Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Clin. transl. oncol. (Print) ; 26(1): 52-68, jan. 2024.
Article in English | IBECS | ID: ibc-229146

ABSTRACT

The MAF bZIP transcription factor G-antisense RNA 1 (MAFG-AS1) is located on chromosome 17. MAFG-AS1 was upregulated in 15 human cancers. MAFG-AS1 not only suppresses 16 miRNAs but also directly impacts 22 protein-coding genes' expression. Notably, abnormal MAFG-AS1 expression is connected to clinicopathological characteristics and a worse prognosis in a variety of cancers. Moreover, MAFG-AS1 takes its part in the tumorigenesis and progression of various human malignancies by suppressing apoptosis and promoting proliferation, migration, invasion, aerobic glycolysis, ferroptosis, angiogenesis, EMT, and metastasis. Besides, it can predict treatment effectiveness in ER + breast cancer, urothelial bladder carcinoma, and liver cancer by functioning as a trigger of resistance to tamoxifen, sorafenib, and cisplatin. This study systematically presents the functions of MAFG-AS1 in various cancers, as well as the findings of bioinformatics analyses of the MAFG-AS1, which should give clear advice for future research (AU)


Subject(s)
Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic
2.
Clin Transl Oncol ; 26(1): 52-68, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37351806

ABSTRACT

The MAF bZIP transcription factor G-antisense RNA 1 (MAFG-AS1) is located on chromosome 17. MAFG-AS1 was upregulated in 15 human cancers. MAFG-AS1 not only suppresses 16 miRNAs but also directly impacts 22 protein-coding genes' expression. Notably, abnormal MAFG-AS1 expression is connected to clinicopathological characteristics and a worse prognosis in a variety of cancers. Moreover, MAFG-AS1 takes its part in the tumorigenesis and progression of various human malignancies by suppressing apoptosis and promoting proliferation, migration, invasion, aerobic glycolysis, ferroptosis, angiogenesis, EMT, and metastasis. Besides, it can predict treatment effectiveness in ER + breast cancer, urothelial bladder carcinoma, and liver cancer by functioning as a trigger of resistance to tamoxifen, sorafenib, and cisplatin. This study systematically presents the functions of MAFG-AS1 in various cancers, as well as the findings of bioinformatics analyses of the MAFG-AS1, which should give clear advice for future research.


Subject(s)
Breast Neoplasms , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Female , Carcinogens , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Antisense/genetics , Liver Neoplasms/genetics , Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Cell Movement/genetics , Repressor Proteins/genetics , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism
3.
Eur J Med Res ; 28(1): 497, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37941063

ABSTRACT

Long noncoding RNAs (lncRNAs) refer to a type of non-protein-coding transcript of more than 200 nucleotides. LncRNAs play fundamental roles in disease development and progression, and lncRNAs are dysregulated in many pathophysiological processes. Thus, lncRNAs may have potential value in clinical applications. The lncRNA, MAF BZIP Transcription Factor G (MAFG)-AS1, is dysregulated in several cancer, including breast cancer, lung cancer, liver cancer, bladder cancer, colorectal cancer, gastric cancer, esophagus cancer, prostate cancer, pancreatic cancer, ovarian cancer, and glioma. Altered MAFG-AS1 levels are also associated with diverse clinical characteristics and patient outcomes. Mechanistically, MAFG-AS1 mediates a variety of cellular processes via the regulation of target gene expression. Therefore, the diagnostic, prognostic, and therapeutic aspects of MAFG-AS1 have been widely explored. In this review, we discuss the expression, major roles, and molecular mechanisms of MAFG-AS1, the relationship between MAFG-AS1 and clinical features of diseases, and the clinical applications of MAFG-AS1.


