J Korean Neurosurg Soc.  2023 Nov;66(6):642-651. 10.3340/jkns.2023.0005.

Cyclin-Dependent Kinase Inhibitor 2A is a Key Regulator of Cell Cycle Arrest and Senescence in Endothelial Colony-Forming Cells in Moyamoya Disease

Affiliations
  • 1Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children’s Hospital, Seoul, Korea
  • 2Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
  • 3Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea
  • 4Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea

Abstract


Objective
: Endothelial colony-forming cells (ECFCs) have been reported to play an important role in the pathogenesis of moyamoya disease (MMD). We have previously observed stagnant growth in MMD ECFCs with functional impairment of tubule formation. We aimed to verify the key regulators and related signaling pathways involved in the functional defects of MMD ECFCs.
Methods
: ECFCs were cultured from peripheral blood mononuclear cells of healthy volunteers (normal) and MMD patients. Low-density lipoproteins uptake, flow cytometry, high content screening, senescence-associated β-galactosidase, immunofluorescence, cell cycle, tubule formation, microarray, real-time quantitative polymerase chain reaction, small interfering RNA transfection, and western blot analyses were performed.
Results
: The acquisition of cells that can be cultured for a long time with the characteristics of late ECFCs was significantly lower in the MMD patients than the normal. Importantly, the MMD ECFCs showed decreased cellular proliferation with G1 cell cycle arrest and cellular senescence compared to the normal ECFCs. A pathway enrichment analysis demonstrated that the cell cycle pathway was the major enriched pathway, which is consistent with the results of the functional analysis of ECFCs. Among the genes associated with the cell cycle, cyclin-dependent kinase inhibitor 2A (CDKN2A) showed the highest expression in MMD ECFCs. Knockdown of CDKN2A in MMD ECFCs enhanced proliferation by reducing G1 cell cycle arrest and inhibiting senescence through the regulation of CDK4 and phospho retinoblastoma protein.
Conclusion
: Our study suggests that CDKN2A plays an important role in the growth retardation of MMD ECFCs by inducing cell cycle arrest and senescence.

Keyword

Moyamoya disease; Endothelial colony-forming cells; Cyclin dependent kinase inhibitor 2A; Cell cycle; Cellular senescence

Figure

  • Fig. 1. Cell growth arrest and senescence of MMD ECFCs. A : Cell proliferation is assessed via a high-throughput screening (HCS) assay. Representative growth curves (nonexpanded cells) of ECFCs are generated from cell images taken every 4 hours using the imaging system and its cell density algorithm. B : Doubling time results suggest that MMD ECFCs are significantly slower than normal ECFCs. C : Cell cycle analysis by flow cytometry shows that G1 arrest is higher in MMD ECFCs than in normal ECFCs. D and E : Senescence-associated β-galactosidase (SA-β-gal) staining (×100) reveals more SA-β-gal-positive cells in MMD ECFCs than in normal ECFCs. F and G : In both MMD ECFCs and normal ECFCs, few caspase-3-positive cells are observed (×100). Scale bars : 100 μm. *p<0.05, **p<0.01, and ***p<0.001. ECFCs : endothelial colony-forming cells, MMD : Moyamoya disease. DAPI : 4',6-diamidino-2-phenylindole.

  • Fig. 2. Analysis of the major biological pathways of MMD ECFCs and verification of the associated genes. A : Functional gene enrichment analysis of identified differentially expressed genes (DEGs) as biological pathways. B : Validation of cell cycle-related genes by RT-qPCR. *p<0.05. ATM : ataxia telangiectasia mutated, ECFCs : endothelial colony-forming cells, MMD : Moyamoya disease, CDKN : cyclin-dependent kinase inhibitor, CCNA : cyclin A, CCNB : cyclin B, CCND : cyclin D, SIRT1 : sirtuin 1, RB1 : retinoblastoma, RT-qPCR : real-time quantitative polymerase chain reaction.

