Attenuation of acute stroke injury in rat brain by minocycline promotes blood-brain barrier remodeling and alternative microglia/macrophage activation during recovery

Yirong Yang, Victor M Salayandia, Jeffrey F Thompson, Lisa Y Yang, Eduardo Y Estrada, Yi Yang, Yirong Yang, Victor M Salayandia, Jeffrey F Thompson, Lisa Y Yang, Eduardo Y Estrada, Yi Yang

Abstract

Background: Minocycline reduces reperfusion injury by inhibiting matrix metalloproteinases (MMPs) and microglia activity after cerebral ischemia. Prior studies of minocycline investigated short-term neuroprotective effects during subacute stage of stroke; however, the late effects of minocycline against early reperfusion injury on neurovascular remodeling are less well studied. We have shown that spontaneous angiogenesis vessels in ischemic brain regions have high blood-brain barrier (BBB) permeability due to lack of major tight junction proteins (TJPs) in endothelial cells at three weeks. In the present study, we longitudinally investigated neurological outcome, neurovascular remodeling and microglia/macrophage alternative activation after spontaneous and minocycline-induced stroke recovery.

Methods: Adult spontaneously hypertensive rats had a 90 minute transient middle cerebral artery occlusion. At the onset of reperfusion they received a single dose of minocycline (3 mg/kg intravenously) or a vehicle. They were studied at multiple time points up to four weeks with magnetic resonance imaging (MRI), immunohistochemistry and biochemistry.

Results: Minocycline significantly reduced the infarct size and prevented tissue loss in the ischemic hemispheres compared to vehicle-treated rats from two to four weeks as measured with MRI. Cerebral blood flow measured with arterial spin labeling (ASL) showed that minocycline improved perfusion. Dynamic contrast-enhanced MRI indicated that minocycline reduced BBB permeability accompanied with higher levels of TJPs measured with Western blot. Increased MMP-2 and -3 were detected at four weeks. Active microglia/macrophage, surrounding and within the peri-infarct areas, expressed YM1, a marker of M2 microglia/macrophage activation, at four weeks. These microglia/macrophage expressed both pro-inflammatory factors tumor necrosis factors-α (TNF-α) and interleukin-1β (IL-1β) and anti-inflammatory factors transforming growth factor-β (TGF-β) and interleukin-10 (IL-10). Treatment with minocycline significantly reduced levels of TNF-α and IL-1β, and increased levels of TGF-β, IL-10 and YM1.

Conclusions: Early minocycline treatment against reperfusion injury significantly promotes neurovascular remodeling during stroke recovery by reducing brain tissue loss, enhancing TJP expression in ischemic brains and facilitating neuroprotective phenotype alternative activation of microglia/macrophages.