Subject(s)
Breast Neoplasms , Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Male , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , MicroRNAs/genetics , Lung Neoplasms/genetics , Breast Neoplasms/genetics , Prognosis , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Cell Line, Tumor , Repressor Proteins/genetics , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism
4.
Growth Factors ; 41(3): 152-164, 2023 08.
Article in English | MEDLINE | ID: mdl-37428861

ABSTRACT

We investigated the mechanism of ETS-translocation variant 1 (ETV1)/lncRNA-MAFG-AS1 in pancreatic cancer (PC). MAFG-AS1 and ETV1 levels in PC cell lines and HPNE cells were determined using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting (WB). After transfection with sh-MAFG-AS1, PC cell invasion, migration, proliferation, and epithelial-mesenchymal transition (EMT)-related proteins were measured by 5-ethynyl-2'-deoxyuridine (EdU), Transwell assay, and WB. The binding between ETV1 and MAFG-AS1 was studied using dual-luciferase assay and chromatin immunoprecipitation. The interactions between MAFG-AS1, IGF2BP2, and ETV1 were tested. Combined experiments were further performed using sh-MAFG-AS1 and pcDNA-ETV1 simultaneously. ETV1/MAFG-AS1 was highly expressed in PC cells. Blocking MAFG-AS1 inhibited the malignant behaviors of PC cells. ETV1 induced MAFG-AS1 transcription in PC cells. MAFG-AS1 stabilized ETV1 mRNA by recruiting IGF2BP2. ETV1 overexpression partially antagonized the suppression of silencing MAFG-AS1 on PC cells. ETV1-induced MAFG-AS1 stabilized the ETV1 expression by recruiting IGF2BP2 and promoted PC cell migration, invasion, proliferation, and EMT.


Subject(s)
MicroRNAs , Pancreatic Neoplasms , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Transcription Factors/genetics , Transcription Factors/metabolism , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Repressor Proteins/genetics , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Pancreatic Neoplasms
5.
J Med Chem ; 66(9): 6184-6192, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37097833

ABSTRACT

Nuclear factor erythroid-related 2-factor 2 (Nrf2) is a transcription factor traditionally thought of as a cellular protector. However, in many cancers, Nrf2 is constitutively activated and correlated with therapeutic resistance. Nrf2 heterodimerizes with small musculoaponeurotic fibrosarcoma Maf (sMAF) transcription factors, allowing binding to the antioxidant responsive element (ARE) and induction of transcription of Nrf2 target genes. While transcription factors are historically challenging to target, stapled peptides have shown great promise for inhibiting these protein-protein interactions. Herein, we describe the first direct cell-permeable inhibitor of Nrf2/sMAF heterodimerization. N1S is a stapled peptide designed based on AlphaFold predictions of the interactions between Nrf2 and sMAF MafG. A cell-based reporter assay combined with in vitro biophysical assays demonstrates that N1S directly inhibits Nrf2/MafG heterodimerization. N1S treatment decreases the transcription of Nrf2-dependent genes and sensitizes Nrf2-dependent cancer cells to cisplatin. Overall, N1S is a promising lead for the sensitization of Nrf2-addicted cancers.


Subject(s)
NF-E2-Related Factor 2 , Repressor Proteins , NF-E2-Related Factor 2/metabolism , Repressor Proteins/metabolism , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism , Gene Expression Regulation , Peptides/metabolism
6.
J Med Chem ; 66(9): 6070-6081, 2023 05 11.
Article in English | MEDLINE | ID: mdl-36892138

ABSTRACT

Proteolysis-targeting chimera (PROTAC) technology has emerged as a potential strategy to degrade "undruggable" proteins in recent years. Nrf2, an aberrantly activated transcription factor in cancer, is generally considered undruggable as lacking active sites or allosteric pockets. Here, we constructed the chimeric molecule C2, which consists of an Nrf2-binding element and a CRBN ligand, as a first-in-class Nrf2 degrader. Surprisingly, C2 was found to selectively degrade an Nrf2-MafG heterodimer simultaneously via the ubiquitin-proteasome system. C2 impeded Nrf2-ARE transcriptional activity significantly and improved the sensitivity of NSCLC cells to ferroptosis and therapeutic drugs. The degradation character of ARE-PROTACs suggests that the PROTAC hijacking the transcription element of TFs could achieve co-degradation of the transcription complex.


Subject(s)
NF-E2-Related Factor 2 , Proteolysis Targeting Chimera , Gene Expression Regulation , NF-E2-Related Factor 2/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , MafG Transcription Factor/metabolism
7.
Pathol Res Pract ; 243: 154348, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36736142

ABSTRACT

Long non-coding RNAs (lncRNAs) have more than 200 nucleotides and do not encode proteins. At the same time, they can regulate various biological functions and therefore play an essential role as oncogenes or tumor suppressors in human cancers. MAFG-AS1 is an antisense RNA of MAF BZIP Transcription Factor G (MAFG) located at chromosome 17q25.3 head-to-head with the MAFG encoding gene containing a transcript size of 1895 bp. Accumulating evidence shows that MAFG-AS1 is overexpressed in many cancers, functions as an oncogene, and is significantly associated with poor clinical characteristics and prognosis. In this review, we first discuss the recent literature regarding the role of MAFG-AS1 in different cancers as well as its diagnostic and prognostic values. Then we will provide insights into its biological functions, such as its role in cancer progression, competing endogenous RNA (ceRNA) activity, regulation of EMT, glycolysis, energy metabolism, transcription factors, proteasomal degradation, and signaling pathways.


Subject(s)
MicroRNAs , Neoplasms , RNA, Long Noncoding , Humans , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Neoplasms/genetics , Oncogenes , RNA, Antisense/metabolism , RNA, Long Noncoding/metabolism , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Cell Proliferation/genetics , Repressor Proteins/genetics , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism
8.
Environ Sci Pollut Res Int ; 30(7): 19250-19258, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36229729

ABSTRACT

Recent references discovered that lncRNAs acted roles in malignant cancer development. However, the role of MAFG-AS1 in acute myeloid leukemia (AML) development remains unknown. MAFG-AS1 and miR-147b were determined in AML cells and specimens using qRT-PCR assay. Cell proliferation was detected by CCK-8 analysis and flow cytometry was carried out to measure cell cycle. Luciferase reporter analysis was done to determine the mechanism of MAFG-AS1 and miR-147b. We noted that MAFG-AS1 was overexpressed in AML cells and in serum and bone narrow from AML compared with normal controls specimen. Elevated expression of MAFG-AS1 increased cell growth, cycle and EMT in AML cell HL-60 cell. MAFG-AS1 sponged miR-147b expression in HL-60 cell. Moreover, miR-147b was downregulated in AML cells and in serum and bone narrow from AML compared with normal control specimen. miR-147b was negatively correlated with MAFG-AS1 in the serum and bone narrow of AML cases. We illustrated that HOXA9 was one target of miR-147b and ectopic expression of MAFG-AS1 enhanced HOXA9 expression HL-60 cell. Forced expression of MAFG-AS1 induced cell growth, cycle, and EMT via promoting HOXA9. These data illustrated that MAFG-AS1 acted as one oncogenic gene and accelerated AML progression via modulating miR-147b/HOXA9 axis.


Subject(s)
Leukemia, Myeloid, Acute , MicroRNAs , RNA, Long Noncoding , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Cell Movement/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Cell Proliferation/genetics , Cell Line, Tumor , Repressor Proteins/genetics , MafG Transcription Factor/genetics
9.
Acta Neurobiol Exp (Wars) ; 82(3): 315-326, 2022.
Article in English | MEDLINE | ID: mdl-36214714

ABSTRACT

This study was designed to explore the function of lncRNA MAFG­AS1/miR­642a­5p/Notch1 in glioblastoma (GBM) cells under radiation. GBM cells (M059K and M059J) were transfected or/and irradiated. Western blotting was used to detect Notch1 protein and its downstream Hes1 protein. Cell Counting Kit-8 assay and flow cytometry were applied for viability and apoptosis tests, respectively. Luciferase reporter plasmids and Ago2 antibody were used to verify the predicted binding of miR­642a­5p to MAFG­AS1 or Notch1 mRNA. Notch1 and MAFG­AS1 were highly expressed and miR­642a­5p was lowly expressed in radioresistant M059K cells. Knockdown of Notch1 inhibited radioresistance and promoted apoptosis in M059K cells. MAFG­AS1 competed with Notch1 mRNA for binding of miR­642a­5p and therefore promoted the expression of Notch1. Overexpression of miR­642a­5p or silencing of MAFG­AS1 inhibited the radioresistance of M059K cells. Knockdown of Notch1 or overexpression of miR­642a­5p reduced viability and increased apoptosis in irradiated M059J cells overexpressing MAFG­AS1. MAFG­AS1 reduces the radiosensitivity of GBM cells via miR­642a­5p/Notch1 axis.


Subject(s)
Glioblastoma , MicroRNAs , RNA, Long Noncoding , Receptor, Notch1 , Cell Line, Tumor , Cell Proliferation/genetics , Glioblastoma/genetics , Glioblastoma/radiotherapy , Humans , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Messenger , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Repressor Proteins
10.
Biochem Biophys Res Commun ; 616: 95-103, 2022 08 06.
Article in English | MEDLINE | ID: mdl-35653827

ABSTRACT

PURPOSE: We aimed to explore the function and competing endogenous RNA (ceRNA) pathway of MAFG-AS1 in breast cancer. METHODS: qRT-PCR assay identified the expression of MAFG-AS1, miR-3612 and FKBP4. We used Western blot analysis to test the autophagy related protein levels in breast cancer cells. Functional assays such as Cell Counting Kit-8 (CCK8) assay, BrdU proliferation assay, Caspase-3 activity detection were used to identify the function of MAFG-AS1, miR-3612 and FKBP4 in breast cancer cells. Mechanism assays were used to verify the interacting relationship among MAFG-AS1, miR-3612 and FKBP4, including RNA pull down assay, RNA immunoprecipitation (RIP) assay and luciferase reporter assay. RESULTS: MAFG-AS1 and FKBP4 were both up-regulated in breast cancer tissues. MAFG-AS1 could function as an oncogene in breast cancer to activate cell proliferation, and inhibit cell apoptosis and autophagy. Meanwhile, MAFG-AS1 could sponge miR-3612 to elevate the expression of FKBP4. Besides, FKBP4 could activate the cell proliferation and inhibit cell apoptosis and autophagy, which could relieve the inhibitory effect of miR-3612 on breast cancer cells. CONCLUSION: MAFG-AS1 could activate breast cancer progression via modulating miR-3612/FKBP4 axis in vitro.


Subject(s)
Breast Neoplasms , MicroRNAs , RNA, Long Noncoding , Tacrolimus Binding Proteins , Autophagy/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/genetics , Disease Progression , Female , Humans , MafG Transcription Factor/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Repressor Proteins/metabolism
11.
Oncogene ; 41(31): 3846-3858, 2022 07.
Article in English | MEDLINE | ID: mdl-35773412

ABSTRACT

Aldo-keto reductase family 1 member C3 (AKR1C3) serves as a contributor to numerous kinds of tumors, and its expression is elevated in patients with hepatocellular carcinoma (HCC). However, the biological function of AKR1C3 in HCC remains unclear. Here we investigated the role of AKR1C3 in liver carcinogenesis using in vitro and in vivo models. We determined that AKR1C3 is frequently increased in HCC tissues with poor prognosis. Genetically manipulated cells with AKR1C3 construction were examined to highlight the pro-tumoral growth of both wild-type AKR1C3 and mutant in vitro and in vivo. We observed promising treatment effects of AKR1C3 shRNA by intratumoral injection in mice. Mechanically, we demonstrated that the transcription factor heterodimer NRF2/MAFG was able to bind directly to AKR1C3 promoter to activate its transcription. Further, AKR1C3 stabilized PARP1 by decreasing its ubiquitination, which resulted in HCC cell proliferation and low sensitivity of Cisplatin. Moreover, we discovered that the tumorigenic role of AKR1C3 was non-catalytic dependent and the NRF2/MAFG-AKR1C3-PARP1 axis might be one of the important proliferation pathways in HCC. In conclusion, blockage of AKR1C3 expression provides potential therapeutic benefits against HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , 3-Hydroxysteroid Dehydrogenases/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Hydroxyprostaglandin Dehydrogenases/genetics , Liver Neoplasms/genetics , MafG Transcription Factor/metabolism , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Repressor Proteins/metabolism
12.
Mol Biotechnol ; 64(9): 970-983, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35275356

