General Information of the Disease (ID: M6ADIS0001)
Name
Brain cancer
ICD
ICD-11: 2A00
Full List of Target Gene(s) of This m6A-centered Disease Response
Beta-catenin-interacting protein 1 (CTNNBIP1)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [1]
Response Summary METTL3-mediated m6A modification upregulated circDLC1 expression, and circDLC1 promoted Beta-catenin-interacting protein 1 (CTNNBIP1) transcription by sponging miR-671-5p, thus repressing the malignant proliferation of glioma.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
In-vitro Model T98G Glioblastoma Homo sapiens CVCL_0556
LN-229 Glioblastoma Homo sapiens CVCL_0393
LN-18 Glioblastoma Homo sapiens CVCL_0392
HEB (human normal glial cell line HEB were obtained from Tongpai (Shanghai) biotechnology co., LTD (Shanghai, China))
A-172 Glioblastoma Homo sapiens CVCL_0131
CCAAT/enhancer-binding protein alpha (CEBPA)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [2]
Response Summary This work demonstrates anti-tumor effects of 2HG in inhibiting proliferation/survival of FTO-high cancer cells via targeting FTO/m6A/MYC/CCAAT/enhancer-binding protein alpha (CEBPA) signaling.High levels of FTO sensitize leukemia cells to R-2HG, whereas hyperactivation of MYC signaling confers resistance that can be reversed by the inhibition of MYC signaling. R-2HG also displays anti-tumor activity in glioma. High levels of FTO sensitize leukemic cells to R-2HG, whereas hyperactivation of MYC signaling confers resistance that can be reversed by the inhibition of MYC signaling.
Responsed Disease Glioma [ICD-11: 2A00.0]
Responsed Drug R-2HG Investigative
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Down regulation
Cell Process Glutamine metabolism
Cell apoptosis
In-vitro Model 8-MG-BA Glioblastoma Homo sapiens CVCL_1052
A-172 Glioblastoma Homo sapiens CVCL_0131
DK-MG Glioblastoma Homo sapiens CVCL_1173
GaMG Glioblastoma Homo sapiens CVCL_1226
HEL Erythroleukemia Homo sapiens CVCL_0001
Jurkat T acute lymphoblastic leukemia Homo sapiens CVCL_0065
KOCL-45 B acute lymphoblastic leukemia Homo sapiens CVCL_3993
KOCL-48 Childhood acute monocytic leukemia Homo sapiens CVCL_6867
KOCL-50 B acute lymphoblastic leukemia Homo sapiens CVCL_6866
KOCL-51 B acute lymphoblastic leukemia Homo sapiens CVCL_6865
KOCL-69 B acute lymphoblastic leukemia Homo sapiens CVCL_3995
KOPN-1 B acute lymphoblastic leukemia Homo sapiens CVCL_3937
LN-18 Glioblastoma Homo sapiens CVCL_0392
LN-229 Glioblastoma Homo sapiens CVCL_0393
MA9.3 (MA9.3)
MA9.6ITD (MLL-AF9 plus FLT3-ITD)
MA9.6RAS (MLL-AF9 plus NRasG12D)
MA9.6 (MLL-AF9)
MA9.6ITD (MLL-AF9 plus FLT3-ITD)
MA9.6RAS (MLL-AF9 plus NRasG12D)
ME-1 [Human leukemia] Adult acute myeloid leukemia Homo sapiens CVCL_2110
ML-2 Adult acute myeloid leukemia Homo sapiens CVCL_1418
MV4-11 Childhood acute monocytic leukemia Homo sapiens CVCL_0064
NB4 Acute promyelocytic leukemia Homo sapiens CVCL_0005
NOMO-1 Adult acute monocytic leukemia Homo sapiens CVCL_1609
PL21 Familial adenomatous polyposis Homo sapiens CVCL_JM48
T98G Glioblastoma Homo sapiens CVCL_0556
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
U-937 Adult acute monocytic leukemia Homo sapiens CVCL_0007
In-vivo Model For R-2HG injection mouse models, sensitive (NOMO-1 and MA9.3ITD) or resistant (MA9.3RAS) cells were injected into NSGS or NRGS intravenously, and then R-2HG (6mg/kg body weight) or PBS were injected once daily through tail vein for 12 consecutive days starting from day 11 post xeno-transplantation.
CD44 antigen (CD44)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [3]
Response Summary HNRNPA2B1 and HNRNPC were extensively expressed in the Glioblastoma multiforme(GBM) microenvironment.m6A regulators promoted the stemness state in GBM cancer cells. Cell communication analysis identified genes in the GALECTIN signaling network in GBM samples, and expression of these genes (LGALS9, CD44 antigen (CD44), CD45, and HAVCR2) correlated with that of m6A regulators.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Heterogeneous nuclear ribonucleoproteins A2/B1 (HNRNPA2B1) READER
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [3]
Response Summary HNRNPA2B1 and HNRNPC were extensively expressed in the Glioblastoma multiforme(GBM) microenvironment. m6A regulators promoted the stemness state in GBM cancer cells. Cell communication analysis identified genes in the GALECTIN signaling network in GBM samples, and expression of these genes (LGALS9, CD44 antigen (CD44), CD45, and HAVCR2) correlated with that of m6A regulators.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Heterogeneous nuclear ribonucleoproteins C1/C2 (HNRNPC) READER
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Cystine/glutamate transporter (SLC7A11)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [4]
Response Summary This study NKAP knockdown induced cell death in glioblastoma cells. NKAP acted as a new ferroptosis suppressor by binding to m6A and then promoting Cystine/glutamate transporter (SLC7A11) mRNA splicing and maturation.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator NF-kappa-B-activating protein (NKAP) READER
Target Regulation Up regulation
Pathway Response Ferroptosis hsa04216
Cell Process Ferroptosis
In-vitro Model U87MG (Astroblastoma cells from human brain)
U251 (Fibroblasts or fibroblast like cells)
In-vivo Model The male BALB/c nude mice were randomized divide into two groups, each group including six 4 weeks old nude mice. Investigators were blinded to the treatment groups during data collection and subsequent data analysis. In the subcutaneous xenograft model, 5 × 105 cells were subcutaneously injected in the right flanks of nude mice. In the orthotopic intracranial mouse model, each mouse was intracranially injected with 1 × 105 luciferase transfected U87MG cells in 10 uL PBS solution.
DNA-3-methyladenine glycosylase (ANPG/MPG)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [5]
Response Summary Two critical DNA repair genes (MGMT and DNA-3-methyladenine glycosylase (ANPG/MPG)) were m6A-modified by METTL3, whereas inhibited by METTL3 silencing or DAA-mediated total methylation inhibition, which is crucial for METTL3-improved temozolomide resistance in glioblastoma cells.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Responsed Drug Temozolomide Approved
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Down regulation
Pathway Response Nucleotide excision repair hsa03420
Cell Process DNA repair
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model Subcutaneously injected shMETTL3 or shNC-expressing U87-MG-TMZ cells into BALB/c NOD mice. After confirmation of GBM implantation, mice were treated with TMZ (66 mg/kg/d, 5 d per week, for 3 cycles).
Ephrin type-B receptor 3 (EPHB3)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [6]
Response Summary YTHDF2 enhanced TMZ resistance in GBM by activation of the PI3K/Akt and NF-Kappa-B signalling pathways via inhibition of Ephrin type-B receptor 3 (EPHB3) and TNFAIP3.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Responsed Drug Temozolomide Approved
Target Regulator YTH domain-containing family protein 2 (YTHDF2) READER
Target Regulation Down regulation
Pathway Response PI3K-Akt signaling pathway hsa04151
Cell Process RNA stability
In-vitro Model T98G Glioblastoma Homo sapiens CVCL_0556
LN-229 Glioblastoma Homo sapiens CVCL_0393
In-vivo Model 5 × 106 infected T98G cells (LV-NC or LV-YTHDF2) were injected into the flanks of mice through subcutaneous.