  • Fig. 3. Inhibition effect of CDKN2A in MMD ECFCs. A : CDKN2A-siRNA treatment effectively reduces CDKN2A mRNA expression by 70%. B : Cell viability analysis reveals increased cell growth of MMD ECFCs by knockdown of CDKN2A. C : Cell cycle analysis confirms inhibition of G1 arrest by knockdown of CDKN2A in MMD ECFCs. D and E : Senescence-associated β-galactosidase (SA-β-gal) assay (×100) shows a significantly decreased the cellular senescence of MMD ECFCs by knockdown of CDKN2A. F and G : Caspase-3 staining (×100) indicates that knockdown of CDKN2NA does not affect apoptosis in MMD ECFCs. H and I : Tube formation analysis demonstrates that knockdown of CDKN2A significantly improves the tubule formation capability of MMD ECFCs (×100). Scale bars : 100 μm. ***p<0.001. CDKN2A : cyclin-dependent kinase inhibitor 2A, NC: negative control, MMD : Moyamoya disease, ECFCs : endothelial colony-forming cells.

  • Fig. 4. Cellular senescence and cell cycle-related signaling pathways affected by inhibition of CDKN2A in MMD ECFCs. A : Representative western blot results indicate a significant increase in CDK4 and pRB by knockdown of CDKN2A in MMD ECFCs at 24 hours. On the other hand, CDKN1A, CDK2, and CDK6 are not affected by the knockdown of CDKN2A in MMD ECFCs. B : Densitometry graphs of the western blots are quantified, and the data are expressed as the mean±standard deviation. *p<0.05, ***p<0.001. NC : negative control, CDKN2A : cyclin-dependent kinase inhibitor 2A, pRB1 : phospho retinoblastoma1, MMD : Moyamoya disease, ECFCs : endothelial colony-forming cells.