Figures

Figure 1
Figure 1
Stroke recovery monitored by magnetic resonance imaging. A. Anatomical T2 MR images. Arrows indicate ischemic hemispheres. Line graph demonstrates quantification of infarct volumes in ischemic hemispheres. B. ADC maps. Line graph demonstrates quantification of edema (acute stage) and tissue loss (late stage) in ischemic hemispheres. C. FA maps. Line graph demonstrates quantification of white matter change in ischemic hemispheres. *P <0.05, ***P <0.001 versus vehicle group, n = 8 in each group.
Figure 2
Figure 2
ASL maps monitored by MRI at 48 hours and one, two and four weeks after stroke. Arrows indicate ischemic hemispheres. Line graph demonstrates quantification of blood flow in peri-infarct areas. *P <0.05 versus vehicle group, n = 8 in each group.
Figure 3
Figure 3
Blood–brain barrier permeability monitored by magnetic resonance imaging. A. Parametric image Ki map by DCE-MRI represents BBB transfer rate at four weeks after stroke. Color-coded permeability coefficient maps reconstructed from contrast-enhanced MRI data demonstrate the regions of high (red) and low (blue) permeability. B. Histogram demonstrates the quantification of BBB permeability in peri-infarct areas (light blue areas in the brain slice cartoon). *P = 0.0504 versus vehicle group, n = 8 in each group. C. RECA1 (marker of endothelial cells) immunostaining shows increased density of new vessels in the peri-infarct area (arrows), corresponding to the regions where BBB transfer rate and plasma volume were measured.
Figure 4
Figure 4
Expression of tight junction proteins and matrix metalloproteinases at four weeks after stroke. A. Western blot analysis for TJPs. ZO-1: **P <0.05 versus V-I, V-C and Mino-C. Occludin: *P <0.05 versus V-I, V-C and Mino-C. Claudin-5: *P <0.05 versus V-I, V-C and Mino-C. B. Gel zymography analysis for MMP-2 and −9. **P <0.01 versus V-C and Mino-C. n = 8 in the vehicle group, 9 in the minocycline group. C. Western blot analysis for MMP-3. Level of MMP-3 including preform (57 kDa) and active form (45 kDa): *P <0.05 versus V-C and Mino-C. Mino-C: minocycline contralateral. Mino-I: minocycline ipsilateral. V-C: Vehicle contralateral. V-I: Vehicle ipsilateral. n = 8 in the vehicle group, 9 in the minocycline group.
Figure 5
Figure 5
Microglia/macrophage activation by immunofluorescence staining of Iba-1. A. Morphological changes of microglia/macrophage in ischemic hemispheres over reperfusion courses. Scale bar = 100 μm. The right panels at each time point present a higher magnification of the images shown in squares in the left panels. Scale bar = 50 μm. DAPI was used to show nuclei and vessel at one week. core-i: core infarct area; peri-i: peri-infarct area; V: vessel. B. Quantification of Iba-1 fluorescence (FL) intensity in ischemic hemispheres over reperfusion courses. *P <0.05, ***P <0.001 versus vehicle group, n = 5 in each group. The brain cartoon shows the three images measured by ImageJ that were obtained from the infarct areas, indicated by the squares in red.
Figure 6
Figure 6
Phenotype of microglia/macrophage alternative activation at four weeks after stroke. A. Double immunostaining shows expression of YM1 in microglia/macrophage (OX-42). Scale bars = 50. DAPI was used to show nuclei. B. Western blot analysis for protein level of YM1. **P <0.01 versus V-C and Mino-C. ***P <0.001 versus V-C and Mino-C. #P <0.05 versus V-I. n = 8 in the vehicle group, 9 in the minocycline group.
Figure 7
Figure 7
Inflammatory factors expressed by active microglia/macrophage in peri-infarct areas at four weeks after stroke. A. Double immunostaining shows expression of TNF-α, IL-1β, IL-10 and TGF-β in active microglia (Iba-1). B. Analysis and quantification for co-localization of cytokines and Iba-1 in the microglia/macrophages with Fiji-ImageJ. Representative two-dimensional histogram and scatterplots visualize the correlation of the pixel intensities, over all pixels and voxels in the images with different Li’s ICQ values, generated by Fiji-ImageJ. Statistical bar figures demonstrate the quantification of Li’s ICQ values for co-localization of each cytokine with Iba-1 in vehicle- and minocycline-treated ischemic hemispheres. *P <0.05, **P <0.01 versus V-I. n = 5 in each group. C. Double immunostaining shows little expression of TNF-α and IL-1β in active microglia/macrophage, with extending processes in the peri-infarct areas bordering with intact tissues. Scale bars = 50 μm. D. Double immunostaining shows expression of TGF-β in PDGFR-β-positive pericytes that closely surround the vessels (V). Scale bars = 50 and 100 μm.
Figure 8
Figure 8
Western blot analysis for active forms of inflammatory factors in rat brains at four weeks after stroke. TNF-α: levels of TNF-α were decreased in ischemic rat brains and treatment with minocycline facilitated the reduction. IL-1β: *P <0.05 versus V-I. IL-10: *P <0.05 versus V-I. TGF-α: *P <0.05 versus V-I; ##P <0.01 versus V-C and Mino-C. n = 8 in the vehicle group, 9 in the minocycline group.

References

    1. Yang Y, Candelario-Jalil E, Thompson JF, Cuadrado E, Estrada EY, Rosell A, et al. Increased intranuclear matrix metalloproteinase activity in neurons interferes with oxidative DNA repair in focal cerebral ischemia. J Neurochem. 2010;112:134–49. doi: 10.1111/j.1471-4159.2009.06433.x.
    1. Yang Y, Estrada EY, Thompson JF, Liu W, Rosenberg GA. Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. J Cereb Blood Flow Metab. 2007;27:697–709. doi: 10.1038/sj.jcbfm.9600440.
    1. Yang Y, Thompson JF, Taheri S, Salayandia VM, McAvoy TA, Hill JW, et al. Early inhibition of MMP activity in ischemic rat brain promotes expression of tight junction proteins and angiogenesis during recovery. J Cereb Blood Flow Metab. 2013;33:1104–14. doi: 10.1038/jcbfm.2013.56.
    1. Fagan SC, Cronic LE, Hess DC. Minocycline development for acute ischemic stroke. Transl Stroke Res. 2011;2:202–8. doi: 10.1007/s12975-011-0072-6.
    1. Murata Y, Rosell A, Scannevin RH, Rhodes KJ, Wang X, Lo EH. Extension of the thrombolytic time window with minocycline in experimental stroke. Stroke. 2008;39:3372–7. doi: 10.1161/STROKEAHA.108.514026.
    1. Fagan SC, Waller JL, Nichols FT, Edwards DJ, Pettigrew LC, Clark WM, et al. Minocycline to improve neurologic outcome in stroke (MINOS): a dose-finding study. Stroke. 2010;41:2283–7. doi: 10.1161/STROKEAHA.110.582601.
    1. Switzer JA, Hess DC, Ergul A, Waller JL, Machado LS, Portik-Dobos V, et al. Matrix metalloproteinase-9 in an exploratory trial of intravenous minocycline for acute ischemic stroke. Stroke. 2011;42:2633–5. doi: 10.1161/STROKEAHA.111.618215.
    1. Machado LS, Kozak A, Ergul A, Hess DC, Borlongan CV, Fagan SC. Delayed minocycline inhibits ischemia-activated matrix metalloproteinases 2 and 9 after experimental stroke. BMC Neurosci. 2006;7:56. doi: 10.1186/1471-2202-7-56.
    1. Machado LS, Sazonova IY, Kozak A, Wiley DC, El-Remessy AB, Ergul A, et al. Minocycline and tissue-type plasminogen activator for stroke: assessment of interaction potential. Stroke. 2009;40:3028–33. doi: 10.1161/STROKEAHA.109.556852.
    1. Lee CZ, Yao JS, Huang Y, Zhai W, Liu W, Guglielmo BJ, et al. Dose–response effect of tetracyclines on cerebral matrix metalloproteinase-9 after vascular endothelial growth factor hyperstimulation. J Cereb Blood Flow Metab. 2006;26:1157–64.
    1. Jin X, Liu J, Liu KJ, Rosenberg GA, Yang Y, Liu W. Normobaric hyperoxia combined with minocycline provides greater neuroprotection than either alone in transient focal cerebral ischemia. Exp Neurol. 2013;240:9–16. doi: 10.1016/j.expneurol.2012.11.018.
    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;37:1087–93. doi: 10.1161/.
    1. Colton CA. Heterogeneity of microglial activation in the innate immune response in the brain. J Neuroimmune Pharmacol. 2009;4:399–418. doi: 10.1007/s11481-009-9164-4.
    1. Emsley HC, Tyrrell PJ. Inflammation and infection in clinical stroke. J Cereb Blood Flow Metab. 2002;22:1399–419. doi: 10.1097/00004647-200212000-00001.
    1. Jacobs AH, Tavitian B. Noninvasive molecular imaging of neuroinflammation. J Cereb Blood Flow Metab. 2012;32:1393–415. doi: 10.1038/jcbfm.2012.53.
    1. Oliveira GB, Fontes Ede Jr A, de Carvalho S, da Silva JB, Fernandes LM, Oliveira MC, et al. Minocycline mitigates motor impairments and cortical neuronal loss induced by focal ischemia in rats chronically exposed to ethanol during adolescence. Brain Res. 2014;1561:23–34. doi: 10.1016/j.brainres.2014.03.005.
    1. Lartey FM, Ahn GO, Ali R, Rosenblum S, Miao Z, Arksey N, et al. The relationship between serial [(18) F]PBR06 PET imaging of microglial activation and motor function following stroke in mice. Mol Imaging Biol. 2014;16:81–9.
    1. Lampron A, Elali A, Rivest S. Innate immunity in the CNS: redefining the relationship between the CNS and its environment. Neuron. 2013;78:214–32. doi: 10.1016/j.neuron.2013.04.005.
    1. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32:593–604. doi: 10.1016/j.immuni.2010.05.007.
    1. Taylor RA, Sansing LH. Microglial responses after ischemic stroke and intracerebral hemorrhage. Clin Dev Immunol. 2013;2013:746068. doi: 10.1155/2013/746068.
    1. Giunti D, Parodi B, Cordano C, Uccelli A, Kerlero de Rosbo N. Can we switch microglia’s phenotype to foster neuroprotection? Focus on multiple sclerosis. Immunology. 2014;141:328–39. doi: 10.1111/imm.12177.
    1. Cramer SC. Repairing the human brain after stroke: I. Mechanisms of spontaneous recovery. Ann Neurol. 2008;63:272–87. doi: 10.1002/ana.21393.
    1. Cramer SC. Repairing the human brain after stroke: II. Restorative therapies. Ann Neurol. 2008;63:549–60. doi: 10.1002/ana.21412.
    1. Yang Y, Hill JW, Rosenberg GA. Multiple roles of metalloproteinases in neurological disorders. Prog Mol Biol Transl Sci. 2011;99:241–63. doi: 10.1016/B978-0-12-385504-6.00006-3.
    1. Xu L, Fagan SC, Waller JL, Edwards D, Borlongan CV, Zheng J, et al. Low dose intravenous minocycline is neuroprotective after middle cerebral artery occlusion-reperfusion in rats. BMC Neurol. 2004;4:7. doi: 10.1186/1471-2377-4-7.
    1. Hill JW, Poddar R, Thompson JF, Rosenberg GA, Yang Y. Intranuclear matrix metalloproteinases promote DNA damage and apoptosis induced by oxygen-glucose deprivation in neurons. Neuroscience. 2012;220:277–90. doi: 10.1016/j.neuroscience.2012.06.019.
    1. Sood R, Yang Y, Taheri S, Candelario-Jalil E, Estrada EY, Walker EJ, et al. Increased apparent diffusion coefficients on MRI linked with matrix metalloproteinases and edema in white matter after bilateral carotid artery occlusion in rats. J Cereb Blood Flow Metab. 2009;29:308–16. doi: 10.1038/jcbfm.2008.121.
    1. Taheri S, Sood R. Partial volume effect compensation for improved reliability of quantitative blood–brain barrier permeability. Magn Reson Imaging. 2007;25:613–25. doi: 10.1016/j.mri.2006.09.029.
    1. Patlak CS, Blasberg RG, Fenstermacher JD. Graphical evaluation of blood-to-brain transfer constants from multiple-time uptake data. J Cereb Blood Flow Metab. 1983;3:1–7. doi: 10.1038/jcbfm.1983.1.
    1. Ewing JR, Knight RA, Nagaraja TN, Yee JS, Nagesh V, Whitton PA, et al. Patlak plots of Gd-DTPA MRI data yield blood–brain transfer constants concordant with those of 14C-sucrose in areas of blood–brain opening. Magn Reson Med. 2003;50:283–92. doi: 10.1002/mrm.10524.
    1. Ito D, Tanaka K, Suzuki S, Dembo T, Fukuuchi Y. Enhanced expression of Iba1, ionized calcium-binding adapter molecule 1, after transient focal cerebral ischemia in rat brain. Stroke. 2001;32:1208–15. doi: 10.1161/01.STR.32.5.1208.
    1. Patel AR, Ritzel R, McCullough LD, Liu F. Microglia and ischemic stroke: a double-edged sword. Int J Physiol Pathophysiol Pharmacol. 2013;5:73–90.
    1. Li Q, Lau A, Morris TJ, Guo L, Fordyce CB, Stanley EF. A syntaxin 1, Galpha(o), and N-type calcium channel complex at a presynaptic nerve terminal: analysis by quantitative immunocolocalization. J Neurosci. 2004;24:4070–81. doi: 10.1523/JNEUROSCI.0346-04.2004.
    1. Charlton RA, Barrick TR, McIntyre DJ, Shen Y, O’Sullivan M, Howe FA, et al. White matter damage on diffusion tensor imaging correlates with age-related cognitive decline. Neurology. 2006;66:217–22. doi: 10.1212/01.wnl.0000194256.15247.83.
