Targeting c-Myc transactivation by LMNA inhibits tRNA processing essential for malate-aspartate shuttle and tumour progression

Jianqun Wang , Mei Hong , Yang Cheng , Xiaojing Wang , Dan Li , Guo Chen , Banghe Bao , Jiyu Song , Xinyi Du , Chunhui Yang , Liduan Zheng , Qiangsong Tong

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (5) : e1680

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (5) : e1680 DOI: 10.1002/ctm2.1680
RESEARCH ARTICLE

Targeting c-Myc transactivation by LMNA inhibits tRNA processing essential for malate-aspartate shuttle and tumour progression

Author information +
History +
PDF

Abstract

Background: A series of studies have demonstrated the emerging involvement of transfer RNA (tRNA) processing during the progression of tumours. Nevertheless, the roles and regulating mechanisms of tRNA processing genes in neuroblastoma (NB), the prevalent malignant tumour outside the brain in children, are yet unknown.

Methods: Analysis of multi-omics results was conducted to identify crucial regulators of downstream tRNA processing genes. Co-immunoprecipitation and mass spectrometry methods were utilised to measure interaction between proteins. The impact of transcriptional regulators on expression of downstream genes was measured by dual-luciferase reporter, chromatin immunoprecipitation, western blotting and real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR) methods. Studies have been conducted to reveal impact and mechanisms of transcriptional regulators on biological processes of NB. Survival differences were analysed using the log-rank test.

Results: c-Myc was identified as a transcription factor driving tRNA processing gene expression and subsequent malate-aspartate shuttle (MAS) in NB cells. Mechanistically, c-Myc directly promoted the expression of glutamyl-prolyl-tRNA synthetase (EPRS) and leucyl-tRNA synthetase (LARS), resulting in translational up-regulation of glutamic-oxaloacetic transaminase 1 (GOT1) as well as malate dehydrogenase 1 (MDH1) via inhibiting general control nonrepressed 2 or activating mechanistic target of rapamycin signalling. Meanwhile, lamin A (LMNA) inhibited c-Myc transactivation via physical interaction, leading to suppression of MAS, aerobic glycolysis, tumourigenesis and aggressiveness. Pre-clinically, lobeline was discovered as a LMNA-binding compound to facilitate its interaction with c-Myc, which inhibited aminoacyl-tRNA synthetase expression, MAS and tumour progression of NB, as well as growth of organoid derived from c-Myc knock-in mice. Low levels of LMNA or elevated expression of c-Myc, EPRS, LARS, GOT1 or MDH1 were linked to a worse outcome and a shorter survival time of clinical NB patients.

Conclusions: These results suggest that targeting c-Myc transactivation by LMNA inhibits tRNA processing essential for MAS and tumour progression.

Keywords

c-Myc / lamin A / malate-aspartate shuttle / neuroblastoma / transfer RNA processing / tumour progression

Cite this article

Download citation ▾
Jianqun Wang, Mei Hong, Yang Cheng, Xiaojing Wang, Dan Li, Guo Chen, Banghe Bao, Jiyu Song, Xinyi Du, Chunhui Yang, Liduan Zheng, Qiangsong Tong. Targeting c-Myc transactivation by LMNA inhibits tRNA processing essential for malate-aspartate shuttle and tumour progression. Clinical and Translational Medicine, 2024, 14(5): e1680 DOI:10.1002/ctm2.1680

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Giegé R, Eriani G. The tRNA identity landscape for aminoacylation and beyond. Nucleic Acids Res. 2023;51:1528-1570.

[2]

Kwon NH, Fox PL, Kim S. Aminoacyl-tRNA synthetases as therapeutic targets. Nat Rev Drug Discov. 2019;18:629-650.

[3]

Liu H, Fredimoses M, Niu P, et al. EPRS/GluRS promotes gastric cancer development via WNT/GSK-3β/β-catenin signaling pathway. Gastric Cancer. 2021;24:1021-1036.

[4]

Gao G, Yao Y, Li K, et al. A human leucyl-tRNA synthetase as an anticancer target. Onco Targets Ther. 2015;8:2933-2942.

[5]

Borst P. The malate-aspartate shuttle (Borst cycle): how it started and developed into a major metabolic pathway. IUBMB Life. 2020;72:2241-2259.

[6]

Wang C, Chen H, Zhang M, Zhang J, Wei X, Ying W. Malate-aspartate shuttle inhibitor aminooxyacetic acid leads to decreased intracellular ATP levels and altered cell cycle of C6 glioma cells by inhibiting glycolysis. Cancer Lett. 2016;378:1-71.

[7]

Qiu B, Matthay KK. Advancing therapy for neuroblastoma. Nat Rev Clin Oncol. 2022;19:515-533.

