Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2021, Vol. 15 Issue (6) : 887-902    https://doi.org/10.1007/s11684-021-0879-9
RESEARCH ARTICLE
Loss of monocarboxylate transporter 1 aggravates white matter injury after experimental subarachnoid hemorrhage in rats
Xin Wu, Zongqi Wang, Haiying Li, Xueshun Xie, Jiang Wu, Haitao Shen, Xiang Li, Zhong Wang(), Gang Chen()
Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
 Download: PDF(3312 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Monocarboxylic acid transporter 1 (MCT1) maintains axonal function by transferring lactic acid from oligodendrocytes to axons. Subarachnoid hemorrhage (SAH) induces white matter injury, but the involvement of MCT1 is unclear. In this study, the SAH model of adult male Sprague-Dawley rats was used to explore the role of MCT1 in white matter injury after SAH. At 48 h after SAH, oligodendrocyte MCT1 was significantly reduced, and the exogenous overexpression of MCT1 significantly improved white matter integrity and long-term cognitive function. Motor training after SAH significantly increased the number of ITPR2+SOX10+ oligodendrocytes and upregulated the level of MCT1, which was positively correlated with the behavioral ability of rats. In addition, miR-29b and miR-124 levels were significantly increased in SAH rats compared with non-SAH rats. Further intervention experiments showed that miR-29b and miR-124 could negatively regulate the level of MCT1. This study confirmed that the loss of MCT1 may be one of the mechanisms of white matter damage after SAH and may be caused by the negative regulation of miR-29b and miR-124. MCT1 may be involved in the neurological improvement of rehabilitation training after SAH.

Keywords microRNAs      monocarboxylate transporter 1      motor training      subarachnoid hemorrhage      white matter injury     
Corresponding Author(s): Zhong Wang,Gang Chen   
Just Accepted Date: 09 November 2021   Online First Date: 06 December 2021    Issue Date: 27 December 2021
 Cite this article:   
Xin Wu,Zongqi Wang,Haiying Li, et al. Loss of monocarboxylate transporter 1 aggravates white matter injury after experimental subarachnoid hemorrhage in rats[J]. Front. Med., 2021, 15(6): 887-902.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-021-0879-9
https://academic.hep.com.cn/fmd/EN/Y2021/V15/I6/887
Fig.1  Levels of MCT1 were significantly decreased in brain tissues after SAH induction for 48 h. (A) Time course of MCT1 expression in the rat brain tissues after SAH. Top: representative Western blot bands of MCT1 at 3, 6, 12, 24, and 48 h after SAH. Bottom: quantitative analysis of the relative protein level. (B) Mean density (sum integrated optical density/sum pixel area) of MCT1. (C) Double-immunofluorescent analysis was performed with antibodies against MCT1 (green) and Olig2 (oligodendrocyte marker, red), and nuclei were fluorescently labeled with DAPI (blue). Representative images of the expression of MCT1 in rat corpus callosum oligodendrocytes at 48 h after SAH. Scale bar= 100 μm. Data are presented as mean±SD. n = 10 in A, n = 6 in B. *P≤0.05, **P≤0.01 vs. the sham group. N.S., not significant.
Fig.2  Effect of MCT1 overexpression on acute axonal injury after SAH. (A) Efficacy of MCT1 overexpression in brain tissues based on Western blot analysis. Top: representative Western blot bands of MCT1 in the sham, SAH, SAH+ Vector, and SAH+ Over-MCT1 groups. Bottom: quantitative analysis of the relative protein level. (B) Double-immunofluorescent analysis was performed with antibodies against MCT1 (green) and Olig2 (oligodendrocyte marker, red), and nuclei were fluorescently labeled with DAPI (blue). Representative images of the expression of MCT1 in rat corpus callosum oligodendrocytes at 48 h after SAH. Scale bar= 100 μm. (C) Quantification of the relative fluorescence intensity of MCT1. (D) Representative images of β-APP (green) and 200 kDa neurofilament heavy antibody (NF200, red) in the corpus callosum at 48 h after SAH or sham operation. Scale bar= 100 μm. (E) Quantification of the number of β-APP per square millimeter. Data are presented as mean±SD. n = 6 per group. *P≤0.05, ***P≤0.001.
