Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2024

Open Access 01-12-2024 | Gastric Cancer | Research

MAGOH promotes gastric cancer progression via hnRNPA1 expression inhibition-mediated RONΔ160/PI3K/AKT signaling pathway activation

Authors: Shanshan Yu, Cheng Chen, Ming Chen, Jinxiao Liang, Kecheng Jiang, Bin Lou, Jun Lu, Xiaohua Zhu, Donghui Zhou

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2024

Login to get access

Abstract

Background

Gastric cancer (GC) is associated with high mortality and heterogeneity and poses a great threat to humans. Gene therapies for the receptor tyrosine kinase RON and its spliceosomes are attracting increasing amounts of attention due to their unique characteristics. However, little is known about the mechanism involved in the formation of the RON mRNA alternative spliceosome RONΔ160.

Methods

Fourteen human GC tissue samples and six normal gastric tissue samples were subjected to label-free relative quantitative proteomics analysis, and MAGOH was identified as a candidate protein for subsequent studies. The expression of MAGOH in clinical specimens was verified by quantitative real-time PCR and western blotting. We then determined the biological function of MAGOH in GC through in vitro and in vivo experiments. RNA pulldown, RNA sequencing and RNA immunoprecipitation (RIP) were subsequently conducted to uncover the underlying mechanism by which MAGOH regulated the formation of RONΔ160.

Results

Proteomic analysis revealed that MAGOH, which is located at key nodes and participates in RNA processing and mRNA splicing, was upregulated in GC tissue and GC cell lines and was associated with poor prognosis. Functional analysis showed that MAGOH promoted the proliferation, migration and invasion of GC cells in vitro and in vivo. Mechanistically, MAGOH inhibited the expression of hnRNPA1 and reduced the binding of hnRNPA1 to RON mRNA, thereby promoting the formation of RONΔ160 to activate the PI3K/AKT signaling pathway and consequently facilitating GC progression.

