LATS1 Promotes B-ALL Tumorigenesis by Regulating YAP1 Phosphorylation and Subcellular Localization

Feng Zhang1(), Mohammed Awal Issah2, Hai-ying Fu3, Hua-rong Zhou1, Ting-bo Liu1, Jian-zhen Shen1()

PDF
Current Medical Science ›› 2024, Vol. 44 ›› Issue (1) : 81-92. DOI: 10.1007/s11596-023-2821-7
Original Article

LATS1 Promotes B-ALL Tumorigenesis by Regulating YAP1 Phosphorylation and Subcellular Localization

  • Feng Zhang1(), Mohammed Awal Issah2, Hai-ying Fu3, Hua-rong Zhou1, Ting-bo Liu1, Jian-zhen Shen1()
Author information +
History +

Abstract

Abstract
Objective

YAP1 plays a dual role as an oncogene and tumor suppressor gene in several tumors; differentiating between these roles may depend on the YAP1 phosphorylation pattern. The specific function of YAP1 in B cell acute lymphoblastic leukemia (B-ALL), however, is currently unclear. Thus, in the present study, the role of YAP1 in B-ALL was investigated using relevant cell lines and patient datasets.

Methods

The effects of shRNA-mediated knockdown on YAP1 and LATS1 levels in the NALM6 and MOLT-4 cell lines were examined using Western blotting, quantitative real-time polymerase chain reaction, flow cytometry, immunostaining, and nude mouse subcutaneous tumorigenesis experiments. Gene expression levels of Hippo pathway-related molecules before and after verteporfin (VP) treatment were compared using RNA-Seq to identify significant Hippo pathway-related genes in NALM6 cells.

Results

Patients with ALL showing high YAP1 expression and low YAP1-Ser127 phosphorylation levels had worse prognoses than those with low YAP1 protein expression and high YAP1-Ser127 phosphorylation levels. YAP1-Ser127 phosphorylation levels were lower in NALM6 cells than in MOLT-4 and control cells; YAP1 was distributed in the nuclei in NALM6 cells. Knockdown of YAP1 inhibited MOLT-4 and NALM6 cell proliferation and arrested the NALM6 cell cycle in the G0/G1 phase. Before and after VP treatment, the expression of the upstream gene LATS1 was upregulated; its overexpression promoted YAP1-Ser127 phosphorylation. Further, YAP1 was distributed in the plasma.

Conclusion

LATS1 may downregulate YAP1-Ser127 phosphorylation and maintain B-ALL cell function; thus, VP, which targets this axis, may serve as a new therapeutic method for improving the outcomes for B-ALL patients.

Keywords

acute lymphoblastic leukemia / large tumor suppressor kinase 1 / phosphorylation / RNA-Seq / Yes1-associated protein

Cite this article

Download citation ▾
Feng Zhang, Mohammed Awal Issah, Hai-ying Fu, Hua-rong Zhou, Ting-bo Liu, Jian-zhen Shen. LATS1 Promotes B-ALL Tumorigenesis by Regulating YAP1 Phosphorylation and Subcellular Localization. Current Medical Science, 2024, 44(1): 81‒92 https://doi.org/10.1007/s11596-023-2821-7

References

[1]
Oriol A, Vives S, Hernández-Rivas JM, et al. Outcome after relapse of acute lymphoblastic leukemia in adult patients included in four consecutive risk-adapted trials by the PETHEMA Study Group. Haematologica, 2010,95(4):589–596
[2]
Zhang M, Huang H. How to combine the two landmark treatment methods-allogeneic hematopoietic stem cell transplantation and chimeric antigen receptor T cell therapy together to cure high-risk B cell acute lymphoblastic leukemia? Front Immunol, 2020,11: 611710
[3]
Hayden PJ, Roddie C, Bader P, et al. Management of adults and children receiving CAR T-cell therapy: 2021 best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association (EHA). Ann Oncol, 2022,33(3):259–275
[4]
Tan X, Wang XQ, Zhang C, et al. Donor-derived CD19 CAR-T Cells versus Chemotherapy Plus Donor Lymphocyte Infusion for Treatment of Recurrent CD19-positive B-ALL After Allogeneic Hematopoietic Stem Cell Transplantation. Curr Med Sci, 2023,43(4):733–740
[5]
Wudhikarn K, King AC, Geyer MB, et al. Outcomes of relapsed B-cell acute lymphoblastic leukemia after sequential treatment with Blinatumomab and inotuzumab. Blood Adv, 2022,6(5):1432–1443
[6]
Cunningham R, Hansen CG. The Hippo pathway in cancer: YAP/TAZ and TEAD as therapeutic targets in cancer. Clin Sci (Lond), 2022,136(3):197–222
[7]
Fu V, Plouffe SW, Guan KL. The Hippo pathway in organ development, homeostasis, and regeneration. Curr Opin Cell Biol, 2017,49:99–107
[8]
Bai X, Huang L, Niu L, et al. Mst1 positively regulates B-cell receptor signaling via CD19 transcriptional levels. Blood Adv, 2016,1(3):219–230
[9]
Alsufyani F, Mattoo H, Zhou D, et al. The Mst1 kinase is required for follicular B cell homing and B-1 B cell development. Front Immunol, 2018,9:2393
[10]
Zhou X, Chen N, Xu H, et al. Regulation of Hippo-YAP signaling by insulin-like growth factor-1 receptor in the tumorigenesis of diffuse large B-cell lymphoma. J Hematol Oncol, 2020,13(1):77
[11]
Wang Z, Ran X, Qian S, et al. GPNMB promotes the progression of diffuse large B cell lymphoma via YAP1-mediated activation of the Wnt/β-catenin signaling pathway. Arch Biochem Biophys, 2021,710:108998
[12]
Fan S, Price T, Huang W, et al. PINK1-dependent mitophagy regulates the migration and homing of multiple myeloma cells via the MOB1B-mediated hippo-YAP/TAZ pathway. Adv Sci (Weinh), 2020,7(5): 1900860
[13]
Grieve S, Wajnberg G, Lees M, et al. TAZ functions as a tumor suppressor in multiple myeloma by downregulating MYC. Blood Adv, 2019,3(22):3613–3625
[14]
Donato E, Biagioni F, Bisso A, et al. YAP and TAZ are dispensable for physiological and malignant haematopoiesis. Leukemia, 2018,32(9):2037–2040
[15]
Moon S, Yeon Park S, Woo Park H. Regulation of the Hippo pathway in cancer biology. Cell Mol Life Sci, 2018,75(13):2303–2319
[16]
Machado-Neto JA, de Melo Campos P, Olalla Saad ST, et al. YAP1 expression in myelodysplastic syndromes and acute leukemias. Leuk Lymphoma, 2014,55(10):2413–2415
[17]
Cottini F, Hideshima T, Xu C, et al. Rescue of Hippo coactivator YAP1 triggers DNA damage-induced apoptosis in hematological cancers. Nat Med, 2014,20(6):599–606
[18]
Wei W, Huang S, Ling Q, et al. Homoharringtonine is synergistically lethal with BCL-2 inhibitor APG-2575 in acute myeloid leukemia. J Transl Med, 2022,20(1):299
[19]
Zhu B, Pan S, Liu J, et al. HIF-1α forms regulatory loop with YAP to coordinate hypoxia-induced adriamycin resistance in acute myeloid leukemia cells. Cell Biol Int, 2020,44(2):456–466
[20]
Chen M, Wang J, Yao SF, et al. Effect of YAP inhibition on human leukemia HL-60 cells. Int J Med Sci, 2017,14(9):902–910
[21]
Liu H, Du S, Lei T, et al. Multifaceted regulation and functions of YAP/TAZ in tumors. Oncol Rep, 2018,40(1):16–28
[22]
Barry ER, Morikawa T, Butler BL, et al. Restriction of intestinal stem cell expansion and the regenerative response by YAP. Nature, 2013,493(7430):106–110
[23]
Wu R, Yang H, Wan J, et al. Knockdown of the Hippo transducer YAP reduces proliferation and promotes apoptosis in the Jurkat leukemia cell. Mol Med Rep, 2018,18(6):5379–5388
[24]
Moriyama K, Hori T. BCR-ABL induces tyrosine phosphorylation of YAP leading to expression of survivin and cyclin D1 in chronic myeloid leukemia cells. Int J Hematol, 2019,110(5):591–598
[25]
Zhao B, Li L, Tumaneng K, et al. A coordinated phosphorylation by Lats and CK1 regulates YAP stability through SCF(beta-TrCP). Genes Dev, 2010,24(1):72–85
[26]
Liu CY, Zha ZY, Zhou X, et al. The hippo tumor pathway promotes TAZ degradation by phosphorylating a phosphodegron and recruiting the SCFta-TrCP E3 ligase. J Biol Chem, 2010,285(48):37159–37169
[27]
Kim MK, Jang JW, Bae SC. DNA binding partners of YAP/TAZ. BMB Rep, 2018,51(3):126–133
[28]
Lin KC, Park HW, Guan KL. Regulation of the hippo pathway transcription factor TEAD. Trends Biochem Sci, 2017,42(11):862–872
[29]
He L, Pratt H, Gao M, et al. YAP and TAZ are transcriptional co-activators of AP-1 proteins and STAT3 during breast cellular transformation. eLife, 2021,10:e67312
[30]
Wang Y, Chen H, Liu W, et al. MCM6 is a critical transcriptional target of YAP to promote gastric tumorigenesis and serves as a therapeutic target. Theranostics, 2022,12(15):6509–6526
[31]
Manning SA, Dent LG, Kondo S, et al. Dynamic fluctuations in subcellular localization of the hippo pathway effector Yorkie in vivo. Curr Biol, 2018,28(10):1651–1660.e4
[32]
Thomasy SM, Morgan JT, Wood JA, et al. Substratum stiffness and latrunculin B modulate the gene expression of the mechanotransducers YAP and TAZ in human trabecular meshwork cells. Exp Eye Res, 2013,113:66–73
[33]
Das A, Fischer RS, Pan D, et al. YAP nuclear localization in the absence of cell-cell contact is mediated by a filamentous actin-dependent, myosin II - and phospho-YAP-independent pathway during extracellular matrix mechanosensing. J Biol Chem, 2016,291(12):6096–6110
[34]
Fu M, Hu Y, Lan T, et al. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct Target Ther, 2022,7(1):376
[35]
Calses PC, Crawford JJ, Lill JR, et al. Hippo pathway in cancer: Aberrant regulation and therapeutic opportunities. Trends Cancer, 2019,5(5):297–307
[36]
Plouffe SW, Meng Z, Lin KC, et al. Characterization of hippo pathway components by gene inactivation. Mol Cell, 2016,64(5):993–1008
[37]
Pantziarka P, Verbaanderd C, Sukhatme V, et al. ReDO_DB: the repurposing drugs in oncology database. Ecancermedicalscience, 2018,6:886
[38]
Liu-Chittenden Y, Huang B, Shim JS, et al. Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev, 2012,26(12):1300–1305
[39]
Lin CH, Pelissier FA, Zhang H, et al. Microenvironment rigidity modulates responses to the HER2 receptor tyrosine kinase inhibitor lapatinib via YAP and TAZ transcription factors. Mol Biol Cell, 2015,26(22):3946–3953
[40]
Acedo P, Fernandes A, Zawacka-Pankau J. Activation of TAp73 and inhibition of TrxR by Verteporfin for improved cancer therapy in TP53 mutant pancreatic tumors. Future Sci OA, 2019,5(2):FSO366
[41]
Patel SH, Camargo FD, Yimlamai D. Hippo signaling in the liver regulates organ size, cell fate, and carcinogenesis. Gastroenterology, 2017,152(3):533–545
[42]
Guillermin O, Angelis N, Sidor CM, et al. Wnt and Src signals converge on YAP-TEAD to drive intestinal regeneration. EMBO J, 2021,40(13):e105770
[43]
Li Q, Sun Y, Jarugumilli GK, et al. Lats1/2 sustain intestinal stem cells and Wnt activation through TEAD-dependent and independent transcription. Cell Stem Cell, 2020,26(5):675–692.e8
PDF

Accesses

Citations

Detail

Sections
Recommended

/