    1. Tanaka Y, Nagaoka T, Nair G, Ohno K, Duong TQ. Arterial spin labeling and dynamic susceptibility contrast CBF MRI in postischemic hyperperfusion, hypercapnia, and after mannitol injection. J Cereb Blood Flow Metab. 2011;31:1403–11. doi: 10.1038/jcbfm.2010.228.
    1. Iadecola C, Anrather J. The immunology of stroke: from mechanisms to translation. Nat Med. 2011;17:796–808. doi: 10.1038/nm.2399.
    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;196:290–7. doi: 10.1016/j.expneurol.2005.08.004.
    1. Snook ER, Fisher-Perkins JM, Sansing HA, Lee KM, Alvarez X, MacLean AG, et al. Innate immune activation in the pathogenesis of a murine model of globoid cell leukodystrophy. Am J Pathol. 2014;184:382–96. doi: 10.1016/j.ajpath.2013.10.011.
    1. McColl BW, Rothwell NJ, Allan SM. Systemic inflammation alters the kinetics of cerebrovascular tight junction disruption after experimental stroke in mice. J Neurosci. 2008;28:9451–62. doi: 10.1523/JNEUROSCI.2674-08.2008.
    1. Rosell A, Cuadrado E, Ortega-Aznar A, Hernandez-Guillamon M, Lo EH, Montaner J. MMP-9-positive neutrophil infiltration is associated to blood–brain barrier breakdown and basal lamina type IV collagen degradation during hemorrhagic transformation after human ischemic stroke. Stroke. 2008;39:1121–6. doi: 10.1161/STROKEAHA.107.500868.
    1. Schaller B, Graf R. Cerebral ischemia and reperfusion: the pathophysiologic concept as a basis for clinical therapy. J Cereb Blood Flow Metab. 2004;24:351–71. doi: 10.1097/00004647-200404000-00001.
    1. Pan J, Konstas AA, Bateman B, Ortolano GA, Pile-Spellman J. Reperfusion injury following cerebral ischemia: pathophysiology, MR imaging, and potential therapies. Neuroradiology. 2007;49:93–102. doi: 10.1007/s00234-006-0183-z.
    1. Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. 2010;87:779–89. doi: 10.1189/jlb.1109766.
    1. Liu Z, Fan Y, Won SJ, Neumann M, Hu D, Zhou L, et al. Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke. 2007;38:146–52. doi: 10.1161/.
    1. Weng YC, Kriz J. Differential neuroprotective effects of a minocycline-based drug cocktail in transient and permanent focal cerebral ischemia. Exp Neurol. 2007;204:433–42. doi: 10.1016/j.expneurol.2006.12.003.
    1. Kobayashi K, Imagama S, Ohgomori T, Hirano K, Uchimura K, Sakamoto K, et al. Minocycline selectively inhibits M1 polarization of microglia. Cell Death Dis. 2013;4:e525. doi: 10.1038/cddis.2013.54.
    1. Hewlett KA, Corbett D. Delayed minocycline treatment reduces long-term functional deficits and histological injury in a rodent model of focal ischemia. Neuroscience. 2006;141:27–33. doi: 10.1016/j.neuroscience.2006.03.071.
    1. Ozen I, Deierborg T, Miharada K, Padel T, Englund E, Genove G, et al. Brain pericytes acquire a microglial phenotype after stroke. Acta Neuropathol. 2014;128:381–96. doi: 10.1007/s00401-014-1295-x.
    1. Ronaldson PT, Demarco KM, Sanchez-Covarrubias L, Solinsky CM, Davis TP. Transforming growth factor-beta signaling alters substrate permeability and tight junction protein expression at the blood–brain barrier during inflammatory pain. J Cereb Blood Flow Metab. 2009;29:1084–98. doi: 10.1038/jcbfm.2009.32.
    1. Dohgu S, Yamauchi A, Takata F, Naito M, Tsuruo T, Higuchi S, et al. Transforming growth factor-beta1 upregulates the tight junction and P-glycoprotein of brain microvascular endothelial cells. Cell Mol Neurobiol. 2004;24:491–7. doi: 10.1023/B:CEMN.0000022776.47302.ce.

Source: PubMed

3
Abonnieren