Reference

References

1. Bang OY, Fujimura M, Kim SK. The pathophysiology of moyamoya disease: an update. J Stroke. 18:12–20. 2016.
Article
2. Chang TY, Tsai WC, Huang TS, Su SH, Chang CY, Ma HY, et al. Dysregulation of endothelial colony-forming cell function by a negative feedback loop of circulating miR-146a and -146b in cardiovascular disease patients. PLoS One. 12:e0181562. 2017.
Article
3. Choi SA, Chong S, Kwak PA, Moon YJ, Jangra A, Phi JH, et al. Impaired functional recovery of endothelial colony-forming cells from moyamoya disease in a chronic cerebral hypoperfusion rat model. J Neurosurg Pediatr. 23:204–213. 2018.
Article
4. Guey S, Tournier-Lasserve E, Hervé D, Kossorotoff M. Moyamoya disease and syndromes: from genetics to clinical management. Appl Clin Genet. 8:49–68. 2015.
5. Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, et al. Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine. 47:446–456. 2019.
Article
6. Jangra A, Choi SA, Koh EJ, Moon YJ, Wang KC, Phi JH, et al. Panobinostat, a histone deacetylase inhibitor, rescues the angiogenic potential of endothelial colony-forming cells in moyamoya disease. Childs Nerv Syst. 35:823–831. 2019.
Article
7. Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, et al. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine. 40:554–563. 2019.
Article
8. Kang HS, Wang KC, Kim SK. Circulating vascular progenitor cells in moyamoya disease. J Korean Neurosurg Soc. 57:428–431. 2015.
Article
9. Kim JH, Jung JH, Phi JH, Kang HS, Kim JE, Chae JH, et al. Decreased level and defective function of circulating endothelial progenitor cells in children with moyamoya disease. J Neurosci Res. 88:510–518. 2010.
Article
10. Kim SK, Cho BK, Phi JH, Lee JY, Chae JH, Kim KJ, et al. Pediatric moyamoya disease: an analysis of 410 consecutive cases. Ann Neurol. 68:92–101. 2010.
Article
11. Kirkland JL, Tchkonia T. Cellular senescence: a translational perspective. EBioMedicine. 21:21–28. 2017.
Article
12. Lee JY, Moon YJ, Lee HO, Park AK, Choi SA, Wang KC, et al. Deregulation of retinaldehyde dehydrogenase 2 leads to defective angiogenic function of endothelial colony-forming cells in pediatric moyamoya disease. Arterioscler Thromb Vasc Biol. 35:1670–1677. 2015.
Article
13. Liu W, Morito D, Takashima S, Mineharu Y, Kobayashi H, Hitomi T, et al. Identification of RNF213 as a susceptibility gene for moyamoya disease and its possible role in vascular development. PLoS One. 6:e22542. 2011.
Article
14. Ma Q, Xu Y, Liao H, Cai Y, Xu L, Xiao D, et al. Identification and validation of key genes associated with non-small-cell lung cancer. J Cell Physiol. 234:22742–22752. 2019.
Article
15. Medina RJ, Barber CL, Sabatier F, Dignat-George F, Melero-Martin JM, Khosrotehrani K, et al. Endothelial progenitors: a consensus statement on nomenclature. Stem Cells Transl Med. 6:1316–1320. 2017.
Article
16. Oh JW, Oh YJ, Han S, Her NG, Nam DH. High-content analysis-based sensitivity prediction and novel therapeutics screening for c-Met-addicted glioblastoma. Cancers (Basel). 13:372. 2021.
Article
17. Phi JH, Choi SA, Lim SH, Lee J, Wang KC, Park SH, et al. ID3 contributes to cerebrospinal fluid seeding and poor prognosis in medulloblastoma. BMC Cancer. 13:291. 2013.
Article
18. Phi JH, Suzuki N, Moon YJ, Park AK, Wang KC, Lee JY, et al. Chemokine ligand 5 (CCL5) derived from endothelial colony-forming cells (ECFCs) mediates recruitment of smooth muscle progenitor cells (SPCs) toward critical vascular locations in moyamoya disease. PLoS One. 12:e0169714. 2017.
Article
19. Rafat N, Beck GCh, Peña-Tapia PG, Schmiedek P, Vajkoczy P. Increased levels of circulating endothelial progenitor cells in patients with moyamoya disease. Stroke. 40:432–438. 2009.
Article
20. Regnault V, Challande P, Pinet F, Li Z, Lacolley P. Cell senescence: basic mechanisms and the need for computational networks in vascular ageing. Cardiovasc Res. 117:1841–1858. 2021.
Article
21. Roder C, Nayak NR, Khan N, Tatagiba M, Inoue I, Krischek B. Genetics of moyamoya disease. J Hum Genet. 55:711–716. 2010.
Article
22. Schira-Heinen J, Czapla A, Hendricks M, Kloetgen A, Wruck W, Adjaye J, et al. Functional omics analyses reveal only minor effects of microRNAs on human somatic stem cell differentiation. Sci Rep. 10:3284. 2020.
Article
23. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 13:1501–1512. 1999.
Article
24. Urbich C, Dimmeler S. Endothelial progenitor cells: characterization and role in vascular biology. Circ Res. 95:343–353. 2004.
25. Wang HH, Lee YN, Su CH, Shu KT, Liu WT, Hsieh CL, et al. S-phase kinase-associated protein-2 rejuvenates senescent endothelial progenitor cells and induces angiogenesis in vivo. Sci Rep. 10:6646. 2020.
26. Wissler Gerdes EO, Misra A, Netto JME, Tchkonia T, Kirkland JL. Strategies for late phase preclinical and early clinical trials of senolytics. Mech Ageing Dev. 200:111591. 2021.
Article
27. Witt G, Keminer O, Leu J, Tandon R, Meiser I, Willing A, et al. An automated and high-throughput-screening compatible pluripotent stem cell-based test platform for developmental and reproductive toxicity assessment of small molecule compounds. Cell Biol Toxicol. 37:229–243. 2021.
Article
28. Yu J, Du Q, Hu M, Zhang J, Chen J. Endothelial progenitor cells in moyamoya disease: current situation and controversial issues. Cell Transplant. 29:963689720913259. 2020.
Article
Full Text Links
  • JKNS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr