an NKX3-2-dependent activation of 47S gene promotor activity. This finding connects morphogen-mediated changes in cellular differentiation to an aspect of ribosome biogenesis via key transcription factors central to determining the chondrocyte phenotype.


Sujet(s)
Protéine morphogénétique osseuse de type 7/physiologie , Chondrocytes/métabolisme , Protéines à homéodomaine/physiologie , Biosynthèse des protéines/génétique , ARN ribosomique/métabolisme , Facteurs de transcription/physiologie , Protéine morphogénétique osseuse de type 7/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Cellules cultivées , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/physiologie , Chondrogenèse/effets des médicaments et des substances chimiques , Chondrogenèse/génétique , Sous-unité alpha 1 du facteur CBF/génétique , Sous-unité alpha 1 du facteur CBF/métabolisme , Humains , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Biosynthèse des protéines/effets des médicaments et des substances chimiques , ARN ribosomique/génétique , Transcription génétique/effets des médicaments et des substances chimiques , Transcription génétique/génétique , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/génétique
14.
Int J Mol Sci ; 23(3)2022 Jan 20.
Article de Anglais | MEDLINE | ID: mdl-35163072

RÉSUMÉ

The TIR-containing protein C (TcpC) of the uropathogenic Escherichia coli strain CFT073 modulates innate immunity by interfering with the Toll-like receptor and NALP3 inflammasome signaling cascade. During a urinary tract infection the pathogen encounters epithelial and innate immune cells and replicates by several orders of magnitude. We therefore analyzed whether these cell types and also the density of the pathogen would induce the recently defined promoter of the CFT073 tcpC gene to, in time, dampen innate immune responses. Using reporter constructs we found that the uroepithelial cell line T24/83 and the monocytic cell line THP-1 induced the tcpC promoter. Differentiation of monocytic THP-1 cells to macrophages increased their potential to switch on the promoter. Cell-associated CFT073 displayed the highest promoter activity. Since potassium represents the most abundant intracellular ion and is secreted to induce the NLRP3 inflammasome, we tested its ability to activate the tcpC promoter. Potassium induced the promoter with high efficiency. Sodium, which is enriched in the renal cortex generating an antibacterial hypersalinity, also induced the tcpC promoter. Finally, the bacterial density modulated the tcpC promoter activity. In the search for promoter-regulating proteins, we found that the DNA-binding protein H-NS dampens the promoter activity. Taken together, different cell types and salts, present in the kidney, are able to induce the tcpC promoter and might explain the mechanism of TcpC induction during a kidney infection with uropathogenic E. coli strains.


Sujet(s)
Protéines Escherichia coli/génétique , Protéines Escherichia coli/métabolisme , Protéines de fimbriae/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Infections urinaires/microbiologie , Escherichia coli uropathogène/pathogénicité , Facteurs de virulence/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Régulation de l'expression des gènes bactériens , Humains , Inflammasomes/métabolisme , Modèles biologiques , Potassium/pharmacologie , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Transduction du signal , Sodium/pharmacologie , Cellules THP-1 , Infections urinaires/métabolisme , Escherichia coli uropathogène/génétique , Facteurs de virulence/métabolisme
15.
Int J Mol Sci ; 23(3)2022 Jan 28.
Article de Anglais | MEDLINE | ID: mdl-35163437

