AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (5.1 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Full Length Article | Open Access

Multi-omics integration reveals potential stage-specific druggable targets in T-cell acute lymphoblastic leukemia

Zijun Yana,bJie XiaeZiyang Caoa,bHongyang Zhanga,bJinxia Wanga,bTienan Fenga,dYi Shuc( )Lin Zoua,b,c( )
Clinical Research Unit, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
Center for Clinical Molecular Laboratory Medicine of Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
Bioinformatics and BioMedical Bigdata Mining Laboratory, School of Big Health, Guizhou Medical University, Guiyang, Guizhou 554300, China

Peer review under responsibility of Chongqing Medical University.

Show Author Information

Abstract

T-cell acute lymphoblastic leukemia (T-ALL), a heterogeneous hematological malignancy, is caused by the developmental arrest of normal T-cell progenitors. The development of targeted therapeutic regimens is impeded by poor knowledge of the stage-specific aberrances in this disease. In this study, we performed multi-omics integration analysis, which included mRNA expression, chromatin accessibility, and gene-dependency database analyses, to identify potential stage-specific druggable targets and repositioned drugs for this disease. This multi-omics integration helped identify 29 potential pathological genes for T-ALL. These genes exhibited tissue-specific expression profiles and were enriched in the cell cycle, hematopoietic stem cell differentiation, and the AMPK signaling pathway. Of these, four known druggable targets (CDK6, TUBA1A, TUBB, and TYMS) showed dysregulated and stage-specific expression in malignant T cells and may serve as stage-specific targets in T-ALL. The TUBA1A expression level was higher in the early T cell precursor (ETP)-ALL cells, while TUBB and TYMS were mainly highly expressed in malignant T cells arrested at the CD4 and CD8 double-positive or single-positive stage. CDK6 exhibited a U-shaped expression pattern in malignant T cells along the naïve to maturation stages. Furthermore, mebendazole and gemcitabine, which target TUBA1A and TYMS, respectively, exerted stage-specific inhibitory effects on T-ALL cell lines, indicating their potential stage-specific antileukemic role in T-ALL. Collectively, our findings might aid in identifying potential stage-specific druggable targets and are promising for achieving more precise therapeutic strategies for T-ALL.

References

1

Pui CH, Robison LL, Look AT. Acute lymphoblastic leukaemia. Lancet. 2008;371(9617):1030–1043.

2

Tang J, Yu J, Cai J, et al. Prognostic factors for CNS control in children with acute lymphoblastic leukemia treated without cranial irradiation. Blood. 2021;138(4):331–343.

3

Goldstone AH, Richards SM, Lazarus HM, et al. In adults with standard-risk acute lymphoblastic leukemia, the greatest benefit is achieved from a matched sibling allogeneic transplantation in first complete remission, and an autologous transplantation is less effective than conventional consolidation/maintenance chemotherapy in all patients: final results of the International ALL Trial (MRC UKALL XII/ECOG E2993). Blood. 2008;111(4):1827–1833.

4

Kozlowski P, Åström M, Ahlberg L, et al. High relapse rate of T cell acute lymphoblastic leukemia in adults treated with Hyper-CVAD chemotherapy in Sweden. Eur J Haematol. 2014;92(5):377–381.

5

Bene MC, Castoldi G, Knapp W, et al. Proposals for the immunological classification of acute leukemias. European group for the immunological characterization of leukemias (EGIL). Leukemia. 1995;9(10):1783–1786.

6

Liu Y, Easton J, Shao Y, et al. The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat Genet. 2017;49(8):1211–1218.

7

Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol. 2009;10(2):147–156.

8

Weng AP, Ferrando AA, Lee W, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science. 2004;306(5694):269–271.

9

Graux C, Cools J, Melotte C, et al. Fusion of NUP214 to ABL1 on amplified episomes in T-cell acute lymphoblastic leukemia. Nat Genet. 2004;36(10):1084–1089.

10

Flex E, Petrangeli V, Stella L, et al. Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia. J Exp Med. 2008;205(4):751–758.

11

Shochat C, Tal N, Bandapalli OR, et al. Gain-of-function mutations in interleukin-7 receptor-alpha (IL7R) in childhood acute lymphoblastic leukemias. J Exp Med. 2011;208(5):901–908.

12

Zenatti PP, Ribeiro D, Li W, et al. Oncogenic IL7R gain-of-function mutations in childhood T-cell acute lymphoblastic leukemia. Nat Genet. 2011;43(10):932–939.

13

Knoechel B, Roderick JE, Williamson KE, et al. An epigenetic mechanism of resistance to targeted therapy in T cell acute lymphoblastic leukemia. Nat Genet. 2014;46(4):364–370.

14

Ishio T, Kumar S, Shimono J, et al. Genome-wide CRISPR screen identifies CDK6 as a therapeutic target in adult T-cell leukemia/lymphoma. Blood. 2022;139(10):1541–1556.

15

Rodriguez S, Abundis C, Boccalatte F, et al. Therapeutic targeting of the E3 ubiquitin ligase SKP2 in T-ALL. Leukemia. 2020;34(5):1241–1252.

16

Subramaniam PS, Whye DW, Efimenko E, et al. Targeting nonclassical oncogenes for therapy in T-ALL. Cancer Cell. 2012;21(4):459–472.

17

Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol. 2017;18(1):83.

18

Wang Y, Yang Y, Chen S, et al. DeepDRK: a deep learning framework for drug repurposing through kernel-based multi-omics integration. Briefings Bioinf. 2021;22(5):bbab048.

19

Aydin B, Beklen H, Arga KY, et al. Epigenomic and transcriptomic landscaping unraveled candidate repositioned therapeutics for non-functioning pituitary neuroendocrine tumors. J Endocrinol Invest. 2023;46(4):727–747.

20

Tang F, Barbacioru C, Wang Y, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. 2009;6(5):377–382.

21

Homminga I, Pieters R, Langerak AW, et al. Integrated transcript and genome analyses reveal NKX2-1 and MEF2C as potential oncogenes in T cell acute lymphoblastic leukemia. Cancer Cell. 2011;19(4):484–497.

22

Cramer-Morales K, Nieborowska-Skorska M, Scheibner K, et al. Personalized synthetic lethality induced by targeting RAD52 in leukemias identified by gene mutation and expression profile. Blood. 2013;122(7):1293–1304.

23

Erarslan-Uysal B, Kunz JB, Rausch T, et al. Chromatin accessibility landscape of pediatric T-lymphoblastic leukemia and human T-cell precursors. EMBO Mol Med. 2020;12(9):e12104.

24

Shu Y, Wang Y, Lv WQ, et al. ARRB1-promoted NOTCH1 degradation is suppressed by OncomiR miR-223 in T-cell acute lymphoblastic leukemia. Cancer Res. 2020;80(5):988–998.

25

Van Thillo Q, De Bie J, Seneviratne JA, et al. Oncogenic cooperation between TCF7-SPI1 and NRAS(G12D) requires β-catenin activity to drive T-cell acute lymphoblastic leukemia. Nat Commun. 2021;12(1):4164.

26

Park JE, Botting RA, Domínguez Conde C, et al. A cell atlas of human thymic development defines T cell repertoire formation. Science. 2020;367(6480):eaay3224.

27

Ritchie ME, Phipson B, Wu D, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.

28

Tsherniak A, Vazquez F, Montgomery PG, et al. Defining a cancer dependency map. Cell. 2017;170(3):564–576.e16.

29

Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607-D613.

30

Warde-Farley D, Donaldson SL, Comes O, et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010;38(Web Server issue):W214-W220.

31

Yu G, Wang LG, Han Y, et al. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16(5):284–287.

32

Ni Q, Su X, Chen J, et al. Prediction of metabolic gene biomarkers for neurodegenerative disease by an integrated network-based approach. BioMed Res Int. 2015;2015:432012.

33

Becker KG, Barnes KC, Bright TJ, et al. The genetic association database. Nat Genet. 2004;36(5):431–432.

34

Alexeyenko A, Schmitt T, Tjärnberg A, et al. Comparative interactomics with funcoup 2.0. Nucleic Acids Res. 2012;40(Database issue):D821-D828.

35

Law V, Knox C, Djoumbou Y, et al. DrugBank 4.0: shedding new light on drug metabolism. Nucleic Acids Res. 2014;42(Database issue):D1091-D1097.

36

Ghandi M, Huang FW, Jané-Valbuena J, et al. Next-generation characterization of the cancer cell line encyclopedia. Nature. 2019;569(7757):503–508.

37

Reinhold WC, Sunshine M, Liu H, et al. CellMiner: a web-based suite of genomic and pharmacologic tools to explore transcript and drug patterns in the NCI-60 cell line set. Cancer Res. 2012;72(14):3499–3511.

38

Burley SK, Berman HM, Christie C, et al. RCSB Protein Data Bank: sustaining a living digital data resource that enables breakthroughs in scientific research and biomedical education. Protein Sci. 2018;27(1):316–330.

39
Biovia DS. Discovery Studio Modeling Environment, Release2017. San Diego: DassaultSystèmes; 2016.
40

Sterling T, Irwin JJ. ZINC 15: ligand discovery for everyone. J Chem Inf Model. 2015;55(11):2324–2337.

41

Kim S, Thiessen PA, Bolton EE, et al. PubChem substance and compound databases. Nucleic Acids Res. 2016;44(D1):D1202-D1213.

42

Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010;31(2):455–461.

43

Yao H, Xie S, Ma X, et al. Identification of a potent oridonin analogue for treatment of triple-negative breast cancer. J Med Chem. 2020;63(15):8157–8178.

44

Butler A, Hoffman P, Smibert P, et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36(5):411–420.

45

Bray NL, Pimentel H, Melsted P, et al. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol. 2016;34(5):525–527.

46

de Bock CE, Demeyer S, Degryse S, et al. HOXA9 cooperates with activated JAK/STAT signaling to drive leukemia development. Cancer Discov. 2018;8(5):616–631.

47

Zhang J, Ding L, Holmfeldt L, et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature. 2012;481(7380):157–163.

48

Kraszewska MD, Dawidowska M, Szczepański T, et al. T-cell acute lymphoblastic leukaemia: recent molecular biology findings. Br J Haematol. 2012;156(3):303–315.

49

Greenberg JM, Gonzalez-Sarmiento R, Arthur DC, et al. Immunophenotypic and cytogenetic analysis of Molt-3 and Molt-4:human T-lymphoid cell lines with rearrangement of chromosome 7. Blood. 1988;72(5):1755–1760.

50

De Bie J, Demeyer S, Alberti-Servera L, et al. Single-cell sequencing reveals the origin and the order of mutation acquisition in T-cell acute lymphoblastic leukemia. Leukemia. 2018;32(6):1358–1369.

51

Karczewski KJ, Snyder MP. Integrative omics for health and disease. Nat Rev Genet. 2018;19(5):299–310.

52

Aifantis I, Raetz E, Buonamici S. Molecular pathogenesis of T-cell leukaemia and lymphoma. Nat Rev Immunol. 2008;8(5):380–390.

53

Hammond JW, Cai D, Verhey KJ. Tubulin modifications and their cellular functions. Curr Opin Cell Biol. 2008;20(1):71–76.

54

Mandelkow E, Mandelkow EM. Microtubules and microtubule-associated proteins. Curr Opin Cell Biol. 1995;7(1):72–81.

55

Duijf PHG, Schultz N, Benezra R. Cancer cells preferentially lose small chromosomes. Int J Cancer. 2013;132(10):2316–2326.

56

Nami B, Wang Z. Genetics and expression profile of the tubulin gene superfamily in breast cancer subtypes and its relation to taxane resistance. Cancers. 2018;10(8):274.

57

Wang D, Jiao Z, Ji Y, et al. Elevated TUBA1A might indicate the clinical outcomes of patients with gastric cancer, being associated with the infiltration of macrophages in the tumor immune microenvironment. J Gastrointestin Liver Dis. 2020;29(4):509–522.

58

Boggs AE, Vitolo MI, Whipple RA, et al. α-Tubulin acetylation elevated in metastatic and basal-like breast cancer cells promotes microtentacle formation, adhesion, and invasive migration. Cancer Res. 2015;75(1):203–215.

59

Wolfe MS, Wershing JM. Mebendazole. Treatment of trichuriasis and ascariasis in Bahamian children. JAMA. 1974;230(10):1408–1411.

60

Bai RY, Staedtke V, Rudin CM, et al. Effective treatment of diverse medulloblastoma models with mebendazole and its impact on tumor angiogenesis. Neuro Oncol. 2015;17(4):545–554.

61

Bai RY, Staedtke V, Aprhys CM, et al. Antiparasitic mebendazole shows survival benefit in 2 preclinical models of glioblastoma multiforme. Neuro Oncol. 2011;13(9):974–982.

62

Ntavatzikos A, Spathis A, Patapis P, et al. TYMS/KRAS/BRAF molecular profiling predicts survival following adjuvant chemotherapy in colorectal cancer. World J Gastrointest Oncol. 2019;11(7):551–566.

63

Denechaud PD, Fajas L, Giralt A. E2F1, a novel regulator of metabolism. Front Endocrinol(Lausanne). 2017;8:311.

64

Zhang F, Ye J, Guo W, et al. TYMS-TM4SF4 axis promotes the progression of colorectal cancer by EMT and upregulating stem cell marker. Am J Cancer Res. 2022;12(3):1009–1026.

65

Lu Y, Zhuo C, Cui B, et al. TYMS serves as a prognostic indicator to predict the lymph node metastasis in Chinese patients with colorectal cancer. Clin Biochem. 2013;46(15):1478–1483.

66

Taddia L, D’Arca D, Ferrari S, et al. Inside the biochemical pathways of thymidylate synthase perturbed by anticancer drugs: novel strategies to overcome cancer chemoresistance. Drug Resist Updat. 2015;23:20–54.

67

Fu F, Zhang Y, Gao Z, et al. Development and validation of a five-gene model to predict postoperative brain metastasis in operable lung adenocarcinoma. Int J Cancer. 2020;147(2):584–592.

68

Plunkett W, Huang P, Gandhi V. Preclinical characteristics of gemcitabine. Anti Cancer Drugs. 1995;6(Suppl 6):7–13.

69

Malumbres M, Barbacid M. Mammalian cyclin-dependent kinases. Trends Biochem Sci. 2005;30(11):630–641.

70

Hu MG, Deshpande A, Schlichting N, et al. CDK6 kinase activity is required for thymocyte development. Blood. 2011;117(23):6120–6131.

71

Bride KL, Hu H, Tikhonova A, et al. Rational drug combinations with CDK4/6 inhibitors in acute lymphoblastic leukemia. Haematologica. 2022;107(8):1746–1757.

72

Jena N, Sheng J, Hu JK, et al. CDK6-mediated repression of CD25 is required for induction and maintenance of Notch1-induced T-cell acute lymphoblastic leukemia. Leukemia. 2016;30(5):1033–1043.

73

Mesquita B, Veiga I, Pereira D, et al. No significant role for beta tubulin mutations and mismatch repair defects in ovarian cancer resistance to paclitaxel/cisplatin. BMC Cancer. 2005;5:101.

74

Liu Y, Ma S, Ma Q, et al. Silencing LINC00665 inhibits cutaneous melanoma in vitro progression and induces apoptosis via the miR-339-3p/TUBB. J Clin Lab Anal. 2022;36(9):e24630.

Genes & Diseases
Article number: 100949
Cite this article:
Yan Z, Xia J, Cao Z, et al. Multi-omics integration reveals potential stage-specific druggable targets in T-cell acute lymphoblastic leukemia. Genes & Diseases, 2024, 11(5): 100949. https://doi.org/10.1016/j.gendis.2023.03.022

224

Views

2

Downloads

0

Crossref

0

Web of Science

0

Scopus

0

CSCD

Altmetrics

Received: 20 October 2022
Accepted: 11 March 2023
Published: 26 April 2023
© 2023 The Authors.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return