Forkhead box protein M1 (FOXM1)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [7]
Response Summary ALKBH5 and FOXM1-AS disrupted GSC tumorigenesis through the Forkhead box protein M1 (FOXM1) axis.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator RNA demethylase ALKBH5 (ALKBH5) ERASER
Target Regulation Up regulation
Pathway Response Glioma hsa05214
Cell Process Cells proliferation
Signaling pathways regulating pluripotency of stem cells (hsa04550)
In-vitro Model LN-229 Glioblastoma Homo sapiens CVCL_0393
Hs 683 Oligodendroglioma Homo sapiens CVCL_0844
SW1783 Anaplastic astrocytoma Homo sapiens CVCL_1722
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model For the animal survival analysis, mice were intracranially injected with 10,000 GSCs and maintained until moribund or 80 days after injection. For the rescue studies, GSCs with ALKBH5 or FOXM1-AS shRNAs were co-transfected with a FOXM1, ALKBH5 wild-type or mutant expression construct.
Galectin-9 (LGALS9)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [3]
Response Summary HNRNPA2B1 and HNRNPC were extensively expressed in the Glioblastoma multiforme(GBM) microenvironment.m6A regulators promoted the stemness state in GBM cancer cells. Cell communication analysis identified genes in the GALECTIN signaling network in GBM samples, and expression of these genes (Galectin-9 (LGALS9), CD44, CD45, and HAVCR2) correlated with that of m6A regulators.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Heterogeneous nuclear ribonucleoproteins A2/B1 (HNRNPA2B1) READER
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [3]
Response Summary HNRNPA2B1 and HNRNPC were extensively expressed in the Glioblastoma multiforme(GBM) microenvironment. m6A regulators promoted the stemness state in GBM cancer cells. Cell communication analysis identified genes in the GALECTIN signaling network in GBM samples, and expression of these genes (Galectin-9 (LGALS9), CD44, CD45, and HAVCR2) correlated with that of m6A regulators.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Heterogeneous nuclear ribonucleoproteins C1/C2 (HNRNPC) READER
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Heat shock protein HSP 90-alpha (HSP90/HSP90AA1)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [8]
Response Summary m6A regulated cell proliferation by influencing apoptosis of U251 cells through regulating Heat shock protein HSP 90-alpha (HSP90/HSP90AA1) expression. m6A level was decreased in glioma tissue, which was caused by decreased METTL3 and increased FTO levels.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Down regulation
Cell Process Cell migration and proliferation
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [8]
Response Summary m6A regulated cell proliferation by influencing apoptosis of U251 cells through regulating Heat shock protein HSP 90-alpha (HSP90/HSP90AA1) expression.m6A level was decreased in glioma tissue, which was caused by decreased METTL3 and increased FTO levels.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Down regulation
Cell Process Cell migration and proliferation
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
Hepatitis A virus cellular receptor 2 (HAVCR2)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [3]
Response Summary HNRNPA2B1 and HNRNPC were extensively expressed in the Glioblastoma multiforme(GBM) microenvironment.m6A regulators promoted the stemness state in GBM cancer cells. Cell communication analysis identified genes in the GALECTIN signaling network in GBM samples, and expression of these genes (LGALS9, CD44, CD45, and Hepatitis A virus cellular receptor 2 (HAVCR2)) correlated with that of m6A regulators.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Heterogeneous nuclear ribonucleoproteins A2/B1 (HNRNPA2B1) READER
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [3]
Response Summary HNRNPA2B1 and HNRNPC were extensively expressed in the Glioblastoma multiforme(GBM) microenvironment. m6A regulators promoted the stemness state in GBM cancer cells. Cell communication analysis identified genes in the GALECTIN signaling network in GBM samples, and expression of these genes (LGALS9, CD44, CD45, and Hepatitis A virus cellular receptor 2 (HAVCR2)) correlated with that of m6A regulators.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Heterogeneous nuclear ribonucleoproteins C1/C2 (HNRNPC) READER
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Homeobox protein NANOG (NANOG)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [9]
Response Summary In glioma, hsa_circ_0072083 could regulate Homeobox protein NANOG (NANOG) and ALKBH5 via targeting miR-1252-5p to control temozolomide resistance. circ_0072083 silence reduced NANOG expression via blocking ALKBH5-mediated demethylation.
Responsed Disease Glioma [ICD-11: 2A00.0]
Responsed Drug Temozolomide Approved
Target Regulator RNA demethylase ALKBH5 (ALKBH5) ERASER
Target Regulation Up regulation
Cell Process Cellular Processes
Cell growth and death
Cell apoptosis
In-vitro Model HEK293T Normal Homo sapiens CVCL_0063
U251 (Fibroblasts or fibroblast like cells)
U87 (A primary glioblastoma cell line)
In-vivo Model U251/TR cells (2 × 106 per mouse) with stable transfection of sh-circ_0072083 or sh-NC were subcutaneously injected into mice.
Interferon-inducible protein 4 (ADAR1)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [10]
Response Summary METTL3, upregulated in glioblastoma, methylates Interferon-inducible protein 4 (ADAR1) mRNA and increases its protein level leading to a pro-tumorigenic mechanism connecting METTL3, YTHDF1, and ADAR1.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
Pathway Response mRNA surveillance pathway hsa03015
RNA degradation hsa03018
Cell Process RNA stability
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
U-118MG Astrocytoma Homo sapiens CVCL_0633
T98G Glioblastoma Homo sapiens CVCL_0556
A-172 Glioblastoma Homo sapiens CVCL_0131
In-vivo Model 2 × 106 U87MG cells already expressing shscr or shADAR1 were subcutaneously injected in the flank of 6-week-old nude mice (nu/nu, Charles River, Wilmington, MA, USA).
Interleukin enhancer-binding factor 3 (ILF3)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [11]
Response Summary loss of m6A RNA methylation and increased translation in human glioblastoma cells as well as a role for miRNAs in the modulation of m6A RNA demethylation in genes that are most efficiently translated during glioma stem cells differentiation. Ectopic expression of the RRACH-binding miR-145 induces loss of m6A, formation of FTO/AGO1/Interleukin enhancer-binding factor 3 (ILF3)/miR-145 complexes on a clinically relevant tumor suppressor gene (CLIP3) and significant increase in its nascent translation.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Up regulation
In-vitro Model GBM1 Glioblastoma Homo sapiens CVCL_DG57
GBM2 Glioblastoma Homo sapiens CVCL_DG58
GBM3 Glioblastoma Homo sapiens CVCL_DG59
Meltrin-beta (ADAM19)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [12]
Response Summary Knockdown of METTL3 or METTL14 induced changes in mRNA m6A enrichment and altered mRNA expression of genes (e.g., Meltrin-beta (ADAM19)) with critical biological functions in GSCs. Treatment with MA2, a chemical inhibitor of FTO, dramatically suppressed GSC-induced tumorigenesis and prolonged lifespan in GSC-grafted animals.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Down regulation
Cell Process Cells growth
Cells self-renewal
Tumorigenesis
MicroRNAs in cancer (hsa05206)
In-vitro Model GSC Glioma Epinephelus akaara CVCL_M752
In-vivo Model 2 × 105 dissociated cells in 2 uL PBS were injected into the following site (anteroposterior [AP] +0.6 mm, mediolateral [ML] +1.6 mm, and dorsoventricular [DV] 2.6 mm) with a rate of 1 uL/min.
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [12]
Response Summary Knockdown of METTL3 or METTL14 induced changes in mRNA m6A enrichment and altered mRNA expression of genes (e.g., Meltrin-beta (ADAM19)) with critical biological functions in GSCs. Treatment with MA2, a chemical inhibitor of FTO, dramatically suppressed GSC-induced tumorigenesis and prolonged lifespan in GSC-grafted animals.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Methyltransferase-like 14 (METTL14) WRITER
Target Regulation Down regulation
Cell Process Cells growth
Cells self-renewal
Tumorigenesis
MicroRNAs in cancer (hsa05206)
In-vitro Model GSC Glioma Epinephelus akaara CVCL_M752
In-vivo Model 2 × 105 dissociated cells in 2 uL PBS were injected into the following site (anteroposterior [AP] +0.6 mm, mediolateral [ML] +1.6 mm, and dorsoventricular [DV] 2.6 mm) with a rate of 1 uL/min.