Fig.3  Effect of MCT1 overexpression on acute white matter injury after SAH. (A) Effect of MCT1 overexpression on the level of myelin basic protein (MBP) as shown by Western blot analysis. Top: representative Western blot bands of MBP in the sham, SAH, SAH+ Vector, and SAH+ Over-MCT1 groups. Bottom: quantitative analysis of the relative protein level. (B) Effect of MCT1 overexpression in the level of non-phosphorylated neurofilament H (monoclonal clone ID: SMI 32) based on Western blot analysis. Top: representative Western blot bands of SMI 32 in the sham, SAH, SAH+ Vector, and SAH+ Over-MCT1 groups. Bottom: quantitative analysis of the relative protein level. (C) Representative images of MBP (green) and SMI 32 (red) in the corpus callosum at 48 h after SAH or sham operation. Scale bar= 100 μm. (D) Quantification of the ratios of SMI 32 to MBP fluorescence intensity in the corpus callosum, illustrated as fold change compared with the sham group average value. Data are presented as mean±SD. n = 6 per group. *P≤0.05, ***P≤0.001.
Fig.4  Effect of MCT1 overexpression on SAH-induced long-term neurobehavioral outcomes. (A) Latency to fall in the rotarod test before (3 days of pretraining before SAH) and up to 35 days after SAH. (B) Time to remove the adhesive tapes in the adhesive removal test before (3 days of pretraining before SAH) and up to 35 days after SAH. (C) Escape latency in the Morris water maze test at 29 to 33 days after SAH. (D) Representative images illustrate swim paths on 33 days (learning) and 34 days (memory) after SAH or sham operation. (E) Target quadrant time in the Morris water maze test at 34 days after SAH. (F) Swimming speed in the Morris water maze test at 34 days after SAH. Data are presented as mean±SD. n = 10–12 per group. *P≤0.05, **P≤0.01. N.S., not significant.
Fig.5  Rotarod training increases the level of MCT1 in ITPR2+SOX10+ oligodendrocytes. (A) Representative images of ITPR2+ (green) out of SOX10+ (red) cells; nuclei were fluorescently labeled with DAPI (blue) in the corpus callosum in the sham, sham+ training, SAH, and SAH+ training groups after 2 days of rotarod training. Scale bar= 100 μm. (B) Quantification of ITPR2+ of SOX10+ cells in the corpus callosum. (C) Representative Western blot bands of MCT1 in the corpus callosum region in the sham, sham+ training, SAH, and SAH+ training groups after 7 days of rotarod training. (D) Quantitative analysis of the relative protein level. (E) Representative images of MCT1+ (green) out of SOX10+ (red) cells in the corpus callosum in the sham, sham+ training, SAH, and SAH+ training groups after 7 days of rotarod training. Scale bar= 100 μm. (F) Quantification of MCT1+ out of SOX10+ cells in the corpus callosum. Data are presented as mean±SD. n = 4–6 per group. *P≤0.05, **P≤0.01.
Fig.6  Rotarod training accelerates long-term neurobehavioral recovery after SAH. (A) Correlation between the percentage of MCT1+ out of SOX10+ cells and the latency to fall in the rotarod test on day 35 after sham operation. (B) Correlation between the percentage of MCT1+ out of SOX10+ cells and the latency to fall in the rotarod test on day 35 after SAH operation. (C) Correlation between the percentage of MCT1+ out of SOX10+ cells and the time to remove the adhesive tapes in the adhesive removal test on day 35 after sham operation. (D) Correlation between the percentage of MCT1+ out of SOX10+ cells and the time to remove the adhesive tapes in the adhesive removal test on day 35 after SAH operation. n = 9–10 per group.
Fig.7  Role of miRNA-29b and miRNA-124 in regulating MCT1. (A) Relative expression level of miR-29b-3p in the CSF of SD rats in the sham and SAH groups. (B) Relative expression level of miR-124-3p in the CSF of SD rats in the sham and SAH groups. (C) Top: representative Western blot bands of MCT1 in the corpus callosum region in five different groups at 48 h after experimental SAH. Bottom: quantitative analysis of the relative protein level. (D) Double-immunofluorescent analysis was performed with antibodies against MCT1 (green) and Olig2 (red), and nuclei were fluorescently labeled with DAPI (blue). Representative images of the expressions of MCT1 in rat corpus callosum oligodendrocytes at 48 h after SAH. Scale bar= 100 μm. (E) Quantification of the relative fluorescence intensity of MCT1. (F) Schematic diagram for the role of MCT1 in white matter injury after experimental subarachnoid hemorrhage. After the onset of SAH, a rapid degeneration and demyelination of white matter occurs; however, the decrease of MCT1in oligodendrocytes in the white matter region interferes with the energy supply of oligodendrocytes to axons, thus further aggravating white matter injury. The upregulation of miR-29b and miR-124 may be entirely or partially involved in the decrease of MCT1 after SAH, and motor training might be a promising rescue conversely. Data are presented as mean±SD. n = 6–8 per group. *P≤0.05, **P≤0.01, ***P≤0.001.
