Early treatment with minocycline following stroke in rats improves functional recovery and differentially modifies responses of peri-infarct microglia and astrocytes

Wai Ping Yew, Natalia D Djukic, Jaya S P Jayaseelan, Frederick R Walker, Karl A A Roos, Timothy K Chataway, Hakan Muyderman, Neil R Sims, Wai Ping Yew, Natalia D Djukic, Jaya S P Jayaseelan, Frederick R Walker, Karl A A Roos, Timothy K Chataway, Hakan Muyderman, Neil R Sims

Abstract

Background: Altered neuronal connectivity in peri-infarct tissue is an important contributor to both the spontaneous recovery of neurological function that commonly develops after stroke and improvements in recovery that have been induced by experimental treatments in animal models. Microglia and astrocytes are primary determinants of the environment in peri-infarct tissue and hence strongly influence the potential for neuronal plasticity. However, the specific roles of these cells and the timing of critical changes in their function are not well understood. Minocycline can protect against ischemic damage and promote recovery. These effects are usually attributed, at least partially, to the ability of this drug to suppress microglial activation. This study tested the ability of minocycline treatment early after stroke to modify reactive responses in microglia and astrocytes and improve recovery.

Methods: Stroke was induced by photothrombosis in the forelimb sensorimotor cortex of Sprague-Dawley rats. Minocycline was administered for 2 days after stroke induction and the effects on forelimb function assessed up to 28 days. The responses of peri-infarct Iba1-positive cells and astrocytes were evaluated using immunohistochemistry and Western blots.

Results: Initial characterization showed that the numbers of Iba1-positive microglia and macrophages decreased in peri-infarct tissue at 24 h then increased markedly over the next few days. Morphological changes characteristic of activation were readily apparent by 3 h and increased by 24 h. Minocycline treatment improved the rate of recovery of motor function as measured by a forelimb placing test but did not alter infarct volume. At 3 days, there were only minor effects on core features of peri-infarct microglial reactivity including the morphological changes and increased density of Iba1-positive cells. The treatment caused a decrease of 57% in the small subpopulation of cells that expressed CD68, a marker of phagocytosis. At 7 days, the expression of glial fibrillary acidic protein and vimentin was markedly increased by minocycline treatment, indicating enhanced reactive astrogliosis.

Conclusions: Early post-stroke treatment with minocycline improved recovery but had little effect on key features of microglial activation. Both the decrease in CD68-positive cells and the increased activation of astrogliosis could influence neuronal plasticity and contribute to the improved recovery.

Keywords: Astrocytes; Focal ischemia; Functional recovery; Microglia; Minocycline; Peri-infarct; Photothrombosis; Stroke.

Conflict of interest statement

Ethics approval and consent to participate

All studies involving rats were conducted according to the “Australian code for the care and use of animals for scientific purposes, 2013” published by the National Health and Medical Research Council, Australia. The investigations were approved by the Animal Welfare Committee of Flinders University, Adelaide, South Australia (Project number 754/10).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Summary of experimental design for the three major phases of the study. a Characterization of responses of Iba1-positive cells in peri-infarct tissue during the first week following photothrombotic stroke. The numbers of rats shown were all assessed using the forelimb placing test. Perfusion-fixed tissue from four to six rats per time point was used to assess infarct volume and that from four rats per time point was used for immunohistochemistry. b Effects of lower-dose minocycline treatment on forelimb function and cellular changes during the first week following photothrombotic stroke. c Effects of higher-dose minocycline treatment on forelimb function (up to 28 days) and cellular changes following photothrombotic stroke
Fig. 2
Fig. 2
