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 (1.2 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Increased plasma periostin concentration predicts angiographic vasospasm development in non-severe aneurysmal subarachnoid hemorrhage

Hiroki OinakaFumihiro Kawakita( )Hideki NakajimaYume SuzukiMai NampeiTakeshi OkadaRyuta YasudaNaoki TomaHidenori SuzukipSEED Group,1
Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan

1 Members along with their affiliations listed in Appendix.

Show Author Information

Abstract

It is unknown whether plasma concentrations of a matricellular protein periostin change in association with the development of angiographic vasospasm after aneurysmal subarachnoid hemorrhage (SAH). In 113 patients with aneurysmal SAH of World Federation of Neurological Surgeons grades 1–3 at admission, plasma periostin concentrations were serially measured at days 1–3, 4–6, 7–9 and 10–12 after SAH onset. Measured periostin levels and clinical variables were compared between patients with and without angiographic vasospasm. Periostin concentrations were significantly higher in patients with angiographic vasospasm at days 4–6 and 7–9. Receiver operating characteristic curve analyses indicated that cutoff plasma periostin values of 54.3 ng/ml at days 4–6 and 58.1 ng/ml at days 7–9 predicted or diagnosed angiographic vasospasm development with a specificity of 66.0 % and a sensitivity of 72.7 %, and a specificity of 75.0 % and a sensitivity of 55.0 %, respectively. Multivariate analyses also revealed that increased plasma periostin concentrations at days 7–9 was independently associated with angiographic vasospasm development. This study showed for the first time that plasma periostin levels were increased in patients with angiographic vasospasm. These findings suggest that plasma periostin can serve as a biomarker and may be a new therapeutic target for angiographic vasospasm after SAH.

References

1

Vergouwen MD, Vermeulen M, van Gijn J, et al. Definition of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage as an outcome event in clinical trials and observational studies: proposal of a multidisciplinary research group. Stroke. 2010;41:2391–2395. https://doi.org/10.1161/STROKEAHA.110.589275.

2

Suzuki H, Nakatsuka Y, Yasuda R, et al. Dose-dependent inhibitory effects of cilostazol on delayed cerebral infarction after aneurysmal subarachnoid hemorrhage. Transl Stroke Res. 2019;10:381–388. https://doi.org/10.1007/s12975-018-0650-y.

3

Suzuki H, Siba M, Nakatsuka Y, et al. Higher cerebrospinal fluid pH may contribute to the development of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Transl Stroke Res. 2017;8:165–173. https://doi.org/10.1007/s12975-016-0500-8.

4

Okada T, Suzuki H. Toll-like receptor 4 as a possible therapeutic target for delayed brain injuries after aneurysmal subarachnoid hemorrhage. Neural Regen Res. 2017;12:193–196. https://doi.org/10.4103/1673-5374.200795.

5

Atangana E, Schneider UC, Blecharz K, et al. Intravascular inflammation triggers intracerebral activated microglia and contributes to secondary brain injury after experimental subarachnoid hemorrhage (eSAH). Transl Stroke Res. 2017;8:144–156. https://doi.org/10.1007/s12975-016-0485-3.

6

Nishikawa H, Suzuki H. Possible role of inflammation and galectin-3 in brain injury after subarachnoid hemorrhage. Brain Sci. 2018;8:30. https://doi.org/10.3390/brainsci8020030.

7

De Oliveira Manoel AL, Macdonald RL. Neuroinflammation as a target for intervention in subarachnoid hemorrhage. Front Neurol. 2018;9:292. https://doi.org/10.3389/fneur.2018.00292.

8

Zheng VZ, Wong GKC. Neuroinflammation responses after subarachnoid hemorrhage: a review. J Clin Neurosci. 2017;42:7–11. https://doi.org/10.1016/j.jocn.2017.02.001.

9

Lucke-World BP, Logsdon AF, Manoranjan B, et al. Aneurysmal subarachnoid hemorrhage and neuroinflammation: a comprehensive review. Int J Mol Sci. 2016;17:497. https://doi.org/10.3390/ijms17040497.

10

Hu F, Shang XF, Wang W, et al. High-levels expression of periostin is significantly correlated with tumour angiogenesis and poor prognosis in osteosarcoma. Int J Exp Pathol. 2016;97:86–92. https://doi.org/10.1111/iep.12171.