RÉSUMÉ

Insulin-like growth factor 1 (IGF-1) not only regulates neuronal function and development but also is neuroprotective in the setting of acute ischemic stroke. G-protein-coupled receptor 17 (GPR17) expression in brain tissue serves as an indicator of brain damage. As whether IGF-1 regulates GPR17 expression remains unknown, the aim of this study is to investigate how IGF-1 regulates GPR17 expression in vitro. Human neuroblastoma SK-N-SH cells were used. Lentivirus-mediated short hairpin RNA (shRNA) was constructed to mediate the silencing of FoxO1, while adenoviral vectors were used for its overexpression. Verification of the relevant signaling cascade was performed using a FoxO1 inhibitor (AS1842856), a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002), and a GPR17 antagonist (cangrelor). Cell proliferation was analyzed using EdU staining; immunofluorescence staining was used to detect the expression and subcellular localization of FoxO1. Chromatin immunoprecipitation was used to analyze the binding of FoxO1 to the GPR17 promoter in SK-N-SH cells. The expression of FoxO1, GPR17, and protein kinase B (also known as Akt) mRNA and protein as well as the levels of FoxO1 and Akt phosphorylation were investigated in this study. IGF-1 was found to downregulate FoxO1 and GPR17 expression in SK-N-SH cells while promoting cell viability and proliferation. Inhibition of FoxO1 and antagonism of GPR17 were found to play a role similar to that of IGF-1. Silencing of FoxO1 by lentivirus-mediated shRNA resulted in the downregulation of FoxO1 and GPR17 expression. The overexpression of FoxO1 via adenoviral vectors resulted in the upregulation of FoxO1 and GPR17 expression. Blocking of PI3K signaling by LY294002 inhibited the effect of IGF-1 on GPR17 suppression. Results from chromatin immunoprecipitation revealed that IGF-1 promotes FoxO1 nuclear export and reduces FoxO1 binding to the GPR17 promoter in SK-N-SH cells. Here, we conclude that IGF-1 enhances cell viability and proliferation in SK-N-SH cells via the promotion of FoxO1 nuclear export and reduction of FoxO1 binding to the GPR17 promoter via PI3K/Akt signaling. Our findings suggest that the enhancement of IGF-1 signaling to antagonize GPR17 serves as a potential therapeutic strategy in the management of acute ischemic stroke.


Sujet(s)
Régulation négative , Protéine O1 à motif en tête de fourche/génétique , Facteur de croissance IGF-I/métabolisme , Neurones/cytologie , Récepteurs couplés aux protéines G/génétique , AMP/analogues et dérivés , AMP/pharmacologie , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , 4H-1-Benzopyran-4-ones/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Protéine O1 à motif en tête de fourche/métabolisme , Techniques de knock-out de gènes , Humains , Lentivirus/physiologie , Morpholines/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Quinolinone/pharmacologie , Petit ARN interférent/pharmacologie , Récepteurs couplés aux protéines G/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
16.
FASEB J ; 36(2): e22162, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35061304

RÉSUMÉ

Apoptosis of alveolar epithelial cells is a critical initial link in the pathogenesis of acute lung injury (ALI), recent studies have revealed that Methyl-CpG binding domain protein 2 (MBD2) was involved in the execution of apoptosis, yet its role in ALI remained unclear. In the present study, we aim to explore the role and mechanism of MBD2 in the pathogenesis of ALI. We have found that MBD2 expression, in parallel to apoptosis, increased in alveolar epithelial cells of mice treated with LPS, knockout of MBD2 reduced apoptosis and protected mice from LPS-induced ALI. In MLE-12 cells, a cell line of murine alveolar epithelial cells, LPS induced MBD2 expression and apoptosis in a dose- and time-dependent manner. Knockdown of MBD2 with shRNA alleviated, while overexpression of MBD2 increased LPS-induced apoptosis. Mechanistically, intracellular zinc level decreased when MLE-12 cells were treated with LPS. MBD2 knockdown restored intracellular zinc level after LPS treatment, and MBD2 overexpression further aggravated LPS-induced intracellular zinc loss. Metal transcription factor 1 (MTF1) is a critical transcription factor in charge of intracellular zinc efflux. LPS treatment induced MTF1 expression both in vivo and in vitro. Inhibition of MTF1 reduced LPS-induced apoptosis in MLE-12 cells. MBD2 could bind to the promoter region of MTF1 and promote MTF1 expression. Collectively, these data indicated that loss of MBD2-ameliorated LPS-induced alveolar epithelial cell apoptosis and ALI in mice via modulating intracellular zinc homeostasis by upregulating MTF1.