Tissue damage and functional changes following photothrombotic stroke. a Cresyl-violet stained coronal section of a rat brain at 24 h after induction of photothrombotic stroke showing the clearly defined infarct and the ROIs used for analysis of sections immunolabeled to detect Iba1. The scale is in millimeters. b Cresyl-violet stained coronal section at 3 h after stroke induction. c Infarct volume over 7 days after stroke induction. The infarct is fully developed by 24 h and then contracts substantially by 7 days (n = 4–6 per time point). d Coronal section of cerebral cortex immunolabeled for NeuN at 3 h after stroke induction (scale bar = 500 μm). e Box plot of forelimb placing scores following stroke. Outlier values are shown as circles; Blue = forelimb ipsilateral to infarct, Red = forelimb contralateral to infarct (n ≥ 5 per group)
Fig. 3
Fig. 3
Circularity of Iba1-positive cells after photothrombotic stroke. This parameter was measured in ROIs as defined in Fig. 2a. a Comparison of Iba1-immunolabeled cells in peri-infarct tissue (Ipsi01A) and equivalent tissue (Contra01) in the hemisphere contralateral to the infarct at 3 h after stroke induction. The scale bar = 100 μm. b Regional differences at 3 h. Insets show examples of processed images of Iba1-positive cells at an ROI adjacent to the infarct (Ipsi01A) and in corresponding contralateral tissue (Contra01). c Regional differences at 24 h. Inset shows an example of a processed image at Ipsi01A. d Time course of changes in peri-infarct regions (Ipsi01A and Ipsi01B), tissue 1 mm from the infarct (ipsi01A + 1 mm) and corresponding contralateral tissue (Contra01) over the first 7 days. For panels bd, n = 4 for each data point; *p < 0.05, **p < 0.01, ***p < 0.001 compared with the corresponding contralateral region (one-way ANOVA with Tukey’s HSD post-hoc test)
Fig. 4
Fig. 4
Additional morphological features of Iba1-positive cells at 24 h and 7 days after photothrombotic stroke. a Cell area, b cell perimeter, and c cell soma area. For panels ac, n = 3 (7 days) or 4 (24 h); *p < 0.05, **p < 0.01, ***p < 0.001 compared with the corresponding contralateral region, contra01 (one-way ANOVA with Tukey’s HSD post-hoc test). The ROIs that were analyzed are identified in Fig. 2a
Fig. 5
Fig. 5
Changes in the pattern of Iba1-immunolabeling after photothrombotic stroke. a Regional differences in area fraction at 24 h. b–d Changes in b area fraction, c Iba1-positive particle count, and d Iba1-positive particle size in peri-infarct tissue, tissue 1 mm from the infarct and corresponding contralateral tissue between 3 h and 7 days. For all panels, n = 4 per data point; *p < 0.05, **p < 0.01, ***p < 0.001 compared with Contra01 (one-way ANOVA with Tukey’s HSD post-hoc test). At 24 h, there was also a significant difference between each of the peri-infarct regions and tissue located 1 mm from the infarct (at ipsi01a + 1 mm) for area fraction (Ipsi01a: p < 0.05; Ipsi01b: p < 0.01) and particle count (p < 0.01 for each of the peri-infarct regions). The ROIs that were analyzed are identified in Fig. 2a
Fig. 6
Fig. 6
Effects of lower dose minocycline treatment on infarct volume and forelimb function after photothrombotic stroke. a Infarct volume: Two-way analysis of variance revealed no significant effect of the minocycline treatment but a significant effect of time after stroke (p < 0.01; n = 4–8 per group). b Box plot of results from the forelimb placing test. Outliers are indicated with open circles. n = 6 per group for all times except 3 h which shows the pooled data for rats in each of the subsequent groups. Rate of recovery was faster at 3 days *p < 0.05 (Minocycline vs vehicle, Mann-Whitney U test analysis of area under curve for those rats tested at both 3 h and 3 days). No significant differences were detected at other times
Fig. 7
Fig. 7
Effects of lower dose minocycline treatment on responses of Iba1-positive cells after photothrombotic stroke. a Circularity and b area fraction of Iba1-positive cells at 24 h, 3 days and 7 days following photothrombotic stroke (n = 4 to 8 per group). Two-way analysis of variance detected highly significant differences between the contralateral and peri-infarct regions for both circularity and area fraction at all three time points (p < 0.001). However, there were no significant effects of treatment on these parameters
Fig. 8
Fig. 8
