CC BY 4.0 · Glob Med Genet 2024; 11(02): 187-195
DOI: 10.1055/s-0044-1787752
Original Article

Clinical and Molecular Characteristics of Megakaryocytes in Myelodysplastic Syndrome

Fangxiu Luo*
3   Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
,
Jialu Zhao*
1   Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
,
Yubao Chen
4   Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
,
Zhenping Peng
1   Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
,
Ran An
4   Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
,
Yeling Lu
1   Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
,
Jiaming Li
1   Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
2   Transfusion Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
› Author Affiliations
Funding This work was supported in part by grants from the National Natural Science Foundation of China (No.82070194 and No.8237022).

Abstract

Objective Myelodysplastic syndrome (MDS) is a malignant clonal disorder of hematopoietic stem cells which is characterized by morphologic dysplasia. However, the pathological characteristics of megakaryocytes (MKs) in MDS patients with gene mutation are not well established.

Methods Bone marrow MK specimens from 104 patients with primary MDS were evaluated, and all patients were distributed into two groups according to gene mutation associated with functional MKs. The morphologic and cellular characteristics of MKs and platelets were recorded and compared.

Results The more frequently mutated genes in MDS patients were TUBB1 (11.54%), VWF (8.65%), NBEAL2 (5.77%), and the most common point mutation was TUBB1 p.(R307H) and p.(Q43P). Patients with MK mutation showed a decrease in adenosine diphosphate-induced platelet aggregation, high proportion of CD34+ CD61+ MKs (10.00 vs. 4.00%, p = 0.012), and short overall survival (33.15 vs. 40.50 months, p = 0.013). Further, patients with a higher percent of CD34+ CD61+ MKs (≧20.00%) had lower platelet counts (36.00 × 109/L vs. 88.50 × 109/L, p = 0.015) and more profound emperipolesis (p = 0.001). By analyzing RNA-sequencing of MKs, differentially expressed mRNA was involved in physiological processes including platelet function and platelet activation, especially for MDS patients with high percent of CD34+CD61+MKs. The high levels of expression of CD62P, CXCL10, and S100A9 mRNA, shown by RNA sequencing, were validated by PCR assay.

Conclusion High proportion of CD34+ CD61+ MKs was a poor prognostic factor in MDS patients with MK mutation. CD62P, CXCL10, and S100A9 may be the potential targets to evaluate the molecular link between gene defects and platelet function.

Author Contribution Statement

J.M.L. performed the research and wrote the manuscript. Y.L.L. assisted in performing the research. F.X.L. and J.L.Z. assisted in analyzing the data and writing the manuscript. Z.P.P.; J.L.Z.; R.A.; and Y.B.C. assisted in the data analysis.


Consent for Publication

All authors approved the submission of the paper.


Data Availability Statement

Data will be made available on request.


* Fangxiu Luo and Jialu Zhao contributed equally to this work.




Publication History

Article published online:
10 June 2024

© 2024. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Sandes AF, Yamamoto M, Matarraz S. et al. Altered immunophenotypic features of peripheral blood platelets in myelodysplastic syndromes. Haematologica 2012; 97 (06) 895-902
  • 2 Liu J, Niu H, Yue L. et al. Abnormal platelet phenotypes in patients with myelodysplastic syndromes. Int J Lab Hematol 2020; 42 (04) 371-379
  • 3 Lee K, Ahn HS, Estevez B, Poncz M. RUNX1-deficient human megakaryocytes demonstrate thrombopoietic and platelet half-life and functional defects. Blood 2023; 141 (03) 260-270
  • 4 Arkoun B, Robert E, Boudia F. et al. Stepwise GATA1 and SMC3 mutations alter megakaryocyte differentiation in a Down syndrome leukemia model. J Clin Invest 2022; 132 (14) 132
  • 5 Rosa JP, Raslova H, Bryckaert M. Filamin A: key actor in platelet biology. Blood 2019; 134 (16) 1279-1288
  • 6 Johnson B, Fletcher SJ, Morgan NV. Inherited thrombocytopenia: novel insights into megakaryocyte maturation, proplatelet formation and platelet lifespan. Platelets 2016; 27 (06) 519-525
  • 7 Debili N, Issaad C, Massé JM. et al. Expression of CD34 and platelet glycoproteins during human megakaryocytic differentiation. Blood 1992; 80 (12) 3022-3035
  • 8 Tang G, Wang SA, Menon M, Dresser K, Woda BA, Hao S. High-level CD34 expression on megakaryocytes independently predicts an adverse outcome in patients with myelodysplastic syndromes. Leuk Res 2011; 35 (06) 766-770
  • 9 Pellegrini W, Facchetti F, Marocolo D, Pelizzari AM, Capucci A, Rossi G. Expression of CD34 by megakaryocytes in myelodysplastic syndromes. Haematologica 2000; 85 (10) 1117-1118
  • 10 Martín M, de Paz R, Jiménez-Yuste V. et al. Platelet apoptosis and agonist-mediated activation in myelodysplastic syndromes. Thromb Haemost 2013; 109 (05) 909-919
  • 11 Girtovitis FI, Ntaios G, Papadopoulos A, Ioannidis G, Makris PE. Defective platelet aggregation in myelodysplastic syndromes. Acta Haematol 2007; 118 (02) 117-122
  • 12 Zhang Y, Wu J, Qin T. et al. Comparison of the revised 4th (2016) and 5th (2022) editions of the World Health Organization classification of myelodysplastic neoplasms. Leukemia 2022; 36 (12) 2875-2882
  • 13 Greenberg PL, Tuechler H, Schanz J. et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood 2012; 120 (12) 2454-2465
  • 14 Li J, Peng Z, Luo F, Chen Y. SET domain containing 2 deficiency in myelodysplastic syndrome. Front Genet 2020; 11: 794
  • 15 Heililahong H, Jin P, Lei H. et al. Whole transcriptome analysis of platelet concentrates during storage. Blood Transfus 2023; 21 (02) 146-156
  • 16 Matsumura T, Nakamura-Ishizu A, Takaoka K. et al. TUBB1 dysfunction in inherited thrombocytopenia causes genome instability. Br J Haematol 2019; 185 (05) 888-902
  • 17 Kreft IC, Huisman EJ, Cnossen MH. et al. Proteomic landscapes of inherited platelet disorders with different etiologies. J Thromb Haemost 2023; 21 (02) 359-372.e3
  • 18 Larocca LM, Heller PG, Podda G. et al. Megakaryocytic emperipolesis and platelet function abnormalities in five patients with gray platelet syndrome. Platelets 2015; 26 (08) 751-757
  • 19 Kahr WH, Lo RW, Li L. et al. Abnormal megakaryocyte development and platelet function in Nbeal2(-/-) mice. Blood 2013; 122 (19) 3349-3358
  • 20 Bury L, Malara A, Momi S, Petito E, Balduini A, Gresele P. Mechanisms of thrombocytopenia in platelet-type von Willebrand disease. Haematologica 2019; 104 (07) 1473-1481
  • 21 Warren JT, Di Paola J. Genetics of inherited thrombocytopenias. Blood 2022; 139 (22) 3264-3277
  • 22 Galera P, Dulau-Florea A, Calvo KR. Inherited thrombocytopenia and platelet disorders with germline predisposition to myeloid neoplasia. Int J Lab Hematol 2019; 41 (Suppl. 01) 131-141
  • 23 Duarte FB, DE Jesus Dos Santos TE, Barbosa MC. et al. Presence of CD34+ in megakaryocytes in association with p53 expression predicts unfavorable prognosis in low-risk myelodysplastic syndrome patients. In Vivo 2019; 33 (01) 277-280
  • 24 Marneth AE, van Heerde WL, Hebeda KM. et al. Platelet CD34 expression and α/δ-granule abnormalities in GFI1B- and RUNX1-related familial bleeding disorders. Blood 2017; 129 (12) 1733-1736
  • 25 Rabbolini DJ, Morel-Kopp MC, Chen Q. et al. Thrombocytopenia and CD34 expression is decoupled from α-granule deficiency with mutation of the first growth factor-independent 1B zinc finger. J Thromb Haemost 2017; 15 (11) 2245-2258
  • 26 van Grinsven E, Udalova IA. You reap what you sow: neutrophils “plucking” platelets harvest prothrombotic effects. Immunity 2022; 55 (12) 2217-2219
  • 27 Cunin P, Nigrovic PA. Megakaryocyte emperipolesis: a new frontier in cell-in-cell interaction. Platelets 2020; 31 (06) 700-706
  • 28 Köhler A, De Filippo K, Hasenberg M. et al. G-CSF-mediated thrombopoietin release triggers neutrophil motility and mobilization from bone marrow via induction of Cxcr2 ligands. Blood 2011; 117 (16) 4349-4357
  • 29 Cunin P, Bouslama R, Machlus KR. et al. Megakaryocyte emperipolesis mediates membrane transfer from intracytoplasmic neutrophils to platelets. eLife 2019; 8: e44031
  • 30 Brennan Y, Levade M, Ward CM. Acquired platelet function disorders. Thromb Res 2020; 196: 561-568
  • 31 Baharoglu MI, Cordonnier C, Al-Shahi Salman R. et al; PATCH Investigators. Platelet transfusion versus standard care after acute stroke due to spontaneous cerebral haemorrhage associated with antiplatelet therapy (PATCH): a randomised, open-label, phase 3 trial. Lancet 2016; 387 (10038): 2605-2613