Conclusions

Our study revealed that MAGOH could promote the formation of RONΔ160 and activate the PI3K/AKT signaling pathway through the inhibition of hnRNPA1 expression. We elucidate a novel mechanism and potential therapeutic targets for the growth and metastasis of GC based on the MAGOH-RONΔ160 axis, and these findings have important guiding significance and clinical value for the future development of effective therapeutic strategies for GC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sung H, Ferlay J, Siegel R, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef Sung H, Ferlay J, Siegel R, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef
2.
go back to reference In H, Solsky I, Palis B, Langdon-Embry M, Ajani J, Sano T. Validation of the 8th edition of the AJCC TNM staging system for gastric cancer using the national cancer database. Ann Surg Oncol. 2017;24(12):3683–91.PubMedCrossRef In H, Solsky I, Palis B, Langdon-Embry M, Ajani J, Sano T. Validation of the 8th edition of the AJCC TNM staging system for gastric cancer using the national cancer database. Ann Surg Oncol. 2017;24(12):3683–91.PubMedCrossRef
3.
go back to reference Smyth E, Nilsson M, Grabsch H, van Grieken N, Lordick F. Gastric cancer. Lancet (London, England). 2020;396(10251):635–48.PubMedCrossRef Smyth E, Nilsson M, Grabsch H, van Grieken N, Lordick F. Gastric cancer. Lancet (London, England). 2020;396(10251):635–48.PubMedCrossRef
4.
go back to reference Zhou Y, Chen Y, Shi Y, Wu L, Tan Y, Li T, et al. FAM117B promotes gastric cancer growth and drug resistance by targeting the KEAP1/NRF2 signaling pathway. J Clin Invest. 2023;133(3):e158705.PubMedPubMedCentralCrossRef Zhou Y, Chen Y, Shi Y, Wu L, Tan Y, Li T, et al. FAM117B promotes gastric cancer growth and drug resistance by targeting the KEAP1/NRF2 signaling pathway. J Clin Invest. 2023;133(3):e158705.PubMedPubMedCentralCrossRef
6.
go back to reference Chen Z, Li Y, Tan B, Zhao Q, Fan L, Li F, et al. Progress and current status of molecule-targeted therapy and drug resistance in gastric cancer. Drugs Today (Barc). 2020;56(7):469–82.PubMedCrossRef Chen Z, Li Y, Tan B, Zhao Q, Fan L, Li F, et al. Progress and current status of molecule-targeted therapy and drug resistance in gastric cancer. Drugs Today (Barc). 2020;56(7):469–82.PubMedCrossRef
7.
go back to reference Kawakami H, Okamoto I. MET-targeted therapy for gastric cancer: the importance of a biomarker-based strategy. Gastric Cancer. 2016;19(3):687–95.PubMedCrossRef Kawakami H, Okamoto I. MET-targeted therapy for gastric cancer: the importance of a biomarker-based strategy. Gastric Cancer. 2016;19(3):687–95.PubMedCrossRef
8.
go back to reference Allegra A, Alonci A, Penna G, Innao V, Gerace D, Rotondo F, et al. The cancer stem cell hypothesis: a guide to potential molecular targets. Cancer Invest. 2014;32(9):470–95.PubMedCrossRef Allegra A, Alonci A, Penna G, Innao V, Gerace D, Rotondo F, et al. The cancer stem cell hypothesis: a guide to potential molecular targets. Cancer Invest. 2014;32(9):470–95.PubMedCrossRef
9.
go back to reference Lu H, Clauser K, Tam W, Fröse J, Ye X, Eaton E, et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat Cell Biol. 2014;16(11):1105–17.PubMedPubMedCentralCrossRef Lu H, Clauser K, Tam W, Fröse J, Ye X, Eaton E, et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat Cell Biol. 2014;16(11):1105–17.PubMedPubMedCentralCrossRef
10.
go back to reference Jiang J, Zhang Y, Chuai S, Wang Z, Zheng D, Xu F, et al. Trastuzumab (herceptin) targets gastric cancer stem cells characterized by CD90 phenotype. Oncogene. 2012;31(6):671–82.PubMedCrossRef Jiang J, Zhang Y, Chuai S, Wang Z, Zheng D, Xu F, et al. Trastuzumab (herceptin) targets gastric cancer stem cells characterized by CD90 phenotype. Oncogene. 2012;31(6):671–82.PubMedCrossRef
12.
go back to reference Black D. Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem. 2003;72:291–336.PubMedCrossRef Black D. Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem. 2003;72:291–336.PubMedCrossRef
13.
go back to reference Climente-González H, Porta-Pardo E, Godzik A, Eyras E. The functional impact of alternative splicing in cancer. Cell Rep. 2017;20(9):2215–26.PubMedCrossRef Climente-González H, Porta-Pardo E, Godzik A, Eyras E. The functional impact of alternative splicing in cancer. Cell Rep. 2017;20(9):2215–26.PubMedCrossRef
14.
go back to reference Ma Q, Tatsuno T, Nakamura Y, Ishigaki Y. The stability of Magoh and Y14 depends on their heterodimer formation and nuclear localization. Biochem Biophys Res Commun. 2019;511(3):631–6.PubMedCrossRef Ma Q, Tatsuno T, Nakamura Y, Ishigaki Y. The stability of Magoh and Y14 depends on their heterodimer formation and nuclear localization. Biochem Biophys Res Commun. 2019;511(3):631–6.PubMedCrossRef
15.
go back to reference Singh K, Wachsmuth L, Kulozik A, Gehring N. Two mammalian MAGOH genes contribute to exon junction complex composition and nonsense-mediated decay. RNA Biol. 2013;10(8):1291–8.PubMedPubMedCentralCrossRef Singh K, Wachsmuth L, Kulozik A, Gehring N. Two mammalian MAGOH genes contribute to exon junction complex composition and nonsense-mediated decay. RNA Biol. 2013;10(8):1291–8.PubMedPubMedCentralCrossRef
16.