Effect of higher-dose minocycline treatment on recovery of neurological function after photothrombotic stroke. Box plots of dysfunction and recovery up to 28 days after photothrombotic stroke in a the forelimb placing test and b the cylinder test. Outliers are shown as open circles (n = 8 per group). Recovery in the forelimb placing test was more rapid in minocycline-treated rats (p < 0.05; Mann-Whitney U test comparison of area under curve for time points between 3 h and 28 days.) There was no significant difference in recovery between 24 h and 28 days for performance in the cylinder test
Fig. 9
Fig. 9
Effect of higher-dose minocycline treatment on Iba1-positive cells at 3 days after photothrombotic stroke. a Circularity and b area fraction of immunolabeling (n = 6 to 7 per group). Two-way analysis of variance revealed a statistically significant difference between the hemispheres for both circularity and area fraction (p < 0.01) and a significant effect of the minocycline treatment on Iba1 area fraction (p < 0.05) but not on circularity. For area fraction, there was no significant interaction between hemisphere and minocycline treatment
Fig. 10
Fig. 10
Effect of higher-dose minocycline treatment on CD68-immunolabeled cells at 3 days after photothrombotic stroke. a Representative image of CD68 (red) and NeuN (green) double-immunolabeled coronal section at the infarct boundary of a vehicle-treated rat. The infarct is visible in the upper part of the image. The scale bar = 200 μm. b Effects of the higher-dose minocycline treatment on CD68-positive cells within the peri-infarct tissue at 3 days following photothrombotic stroke (n = 6 to 7 per group). The CD68-positive particle count was assessed within 0.3 mm of the infarct (Peri-infarct) and between 0.3 and 0.6 mm from the infarct (Peri-infarct + 0.3 mm). Two-way analysis of variance revealed a highly significant effect of both treatment (p < 0.01) and tissue location (p < 0.001) on CD68-positive particle count at 3 days after stroke. There was also a strong interaction between the two factors (p < 0.001) reflecting the larger decrease in particle density induced by minocycline in the tissue immediately adjacent to the infarct compared with the other regions examined
Fig. 11
Fig. 11
Effects of higher-dose minocycline treatment on markers of reactive astrogliosis after photothrombotic stroke. a, b A large increase in vimentin content in peri-infarct tissue over the first 7 days following photothrombotic stroke in untreated rats was detected by a area fraction of vimentin-positive cells and b Western blotting. The area fraction of vimentin immunolabeling was markedly increased by 2 days and further increased at 3 and 7 days after stroke (n = 4 per group at each time); *p < 0.05, **p < 0.01 vs. corresponding contralateral region (one-way ANOVA with Tukey’s HSD). c Double immunolabeling of GFAP and vimentin in peri-infarct tissue at 3 days after photothrombotic stroke. Vimentin (red) is expressed almost exclusively in cells also expressing GFAP (green). Yellow shows colocalization. The scale bar = 100 μm. de Effect of higher-dose minocycline treatment on expression of astroglial proteins in samples containing infarct plus peri-infarct tissue at 7 days following photothrombotic stroke (n = 6 per group). d The intermediate filament proteins, vimentin and GFAP. e Neurocan. The band intensities for vimentin (54 kDa) and GFAP (50 kDa) were normalized to actin (45 kDa). There was a significant effect of minocycline treatment on both vimentin and GFAP content (**p < 0.01). The intensity of bands for full-length neurocan (approx. 250 kDa) were determined relative to that of a proteolytic fragment of this protein (approx. 150 kDa). No statistically significant effects were detected in the neurocan protein ratio in tissue from the minocycline-treated rats compared with the vehicle-treated rats

References

    1. Cramer SC. Repairing the human brain after stroke: I. mechanisms of spontaneous recovery. Ann Neurol. 2008;63:272–287. doi: 10.1002/ana.21393.
    1. Murphy TH, Corbett D. Plasticity during stroke recovery: from synapse to behaviour. Nat Rev Neurosci. 2009;10:861–872. doi: 10.1038/nrn2735.
    1. Carmichael ST, Kathirvelu B, Schweppe CA, Nie EH. Molecular, cellular and functional events in axonal sprouting after stroke. Exp Neurol. 2017;287:384–394. doi: 10.1016/j.expneurol.2016.02.007.