[8]

Su Z, Fang H, Hong H, et al. An investigation of biomarkers derived from legacy microarray data for their utility in the RNA-seq era. Genome Biol. 2014;15:523.

[9]

Kildisiute G, Kholosy WM, Young MD, et al. Tumor to normal single-cell mRNA comparisons reveal a pan-neuroblastoma cancer cell. Sci Adv. 2021;7:eabd3311.

[10]

Lachmann A, Xu H, Krishnan J, Berger SI, Mazloom AR, Ma'ayan A. ChEA: transcription factor regulation inferred from integrating genome-wide ChIP-X experiments. Bioinformatics. 2010;26:2438-2444.

[11]

Keller TL, Zocco D, Sundrud MS, et al. Halofuginone and other febrifugine derivatives inhibit prolyl-tRNA synthetase. Nat Chem Biol. 2012;8:311-317.

[12]

Stark C, Breitkreutz B-J, Reguly T, Boucher L, Breitkreutz A, Tyers M. BioGRID: a general repository for interaction datasets. Nucleic Acids Res. 2006;34:D535-D539.

[13]

Pierce BG, Wiehe K, Hwang H, Kim BH, Vreven T, Weng Z. ZDOCK server: interactive docking prediction of protein-protein complexes and symmetric multimers. Bioinformatics. 2014;30:1771-1773.

[14]

Yamanishi Y, Kotera M, Moriya Y, Sawada R, Kanehisa M, Goto S. DINIES: drug‒target interaction network inference engine based on supervised analysis. Nucleic Acids Res. 2014;42:W39-W45.

[15]

Mendez D, Gaulton A, Bento AP, et al. ChEMBL: towards direct deposition of bioassay data. Nucleic Acids Res. 2018;47:D930-D940.

[16]

Ito T, Ando H, Suzuki T, et al. Identification of a primary target of thalidomide teratogenicity. Science. 2010;327:1345-1350.

[17]

Madden SK, de Araujo AD, Gerhardt M, Fairlie DP, Mason JM. Taking the Myc out of cancer: toward therapeutic strategies to directly inhibit c-Myc. Mol Cancer. 2021;20:3.

[18]

Wang LL, Teshiba R, Ikegaki N, et al. Augmented expression of MYC and/or MYCN protein defines highly aggressive MYC-driven neuroblastoma: a Children's Oncology Group study. Br J Cancer. 2015;113:57-63.

[19]

Zimmerman MW, Liu Y, He S, et al. MYC drives a subset of high-risk pediatric neuroblastomas and is activated through mechanisms including enhancer hijacking and focal enhancer amplification. Cancer Discov. 2018;8:320-335.

[20]

Zirin J, Ni X, Sack LM, et al. Interspecies analysis of MYC targets identifies tRNA synthetases as mediators of growth and survival in MYC-overexpressing cells. Proc Natl Acad Sci U S A. 2019;116:14614-14619.

[21]

Ye J, Palm W, Peng M, et al. GCN2 sustains mTORC1 suppression upon amino acid deprivation by inducing Sestrin2. Genes Dev. 2015;29:2331-2336.

[22]

Gold LT, Masson GR. GCN2: roles in tumour development and progression. Biochem Soc Trans. 2022;50:737-745.

[23]

Goberdhan DC, Wilson C, Harris AL. Amino acid sensing by mTORC1: intracellular transporters mark the spot. Cell Metab. 2016;23:580-589.

[24]

Han JM, Jeong SJ, Park MC, et al. Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway. Cell. 2012;149:410-424.

[25]

Shin SH, Kim HS, Jung SH, Xu HD, Jeong YB, Chung YJ. Implication of leucyl-tRNA synthetase 1 (LARS1) over-expression in growth and migration of lung cancer cells detected by siRNA targeted knock-down analysis. Exp Mol Med. 2008;40:229-236.

[26]

Reddy KL, Zullo JM, Bertolino E, Singh H. Transcriptional repression mediated by repositioning of genes to the nuclear lamina. Nature. 2008;452:243-247.

[27]

Johnson BR, Nitta RT, Frock RL, et al. A-type lamins regulate retinoblastoma protein function by promoting subnuclear localization and preventing proteasomal degradation. Proc Natl Acad Sci U S A. 2004;101:9677-9682.

[28]

Gonzalez JM, Navarro-Puche A, Casar B, Crespo P, Andres V. Fast regulation of AP-1 activity through interaction of lamin A/C, ERK1/2, and c-Fos at the nuclear envelope. J Cell Biol. 2008;183:653-666.

[29]

Bermeo S, Vidal C, Zhou H, Duque G. Lamin A/C acts as an essential factor in mesenchymal stem cell differentiation through the regulation of the dynamics of the Wnt/β-catenin pathway. J Cell Biochem. 2015;116:2344-2353.

[30]

Van Berlo JH, Voncken JW, Kubben N, et al. A-type lamins are essential for TGF-beta1 induced PP2A to dephosphorylate transcription factors. Hum Mol Genet. 2005;14:2839-2849.

[31]

de Leeuw R, Gruenbaum Y, Medalia O. Nuclear lamins: thin filaments with major functions. Trends Cell Biol. 2018;28:34-45.

[32]

Rauschert I, Aldunate F, Preussner J, et al. Promoter hypermethylation as a mechanism for lamin A/C silencing in a subset of neuroblastoma cells. PLoS One. 2017;12:e0175953.

[33]

Funke L, Bracht T, Oeck S, et al. NTRK1/TrkA signaling in neuroblastoma cells induces nuclear reorganization and intra-nuclear aggregation of lamin A/C. Cancers. 2021;13:5293.

[34]

Hann SR. Role of post-translational modifications in regulating c-Myc proteolysis, transcriptional activity and biological function. Semin Cancer Biol. 2006;16:288-302.

[35]

Myant K, Qiao X, Halonen T, et al. Serine 62-phosphorylated MYC associates with nuclear lamins and its regulation by CIP2A is essential for regenerative proliferation. Cell Rep. 2015;12:1019-1031.

[36]

Miller DK, Crooks PA, Zheng G, et al. Lobeline analogs with enhanced affinity and selectivity for plasmalemma and vesicular monoamine transporters. J Pharmacol Exp Ther. 2004;310:1035-1045.

[37]

Remya C, Dileep KV, Variyar EJ, Omkumar RV, Sadasivan C. Lobeline: a multifunctional alkaloid modulates cholinergic and glutamatergic activities. IUBMB Life. 2023;75:844-855.

[38]

Dwoskin LP, Crooks PA. A novel mechanism of action and potential use for lobeline as a treatment for psychostimulant abuse. Biochem Pharmacol. 2002;63:89-98.

[39]

Stead LF, Hughes JR. Lobeline for smoking cessation. Cochrane Database Syst Rev. 2012;2012:CD000124.

[40]

Ma Y, Wink M. Lobeline, a piperidine alkaloid from Lobelia can reverse P-gp dependent multidrug resistance in tumor cells. Phytomedicine. 2008;15:754-758.

[41]

Wang X, Guo Y, Chen G, et al. Therapeutic targeting of FUBP3 phase separation by GATA2-AS1 inhibits malate-aspartate shuttle and neuroblastoma progression via modulating SUZ12 activity. Oncogene. 2023;42:2673-2687.

[42]

Li H, Yang F, Hu A, et al. Therapeutic targeting of circ-CUX1/EWSR1/MAZ axis inhibits glycolysis and neuroblastoma progression. EMBO Mol Med. 2019;11:e10835.

[43]

Song H, Wang J, Wang X, et al. HNF4A-AS1-encoded small peptide promotes self-renewal and aggressiveness of neuroblastoma stem cells via eEF1A1-repressed SMAD4 transactivation. Oncogene. 2022;41:2505-2519.

[44]

Hu A, Chen G, Bao B, et al. Therapeutic targeting of CNBP phase separation inhibits ribosome biogenesis and neuroblastoma progression via modulating SWI/SNF complex activity. Clin Transl Med. 2023;13:e1235.

[45]

Li H, Jiao W, Song J, et al. circ-hnRNPU inhibits NONO-mediated c-Myc transactivation and mRNA stabilization essential for glycosylation and cancer progression. J Exp Clin Cancer Res. 2023;42:313.

[46]

Yang J, Smith DK, Ni H, et al. SOX4-mediated repression of specific tRNAs inhibits proliferation of human glioblastoma cells. Proc Natl Acad Sci U S A. 2020;117:5782-5790.

[47]

Nottrott S, Simard MJ, Richter JD. Human let-7a miRNA blocks protein production on actively translating polyribosomes. Nat Struct Mol Biol. 2006;13:1108-1114.

[48]

Hidalgo San Jose L, Signer RAJ. Cell-type-specific quantification of protein synthesis in vivo. Nat Protoc. 2019;14:441-460.

[49]

Son MJ, Ryu JS, Kim JY, et al. Upregulation of mitochondrial NAD(+) levels impairs the clonogenicity of SSEA1(+) glioblastoma tumor-initiating cells. Exp Mol Med. 2017;49:e344.

[50]

Chen Y, Yang F, Fang E, et al. Circular RNA circAGO2 drives cancer progression through facilitating HuR-repressed functions of AGO2-miRNA complexes. Cell Death Differ. 2019;26:1346-1364.

[51]

Poli G, Sarchielli E, Guasti D, et al. Human fetal adrenal cells retain age-related stem- and endocrine-differentiation potential in culture. FASEB J. 2019;33:2263-2277.

RIGHTS & PERMISSIONS

2024 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

145

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/