ABSTRACT

Lung adenocarcinoma (LUAD) patients exhibit poor prognosis, primarily due to metastasis. Emerging studies have demonstrated that long noncoding RNAs (lncRNAs) play critical roles in cancer progression and metastasis besides their physiological function. Here, we investigated the potential role of lncRNA MAF BZIP Transcription Factor G Antisense RNA 1 (MAFG-AS1) in LUAD metastasis by analyzing its expression in The Cancer Genome Atlas (TCGA) LUAD database, and its function in LUAD using in vitro and in vivo experiments. We performed bioinformatics analysis, western blotting, dual-luciferase reporter gene assay, RNA immunoprecipitation (RIP), and rescue assays to reveal the molecular mechanisms underlying MAFG-AS1 function. We observed augmented expression of MAFG-AS1 in LUAD tissues compared with normal adjacent tissues, and its association with poor prognosis. Furthermore, MAFG-AS1 overexpression promoted LUAD cell migration, proliferation, invasion, and epithelial mesenchymal transition (EMT). Besides, MAFG-AS1 also targeted miR-3196 directly by acting as an endogenous sponge, thereby rescuing the inhibition of SOX12, a target of miR-3196. Thus, the rescue assays demonstrated that MAFG-AS1 promotes cell migration, invasion, and EMT by modulating the miR-3196/SOX12 pathway. In conclusion, our findings suggest that MAFG-AS1/miR-3196/SOX12 axis regulates LUAD progression and is a potential therapeutic target for LUAD.


Subject(s)
Adenocarcinoma , Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Adenocarcinoma/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Repressor Proteins/genetics , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
13.
Mol Cell Biol ; 42(3): e0052021, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35129372

ABSTRACT

Members of the cap'n'collar (CNC) family of transcription factors, including Nrf1 and Nrf2, heterodimerize with small Maf (sMaf) proteins (MafF, MafG, and MafK) and regulate target gene expression through CNC-sMaf-binding elements (CsMBEs). We recently developed a unique tethered dimer assessment system combined with small Maf triple-knockout fibroblasts, which enabled the characterization of specific CNC-sMaf heterodimer functions. In this study, we evaluated the molecular function of the tethered Nrf1-MafG (T-N1G) heterodimer. We found that T-N1G activates the expression of proteasome subunit genes, well-known Nrf1 target genes, and binds specifically to CsMBEs in the proximity of these genes. T-N1G was also found to activate genes involved in proteostasis-related pathways, including endoplasmic reticulum-associated degradation, chaperone, and ubiquitin-mediated degradation pathways, indicating that the Nrf1-MafG heterodimer regulates a wide range of proteostatic stress response genes. By taking advantage of this assessment system, we found that Nrf1 has the potential to activate canonical Nrf2 target cytoprotective genes when strongly induced. Our results also revealed that transposable SINE B2 repeats harbor CsMBEs with high frequency and contribute to the target gene diversity of CNC-sMaf transcription factors.


Subject(s)
MafG Transcription Factor , NF-E2-Related Factor 2 , Endoplasmic Reticulum-Associated Degradation , MafG Transcription Factor/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Repressor Proteins/metabolism
14.
Cancer Gene Ther ; 29(3-4): 277-291, 2022 03.
Article in English | MEDLINE | ID: mdl-34035482

ABSTRACT

Long non-coding RNAs (lncRNAs) were recently recognized to vitally function in a variety of cancer cellular events, including epithelial-mesenchymal transition (EMT), invasion, and migration, particularly in ovarian cancer (OC). Herein, we sought to investigate the potential role of MAFG-AS1 in the malignant behaviors of OC cells. The binding affinity between MAFG-AS1, miR-339-5p, NFKB1 or IGF1 was characterized so as to identify the underlying mechanism of corresponding their interactions. We conducted MAFG-AS1 overexpression or knockdown along with NFKB1 and IGF1 silencing to examine their effects on the EMT, migration, and invasion of OC cells. Tumors were xenografted in nude mice to validate the in vitro findings. Our data showed significantly high expression pattern of MAFG-AS1 in the OC tissues and cells. Further mechanistic investigations revealed that MAFG-AS1 upregulated the IGF1 expression pattern through recruitment of NFKB1, whereas MAFG-AS1 upregulated the NFKB1 expression pattern through binding to miR-339-5p. Thus, MAFG-AS1 overexpression accelerated the EMT, invasion, and migration of OC cells, which could be annulled by silencing of IGF1 or NFKB1. Besides, our in vitro findings were successfully recapitulated in the xenograft mice. These results determined that MAFG-AS1 stimulated the OC malignant progression by upregulating the NFKB1-mediated IGF1 via miR-339-5p, thus highlighting a novel potential therapeutic target against OC.


Subject(s)
MicroRNAs , Ovarian Neoplasms , RNA, Long Noncoding , Animals , Carcinoma, Ovarian Epithelial/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor I , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B p50 Subunit , Neoplasm Invasiveness/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phenotype , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Repressor Proteins/genetics
15.
Crit Rev Eukaryot Gene Expr ; 31(5): 27-32, 2021.
Article in English | MEDLINE | ID: mdl-34591387

ABSTRACT

Gastric cancer is a commonly diagnosed, often fatal malignancy and requires novel anticancer therapies and preventative approaches. This study described the involvement of MAFG-AS1, a lncRNA with important functions in cancer biology, in gastric adenocarcinoma (GA). Thirty-six male and forty-two female GA patients with an average age of 51.9 ± 5.7 years in the range of 35 to 68 years were enrolled. Paired gastric cancer (GC) and non-tumor tissues were collected from each patient. MAFG-AS1 expression was determined. RNA interaction prediction, dual luciferase reporter assay, RT-qPCR assay, Western blot, and CCK-8 assay were conducted. The results indicated that MAFG-AS1 was highly expressed in GA and closely correlated with poor survival. MAFG-AS1 interacted with miR-505, but MAFG-AS1 and miR-505 overexpression showed no significant effects on each other's expression. In addition, MAFG-AS1 increased the expression of PLK1, a miR-505 target. MAFG-AS1 and PLK1 overexpression increased GC cell proliferation rate. MiR-505 overexpression reduced the effects of MAFG-AS1 and PLK1 overexpression on cell proliferation. Therefore, MAFG-AS1 might upregulate PLK1 by sponging miR-505 to promote GA cell proliferation.


Subject(s)
Cell Cycle Proteins/metabolism , MafG Transcription Factor/metabolism , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Long Noncoding/metabolism , Repressor Proteins/metabolism , Stomach Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adult , Aged , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , MafG Transcription Factor/genetics , Male , Middle Aged , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Stomach Neoplasms/genetics , Polo-Like Kinase 1
16.
BMC Cardiovasc Disord ; 21(1): 448, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34535081

ABSTRACT

BACKGROUND: Vascular endothelial cell apoptosis is the leading risk factor of atherosclerosis (AS). The purpose of our study was to use a new generation high-throughput transcription factor (TF) detection method to identify novel key TFs in vascular endothelial cell apoptosis induced by palmitic acid (PA). METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with 0, 300, or 500 µM PA. Candidate TFs in the three groups were identified by differential expression, pathway enrichment, Western Blot (WB), and RT-qPCR analyses. Apoptosis was assessed by fluorescence-activated cell sorting (FACS) using FITC-annexin V and propidium iodide staining. RESULTS: We established a HUVEC apoptosis model to simulate the process of atherosclerosis onset and identified 51 significant TFs. of the 51 TFs, v-maf musculoaponeurotic fibrosarcoma oncogene family protein G (MAFG) and v-maf musculoaponeurotic fibrosarcoma oncogene family protein F (MAFF), were matched to known AS signalling pathways and were validated by WB and RT-qPCR analyses in our study. Overexpression of MAFG or MAFF in HUVECs significantly inhibited PA-induced early apoptosis. CONCLUSIONS: We identified MAFF and MAFG as novel key TFs in vascular endothelial cell apoptosis.


Subject(s)
Apoptosis/drug effects , Atherosclerosis/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , MafF Transcription Factor/metabolism , MafG Transcription Factor/metabolism , Nuclear Proteins/metabolism , Palmitic Acid/toxicity , Proteome , Proteomics , Repressor Proteins/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Chromatography, Liquid , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , MafF Transcription Factor/genetics , MafG Transcription Factor/genetics , Nuclear Proteins/genetics , Protein Interaction Maps , Repressor Proteins/genetics , Signal Transduction , Tandem Mass Spectrometry , Transcription, Genetic
17.
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
18.
Biochem Biophys Res Commun ; 555: 175-181, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33819748

ABSTRACT

Microgravity and radiation exposure-induced bone damage is one of the most significant alterations in astronauts after long-term spaceflight. However, the underlying mechanism is still largely unknown. Recent ground-based simulation studies have suggested that this impairment is likely mediated by increased production of reactive oxygen species (ROS) during spaceflight. The small Maf protein MafG is a basic-region leucine zipper-type transcription factor, and it globally contributes to regulation of antioxidant and metabolic networks. Our research investigated the role of MafG in the process of apoptosis induced by simulated microgravity and radiation in MC3T3-E1 cells. We found that simulated microgravity or radiation alone decreased MafG expression and elevated apoptosis in MC3T3-E1 cells, and combined simulated microgravity and radiation treatment aggravated apoptosis. Meanwhile, under normal conditions, increased ROS levels facilitated apoptosis and downregulated the expression of MafG in MC3T3-E1 cells. Overexpression of MafG decreased apoptosis induced by simulated microgravity and radiation. These findings provide new insight into the mechanism of bone damage induced by microgravity and radiation during space flight.


Subject(s)
Apoptosis/radiation effects , MafG Transcription Factor/metabolism , Osteoblasts/cytology , Osteoblasts/radiation effects , Repressor Proteins/metabolism , Apoptosis/physiology , Cell Line , Down-Regulation , Gene Expression Regulation/radiation effects , Humans , MafG Transcription Factor/genetics , Osteoblasts/physiology , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Weightlessness Simulation , X-Rays
19.
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
20.
Hum Cell ; 34(2): 588-597, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33400245

ABSTRACT

MAFG-AS1 is an oncogenic lncRNA in multiple types of cancer. However, its role in bladder cancer (BC) remains unclear. The present study aimed to investigate the function of MAFG-AS1 in BC. BC and paired non-tumor tissues were collected. Two BC cell lines HT01197 and HT-1376 were used. Dual luciferase activity assay, RT-qPCR, western blot, CCK-8, transwell invasion assay, and wound healing assay were performed. We found that MAFG-AS1 was significantly up-regulated in BC tissues and predicted a poor survival rate. MAFG-AS1 interacted with miR-125b-5p. However, the expression levels of MAFG­AS1 and miR-125b-5p were not obviously correlated in BC tissues, and MAFG­AS1 and miR-125b-5p did not regulate the expression of each other. Interestingly, we found that SphK1, a downstream target of miR-125b-5p, was negatively correlated with miR-125b-5p, while it was positively correlated with MAFG-AS1 across BC tissues. In addition, overexpression of MAFG­AS1 upregulated the expression of SphK1 in BC cells, and attenuated the inhibitory effects of miR-125b-5p on the expression of SphK1. Functional assays showed that overexpression of MAFG­AS1 promoted BC cell proliferation, migration, and invasion, while its effects were attenuated by overexpression of miR-125b-5p. Moreover, overexpression of miR-125b-5p inhibited BC cell proliferation, migration, and invasion, while its effects were alleviated by overexpression of SphK1. Taken together, our findings demonstrated that MAFG-AS1 has an oncogenic role in BC by regulating the miR-125b-5p/SphK1 axis. MAFG-AS1 might serve as a good diagnostic marker and a potential therapeutic target of BC.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , MafG Transcription Factor/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA, Long Noncoding/physiology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Molecular Targeted Therapy , Urinary Bladder Neoplasms/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...