    1. Pekna M, Pekny M, Nilsson M. Modulation of neural plasticity as a basis for stroke rehabilitation. Stroke. 2012;43:2819–2828. doi: 10.1161/STROKEAHA.112.654228.
    1. Benowitz LI, Carmichael ST. Promoting axonal rewiring to improve outcome after stroke. Neurobiol Dis. 2010;37:259–266. doi: 10.1016/j.nbd.2009.11.009.
    1. Hermann DM, Chopp M. Promoting brain remodelling and plasticity for stroke recovery: therapeutic promise and potential pitfalls of clinical translation. Lancet Neurol. 2012;11:369–380. doi: 10.1016/S1474-4422(12)70039-X.
    1. Anttila JE, Whitaker KW, Wires ES, Harvey BK, Airavaara M. Role of microglia in ischemic focal stroke and recovery: focus on toll-like receptors. Prog Neuro-Psychopharmacol Biol Psychiatry. 2017;79:3–14. doi: 10.1016/j.pnpbp.2016.07.003.
    1. Morrison HW, Filosa JA. A quantitative spatiotemporal analysis of microglia morphology during ischemic stroke and reperfusion. J Neuroinflammation. 2013;10:4. doi: 10.1186/1742-2094-10-4.
    1. Schroeter M, Jander S, Stoll G. Non-invasive induction of focal cerebral ischemia in mice by photothrombosis of cortical microvessels: characterization of inflammatory responses. J Neurosci Methods. 2002;117:43–49. doi: 10.1016/S0165-0270(02)00072-9.
    1. Yenari MA, Kauppinen TM, Swanson RA. Microglial activation in stroke: therapeutic targets. Neurotherapeutics. 2010;7:378–91.
    1. Ma YY, Wang JX, Wang YT, Yang GY. The biphasic function of microglia in ischemic stroke. Prog Neurobiol. 2017;157:247–272. doi: 10.1016/j.pneurobio.2016.01.005.
    1. Liu ZW, Chopp M. Astrocytes, therapeutic targets for neuroprotection and neurorestoration in ischemic stroke. Prog Neurobiol. 2016;144:103–120. doi: 10.1016/j.pneurobio.2015.09.008.
    1. Pekny M, Pekna M. Reactive gliosis in the pathogenesis of CNS diseases. Biochim Biophys Acta. 2016;1862:483–491. doi: 10.1016/j.bbadis.2015.11.014.
    1. Burda JE, Sofroniew MV. Reactive gliosis and the multicellular response to CNS damage and disease. Neuron. 2014;81:229–248. doi: 10.1016/j.neuron.2013.12.034.
    1. Sofroniew MV. Astrocyte barriers to neurotoxic inflammation. Nat Rev Neurosci. 2015;16:249–263. doi: 10.1038/nrn3898.
    1. Sims NR, Yew WP. Reactive astrogliosis in stroke: contributions of astrocytes to recovery of neurological function. Neurochem Int. 2017;107:88–103. doi: 10.1016/j.neuint.2016.12.016.
    1. Li T, Pang SR, Yu YP, Wu XQ, Guo J, Zhang SX. Proliferation of parenchymal microglia is the main source of microgliosis after ischaemic stroke. Brain. 2013;136:3578–3588. doi: 10.1093/brain/awt287.
    1. Schilling M, Besselmann M, Muller M, Strecker JK, Ringelstein EB, Kiefer R. Predominant phagocytic activity of resident microglia over hematogenous macrophages following transient focal cerebral ischemia: an investigation using green fluorescent protein transgenic bone marrow chimeric mice. Exp Neurol. 2005;196:290–297. doi: 10.1016/j.expneurol.2005.08.004.
    1. Zhou W, Liesz A, Bauer H, Sommer C, Lahrmann B, Valous N, Grabe N, Veltkamp R. Postischemic brain infiltration of leukocyte subpopulations differs among murine permanent and transient focal cerebral ischemia models. Brain Pathol. 2013;23:34–44. doi: 10.1111/j.1750-3639.2012.00614.x.
    1. Yrjanheikki J, Tikka T, Keinanen R, Goldsteins G, Chan PH, Koistinaho J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci U S A. 1999;96:13496–13500. doi: 10.1073/pnas.96.23.13496.
    1. Yenari MA, Xu L, Tang XN, Qiao Y, Giffard RG. Microglia potentiate damage to blood-brain barrier constituents: improvement by minocycline in vivo and in vitro. Stroke. 2006;37:1087–1093. doi: 10.1161/.
    1. Franco EC, Cardoso MM, Gouveia A, Pereira A, Gomes-Leal W. Modulation of microglial activation enhances neuroprotection and functional recovery derived from bone marrow mononuclear cell transplantation after cortical ischemia. Neurosci Res. 2012;73:122–132. doi: 10.1016/j.neures.2012.03.006.
    1. Yang YR, Salayandia VM, Thompson JF, Yang LY, Estrada EY, Yang Y. Attenuation of acute stroke injury in rat brain by minocycline promotes blood-brain barrier remodeling and alternative microglia/macrophage activation during recovery. J Neuroinflammation. 2015;12:26. doi: 10.1186/s12974-015-0245-4.
    1. Li J, McCullough LD. Sex differences in minocycline-induced neuroprotection after experimental stroke. J Cereb Blood Flow Metab. 2009;29:670–674. doi: 10.1038/jcbfm.2009.3.
    1. Chu LS, Fang SH, Zhou Y, Yin YJ, Chen WY, Li JH, Sun J, Wang ML, Zhang WP, Wei EQ. Minocycline inhibits 5-lipoxygenase expression and accelerates functional recovery in chronic phase of focal cerebral ischemia in rats. Life Sci. 2010;86:170–177. doi: 10.1016/j.lfs.2009.12.001.
    1. Liebigt S, Schlegel N, Oberland J, Witte OW, Redecker C, Keiner S. Effects of rehabilitative training and anti-inflammatory treatment on functional recovery and cellular reorganization following stroke. Exp Neurol. 2012;233:776–782. doi: 10.1016/j.expneurol.2011.11.037.
    1. Liu ZY, Fan Y, Won SJ, Neumann M, Hu DZ, Zhou LF, Weinstein PR, Liu J. Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke. 2007;38:146–152. doi: 10.1161/.
    1. Garrido-Mesa N, Zarzuelo A, Galvez J. Minocycline: far beyond an antibiotic. Br J Pharmacol. 2013;169:337–352. doi: 10.1111/bph.12139.
    1. Alaverdashvili M, Moon SK, Beckman CD, Virag A, Whishaw IQ. Acute but not chronic differences in skilled reaching for food following motor cortex devascularization vs. photothrombotic stroke in the rat. Neuroscience. 2008;157:297–308. doi: 10.1016/j.neuroscience.2008.09.015.
    1. Watson BD, Dietrich WD, Busto R, Wachtel MS, Ginsberg MD. Induction of reproducible brain infarction by photochemically initiated thrombosis. Ann Neurol. 1985;17:497–504. doi: 10.1002/ana.410170513.
    1. Carmichael ST. Rodent models of focal stroke: size, mechanism, and purpose. NeuroRx. 2005;2:396–409. doi: 10.1602/neurorx.2.3.396.
    1. Schroeter M, Schiene K, Kraemer M, Hagemann G, Weigel H, Eysel UT, Witte OW, Stoll G. Astroglial responses in photochemically induced focal ischemia of the rat cortex. Exp Brain Res. 1995;106:1–6. doi: 10.1007/BF00241351.
    1. Schroeter M, Jander S, Witte OW, Stoll G. Heterogeneity of the microglial response in photochemically induced focal ischemia of the rat cerebral cortex. Neuroscience. 1999;89:1367–1377. doi: 10.1016/S0306-4522(98)00398-4.
    1. Nowicka D, Rogozinska K, Aleksy M, Witte OW, Skangiel-Kramska J. Spatiotemporal dynamics of astroglial and microglial responses after photothrombotic stroke in the rat brain. Acta Neurobiol Exp. 2008;68:155–168.
    1. Li HL, Zhang NN, Lin HY, Yu Y, Cai QY, Ma LX, Ding SH. Histological, cellular and behavioral assessments of stroke outcomes after photothrombosis-induced ischemia in adult mice. BMC Neurosci. 2014;15:58. doi: 10.1186/1471-2202-15-58.
    1. Cardoso MM, Franco EC, de Souza CC, da Silva MC, Gouveia A, Gomes-Leal W. Minocycline treatment and bone marrow mononuclear cell transplantation after endothelin-1 induced striatal ischemia. Inflammation. 2013;36:197–205. doi: 10.1007/s10753-012-9535-5.
    1. Kim BJ, Kim MJ, Park JM, Lee SH, Kim YJ, Ryu S, Kim YH, Yoon BW. Reduced neurogenesis after suppressed inflammation by minocycline in transient cerebral ischemia in rat. J Neurol Sci. 2009;279:70–75. doi: 10.1016/j.jns.2008.12.025.
    1. Schallert T, Woodlee MT. Orienting and placing. In: Whishaw IQ, Kolb B, editors. The behavior of the laboratory rat. Oxford: Oxford University Press; 2005. pp. 129–140.
    1. Schallert T, Fleming SM, Leasure JL, Tillerson JL, Bland ST. CNS plasticity and assessment of forelimb sensorimotor outcome in unilateral rat models of stroke, cortical ablation, parkinsonism and spinal cord injury. Neuropharmacology. 2000;39:777–787. doi: 10.1016/S0028-3908(00)00005-8.
    1. Metz GA, Kolb B, Whishaw IQ. Neuropsychological tests. In: Whishaw IQ, Kolb B, editors. The behavior of the laboratory rat. Oxford: Oxford University Press; 2005. pp. 475–498.
    1. Rasband WS: ImageJ. U. S. National Institutes of Health, Bethesda, Maryland, USA. 1997-2018.
    1. Kongsui R, Johnson SJ, Graham BA, Nilsson M, Walker FR. A combined cumulative threshold spectra and digital reconstruction analysis reveal structural alterations of microglia within the prefrontal cortex following low-dose LPS administration. Neuroscience. 2015;310:629–640. doi: 10.1016/j.neuroscience.2015.09.061.
    1. Harris NG, Carmichael ST, Hovda DA, Sutton RL. Traumatic brain injury results in disparate regions of chondroitin sulfate proteoglycan expression that are temporally limited. J Neurosci Res. 2009;87:2937–2950. doi: 10.1002/jnr.22115.
    1. Matsui F, Watanabe E, Oohira A. Immunological identification of 2 proteoglycan fragments derived from neurocan, a brain-specific chondroitin sulfate proteoglycan. Neurochem Int. 1994;25:425–431. doi: 10.1016/0197-0186(94)90018-3.
    1. Asher RA, Morgenstern DA, Fidler PS, Adcock KH, Oohira A, Braistead JE, Levine JM, Margolis RU, Rogers JH, Fawcett JW. Neurocan is upregulated in injured brain and in cytokine-treated astrocytes. J Neurosci. 2000;20:2427–2438. doi: 10.1523/JNEUROSCI.20-07-02427.2000.
    1. Deguchi K, Takaishi M, Hayashi T, Oohira A, Nagotani S, Li F, Jin G, Nagano I, Shoji M, Miyazaki M, et al. Expression of neurocan after transient middle cerebral artery occlusion in adult rat brain. Brain Res. 2005;1037:194–199. doi: 10.1016/j.brainres.2004.12.016.
    1. Matsui F, Kawashima S, Shuo T, Yamauchi S, Tokita Y, Aono S, Keino H, Oohira A. Transient expression of juvenile-type neurocan by reactive astrocytes in adult rat brains injured by kainate-induced seizures as well as surgical incision. Neuroscience. 2002;112:773–781. doi: 10.1016/S0306-4522(02)00136-7.
    1. Petito CK, Morgello S, Felix JC, Lesser ML. The 2 patterns of reactive astrocytosis in postischemic rat-brain. J Cereb Blood Flow Metab. 1990;10:850–859. doi: 10.1038/jcbfm.1990.141.
