Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Med Chem ; 224: 113686, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34303079

ABSTRACT

Pathway activating mutations of the transcription factor NRF2 and its negative regulator KEAP1 are strongly correlative with poor clinical outcome with pemetrexed/carbo(cis)platin/pembrolizumab (PCP) chemo-immunotherapy in lung cancer. Despite the strong genetic support and therapeutic potential for a NRF2 transcriptional inhibitor, currently there are no known direct inhibitors of the NRF2 protein or its complexes with MAF and/or DNA. Herein we describe the design of a novel and high-confidence homology model to guide a medicinal chemistry effort that resulted in the discovery of a series of peptides that demonstrate high affinity, selective binding to the Antioxidant Response Element (ARE) DNA and thereby displace NRF2-MAFG from its promoter, which is an inhibitory mechanism that to our knowledge has not been previously described. In addition to their activity in electrophoretic mobility shift (EMSA) and TR-FRET-based assays, we show significant dose-dependent ternary complex disruption of NRF2-MAFG binding to DNA by SPR, as well as cellular target engagement by thermal destabilization of HiBiT-tagged NRF2 in the NCI-H1944 NSCLC cell line upon digitonin permeabilization, and SAR studies leading to improved cellular stability. We report the characterization and unique profile of lead peptide 18, which we believe to be a useful in vitro tool to probe NRF2 biology in cancer cell lines and models, while also serving as an excellent starting point for additional in vivo optimization toward inhibition of NRF2-driven transcription to address a significant unmet medical need in non-small cell lung cancer (NSCLC).


Subject(s)
DNA/chemistry , MafG Transcription Factor/antagonists & inhibitors , NF-E2-Related Factor 2/antagonists & inhibitors , Peptides/chemistry , Antioxidant Response Elements/drug effects , DNA/metabolism , Drug Design , Drug Stability , Electrophoretic Mobility Shift Assay , Half-Life , HeLa Cells , Humans , MafG Transcription Factor/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Peptides/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Structure-Activity Relationship
2.
FASEB J ; 35(5): e21529, 2021 05.
Article in English | MEDLINE | ID: mdl-33813778

ABSTRACT

To identify hepatitis B virus (HBV)-related lncRNA(s), we previously examined the transcription profiles of the HBV-transgenic cell line HepG2-4D14 and parental HepG2 cells by RNA deep sequencing and identified 38 upregulated long noncoding RNAs (lncRNAs). In the present study, the lncRNA MAFG-AS1 is investigated in detail because its gene is located adjacent to the MAFG gene, which is an important transcription factor involved in cell proliferation. The level of MAFG-AS1 is significantly higher in HCC tissue than in nontumor tissues. TCGA data show that the expression level of MAFG-AS1 is negatively correlated with survival of HCC patients. GEO cohort data show that compared with healthy tissues, the expression level of MAFG-AS1 is significantly higher in HBV-infected liver tissues. Real-time PCR and luciferase reporter assay data show that HBx can enhance the transcription of MAFG-AS1. Gain-of-function and loss-of-function experiments indicate that MAFG-AS1 promotes proliferation, migration, and invasion of HCC cells. Tumor formation assay results demonstrate that knockdown of MAFG-AS1 significantly inhibits cell proliferation in nude mice. Furthermore, MAFG-AS1 enhances the transcription of adjacent MAFG via E2F1. Additionally, MAFG-AS1 interacts with three subunits (MYH9, MYL12B, and MYL6) of nonmuscle myosin IIA (NM IIA). Knockdown of MAFG-AS1 inhibits ATPase activity of MYH9, interaction of NM IIA subunits, and cell cycle progression. Thus, the lncRNA MAFG-AS1 is upregulated by HBV and promotes proliferation and migration of HCC cells. Our findings suggest that MAFG-AS1 is a potential oncogene that may contribute to HBV-related HCC development.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , MafG Transcription Factor/metabolism , Nonmuscle Myosin Type IIA/chemistry , Repressor Proteins/metabolism , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MafG Transcription Factor/antagonists & inhibitors , MafG Transcription Factor/genetics , Nonmuscle Myosin Type IIA/genetics , Nonmuscle Myosin Type IIA/metabolism , Oligonucleotides, Antisense/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins/genetics
3.
Article in English | MEDLINE | ID: mdl-32371093

ABSTRACT

The farnesoid-X-receptor (FXR) is validated target in the cholestatic disorders treatment. Obeticholic acid (OCA), the first in class of FXR agonist approved for clinical use, causes side effects including acute liver decompensation when administered to cirrhotic patients with primary biliary cholangitis at higher than recommended doses. The V-Maf avian-musculoaponeurotic-fibrosarcoma-oncogene-homolog-G (Mafg) and nuclear factor-erythroid-2-related-factor-2 (Nrf2) mediates some of the downstream effects of FXR. In the present study we have investigated the role of FXR/MafG/NRF2 pathway in the development of liver toxicity caused by OCA in rodent models of cholestasis. Cholestasis was induced by bile duct ligation (BDL) or administration of α-naphtyl-isothiocyanate (ANIT) to male Wistar rats and FXR-/- and FXR+/+ mice. Treating BDL and ANIT rats with OCA exacerbated the severity of cholestasis, hepatocytes injury and severely downregulated the expression of basolateral transporters. In mice, genetic ablation FXR or its pharmacological inhibition by 3-(naphthalen-2-yl)-5-(piperidin-4-yl)-1,2,4-oxadiazole rescued from negative regulation of MRP4 and protected against liver injury caused by ANIT. By RNAseq analysis we found that FXR antagonism effectively reversed the transcription of over 2100 genes modulated by OCA/ANIT treatment, including Mafg and Nrf2 and their target genes Cyp7a1, Cyp8b1, Mat1a, Mat2a, Gss. Genetic and pharmacological Mafg inhibition by liver delivery of siRNA antisense or S-adenosylmethionine effectively rescued from damage caused by ANIT/OCA. In contrast, Nrf2 induction by sulforaphane was protective. CONCLUSIONS: Liver injury caused by FXR agonism in cholestasis is FXR-dependent and is reversed by FXR and Mafg antagonism or Nrf2 induction.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Cholestasis/metabolism , Liver Diseases/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Chenodeoxycholic Acid/pharmacology , Cholestasis/complications , Cholestasis/genetics , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Liver Diseases/etiology , Liver Diseases/genetics , MafG Transcription Factor/antagonists & inhibitors , MafG Transcription Factor/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/genetics , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics
4.
Transl Res ; 200: 1-17, 2018 10.
Article in English | MEDLINE | ID: mdl-30053382

ABSTRACT

Adjuvant chemotherapy for solid tumors based on platinum-derived compounds such as cisplatin is the treatment of choice in most cases. Cisplatin triggers signaling pathways that lead to cell death, but it also induces changes in tumor cells that modify the therapeutic response, thereby leading to cisplatin resistance. We have recently reported that microRNA-7 is silenced by DNA methylation and is involved in the resistance to platinum in cancer cells through the action of the musculoaponeurotic fibrosarcoma oncogene family, protein G (MAFG). In the present study, we first confirm the miR-7 epigenetic regulation of MAFG in 44 normal- and/or tumor-paired samples in non-small-cell lung cancer (NSCLC). We also provide translational evidence of the role of MAFG and the clinical outcome in NSCLC by the interrogation of two extensive in silico databases of 2019 patients. Moreover, we propose that MAFG-mediated resistance could be conferred due to lower reactive oxygen species production after cisplatin exposure. We developed specifically selected aptamers against MAFG, with high sensitivity to detect the protein at a nuclear level probed by aptacytochemistry and histochemistry analyses. The inhibition of MAFG activity through the action of the specific aptamer apMAFG6F increased the levels of reactive oxygen species production and the sensitivity to cisplatin. We report first the specific nuclear identification of MAFG as a novel detection method for diagnosis in NSCLC, and then we report that MAFG modulates the redox response and confers cell protection against free radicals generated after platinum administration, thus also being a promising therapeutic target.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , MafG Transcription Factor/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/pharmacology , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cloning, Molecular , DNA Methylation , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Epigenesis, Genetic/genetics , Gene Expression , Gene Silencing , HEK293 Cells , Humans , Lung Neoplasms/genetics , MafG Transcription Factor/genetics , MafG Transcription Factor/physiology , MicroRNAs/genetics , MicroRNAs/physiology , Oxidation-Reduction , Prognosis , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Repressor Proteins/physiology , Sequence Analysis, DNA , Transfection
5.
FEBS Lett ; 582(16): 2357-64, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18538669

ABSTRACT

We identified MafG as a protein that interacts with HIF-1alpha, a key factor in hypoxic response, using the yeast two-hybrid system. Interaction between MafG and HIF-1alpha was confirmed by surface plasmon resonance and by translocation to the nucleolus with the NoLS signal. A knockdown of MafG reduced erythropoietin (EPO) mRNA levels as well as luciferase reporter activity with the hypoxia response element. The knockdown of MafG did not change total HIF-1alpha protein, but reduced the accumulation of HIF-1alpha in the nuclei. These results suggest that MafG regulates the hypoxic response of cells by detaining HIF-1alpha in the nuclei.


Subject(s)
Cell Nucleus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MafG Transcription Factor/physiology , Repressor Proteins/physiology , Cell Hypoxia , Cell Line, Tumor , Genes, Reporter , Humans , MafG Transcription Factor/antagonists & inhibitors , MafG Transcription Factor/genetics , RNA Interference , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...