Methylated-DNA--protein-cysteine methyltransferase (MGMT)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [5]
Response Summary Two critical DNA repair genes (Methylated-DNA--protein-cysteine methyltransferase (MGMT) and APNG) were m6A-modified by METTL3, whereas inhibited by METTL3 silencing or DAA-mediated total methylation inhibition, which is crucial for METTL3-improved temozolomide resistance in glioblastoma cells.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Responsed Drug Temozolomide Approved
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Down regulation
Pathway Response Nucleotide excision repair hsa03420
Cell Process DNA repair
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model Subcutaneously injected shMETTL3 or shNC-expressing U87-MG-TMZ cells into BALB/c NOD mice. After confirmation of GBM implantation, mice were treated with TMZ (66 mg/kg/d, 5 d per week, for 3 cycles).
Myc proto-oncogene protein (MYC)
In total 5 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [13]
Response Summary The IGF1/IGF1R inhibitor, linsitinib for further investigation based upon the role of the IGF pathway member, IGFBP3, as a downstream effector of YTHDF2-Myc proto-oncogene protein (MYC) axis in GSCs. Inhibiting glioblastoma stem cells viability without affecting NSCs and impairing in vivo glioblastoma growth.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Responsed Drug Linsitinib Phase 3
Target Regulator YTH domain-containing family protein 2 (YTHDF2) READER
Target Regulation Up regulation
In-vitro Model NSC11 (Pluripotent derived neural progenitor cell)
NHA (Normal human astrocytes)
HNP1 (A human neural progenitor cell)
ENSA (A human embryonic stem derived neural progenitor cell)
In-vivo Model Implanting 5000 human derived GSCs into the right cerebral cortex of NSG mice at a depth of 3.5 mm under a University of California, San Diego Institutional Animal Care and Use Committee (IACUC) approved protocol. Brains were harvested and fixed in 4% formaldehyde, cryopreserved in 30% sucrose, and then cryosectioned. Hematoxylin and eosin (H&E) staining was performed on sections for histological analysis. In parallel survival experiments, mice were observed until the development of neurological signs. For in vivo drug treatment studies, intracranial xenografts were generated by implanting 5000 patient-derived GSCs (387 and 4121) into the right cerebral cortex of NSG mice as described above. Mice recovered for 7 days were randomly assigned into drug vs. treatment group by a blinded investigator. Mice were then treated daily with either vehicle (25 mM Tartaric acid) or 50 mg/kg linsitinib by oral gavage.
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [2]
Response Summary This work demonstrates anti-tumor effects of 2HG in inhibiting proliferation/survival of FTO-high cancer cells via targeting FTO/m6A/Myc proto-oncogene protein (MYC)/CEBPA signaling.High levels of FTO sensitize leukemia cells to R-2HG, whereas hyperactivation of MYC signaling confers resistance that can be reversed by the inhibition of MYC signaling. R-2HG also displays anti-tumor activity in glioma. High levels of FTO sensitize leukemic cells to R-2HG, whereas hyperactivation of MYC signaling confers resistance that can be reversed by the inhibition of MYC signaling.
Responsed Disease Glioma [ICD-11: 2A00.0]
Responsed Drug R-2HG Investigative
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Down regulation
Cell Process Glutamine metabolism
Cell apoptosis
In-vitro Model 8-MG-BA Glioblastoma Homo sapiens CVCL_1052
A-172 Glioblastoma Homo sapiens CVCL_0131
DK-MG Glioblastoma Homo sapiens CVCL_1173
GaMG Glioblastoma Homo sapiens CVCL_1226
HEL Erythroleukemia Homo sapiens CVCL_0001
Jurkat T acute lymphoblastic leukemia Homo sapiens CVCL_0065
KOCL-45 B acute lymphoblastic leukemia Homo sapiens CVCL_3993
KOCL-48 Childhood acute monocytic leukemia Homo sapiens CVCL_6867
KOCL-50 B acute lymphoblastic leukemia Homo sapiens CVCL_6866
KOCL-51 B acute lymphoblastic leukemia Homo sapiens CVCL_6865
KOCL-69 B acute lymphoblastic leukemia Homo sapiens CVCL_3995
KOPN-1 B acute lymphoblastic leukemia Homo sapiens CVCL_3937
LN-18 Glioblastoma Homo sapiens CVCL_0392
LN-229 Glioblastoma Homo sapiens CVCL_0393
MA9.3 (MA9.3)
MA9.6ITD (MLL-AF9 plus FLT3-ITD)
MA9.6RAS (MLL-AF9 plus NRasG12D)
MA9.6 (MLL-AF9)
MA9.6ITD (MLL-AF9 plus FLT3-ITD)
MA9.6RAS (MLL-AF9 plus NRasG12D)
ME-1 [Human leukemia] Adult acute myeloid leukemia Homo sapiens CVCL_2110
ML-2 Adult acute myeloid leukemia Homo sapiens CVCL_1418
MV4-11 Childhood acute monocytic leukemia Homo sapiens CVCL_0064
NB4 Acute promyelocytic leukemia Homo sapiens CVCL_0005
NOMO-1 Adult acute monocytic leukemia Homo sapiens CVCL_1609
PL21 Familial adenomatous polyposis Homo sapiens CVCL_JM48
T98G Glioblastoma Homo sapiens CVCL_0556
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
U-937 Adult acute monocytic leukemia Homo sapiens CVCL_0007
In-vivo Model For R-2HG injection mouse models, sensitive (NOMO-1 and MA9.3ITD) or resistant (MA9.3RAS) cells were injected into NSGS or NRGS intravenously, and then R-2HG (6mg/kg body weight) or PBS were injected once daily through tail vein for 12 consecutive days starting from day 11 post xeno-transplantation.
Experiment 3 Reporting the m6A-centered Disease Response by This Target Gene [13]
Response Summary YTHDF2 depletion downregulated IGFBP3 mRNA and protein levels, without affecting its mRNA stability. YTHDF2 regulated IGFBP3 levels via Myc proto-oncogene protein (MYC) in glioblastoma stem cells.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Insulin-like growth factor-binding protein 3 (IGFBP3) READER
In-vitro Model ENSA (A human embryonic stem derived neural progenitor cell)
HNP1 (A human neural progenitor cell)
NSC11 (Pluripotent derived neural progenitor cell)
NHA (Normal human astrocytes)
In-vivo Model Implanting 5000 human derived GSCs into the right cerebral cortex of NSG mice at a depth of 3.5 mm under a University of California, San Diego Institutional Animal Care and Use Committee (IACUC) approved protocol. Brains were harvested and fixed in 4% formaldehyde, cryopreserved in 30% sucrose, and then cryosectioned. Hematoxylin and eosin (H&E) staining was performed on sections for histological analysis. In parallel survival experiments, mice were observed until the development of neurological signs. For in vivo drug treatment studies, intracranial xenografts were generated by implanting 5000 patient-derived GSCs (387 and 4121) into the right cerebral cortex of NSG mice as described above. Mice recovered for 7 days were randomly assigned into drug vs. treatment group by a blinded investigator. Mice were then treated daily with either vehicle (25 mM Tartaric acid) or 50 mg/kg linsitinib by oral gavage.
Experiment 4 Reporting the m6A-centered Disease Response by This Target Gene [13]
Response Summary The m6A reader YTHDF2 stabilized Myc proto-oncogene protein (MYC) mRNA specifically in cancer stem cells. Given the challenge of targeting MYC, YTHDF2 presents a therapeutic target to perturb MYC signaling in glioblastoma. The IGF1/IGF1R inhibitor linsitinib preferentially targeted YTHDF2-expressing cells, inhibiting GSC viability without affecting NSCs and impairing in vivo glioblastoma growth. YTHDF2 links RNA epitranscriptomic modifications and GSC growth, laying the foundation for the YTHDF2-MYC-IGFBP3 axis as a specific and novel therapeutic target in glioblastoma.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator YTH domain-containing family protein 2 (YTHDF2) READER
Target Regulation Up regulation
Pathway Response RNA degradation hsa03018
Cell Process RNA stability
In-vitro Model ()
HNP1 (A human neural progenitor cell)
NHA (Normal human astrocytes)
NSC11 (Pluripotent derived neural progenitor cell)
In-vivo Model For in vivo drug treatment studies, intracranial xenografts were generated by implanting 5000 patient-derived GSCs (387 and 4121) into the right cerebral cortex of NSG mice as described above.
Experiment 5 Reporting the m6A-centered Disease Response by This Target Gene [13]
Response Summary The m6A reader YTHDF2 stabilized Myc proto-oncogene protein (MYC) mRNA specifically in cancer stem cells. Given the challenge of targeting MYC, YTHDF2 presents a therapeutic target to perturb MYC signaling in glioblastoma. The IGF1/IGF1R inhibitor linsitinib preferentially targeted YTHDF2-expressing cells, inhibiting GSC viability without affecting NSCs and impairing in vivo glioblastoma growth. YTHDF2 links RNA epitranscriptomic modifications and GSC growth, laying the foundation for the YTHDF2-MYC-IGFBP3 axis as a specific and novel therapeutic target in glioblastoma.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) READER
Target Regulation Up regulation
Pathway Response RNA degradation hsa03018
Cell Process RNA stability
In-vitro Model ()
HNP1 (A human neural progenitor cell)
NHA (Normal human astrocytes)
NSC11 (Pluripotent derived neural progenitor cell)
In-vivo Model For in vivo drug treatment studies, intracranial xenografts were generated by implanting 5000 patient-derived GSCs (387 and 4121) into the right cerebral cortex of NSG mice as described above.
Nuclear factor NF-kappa-B p105 subunit (NF-Kappa-B/NFKB1)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [14]
Response Summary YTHDF2 accelerated UBXN1 mRNA degradation via METTL3-mediated m6A, which, in turn, promoted Nuclear factor NF-kappa-B p105 subunit (NF-Kappa-B/NFKB1) activation. YTHDF2 promotes the malignant progression of gliomas and revealed important insight into the upstream regulatory mechanism of NF-Kappa-B activation via UBXN1 with a primary focus on m6A modification.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator YTH domain-containing family protein 2 (YTHDF2) READER
Target Regulation Up regulation
Pathway Response NF-kappa B signaling pathway hsa04064
In-vitro Model U87 (A primary glioblastoma cell line)
N33 (The GBM patient-derived cell line)
LN-229 Glioblastoma Homo sapiens CVCL_0393
H4 Astrocytoma Homo sapiens CVCL_1239
In-vivo Model Five-week-old female BALB/c nude mice (Charles Rivers, Beijing, China) were selected for the experiments. U87 cells (5 × 105) transfected with an empty vector, YTHDF2 overexpression, or METTL3 overexpression vectors were suspended in PBS and injected into the right frontal node of nude mice. The inoculation position was 2 mm lateral and 2 mm posterior to the anterior fontanel. Tumor size was estimated from luciferase volume measurements and MRI. The mice were sacrificed when they exhibited disturbed activity or convulsion. The brain was then harvested and embedded in paraffin.
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [14]
Response Summary YTHDF2 accelerated UBXN1 mRNA degradation via METTL3-mediated m6A, which, in turn, promoted Nuclear factor NF-kappa-B p105 subunit (NF-Kappa-B/NFKB1) activation. YTHDF2 promotes the malignant progression of gliomas and revealed important insight into the upstream regulatory mechanism of NF-Kappa-B activation via UBXN1 with a primary focus on m6A modification.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
Pathway Response NF-kappa B signaling pathway hsa04064
In-vitro Model U87 (A primary glioblastoma cell line)
N33 (The GBM patient-derived cell line)
LN-229 Glioblastoma Homo sapiens CVCL_0393
H4 Astrocytoma Homo sapiens CVCL_1239
In-vivo Model Five-week-old female BALB/c nude mice (Charles Rivers, Beijing, China) were selected for the experiments. U87 cells (5 × 105) transfected with an empty vector, YTHDF2 overexpression, or METTL3 overexpression vectors were suspended in PBS and injected into the right frontal node of nude mice. The inoculation position was 2 mm lateral and 2 mm posterior to the anterior fontanel. Tumor size was estimated from luciferase volume measurements and MRI. The mice were sacrificed when they exhibited disturbed activity or convulsion. The brain was then harvested and embedded in paraffin.
Oxysterols receptor LXR-alpha (LXRA)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [15]
Response Summary YTHDF2 facilitates m6A-dependent mRNA decay of Oxysterols receptor LXR-alpha (LXRA) and HIVEP2, which impacts the glioma patient survival. YTHDF2 promotes tumorigenesis of GBM cells, largely through the downregulation of LXRA and HIVEP2.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator YTH domain-containing family protein 2 (YTHDF2) READER
Target Regulation Down regulation
Pathway Response mRNA surveillance pathway hsa03015), RNA degradation
Cell Process RNA stability
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
U-251MG Astrocytoma Homo sapiens CVCL_0021
T98G Glioblastoma Homo sapiens CVCL_0556
SW1783 Anaplastic astrocytoma Homo sapiens CVCL_1722
LN-229 Glioblastoma Homo sapiens CVCL_0393
Hs 683 Oligodendroglioma Homo sapiens CVCL_0844
GSC7-2 (GSC7-2 were obtained from fresh surgical specimens of human primary and recurrent glioma)
GSC6-27 (GSC6-27 were obtained from fresh surgical specimens of human primary and recurrent glioma)
GSC23 Glioblastoma Homo sapiens CVCL_DR59
GSC20 (GSC20 were obtained from fresh surgical specimens of human primary and recurrent glioma)
GSC17 Glioblastoma Homo sapiens CVCL_DR57
GSC11 Glioblastoma Homo sapiens CVCL_DR55
In-vivo Model For the studies of investigating mice survival, mice were intracranially injected with 10,000 GSC11, 10,000 GSC7-2, or 500,000 LN229 cells.
PDH kinase 1 (PDK1)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [16]
Response Summary Long noncoding RNA just proximal to X-inactive specific transcript facilitates aerobic glycolysis and temozolomide chemoresistance by promoting stability of PDH kinase 1 (PDK1) mRNA in an m6A-dependent manner in glioblastoma multiforme cells. JPX interacted with N6-methyladenosine (m6A) demethylase FTO alpha-ketoglutarate dependent dioxygenase (FTO) and enhanced FTO-mediated PDK1 mRNA demethylation.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Responsed Drug Temozolomide Approved
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Up regulation
Pathway Response Citrate cycle hsa00020
Central carbon metabolism in cancer hsa05230
Cell Process Aerobic glycolysis
Cell apoptosis
In-vitro Model LN-18 Glioblastoma Homo sapiens CVCL_0392
LN-229 Glioblastoma Homo sapiens CVCL_0393
SHG-44 Astrocytoma Homo sapiens CVCL_6728
U251 (Fibroblasts or fibroblast like cells)
U87 (A primary glioblastoma cell line)
Protein argonaute-1 (AGO1)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [11]
Response Summary loss of m6A RNA methylation and increased translation in human glioblastoma cells as well as a role for miRNAs in the modulation of m6A RNA demethylation in genes that are most efficiently translated during glioma stem cells differentiation. Ectopic expression of the RRACH-binding miR-145 induces loss of m6A, formation of FTO/Protein argonaute-1 (AGO1)/ILF3/miR-145 complexes on a clinically relevant tumor suppressor gene (CLIP3) and significant increase in its nascent translation.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Up regulation
In-vitro Model GBM1 Glioblastoma Homo sapiens CVCL_DG57
GBM2 Glioblastoma Homo sapiens CVCL_DG58
GBM3 Glioblastoma Homo sapiens CVCL_DG59
Putative heat shock 70 kDa protein 7 (HSPA7)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [17]
Response Summary Putative heat shock 70 kDa protein 7 (HSPA7) promoted macrophage infiltration and SPP1 expression via upregulating the YAP1 and LOX expression of glioblastoma stem cells in vitro and in clinical glioblastoma tumor samples. HSPA7 is expected to predict the ability of PD-1 inhibitors to respond to treatment in glioblastoma.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Responsed Drug Pembrolizumab Approved
Pathway Response PD-L1 expression and PD-1 checkpoint pathway in cancer hsa05235
In-vitro Model A-172 Glioblastoma Homo sapiens CVCL_0131
GSC 20 (Mesenchymal (MES) subtype GSC cell lines)
GSC 20 (Mesenchymal (MES) subtype GSC cell lines)
GSC 8-11 (Proneural (PN) subtype GSC cell lines)
LN-229 Glioblastoma Homo sapiens CVCL_0393
Neural progenitor cells (NPCs) (The progenitor cells of the CNS)
U-251MG Astrocytoma Homo sapiens CVCL_0021
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
Receptor-type tyrosine-protein phosphatase C (CD45)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [3]
Response Summary HNRNPA2B1 and HNRNPC were extensively expressed in the Glioblastoma multiforme(GBM) microenvironment.m6A regulators promoted the stemness state in GBM cancer cells. Cell communication analysis identified genes in the GALECTIN signaling network in GBM samples, and expression of these genes (LGALS9, CD44, Receptor-type tyrosine-protein phosphatase C (CD45), and HAVCR2) correlated with that of m6A regulators.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Heterogeneous nuclear ribonucleoproteins A2/B1 (HNRNPA2B1) READER
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [3]
Response Summary HNRNPA2B1 and HNRNPC were extensively expressed in the Glioblastoma multiforme(GBM) microenvironment. m6A regulators promoted the stemness state in GBM cancer cells. Cell communication analysis identified genes in the GALECTIN signaling network in GBM samples, and expression of these genes (LGALS9, CD44, Receptor-type tyrosine-protein phosphatase C (CD45), and HAVCR2) correlated with that of m6A regulators.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Heterogeneous nuclear ribonucleoproteins C1/C2 (HNRNPC) READER
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
THP-1 Childhood acute monocytic leukemia Homo sapiens CVCL_0006
Serine/arginine-rich splicing factor 11 (SRSF11)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [18]
Response Summary Silencing METTL3 or overexpressing dominant-negative mutant METTL3 suppressed the growth and self-renewal of Glioblastoma cells. Integrated transcriptome and MeRIP-seq analyses revealed that downregulating the expression of METTL3 decreased m6A modification levels of Serine/arginine-rich splicing factor 11 (SRSF11), which led to YTHDC1-dependent NMD of SRSF transcripts and decreased SRSF protein expression.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
Pathway Response RNA degradation hsa03018
Cell Process mRNA decay
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model For subcutaneous tumor model, each mouse was injected subcutaneously in the right flank with 2 × 106 U87MG cells (METTL3-KD or control) in 100 uL PBS.
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [18]
Response Summary Silencing METTL3 or overexpressing dominant-negative mutant METTL3 suppressed the growth and self-renewal of Glioblastoma cells. Integrated transcriptome and MeRIP-seq analyses revealed that downregulating the expression of METTL3 decreased m6A modification levels of Serine/arginine-rich splicing factor 11 (SRSF11), which led to YTHDC1-dependent NMD of SRSF transcripts and decreased SRSF protein expression.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator YTH domain-containing protein 1 (YTHDC1) READER
Target Regulation Up regulation
Pathway Response RNA degradation hsa03018
Cell Process mRNA decay
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model For subcutaneous tumor model, each mouse was injected subcutaneously in the right flank with 2 × 106 U87MG cells (METTL3-KD or control) in 100 uL PBS.
Serine/arginine-rich splicing factor 3 (SRSF3)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [18]
Response Summary Silencing METTL3 or overexpressing dominant-negative mutant METTL3 suppressed the growth and self-renewal of Glioblastoma cells. Integrated transcriptome and MeRIP-seq analyses revealed that downregulating the expression of METTL3 decreased m6A modification levels of Serine/arginine-rich splicing factor 3 (SRSF3), which led to YTHDC1-dependent NMD of SRSF transcripts and decreased SRSF protein expression.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
Pathway Response RNA degradation hsa03018
Cell Process mRNA decay
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model For subcutaneous tumor model, each mouse was injected subcutaneously in the right flank with 2 × 106 U87MG cells (METTL3-KD or control) in 100 uL PBS.
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [18]
Response Summary Silencing METTL3 or overexpressing dominant-negative mutant METTL3 suppressed the growth and self-renewal of Glioblastoma cells. Integrated transcriptome and MeRIP-seq analyses revealed that downregulating the expression of METTL3 decreased m6A modification levels of Serine/arginine-rich splicing factor 3 (SRSF3), which led to YTHDC1-dependent NMD of SRSF transcripts and decreased SRSF protein expression.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator YTH domain-containing protein 1 (YTHDC1) READER
Target Regulation Up regulation
Pathway Response RNA degradation hsa03018
Cell Process mRNA decay
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model For subcutaneous tumor model, each mouse was injected subcutaneously in the right flank with 2 × 106 U87MG cells (METTL3-KD or control) in 100 uL PBS.
Serine/arginine-rich splicing factor 6 (SRSF6)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [18]
Response Summary Silencing METTL3 or overexpressing dominant-negative mutant METTL3 suppressed the growth and self-renewal of Glioblastoma cells. Integrated transcriptome and MeRIP-seq analyses revealed that downregulating the expression of METTL3 decreased m6A modification levels of Serine/arginine-rich splicing factor 6 (SRSF6), which led to YTHDC1-dependent NMD of SRSF transcripts and decreased SRSF protein expression.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
Pathway Response RNA degradation hsa03018
Cell Process mRNA decay
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model For subcutaneous tumor model, each mouse was injected subcutaneously in the right flank with 2 × 106 U87MG cells (METTL3-KD or control) in 100 uL PBS.
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [18]
Response Summary Silencing METTL3 or overexpressing dominant-negative mutant METTL3 suppressed the growth and self-renewal of Glioblastoma cells. Integrated transcriptome and MeRIP-seq analyses revealed that downregulating the expression of METTL3 decreased m6A modification levels of Serine/arginine-rich splicing factor 6 (SRSF6), which led to YTHDC1-dependent NMD of SRSF transcripts and decreased SRSF protein expression.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator YTH domain-containing protein 1 (YTHDC1) READER
Target Regulation Up regulation
Pathway Response RNA degradation hsa03018
Cell Process mRNA decay
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model For subcutaneous tumor model, each mouse was injected subcutaneously in the right flank with 2 × 106 U87MG cells (METTL3-KD or control) in 100 uL PBS.
Serine/arginine-rich splicing factor 7 (SRSF7)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [19]
Response Summary The gene expression of Serine/arginine-rich splicing factor 7 (SRSF7) is positively correlated with glioblastoma (GBM) cell-specific m6A methylation. The two m6A sites on PDZ-binding kinase (PBK) are regulated by SRSF7 and partially mediate the effects of SRSF7 in GBM cells through recognition by IGF2BP2.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) READER
Pathway Response Spliceosome hsa03040
In-vitro Model HEK293T Normal Homo sapiens CVCL_0063
U87MG (Astroblastoma cells from human brain)
LN-229 Glioblastoma Homo sapiens CVCL_0393
A-172 Glioblastoma Homo sapiens CVCL_0131
LN-18 Glioblastoma Homo sapiens CVCL_0392
LN-428 Glioblastoma Homo sapiens CVCL_3959
LN-443 Glioblastoma Homo sapiens CVCL_3960
SNB-19 Astrocytoma Homo sapiens CVCL_0535
T98G Glioblastoma Homo sapiens CVCL_0556
U-118MG Astrocytoma Homo sapiens CVCL_0633
U251 (Fibroblasts or fibroblast like cells)
U-138MG Astrocytoma Homo sapiens CVCL_0020
Suppressor of cytokine signaling 2 (SOCS2)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [20]
Response Summary miR-302a was identified to target METTL3, which could inhibit Suppressor of cytokine signaling 2 (SOCS2) expression via m6A modification in glioma.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Down regulation
Pathway Response Transcriptional misregulation in cancer hsa05202
Cell Process RNA stability
In-vitro Model LN-229 Glioblastoma Homo sapiens CVCL_0393
LN-18 Glioblastoma Homo sapiens CVCL_0392
HEB (human normal glial cell line HEB were obtained from Tongpai (Shanghai) biotechnology co., LTD (Shanghai, China))
In-vivo Model Mice were subcutaneously injected with 2.5 × 106 U251 cells stably infected with OE-NC, OE-JMJD1C, OE-NC and sh-NC, OE-JMJD1C and sh-NC or OE-JMJD1C and sh-SOCS2 (n = 10 in each group) in 0.1 ml PBS.
TNF alpha-induced protein 3 (TNFAIP3)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [6]
Response Summary YTHDF2 enhanced TMZ resistance in GBM by activation of the PI3K/Akt and NF-Kappa-B signalling pathways via inhibition of EPHB3 and TNF alpha-induced protein 3 (TNFAIP3).
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Responsed Drug Temozolomide Approved
Target Regulator YTH domain-containing family protein 2 (YTHDF2) READER
Target Regulation Down regulation
Pathway Response PI3K-Akt signaling pathway hsa04151
Cell Process RNA stability
In-vitro Model T98G Glioblastoma Homo sapiens CVCL_0556
LN-229 Glioblastoma Homo sapiens CVCL_0393
In-vivo Model 5 × 106 infected T98G cells (LV-NC or LV-YTHDF2) were injected into the flanks of mice through subcutaneous.
Transcription factor HIVEP2 (HIVEP2)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [15]
Response Summary YTHDF2 facilitates m6A-dependent mRNA decay of LXRA and Transcription factor HIVEP2 (HIVEP2), which impacts the glioma patient survival. YTHDF2 promotes tumorigenesis of GBM cells, largely through the downregulation of LXRA and HIVEP2.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator YTH domain-containing family protein 2 (YTHDF2) READER
Target Regulation Down regulation
Pathway Response mRNA surveillance pathway hsa03015), RNA degradation
Cell Process RNA stability
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
U-251MG Astrocytoma Homo sapiens CVCL_0021
T98G Glioblastoma Homo sapiens CVCL_0556
SW1783 Anaplastic astrocytoma Homo sapiens CVCL_1722
LN-229 Glioblastoma Homo sapiens CVCL_0393
Hs 683 Oligodendroglioma Homo sapiens CVCL_0844
GSC7-2 (GSC7-2 were obtained from fresh surgical specimens of human primary and recurrent glioma)
GSC6-27 (GSC6-27 were obtained from fresh surgical specimens of human primary and recurrent glioma)
GSC23 Glioblastoma Homo sapiens CVCL_DR59
GSC20 (GSC20 were obtained from fresh surgical specimens of human primary and recurrent glioma)
GSC17 Glioblastoma Homo sapiens CVCL_DR57
GSC11 Glioblastoma Homo sapiens CVCL_DR55
In-vivo Model For the studies of investigating mice survival, mice were intracranially injected with 10,000 GSC11, 10,000 GSC7-2, or 500,000 LN229 cells.
Transcription factor PU.1 (SPI1)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [21]
Response Summary FTO inhibited growth, migration and invasion of GBM cells in vitro and in vivo.decreased FTO expression could induce the downregulation of MTMR3 expression by modulating the processing of pri-miR-10a in an m6A/HNRNPA2B1-dependent manner in GBM cells. Furthermore, the transcriptional activity of FTO was inhibited by the transcription factor Transcription factor PU.1 (SPI1).
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Down regulation
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
U251 (Fibroblasts or fibroblast like cells)
U-118MG Astrocytoma Homo sapiens CVCL_0633
LN-229 Glioblastoma Homo sapiens CVCL_0393
A-172 Glioblastoma Homo sapiens CVCL_0131
In-vivo Model BALB/c male nude mice were 4 weeks old. GBM cells with stable overexpression or knockdown of FTO and ovNC or shNC were transduced with lentivirus expressing luciferase. The cells were intracranially injected at a density of 5 × 105/10 uL into every mouse to form an orthotopic xenograft model. Coordinates of injection were 1 mm anterior and 2.5 mm right to the bregma, at a depth of 3.5 mm (the right frontal lobes of the mouse). Every 6 days, bioluminescence imaging (IVIS Lumina Series III; PerkinElmer, Waltham, MA) was used to image the mouse. At 8 days, we randomly chose 5 mice from each group to euthanize them, and their brain tissues were fixed with paraformaldehyde for further study. Another 5 mice were used for survival time analysis. For DB2313 (563801; MedKoo) anti-tumor research, male nude mice were subcutaneously injected with 5 × 106 U87MG cells suspended in 0.1 mL PBS. After 7 days, mice were intraperitoneally injected with DB2313 at density of 10 mg/kg/day dissolved in PBS solvent containing 10% DMSO for 7 days. The other group treated with vehicle only was set as the control group.
Transcription factor SOX-2 (SOX2)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [22]
Response Summary GBM tumors have elevated levels of METTL3 transcripts and silencing METTL3 in U87/TIC inhibited tumor growth in an intracranial orthotopic mouse model with prolonged mice survival. The exogenous overexpression of 3'UTR-less Transcription factor SOX-2 (SOX2) significantly alleviated the inhibition of neurosphere formation observed in METTL3 silenced GSCs.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
Pathway Response Signaling pathways regulating pluripotency of stem cells hsa04550
Cell Process DNA repair
Nucleotide excision repair (hsa03420)
In-vitro Model Mouse immortalized astrocytes (A type of glial cell)
UBX domain-containing protein 1 (UBXN1)
In total 2 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [14]
Response Summary YTHDF2 accelerated UBX domain-containing protein 1 (UBXN1) mRNA degradation via METTL3-mediated m6A, which, in turn, promoted NF-Kappa-B activation. YTHDF2 promotes the malignant progression of gliomas and revealed important insight into the upstream regulatory mechanism of NF-Kappa-B activation via UBXN1 with a primary focus on m6A modification.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator YTH domain-containing family protein 2 (YTHDF2) READER
Target Regulation Down regulation
Pathway Response NF-kappa B signaling pathway hsa04064
In-vitro Model U87 (A primary glioblastoma cell line)
N33 (The GBM patient-derived cell line)
LN-229 Glioblastoma Homo sapiens CVCL_0393
H4 Astrocytoma Homo sapiens CVCL_1239
In-vivo Model Five-week-old female BALB/c nude mice (Charles Rivers, Beijing, China) were selected for the experiments. U87 cells (5 × 105) transfected with an empty vector, YTHDF2 overexpression, or METTL3 overexpression vectors were suspended in PBS and injected into the right frontal node of nude mice. The inoculation position was 2 mm lateral and 2 mm posterior to the anterior fontanel. Tumor size was estimated from luciferase volume measurements and MRI. The mice were sacrificed when they exhibited disturbed activity or convulsion. The brain was then harvested and embedded in paraffin.
Experiment 2 Reporting the m6A-centered Disease Response by This Target Gene [14]
Response Summary YTHDF2 accelerated UBX domain-containing protein 1 (UBXN1) mRNA degradation via METTL3-mediated m6A, which, in turn, promoted NF-Kappa-B activation. YTHDF2 promotes the malignant progression of gliomas and revealed important insight into the upstream regulatory mechanism of NF-Kappa-B activation via UBXN1 with a primary focus on m6A modification.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Down regulation
Pathway Response NF-kappa B signaling pathway hsa04064
In-vitro Model U87 (A primary glioblastoma cell line)
N33 (The GBM patient-derived cell line)
LN-229 Glioblastoma Homo sapiens CVCL_0393
H4 Astrocytoma Homo sapiens CVCL_1239
In-vivo Model Five-week-old female BALB/c nude mice (Charles Rivers, Beijing, China) were selected for the experiments. U87 cells (5 × 105) transfected with an empty vector, YTHDF2 overexpression, or METTL3 overexpression vectors were suspended in PBS and injected into the right frontal node of nude mice. The inoculation position was 2 mm lateral and 2 mm posterior to the anterior fontanel. Tumor size was estimated from luciferase volume measurements and MRI. The mice were sacrificed when they exhibited disturbed activity or convulsion. The brain was then harvested and embedded in paraffin.
Vacuolar protein-sorting-associated protein 25 (VPS25)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [23]
Response Summary YTHDC1 inhibited glioma proliferation by reducing the expression of Vacuolar protein-sorting-associated protein 25 (VPS25).
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator YTH domain-containing protein 1 (YTHDC1) READER
Target Regulation Down regulation
Pathway Response JAK-STAT signaling pathway hsa04630
Cell Process Arrest cell cycle at G0/G1 phase
Cell apoptosis
In-vitro Model U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
In-vivo Model The U87MG cells (1 × 107 cells in 0.1 ml PBS) were injected subcutaneously into BALB/c nude mice. Tumor width and length were recorded every 5 days.
Cancer susceptibility 9 (CASC9)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [24]
Response Summary Cancer susceptibility 9 (CASC9)/IGF2BP2/HK2 axis promotes the aerobic glycolysis of glioblastoma multiforme.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) READER
Target Regulation Up regulation
Pathway Response Glycolysis / Gluconeogenesis hsa00010
Cell Process Aerobic glycolysis
In-vitro Model NHA (Normal human astrocytes)
U251 (Fibroblasts or fibroblast like cells)
U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
Metastasis associated lung adenocarcinoma transcript 1 (MALAT1)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [25]
Response Summary METTL3 promoted the malignant progression of IDH-wildtype gliomas and revealed important insight into the upstream regulatory mechanism of Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) and NF-Kappa-B with a primary focus on m6A modification.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
Pathway Response TNF signaling pathway hsa04668
Cell Process Cell proliferation and metastasis
In-vitro Model H4 Astrocytoma Homo sapiens CVCL_1239
LN-229 Glioblastoma Homo sapiens CVCL_0393
U87 (A primary glioblastoma cell line)
In-vivo Model U87 cells (5 × 105) transfected with an empty vector, METTL3 shRNA, or METTL3 overexpression vector were inoculated into the right frontal node of nude mice.
Nuclear paraspeckle assembly transcript 1 (NEAT1)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [26]
Response Summary Hypoxia-induced ALKBH5 erased m6A deposition from the lncRNA NEAT1, stabilizing the transcript and facilitating Nuclear paraspeckle assembly transcript 1 (NEAT1)-mediated paraspeckle assembly. Ectopic expression of CXCL8 in ALKBH5-deficient glioblastoma multiforme cells partially restored TAM recruitment and tumor progression.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator RNA demethylase ALKBH5 (ALKBH5) ERASER
Target Regulation Down regulation
Pathway Response Transcriptional misregulation in cancer hsa05202
Cell Process Immune
In-vitro Model GL261 Mouse glioblastoma Mus musculus CVCL_Y003
U87 (A primary glioblastoma cell line)
In-vivo Model Male C57BL/6 mice (4-6 weeks old) were used in all tumor allografting experiments and transplanted with GL261-luc cells (1×105) into the frontal lobes of brains.
microRNA 145 (MIR145)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [11]
Response Summary loss of m6A RNA methylation and increased translation in human glioblastoma cells as well as a role for miRNAs in the modulation of m6A RNA demethylation in genes that are most efficiently translated during glioma stem cells differentiation. Ectopic expression of the RRACH-binding miR-145 induces loss of m6A, formation of FTO/AGO1/ILF3/microRNA 145 (MIR145) complexes on a clinically relevant tumor suppressor gene (CLIP3) and significant increase in its nascent translation.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Up regulation
In-vitro Model GBM1 Glioblastoma Homo sapiens CVCL_DG57
GBM2 Glioblastoma Homo sapiens CVCL_DG58
GBM3 Glioblastoma Homo sapiens CVCL_DG59
microRNA 155 (MIR155)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [27]
Response Summary FTO Inhibition Enhances the Antitumor Effect of Temozolomide by Targeting MYC-microRNA 155 (MIR155)/miR-23a Cluster-MXI1 Feedback Circuit in Glioma.
Responsed Disease Glioma [ICD-11: 2A00.0]
Responsed Drug Temozolomide Approved
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
In-vitro Model A-172 Glioblastoma Homo sapiens CVCL_0131
HEK293T Normal Homo sapiens CVCL_0063
U251 (Fibroblasts or fibroblast like cells)
U87 (A primary glioblastoma cell line)
In-vivo Model Previously prepared U87 cells (5 × 106 cells, 60 uL) stably infected with miRNA were injected subcutaneously in the right flank of the mice, whereas control U87 cells infected with empty vector were injected in the left flank.
microRNA 23a (MIR23A)
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [27]
Response Summary FTO Inhibition Enhances the Antitumor Effect of Temozolomide by Targeting MYC-miR-155/microRNA 23a (MIR23A) Cluster-MXI1 Feedback Circuit in Glioma.
Responsed Disease Glioma [ICD-11: 2A00.0]
Responsed Drug Temozolomide Approved
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
In-vitro Model A-172 Glioblastoma Homo sapiens CVCL_0131
HEK293T Normal Homo sapiens CVCL_0063
U251 (Fibroblasts or fibroblast like cells)
U87 (A primary glioblastoma cell line)
In-vivo Model Previously prepared U87 cells (5 × 106 cells, 60 uL) stably infected with miRNA were injected subcutaneously in the right flank of the mice, whereas control U87 cells infected with empty vector were injected in the left flank.
hsa-miR-671-5p
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [1]
Response Summary METTL3-mediated m6A modification upregulated circDLC1 expression, and circDLC1 promoted CTNNBIP1 transcription by sponging hsa-miR-671-5p, thus repressing the malignant proliferation of glioma.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
In-vitro Model T98G Glioblastoma Homo sapiens CVCL_0556
LN-229 Glioblastoma Homo sapiens CVCL_0393
LN-18 Glioblastoma Homo sapiens CVCL_0392
HEB (human normal glial cell line HEB were obtained from Tongpai (Shanghai) biotechnology co., LTD (Shanghai, China))
A-172 Glioblastoma Homo sapiens CVCL_0131
Circ_DLC1
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [1]
Response Summary METTL3-mediated m6A modification upregulated Circ_DLC1 expression, and circDLC1 promoted CTNNBIP1 transcription by sponging miR-671-5p, thus repressing the malignant proliferation of glioma.
Responsed Disease Glioma [ICD-11: 2A00.0]
Target Regulator Methyltransferase-like 3 (METTL3) WRITER
Target Regulation Up regulation
In-vitro Model T98G Glioblastoma Homo sapiens CVCL_0556
LN-229 Glioblastoma Homo sapiens CVCL_0393
LN-18 Glioblastoma Homo sapiens CVCL_0392
HEB (human normal glial cell line HEB were obtained from Tongpai (Shanghai) biotechnology co., LTD (Shanghai, China))
A-172 Glioblastoma Homo sapiens CVCL_0131
pri-miR-10a
In total 1 item(s) under this target gene
Experiment 1 Reporting the m6A-centered Disease Response by This Target Gene [21]
Response Summary FTO inhibited growth, migration and invasion of GBM cells in vitro and in vivo.decreased FTO expression could induce the downregulation of MTMR3 expression by modulating the processing of pri-miR-10a in an m6A/HNRNPA2B1-dependent manner in GBM cells. Furthermore, the transcriptional activity of FTO was inhibited by the transcription factor SPI1.
Responsed Disease Glioblastoma [ICD-11: 2A00.00]
Target Regulator Fat mass and obesity-associated protein (FTO) ERASER
Target Regulation Up regulation
In-vitro Model U-87MG ATCC Glioblastoma Homo sapiens CVCL_0022
U251 (Fibroblasts or fibroblast like cells)
U-118MG Astrocytoma Homo sapiens CVCL_0633
LN-229 Glioblastoma Homo sapiens CVCL_0393
A-172 Glioblastoma Homo sapiens CVCL_0131
In-vivo Model BALB/c male nude mice were 4 weeks old. GBM cells with stable overexpression or knockdown of FTO and ovNC or shNC were transduced with lentivirus expressing luciferase. The cells were intracranially injected at a density of 5 × 105/10 uL into every mouse to form an orthotopic xenograft model. Coordinates of injection were 1 mm anterior and 2.5 mm right to the bregma, at a depth of 3.5 mm (the right frontal lobes of the mouse). Every 6 days, bioluminescence imaging (IVIS Lumina Series III; PerkinElmer, Waltham, MA) was used to image the mouse. At 8 days, we randomly chose 5 mice from each group to euthanize them, and their brain tissues were fixed with paraformaldehyde for further study. Another 5 mice were used for survival time analysis. For DB2313 (563801; MedKoo) anti-tumor research, male nude mice were subcutaneously injected with 5 × 106 U87MG cells suspended in 0.1 mL PBS. After 7 days, mice were intraperitoneally injected with DB2313 at density of 10 mg/kg/day dissolved in PBS solvent containing 10% DMSO for 7 days. The other group treated with vehicle only was set as the control group.