    1. Korzhevskii DE, Lentsman MV, Kirik OV, Otellin VA. Vimentin-immunopositive cells in the rat telencephalon after experimental ischemic stroke. Neurosci Behav Physiol. 2008;38:845. doi: 10.1007/s11055-008-9061-y.
    1. Lampl Y, Boaz M, Gilad R, Lorberboym M, Dabby R, Rapoport A, Anca-Hershkowitz M, Sadeh M. Minocycline treatment in acute stroke - an open-label, evaluator-blinded study. Neurology. 2007;69:1404–1410. doi: 10.1212/01.wnl.0000277487.04281.db.
    1. Madinier A, Quattromani MJ, Sjolund C, Ruscher K, Wieloch T. Enriched housing enhances recovery of limb placement ability and reduces aggrecan-containing perineuronal nets in the rat somatosensory cortex after experimental stroke. PLoS One. 2014;9:e93121. doi: 10.1371/journal.pone.0093121.
    1. Schroeter M, Jander S, Huitinga I, Witte OW, Stoll G. Phagocytic response in photochemically induced infarction of rat cerebral cortex—the role of resident microglia. Stroke. 1997;28:382–386. doi: 10.1161/01.STR.28.2.382.
    1. Yrjanheikki J, Keinanen R, Pellikka M, Hokfelt T, Koistinaho J. Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc Natl Acad Sci U S A. 1998;95:15769–15774. doi: 10.1073/pnas.95.26.15769.
    1. Nagai N, Kawao N, Okada K, Ishida C, Okumoto K, Ueshima S, Suzuki Y, Umemura K, Matsuo O. Initial brain lesion size affects the extent of subsequent pathophysiological responses. Brain Res. 2010;1322:109–117. doi: 10.1016/j.brainres.2010.01.077.
    1. Abeysinghe HCS, Bokhari L, Dusting GJ, Roulston CL. Brain remodelling following endothelin-1 induced stroke in conscious rats. PLoS One. 2014;9:e97007. doi: 10.1371/journal.pone.0097007.
    1. Keiner S, Wurm F, Kunze A, Witte OW, Redeckew C. Rehabilitative therapies differentially alter proliferation and survival of glial cell populations in the perilesional zone of cortical infarcts. Glia. 2008;56:516–527. doi: 10.1002/glia.20632.
    1. Pena-Philippides JC, Caballero-Garrido E, Lordkipanidze T, Roitbak T. In vivo inhibition of miR-155 significantly alters post-stroke inflammatory response. J Neuroinflammation. 2016;13:287. doi: 10.1186/s12974-016-0753-x.
    1. Hoehn BD, Palmer TD, Steinberg GK. Neurogenesis in rats after focal cerebral ischemia is enhanced by indomethacin. Stroke. 2005;36:2718–2724. doi: 10.1161/.
    1. Zarruk JG, Greenhalgh AD, David S. Microglia and macrophages differ in their inflammatory profile after permanent brain ischemia. Exp Neurol. 2018;301:120–132. doi: 10.1016/j.expneurol.2017.08.011.
    1. Fagan SC, Edwards DJ, Borlongan CV, Xu L, Arora A, Feuerstein G, Hess DC. Optimal delivery of minocycline to the brain: implication for human studies of acute neuroprotection. Exp Neurol. 2004;186:248–251. doi: 10.1016/j.expneurol.2003.12.006.
    1. Wattananit S, Tornero D, Graubardt N, Memanishvili T, Monni E, Tatarishvili J, Miskinyte G, Ge R, Ahlenius H, Lindvall O, et al. Monocyte-derived macrophages contribute to spontaneous long-term functional recovery after stroke in mice. J Neurosci. 2016;36:4182–4195. doi: 10.1523/JNEUROSCI.4317-15.2016.
    1. Schmidt A, Strecker JK, Hucke S, Bruckmann NM, Herold M, Mack M, Diederich K, Schabitz WR, Wiendl H, Klotz L, Minnerup J. Targeting different monocyte/macrophage subsets has no impact on outcome in experimental stroke. Stroke. 2017;48:1061–1069. doi: 10.1161/STROKEAHA.116.015577.
    1. Cherry JD, Olschowka JA, O'Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation. 2014;11:98. doi: 10.1186/1742-2094-11-98.