go back to reference Viswanathan S, Nogueira M, Buss C, Krill-Burger J, Wawer M, Malolepsza E, et al. Genome-scale analysis identifies paralog lethality as a vulnerability of chromosome 1p loss in cancer. Nat Genet. 2018;50(7):937–43.PubMedPubMedCentralCrossRef Viswanathan S, Nogueira M, Buss C, Krill-Burger J, Wawer M, Malolepsza E, et al. Genome-scale analysis identifies paralog lethality as a vulnerability of chromosome 1p loss in cancer. Nat Genet. 2018;50(7):937–43.PubMedPubMedCentralCrossRef
17.
go back to reference Muromoto R, Taira N, Ikeda O, Shiga K, Kamitani S, Togi S, et al. The exon-junction complex proteins, Y14 and MAGOH regulate STAT3 activation. Biochem Biophys Res Commun. 2009;382(1):63–8.PubMedCrossRef Muromoto R, Taira N, Ikeda O, Shiga K, Kamitani S, Togi S, et al. The exon-junction complex proteins, Y14 and MAGOH regulate STAT3 activation. Biochem Biophys Res Commun. 2009;382(1):63–8.PubMedCrossRef
18.
go back to reference Kataoka N, Diem M, Kim V, Yong J, Dreyfuss G. Magoh, a human homolog of Drosophila mago nashi protein, is a component of the splicing-dependent exon-exon junction complex. EMBO J. 2001;20(22):6424–33.PubMedPubMedCentralCrossRef Kataoka N, Diem M, Kim V, Yong J, Dreyfuss G. Magoh, a human homolog of Drosophila mago nashi protein, is a component of the splicing-dependent exon-exon junction complex. EMBO J. 2001;20(22):6424–33.PubMedPubMedCentralCrossRef
19.
go back to reference Lau C, Diem M, Dreyfuss G, Van Duyne G. Structure of the Y14-Magoh core of the exon junction complex. Curr Biol. 2003;13(11):933–41.PubMedCrossRef Lau C, Diem M, Dreyfuss G, Van Duyne G. Structure of the Y14-Magoh core of the exon junction complex. Curr Biol. 2003;13(11):933–41.PubMedCrossRef
20.
go back to reference Le Hir H, Gatfield D, Braun I, Forler D, Izaurralde E. The protein Mago provides a link between splicing and mRNA localization. EMBO Rep. 2001;2(12):1119–24.PubMedPubMedCentralCrossRef Le Hir H, Gatfield D, Braun I, Forler D, Izaurralde E. The protein Mago provides a link between splicing and mRNA localization. EMBO Rep. 2001;2(12):1119–24.PubMedPubMedCentralCrossRef
21.
go back to reference Ballut L, Marchadier B, Baguet A, Tomasetto C, Séraphin B, Le Hir H. The exon junction core complex is locked onto RNA by inhibition of eIF4AIII ATPase activity. Nat Struct Mol Biol. 2005;12(10):861–9.PubMedCrossRef Ballut L, Marchadier B, Baguet A, Tomasetto C, Séraphin B, Le Hir H. The exon junction core complex is locked onto RNA by inhibition of eIF4AIII ATPase activity. Nat Struct Mol Biol. 2005;12(10):861–9.PubMedCrossRef
22.
go back to reference Soederberg A, Meißgeier T, Bosserhoff A, Linck-Paulus L. MAGOH and MAGOHB knockdown in melanoma cells decreases nonsense-mediated decay activity and promotes apoptosis via upregulation of GADD45A. Cells. 2022;11(23):3859.PubMedPubMedCentralCrossRef Soederberg A, Meißgeier T, Bosserhoff A, Linck-Paulus L. MAGOH and MAGOHB knockdown in melanoma cells decreases nonsense-mediated decay activity and promotes apoptosis via upregulation of GADD45A. Cells. 2022;11(23):3859.PubMedPubMedCentralCrossRef
23.
go back to reference Barreiro R, Guardia G, Meliso F, Lei X, Li W, Savio A, et al. The paralogues MAGOH and MAGOHB are oncogenic factors in high-grade gliomas and safeguard the splicing of cell division and cell cycle genes. RNA Biol. 2023;20(1):311–22.PubMedPubMedCentralCrossRef Barreiro R, Guardia G, Meliso F, Lei X, Li W, Savio A, et al. The paralogues MAGOH and MAGOHB are oncogenic factors in high-grade gliomas and safeguard the splicing of cell division and cell cycle genes. RNA Biol. 2023;20(1):311–22.PubMedPubMedCentralCrossRef
24.
go back to reference Zhou Y, Li Z, Wu X, Tou L, Zheng J, Zhou D. MAGOH/MAGOHB inhibits the tumorigenesis of gastric cancer via inactivation of b-RAF/MEK/ERK signaling. Onco Targets Ther. 2020;13:12723–35.PubMedPubMedCentralCrossRef Zhou Y, Li Z, Wu X, Tou L, Zheng J, Zhou D. MAGOH/MAGOHB inhibits the tumorigenesis of gastric cancer via inactivation of b-RAF/MEK/ERK signaling. Onco Targets Ther. 2020;13:12723–35.PubMedPubMedCentralCrossRef
25.
go back to reference Krishnaswamy S, Mohammed A, Tripathi G, Alokail M, Al-Daghri N. Splice variants of the extracellular region of RON receptor tyrosine kinase in lung cancer cell lines identified by PCR and sequencing. BMC Cancer. 2017;17(1):738.PubMedPubMedCentralCrossRef Krishnaswamy S, Mohammed A, Tripathi G, Alokail M, Al-Daghri N. Splice variants of the extracellular region of RON receptor tyrosine kinase in lung cancer cell lines identified by PCR and sequencing. BMC Cancer. 2017;17(1):738.PubMedPubMedCentralCrossRef
26.
go back to reference Yao H, Zhou Y, Zhang R, Wang M. MSP-RON signalling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 2013;13(7):466–81.PubMedCrossRef Yao H, Zhou Y, Zhang R, Wang M. MSP-RON signalling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 2013;13(7):466–81.PubMedCrossRef
27.
go back to reference Zhou D, Pan G, Zheng C, Zheng J, Yian L, Teng X. Expression of the RON receptor tyrosine kinase and its association with gastric carcinoma versus normal gastric tissues. BMC Cancer. 2008;8:353.PubMedPubMedCentralCrossRef Zhou D, Pan G, Zheng C, Zheng J, Yian L, Teng X. Expression of the RON receptor tyrosine kinase and its association with gastric carcinoma versus normal gastric tissues. BMC Cancer. 2008;8:353.PubMedPubMedCentralCrossRef
28.