1 CVB Hviid, SV Lauridsen, T Gyldenholm, N Sunde, T Parkner, AM Hvas. Plasma neurofilament light chain is associated with poor functional outcome and mortality rate after spontaneous subarachnoid hemorrhage. Transl Stroke Res 2020; 11(4): 671–677
https://doi.org/10.1007/s12975-019-00761-4 pmid: 31808039
2 W Wang, B Jiang, H Sun, X Ru, D Sun, L Wang, L Wang, Y Jiang, Y Li, Y Wang, Z Chen, S Wu, Y Zhang, D Wang, Y Wang, VL; the NESS-China Investigators Feigin. Prevalence, incidence, and mortality of stroke in China: results from a nationwide population-based survey of 480 687 adults. Circulation 2017; 135(8): 759–771
https://doi.org/10.1161/CIRCULATIONAHA.116.025250 pmid: 28052979
3 M Balbi, MJ Vega, A Lourbopoulos, NA Terpolilli, N Plesnila. Long-term impairment of neurovascular coupling following experimental subarachnoid hemorrhage. J Cereb Blood Flow Metab 2020; 40(6): 1193–1202
https://doi.org/10.1177/0271678X19863021 pmid: 31296132
4 H Nishikawa, Y Nakatsuka, M Shiba, F Kawakita, M Fujimoto, H Suzuki; pSEED group. Increased plasma galectin-3 preceding the development of delayed cerebral infarction and eventual poor outcome in non-severe aneurysmal subarachnoid hemorrhage. Transl Stroke Res 2018; 9(2): 110–119
https://doi.org/10.1007/s12975-017-0564-0 pmid: 28831694
5 J Pang, J Peng, P Yang, L Kuai, L Chen, JH Zhang, Y Jiang. White matter injury in early brain injury after subarachnoid hemorrhage. Cell Transplant 2019; 28(1): 26–35
https://doi.org/10.1177/0963689718812054 pmid: 30442028
6 YD Reijmer, MS van den Heerik, R Heinen, A Leemans, J Hendrikse, JB de Vis, LA van der Kleij, C Lucci, ME Hendriks, MJE van Zandvoort, IMC Huenges Wajer, JMA Visser-Meily, GJE Rinkel, GJ Biessels, MDI Vergouwen. Microstructural white matter abnormalities and cognitive impairment after aneurysmal subarachnoid hemorrhage. Stroke 2018; 49(9): 2040–2045
https://doi.org/10.1161/STROKEAHA.118.021622 pmid: 30354997
7 MR Etherton, O Wu, AK Giese, A Lauer, G Boulouis, B Mills, L Cloonan, KL Donahue, W Copen, P Schaefer, NS Rost. White matter integrity and early outcomes after acute ischemic stroke. Transl Stroke Res 2019; 10(6): 630–638
https://doi.org/10.1007/s12975-019-0689-4 pmid: 30693424
8 J Peng, J Pang, L Huang, B Enkhjargal, T Zhang, J Mo, P Wu, W Xu, Y Zuo, J Peng, G Zuo, L Chen, J Tang, JH Zhang, Y Jiang. LRP1 activation attenuates white matter injury by modulating microglial polarization through Shc1/PI3K/Akt pathway after subarachnoid hemorrhage in rats. Redox Biol 2019; 21: 101121
https://doi.org/10.1016/j.redox.2019.101121 pmid: 30703614
9 Y Egashira, Y Hua, RF Keep, T Iwama, G Xi. Lipocalin 2 and blood-brain barrier disruption in white matter after experimental subarachnoid hemorrhage. Acta Neurochir Suppl (Wien) 2016; 121: 131–134
https://doi.org/10.1007/978-3-319-18497-5_23 pmid: 26463936
10 I Micu, JR Plemel, C Lachance, J Proft, AJ Jansen, K Cummins, J van Minnen, PK Stys. The molecular physiology of the axo-myelinic synapse. Exp Neurol 2016; 276: 41–50
https://doi.org/10.1016/j.expneurol.2015.10.006 pmid: 26515690
11 I Micu, JR Plemel, AV Caprariello, KA Nave, PK Stys. Axo-myelinic neurotransmission: a novel mode of cell signalling in the central nervous system. Nat Rev Neurosci 2018; 19(1): 49–58
https://doi.org/10.1038/nrn.2017.128 pmid: 29118449
12 K Pierre, L Pellerin. Monocarboxylate transporters in the central nervous system: distribution, regulation and function. J Neurochem 2005; 94(1): 1–14
