CC BY-NC-ND 4.0 · Thromb Haemost 2020; 120(04): 671-686
DOI: 10.1055/s-0040-1708483
Cellular Signaling and Proteolysis
Georg Thieme Verlag KG Stuttgart · New York

N-Methyl-D-Aspartate Receptor Hypofunction in Meg-01 Cells Reveals a Role for Intracellular Calcium Homeostasis in Balancing Megakaryocytic-Erythroid Differentiation

1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
Taryn N. Green
1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
Martin Chopra
1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
Yohanes N. S. Nursalim
1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
Leandro Ladvanszky
1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
Nicholas Knowlton
1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
Cherie Blenkiron
1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
Raewyn C. Poulsen
2   Department of Medicine, School of Medicine, University of Auckland, Auckland, New Zealand
3   Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
Dean C. Singleton
4   Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
,
Stefan K. Bohlander
1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
,
1   Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
5   LabPlus Haematology, Auckland City Hospital, Auckland, New Zealand
› Author Affiliations
Funding This work was funded by Auckland Medical Research Foundation, Leukemia & Blood Cancer New Zealand, and Cancer Research Trust.
Further Information

Publication History

21 September 2019

04 February 2020

Publication Date:
14 April 2020 (online)

Abstract

The release of calcium ions (Ca2+) from the endoplasmic reticulum (ER) and related store-operated calcium entry (SOCE) regulate maturation of normal megakaryocytes. The N-methyl-D-aspartate (NMDA) receptor (NMDAR) provides an additional mechanism for Ca2+ influx in megakaryocytic cells, but its role remains unclear. We created a model of NMDAR hypofunction in Meg-01 cells using CRISPR-Cas9 mediated knockout of the GRIN1 gene, which encodes an obligate, GluN1 subunit of the NMDAR. We found that compared with unmodified Meg-01 cells, Meg-01-GRIN1 −/− cells underwent atypical differentiation biased toward erythropoiesis, associated with increased basal ER stress and cell death. Resting cytoplasmic Ca2+ levels were higher in Meg-01-GRIN1 −/− cells, but ER Ca2+ release and SOCE were lower after activation. Lysosome-related organelles accumulated including immature dense granules that may have contributed an alternative source of intracellular Ca2+. Microarray analysis revealed that Meg-01-GRIN1 −/− cells had deregulated expression of transcripts involved in Ca2+ metabolism, together with a shift in the pattern of hematopoietic transcription factors toward erythropoiesis. In keeping with the observed pro-cell death phenotype induced by GRIN1 deletion, memantine (NMDAR inhibitor) increased cytotoxic effects of cytarabine in unmodified Meg-01 cells. In conclusion, NMDARs comprise an integral component of the Ca2+ regulatory network in Meg-01 cells that help balance ER stress and megakaryocytic-erythroid differentiation. We also provide the first evidence that megakaryocytic NMDARs regulate biogenesis of lysosome-related organelles, including dense granules. Our results argue that intracellular Ca2+ homeostasis may be more important for normal megakaryocytic and erythroid differentiation than currently recognized; thus, modulation may offer therapeutic opportunities.

Authors' Contributions

M.L.K.Z designed the study. J.I.H., T.N.G., M.C., Y.N.S.N., L.L. and M.L.K.Z generated and analysed data. N.C., C.B., R.C.P., D.C.S., S.K.B. and M.L.K.Z provided methodology advice, supervision and mentorship. J.I.H., T.N.G. and M.L.K.Z wrote the manuscript. All authors edited the manuscript and approved the final version for submission.


Note

J.I.H. received PhD scholarship from Anne and Victoria Norman, supplemented by payments from the Marijana Kumerich Trust.


 
  • References

  • 1 Berridge MJ, Bootman MD, Roderick HL. Calcium signalling: dynamics, homeostasis and remodelling. Nat Rev Mol Cell Biol 2003; 04 (07) 517-529
  • 2 Di Buduo CA, Balduini A, Moccia F. Pathophysiological significance of store-operated calcium entry in megakaryocyte function: opening new paths for understanding the role of calcium in thrombopoiesis. Int J Mol Sci 2016; 17 (12) 17
  • 3 Berridge MJ. Inositol trisphosphate and calcium signalling mechanisms. Biochim Biophys Acta 2009; 1793 (06) 933-940
  • 4 Clapham DE. Calcium signaling. Cell 2007; 131 (06) 1047-1058
  • 5 Zaslavsky A, Chou ST, Schadler K. , et al. The calcineurin-NFAT pathway negatively regulates megakaryopoiesis. Blood 2013; 121 (16) 3205-3215
  • 6 Di Buduo CA, Moccia F, Battiston M. , et al. The importance of calcium in the regulation of megakaryocyte function. Haematologica 2014; 99 (04) 769-778
  • 7 Wang JX, Furukawa H. Dissecting diverse functions of NMDA receptors by structural biology. Curr Opin Struct Biol 2019; 54: 34-42
  • 8 Genever PG, Wilkinson DJ, Patton AJ. , et al. Expression of a functional N-methyl-D-aspartate-type glutamate receptor by bone marrow megakaryocytes. Blood 1999; 93 (09) 2876-2883
  • 9 Traynelis SF, Wollmuth LP, McBain CJ. , et al. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 2010; 62 (03) 405-496
  • 10 Kamal T, Green TN, Morel-Kopp MC. , et al. Inhibition of glutamate regulated calcium entry into leukemic megakaryoblasts reduces cell proliferation and supports differentiation. Cell Signal 2015; 27 (09) 1860-1872
  • 11 Kamal T, Green TN, Hearn JI. , et al. N-methyl-d-aspartate receptor mediated calcium influx supports in vitro differentiation of normal mouse megakaryocytes but proliferation of leukemic cell lines. Res Pract Thromb Haemost 2017; 02 (01) 125-138
  • 12 Hitchcock IS, Skerry TM, Howard MR, Genever PG. NMDA receptor-mediated regulation of human megakaryocytopoiesis. Blood 2003; 102 (04) 1254-1259
  • 13 Ogura M, Morishima Y, Ohno R. , et al. Establishment of a novel human megakaryoblastic leukemia cell line, MEG-01, with positive Philadelphia chromosome. Blood 1985; 66 (06) 1384-1392
  • 14 Ogura M, Morishima Y, Okumura M. , et al. Functional and morphological differentiation induction of a human megakaryoblastic leukemia cell line (MEG-01s) by phorbol diesters. Blood 1988; 72 (01) 49-60
  • 15 Trécul A, Morceau F, Gaigneaux A, Schnekenburger M, Dicato M, Diederich M. Valproic acid regulates erythro-megakaryocytic differentiation through the modulation of transcription factors and microRNA regulatory micro-networks. Biochem Pharmacol 2014; 92 (02) 299-311
  • 16 Chen W, Arroyo JD, Timmons JC, Possemato R, Hahn WC. Cancer-associated PP2A Aalpha subunits induce functional haploinsufficiency and tumorigenicity. Cancer Res 2005; 65 (18) 8183-8192
  • 17 Kalev-Zylinska ML, Hearn JI, Rong J. , et al. Altered N-methyl D-aspartate receptor subunit expression causes changes to the circadian clock and cell phenotype in osteoarthritic chondrocytes. Osteoarthritis Cartilage 2018; 26 (11) 1518-1530
  • 18 Kalev-Zylinska ML, Green TN, Morel-Kopp MC. , et al. N-methyl-D-aspartate receptors amplify activation and aggregation of human platelets. Thromb Res 2014; 133 (05) 837-847
  • 19 Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001; 25 (04) 402-408
  • 20 van Galen P, Kreso A, Mbong N. , et al. The unfolded protein response governs integrity of the haematopoietic stem-cell pool during stress. Nature 2014; 510 (7504): 268-272
  • 21 Lasham A, Herbert M, Coppieters 't Wallant N. , et al. A rapid and sensitive method to detect siRNA-mediated mRNA cleavage in vivo using 5′ RACE and a molecular beacon probe. Nucleic Acids Res 2010; 38 (03) e19
  • 22 Singleton DC, Rouhi P, Zois CE. , et al. Hypoxic regulation of RIOK3 is a major mechanism for cancer cell invasion and metastasis. Oncogene 2015; 34 (36) 4713-4722
  • 23 Irizarry RA, Hobbs B, Collin F. , et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 2003; 04 (02) 249-264
  • 24 Ambrosio AL, Boyle JA, Di Pietro SM. Mechanism of platelet dense granule biogenesis: study of cargo transport and function of Rab32 and Rab38 in a model system. Blood 2012; 120 (19) 4072-4081
  • 25 Pérez-Riesgo E, Gutiérrez LG, Ubierna D. , et al. Transcriptomic analysis of calcium remodeling in colorectal cancer. Int J Mol Sci 2017; 18 (05) 18
  • 26 Ramanathan G, Mannhalter C. Increased expression of transient receptor potential canonical 6 (TRPC6) in differentiating human megakaryocytes. Cell Biol Int 2016; 40 (02) 223-231
  • 27 Kaestner L, Wang X, Hertz L, Bernhardt I. Voltage-activated ion channels in non-excitable cells-a viewpoint regarding their physiological justification. Front Physiol 2018; 09: 450
  • 28 Zhao P, Xiao X, Kim AS. , et al. c-Jun inhibits thapsigargin-induced ER stress through up-regulation of DSCR1/Adapt78. Exp Biol Med (Maywood) 2008; 233 (10) 1289-1300
  • 29 Isakari Y, Sogo S, Ishida T. , et al. Gene expression analysis during platelet-like particle production in phorbol myristate acetate-treated MEG-01 cells. Biol Pharm Bull 2009; 32 (03) 354-358
  • 30 Brignall R, Cauchy P, Bevington SL. , et al. Integration of kinase and calcium signaling at the level of chromatin underlies inducible gene activation in T cells. J Immunol 2017; 199 (08) 2652-2667
  • 31 Kinney MA, Vo LT, Frame JM. , et al. A systems biology pipeline identifies regulatory networks for stem cell engineering. Nat Biotechnol 2019; 37 (07) 810-818
  • 32 Zhu F, Feng M, Sinha R, Seita J, Mori Y, Weissman IL. Screening for genes that regulate the differentiation of human megakaryocytic lineage cells. Proc Natl Acad Sci U S A 2018; 115 (40) E9308-E9316
  • 33 Kuvardina ON, Herglotz J, Kolodziej S. , et al. RUNX1 represses the erythroid gene expression program during megakaryocytic differentiation. Blood 2015; 125 (23) 3570-3579
  • 34 Frontelo P, Manwani D, Galdass M. , et al. Novel role for EKLF in megakaryocyte lineage commitment. Blood 2007; 110 (12) 3871-3880
  • 35 Cohen S, Greenberg ME. Communication between the synapse and the nucleus in neuronal development, plasticity, and disease. Annu Rev Cell Dev Biol 2008; 24: 183-209
  • 36 Albarrán L, Lopez JJ, Dionisio N, Smani T, Salido GM, Rosado JA. Transient receptor potential ankyrin-1 (TRPA1) modulates store-operated Ca(2+) entry by regulation of STIM1-Orai1 association. Biochim Biophys Acta 2013; 1833 (12) 3025-3034
  • 37 Chen JL, Ping YH, Tseng MJ. , et al. Notch1-promoted TRPA1 expression in erythroleukemic cells suppresses erythroid but enhances megakaryocyte differentiation. Sci Rep 2017; 07: 42883
  • 38 Kobuke K, Oki K, Gomez-Sanchez CE. , et al. Calneuron 1 increased Ca(2+) in the endoplasmic reticulum and aldosterone production in aldosterone-producing adenoma. Hypertension 2018; 71 (01) 125-133
  • 39 Kim HJ, Soyombo AA, Tjon-Kon-Sang S, So I, Muallem S. The Ca(2+) channel TRPML3 regulates membrane trafficking and autophagy. Traffic 2009; 10 (08) 1157-1167
  • 40 Pandey V, Chuang CC, Lewis AM. , et al. Recruitment of NAADP-sensitive acidic Ca2+ stores by glutamate. Biochem J 2009; 422 (03) 503-512
  • 41 Kook SY, Jeong H, Kang MJ. , et al. Crucial role of calbindin-D28k in the pathogenesis of Alzheimer's disease mouse model. Cell Death Differ 2014; 21 (10) 1575-1587
  • 42 Bellido T, Huening M, Raval-Pandya M, Manolagas SC, Christakos S. Calbindin-D28k is expressed in osteoblastic cells and suppresses their apoptosis by inhibiting caspase-3 activity. J Biol Chem 2000; 275 (34) 26328-26332
  • 43 Zunino R, Li Q, Rosé SD. , et al. Expression of scinderin in megakaryoblastic leukemia cells induces differentiation, maturation, and apoptosis with release of plateletlike particles and inhibits proliferation and tumorigenesis. Blood 2001; 98 (07) 2210-2219
  • 44 Polasek J. Platelet secretory granules or secretory lysosomes?. Platelets 2005; 16 (08) 500-501
  • 45 Ambrosio AL, Di Pietro SM. Storage pool diseases illuminate platelet dense granule biogenesis. Platelets 2017; 28 (02) 138-146
  • 46 Ambrosio AL, Boyle JA, Di Pietro SM. TPC2 mediates new mechanisms of platelet dense granule membrane dynamics through regulation of Ca2+ release. Mol Biol Cell 2015; 26 (18) 3263-3274
  • 47 Shivarov V, Ivanova M, Tiu RV. Mutated calreticulin retains structurally disordered C terminus that cannot bind Ca(2+): some mechanistic and therapeutic implications. Blood Cancer J 2014; 04: e185
  • 48 Nam AS, Kim KT, Chaligne R. , et al. Somatic mutations and cell identity linked by Genotyping of Transcriptomes. Nature 2019; 571 (7765): 355-360