References
Ref 1 Molecular mechanism of m(6)A methylation of circDLC1 mediated by RNA methyltransferase METTL3 in the malignant proliferation of glioma cells. Cell Death Discov. 2022 Apr 26;8(1):229. doi: 10.1038/s41420-022-00979-6.
Ref 2 R-2HG Exhibits Anti-tumor Activity by Targeting FTO/m(6)A/MYC/CEBPA Signaling. Cell. 2018 Jan 11;172(1-2):90-105.e23. doi: 10.1016/j.cell.2017.11.031. Epub 2017 Dec 14.
Ref 3 Roles of the m(6)A Modification of RNA in the Glioblastoma Microenvironment as Revealed by Single-Cell Analyses. Front Immunol. 2022 Apr 26;13:798583. doi: 10.3389/fimmu.2022.798583. eCollection 2022.
Ref 4 RNA binding protein NKAP protects glioblastoma cells from ferroptosis by promoting SLC7A11 mRNA splicing in an m(6)A-dependent manner. Cell Death Dis. 2022 Jan 21;13(1):73. doi: 10.1038/s41419-022-04524-2.
Ref 5 METTL3 Promotes the Resistance of Glioma to Temozolomide via Increasing MGMT and ANPG in a m(6)A Dependent Manner. Front Oncol. 2021 Jul 15;11:702983. doi: 10.3389/fonc.2021.702983. eCollection 2021.
Ref 6 YTHDF2 promotes temozolomide resistance in glioblastoma by activation of the Akt and NF-KappaB signalling pathways via inhibiting EPHB3 and TNFAIP3. Clin Transl Immunology. 2022 May 9;11(5):e1393. doi: 10.1002/cti2.1393. eCollection 2022.
Ref 7 m(6)A Demethylase ALKBH5 Maintains Tumorigenicity of Glioblastoma Stem-like Cells by Sustaining FOXM1 Expression and Cell Proliferation Program. Cancer Cell. 2017 Apr 10;31(4):591-606.e6. doi: 10.1016/j.ccell.2017.02.013. Epub 2017 Mar 23.
Ref 8 m6A RNA Methylation Controls Proliferation of Human Glioma Cells by Influencing Cell Apoptosis. Cytogenet Genome Res. 2019;159(3):119-125. doi: 10.1159/000499062. Epub 2019 Oct 23.
Ref 9 Warburg effect-promoted exosomal circ_0072083 releasing up-regulates NANGO expression through multiple pathways and enhances temozolomide resistance in glioma. J Exp Clin Cancer Res. 2021 May 11;40(1):164. doi: 10.1186/s13046-021-01942-6.
Ref 10 ADAR1 is a new target of METTL3 and plays a pro-oncogenic role in glioblastoma by an editing-independent mechanism. Genome Biol. 2021 Jan 28;22(1):51. doi: 10.1186/s13059-021-02271-9.
Ref 11 miRNA-mediated loss of m6A increases nascent translation in glioblastoma. PLoS Genet. 2021 Mar 8;17(3):e1009086. doi: 10.1371/journal.pgen.1009086. eCollection 2021 Mar.
Ref 12 m(6)A RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells. Cell Rep. 2017 Mar 14;18(11):2622-2634. doi: 10.1016/j.celrep.2017.02.059.
Ref 13 The RNA m6A Reader YTHDF2 Maintains Oncogene Expression and Is a Targetable Dependency in Glioblastoma Stem Cells. Cancer Discov. 2021 Feb;11(2):480-499. doi: 10.1158/2159-8290.CD-20-0331. Epub 2020 Oct 6.
Ref 14 YTHDF2 facilitates UBXN1 mRNA decay by recognizing METTL3-mediated m(6)A modification to activate NF-KappaB and promote the malignant progression of glioma. J Hematol Oncol. 2021 Jul 10;14(1):109. doi: 10.1186/s13045-021-01124-z.
Ref 15 EGFR/SRC/ERK-stabilized YTHDF2 promotes cholesterol dysregulation and invasive growth of glioblastoma. Nat Commun. 2021 Jan 8;12(1):177. doi: 10.1038/s41467-020-20379-7.
Ref 16 Long noncoding RNA just proximal to X-inactive specific transcript facilitates aerobic glycolysis and temozolomide chemoresistance by promoting stability of PDK1 mRNA in an m6A-dependent manner in glioblastoma multiforme cells. Cancer Sci. 2021 Nov;112(11):4543-4552. doi: 10.1111/cas.15072. Epub 2021 Aug 30.
Ref 17 The N(6)-Methyladenosine-Modified Pseudogene HSPA7 Correlates With the Tumor Microenvironment and Predicts the Response to Immune Checkpoint Therapy in Glioblastoma. Front Immunol. 2021 Jul 20;12:653711. doi: 10.3389/fimmu.2021.653711. eCollection 2021.
Ref 18 N(6)-Methyladenosine Modulates Nonsense-Mediated mRNA Decay in Human Glioblastoma. Cancer Res. 2019 Nov 15;79(22):5785-5798. doi: 10.1158/0008-5472.CAN-18-2868. Epub 2019 Sep 17.
Ref 19 Specific Regulation of m(6)A by SRSF7 Promotes the Progression of Glioblastoma. Genomics Proteomics Bioinformatics. 2021 Dec 23:S1672-0229(21)00252-7. doi: 10.1016/j.gpb.2021.11.001. Online ahead of print.
Ref 20 Histone demethylase JMJD1C promotes the polarization of M1 macrophages to prevent glioma by upregulating miR-302a. Clin Transl Med. 2021 Sep;11(9):e424. doi: 10.1002/ctm2.424.
Ref 21 SPI1-induced?downregulation of FTO promotes GBM progression by regulating pri-miR-10a processing in an m6A-dependent manner. Mol Ther Nucleic Acids. 2022 Jan 1;27:699-717. doi: 10.1016/j.omtn.2021.12.035. eCollection 2022 Mar 8.
Ref 22 Essential role of METTL3-mediated m(6)A modification in glioma stem-like cells maintenance and radioresistance. Oncogene. 2018 Jan 25;37(4):522-533. doi: 10.1038/onc.2017.351. Epub 2017 Oct 9.
Ref 23 YTHDC1-mediated VPS25 regulates cell cycle by targeting JAK-STAT signaling in human glioma cells. Cancer Cell Int. 2021 Dec 4;21(1):645. doi: 10.1186/s12935-021-02304-0.
Ref 24 m(6)A reader IGF2BP2-stabilized CASC9 accelerates glioblastoma aerobic glycolysis by enhancing HK2 mRNA stability. Cell Death Discov. 2021 Oct 13;7(1):292. doi: 10.1038/s41420-021-00674-y.
Ref 25 METTL3 enhances the stability of MALAT1 with the assistance of HuR via m6A modification and activates NF-KappaB to promote the malignant progression of IDH-wildtype glioma. Cancer Lett. 2021 Jul 28;511:36-46. doi: 10.1016/j.canlet.2021.04.020. Epub 2021 Apr 29.
Ref 26 ALKBH5 Facilitates Hypoxia-Induced Paraspeckle Assembly and IL8 Secretion to Generate an Immunosuppressive Tumor Microenvironment. Cancer Res. 2021 Dec 1;81(23):5876-5888. doi: 10.1158/0008-5472.CAN-21-1456. Epub 2021 Oct 20.
Ref 27 FTO Inhibition Enhances the Antitumor Effect of Temozolomide by Targeting MYC-miR-155/23a Cluster-MXI1 Feedback Circuit in Glioma. Cancer Res. 2020 Sep 15;80(18):3945-3958. doi: 10.1158/0008-5472.CAN-20-0132. Epub 2020 Jul 17.