https://doi.org/10.1111/j.1471-4159.2005.03168.x pmid: 15953344
13 JE Rinholm, NB Hamilton, N Kessaris, WD Richardson, LH Bergersen, D Attwell. Regulation of oligodendrocyte development and myelination by glucose and lactate. J Neurosci 2011; 31(2): 538–548
https://doi.org/10.1523/JNEUROSCI.3516-10.2011 pmid: 21228163
14 ED Ponomarev, T Veremeyko, HL Weiner. MicroRNAs are universal regulators of differentiation, activation, and polarization of microglia and macrophages in normal and diseased CNS. Glia 2013; 61(1): 91–103
https://doi.org/10.1002/glia.22363 pmid: 22653784
15 TJ Pullen, G da Silva Xavier, G Kelsey, GA Rutter. miR-29a and miR-29b contribute to pancreatic β-cell-specific silencing of monocarboxylate transporter 1 (Mct1). Mol Cell Biol 2011; 31(15): 3182–3194
https://doi.org/10.1128/MCB.01433-10 pmid: 21646425
16 SY Xu, XL Jiang, Q Liu, J Xu, J Huang, SW Gan, WT Lu, F Zhuo, M Yang, SQ Sun. Role of rno-miR-124-3p in regulating MCT1 expression in rat brain after permanent focal cerebral ischemia. Genes Dis 2019; 6(4): 398–406
https://doi.org/10.1016/j.gendis.2019.01.002 pmid: 31832520
17 M Ziu, L Fletcher, S Rana, DF Jimenez, M Digicaylioglu. Temporal differences in microRNA expression patterns in astrocytes and neurons after ischemic injury. PLoS One 2011; 6(2): e14724
https://doi.org/10.1371/journal.pone.0014724 pmid: 21373187
18 Q Ji, Y Ji, J Peng, X Zhou, X Chen, H Zhao, T Xu, L Chen, Y Xu. Increased brain-specific miR-9 and miR-124 in the serum exosomes of acute ischemic stroke patients. PLoS One 2016; 11(9): e0163645
https://doi.org/10.1371/journal.pone.0163645 pmid: 27661079
19 TB Massoto, ACR Santos, BS Ramalho, FM Almeida, AMB Martinez, SA Marques. Mesenchymal stem cells and treadmill training enhance function and promote tissue preservation after spinal cord injury. Brain Res 2020; 1726: 146494
https://doi.org/10.1016/j.brainres.2019.146494 pmid: 31586628
20 T Sun, C Ye, Z Zhang, J Wu, H Huang. Cotransplantation of olfactory ensheathing cells and Schwann cells combined with treadmill training promotes functional recovery in rats with contused spinal cords. Cell Transplant 2013; 22(Suppl 1): S27–S38
https://doi.org/10.3727/096368913X672118 pmid: 24044361
21 S Marques, A Zeisel, S Codeluppi, D van Bruggen, A Mendanha Falcão, L Xiao, H Li, M Häring, H Hochgerner, RA Romanov, D Gyllborg, A Muñoz Manchado, G La Manno, P Lönnerberg, EM Floriddia, F Rezayee, P Ernfors, E Arenas, J Hjerling-Leffler, T Harkany, WD Richardson, S Linnarsson, G Castelo-Branco. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 2016; 352(6291): 1326–1329
https://doi.org/10.1126/science.aaf6463 pmid: 27284195
22 K Hamano, T Takeya, N Iwasaki, J Nakayama, T Ohto, Y Okada. A quantitative study of the progress of myelination in the rat central nervous system, using the immunohistochemical method for proteolipid protein. Brain Res Dev Brain Res 1998; 108(1–2): 287–293
https://doi.org/10.1016/S0165-3806(98)00063-7 pmid: 9693804
23 Y Dou, H Shen, D Feng, H Li, X Tian, J Zhang, Z Wang, G Chen. Tumor necrosis factor receptor-associated factor 6 participates in early brain injury after subarachnoid hemorrhage in rats through inhibiting autophagy and promoting oxidative stress. J Neurochem 2017; 142(3): 478–492
https://doi.org/10.1111/jnc.14075 pmid: 28543180
24 T Sugawara, R Ayer, V Jadhav, JH Zhang. A new grading system evaluating bleeding scale in filament perforation subarachnoid hemorrhage rat model. J Neurosci Methods 2008; 167(2): 327–334
https://doi.org/10.1016/j.jneumeth.2007.08.004 pmid: 17870179
25 H Shen, Z Chen, Y Wang, A Gao, H Li, Y Cui, L Zhang, X Xu, Z Wang, G Chen. Role of neurexin-1β and neuroligin-1 in cognitive dysfunction after subarachnoid hemorrhage in rats. Stroke 2015; 46(9): 2607–2615
https://doi.org/10.1161/STROKEAHA.115.009729 pmid: 26219651
26 P Zhang, T Wang, D Zhang, Z Zhang, S Yuan, J Zhang, J Cao, H Li, X Li, H Shen, G Chen. Exploration of MST1-mediated secondary brain injury induced by intracerebral hemorrhage in rats via Hippo signaling pathway. Transl Stroke Res 2019; 10(6): 729–743
https://doi.org/10.1007/s12975-019-00702-1 pmid: 30941717
27 H Zhao, G Li, S Zhang, F Li, R Wang, Z Tao, Q Ma, Z Han, F Yan, J Fan, L Li, X Ji, Y Luo. Inhibition of histone deacetylase 3 by miR-494 alleviates neuronal loss and improves neurological recovery in experimental stroke. J Cereb Blood Flow Metab 2019; 39(12): 2392–2405
https://doi.org/10.1177/0271678X19875201 pmid: 31510852
28 L Mao, T Yang, X Li, X Lei, Y Sun, Y Zhao, W Zhang, Y Gao, B Sun, F Zhang. Protective effects of sulforaphane in experimental vascular cognitive impairment: contribution of the Nrf2 pathway. J Cereb Blood Flow Metab 2019; 39(2): 352–366
https://doi.org/10.1177/0271678X18764083 pmid: 29533123
29 V Bouet, M Boulouard, J Toutain, D Divoux, M Bernaudin, P Schumann-Bard, T Freret. The adhesive removal test: a sensitive method to assess sensorimotor deficits in mice. Nat Protoc 2009; 4(10): 1560–1564
https://doi.org/10.1038/nprot.2009.125 pmid: 19798088
30 ZF Deng, HL Zheng, JG Chen, Y Luo, JF Xu, G Zhao, JJ Lu, HH Li, SQ Gao, DZ Zhang, LQ Zhu, YH Zhang, F Wang. miR-214-3p targets β-catenin to regulate depressive-like behaviors induced by chronic social defeat stress in mice. Cereb Cortex 2019; 29(4): 1509–1519
https://doi.org/10.1093/cercor/bhy047 pmid: 29522177
31 J Zhao, H Mu, L Liu, X Jiang, D Wu, Y Shi, RK Leak, X Ji. Transient selective brain cooling confers neurovascular and functional protection from acute to chronic stages of ischemia/reperfusion brain injury. J Cereb Blood Flow Metab 2019; 39(7): 1215–1231
https://doi.org/10.1177/0271678X18808174 pmid: 30334662
32 X Dai, J Chen, F Xu, J Zhao, W Cai, Z Sun, TK Hitchens, LM Foley, RK Leak, J Chen, X Hu. TGFα preserves oligodendrocyte lineage cells and improves white matter integrity after cerebral ischemia. J Cereb Blood Flow Metab 2020; 40(3): 639–655
https://doi.org/10.1177/0271678X19830791 pmid: 30834805
33 H Shi, X Hu, RK Leak, Y Shi, C An, J Suenaga, J Chen, Y Gao. Demyelination as a rational therapeutic target for ischemic or traumatic brain injury. Exp Neurol 2015; 272: 17–25
https://doi.org/10.1016/j.expneurol.2015.03.017 pmid: 25819104
34 Y Lee, BM Morrison, Y Li, S Lengacher, MH Farah, PN Hoffman, Y Liu, A Tsingalia, L Jin, PW Zhang, L Pellerin, PJ Magistretti, JD Rothstein. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature 2012; 487(7408): 443–448
https://doi.org/10.1038/nature11314 pmid: 22801498
35 AS Wahl, W Omlor, JC Rubio, JL Chen, H Zheng, A Schröter, M Gullo, O Weinmann, K Kobayashi, F Helmchen, B Ommer, ME Schwab. Asynchronous therapy restores motor control by rewiring of the rat corticospinal tract after stroke. Science 2014; 344(6189): 1250–1255
https://doi.org/10.1126/science.1253050 pmid: 24926013
36 BA Brody, HC Kinney, AS Kloman, FH Gilles. Sequence of central nervous system myelination in human infancy. I. An autopsy study of myelination. J Neuropathol Exp Neurol 1987; 46(3): 283–301
https://doi.org/10.1097/00005072-198705000-00005 pmid: 3559630
37 HC Kinney, BA Brody, AS Kloman, FH Gilles. Sequence of central nervous system myelination in human infancy. II. Patterns of myelination in autopsied infants. J Neuropathol Exp Neurol 1988; 47(3): 217–234
https://doi.org/10.1097/00005072-198805000-00003 pmid: 3367155
38 BF Olkowski, MA Devine, LE Slotnick, E Veznedaroglu, KM Liebman, ML Arcaro, MJ Binning. Safety and feasibility of an early mobilization program for patients with aneurysmal subarachnoid hemorrhage. Phys Ther 2013; 93(2): 208–215
https://doi.org/10.2522/ptj.20110334 pmid: 22652987
39 S Lengacher, T Nehiri-Sitayeb, N Steiner, L Carneiro, C Favrod, F Preitner, B Thorens, JC Stehle, L Dix, F Pralong, PJ Magistretti, L Pellerin. Resistance to diet-induced obesity and associated metabolic perturbations in haploinsufficient monocarboxylate transporter 1 mice. PLoS One 2013; 8(12): e82505
https://doi.org/10.1371/journal.pone.0082505 pmid: 24367518
40 B Chatel, D Bendahan, C Hourdé, L Pellerin, S Lengacher, P Magistretti, Y Le Fur, C Vilmen, M Bernard, LA Messonnier. Role of MCT1 and CAII in skeletal muscle pH homeostasis, energetics, and function: in vivo insights from MCT1 haploinsufficient mice. FASEB J 2017; 31(6): 2562–2575
https://doi.org/10.1096/fj.201601259R pmid: 28254758
41 M Zhang, Z Ma, H Qin, Z Yao. Monocarboxylate transporter 1 in the medial prefrontal cortex developmentally expresses in oligodendrocytes and associates with neuronal amounts. Mol Neurobiol 2017; 54(3): 2315–2326
https://doi.org/10.1007/s12035-016-9820-7 pmid: 26957300
42 S Wu, Y Lin, D Xu, J Chen, M Shu, Y Zhou, W Zhu, X Su, Y Zhou, P Qiu, G Yan. miR-135a functions as a selective killer of malignant glioma. Oncogene 2012; 31(34): 3866–3874
https://doi.org/10.1038/onc.2011.551 pmid: 22139076
43 F Li, MW Zhou, N Liu, YY Yang, HY Xing, Y Lu, XX Liu. MicroRNA-219 inhibits proliferation and induces differentiation of oligodendrocyte precursor cells after contusion spinal cord injury in rats. Neural Plast 2019; 2019: 9610687
https://doi.org/10.1155/2019/9610687 pmid: 30911293
44 Y Toyota, J Wei, G Xi, RF Keep, Y Hua. White matter T2 hyperintensities and blood-brain barrier disruption in the hyperacute stage of subarachnoid hemorrhage in male mice: the role of lipocalin-2. CNS Neurosci Ther 2019; 25(10): 1207–1214
https://doi.org/10.1111/cns.13221 pmid: 31568658
45 J Li, MC Chan, Y Yu, Y Bei, P Chen, Q Zhou, L Cheng, L Chen, O Ziegler, GC Rowe, S Das, J Xiao. miR-29b contributes to multiple types of muscle atrophy. Nat Commun 2017; 8: 15201
https://doi.org/10.1038/ncomms15201 pmid: 28541289
46 WD Bao, XT Zhou, LT Zhou, F Wang, X Yin, Y Lu, LQ Zhu, D Liu. Targeting miR-124/ferroportin signaling ameliorated neuronal cell death through inhibiting apoptosis and ferroptosis in aged intracerebral hemorrhage murine model. Aging Cell 2020; 19(11): e13235
https://doi.org/10.1111/acel.13235 pmid: 33068460
47 A Gupta, L Pulliam. Exosomes as mediators of neuroinflammation. J Neuroinflammation 2014; 11: 68
https://doi.org/10.1186/1742-2094-11-68 pmid: 24694258
[1] FMD-21033-OF-CG_suppl_1 Download
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed