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
Review Article | Open Access

Phenotype change of polarized microglia after intracerebral hemorrhage: Advances in research

Ran TangaZhuyi HuangbHeling Chuc( )
Nanjing Medical University, Nanjing 210000, China
Department of Neurology, Shanghai East Hospital, Tongji University, Shanghai 200120, China
Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
Show Author Information

Abstract

Spontaneous intracerebral hemorrhage (ICH) accounts for approximately 10–15% of all strokes with high rates of mortality and residual disability. Multiple studies have demonstrated that microglia-mediated neuroinflammation exerts a significant function on the secondary injury of ICH, which may cause severe brain damage or potential neurorestoration in different conditions. Microglia are the major phagocytes in central nervous system. In different time courses after ICH, microglia can be activated and polarized into two different states: classic M1-like phenotypes in the early stage and alternative M2-like phenotypes in long-term recovery. The mechanism of this phenotypic change is very complicated, including the influence of inflammatory mediators and microglia-astrocyte crosstalk. In this work, we encapsulate the M1-to-M2 microglial phenotype transition as well as its effects on post-ICH neuroinflammation, and describe potential therapeutic targets based on this feature.

References

1

An SJ, Kim TJ, Yoon BW. Epidemiology, risk factors, and clinical features of intracerebral hemorrhage: an update. J Stroke. 2017;19: 3–10.

2

Qureshi AI, Mendelow AD, Hanley DF. Intracerebral haemorrhage. Lancet. 2009;373: 1632–1644.

3

Ziai WC, Carhuapoma JR. Intracerebral hemorrhage. Continuum (Minneap Minn). 2018;24(6): 1603–1622.

4

Mracsko E, Veltkamp R. Neuroinflammation after intracerebral hemorrhage. Front Cell Neurosci. 2014;8: 388.

5

Colonna M, Butovsky O. Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol. 2017;35: 441–468.

6

Hu X, Leak RK, Shi Y, et al. Microglial and macrophage polarization—new prospects for brain repair. Nat Rev Neurol. 2015;11: 56–64.

7

Wan S, Cheng Y, Jin H, et al. Microglia activation and polarization after intracerebral hemorrhage in mice: the role of protease-activated receptor-1. Transl Stroke Res. 2016;7: 478–487.

8

Zhang Z, Zhang Z, Zhang Z, et al. Microglial polarization and inflammatory mediators after intracerebral hemorrhage. Mol Neurobiol. 2017;54:1874–1886.

9
Lin X, Ye H, Siaw-Debrah F, et al. AC-YVAD-CMK Inhibits Pyroptosis and Improves Functional Outcome after Intracerebral Hemorrhage. Biomed Res Int. 2018;2018:3706047. Published 2018 Oct 16. doi: 10.1155/2018/3706047.
10

Ye X, Zuo D, Yu L, et al. ROS/TXNIP pathway contributes to thrombin induced NLRP3 inflammasome activation and cell apoptosis in microglia. Biochem Biophys Res Commun. 2017;485(2):499–505.

11

Lan X, Han X, Li Q, et al. Pinocembrin protects hemorrhagic brain primarily by inhibiting toll-like receptor 4 and reducing M1 phenotype microglia. Brain Behav Immun. 2016;61:326–339.

12

Xiong XY, Liu L, Wang FX, et al. Toll-like receptor 4/MyD88-mediated signaling of hepcidin expression causing brain iron accumulation, oxidative injury, and cognitive impairment after intracerebral hemorrhage. Circulation. 2016;134:1025–1038.

13

Wang Y, Su L, Morin MD, et al. TLR4/MD-2 activation by a synthetic agonist with no similarity to LPS. Proc Natl Acad Sci USA. 2016;113:E884–E893.

14

Fang H, Wang PF, Zhou Y, et al. Toll-like receptor 4 signaling in intracerebral hemorrhage-induced inflammation and injury. J Neuroinflammation. 2013;10:27.

15

Lin S, Yin Q, Zhong Q, et al. Heme activates TLR4-mediated inflammatory injury via MyD88/TRIF signaling pathway in intracerebral hemorrhage. J Neuroinflammation. 2012;9:46.

16

Yang Y, Tan X, Xu J, et al. Luteolin alleviates neuroinflammation via downregulating the TLR4/TRAF6/NF-κB pathway after intracerebral hemorrhage. Biomed Pharmacother. 2020;126:110044.

17

Rodriguez-Yanez M, Brea D, Arias S, et al. Increased expression of Toll-like receptors 2 and 4 is associated with poor outcome in intracerebral hemorrhage. J Neuroimmunol. 2012;247:75–80.

18

Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol. 2016;173:649–665.

19

Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation. 2014;11:98.

20

Mantovani A, Sica A, Sozzani S, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25:677–686.

21

Taylor RA, Chang CF, Goods BA, et al. TGF-beta1 modulates microglial phenotype and promotes recovery after intracerebral hemorrhage. J Clin Invest. 2017;127:280–292.

22

Jiang C, Wang Y, Hu Q, et al. Immune changes in peripheral blood and hematoma of patients with intracerebral hemorrhage. FASEB J. 2020.

23

Yang J, Ding S, Huang W, et al. Interleukin-4 Ameliorates the functional recovery of intracerebral hemorrhage through the alternative activation of microglia/macrophage. Front Neurosci. 2016;10:61.

24

Flores JJ, Klebe D, Rolland WB, Lekic T, Krafft PR, Zhang JH. PPARγ-induced upregulation of CD36 enhances hematoma resolution and attenuates longterm neurological deficits after germinal matrix hemorrhage in neonatal rats. Neurobiol Dis. 2016;87:124–133.

25
Kaiser S, Selzner L, Weber J, Schallner N. Carbon monoxide controls microglial erythrophagocytosis by regulating CD36 surface expression to reduce the severity of hemorrhagic injury[published online ahead of print, 2020 May 31]. Glia. 2020;10.1002/glia.23864. doi: 10.1002/glia.23864.
26

Zhao JL, Chen YJ, Yu J, et al. ISO-alpha-acids improve the hematoma resolution and prevent peri-hematoma inflammations by transforming microglia via PPARgamma-CD36 axis in ICH rats. Int Immunopharmacol. 2020;83:106396.

27

Mu Q, Wang L, Hang H, Liu C, Wu G. Rosiglitazone pretreatment influences thrombin-induced phagocytosis by rat microglia via activating PPARγ and CD36. Neurosci Lett. 2017;651:159–164.

28

Lan X, Han X, Li Q, et al. Modulators of microglial activation and polarization after intracerebral haemorrhage. Nat Rev Neurol. 2017;13:420–433.

29

Bai Q, Xue M, Yong VW. Microglia and macrophage phenotypes in intracerebral haemorrhage injury: therapeutic opportunities. Brain. 2020;143 (5):1297–1314.

30

Chen Z, Xu N, Dai X, et al. Interleukin-33 reduces neuronal damage and white matter injury via selective microglia M2 polarization after intracerebral hemorrhage in rats. Brain Res Bull. 2019;150:127–135.

31

Yang S, Nakamura T, Hua Y, et al. The role of complement C3 in intracerebral hemorrhage-induced brain injury. J Cereb Blood Flow Metab. 2006;26:1490–1495.

32

Yuan B, Yuan B, Fu F, et al. C5a/C5aR pathway plays a vital role in brain inflammatory injury via initiating Fgl-2 in intracerebral hemorrhage. Mol Neurobiol. 2017;54:6187–6197.

33

Li G, Fan RM, Chen JL, et al. Neuroprotective effects of argatroban and C5a receptor antagonist (PMX53) following intracerebral haemorrhage. Clin Exp Immunol. 2014;175:285–295.

34

Zhou Y, Xiong KL, Lin S, et al. Elevation of high-mobility group protein box-1 in serum correlates with severity of acute intracerebral hemorrhage. Mediat Inflamm. 2010;2010:142458.

35

Lei C, Lin S, Zhang C, et al. High-mobility group box1 protein promotes neuroinflammation after intracerebral hemorrhage in rats. Neuroscience. 2013;228:190–199.

36
Paudel YN, Shaikh MF, Chakraborti A, et al. HMGB1: A Common Biomarker and Potential Target for TBI, Neuroinflammation, Epilepsy, and Cognitive Dysfunction. Front Neurosci. 2018;12: 628. Published 2018 Sep 11. doi: 10.3389/fnins.2018.00628.
37
Wang D, Liu K, Wake H, Teshigawara K, Mori S, Nishibori M. Anti-high mobility group box-1 (HMGB1) antibody inhibits hemorrhage-induced brain injury and improved neurological deficits in rats. Sci Rep. 2017;7: 46243. Published 2017 Apr 10. doi: 10.1038/srep46243.
38

Lan X, Han X, Liu X, et al. Inflammatory responses after intracerebral hemorrhage: from cellular function to therapeutic targets. J Cereb Blood Flow Metab. 2019;39:184–186.

39

He M, Dong H, Huang Y, et al. Astrocyte-derived CCL2 is associated with M1 activation and recruitment of cultured microglial cells. Cell Physiol Biochem. 2016;38:859–870.

40

Hernandez-Rabaza V, Cabrera-Pastor A, Taoro-Gonzalez L, et al. Neuroinflammation increases GABAergic tone and impairs cognitive and motor function in hyperammonemia by increasing GAT-3 membrane expression. Reversal by sulforaphane by promoting M2 polarization of microglia[J]. Journal of neuroinflammation, 2016, 13:83.

41

Eun JJ, Sun G, Bautista GJ, et al. The mitochondria-derived peptide humanin improves recovery from intracerebral hemorrhage: implication of mitochondria transfer and microglia phenotype change. J Neurosci. 2020.

42

Lattanzi S, Brigo F, Trinka E, Cagnetti C, Di Napoli M, Silvestrini M. Neutrophil-to-lymphocyte ratio in acute cerebral hemorrhage: a system review. Transl Stroke Res. 2019;10(2):137–145.

43

Zhou K, Zhong Q, Wang Y, et al. Regulatory T cells ameliorate intracerebral hemorrhage-induced inflammatory injury by modulating microglia/macrophage polarization through the IL-10/GSK3β/PTEN axis. J Cereb Blood Flow Metab. 2017;37:967–979.

44

Yang Z, Yu A, Liu Y, et al. Regulatory T cells inhibit microglia activation and protect against inflammatory injury in intracerebral hemorrhage. Int Immunopharmacol. 2014;22:522–525.

45

Mao L, Yuan H, Wang W, et al. Adoptive regulatory T-cell therapy attenuates perihematomal inflammation in a mouse model of experimental intracerebral hemorrhage. Cell Mol Neurobiol. 2017;37:919–929.

46

Yang H, Gao XJ, Li YJ, et al. Minocycline reduces intracerebral hemorrhage-induced white matter injury in piglets. CNS Neurosci Ther. 2019;25:1195–1206.

47

Miao H, Li R, Han C, et al. Minocycline promotes posthemorrhagic neurogenesis via M2 microglia polarization via upregulation of the TrkB/BDNF pathway in rats. J Neurophysiol. 2018;120:1307–1317.

48

Wang G, Li Z, Li S, et al. Minocycline preserves the integrity and permeability of BBB by altering the activity of DKK1–Wnt signaling in ICH model. Neuroscience. 2019;415:135–146.

49

Dai S, Hua Y, Keep RF, et al. Minocycline attenuates brain injury and iron overload after intracerebral hemorrhage in aged female rats. Neurobiol Dis. 2019;126:76–84.

50

Malhotra K, Chang JJ, Khunger A, et al. Minocycline for acute stroke treatment: a systematic review and meta-analysis of randomized clinical trials. J Neurol. 2018;265:1871–1879.

51

Fouda AY, Newsome AS, Spellicy S, et al. Minocycline in acute cerebral hemorrhage: an early phase randomized trial. Stroke. 2017;48:2885–2887.

Brain Hemorrhages
Pages 161-165
Cite this article:
Tang R, Huang Z, Chu H. Phenotype change of polarized microglia after intracerebral hemorrhage: Advances in research. Brain Hemorrhages, 2020, 1(3): 161-165. https://doi.org/10.1016/j.hest.2020.08.001

56

Views

0

Downloads

1

Crossref

2

Web of Science

2

Scopus

Altmetrics

Received: 09 June 2020
Revised: 14 August 2020
Accepted: 19 August 2020
Published: 01 September 2020
© 2020 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