Sujet(s)
Lésion pulmonaire aigüe/génétique , Pneumocytes/métabolisme , Apoptose/génétique , Protéines de liaison à l'ADN/génétique , Homéostasie/génétique , Zinc/métabolisme , Pneumocytes/effets des médicaments et des substances chimiques , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/génétique , Homéostasie/effets des médicaments et des substances chimiques , Lipopolysaccharides/pharmacologie , Mâle , Souris , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Régions promotrices (génétique)/génétique , Petit ARN interférent/génétique , Facteurs de transcription/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/génétique
17.
Bioengineered ; 13(1): 1476-1490, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34974813

RÉSUMÉ

To investigate the role of hypoxia-inducible factor 1-alpha (HIF1A) in hypoxia/reoxygenation (H/R) injury of cardiomyocytes induced by high glucose (HG). The in vitro model of coronary heart disease with diabetes was that H9c2 cells were stimulated by H/R and HG. Quantitative reverse transcription PCR (RT-qPCR) and Western blot analysis were used to detect the expression of HIF1A and angiopoietin-like protein 2 (ANGPTL2) in H9c2 cells. Cell viability and apoptosis were, respectively, estimated by Cell Counting Kit 8 (CCK-8) and TUNEL assays. Lactate dehydrogenase (LDH) activity, inflammation and oxidative stress were in turn detected by their commercial assay kits. Luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were used to confirm the association between HIF1A and ANGPTL2 promoter. The expression of nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway-related proteins and apoptosis-related proteins were also detected by Western blot analysis. As a result, ANGPTL2 expression was upregulated in H9c2 cells induced by HG or/and H/R. ANGPTL2 positively modulated HIF1A expression in H9c2 cells. HG or/and H/R suppressed the cell viability and promoted apoptosis, inflammatory response and oxidative stress levels in H9c2 cells. However, the knockdown of ANGPTL2 could reverse the above phenomena in H/R-stimulated-H9c2 cells through activation of Nrf2/HO-1 pathway. HIF1A transcriptionally activated ANGPTL2 expression. The effect of knockdown of ANGPTL2 on H/R triggered-H9c2 cells was weakened by HIF1A overexpression. In conclusion, knockdown of HIF1A downregulated ANGPTL2 to alleviate H/R injury in HG-induced H9c2 cells by activating the Nrf2/HO-1 pathway.


Sujet(s)
Protéine-2 de type angiopoïétine/génétique , Glucose/effets indésirables , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Lésion de reperfusion myocardique/génétique , Myocytes cardiaques/cytologie , Protéine-2 de type angiopoïétine/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire , Immunoprécipitation de la chromatine , Techniques de knock-down de gènes , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Modèles biologiques , Lésion de reperfusion myocardique/étiologie , Lésion de reperfusion myocardique/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Stress oxydatif , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Rats , Régulation positive
18.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article de Anglais | MEDLINE | ID: mdl-34983841

RÉSUMÉ

Macrophages induce a number of inflammatory response genes in response to stimulation with microbial ligands. In response to endotoxin Lipid A, a gene-activation cascade of primary followed by secondary-response genes is induced. Epigenetic state is an important regulator of the kinetics, specificity, and mechanism of gene activation of these two classes. In particular, SWI/SNF chromatin-remodeling complexes are required for the induction of secondary-response genes, but not primary-response genes, which generally exhibit open chromatin. Here, we show that a recently discovered variant of the SWI/SNF complex, the noncanonical BAF complex (ncBAF), regulates secondary-response genes in the interferon (IFN) response pathway. Inhibition of bromodomain-containing protein 9 (BRD9), a subunit of the ncBAF complex, with BRD9 bromodomain inhibitors (BRD9i) or a degrader (dBRD9) led to reduction in a number of interferon-stimulated genes (ISGs) following stimulation with endotoxin lipid A. BRD9-dependent genes overlapped highly with a subset of genes differentially regulated by BET protein inhibition with JQ1 following endotoxin stimulation. We find that the BET protein BRD4 is cobound with BRD9 in unstimulated macrophages and corecruited upon stimulation to ISG promoters along with STAT1, STAT2, and IRF9, components of the ISGF3 complex activated downstream of IFN-alpha receptor stimulation. In the presence of BRD9i or dBRD9, STAT1-, STAT2-, and IRF9-binding is reduced, in some cases with reduced binding of BRD4. These results demonstrate a specific role for BRD9 and the ncBAF complex in ISG activation and identify an activity for BRD9 inhibitors and degraders in dampening endotoxin- and IFN-dependent gene expression.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Interférons/métabolisme , Activation des macrophages/effets des médicaments et des substances chimiques , Facteurs de transcription/métabolisme , Antiviraux/pharmacologie , Protéines du cycle cellulaire/génétique , Assemblage et désassemblage de la chromatine/effets des médicaments et des substances chimiques , Humains , Sous-unité gamma du complexe ISGF3/métabolisme , Interféron alpha/pharmacologie , Interférons/génétique , Interférons/pharmacologie , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Domaines protéiques , Facteur de transcription STAT-1/métabolisme , Facteur de transcription STAT-2/métabolisme , Facteurs de transcription/génétique , Activation de la transcription/effets des médicaments et des substances chimiques
19.
Cancer Lett ; 530: 170-180, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35077804

RÉSUMÉ

C/EBPß has recently emerged as a pro-leukemogenic transcription factor that cooperates with oncoprotein MYB to maintain proliferation and differentiation block of AML cells, making C/EBPß an interesting drug target for AML. Here we have studied the inhibitory potential and biological effects of a synthetic analog of the natural product helenalin, a known inhibitor of C/EBPß. The synthetic compound inhibits C/EBPß by covalent binding to cysteine residues in the transactivation domain, thereby causing up-regulation of differentiation-associated genes, cell death and reduced self-renewal potential of AML cells. Suppression of these effects by ectopic expression of C/EBPß or MYB and gene expression profiling validate C/EBPß as a relevant target of the helenalin-mimic and highlight its role as a pro-leukemogenic factor. Overall, our work demonstrates that the synthetic helenalin mimic acts as a covalent inhibitor of C/EBPß and identifies the cysteine residues in the transactivation domain of C/EBPß as ligandable sites. The helenalin mimic can be considered a potential "lead molecule" but needs further development towards more effective C/EBPß inhibitors before being used as a therapeutic agent.


Sujet(s)
Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Sesquiterpènes de type guaïane/pharmacologie , Activation de la transcription/effets des médicaments et des substances chimiques , Cellules 3T3 , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/génétique , Lignée cellulaire , Lignée cellulaire tumorale , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/génétique , Cellules HEK293 , Cellules HL-60 , Humains , Leucémie aigüe myéloïde/génétique , Ligands , Souris , Souris de lignée C57BL , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Régions promotrices (génétique)/génétique , Cellules THP-1
20.
Cancer Sci ; 113(1): 28-40, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-34727389

RÉSUMÉ

Hepatocellular carcinoma (HCC) is one of the commonest lethal malignancies worldwide, and often diagnosed at an advanced stage, without any curative therapy. Immune checkpoint blockers targeting the programmed death receptor 1 (PD-1) have shown impressive antitumor activity in patients with advanced-stage HCC, while the response rate is only 30%. Inducible PD-L1 overexpression may result in a lack of response to cancer immunotherapy, which is attributed to a mechanism of adaptive immune resistance. Our study investigated that the overexpression of PD-L1 promoted the invasion and migration of liver cancer cells in vitro, and the induced overexpression of PD-L1 in the tumor microenvironment could weaken the effects of anti-PD-1 immunotherapy in a BALB/c mouse model of liver cancer. CPI-203, a small-molecule bromodomain-containing protein 4 (BRD4) inhibitor, which can potently inhibit PD-L1 expression in vitro and in vivo, combined with PD-1 antibody improved the response to immunotherapy in a liver cancer model. Cell transfection and chromatin immunoprecipitation assay manifested that BRD4 plays a key role in PD-L1 expression; CPI-203 can inhibit PD-L1 expression by inhibiting the BRD4 occupation of the PD-L1 promoter region. This study indicates a potential clinical immunotherapy method to reduce the incidence of clinical resistance to immunotherapy in patients with HCC.


Sujet(s)
Acétamides/administration et posologie , Azépines/administration et posologie , Antigène CD274/génétique , Protéines du cycle cellulaire/métabolisme , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Tumeurs du poumon/traitement médicamenteux , Facteurs de transcription/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Acétamides/pharmacologie , Animaux , Azépines/pharmacologie , Lignée cellulaire tumorale , Synergie des médicaments , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Cellules HepG2 , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Mâle , Souris , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...