    1. Hu XM, Leak RK, Shi YJ, Suenaga J, Gao YQ, Zheng P, Chen J. Microglial and macrophage polarization -new prospects for brain repair. Nat Rev Neurol. 2015;11:56–64. doi: 10.1038/nrneurol.2014.207.
    1. Gertig U, Hanisch UK. Microglial diversity by responses and responders. Front Cell Neurosci. 2014;8:101. doi: 10.3389/fncel.2014.00101.
    1. Ransohoff RM. A polarizing question: do M1 and M2 microglia exist? Nat Neurosci. 2016;19:987–991. doi: 10.1038/nn.4338.
    1. Zarruk JG, Fernandez-Lopez D, Garcia-Yebenes I, Garcia-Gutierrez MS, Vivancos J, Nombela F, Torres M, Burguete MC, Manzanares J, Lizasoain I, Moro MA. Cannabinoid type 2 receptor activation downregulates stroke-induced classic and alternative brain macrophage/microglial activation concomitant to neuroprotection. Stroke. 2012;43:211–219. doi: 10.1161/STROKEAHA.111.631044.
    1. Xu Y, Qian LL, Zong GJ, Ma K, Zhu X, Zhang H, Li N, Yang Q, Bai H, Ben J, et al. Class A scavenger receptor promotes cerebral ischemic injuy by pivoting microglia/macrophage polarization. Neuroscience. 2012;218:35–48. doi: 10.1016/j.neuroscience.2012.05.036.
    1. Huang M, Wan Y, Mao L, He QW, Xia YP, Li M, Li YN, Jin HJ, Hu B. Inhibiting the migration of M1 microglia at hyperacute period could improve outcome of tMCAO rats. CNS Neurosci Ther. 2017;23:222–232. doi: 10.1111/cns.12665.
    1. Tian DS, Li CY, Qin C, Murugan M, Wu LJ, Liu JL. Deficiency in the voltage-gated proton channel Hv1 increases M2 polarization of microglia and attenuates brain damage from photothrombotic ischemic stroke. J Neurochem. 2016;139:96–105. doi: 10.1111/jnc.13751.
    1. Jin Q, Cheng J, Liu Y, Wu J, Wang XY, Wei SW, Zhou XM, Qin ZH, Jia J, Zhen XC. Improvement of functional recovery by chronic metformin treatment is associated with enhanced alternative activation of microglia/macrophages and increased angiogenesis and neurogenesis following experimental stroke. Brain Behav Immun. 2014;40:131–142. doi: 10.1016/j.bbi.2014.03.003.
    1. Boddaert J, Bielen K, Jongers B, Manocha E, Yperzeele L, Cras P, Pirici D, Kumar-Singh S. CD8 signaling in microglia/macrophage polarization in a rat model of cerebral ischemia. PLoS One. 2018;13:e0186937. doi: 10.1371/journal.pone.0186937.
    1. Peters D, Berger J, Langnaese K, Derst C, Madai VI, Krauss M, Fischer KD, Veh RW, Laube G. Arginase and arginine decarboxylase—where do the putative gate keepers of polyamine synthesis reside in rat brain? PLoS One. 2013;8:e66735.
    1. Quirie A, Demougeot CE, Bertrand N, Mossiat C, Garnier P, Marie C, Prigent-Tessier A. Effect of stroke on arginase expression and localization in the rat brain. Eur J Neurosci. 2013;37:1193–1202. doi: 10.1111/ejn.12111.
    1. Carmichael ST, Archibeque I, Luke L, Nolan T, Momiy J, Li SL. Growth-associated gene expression after stroke: evidence for a growth-promoting region in peri-infarct cortex. Exp Neurol. 2005;193:291–311. doi: 10.1016/j.expneurol.2005.01.004.
    1. Chen XP, Zhang X, Liao WJ, Wan Q. Effect of physical and social components of enriched environment on astrocytes proliferation in rats after cerebral ischemia/reperfusion injury. Neurochem Res. 2017;42:1308–1316. doi: 10.1007/s11064-016-2172-x.

Source: PubMed

3
구독하다