11

Izuhara K, Nunomura S, Nanri Y, et al. Periostin in inflammation and allergy. Cell Mol Life Sci. 2017;74:4293–4303. https://doi.org/10.1007/s00018-017-2648-0.

12

Suzuki H, Nishikawa H, Kawakita F. Matricellular proteins as possible biomarkers for early brain injury after aneurysmal subarachnoid hemorrhage. Neural Regen Res. 2018;13:1175–1178. https://doi.org/10.4103/1673-5374.235022.

13

Liu L, Kawakita F, Fujimoto M, et al. Role of periostin in early brain injury after subarachnoid hemorrhage in mice. Stroke. 2017;48:1108–1111. https://doi.org/10.1161/STROKEAHA.117.016629.

14

Luo W, Wang H, Hu J. Increased concentration of serum periostin is associated rith poor outcome of patients with aneurysmal subarachnoid hemorrhage. J Clin Lab Anal. 2018;32:e22389.

15

Kanamaru H, Kawakita F, Nakano F, et al. Plasma periostin and delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Neurotherapeutics. 2019;16:480–490. https://doi.org/10.1007/s13311-018-00707-y.

16

Tanioka S, Ishida F, Nakano F, et al. Machine learning analysis of matricellular proteins and clinical variables for early prediction of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Mol Neurobiol. 2019;56:7128–7135. https://doi.org/10.1007/s12035-019-1601-7.

17

Suzuki H, Kanamaru H, Kawakita F, et al. Cerebrovascular pathophysiology of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Histol Histopathol. 2021;36:143–158. https://doi.org/10.14670/HH-18-253.

18

Giraldo EA, Mandrekar JN, Rubin MN, et al. Timing of clinical grade assessment and poor outcome in patients with aneurysmal subarachnoid hemorrhage. J Neurosurg. 2012;117:15–19. https://doi.org/10.3171/2012.3.JNS11706.

19

Inoue T, Akashi K, Watanabe M, et al. Periostin as biomarker for the diagnosis of pediatric asthma. Pediatr Allergy Immunol. 2016;27:521–526. https://doi.org/10.1111/pai.12575.

20

Dorafshan RM, Safaei S, et al. Periostin: biology and function in cancer. Cancer Cell Int. 2022;22:315. https://doi.org/10.1186/s12935-022-02714-8.

21

Claassen J, Bernardini GL, Kreiter K, et al. Effect of cisternal and ventricular blood on risk of delayed cerebral ischemia after subarachnoid hemorrhage: the Fisher scale revisited. Stroke. 2001;2012–2020. https://doi.org/10.1161/hs0901.095677.

22

Dupont S, Rabinstein AA. Extent of acute hydrocephalus after subarachnoid hemorrhage as a risk factor for poor functional outcome. Neurol Res. 2013;35:107–110. https://doi.org/10.1179/1743132812Y.0000000122.

23

Liu AY, Zhang H, Ouyang G. Periostin, a multifunctional matricellular protein in inflammatory and tumor microenvironment. Matrix Biol. 2014;37:150–156. https://doi.org/10.1016/j.matbio.2014.04.007.

24

Matsusaka M, Kabata H, Fukunaga K, et al. Phenotype of asthma related with high serum periostin levels. Allergol Int. 2015;64:175–180. https://doi.org/10.1016/j.alit.2014.07.003.

25

Satirapoj B, Wang Y, Chamberlin MP, et al. Periostin: novel tissue and urinary biomarker of progressive renal injury induces a coordinated mesenchymal phenotype in tubular cells. Nephrol Dial Transplant. 2012;27:2702–2711. https://doi.org/10.1093/ndt/gfr670.

26

Bobolea I, Barranco P, Del Pozo V, et al. Sputum periostin in patients with different severe asthma phenotypes. Allergy. 2015;70:540–546. https://doi.org/10.1111/all.12580.

27

Fujishima H, Okada N, Matsumoto K, et al. The usefulness of measuring tear periostin for the diagnosis and management of ocular allergic diseases. J Allergy Clin Immunol. 2016;138:459–467. https://doi.org/10.1016/j.jaci.2015.11.039.

28

Shimamura M, Taniyama Y, Katsuragi N, et al. Role of central nervous system periostin in cerebral ischemia. Stroke. 2012;43:1108–1114. https://doi.org/10.1161/STROKEAHA.111.636662.

29

Findlay M, Nisar J, Darsaut T. Cerebral vasospasm: a review. Can J Neurol Sci. 2016;43:15–32. https://doi.org/10.1017/cjn.2015.288.

30

Kawakita F, Fujimoto M, Liu L, et al. Effects of toll-like receptor 4 antagonists against cerebral vasospasm after experimental subarachnoid hemorrhage in mice. Mol Neurobiol. 2017;54:66254–66633. https://doi.org/10.1007/s12035-016-0178-7.

31

Suzuki H, Hasegawa Y, Kanamaru K, et al. Mitogen-activated protein kinases in cerebral vasospasm after subarachnoid hemorrhage: a review. Acta Neurochir Suppl. 2011;110:133–139. https://doi.org/10.1007/978-3-7091-0353-1_23.

32

Suzuki H. How to promote hemoglobin scavenging or clearance and detoxification in hemorrhagic stroke. Transl Stroke Res. 2023;14:625–627. https://doi.org/10.1007/s12975-022-01075-8.

33

Hasegawa Y, Suzuki H, Uekawa K, et al. Characteristics of cerebrovascular injury in the hyperacute phase after induced severe subarachnoid hemorrhage. Transl Stroke Res. 2015;6:458–466. https://doi.org/10.1007/s12975-015-0423-9.

34

Kanamaru H, Suzuki H. Potential therapeutic molecular targets for blood-brain barrier disruption after subarachnoid hemorrhage. Neural Regen Res. 2019;14:1138–1143. https://doi.org/10.4103/1673-5374.251190.

35

Suzuki H, Fujimoto M, Kawakita F, et al. Tenascin-C in brain injuries and edema after subarachnoid hemorrhage: findings from basic and clinical studies. J Neurosci Res. 2020;98:42–56. https://doi.org/10.1002/jnr.24330.

36

Suzuki H, Kawakita F, Asada R. Neuroelectric mechanisms of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Int J Mol Sci. 2022;23:3102. https://doi.org/10.3390/ijms23063102.

37

Okada T, Kawakita F, Nishikawa H, et al. Selective Toll-like receptor 4 antagonists prevent acute blood-brain barrier disruption after subarachnoid hemorrhage in mice. Mol Neurobiol. 2019;56:976–985. https://doi.org/10.1007/s12035-018-1145-2.

38

Kanamaru H, Kawakita F, Nishikawa H, et al. Clarithromycin ameliorates early brain injury after subarachnoid hemorrhage via suppressing periostin-related pathways in mice. Neurotherapeutics. 2021;18:1880–1890. https://doi.org/10.1007/s13311-021-01050-5.

39

Kawakita F, Nakano F, Kanamaru H, et al. Anti-apoptotic effects of AMPA receptor antagonist perampanel in early brain injury after subarachnoid hemorrhage in mice. Transl Stroke Res. 2023. https://doi.org/10.1007/s12975-023-01138-4.

40

Lindner V, Wang Q, Conley BA, et al. Vascular injury induces expression of periostin: implications for vascular cell differentiation and migration. Arterioscler Thromb Vasc Biol. 2005;25:77–83. https://doi.org/10.1161/01.ATV.0000149141.81230.c6.

41

Kawakita F, Kanamaru H, Asada R, et al. Potential roles of matricellular proteins in stroke. Exp Neurol. 2019;322. https://doi.org/10.1016/j.expneurol.2019.113057 113057.

Brain Hemorrhages
Pages 1-7
Cite this article:
Oinaka H, Kawakita F, Nakajima H, et al. Increased plasma periostin concentration predicts angiographic vasospasm development in non-severe aneurysmal subarachnoid hemorrhage. Brain Hemorrhages, 2024, 5(1): 1-7. https://doi.org/10.1016/j.hest.2023.12.003

121

Views

1

Downloads

2

Crossref

2

Web of Science

2

Scopus

Altmetrics

Received: 08 November 2023
Revised: 11 December 2023
Accepted: 12 December 2023
Published: 16 December 2023
© 2023 International Hemorrhagic Stroke Association.

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

Return