go back to reference Wang D, Shen Q, Chen Y, Wang M. Collaborative activities of macrophage-stimulating protein and transforming growth factor-beta1 in induction of epithelial to mesenchymal transition: roles of the RON receptor tyrosine kinase. Oncogene. 2004;23(9):1668–80.PubMedCrossRef Wang D, Shen Q, Chen Y, Wang M. Collaborative activities of macrophage-stimulating protein and transforming growth factor-beta1 in induction of epithelial to mesenchymal transition: roles of the RON receptor tyrosine kinase. Oncogene. 2004;23(9):1668–80.PubMedCrossRef
29.
go back to reference Wang Q, Quan H, Zhao J, Xie C, Wang L, Lou L. RON confers lapatinib resistance in HER2-positive breast cancer cells. Cancer Lett. 2013;340(1):43–50.PubMedCrossRef Wang Q, Quan H, Zhao J, Xie C, Wang L, Lou L. RON confers lapatinib resistance in HER2-positive breast cancer cells. Cancer Lett. 2013;340(1):43–50.PubMedCrossRef
30.
go back to reference Yang S, Nguyen T, Ung T, Jung Y. Role of recepteur d’origine nantais on gastric cancer development and progression. Chonnam Med J. 2017;53(3):178–86.PubMedPubMedCentralCrossRef Yang S, Nguyen T, Ung T, Jung Y. Role of recepteur d’origine nantais on gastric cancer development and progression. Chonnam Med J. 2017;53(3):178–86.PubMedPubMedCentralCrossRef
31.
go back to reference Lu Y, Yao H, Wang M. Multiple variants of the RON receptor tyrosine kinase: biochemical properties, tumorigenic activities, and potential drug targets. Cancer Lett. 2007;257(2):157–64.PubMedCrossRef Lu Y, Yao H, Wang M. Multiple variants of the RON receptor tyrosine kinase: biochemical properties, tumorigenic activities, and potential drug targets. Cancer Lett. 2007;257(2):157–64.PubMedCrossRef
32.
go back to reference Bardella C, Costa B, Maggiora P, Patane’ S, Olivero M, Ranzani G, et al. Truncated RON tyrosine kinase drives tumor cell progression and abrogates cell-cell adhesion through E-cadherin transcriptional repression. Cancer Res. 2004;64(15):5154–61.PubMedCrossRef Bardella C, Costa B, Maggiora P, Patane’ S, Olivero M, Ranzani G, et al. Truncated RON tyrosine kinase drives tumor cell progression and abrogates cell-cell adhesion through E-cadherin transcriptional repression. Cancer Res. 2004;64(15):5154–61.PubMedCrossRef
33.
go back to reference Zhang K, Zhou Y, Yao H, Wang M. Alterations in a defined extracellular region of the RON receptor tyrosine kinase promote RON-mediated motile and invasive phenotypes in epithelial cells. Int J Oncol. 2010;36(1):255–64.PubMed Zhang K, Zhou Y, Yao H, Wang M. Alterations in a defined extracellular region of the RON receptor tyrosine kinase promote RON-mediated motile and invasive phenotypes in epithelial cells. Int J Oncol. 2010;36(1):255–64.PubMed
34.
go back to reference Ma Q, Zhang K, Guin S, Zhou Y, Wang M. Deletion or insertion in the first immunoglobulin-plexin-transcription (IPT) domain differentially regulates expression and tumorigenic activities of RON receptor tyrosine kinase. Mol Cancer. 2010;9:307.PubMedPubMedCentralCrossRef Ma Q, Zhang K, Guin S, Zhou Y, Wang M. Deletion or insertion in the first immunoglobulin-plexin-transcription (IPT) domain differentially regulates expression and tumorigenic activities of RON receptor tyrosine kinase. Mol Cancer. 2010;9:307.PubMedPubMedCentralCrossRef
35.
go back to reference Ma Q, Zhang K, Yao H, Zhou Y, Padhye S, Wang M. Inhibition of MSP-RON signaling pathway in cancer cells by a novel soluble form of RON comprising the entire sema sequence. Int J Oncol. 2010;36(6):1551–61.PubMed Ma Q, Zhang K, Yao H, Zhou Y, Padhye S, Wang M. Inhibition of MSP-RON signaling pathway in cancer cells by a novel soluble form of RON comprising the entire sema sequence. Int J Oncol. 2010;36(6):1551–61.PubMed
36.
go back to reference Braun S, Enculescu M, Setty S, Cortés-López M, de Almeida B, Sutandy F, et al. Decoding a cancer-relevant splicing decision in the RON proto-oncogene using high-throughput mutagenesis. Nat Commun. 2018;9(1):3315.PubMedPubMedCentralCrossRef Braun S, Enculescu M, Setty S, Cortés-López M, de Almeida B, Sutandy F, et al. Decoding a cancer-relevant splicing decision in the RON proto-oncogene using high-throughput mutagenesis. Nat Commun. 2018;9(1):3315.PubMedPubMedCentralCrossRef
37.
go back to reference Collesi C, Santoro M, Gaudino G, Comoglio P. A splicing variant of the RON transcript induces constitutive tyrosine kinase activity and an invasive phenotype. Mol Cell Biol. 1996;16(10):5518–26.PubMedPubMedCentralCrossRef Collesi C, Santoro M, Gaudino G, Comoglio P. A splicing variant of the RON transcript induces constitutive tyrosine kinase activity and an invasive phenotype. Mol Cell Biol. 1996;16(10):5518–26.PubMedPubMedCentralCrossRef
38.
go back to reference Zhou Y, He C, Chen Y, Wang D, Wang M. Altered expression of the RON receptor tyrosine kinase in primary human colorectal adenocarcinomas: generation of different splicing RON variants and their oncogenic potential. Oncogene. 2003;22(2):186–97.PubMedCrossRef Zhou Y, He C, Chen Y, Wang D, Wang M. Altered expression of the RON receptor tyrosine kinase in primary human colorectal adenocarcinomas: generation of different splicing RON variants and their oncogenic potential. Oncogene. 2003;22(2):186–97.PubMedCrossRef
39.
go back to reference Zhou D, Huang L, Zhou Y, Wei T, Yang L, Li C. RON and RONΔ160 promote gastric cancer cell proliferation, migration, and adaption to hypoxia via interaction with β-catenin. Aging. 2019;11(9):2735–48.PubMedPubMedCentralCrossRef Zhou D, Huang L, Zhou Y, Wei T, Yang L, Li C. RON and RONΔ160 promote gastric cancer cell proliferation, migration, and adaption to hypoxia via interaction with β-catenin. Aging. 2019;11(9):2735–48.PubMedPubMedCentralCrossRef
40.
go back to reference Zhou D, Li C, Yang L. Variant RONΔ160 of the RON receptor tyrosine kinase promotes the growth and invasion in vitro and in vivo in gastric cancer cell lines. Cancer Cell Int. 2015;15(1):9.PubMedPubMedCentralCrossRef Zhou D, Li C, Yang L. Variant RONΔ160 of the RON receptor tyrosine kinase promotes the growth and invasion in vitro and in vivo in gastric cancer cell lines. Cancer Cell Int. 2015;15(1):9.PubMedPubMedCentralCrossRef
41.
go back to reference Wang M, Kurtz A, Chen Y. Identification of a novel splicing product of the RON receptor tyrosine kinase in human colorectal carcinoma cells. Carcinogenesis. 2000;21(8):1507–12.PubMedCrossRef Wang M, Kurtz A, Chen Y. Identification of a novel splicing product of the RON receptor tyrosine kinase in human colorectal carcinoma cells. Carcinogenesis. 2000;21(8):1507–12.PubMedCrossRef
42.
go back to reference Chen Y, Zhou Y, Angeloni D, Kurtz A, Qiang X, Wang M. Overexpression and activation of the RON receptor tyrosine kinase in a panel of human colorectal carcinoma cell lines. Exp Cell Res. 2000;261(1):229–38.PubMedCrossRef Chen Y, Zhou Y, Angeloni D, Kurtz A, Qiang X, Wang M. Overexpression and activation of the RON receptor tyrosine kinase in a panel of human colorectal carcinoma cell lines. Exp Cell Res. 2000;261(1):229–38.PubMedCrossRef
43.
go back to reference Kumar K, Chow C, Ebine K, Arslan A, Kwok B, Bentrem D, et al. Differential regulation of ZEB1 and EMT by MAPK-Interacting Protein Kinases (MNK) and eIF4E in pancreatic cancer. Mol Cancer Res. 2016;14(2):216–27.PubMedCrossRef Kumar K, Chow C, Ebine K, Arslan A, Kwok B, Bentrem D, et al. Differential regulation of ZEB1 and EMT by MAPK-Interacting Protein Kinases (MNK) and eIF4E in pancreatic cancer. Mol Cancer Res. 2016;14(2):216–27.PubMedCrossRef
44.
go back to reference Buxadé M, Parra J, Rousseau S, Shpiro N, Marquez R, Morrice N, et al. The Mnks are novel components in the control of TNF alpha biosynthesis and phosphorylate and regulate hnRNP A1. Immunity. 2005;23(2):177–89.PubMedCrossRef Buxadé M, Parra J, Rousseau S, Shpiro N, Marquez R, Morrice N, et al. The Mnks are novel components in the control of TNF alpha biosynthesis and phosphorylate and regulate hnRNP A1. Immunity. 2005;23(2):177–89.PubMedCrossRef
45.
go back to reference Buxade M, Parra-Palau J, Proud C. The Mnks: MAP kinase-interacting kinases (MAP kinase signal-integrating kinases). Front Biosci. 2008;13:5359–73.PubMedCrossRef Buxade M, Parra-Palau J, Proud C. The Mnks: MAP kinase-interacting kinases (MAP kinase signal-integrating kinases). Front Biosci. 2008;13:5359–73.PubMedCrossRef
46.
go back to reference Bonomi S, di Matteo A, Buratti E, Cabianca D, Baralle F, Ghigna C, et al. HnRNP A1 controls a splicing regulatory circuit promoting mesenchymal-to-epithelial transition. Nucleic Acids Res. 2013;41(18):8665–79.PubMedPubMedCentralCrossRef Bonomi S, di Matteo A, Buratti E, Cabianca D, Baralle F, Ghigna C, et al. HnRNP A1 controls a splicing regulatory circuit promoting mesenchymal-to-epithelial transition. Nucleic Acids Res. 2013;41(18):8665–79.PubMedPubMedCentralCrossRef
48.
go back to reference Mayeda A, Helfman D, Krainer A. Modulation of exon skipping and inclusion by heterogeneous nuclear ribonucleoprotein A1 and pre-mRNA splicing factor SF2/ASF. Mol Cell Biol. 1993;13(5):2993–3001.PubMedPubMedCentral Mayeda A, Helfman D, Krainer A. Modulation of exon skipping and inclusion by heterogeneous nuclear ribonucleoprotein A1 and pre-mRNA splicing factor SF2/ASF. Mol Cell Biol. 1993;13(5):2993–3001.PubMedPubMedCentral
49.
go back to reference Wu S, Chen M, Huang J, Zhang F, Lv Z, Jia Y, et al. ORAI2 Promotes gastric cancer tumorigenicity and metastasis through PI3K/Akt signaling and MAPK-dependent focal adhesion disassembly. Cancer Res. 2021;81(4):986–1000.PubMedCrossRef Wu S, Chen M, Huang J, Zhang F, Lv Z, Jia Y, et al. ORAI2 Promotes gastric cancer tumorigenicity and metastasis through PI3K/Akt signaling and MAPK-dependent focal adhesion disassembly. Cancer Res. 2021;81(4):986–1000.PubMedCrossRef
50.
go back to reference Zhang F, Li K, Yao X, Wang H, Li W, Wu J, et al. A miR-567-PIK3AP1-PI3K/AKT-c-Myc feedback loop regulates tumour growth and chemoresistance in gastric cancer. EBioMedicine. 2019;44:311–21.PubMedPubMedCentralCrossRef Zhang F, Li K, Yao X, Wang H, Li W, Wu J, et al. A miR-567-PIK3AP1-PI3K/AKT-c-Myc feedback loop regulates tumour growth and chemoresistance in gastric cancer. EBioMedicine. 2019;44:311–21.PubMedPubMedCentralCrossRef
51.
go back to reference Huang Y, Zhang J, Hou L, Wang G, Liu H, Zhang R, et al. LncRNA AK023391 promotes tumorigenesis and invasion of gastric cancer through activation of the PI3K/Akt signaling pathway. J Exp Clin Cancer Res. 2017;36(1):194.PubMedPubMedCentralCrossRef Huang Y, Zhang J, Hou L, Wang G, Liu H, Zhang R, et al. LncRNA AK023391 promotes tumorigenesis and invasion of gastric cancer through activation of the PI3K/Akt signaling pathway. J Exp Clin Cancer Res. 2017;36(1):194.PubMedPubMedCentralCrossRef
53.
go back to reference Li X, Sun Z, Peng G, Xiao Y, Guo J, Wu B, et al. Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical outcomes in patients with gastric cancer. Theranostics. 2022;12(2):620–38.PubMedPubMedCentralCrossRef Li X, Sun Z, Peng G, Xiao Y, Guo J, Wu B, et al. Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical outcomes in patients with gastric cancer. Theranostics. 2022;12(2):620–38.PubMedPubMedCentralCrossRef
54.
go back to reference Xiao F, Long Z, Guo Y, Zhu H, Zhang Z, Xiao Y, et al. MAGOH is correlated with poor prognosis and is essential for cell proliferation in lower-grade glioma. Aging. 2023;15(12):5713–33.PubMedPubMedCentral Xiao F, Long Z, Guo Y, Zhu H, Zhang Z, Xiao Y, et al. MAGOH is correlated with poor prognosis and is essential for cell proliferation in lower-grade glioma. Aging. 2023;15(12):5713–33.PubMedPubMedCentral
55.
go back to reference Ishigaki Y, Nakamura Y, Tatsuno T, Hashimoto M, Shimasaki T, Iwabuchi K, et al. Depletion of RNA-binding protein RBM8A (Y14) causes cell cycle deficiency and apoptosis in human cells. Exp Biol Med (Maywood). 2013;238(8):889–97.PubMedCrossRef Ishigaki Y, Nakamura Y, Tatsuno T, Hashimoto M, Shimasaki T, Iwabuchi K, et al. Depletion of RNA-binding protein RBM8A (Y14) causes cell cycle deficiency and apoptosis in human cells. Exp Biol Med (Maywood). 2013;238(8):889–97.PubMedCrossRef
56.
go back to reference Ruiz-Torres S, Bourn J, Benight N, Hunt B, Lester C, Waltz S. Macrophage-mediated RON signaling supports breast cancer growth and progression through modulation of IL-35. Oncogene. 2022;41(3):321–33.PubMedCrossRef Ruiz-Torres S, Bourn J, Benight N, Hunt B, Lester C, Waltz S. Macrophage-mediated RON signaling supports breast cancer growth and progression through modulation of IL-35. Oncogene. 2022;41(3):321–33.PubMedCrossRef
57.
go back to reference Wang Z, Yang Y, Hu S, He J, Wu Z, Qi Z, et al. Short-form RON (sf-RON) enhances glucose metabolism to promote cell proliferation via activating β-catenin/SIX1 signaling pathway in gastric cancer. Cell Biol Toxicol. 2021;37(1):35–49.PubMedCrossRef Wang Z, Yang Y, Hu S, He J, Wu Z, Qi Z, et al. Short-form RON (sf-RON) enhances glucose metabolism to promote cell proliferation via activating β-catenin/SIX1 signaling pathway in gastric cancer. Cell Biol Toxicol. 2021;37(1):35–49.PubMedCrossRef
58.
go back to reference Wu Z, Zhang Z, Ge X, Lin Y, Dai C, Chang J, et al. Identification of short-form RON as a novel intrinsic resistance mechanism for anti-MET therapy in MET-positive gastric cancer. Oncotarget. 2015;6(38):40519–34.PubMedPubMedCentralCrossRef Wu Z, Zhang Z, Ge X, Lin Y, Dai C, Chang J, et al. Identification of short-form RON as a novel intrinsic resistance mechanism for anti-MET therapy in MET-positive gastric cancer. Oncotarget. 2015;6(38):40519–34.PubMedPubMedCentralCrossRef
59.
go back to reference Mayeda A, Krainer A. Regulation of alternative pre-mRNA splicing by hnRNP A1 and splicing factor SF2. Cell. 1992;68(2):365–75.PubMedCrossRef Mayeda A, Krainer A. Regulation of alternative pre-mRNA splicing by hnRNP A1 and splicing factor SF2. Cell. 1992;68(2):365–75.PubMedCrossRef
60.
go back to reference Izaurralde E, Jarmolowski A, Beisel C, Mattaj I, Dreyfuss G, Fischer U. A role for the M9 transport signal of hnRNP A1 in mRNA nuclear export. J Cell Biol. 1997;137(1):27–35.PubMedPubMedCentralCrossRef Izaurralde E, Jarmolowski A, Beisel C, Mattaj I, Dreyfuss G, Fischer U. A role for the M9 transport signal of hnRNP A1 in mRNA nuclear export. J Cell Biol. 1997;137(1):27–35.PubMedPubMedCentralCrossRef
61.
go back to reference Chabot B, LeBel C, Hutchison S, Nasim F, Simard M. Heterogeneous nuclear ribonucleoprotein particle A/B proteins and the control of alternative splicing of the mammalian heterogeneous nuclear ribonucleoprotein particle A1 pre-mRNA. Prog Mol Subcell Biol. 2003;31:59–88.PubMedCrossRef Chabot B, LeBel C, Hutchison S, Nasim F, Simard M. Heterogeneous nuclear ribonucleoprotein particle A/B proteins and the control of alternative splicing of the mammalian heterogeneous nuclear ribonucleoprotein particle A1 pre-mRNA. Prog Mol Subcell Biol. 2003;31:59–88.PubMedCrossRef
62.
go back to reference Dreyfuss G, Kim V, Kataoka N. Messenger-RNA-binding proteins and the messages they carry. Nat Rev Mol Cell Biol. 2002;3(3):195–205.PubMedCrossRef Dreyfuss G, Kim V, Kataoka N. Messenger-RNA-binding proteins and the messages they carry. Nat Rev Mol Cell Biol. 2002;3(3):195–205.PubMedCrossRef
63.
go back to reference Kędzierska H, Piekiełko-Witkowska A. Splicing factors of SR and hnRNP families as regulators of apoptosis in cancer. Cancer Lett. 2017;396:53–65.PubMedCrossRef Kędzierska H, Piekiełko-Witkowska A. Splicing factors of SR and hnRNP families as regulators of apoptosis in cancer. Cancer Lett. 2017;396:53–65.PubMedCrossRef
65.
go back to reference Burd C, Dreyfuss G. RNA binding specificity of hnRNP A1: significance of hnRNP A1 high-affinity binding sites in pre-mRNA splicing. EMBO J. 1994;13(5):1197–204.PubMedPubMedCentralCrossRef Burd C, Dreyfuss G. RNA binding specificity of hnRNP A1: significance of hnRNP A1 high-affinity binding sites in pre-mRNA splicing. EMBO J. 1994;13(5):1197–204.PubMedPubMedCentralCrossRef
66.
go back to reference Moshiri A, Puppo M, Rossi M, Gherzi R, Briata P. Resveratrol limits epithelial to mesenchymal transition through modulation of KHSRP/hnRNPA1-dependent alternative splicing in mammary gland cells. Biochim Biophys Acta. 2017;1860(3):291–8.CrossRef Moshiri A, Puppo M, Rossi M, Gherzi R, Briata P. Resveratrol limits epithelial to mesenchymal transition through modulation of KHSRP/hnRNPA1-dependent alternative splicing in mammary gland cells. Biochim Biophys Acta. 2017;1860(3):291–8.CrossRef
67.
go back to reference Puppo M, Bucci G, Rossi M, Giovarelli M, Bordo D, Moshiri A, et al. miRNA-Mediated KHSRP silencing rewires distinct post-transcriptional programs during TGF-β-induced epithelial-to-mesenchymal transition. Cell Rep. 2016;16(4):967–78.PubMedCrossRef Puppo M, Bucci G, Rossi M, Giovarelli M, Bordo D, Moshiri A, et al. miRNA-Mediated KHSRP silencing rewires distinct post-transcriptional programs during TGF-β-induced epithelial-to-mesenchymal transition. Cell Rep. 2016;16(4):967–78.PubMedCrossRef
68.
go back to reference Han P, Cao P, Yue J, Kong K, Hu S, Deng Y, et al. Knockdown of hnRNPA1 promotes NSCLC metastasis and EMT by regulating alternative splicing of LAS1L exon 9. Front Oncol. 2022;12:837248.PubMedPubMedCentralCrossRef Han P, Cao P, Yue J, Kong K, Hu S, Deng Y, et al. Knockdown of hnRNPA1 promotes NSCLC metastasis and EMT by regulating alternative splicing of LAS1L exon 9. Front Oncol. 2022;12:837248.PubMedPubMedCentralCrossRef
69.
go back to reference David C, Chen M, Assanah M, Canoll P, Manley J. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463(7279):364–8.PubMedCrossRef David C, Chen M, Assanah M, Canoll P, Manley J. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463(7279):364–8.PubMedCrossRef
70.
go back to reference Chen Y, Liu J, Wang W, Xiang L, Wang J, Liu S, et al. High expression of hnRNPA1 promotes cell invasion by inducing EMT in gastric cancer. Oncol Rep. 2018;39(4):1693–701.PubMedPubMedCentral Chen Y, Liu J, Wang W, Xiang L, Wang J, Liu S, et al. High expression of hnRNPA1 promotes cell invasion by inducing EMT in gastric cancer. Oncol Rep. 2018;39(4):1693–701.PubMedPubMedCentral
71.
go back to reference Wen Z, Lian L, Ding H, Hu Y, Xiao Z, Xiong K, et al. LncRNA ANCR promotes hepatocellular carcinoma metastasis through upregulating HNRNPA1 expression. RNA Biol. 2020;17(3):381–94.PubMedPubMedCentralCrossRef Wen Z, Lian L, Ding H, Hu Y, Xiao Z, Xiong K, et al. LncRNA ANCR promotes hepatocellular carcinoma metastasis through upregulating HNRNPA1 expression. RNA Biol. 2020;17(3):381–94.PubMedPubMedCentralCrossRef
72.
go back to reference Lan Z, Yao X, Sun K, Li A, Liu S, Wang X. The interaction between lncRNA SNHG6 and hnRNPA1 contributes to the growth of colorectal cancer by enhancing aerobic glycolysis through the regulation of alternative splicing of PKM. Front Oncol. 2020;10:363.PubMedPubMedCentralCrossRef Lan Z, Yao X, Sun K, Li A, Liu S, Wang X. The interaction between lncRNA SNHG6 and hnRNPA1 contributes to the growth of colorectal cancer by enhancing aerobic glycolysis through the regulation of alternative splicing of PKM. Front Oncol. 2020;10:363.PubMedPubMedCentralCrossRef
73.
go back to reference Zhou B, Wang Y, Jiang J, Jiang H, Song J, Han T, et al. The long noncoding RNA colon cancer-associated transcript-1/miR-490 axis regulates gastric cancer cell migration by targeting hnRNPA1. IUBMB Life. 2016;68(3):201–10.PubMedCrossRef Zhou B, Wang Y, Jiang J, Jiang H, Song J, Han T, et al. The long noncoding RNA colon cancer-associated transcript-1/miR-490 axis regulates gastric cancer cell migration by targeting hnRNPA1. IUBMB Life. 2016;68(3):201–10.PubMedCrossRef
74.
go back to reference Chen F, Sha S, Wang S, Shi H, Dong L, Liu D, et al. RP11–81H3.2 promotes gastric cancer progression through miR-339-HNRNPA1 interaction network. Cancer Med. 2020;9(7):2524–34.PubMedPubMedCentralCrossRef Chen F, Sha S, Wang S, Shi H, Dong L, Liu D, et al. RP11–81H3.2 promotes gastric cancer progression through miR-339-HNRNPA1 interaction network. Cancer Med. 2020;9(7):2524–34.PubMedPubMedCentralCrossRef
75.
go back to reference Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 2020;19(1):43.PubMedPubMedCentralCrossRef Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 2020;19(1):43.PubMedPubMedCentralCrossRef
76.
go back to reference Catenacci D, Cervantes G, Yala S, Nelson E, El-Hashani E, Kanteti R, et al. RON (MST1R) is a novel prognostic marker and therapeutic target for gastroesophageal adenocarcinoma. Cancer Biol Ther. 2011;12(1):9–46.PubMedPubMedCentralCrossRef Catenacci D, Cervantes G, Yala S, Nelson E, El-Hashani E, Kanteti R, et al. RON (MST1R) is a novel prognostic marker and therapeutic target for gastroesophageal adenocarcinoma. Cancer Biol Ther. 2011;12(1):9–46.PubMedPubMedCentralCrossRef
77.
go back to reference Mereiter S, Magalhães A, Adamczyk B, Jin C, Almeida A, Drici L, et al. Glycomic analysis of gastric carcinoma cells discloses glycans as modulators of RON receptor tyrosine kinase activation in cancer. Biochim Biophys Acta. 2016;1860(8):1795–808.PubMedCrossRef Mereiter S, Magalhães A, Adamczyk B, Jin C, Almeida A, Drici L, et al. Glycomic analysis of gastric carcinoma cells discloses glycans as modulators of RON receptor tyrosine kinase activation in cancer. Biochim Biophys Acta. 2016;1860(8):1795–808.PubMedCrossRef
78.
go back to reference Diniz F, Lamas S, Osório H, Aguiar P, Freitas D, Gärtner F, et al. Nanoparticles targeting Sialyl-Tn for efficient tyrosine kinase inhibitor delivery in gastric cancer. Acta Biomater. 2023;170:142–54.PubMedCrossRef Diniz F, Lamas S, Osório H, Aguiar P, Freitas D, Gärtner F, et al. Nanoparticles targeting Sialyl-Tn for efficient tyrosine kinase inhibitor delivery in gastric cancer. Acta Biomater. 2023;170:142–54.PubMedCrossRef
79.
go back to reference Huang Y, Liu Y, Zheng C, Shen C. Investigation of cross-contamination and misidentification of 278 widely used tumor cell lines. PLoS One. 2017;12(1):e0170384.PubMedPubMedCentralCrossRef Huang Y, Liu Y, Zheng C, Shen C. Investigation of cross-contamination and misidentification of 278 widely used tumor cell lines. PLoS One. 2017;12(1):e0170384.PubMedPubMedCentralCrossRef
80.
go back to reference Liu X, Zhou Y, Lou Y, Zhong H. Knockdown of HNRNPA1 inhibits lung adenocarcinoma cell proliferation through cell cycle arrest at G0/G1 phase. Gene. 2016;576:791–7.PubMedCrossRef Liu X, Zhou Y, Lou Y, Zhong H. Knockdown of HNRNPA1 inhibits lung adenocarcinoma cell proliferation through cell cycle arrest at G0/G1 phase. Gene. 2016;576:791–7.PubMedCrossRef
81.
go back to reference Liu W, Gou H, Wang X, Li X, Hu X, Su H, et al. TTPAL promotes gastric tumorigenesis by directly targeting NNMT to activate PI3K/AKT signaling. Oncogene. 2021;40(49):6666–79.PubMedPubMedCentralCrossRef Liu W, Gou H, Wang X, Li X, Hu X, Su H, et al. TTPAL promotes gastric tumorigenesis by directly targeting NNMT to activate PI3K/AKT signaling. Oncogene. 2021;40(49):6666–79.PubMedPubMedCentralCrossRef
82.
go back to reference Zhang F, Li K, Pan M, Li W, Wu J, Li M, et al. miR-589 promotes gastric cancer aggressiveness by a LIFR-PI3K/AKT-c-Jun regulatory feedback loop. J Exp Clin Cancer Res. 2018;37(1):152.PubMedPubMedCentralCrossRef Zhang F, Li K, Pan M, Li W, Wu J, Li M, et al. miR-589 promotes gastric cancer aggressiveness by a LIFR-PI3K/AKT-c-Jun regulatory feedback loop. J Exp Clin Cancer Res. 2018;37(1):152.PubMedPubMedCentralCrossRef
83.
go back to reference Zhang X, Yang S, Kang Z, Ru W, Shen X, Li M, et al. circMEF2D negatively regulated by HNRNPA1 inhibits proliferation and differentiation of myoblasts via miR-486-PI3K/AKT Axis. J Agric Food Chem. 2022;70(26):8145–63.PubMedCrossRef Zhang X, Yang S, Kang Z, Ru W, Shen X, Li M, et al. circMEF2D negatively regulated by HNRNPA1 inhibits proliferation and differentiation of myoblasts via miR-486-PI3K/AKT Axis. J Agric Food Chem. 2022;70(26):8145–63.PubMedCrossRef
84.
go back to reference Perry J, He X, Sugimura R, Grindley J, Haug J, Ding S, et al. Cooperation between both Wnt/{beta}-catenin and PTEN/PI3K/Akt signaling promotes primitive hematopoietic stem cell self-renewal and expansion. Genes Dev. 2011;25(18):1928–42.PubMedPubMedCentralCrossRef Perry J, He X, Sugimura R, Grindley J, Haug J, Ding S, et al. Cooperation between both Wnt/{beta}-catenin and PTEN/PI3K/Akt signaling promotes primitive hematopoietic stem cell self-renewal and expansion. Genes Dev. 2011;25(18):1928–42.PubMedPubMedCentralCrossRef
85.
go back to reference Liu Y, Wu K, Huang J, Liu Y, Wang X, Meng Z, et al. The PTEN/PI3K/Akt and Wnt/β-catenin signaling pathways are involved in the inhibitory effect of resveratrol on human colon cancer cell proliferation. Int J Oncol. 2014;45(1):104–12.PubMedCrossRef Liu Y, Wu K, Huang J, Liu Y, Wang X, Meng Z, et al. The PTEN/PI3K/Akt and Wnt/β-catenin signaling pathways are involved in the inhibitory effect of resveratrol on human colon cancer cell proliferation. Int J Oncol. 2014;45(1):104–12.PubMedCrossRef
86.
go back to reference Hollander M, Blumenthal G, Dennis P. PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nat Rev Cancer. 2011;11(4):289–301.PubMedPubMedCentralCrossRef Hollander M, Blumenthal G, Dennis P. PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nat Rev Cancer. 2011;11(4):289–301.PubMedPubMedCentralCrossRef
87.
go back to reference Persad S, Troussard A, McPhee T, Mulholland D, Dedhar S. Tumor suppressor PTEN inhibits nuclear accumulation of beta-catenin and T cell/lymphoid enhancer factor 1-mediated transcriptional activation. J Cell Biol. 2001;153(6):1161–74.PubMedPubMedCentralCrossRef Persad S, Troussard A, McPhee T, Mulholland D, Dedhar S. Tumor suppressor PTEN inhibits nuclear accumulation of beta-catenin and T cell/lymphoid enhancer factor 1-mediated transcriptional activation. J Cell Biol. 2001;153(6):1161–74.PubMedPubMedCentralCrossRef
88.
go back to reference Reyes R, Wani N, Ghoshal K, Jacob S, Motiwala T. Sorafenib and 2-deoxyglucose synergistically inhibit proliferation of both sorafenib-sensitive and -resistant HCC Cells by inhibiting ATP production. Gene Expr. 2017;17(2):129–40.PubMedCrossRef Reyes R, Wani N, Ghoshal K, Jacob S, Motiwala T. Sorafenib and 2-deoxyglucose synergistically inhibit proliferation of both sorafenib-sensitive and -resistant HCC Cells by inhibiting ATP production. Gene Expr. 2017;17(2):129–40.PubMedCrossRef
89.
go back to reference Bao B, Azmi A, Ali S, Zaiem F, Sarkar F. Metformin may function as anti-cancer agent via targeting cancer stem cells: the potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers. Ann Transl Med. 2014;2(6):59.PubMedPubMedCentral Bao B, Azmi A, Ali S, Zaiem F, Sarkar F. Metformin may function as anti-cancer agent via targeting cancer stem cells: the potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers. Ann Transl Med. 2014;2(6):59.PubMedPubMedCentral
Metadata
Title
MAGOH promotes gastric cancer progression via hnRNPA1 expression inhibition-mediated RONΔ160/PI3K/AKT signaling pathway activation
Authors
Shanshan Yu
Cheng Chen
Ming Chen
Jinxiao Liang
Kecheng Jiang
Bin Lou
Jun Lu
Xiaohua Zhu
Donghui Zhou
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2024
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-024-02946-8

Other articles of this Issue 1/2024

Journal of Experimental & Clinical Cancer Research 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine