G Model
PRONEU 1417 1–25 Progress in Neurobiology xxx (2016) xxx–xxx
Contents lists available at ScienceDirect
Progress in Neurobiology journal homepage: www.elsevier.com/locate/pneurobio 1 2 3 4 5 6 7
The biphasic function of microglia in ischemic stroke Q1 Yuanyuan
Ma a,b, Jixian Wang a,b, Yongting Wang b,*, Guo-Yuan Yang a,b,**
a
Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China b
A R T I C L E I N F O
A B S T R A C T
Article history: Received 15 September 2015 Received in revised form 22 December 2015 Accepted 10 January 2016 Available online xxx
Microglia are brain resident macrophages originated from primitive progenitor cells in the yolk sac. Microglia can be activated within hours and recruited to the lesion site. Traditionally, microglia activation is considered to play a deleterious role in ischemic stroke, as inhibition of microglia activation attenuates ischemia induced brain injury. However, increasing evidence show that microglia activation is critical for attenuating neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after cerebral ischemia. Differential polarization of microglia could likely explain the biphasic role of microglia in ischemia. We comprehensively reviewed the mechanisms involved in regulating microglia activation and polarization. The latest discoveries of microRNAs in modulating microglia function are discussed. In addition, the interaction between microglia and other cells including neurons, astrocytes, oligodendrocytes, and stem cells were also reviewed. Future therapies targeting microglia may not exclusively aim at suppressing microglia activation, but also at modulating microglia polarization at different stages of ischemic stroke. More work is needed to elucidate the cellular and molecular mechanisms of microglia polarization under ischemic environment. The roles of microRNAs and transplanted stem cells in mediating microglia activation and polarization during brain ischemia also need to be further studied. ß 2016 Elsevier Ltd. All rights reserved.
Keywords: Crosstalk Ischemic stroke Microglia microRNAs Polarization
8
Contents 1. 2. 3.
4.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Origin of microglia. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Microglia activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Microglia activation in the brain . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. Microglia activation during ischemic stroke . . . . . . . . . . . . . . . . . 3.2. Microglia activation in human ischemic stroke . . . . . . . 3.2.1. Microglia activation in experimental ischemic stroke . . 3.2.2. Receptors mediating microglia activation and function. 3.2.3. Roles of activated microglia in ischemic stroke (Table 1) . . . . . . . . . . . . Microglia and blood brain barrier permeability . . . . . . . . . . . . . . 4.1. The effect of microglia on neurogenesis . . . . . . . . . . . . . . . . . . . . 4.2.
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
000 000 000 000 000 000 000 000 000 000 000
Abbreviations: AD, Alzheimer’s disease; ALS, amyotrophic lateral sclerosis; BBB, blood brain barrier; CSF-1R, colony stimulating factor-1 receptor; EPCs, endothelia progenitor cells; FcRs, Fc receptors; HMGB1, high mobility group box1; IGF-1, insulin-like growth factor 1; iNOS, inducible nitric oxide synthase; LPS, lipopolysaccharide; MCAO, middle cerebral artery occlusion; MerTK, Mer receptor tyrosine kinase; MFG-E8, milk fat globule-EGF factor-8 protein; MS, multiple sclerosis; MSCs, mesenchymal stem cells; NO, nitric oxide; NOX, NADPH oxidase; NSCs, neural stem cells; OPCs, oligodendrocyte progenitor cells; OGD, oxygen–glucose deprivation; PACAP, pituitary adenylate cyclaseactivating polypeptide; PD, Parkinson’s disease; RAGE, receptor for advanced glycation endproducts; ROS, reactive oxygen species; Runx1, runt-related transcription factor 1; SOCS-1, suppressor of cytokine signaling 1; TLRs, Toll-like receptors; TREM-2, triggering receptor expressed on myeloid cells-2; SVZ, subventricular zone; VZ, ventricular zone. * Corresponding author. Tel.: +86 21 62933186; fax: +00 86 21 62932302. Q2 ** Corresponding author at: Shanghai Jiaotong University, Med-X Reasearch Institute, 1954 Hua Shan Road, Shanghai 200030, China. Tel.: +86 21 62933186; fax: +00 86 21 62932302. E-mail addresses:
[email protected] (Y. Wang),
[email protected] (G.-Y. Yang). http://dx.doi.org/10.1016/j.pneurobio.2016.01.005 0301-0082/ß 2016 Elsevier Ltd. All rights reserved.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 2
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
5.
6.
7. 8. 9.
4.2.1. Microglia influence neurogenesis under physiological condition . . . . Microglia influence neurogenesis during ischemic stroke . . . . . . . . . . 4.2.2. Interaction between activated microglia and NPCs . . . . . . . . . . . . . . . 4.2.3. Microglia polarization during ischemic stroke . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Microglia polarization. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1. Ischemia induces microglia polarization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2. Substances involved in the modulation of microglia polarization . . . . . . . . . . . 5.3. Clinical drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.3.1. 5.3.2. Receptors and small molecules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . microRNAs and microglia (Table 2) . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.3.3. Microglia crosstalk with other cells during ischemic stroke . . . . . . . . . . . . . . . . . . . . . 6.1. Microglia and neurons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Neurons control microglia activation . . . . . . . . . . . . . . . . . . . . . . . . . . 6.1.1. Neurons influence microglial function . . . . . . . . . . . . . . . . . . . . . . . . . 6.1.2. Microglia influence neuronal function . . . . . . . . . . . . . . . . . . . . . . . . . 6.1.3. 6.2. Microglia and astrocytes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Cross-talk between microglia and astrocytes . . . . . . . . . . . . . . . . . . . . 6.2.1. Cross-talk between microglia and astrocytes during ischemic stroke. 6.2.2. Microglia and oligodendrocytes/oligodendrocyte progenitor cells . . . . . . . . . . . 6.3. Relationship between microglia and stem cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Microglia as a potential candidate of cell based therapy for ischemic stroke. . . . . . . . Conclusion and prospect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . . . . . . . . . . . . . . .
000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000 000
9
10 Q3 1. Introduction 11 Q4 In the late 19th century, German anatomist Franz Nissl first 12 described microglia and named them as Staebchenzellen (rod 13 cells) for their rod-shaped nuclei. Nissl explained these cells as 14 reactive glia elements with the capacity of migration, phagocytosis 15 and proliferation (Kim and de Vellis, 2005). Later in 1913, Spanish 16 neuroscientist Santiago Ramo´ny Cajal renamed this type of cells as 17 a ‘‘third element’’. He pointed out that these cells were different 18 from neurons and astrocytes, and probably of mesodermal origin. 19 Further, this ‘‘third element’’ was divided into various cell types 20 based on morphological and functional differences (Ginhoux et al., 21 2013). The main population among the third elements was 22 oligodendrocytes, and the second population was microglia 23 (Cartier et al., 2014). Using a silver staining technique, the 24 presence of microglia in human brain was demonstrated. Microglia 25 were classified into three types based on their morphological 26 differences: amoeboid, ramified, and intermediate microglia (Kim 27 and de Vellis, 2005). Since then, the studies of microglia expanded 28 rapidly and widely (Nayak et al., 2014). 29 In the adult mouse brain, microglia constitute 5–12% of all glia 30 cells. The distribution of microglia is different in different regions 31 of the brain, ranging from 5% in the cerebral cortex and corpus 32 callosum to 12% in the substantia nigra. There are approximately 33 3.5 106 microglia in the adult mouse brain. More microglia are in 34 the gray matter than that in the white matter in the mouse brain 35 (Lawson et al., 1990). In normal human brain, the distribution of 36 microglia varies by up to one order of magnitude. The number of 37 microglia accounts for approximately 0.5% to 16.6% of all brain 38 cells. Different from the case in mouse brain, much more microglia 39 are located in the white matter than in the gray matter in human 40 brain. These findings provide valuable information to evaluate 41 microglial response under pathological conditions (Mittelbronn 42 et al., 2001). 43 Microglia are the resident macrophages and the first line of 44 defense against injury in the central nervous system (Hu et al., 45 2014; Prinz and Priller, 2014). After ischemic brain injury, 46 microglia rapidly migrate toward the lesion site and exacerbate 47 tissue injury by producing inflammatory cytokines and cytotoxic 48 substances. On the other hand, however, microglia also contribute 49 to tissue repair and remodeling by clearing up debris and
producing anti-inflammatory cytokines and growth factors (Shi and Pamer, 2011). Increasing evidence indicate that microglia could be beneficial for the functional recovery after cerebral ischemia (Neumann et al., 2009; Neumann et al., 2006). Microglia activation is a complicated process that can be affected by many substances and surrounding cells such as neurons, astrocytes, and oligodendrocytes. The dual role of microglia is associated with different polarization of microglia under different cellular context and pathological stages after brain injury (Hu et al., 2014; Kim et al., 2014). Elucidating the detailed mechanisms of microglia activation and polarization will help to drive microglia shift from a detrimental phenotype to a protective phenotype. Investigation of how microglia communicate with neurons, astrocytes, and oligodendrocytes will aid our understanding on the role of microglia during ischemic disease.
50 51 52 53 54 55 56 57 58 59 60 61 62 63 64
2. Origin of microglia
65
In the last decades, investigators were devoted to deciphering the origin of microglia since its discovery (Wieghofer et al., 2015). Although it was still under debate regarding what was the precise nature of microglia progenitors, the generally accepted concept is that microglia are of myeloid origin (Ginhoux et al., 2013). Cajal believed that microglia originated from mesodermal progenitors (Kettenmann et al., 2011). Meningeal macrophages could penetrate into the brain during embryonic development and transform into microglia (Cartier et al., 2014; Kim and de Vellis, 2005). Initially, circulating monocytes or precursor cells invaded into the developing brain, changed their amoeboid morphology, and formed resting microglia of ramified shape in the mature brain (Kim and de Vellis, 2005). Studies using bone marrow transplantation technique indicated that microglia derived from blood monocytes. In a model of primary demyelination induced by copper chelator cuprizone and a model of facial nerve axotomy in mice, blood derived Ly-6ChiCCR2+ monocytes could invade into the central nervous system and contribute to the repopulation of microglia after the brain was irradiated (Mildner et al., 2007; Varvel et al., 2012). However, further clarification was needed to identify these invading cells as true microglia (Nayak et al., 2014). Using a model of chimeras between chick embryo and quail yolk sac, a study showed that microglia progenitors originated
66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146
from embryonic yolk sac (Cuadros et al., 1993). Myeloid cells originated from quail yolk sac invaded into the chick embryo during development, which was independent of blood supply. Another study supported this concept with the discovery of microglia precursors presented in the yolk sac and later in the brain rudiment at embryonic day eight (Alliot et al., 1999). Most microglia expressed proliferating cell nuclear antigen (PCNA), a marker of cell proliferation, suggesting that microglia could proliferate in situ. Recently, a study further confirmed that microglia were originated from primitive myeloid progenitors located in the yolk sac using fate mapping technique. Crossing mice expressing tamoxifen-inducible Cre recombinase under runt-related transcription factor 1 (Runx1) promoter with mice expressing loxP Rosa-YFP gene produced transgenic mice in which yolk-sacderived cells were YFP+. Because Runx1 was specifically expressed in yolk sac and embryonic liver hematopoietic progenitors, researchers were able to trace yolk-sac-derived cells (Cartier et al., 2014; Nayak et al., 2014). When tamoxifen was injected into pregnant mice before embryonic day 7.5, Runx1+ cells were restricted in yolk sac first and then migrated into the mouse brain from embryonic day 10 to 6 weeks after birth, in a circulation dependent manner (Samokhvalov et al., 2007). However, when tamoxifen was injected at embryonic day 8.5, when yolk sac hematopoiesis was already replaced by definitive hematopoiesis in the embryo, Runx1+ cells could not be found in the brain. These results collectively suggest that Runx1+ cells enter the brain during early development and give rise to microglia. In addition, microglia and yolk sac macrophages are markedly reduced in colony stimulating factor-1 receptor (CSF-1R) deficient mice while the amount of circulating monocytes is not affected, indicating that CSF-1R is essential for the development of microglia and macrophages in the embryo but not required by circulating monocytes. During embryonic development, definitive hematopoiesis is initiated in the aorta, gonads, and mesonephros (AGM) region, and then occur in the fetal liver, spleen, and bone marrow (Lichanska and Hume, 2000; Orkin and Zon, 2008). Tissue macrophages were thought to derive from bone marrow monocytes. Myb, a transcription factor, is required for the development of definitive hematopoiesis but not for the yolk sac myelopoiesis (Mucenski et al., 1991). It was noted that when PU.1, a transcription factor required for macrophage development, was deleted, all macrophages and microglia were absent (DeKoter et al., 1998; McKercher et al., 1996). These results revealed that yolk sac derived macrophages give rise to microglia in the embryo (Cartier et al., 2014). Another study demonstrated that microglia derive from primitive CD45c-kit+ erythromyeloid precursors in yolk sac. These precursors eventually mature into CD45+/c-kit/CX3CR1+ microglia and invade into the brain via specific matrix metalloproteinase. Transcription factors PU.1 and IRF8 are both essential for the development of microglia derived from yolk sac precursors. When PU.1 and IRF8 are deficient, the number of Iba1+ microglia decreased in mouse brain (Kierdorf et al., 2013). Collectively, all these studies suggested that microglia originate from primitive myeloid progenitors arising from yolk sac before embryonic day 8. In addition, microglia are a genetically distinct cell type in the mononuclear phagocyte system (Ginhoux et al., 2010; Schulz et al., 2012).
147
3. Microglia activation
148
3.1. Microglia activation in the brain
149 150 151
Under physiological condition, microglia display classical ramified morphology with a small cell soma and fine processes, which is referred as ‘‘resting microglia’’ (Kreutzberg, 1996). Resting
3
microglia are defined as having low expression of CD45, MHC II, CD80, CD86, and CD11c (Lynch, 2009; Ponomarev et al., 2005; Prinz et al., 2011). The processes of microglia are constantly in motion and survey the local microenvironment of the brain (Nimmerjahn et al., 2005). Disturbance of brain homeostasis could induce microglia activation (Ransohoff and Perry, 2009). Once activated, microglia undergo typical morphological changes, characterized by retracting and thickening of the processes, and hypertrophy of the cell body (Kreutzberg, 1996). A number of markers such as CD45, MHC II, and CD86 are up-regulated when microglia are activated (Ponomarev et al., 2013). In addition, cell surface markers such as Iba1, IB4, F4/80, and CD68 (ED-1) are commonly used to identify microglia activation. After focal brain ischemia, microglia display different expression patterns of markers depending on whether they are located in the peri-infarct region or in the infarct zone. In the peri-infarct region, microglia are positive for Iba1 but negative for CD68, while in the infarct zone microglia are both Iba1and CD68 positive and displayed enhanced CD11b expression (Ito et al., 2001; Morrison and Filosa, 2013). Activated microglia in the local infarct zone mainly express MHC I and act primarily as scavenger cells (Stoll et al., 1998). While activated microglia in regions far from infarct region mainly express MHC II and are thought to be associated with Wallerian (anterograde) degeneration (Block et al., 2005; Morioka et al., 1993). Therefore, microglia at different locations in respect to brain ischemia exert different function.
152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177
3.2. Microglia activation during ischemic stroke
178
Inflammation is recognized as a vital contributor to the 179 pathophysiology of ischemic stroke (Moskowitz et al., 2010). 180 Microglia activation is the first step of inflammatory response in 181 the brain, followed by infiltration of immune cells such as 182 neutrophils, macrophages/monocytes, natural killer cells, T cells, 183 and other neuronal cell activation (Iadecola and Anrather, 2011; Jin 184 et al., 2010). Microglia could be activated within minutes after 185 ischemic brain injury (Nakajima and Kohsaka, 2004). Activated 186 microglia can produce a variety of mediators including inducible 187 nitric oxide synthase (iNOS) (Iadecola and Ross, 1997), nitric oxide 188 (NO) (Gibson et al., 2005; Nakashima et al., 1995), pro- 189 inflammatory cytokines (such as TNF-a) (Lambertsen et al., 190 2009), anti-inflammatory cytokines (such as TGF-b) (Iadecola 191 and Anrather, 2011), growth and trophic factors such as insulin- 192 like growth factor 1 (IGF-1), basic fibroblast growth factor (bFGF), 193 hepatocyte growth factor (HGF), platelet-derived growth factor 194 (PDGF) (Lai and Todd, 2006), nerve growth factor (NGF), brain- 195 derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), 196 neurotrophin-4/5 (NT-4/5), and plasminogen (PGn) (Elkabes 197 et al., 1996). Whether these mediators exacerbate or alleviate 198 tissue damage during brain ischemia needs to be further 199 investigated. For example, TNF-a, which is a key player in brain 200 inflammation, is up-regulated in microglia both in animals and 201 human after ischemic stroke (Clausen et al., 2008; Dziewulska and 202 Mossakowski, 2003; Gregersen et al., 2000). Administration of 203 recombinase TNF-aprotein before transient middle cerebral artery 204 occlusion (MCAO) exacerbated infarct volume, brain swelling, and 205 neurological deficit. These phenomenon could be reversed by pre- 206 treatment with TNF-a antibody (Yang et al., 1998). Blockage of 207 endogenous TNF-a resulted in a smaller infarct volume compared 208 to the control group (Barone et al., 1997). These results implied Q5209 that TNF-a plays a toxic role during focal cerebral ischemia. 210 However, a study using TNF-a, TNF-areceptor I (TNF-p55R), and 211 TNF-a receptor II knock out mice suggests they could function 212 differently. Mice lacking TNF-aand TNF-p55R develop larger 213 infarct volume and poorer neurological outcome compared to 214 the wild type mice and TNF-a receptor II knock out mice. 215
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 4
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
216 217 218 219 220 221 222 223 224 225 226 227
Furthermore, TNF-ais mainly produced by activated microglia and could enhance the tolerance of cultured neurons and astrocytes against oxidative stress and ischemic injury (Ginis et al., 2002; Goodman and Mattson, 1996; Lambertsen et al., 2009). These results indicated that microglia could facilitate neuroprotective function against brain ischemia via TNF-p55R. The different conclusions mentioned above is due to that TNF-a exerts different function depending on the location of TNF-areceptors (Vexler et al., 2006). Further study is needed to elucidate the exact role and mechanism of mediators produced by microglia in ischemic stroke. The task would pave the road for developing intervention strategies targeting mediators produced by microglia.
228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280
3.2.1. Microglia activation in human ischemic stroke With the rapid development of imaging methods such as positron emission tomography (PET) and MRI, it is feasible to study microglia activation in human. Clinical studies demonstrated that microglia activation is present in the brain at the acute phase (Krupinski et al., 1996; Tomimoto et al., 1996), the sub-acute phase (Price et al., 2006), and the recovery phase after ischemic stroke (Gulyas et al., 2012). Microglia activation can be detected in human brain within 24–48 h after brain ischemia, and mainly present in the ischemic core and extend to the peri-infarct region over time (Krupinski et al., 1996; Tomimoto et al., 1996). The time course of microglia activation in the infarct zone or the peri-infarct region is different in stroke patients (Thiel et al., 2010). Combining PET using radiolabeled ligand 11C-(R)-PK11195 with structural brain imaging by MRI, microglia activation was dynamically evaluated in vivo. Activated microglia were observed in 6 patients from 3 to 150 days after ischemic stroke. Imaging results showed that microglia activation could be observed as early as day 3. One patient was examined at 28 and 150 days after brain ischemia. Microglia accumulated at the primary infarct site at 28 days, and spread to the connected region in the ipsilateral and contralateral thalamus at 150 days (Gerhard et al., 2005). Another study examined 4 patients with ischemic stroke showed that activated microglia increased in the first week both in the infarct zone and peri-infarct region, but decreased over time. Microglia activation could persist for up to 14 weeks in the peri-infarct region, which is longer than that in the infarct zone, indicating a longer neuro-inflammatory activity in the peri-infarct region (Gulyas et al., 2012). A clinical study of 18 patients who experienced a first-ever sub-cortical stroke indicated that activated microglia in the infarct zone or in the area far from infarct zone display different association with anterograde pyramidal tract damage (Thiel et al., 2010). The extent of microglia activation in the remote area is related to the amount of anterograde tract damage in the first week of ischemic stroke, while activated microglia in the infarct zone is related to anterograde tract damage 6 months after brain ischemia. Activated microglia in the infarct zone has a negative correlation with clinical outcome, but in the remote area, activated microglia has a positive correlation with clinical outcome (Thiel et al., 2010). The difference of dynamic changes of microglia activation in the infarct zone and in the peri-infarct area observed in different studies mentioned above might be due to different characteristics of patients recruited. More patients should be recruited to obtain more longitudinal data and information on the spatiotemporal evolution of microglial response (Thiel and Heiss, 2011). Whether microglia activation in the infarct zone or in the peri-infarct region is beneficial or detrimental for the recovery of ischemic stroke is controversial (Lai and Todd, 2006). On one hand, activated microglia in the infarct zone could exacerbate delayed neuronal death via producing toxic substances. On the other hand, activated microglia contribute to neuronal regeneration via producing growth factors such as BDNF (Madinier et al., 2009). Microglia could also protect the brain via removing debris from the infarct
zone (Stoll et al., 1998; Thiel et al., 2010). These findings suggested that activated microglia in the extended period after ischemic stroke might be an intervention target to limit late neuronal damage and improve outcome (Price et al., 2006; Thiel and Heiss, 2011). However, the exact role of microglia activation in the infarct zone and in the peri-infarct area in stroke recovery needs to be further studied.
281 282 283 284 285 286 287
3.2.2. Microglia activation in experimental ischemic stroke
288
3.2.2.1. Diversity of microglia morphology in the brain ischemic region. Microglia activation has been extensively studied in animal models of ischemic stroke. Microglia activation appears as an early, sensitive, and reliable signal for neuronal damage (Gehrmann et al., 1992). During the different courses of reperfusion after brain ischemia (from 1 to 14 days), the number and maximum length of microglia processes change in multiple ways (Korematsu et al., 1994; Korzhevskii et al., 2012; Morrison and Filosa, 2013). The diversity of activated microglia morphology is correlated to microglia function and could reflect the severity of ischemia in certain circumstances. For example, in a mouse model of focal cerebral ischemia, a study observed morphological changes of microglia during the course of brain ischemia. Classical resting microglia with fine ramified processes were found in the normal cerebral cortex. At 1 to 3 days after ischemia, ‘‘stellate’’ microglia with intense and shortened processes were mainly found in the peri-infarct region. At 6 days after MCAO, ‘‘amoeboid’’ microglia with a feature of large and round cell body but no processes predominated the region close to the infarct zone (Schroeter et al., 1997). Consistent with these observations, another study showed that ramified microglia and ‘‘amoeboid’’ microglia were found surrounding intact and dying neurons after ischemic stroke, respectively (Perego et al., 2011; Zhang et al., 1997).
289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311
3.2.2.2. Activation periods of brain resident microglia and blood immune cells. After ischemic brain injury, neuro-inflammation rapidly increases and contributes to irreversible neuronal damage (Tobin et al., 2014). Both brain resident microglia and blood immune cells are involved in mediating post-stroke neuroinflammation (Aktas et al., 2007). However, there are different activated periods of brain resident microglia and blood immune cells after brain ischemia. Studies demonstrated that microglia are the dominating cell type in post-stroke neuro-inflammation in the first week after ischemic stroke (Schilling et al., 2003; Schroeter et al., 1997; Thiel and Heiss, 2011). Using a mouse model of focal cerebral ischemia induced by photo thrombosis, a study showed that brain resident microglia played a major role in phagocytosis within three days, while bone marrow derived macrophages participated in the removal of necrotic tissue at 6 days after cerebral ischemia (Schroeter et al., 1997). Using a bone marrow transplantation approach to distinguish brain resident microglia from bone marrow derived macrophages, a study revealed that brain resident microglia, defined as F4/80+ and GFP, could be activated and proliferate as early as 24 h after ischemic attack. Microglia activation peaks at 4 days and then gradually decreases to baseline level until 28 days. Whereas F4/80+ and GFP+ macrophages begin to invade into the brain starting at 4 days and peaked at 7 days. The majority of macrophages in the infarct zone are GFP, suggesting that brain resident microglia carry out predominant defense function during brain ischemia other than blood derived macrophages (Schilling et al., 2003). Microglia and macrophages can be distinguished from each other based on the different expression levels of CD45 using flow cytometry (Campanella et al., 2002; Sedgwick et al., 1998). Cells identified as CD45low CD11b+, CD45intermediate CD11b+, or CD45high CD11b+ represents resting microglia, activated microglia, or macrophages,
312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359
respectively (Gelderblom et al., 2009). Both microglia and macrophages are present in the ischemic hemisphere 3 days after MCAO (Gelderblom et al., 2009). However, another study using similar method showed different results, in which both microglia (CD45dim/CD11b+) and macrophages (CD45high/CD11b+/CD11c) persistently increase at 7, 14, and 28 days after MCAO (Stubbe et al., 2013). The different results regarding the peak time of microglia activation and infiltration of blood immune cells could be due to differences in detection methods, ischemia time, and experimental models. For example, permanent cerebral ischemia causes more pronounced microglia activation at 1 and 5 days after ischemia than transient focal cerebral ischemia. Moreover, blood immune cells infiltration occur early in permanent cerebral ischemia (Chu et al., 2014; Zhou et al., 2013). It is essential to develop novel and sensitive detection methods to explore the role of microglia activation in comparable models.
360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408
3.2.3. Receptors mediating microglia activation and function As mentioned above, different pathological conditions could affect microglia activation. The question is what controls and mediates microglia activation? In the past few years, scientists found that many surface receptors are involved in mediating microglia activation and its functions including inflammation, migration, phagocytosis, motility, and survival during ischemic stroke. Toll-like receptors (TLRs) and purinergic receptors are two widely studied receptors essential for microglia-mediated inflammation. Stimulation of Toll-like receptor 2 (TLR2) and Toll-like receptor 4 (TLR4) activated microglia, mediated pro-inflammatory cytokine production, and caused brain damage after focal cerebral ischemia (Caso et al., 2007; Kilic et al., 2008; Lehnardt et al., 2003, 2007; Rosenberger et al., 2014; Yenari et al., 2010). Deficiency of TLR2 or TLR4 reduced TNF-a, iNOS, and COX2 production, contributing to reduced brain infarct volume and improved neurological outcome (Pradillo et al., 2009; Tang et al., 2007). Recent study indicated that inhibition of TLR2 and TLR4 could be an effective method to reduce brain damage within 6 h after ischemic stroke (Wang et al., 2014). IL-17, which is mainly secreted by Th17 cells, gd T cells, and innate lymphoid cells group 3, is a major contributor to the delayed inflammatory response of ischemia stroke (Roy et al., 2005). Microglia could secrete IL-17 upon activation of TLR2/IL-23/IL-17 signal pathway after brain ischemia (Lv et al., 2011). Microglia upregulate IL-17 specific receptor IL-17RA when TLR is activated and produce more IL-17 in an autocrine manner, contributing to neuronal damage (Kawanokuchi et al., 2008; Liu et al., 2014a; Sonobe et al., 2008; Wang et al., 2009). Blocking IL-17 reduced brain infarct volume and neurological deficiency (Gelderblom et al., 2012), possibly due to the fact that IL-17 can impair neural stem cells (NSCs) proliferation and differentiation (Li et al., 2013). Therefore, TLR2/IL-23/IL-17 pathway could be a potential target for dampening delayed inflammatory response and improving neurological outcome after brain ischemia. Purinergic receptors are constituted of P1 adenosine receptors and P2 ATP receptors (Hu et al., 2014). Among P1 adenosine receptors, A2A receptor is up-regulated in microglia after focal cerebral ischemia and is involved in controlling microglia proliferation and BDNF release induced by LPS stimulation (Gomes et al., 2013; Trincavelli et al., 2008). A2A receptor antagonists or genetic depletion of A2A receptor attenuated ischemia induced brain damage (Rivera-Oliver and Diaz-Rios, 2014; Yang et al., 2013). P2X receptors are composed of seven distinct subunit subtypes (P2X1 to P2X7) (Abbracchio et al., 2009; Burnstock, 2008). P2X4 and P2X7 receptors are predominantly expressed on microglia and mediate microglia activation after cerebral ischemia, hypoxia or hypoxia-ischemia (Burnstock, 2007; Cavaliere et al., 2003; de Rivero Vaccari et al., 2012; Del Puerto
5
et al., 2013; Li et al., 2011; Wixey et al., 2009). P2X7 receptor activation is responsible for inducing microglia to release proinflammatory cytokines such as TNF-a, IL-1b, NO, CXCL2, and CCL (Gendron et al., 2003; Hide et al., 2000; Kataoka et al., 2009; Shiratori et al., 2010). Blocking P2X7 receptor reduced proinflammatory cytokines IL-1b, TNF-a and IL-6 release, neuronal death, and neurological deficit after global cerebral ischemia (Chu et al., 2012). P2X7 receptor antagonist treatment reduced microglia death under Oxygen–Glucose Deprivation (OGD) (Eyo et al., 2013). In contrast, the exact role of P2X4 receptor in cerebral ischemia is less well studied. P2Y12 is another type of purinergic receptors expressed on microglia. P2Y12 is down-regulated after microglia activation. Deficiency of P2Y12 in mice impaired microglia migration, polarization, and its ability to extend their processes toward the lesion site, where nucleotides are released by damaged brain tissue (Haynes et al., 2006). Recently, P2Y12 was found to mediate microglia neurotoxicity. P2Y12 knockout reduced microglia accumulation in the peri-infarct region and reduced neuronal death after cerebral ischemia in mice (Webster et al., 2013). Collectively, these studies suggested that TLRs and the purinergic receptors could be potential targets for controlling microglia activation and reducing post-stroke inflammation mediated by microglia. The chemokine receptor CCR2 is highly expressed in nearly all immune cells and inflammatory monocytes (Ajami et al., 2011). However, CCR2 is expressed at a low level in the brain microglia under normal condition (Hu et al., 2014; Savarin-Vuaillat and Ransohoff, 2007). After cerebral ischemia, CCR2 and its ligand monocyte chemoattractant protein-1 (MCP-1) are up-regulated in microglia (Inose et al., 2014). Activation of CCR2 is responsible for enhancing cerebral inflammation and brain infarct volume (Dimitrijevic et al., 2007). In a model of hypoxia in neonatal rats, administration of MCP-1 induced microglia migration from adjacent area toward the injection site in peri-ventricular white matter (Deng et al., 2009). However, absence of CCR2 in mice mainly reduced the recruitment of blood immune cells but did not affect brain resident microglia activation after transient MCAO (Schilling et al., 2009). Thus, CCR2 seems to be critical for blood immune cells recruitment but not essential for microglia activation after focal brain ischemia. Triggering receptor expressed on myeloid cells-2 (TREM-2), which is a member of triggering receptors expressed on myeloid cells (TREM) family, is expressed on microglia (Schmid et al., 2002). TREM-2 activation promotes microglia migration and phagocytosis of apoptotic neurons without inducing inflammatory cytokines expression (Takahashi et al., 2005). After cerebral ischemia, TREM-2 is up-regulated in microglia (Heldmann et al., 2011; Sugimoto et al., 2014). However, up to now, the role of TREM-2 in cerebral ischemia is largely unknown. Fc receptors (FcRs) are also involved in regulating microglia phagocytosis. In a mouse model of Alzheimer’s disease (AD), microglia were found capable of phagocytose extracellular amyloid plaques via FcRs (Bard et al., 2000). In addition, FcRs expressed on microglia also mediate inflammatory cascade and produce superoxide and NO (Le et al., 2001; Ravetch, 1994; Sylvestre and Ravetch, 1994). FcRs knockout in a transient MCAO model inhibited microglia activation and iNOS production, contributing to reduced infarct volume and mouse mortality at 3 days after MCAO (Komine-Kobayashi et al., 2004). This study suggested that interventions targeting FcRs in the acute phase of brain ischemia might be beneficial for attenuating ischemia induced brain injury. However, whether FcRs knockout exerts beneficial function during the recovery phase of brain ischemia is still unknown and needs further study. The receptor for advanced glycation end products (RAGE) is another receptor which mediates microglia activation and inflammatory response in many diseases (Fang et al., 2010; Lue
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474
G Model
PRONEU 1417 1–25 6
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529
et al., 2001; Ramasamy et al., 2009; Yamagishi and Matsui, 2010; Zhai et al., 2008). In ischemic stroke patients, RAGE is up-regulated in the brain and plasma (Menini et al., 2014; Zhai et al., 2008). The interaction between RAGE and its ligand high mobility group box1 (HMGB1) is involved in exacerbating ischemia induced brain damage, since anti-HMGB1 or RAGE depletion treatment reduced brain infarct volume (Muhammad et al., 2008). In vitro study showed that HMGB1-RAGE interaction is essential for microglia activation induced neuronal death (Muhammad et al., 2008). Recent studies indicated that cysteinyl leukotriene receptor 2 is also involved in microglia mediated inflammatory response and neuronal death after brain ischemia. Selective inhibition of cysteinyl leukotriene receptor 2 using HAMI 3379 via intraventricular injection before ischemia reduced microglia activation and brain injury within 3 days after MCAO (Shi et al., 2012, 2015; Zhao et al., 2011). Thus, HAMI 3379 treatment could be a protective strategy for attenuating microglia mediated inflammatory response at the acute and subacute phase of ischemic stroke. Both Galectin-3 and CD36 belong to pattern recognition receptors (PRR) expressed on microglia. Galectin-3 is required for microglia proliferation, while CD36 is involved in phagocytosis of dying neuronal cells and mediates inflammatory response. Lack of Galectin-3 reduced microglia proliferation, which is associated with enhanced infarct volume and neuronal apoptosis after MCAO (Lalancette-Hebert et al., 2012). Similarly, lack of CD36 in mice caused poorer neurological outcome at 1 day after MCAO, as a result of exacerbated inflammatory response and reduced removal of neuronal debris (Woo et al., 2012). These studies suggested that Galectin-3 and CD36 play a positive role in acute stroke. Sema4D, another molecule found to mediate microglia activation via interacting with its receptors Plexin B1 and CD72 expressed on microglia (Okuno et al., 2010). Sema4D is up-regulated in the periinfarct region of the ischemic brain (Taniguchi et al., 2009). Sema4D deficiency caused an increase in the number of oligodendrocytes in healthy and injured mouse brain. In Sema4D/ mice, the number of amoeboid microglia and iNOS production in the peri-infarct area of the cortex were both reduced after permanent cerebral ischemia, accompanied by better neurological outcome (Sawano et al., 2015). These studies demonstrated that a large number of receptors expressed on microglia contribute to regulating microglia activation under ischemic environment. Many studies investigated the potential of targeting these receptors to treat neurological diseases (Binder et al., 2011; Clark et al., 2007; Tsiperson et al., 2010; Yenari et al., 2010). However, whether treatment strategies targeting these receptors could be developed into effective treatments for brain ischemic disease awaits further study because other cell expressing the identical receptors would also be affected. Furthermore, when and how long should these receptors be blocked or activated also need to be considered. Inhibition of microglia activation for a long term might cause dysfunction of innate immune response in the brain (Hu et al., 2014). Further understanding of the activity of receptors expressed on microglia would facilitate the development of novel and effective therapeutic strategies for the treatment of ischemic stroke.
530
4. Roles of activated microglia in ischemic stroke (Table 1)
531
4.1. Microglia and blood brain barrier permeability
532 533 534 535 536 537
Blood brain barrier (BBB) disruption plays a critical role in the pathogenesis of ischemic stroke (da Fonseca et al., 2014; Yang and Rosenberg, 2011). BBB disruption leads to cerebral vasogenic edema, hemorrhagic transformation, leukocytes infiltration, and the leakage of toxic molecules into the brain. Early phase BBB disruption occurs at 12–24 h after ischemia, and the delayed
secondary phase occurs at 48–72 h. During this period, microglia are activated and affect BBB disruption (da Fonseca et al., 2014). A recent study dynamically observed microglia activation within 72 h after MCAO using time lapse two-photon imaging (Jolivel et al., 2015). Microglia in the penumbra rapidly expanded cellular protrusions toward blood vessels, with morphological changed over time. Increased number of microglia was accompanied by decreased blood vessels. Immunostaining results showed that Iba1+ microglia colocalize with CD31+/Gul+/caveolin-1+/claudin5+/podocalyxin+ blood vessels, indicating that activated microglia phagocytosed endothelial cells. This phenomenon is associated with BBB disruption, confirmed by high levels of IgG and Evans blue extravasation, and high matrix metalloproteinase-9 expression. CX3CR1 knockout mice, in which microglia function is impaired, were used to confirm the role of microglia in BBB disruption after focal cerebral ischemia. CX3CR1 knockout mice displayed a reduced infarct size and contrast agent extravasation (observed by MRI). Inhibition of microglia activation by minocycline treatment after MCAO reduced infarct volume, hemorrhagic transformation, and BBB permeability, partly via preventing the degradation of collagen IV and laminin (Machado et al., 2009; Tikka et al., 2001; Yenari et al., 2006). Moreover, inhibiting microglia activation increased blood vessel integrity (Jolivel et al., 2015). Collectively, these studies suggested that microglia activation could induce BBB disruption after brain ischemia through disintegration of blood vessels in the penumbra. Tables 1 and 2. Several mechanisms and molecules are associated with microglia mediated BBB disruption. Oxidative stress, generally caused by excess production of reactive oxygen species (ROS), is considered a key contributor to the early phase of BBB disruption (Obermeier et al., 2013). Microglia can be activated by ROS and subsequently produce more ROS. The high amount of ROS could in turn amplify microglia activation, injure endothelial cells, and exacerbate BBB disruption (Kacimi et al., 2011). Hypoxia-inducible factor-1 and NF-kB are up-regulated in microglia after cerebral ischemia. The up-regulation of Hypoxia-inducible factor-1 and NFkB could stimulate microglia to produce matrix metalloproteinases, which are key mediators in the delayed secondary phase of BBB disruption (da Fonseca et al., 2014). Pro-inflammatory cytokines such as IL-1b, IL-6, and TNF-a, are released by microglia and up-regulate after ischemic stroke (Lee et al., 2014; Zhou et al., 2013). Recently, our laboratory showed that microglia activation could up-regulate aquaporin-4 expression in astrocytes by releasing IL-1b, IL-6, and TNF-a, causing enhanced astrocyte apoptosis and BBB disruption. P38 signaling pathway was found to be involved in microglia mediated BBB disruption. P38 inhibitor treatment immediately after ischemic reperfusion reduced aquaporin-4 expression in the mouse brain. MSCs transplantation inhibits the microglia mediated aquaporin-4 up-regulation and astrocyte apoptosis, thus contributing to maintaining the integrity of BBB (Tang et al., 2014). Up to now, there are only limited studies focusing on the interaction between microglia activation and BBB disruption. Some studies showed that blood protein fibrinogen could deposit in the area of activated microglia after BBB disruption and cause microglia activation through interacting with Mac-1 on microglia (Adams et al., 2007; Gay et al., 1997), Thus, BBB disruption could cause microglia activation in reverse. However, microglia possibly play a protective role on attenuating BBB disruption via secreting progranulin, which is involved in reducing BBB disruption and brain edema after ischemic stroke (Egashira et al., 2013; Jackman et al., 2013; Kanazawa et al., 2015). Therefore, the causal relationship between microglia activation and BBB disruption is still elusive. The effect of microglia activation on BBB disruption remains to be further explored.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603
Model
Methods or treatment
Injection time
marker
Observe location
Observe time
outcome
Beneficial/ detrimental
Reference
Rats PGRN/ mice
tMCAO 1.5 h
100 mg of PGRN was injected via inguinal vein
4 h after MCAO
Iba1
Peri-infarct zone
4,10,18,24,72 h
Beneficial
Kanazawa et al. (2015)
Rats
tMCAO 2 h
4 h after MCAO
Iba1
Peri-infarct zone of cortex
0.5,1,3,7 d
Detrimental
van Minnen et al.
Sema4D/ mice
pMCAO
Ginsenoside Rd treatment via intralventrical injection Depletion of Sema4D in mice
Detrimental
Sawano et al. (2015)
Rats
tMCAO 1 h
CYSLT2R antagonist administraion
1, 3 d before MCAO Or 0.5 h before MCAO
ED1 Iba1
Ischemic core peri-infart zone in the cortex
1,3 d
Detrimental
Haan et al. (2015)
Mice
20 min BCCAO
Minocycline treatment via intralventrical injection
Immediately after reperfusion
OX-42
Dentate gyrus
1,3,7,14,21,28 d
PGRN knock out do not affect the number of microglia, but reduce brain edema Reduced microglia is accompanied by enhanced neurological function Reduce iNOS production and neurological function deficency Reduce microglia activation, neurological deficit, and infarct volume Reduce neurogenesis
Beneficial
Choi et al. (2015)
18 month-old rats
tMCAO 50 min
ED1 Iba1
peri-infarct area
3,14&28 d
Detrimental
Titova et al. (2014)
Postnatal rats
Normal
Beneficial
ShigemotoMogami et al. (2014)
Mice
tMCAO 1.5 h
Detrimental
Li et al. (2014)
MKP-1 knockout mice
tMCAO 1 h
Beneficial
Li et al. (2014)
Rats CX3CR1-GFP mice Nrf2/ mice and LysMCreHmox 1D/D mice Galectin-3 knockout mice
pMCAO
Detrimental
Gelosa et al. (2014) Parada et al. (2013)
Q11
Photothrombotic stroke
Peri-infarct zone of cortex
Minocycline (30 mg/ kg) treatment via intraperitoneal injection Adjudin (50 mg/kg) treatment via intraperitoneal injection Depletion of MKP-1 in microglia
Begin on postnatal day 2 (P2).for 3 d
CD11b CD68
SVZ
P1,P4,P10,P14,P30
Immediately,5&24 h after reperfusion
CD11b
Cortex striatum
3d
Ticagrelor 3 mg/kg treatment per os a7 nAChR agonist PNU282987 treatment
10 min, 22&36 h after MCAO 1h postphotothrombosis
tMCAO 1 h
Depletion of Galectin3
Rats
Endothelin-1 induced cerebral ischemia
Seven-day-old rats
tMCAO 1.5 h
BMMCs transplantation or compared with minocycline via caudal vein, CB2R and CB1R agonist WIN 55,212-2 (WIN) treatment via subcutaneous injection
Iba1
ED1/Iba1
3d
Peri-infarct area infarct core
Iba1
2, 24, 48 & 72 h 24 h
Reduced microglia is accompanied by enhanced neovascularization Microglia inhibition reduce neurogenesis and oligodendrogenesis Reduced microglia activation is associated with reduced BBB disruption Depletion of MKP-1 in microglia enhance infarct size and neurological deficit Enhance neurological deficit Reduce infarct size and improve motor skills
Beneficial
Mac-2 Iba1
Infarct region
1&3 d
Reduce infarct size and improve motor skills
Beneficial
5 106 cells, 24 h postischemia
ED1
Cortex
7,14&21 d
Reduce infarct size and improve functional recovery
Detrimental
Immediately,4 h after reperfusion twice daily
Iba1
Cortex
24&72 h
Reduce infarct size
Detrimental
LalancetteHebert et al. (2012) Franco et al. (2012)
FernandezLopez et al. (2012)
G Model
PRONEU 1417 1–25
Animal
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
Table 1 Biphasic function of microglia in different models and periods of ischemic stroke.
7
Methods or treatment
Injection time
marker
Observe location
Observe time
outcome
Beneficial/ detrimental
Reference
Seven-day-old rats
tMCAO 3 h
2 d before MCAO
Iba1 IB4 ED1
Ischemic core penumbra
8,24&72 h
Enhance infarct size
Benificial
Faustino et al. (2011)
Rats mice
tMCAO 2 h tMCAO 0.5 h
Immediately after reperfusion; immediately& 1 week later
Mac-1 Iba1 ED1 TREM-2
SVZ striatum
7,14&28 d
Do not affect neurogenesis
No effect
Heldmann et al. (2011)
Hv1 knockout mice CX3CR1-GFP mice Rats
pMCAO tMCAO 2 h
Peri-infarct area
24&72 h
Reduce infarct size and neurological scores
Detrimental
Lozano et al. (2012)
tMCAO 1.5 h
3,5,7&14 d
Reduce infarct size and apoptotic cell
Beneficial
Narantuya et al. (2010a,b)
Rats
tMCAO 2 h
Rats
tMCAO 1.5 h
Rats
Endothelia-1 induced cerebral ischemia tMCAO 1 h
Depletion of microglia via intracerebral injection of liposomeencapsulated clodronate Mac-1-saporin treatment via inntraventricular injection Depletion of voltagegated proton channel Hv1 Microglia transplantation via jugular vein injection Minocycline (90 mg/ kg) treatment via intraperitoneal injection P2X7 receptor agonist BzATP or antagonist OxATP treatment via inntraventricular injection Two-photon microscopy imaging Depletion of CX3CR1
Q12
CX3CR1-GFP CX3CR1 knockout mice Mice
Mongolian gerbil
tMCAO 0.5 h tMCAO 1 h 5 min BCCAO
CD11b-TKmt-30 mice
tMCAO 2 h
Rats
10 min BCCAO
Mice
tMCAO 2 h
Chimeric mice
tMCAO 0.5 h
Rats
tMCAO 2 h
Rats
tMCAO 2 h
3 106 cells,48 h after stroke
ED1
Immediately after reperfusion
Ox6
SVZ peri-infarct zone
1,4&7 d
Reduce neurogenesis
Beneficial
Kim et al. (2009)
0.5 h before reperfusion
CD11b
Cortex striatum
1&3 d
Reduce neurological deficiency
Beneficial
Yanagisawa et al. (2008)
24&48 h after OGD
Reduce neuronal death
Beneficial
Reduce infarct size and apoptotic cells, accelerate recovery Reduced microglia is accompanied by smaller infarct size Increase neuronal survival
Detrimental
Neumann et al. (2008) Denes et al. (2008)
IB4
Magnetic Resonance Imaging
Iba1
Cortex striatum hippocampus
4,24,&72 h
Iba1 lectin
Cortex, striatum
4,24,48&72 h
IB4
Hippocampus
7d
Microglia transplantation via subclavian artery Ganciclovir treatment via intraperitoneal, injection
1 106 cells 24 h before,24 h or 24 h after ischemia, 48 h before&24 to 72 h after stroke
Mac-2 Iba1
Microglia transplantation via subclavian artery injection 45 mg/kg minocycline treatment via intraperitoneal injection
2 106 cells, 4 to 7 days before ischemia
Iba1 Ox42/ CD11b
Hippocampus
2&4 d
0.5&12 h after reperfusion
IB4
Peri-infarct area
24 h
Iba1
Cortex striatum
1,2,4,7,10&14 d
3 d before MCAO, daily
CD11b ED-1
Cortex, striatum
7,14&28 d
5 104 cells, 1 h after reperfusion
Iba1 CD11b
Striatum substantia nigra
8d
Indomethacin (2.5 mg/ kg) treatment via intraperitoneal injection Microglia transplantation via introcerebroventrical injection
3d
Beneficial
Denes et al. (2007)
Beneficial
Imai et al. (2007)
Microglia depletion enhance infarct size and neuronal apoptosis Reduce neuronal death and protect synapse transmission
Beneficial
LalancetteHebert et al. (2007)
Beneficial
Hayashi et al. (2006)
Reduce infarct size, BBB disruption, and neurological deficiency Phagocytosis of neuronal cell debris Enhance neurogenesis
Detrimental
Yenari et al. (2006)
Beneficial
Schilling et al. (2005) Hoehn et al. (2005)
Improve motor function recovery
Beneficial
Detrimental
Kitamura et al. (2005)
G Model
Model
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
Animal
PRONEU 1417 1–25
8
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
Table 1 (Continued )
G Model
PRONEU 1417 1–25
Irradiation Rats
Abbreviations: a7 nAChR, alpha-7 nicotinic acetylcholine receptor; BCCAO, bilateral common carotid artery occlusion; BMNCs, Bone marrow mononuclear cells; BzATP, 20 -30 -O-(4-benzoylbenzoyl)adenosine 50 -triphosphate; CYSLT2R, cysteinyl leukotriene receptor 2; OGD, Oxygen-Glucose Deprivation; OxATP, adenosine 50 -triphosphate-20 ,30 -dialdehyde; PGRN, progranulin; SVZ, sub ventricular zone; tMACO, transient middle cerebral artery occlusion.
Monje et al. (2003) Detrimental 2 months ED1
Dentate gyrus
Microglia activation is accompanied by reduced neurogenesis
Kitamura et al. (2004) Enhance neural survival 3d Infarct core border zone CD11b 4 103 or 4 104 cells, 1 h after reperfusion Microglia transplantation via introcerebroventrical injection
628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667
tMCAO 1 h
605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627
Rats
604
Beneficial
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
9
4.2. The effect of microglia on neurogenesis 4.2.1. Microglia influence neurogenesis under physiological condition Neurogenesis exists in the adult brain, both in human and animals (Ernst et al., 2014; Shors et al., 2001; Tobin et al., 2014). Neurogenesis increases in rodents and primate brain after ischemic stroke (Arvidsson et al., 2002; Jin et al., 2001; Tonchev, 2011; Tsai et al., 2011; Tureyen et al., 2004). Microglia are located in the neurogenic niche and have a complex link with neurogenesis in both healthy and injured brain (Ekdahl, 2012; ShigemotoMogami et al., 2014). Microglia are involved in excise induced neurogenesis in adult mice. The number of brain microglia increased in aged mice and removal of microglia reduced neural progenitor cells (NPCs) activity (Adachi et al., 2010). In postnatal mice, minocycline treatment inhibited microglia activation and reduced neurogenesis in the sub ventricular zone (SVZ) (Shigemoto-Mogami et al., 2014; Vukovic et al., 2012). It is noted that microglia maintain the homeostasis of baseline neurogenic cascade via phagocytosing the new born cells in the sub granular zone of the dentate gyrus in young normal brain. In aged or LPSstimulated rat brain, inhibiting microglia enhanced the number of new born cells and promoted neurogenesis in the sub granular zone (Sierra et al., 2010). These findings indicated that microglia exert different functions on neurogenesis under different physiological conditions. 4.2.2. Microglia influence neurogenesis during ischemic stroke Under pathological conditions including ischemic stroke, microglia exhibit biphasic functions in neurogenesis (Ekdahl et al., 2009). Minocycline treatment for 4 weeks reduced microglia activation, enhanced neurogenesis in dentate gyrus, and improved neurological function after MCAO (Liu et al., 2007). Indomethacin treatment before focal brain ischemia enhanced neurogenesis, which is associated with the inhibition of microglia activation by indomethacin (Hoehn et al., 2005). These findings suggested that microglia activation play a detrimental role in neurogenesis. However, in a rat model of transient cerebral ischemia, minocycline treatment reduced the number of DCX+/BrdU+ cells in the SVZ at 4 days and NeuN+/BrdU+ cells at 7 days after ischemia, though minocycline treatment inhibits microglia activation (Kim et al., 2009). Another study found that microglia accumulating in the peri-infarct striatum peaked at 2 weeks and maintained to 16 weeks after MCAO. IGF-1 mRNA was up-regulated in the SVZ and interestingly only microglia expressed IGF-1 protein in the SVZ. These phenomenon were accompanied by enhanced neurogenesis in the SVZ at 2, 6, and 16 weeks after ischemia, suggesting that microglia are supportive of neurogenesis after ischemic brain injury (Thored et al., 2009). In contrast, a later study showed that microglia depletion did not affect neurogenesis in the SVZ and striatum at 2 weeks after MCAO. DCX+/BrdU+ neuroblasts were examined after the removal of Mac-1+ microglia via Mac-1-saporin intraventricular injection. There was no difference in the number of DCX+/BrdU+ neuroblasts in the SVZ and striatum between Mac1-saporin treated group and control group (Heldmann et al., 2011). These controversial results could be due to the different observation time of neurogenesis, since the later study only focused on two weeks after MCAO, while the former observed up to 16 weeks. A study with longer observation time with multiple time points is needed to clarify the effect of microglia on neurogenesis after ischemic stroke. In addition, different stimulation methods of microglia activation may contribute to these opposite effects on neurogenesis. For example, microglia activation induced by bacterial endotoxin, lipopolysaccharide (LPS), or cranial irradiation all cause severe inflammatory response and inhibit neurogenesis in hippocampus or striatum (Ekdahl et al., 2003). While IL-4 or IFN-g induced microglia activation support neurogenesis or oligoden-
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
10
Table 2 miRNAs associated with microglia activation in different pathological conditions. miRNA
Target
Function
Treatment
Model
Reference
miR-155
SOCS-1
Butovsky et al. (2015)
–
Knock out of miR-155 in mice Ab stimulation
ALS
miR-155
AD
Guedes et al. (2014)
miR-155
c-Maf
Knock out of miR-155 in mice
MCAO
Bsibsi et al. (2014)
miR-155
–
LPS stimulation
Primary microglia
Freilich et al. (2013)
miR-155
SOCS-1
LPS stimulation
N9 cells
Cardoso et al. (2012)
miR-124
–
miR-124 deficiency
zebrafish
Svahn et al. (2015)
miR-Let7A
–
Knock out of miR-Let7A
BV2 cells
Choi et al. (2015)
miR-Let7c-5p
caspase 3
miR-Let7c-5p overexpression
MCAO
miR-204
SIRT1
Inhibition of miR-204 under LPS stimulation
N9 and BV2 cells
Albiero et al. (2015)
miR-203
MyD88
Promote microglia activation Promote IL-6 and IFN-b production Promote proinflammatory cytokines production Mediate proinflammatory pathways Promote iNOS and proinflammatory cytokines production Reduce motility and phagocytosis capacity of microglia Modulate autophagy activity Inhibit microglia activation and reduce brain injury Promote inflammatory response and proliferation of microglia Attenuate brain injury
MCAO
Haan et al. (2015)
miR-Let-7a
-
miR-181c
TLR4
miR-124
p65 TRAF6
inhibit iNOS and IL-6 expression, enhance BDNF expression reduce TNF-a, IL-1b, and iNOS production reduce TNF-a, IL-1b, and IL-6 production
Overexpression of miR203 Overexpression of Let-7a under LPS stimulation
miR-99a, 125-5p, and 342-3p miR-9
–
–
–
Human microglia
Qiu et al. (2015) See comment in PubMed Commons below Butovsky et al. (2014)
MCPIP1
Overexpression of miR-9
Primary microglia
Li et al. (2014)
miR-17
Jadhav et al. (2014b)
Traumatic brain injury
Jadhav et al. (2014a)
miR-124
–
HIV-1 Tat C protein treatment Overexpression of miR200b IL-4 stimulation
HMC3
miR-200b
NOX2 and NOX4 c-Jun
Mediate inflammatory response Reduce ROS production
Primary microglia
Freilich et al. (2013)
miR-29b
TNFAIP3
Japanese encephalitis virus treatment
Primary microglia And BV2 cells
Thounaojam et al. (2014)
miR-29a,b
–
Aged humans microglia and primary microglia MCAO
Fenn et al. (2013)
miR-424
IGF-1 and CX3CL1 –
miR-181c
TNF-a
miR-21
FasL
miR-Let7f
IGF-1
miR-146a
–
Reduce microglia activation, Reduce brain infarct volume –
miR-124
–
Reduce microglia activity
miR-146a
MCP-2
Reduce MCP-2 expression
Reduce iNOS and NO production Mediate antiinflammatory pathways Inhibit iNOS, COX-2, and pro-inflammatory cytokine production Inhibit microglia activation Reduce microglia activation, brain infarct volume, and neuronal apoptosis Reduce TNF-a production
OGD Morphine treatment
Overexpression of miR424
OGD OGD Anti-miR-Let7f treatment Ab42 peptide and TNF-a stimulation Knock out or overexpression of miR124 HIV infection
BV2 cells
Primary microglia and BV2 cells Primary microglia and BV2 cells
van Minnen et al.
Zhao et al. (2013)
Primary microglia and BV2 cells Primary microglia and BV2 cells MCAO
Zhang et al. (2012a)
Selvamani et al. (2012)
HMG
Ajami et al. (2011)
EAE
Ponomarev et al. (2011)
Primary human fetal microglia
Felger et al. (2010)
Zhang et al. (2012b)
Abbreviations: AD, Alzheimer’s disease; ALS, amyotrophic lateral sclerosis; COX-2, cyclooxygenase-2; EAE, Experimental Autoimmune Encephalomyelitis; HMC3, human microglia clone 3 cell lines; HMG, Human microglial cells; FasL, Fas ligand; MCP-2, monocyte chemoattractant protein 2; MCAO, middle cerebral artery occlusion; MCPIP1, monocyte chemotactic protein-induced protein 1; OGD, Oxygen-Glucose Deprivation; SIRT1, Sirtuin1; SOCS-1, suppressor of cytokine signaling 1; TLR4, Toll-like receptor 4; TNFAIP3, tumor necrosis factor alpha-induced protein 3; TRAF6, TNFR-associated factor 6.
668 669 670 671
drogenesis from adult NPCs. Co-culturing NPCs with microglia stimulated with IL-4 or IFN-g promoted NPCs differentiation into Tuj1+ neurons and NG2+ oligodendrocytes. It was suggested that IL-4 stimulated microglia promoted NPCs differentiation into
oligodendrocytes partly via secreting IGF-1, because anti-IGF-1 treatment effectively blocked NPCs differentiation (Butovsky et al., 2006). These studies indicated that different activating stages of microglia may have different effects on neurogenesis.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
672 673 674 675
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
676 677 678 679 680
In conclusion, it is increasingly apparent that microglia activation plays a biphasic role in neurogenesis. Elucidating when and how microglia influence neurogenesis will be helpful for advancing our understanding of brain injury and repair after ischemic stroke (Tobin et al., 2014).
681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728
4.2.3. Interaction between activated microglia and NPCs As mentioned above, microglia exert positive or negative effect on neurogenesis in vivo. A biphasic role regarding the influence of microglia on neurogenesis was also found in vitro. Conditioned medium from primary microglia or co-culturing with microglia enhanced the capacity of NPCs to generate neuroblasts and differentiate into neurons (Aarum et al., 2003; Walton et al., 2006). These studies suggested that microglia could be beneficial and supportive for NPCs differentiation via releasing neurotrophic factors. Another study further supported the paracrine function of microglia on NPCs. Depletion of microglia from cultured hippocampal slice reduced NPCs survival and proliferation while cocultured with microglia or conditioned medium from microglia reversed the phenomenon. In addition, the study found that microglia directly targeted NPCs via releasing IL-4 (Nunan et al., 2014). However, microglia could inhibit the proliferation of NPCs via releasing NO. NPCs co-cultured with microglia from iNOS knockout mice (iNOS/) displayed enhanced proliferation compared with that co-cultured with microglia from wild type mice (iNOS+/+) (Carreira et al., 2014). Interestingly, increasing numbers of studies indicated that microglia activation could be regulated by NPCs. Using GFAPCre/bCateninEx3 transgenic mice, in which CXCL12 expressing progenitor cells were abolished, a study found that microglia were parallelly reduced in the ventricular zone (VZ)/SVZ of developing cerebral cortex. The apoptosis of NPCs in transgenic mice controlled by suicide gene Thymidine Kinase (TK) and Ganciclovir (GCV) treatment induced microglia migration into the VZ/SVZ and exerted phagocytosis function. Moreover, using transgenic mice lacking CSF-1R to deplete microglia inhibited NPCs migration into the cerebral cortex, indicating that NPCs activity could drive microglia migration into the VZ/SVZ in the developing brain (Arno et al., 2014). In turn, microglia are essential for NPCs development. Another study explored the regulation effect of NPCs on microglia activation (Mosher et al., 2012). Microglia and NPCs displayed a close correlation in their positions in the brain. The number of microglia in the SVZ and dentate gyrus, which are origins of NPCs (Kokaia et al., 2012), were higher than that in the cerebral cortex in mice, suggesting a functional relationship between microglia and NPCs. Co-culturing primary microglia or BV2 cells with NPCs enhanced microglia proliferation, migration, and phagocytosis. Further experiments showed that NPCs regulate microglia activation via releasing VEGF. Intrastraital injection of conditioned medium from NPCs or VEGF alone promoted microglia proliferation, which could be blocked by knocking down VEGF in NPCs via lentiviral shRNA. These studies suggested that microglia and NPCs cross-regulate each other, but the regulation mechanisms need to be further explored.
729
5. Microglia polarization during ischemic stroke
730
5.1. Microglia polarization
731 732 733 734 735 736 737
Macrophages could adopt different phenotypes depending on the stimulus, the period, and the environment (Biswas and Mantovani, 2010; Gordon and Taylor, 2005). The process is called polarization. Similar to macrophages, microglia could also polarize to many phenotypes, which was demonstrated in vitro and in vivo (Girard et al., 2013). Based on the stimulation of pro-inflammatory or anti-inflammatory cytokines, microglia could generally polarize
11
into inflammatory and anti-inflammatory phenotypes (Gordon, 2003). Inflammatory phenotype is designated as classically activated microglia and could be induced by IFN-g and LPS stimulation (Nakagawa and Chiba, 2014). Inflammatory phenotype, also known as M1 phenotype, is characterized by the production of a variety of pro-inflammatory cytokines (IL-1b, IL-6, TNF-a, CCL2, and CXCL10), ROS, NO, and proteolytic enzymes matrix metalloproteinase-9 and matrix metalloproteinase-3 (Kettenmann et al., 2011; MacMicking et al., 1997; Saijo and Glass, 2011; Varnum and Ikezu, 2012; Yenari et al., 2010). The functions of classically activated microglia are to present antigens and kill intracellular pathogens to maintain environmental homeostasis (Cherry et al., 2014). However, in vitro studies showed that LPS stimulation induced classically activated microglia (an inflammatory phenotype) exacerbated neuronal injury under OGD condition and inhibited neurogenesis in the hippocampus (Hu et al., 2012; Walter et al., 2011). Anti-inflammatory phenotype, known as M2 phenotype, is the alternatively activated microglia. Based on different stimulation and function, M2 phenotype is further divided into three sub-class including M2a, M2b, and M2c (Chhor et al., 2013; Latta et al., 2015; Sudduth et al., 2013). M2a microglia could be induced by IL-4 or IL13 stimulation and are mainly considered to suppress inflammation. M2a microglia display enhanced expression of Arginase-1, Ym1, IGF-1, CD206, chitinase 3-like 3 and Fizz1 (Latta et al., 2015; Mecha et al., 2015). Intracerebral injection of IL-4 or IL-13 in the brain enhanced Arginase-1, Ym1, CD206 expression (Girard et al., 2013), supporting that IL-4 and IL-13 could promote microglia polarization into M2a phenotype. Unlike M2a phenotype, M2b phenotype does not express Arginase-1, Ym1, and Fizz1. M2b phenotype could be induced by immune complexes and TLRs agonists, and IL-1R ligands and shows increased expression of IL1RA, CD86, and SOCS3 (Chhor et al., 2013; Mecha et al., 2015). However, the role of M2b phenotype is less understood compared to M2a phenotype. The last sub-class is M2c phenotype and could be induced by TGF-b, IL-10, and glucocorticoids. M2c phenotype is also called as ‘‘deactivated microglia’’ (Chhor et al., 2013; Mecha et al., 2015). Instead of having no function, M2c phenotype is involved in tissue regeneration when inflammatory response become weakened (Mantovani et al., 2004). Anti-inflammatory phenotype is capable of producing antiinflammatory cytokines IL-10, TGF-b, IL-4, IL-13, and IGF-1, as well as expressing scavenge receptors, contributing to inhibiting inflammation and promoting tissue repair (Cherry et al., 2014; Ponomarev et al., 2013). Generally, anti-inflammatory phenotype is considered as having a protective function. A study showed that treatment with M2 conditioned media enhanced neuronal survival and process extension after spinal cord injury (Kigerl et al., 2009). Conditioned medium from M2 phenotype reduced neuronal death in brain slices under OGD condition. These results suggested that promoting M2 phenotype polarization is helpful for facilitating tissue regeneration after injury.
738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789
5.2. Ischemia induces microglia polarization
790
Microglia polarization could either exacerbate damage or promote tissue repair, which is partially dependent on different pathophysiological conditions (David and Kroner, 2011; O’Neill and Hardie, 2013). Furthermore, microglia polarization could be affected by the time and severity of injury, aging, and microenvironment (Hu et al., 2015; Norden and Godbout, 2013; Perego et al., 2011). After ischemic stroke, there is dynamic microglia polarization in the peri-infarct region (cortex and striatum). CD206+ M2 microglia were observed in the peri-infarct region as early as day 1, with a transient elevation within 7 days after MCAO, and then gradually decreased. In contrast, CD16/32+ M1 microglia appeared
791 792 793 794 795 796 797 798 799 800 801
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 12
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826
at 3 days and gradually increased until 14 days after MCAO. The mRNA expression of M1 microglia markers iNOS, CD11b, CD16, CD32, and CD86 and M2 microglia markers CD206, Arginase-1, CCL22, Ym1/2, IL-10, and TGF-b were consistent with the immunostaining results mentioned above (Hu et al., 2012). In vitro study showed that neurons injured by ischemia could induce microglia to polarize toward M1 phenotype. Conditioned medium from M1 microglia enhanced neuronal death while M2 microglia protected neurons against OGD (Hu et al., 2012). Different phenotypes of microglia could differentially modulate cell death after brain injury (Girard et al., 2013). Adding M1 or M2 microglia into brain slices after OGD treatment, researchers found that M1 microglia was detrimental for the survival of neurons, while M2 microglia was protective in reducing neuronal death in the CA3 and CA1 region of the hippocampus after OGD treatment. In a transient MCAO model, there was no apparent microglia polarization within 24 h after ischemia (Girard et al., 2013). These results suggested that microglia polarization might exert different functions during the pathology of ischemic stroke. Inhibiting microglia activation as a therapeutic strategy should be thought over, considering it would compromise the beneficial role of microglia for the recovery and tissue repair of ischemia induced brain injury. Future studies are required to explore the function of microglia polarization to guide the development of therapeutic strategies for ischemic stroke.
827
5.3. Substances involved in the modulation of microglia polarization
828 829 830
Many substances could modulate microglia polarization, contributing to improved functional recovery or exacerbated brain injury.
831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864
5.3.1. Clinical drugs Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) is a peptide with neuroprotective function. A recent study used stem cell transplantation to deliver PACAP into mice brain and found that PACAP delivered at 3 days after MCAO could reduce brain infarct size, improve functional recovery, and induce microglia polarization toward M2 phenotype at 7 and 14 days after ischemia (Brifault et al., 2015). This study provided evidence that we could prevent delayed tissue damage through modulating microglia polarization. Metformin, widely used to treat type 2 diabetes, has been shown to be able to reduce the incidence of stroke and promote M2 polarization of macrophages in vivo and in vitro (Jin et al., 2014; Nath et al., 2009; Tsoyi et al., 2011; Zhou et al., 2001). Metformin treatment daily at 24 h after MCAO enhanced M2 polarization related gene expression, promoted angiogenesis and neurogenesis, and improved functional recovery. In vitro study performed by the same group showed that M2 polarization of microglia was essential for Metformin induced angiogenesis (Jin et al., 2014). Two clinical trials showed that Ginsenoside treatment could improve neurological outcome in patients with acute ischemic stroke. The beneficial role of Ginsenoside treatment was associated with inhibiting microglia activation and reducing M1 phenotype related pro-inflammatory cytokines expression (Zhang et al., 2015). Minocycline, which is widely used to inhibit microglia activation, has been used to treat ischemic stroke in clinical trials (Fagan et al., 2010; Lampl et al., 2007). Recent studies showed that minocycline treatment could exert a long-term protective function via attenuating BBB permeability and promoting microglia polarization toward M2 phenotype after focal cerebral ischemia (Yang et al., 2015). Minocycline was also shown to modulate microglia polarization during the early pathogenesis phase of amyotrophic lateral sclerosis (ALS), in which microglia are significantly activated (Kobayashi et al., 2013; Liao et al., 2012). These results suggested that minocycline could be a promising
therapeutic drug for inhibiting microglia activation mediated inflammatory response and tissue injury.
865 866
5.3.2. Receptors and small molecules Recent studies showed that Class A scavenger receptor (SR-A) and cannabinoid type 2 receptor (CB2R) are involved in modulating microglia polarization after ischemic stroke. In SR-A knockout mice, the numbers of F4/80+CD11b+ macrophages and CD16/32+ M1 microglia were reduced in the ischemic brain compared with control mice, while the number of F4/80+CD206+ M2 microglia was increased. Absence of SR-A reduced ischemia induced inflammation and infarct volume, accompanied by lower expression of M1 microglia related genes TNF-a, iNOS, MCP-1, and IL-1b and higher expression of M2 microglia related genes IL-10 (Xu et al., 2012). In CB2R knockout mice, brain infarct volume was increased and neurological impairment was worse. Intraperitoneal injection of CB2R agonist JWH-133 at 10 min or 3 h after MCAO reversed the phenomenon. JWH-133 treatment also reduced Iba1+ microglia activation, concomitant with lower expression of both M1 and M2 microglia markers (Zarruk et al., 2012). Culture supernatants derived from Th1 cells promoted microglia to up-regulate M1 phenotype related genes such as MCP-1 and CCL-2 expression, indicating that Th1 could affect microglia polarization in a paracrine manner. However, the molecular mechanism of the interaction between Th1 cells and microglia in ischemic brain is rarely investigated (Prajeeth et al., 2014). A study in IL-10 knockout mice showed that IL-10 absence exacerbated brain ischemic injury and reduced M2 microglia marker expression, suggesting that IL-10 is critical in modulating microglia polarization toward M2 phenotype (Perez-de Puig et al., 2013). In a model of experimental autoimmune encephalomyelitis (EAE), lentivirus mediated IL-25 gene delivery promoted microglia polarization to M2 phenotype, with high expression of M2 markers Arginase-1, CD206, and Ym1 (Maiorino et al., 2013). Studies showed that TGF-b could induce microglia polarization to M2 phenotype (Blobe et al., 2000; Hanisch and Kettenmann, 2007). On the other hand, M2 polarization of microglia could produce TGFb(Hu et al., 2015). Recent studies found that TGF-b involved in promoting IL-4 mediated M2 polarization of microglia and enhancing the sensitivity of microglia response to IL-4 (Zhou et al., 2012). In summary, microglia polarization is a complicate process and could be modulated by many molecules. Understanding how these molecules modulate microglia polarization under ischemia environment could help us drive microglia toward a protective phenotype, thus protecting brain against ischemic attack.
867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909
5.3.3. microRNAs and microglia (Table 2) Increasing evidence revealed that microRNAs play an important role in the pathophysiology of CNS diseases, including AD, Parkinson’s disease (PD), EAE, multiple sclerosis (MS), ALS, and stroke (Butovsky et al., 2012; Junn and Mouradian, 2012; Moore et al., 2013; Parisi et al., 2013; Ponomarev et al., 2011; Sun et al., 2013; Zhao et al., 2013). Several microRNAs were found to be associated with microglia activation or microglia polarization in CNS diseases. The differential expression of microRNAs could affect microglia polarization and impact the function of microglia in brain injury or tissue repair (Butovsky et al., 2015; Hu et al., 2015; Ponomarev et al., 2011; Su et al., 2015). MiR-124 is unique among microRNAs, with selective expression in the CNS and the highest expression in neurons (Kim et al., 2004; Landgraf et al., 2007; Weng et al., 2011). In a zebrafish model, miR-124 was specifically over expressed in microglia. MiR124 overexpression did not cause morphological changes of microglia, but reduce microglia activity and the capacity of clearing apoptotic cells. Conversely, depletion of miR-124 in
910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994
zebrafish enhanced microglia motility and phagocytosis. These results suggested that stable expression of miR-124 is important for maturity of microglia (Svahn et al., 2015). In a transient cerebral ischemia model in rat, plasma miR-124 was increased at 6 h and continued to elevate at 48 h after ischemia onset, however, there was no correlation between the level of plasma miR-124 and brain infarct volume (Weng et al., 2011). A clinical study of 65 patients with cardiac arrest found that the up-regulation of blood miR-124 at 24 h and 48 h after cardiac arrest was associated with poorer neurological outcome at 6 weeks after cardiac arrest, suggesting that miR-124 could be a biomarker for the prognosis of ischemic stroke and cardiac arrest (Gilje et al., 2014). It remains controversial whether miR-124 exerts beneficial or detrimental role in ischemic brain injury (Doeppner et al., 2013; Liu et al., 2013; Sun et al., 2013; Zhu et al., 2014). One previous study showed that miR-124 delivery could protect cultured neurons against OGD condition. MiR-124 delivery enhanced angiogenesis and neurogenesis at 8 weeks after MCAO, with a mechanism of Usp14-dependent REST degradation in vivo. This concept was supported by the evidence that miR-124 delivery reduced Usp14 and REST expression after MCAO, and REST was down-regulated after Usp14 inhibitor IU-1 treatment (Doeppner et al., 2013). However, another study showed that knocking out miR-124 reduced ischemia induced cell death and improved neurological function. Treatment with miR-124 antagomir enhanced Ku70 expression (a DNA repair protein) in the ischemic region, suggesting that miR-124 promoted ischemia induced brain injury via regulating Ku70 expression (Zhu et al., 2014). Under normal physiological condition, non-activated microglia express high level of miR-124 and mainly express M2 phenotype markers such as Fizz1, Ym1, IL-10 and IL-4 (Ponomarev et al., 2007; Ponomarev et al., 2011). Under pathological condition, with proinflammatory cytokines stimulation or in a model of EAE, activated microglia displayed up-regulated M1 phenotype markers (e.g. MHC II), and down-regulated miR-124. Transfection of miR-124 in mice at different stages of EAE reduced microglia activation and leukocyte infiltration, ameliorated clinical symptoms, and promoted neuronal function recovery. Conversely, repression of miR124 using miR-124 antisense oligonucleotides led to microglia activation. The study indicated that miR-124 is essential for maintaining the quiescent phenotype of microglia and contribute to M2 phenotype in normal condition (Ponomarev et al., 2011; Ponomarev et al., 2013). MiR-124 inhibition reduced inflammation and delayed the course of EAE, suggesting that we might use miR124 as a target for the treatment of inflammation or microglia activation associated diseases including ischemic stroke. MiR-155 is widely accepted as a pro-inflammatory microRNA and has a direct relationship with M1 phenotype (Guedes et al., 2013). MiR-155 expression is increased in microglia after being stimulated by pro-inflammatory cytokines such as LPS, IFN-??, and TNF-?? (Bala et al., 2011; Cardoso et al., 2012; Wang et al., 2010). Knocking down miR-155 by anti-miRNA oligonucleotides enhanced suppressor of cytokine signaling 1 (SOCS-1) protein expression, which is a target of miR-155 and critical for inhibiting inflammation. Knocking down miR-155 also reduced the production of NO as well as the expressions of inflammatory cytokines and iNOS. Conditioned medium collected from microglia, in which miR-155 was inhibited, protected neurons from inflammation mediated injury. This study provided evidence that miR-155 promotes microglia toward pro-inflammatory phenotype (M1 phenotype) via inhibiting SOCS-1 expression (Cardoso et al., 2012). LPS stimulated microglia could inhibit neural stem cells (NSCs) differentiation, which could be reversed via depletion of miR-155 in microglia. When miR-155 was depleted under LPS induced inflammatory environment, NSCs proliferation was restored and the number of amoeboid morphological microglia enhanced in
13
dentate gyrus of mice. Thus, miR-155 in microglia plays an important role in inflammation induced neurogenic deficit (Woodbury et al., 2015). In a mouse model of ALS, microglia were abnormal because many molecules essential for microglia survival were lost. However, the miR-155 expression was increased. The expression of pro-inflammatory genes in microglia was associated with the severity of ALS in mice. MiR-155 ablation suppressed microglia polarization to M1 phenotype and corrected with the abnormality of microglia. In addition, anti-miR-155 treatment delayed the course of ALS and prolonged survival in SOD1 mice, a model used for ALS (Butovsky et al., 2015). Similarly, another study found that miR-155 was highly expressed in cultured M1 phenotype microglia from MS patients. MiR-155 transfection enhanced TNF-a and IL-6 secretion and M1 phenotype marker expression including CD80, CD86, and CCR7 while miR-155 inhibition reduced pro-inflammatory cytokines secretion via inhibiting SOCS-1 expression (Moore et al., 2013). These studies further supported that miR-155 is crucial for M1 polarization of microglia. Therefore, miR-155 inhibition could be neuroprotective for the treatment of neurological diseases where inflammation is excessive and detrimental. Although miR-155 is critical for microglia activation mediated inflammatory response under many pathological conditions, few studies has investigated the role and mechanisms of miR-155 in microglia activation or polarization in ischemic stroke. In a model of permanent MCAO in mice, miR-155 expression was reduced in the brain at 24 h after ischemia, but whether low expression of miR-155 was associated with inflammatory response and microglia activation was not investigated (Liu et al., 2010). Another study indicated that miR-155 in microglia was increased after transient MCAO and induced proinflammatory response in microglia (Su et al., 2014). Combination with these results, whether miR-155 inhibition could restore inflammatory response, modulate microglia polarization, and benefit tissue repair and functional recovery after ischemic stroke needs to be explored in the future. Other microRNAs were also found to have a connection with microglia polarization and activation. MiR-Let7ais involved in modulating microglia polarization toward M2 phenotype. Overexpression of miR-Let7a in BV2 cells, a widely accepted cell line used for investigation of microglia activation, reduced M1 related cytokines including ROS, iNOS, and IL-6 expression, but enhanced M2 related cytokines including IL-10, IL-4, and BNDF expression. MiR-Let7a was found to mediate autophagy activity in microglia under LPS induced inflammation environment (Cho et al., 2015; Song et al., 2015). MiR-Let-7c-5p, a highly conserved miRNA in stroke, is down-regulated in the plasma of patients with ischemic stroke. In mice model of MCAO, miR-Let-7c-5p was demonstrated to be down-regulated in the brain. MiR-Let-7c-5p overexpression in the brain could inhibit microglia activation and promote neurological function recovery. Further experiment showed that ischemia and inflammation could induce miR-Let-7c-5p upregulation in microglia and miR-Let-7c-5p overexpression inhibited microglia activation via suppressing caspase 3 pathway. The study suggested that miR-Let-7c-5p is a promising target for inhibiting microglia activation after cerebral ischemia (Butovsky et al., 2014; Ni et al., 2015; Zhao et al., 2013). MiR-181c is another potential inhibitor of microglia activation. MiR-181c decreased in microglia after OGD treatment. Conditioned medium from miR181c transfected microglia reduced TNF-a production and protected neurons against OGD condition. TNF-a is a direct target of miR-181c, which was elucidated by a luciferase reporter assay. Overexpression of TNF-a compromised the protective effect of miR-181c on neurons under OGD condition. Thus, miR-181c could inhibit microglial production of TNF-a and attenuate ischemiahypoxia induced neuron apoptosis (Zhang et al., 2012a). MiR-9, which is specifically expressed in rodents brain, was used to induce
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060
G Model
PRONEU 1417 1–25 14
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077
transgene expression in brain resident microglia (Akerblom et al., 2013; Deo et al., 2006). MiR-9 was shown to promote microglia activation through down-regulating the expression of miR-9 downstream protein monocyte chemotactic protein-induced protein 1 (MCPIP1) (Yao et al., 2014). MiR-125b-5p, miR-99a, miR-342-3p are highly expressed both in human and mouse brain microglia, but it needs further study to identify the role of these miRNAs in modulating microglia activation or polarization (Butovsky et al., 2014). Collectively, these results indicated that microglia polarization is complicated and could be affected by many substances. Whether inhibition of microglia activation is beneficial or detrimental for the recovery of ischemia induced brain injury might depend on when and how microglia activation is inhibited, and whether microglia polarization is affected. Promoting the switch of microglia toward a neuroprotective M2 phenotype might be a promising approach for ischemic disease therapy.
1078
6. Microglia crosstalk with other cells during ischemic stroke
1079
6.1. Microglia and neurons
1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119
6.1.1. Neurons control microglia activation Neurons could control microglia activation via ‘On’ and ‘Off’ signals. ‘On’ signal is mainly found under pathological condition and included chemokines, purines, glutamate, and matrix metalloproteinase-3. ‘Off’ signal mainly occurs in the healthy brain and is mediated by release of CX3CL1, CD22, neurotransmitters and neurotrophins from neurons, which could bind with receptors on microglia and help microglia exert physiological function (Biber et al., 2007). The interaction between neurons and microglia is complicated and involves a variety of ligands and receptors (Correa et al., 2013). CD200/CD200R and CX3CL1/ CX3CR1 are the two most studied ligands and receptors pairs involved in controlling microglia activation. CD200 is constitutively expressed on neurons and CD200R is expressed mainly on microglia in the brain. Deficiency of CD200 or blockade of CD200R in mice led to enhanced microglia activation and worsened symptoms in the mouse model of EAE, facial nerve transection, retinal inflammation, and encephalitis (Broderick et al., 2002; Deckert et al., 2006; Hoek et al., 2000). These results suggested that CD200/CD200R contributes to maintaining microglia activation at an appropriate level. Similar to the function of CD200/ CD200R, CX3CL1/CX3CR1 could also inhibit microglia activation. CX3CL1 is expressed on neurons and CX3CR1 is only expressed on microglia in the brain. Absence of CX3CR1 in mice enhanced microglia neurotoxicity, which caused more neuronal death in models of PD and ALS (Cardona et al., 2006). In a retinal transplantation study, time-lapse confocal imaging was used to evaluate the influence of absence of CX3CR1 on microglia processes dynamism and mobility. It was observed that the absence of CX3CR1 did not affect microglia processes dynamism including extension and retraction of processes, formation of new processes, and elimination of existing processes, but reduced the rate of movement toward injury induced by laser. Administration of CX3CL1 induced the transformation of microglial spindle processes toward a more branched and shorter counterparts in CX3CR1 heterozygote mice. However, no microglia morphological changes could be detected after the administration of CX3CL1 in CX3CR1 knockout mice, suggesting that CX3CL1/CX3CR1 plays an important role in the interaction between neurons and microglia (Liang et al., 2009).
1120 6.1.2. Neurons influence microglial function 1121 After ischemic stroke, a large number of neuronal cells die 1122 because of reduced blood flow and energy depletion. Dead or dying
cells release damage-associated ligands and excitotoxic glutamate, exacerbating neuronal damage (Neher et al., 2013). However, a recent study showed that injured neurons could promote microglia polarization toward protective phenotype via releasing lipocalin-2. Lipocalin-2 is highly expressed in cultured neurons from human or mice brain after ischemia. Lipocalin-2 treatment induced microglia to secrete IL-10, change from ramified shape to less branches and long-rod shape, and enhance microglia capacity of phagocytosis. Conditioned medium from lipocalin-2 treated microglia enhancedsynaptophysin and post-synaptic density 95 (PSD95) expression and prevented neuronal death from OGD stress (Xing et al., 2014). Damaged neurons induced by ischemia could release IL-4, which could enhance IL-4 receptor expression on microglia and promote microglia polarization to M2 phenotype. IL-4 activated peroxisome proliferator activated receptor g (PPARg) on microglia and enhanced microglia capacity of phagocytosis of apoptotic neurons (Zhao et al., 2015). Glutamate stressed neurons could enhance the capacity of microglia to clear neuronal debris via secreting soluble fractalkine (sFKN) (Noda et al., 2011). These studies implied that injured neurons could promote microglia to exert protective function to help neuron survive under ischemic condition.
1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144
6.1.3. Microglia influence neuronal function Under pathological condition, microglia are traditionally recognized with a function in immunological surveillance (Eyo and Wu, 2013). They could affect disease progression via clearing debris, secreting pro-inflammatory or anti-inflammatory cytokines, and producing growth factors. Microglia are essential for the development and maintenance of healthy brain homeostasis including modulating neural circuit via specifically interacting with neuronal synapse (Eyo and Wu, 2013; Hirasawa et al., 2005; Wake et al., 2013). Two-photon microscopy allows the monitoring of the interaction between neurons and fluorescent-labeled microglia in transgenic mice in vivo (Wake et al., 2009). Microglia directly contacted neuronal synapse through extending processes every 5 min for 1 h. It took microglia in the peri-infarct region longer to connect with neurons, up to approximately 1 h after ischemia. The prolonged connection time after ischemia stimulation caused disappearance of a few presynaptic boutons, suggesting that microglia could monitor the functional state of synapses (Wake et al., 2009). However, the mechanisms driving microglia to make direct contact with neuronal synapse and the exact role of prolonged connection with neuronal synapse after brain ischemia are still unclear. Microglia are professional phagocytes in the brain and could engulf neurons within hours (Neher et al., 2011). Microglial CX3CR1 knockout prevented neuronal loss in a mouse model of AD (Fuhrmann et al., 2010). Microglia play a beneficial role for tissue remodeling and regeneration after ischemic stroke via clearing dead or dying neurons (Brown and Neher, 2014; Neumann et al., 2009). Insufficient clearance of cell debris by microglia could cause failure of tissue repair after focal cerebral ischemia (Stoll et al., 2004). Chimeric mice in which brain resident microglia were GFP, and infiltration of macrophages were GFP+ were used to explore the phagocytosis of brain resident microglia. It was found that brain resident microglia could engulf neuronal debris as early as day 1 and peaked at 2 days, while infiltrated macrophages started to clear neuronal debris at 4 days after MCAO. The study indicated that brain resident microglia are more sensitive and important on the defense of ischemia via clearing dead or dying neurons (Schilling et al., 2009). However, the exact benefit of removing dead or dying neurons by microglia is not clear, maybe partly because it help reduce inflammation (Sierra et al., 2013). In a rat focal cerebral ischemia model induced by endothelin-1, a study
1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253
observed that microglia played anti-inflammatory role through phagocytosing polymorph nuclear granulocytes at 24 h after ischemic injury, and reduced neuronal death (Neumann et al., 2008). Depletion of Mac-2+ resident microglia in the brain caused enhanced neuronal apoptosis and high expression of proinflammatory cytokines at 72 h after transient cerebral ischemia (Lambertsen et al., 2009). These studies suggested that microglia are critical for the neuron survival within the acute phase of ischemic stroke (Lalancette-Hebert et al., 2007). The detrimental role of microglia in neurological diseases had been widely debated over the past several decades (Block et al., 2007; Hanisch and Kettenmann, 2007). Several studies showed that microglia activation is associated with ischemia induced neuronal death (Denes et al., 2008; Franco et al., 2012; Neher et al., 2013; Wu et al., 2012). Inhibition of microglia activation by a number of drugs reduced brain injury (Lai and Todd, 2006). A recent study showed that microglia mediated delayed neuronal death through phagocytosis activity after ischemia. Microglia expressed phagocytosis related proteins Milk fat globule EGF-like factor-8 (MFG-E8) and Mer receptor tyrosine kinase (MerTK) at 3 to 7 days after focal brain ischemia. Blocking microglia phagocytosis via depleting MFG-E8 or MerTK in mice reduced neuronal loss and brain atrophy. This study further demonstrated that MFG-E8 or MerTK mediated phagocytosis of microglia specifically affect the living and viable neurons. In vivo experiment showed that there was no difference in the accumulation of Fluorojade C positive dead neurons between MFG-E8 or MerTK deficient mice and control wild type mice after ischemic injury. Addition of cultured microglia from wild type, MFG-E8, or MerTK deficient mice showed no difference in the loss of untreated neurons, but reduced the loss of viable neurons stressed by glutamate. Moreover, MFG-E8 or MerTK deficiency displayed lower microglial capacity of phagocytosing neurons, suggesting that the interaction between microglia and neurons is a complicated process, and the different functional stages of neurons could influence microglia function differently. Therefore, further study aims at elucidating the role of microglia in different stages of ischemic brain injury would help pave the way in developing microglia targeted treatments for ischemic stroke (Neher et al., 2013). NADPH oxidase (NOX) and NOX induced ROS are common mediators of neuronal damage caused by microglia activation after ischemia (Dringen, 2005). Knocking out NADPH oxidase type 4 (NOX4) reduced BBB leakage and neuronal apoptosis in ischemic brain in mice (Kleinschnitz et al., 2010). Knocking out NADPH oxidase type 2 (NOX2) promoted tissue repair through angiogenesis following ischemic reperfusion in mice (McCann et al., 2014). A recent study found that microglia expressed voltage-gated proton channel Hv1 protein, which is responsible for ischemia induced neuronal death. Microglia cultured from Hv1 knockout mice could not produce NOX and ROS. Treatment of cultured microglia with NOX inhibitor diphenyleneiodonium (DPI, 30 mM) and apocynin (300 mM) or the Hv1 inhibitor Zn2+ suppressed NOX activation and ROS production. In Hv1 knockout mice, brain infarct volume and neurological scores were smaller than wild type mice at 24 and 72 h after MCAO. The number of caspase3+ or TUNEL+ apoptotic neurons decreased in the peri-infarct region. These results suggested that Hv1 contributes to neuronal death via NOX activation and ROS production. In vitro study further supported the hypothesis. Co-culture of neurons with microglia from Hv1 knockout mice reduced neuronal death after 30 min OGD, comparing with neurons co-cultured with microglia from wild type mice. Pre-treatment with NOX inhibitor or Hv1 knockout in microglia protected neurons against OGD. Thus, the study provided a new target protein Hv1 for ischemic stroke therapy (Wu et al., 2012).
15
6.2. Microglia and astrocytes
1254
6.2.1. Cross-talk between microglia and astrocytes Astrocytes are the most abundant cell type in the brain and are thought to serve and protect neurons (Nedergaard and Dirnagl, 2005; Tsai et al., 2012). Both microglia and astrocytes are major components of the innate immune system in the brain. They are able to sense the changes of brain environment and play an important role in various pathological conditions including ischemic stroke (Hanisch and Kettenmann, 2007; Ransohoff and Brown, 2012; Seifert et al., 2006; Tremblay et al., 2011). Based on time-lapse recording from two-photo microscopy imaging, a study observed that microglia directly contacted astrocytes via extending their processes toward astrocytes (Nimmerjahn et al., 2005). Microglia display a comparable density to astrocytes in the brain and express membrane receptors for all known neurotransmitters, which allow microglia to modulate neuronal activity and receive information from astrocytes (Jinno et al., 2007; Pocock and Kettenmann, 2007). Indeed, emerging evidence indicated that there exists a functional communication between microglia and astrocytes, which is crucial for the innate immune response in the brain (Ransohoff and Brown, 2012). The modulators contribute to the communication between microglia and astrocytes including IL1b, TNF-a, TGF-b, adenosine, ATP, and glutamate (Boison et al., 2010; Burnstock et al., 2011; Herrera-Molina and von Bernhardi, 2005; Pascual et al., 2012; Rouach et al., 2002a,b). Recently, several new discoveries further advanced our understanding about the molecular mechanisms regarding the communication between microglia and astrocytes. For example, LPS stimulated microglia could release ATP, and promote astrocytes to release glutamate, which modulated neuronal activity via metabotropic glutamate receptors (mGluRs) (Pascual et al., 2012). LPS stimulation induced a transient spontaneous excitatory postsynaptic currents (EPSCs) in cultured brain slices, which could be blocked by depletion of microglia in brain slices (Pascual et al., 2012). Blocking purinergic receptors via broad spectrum antagonists decreased the frequency of EPSCs, while activating purinergic receptor P2Y1R, which is exclusively expressed on astrocytes in cultured brain slices, reversed the phenomenon. These results collectively suggested that microglia are essential for astrocytes mediated neuronal activity via releasing ATP, which could bind to P2Y1R located on astrocytes. Further experiments found that inhibition of mGluR5 receptor on neurons decreased the effect of LPS stimulation on neuronal activity, implicating that activated microglia induced astrocyte modulated neuronal activity through mGluR5 (Pascual et al., 2012). In conclusion, studies supported that microglia could modulate neuronal activity via communicating with astrocytes in a ATP-P2Y1R-mGluR5-mGluR5 receptor dependent manner. Microglia could trigger astrocytes mediated neuroprotection via an ATPP2Y1R-P38-MAPK-IL-6 dependent signal pathway. Microglia sensed the low concentration of methylmercury (MeHglow), which induced neuronal death, and released ATP. ATP bound to the P2Y1R on astrocytes, which in turn activated P38-MAPKs and promoted astrocytes to produce IL-6. ATP release and IL-6 production could be blocked via inhibition of vesicular nucleotide transporter (VNUT) or P2Y1R. Moreover, additional IL-6 in cultured brain slices attenuated MeHglow induced neuronal death (Shinozaki et al., 2014). These discoveries further implicated that astrocytes are required for microglia mediated neuroprotective function on neurons. Therefore, astrocytes could be a potential choice for regulating microglia function in neurological diseases.
1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313
6.2.2. Cross-talk between microglia and astrocytes during ischemic stroke CX3CL1-CX3CR1 signal in microglia contributes to the regulation of neuronal viability under ischemia condition (Cipriani et al.,
1314 1315 1316 1317
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 16
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339
2011; Denes et al., 2008; Soriano et al., 2002). In a mouse model of permanent MCAO, CX3CL1 and CXCL16 acted synergistically on the crosstalk between neurons, microglia, and astrocytes, and prevented ischemia or glutamate induced neuronal death. Administration of CX3CL1 or CXCL16 at 30 min before MCAO in mice reduced brain infarct volume at 24 h after ischemia. Deficiency of CXCL16 receptor in mice eliminated the protective effect of CX3CL1 or CXCL16. CX3CL1 stimulation promoted cultured astrocytes to release CCL2 and reduced glutamate-excitotoxicity induced neuronal death, which was reversed by depletion of CCL2 receptor A3R or CCL2 antibody treatment. These results suggested that CX3CL1-CXCL16-CCL2 signaling contributes to mitigating ischemia induced brain damage via mediating the interaction between neurons, microglia, and astrocytes (Rosito et al., 2014). Pro-inflammatory mediator lysophosphatidylcholine (LPC) produced by neurons and astrocytes after ischemic stroke stimulated microglia to up-regulate mRNA of MCP-1 and CCR2, which involves in mediating inflammatory response after cerebral ischemia (Inose et al., 2014). Therefore, there are complicated communications between microglia and astrocytes. The understanding of molecular mechanisms of this communication is still limited and calls for the further investigation.
1340
6.3. Microglia and oligodendrocytes/oligodendrocyte progenitor cells
1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381
Oligodendrocytes form the myelin sheath in the CNS and are highly vulnerable to ischemia (Back et al., 2002; Dewar et al., 2003). After 30 min of an artery occlusion, swelling of oligodendrocytes occurs. Most of oligodendrocytes are dead at 3 h after ischemia, which appears earlier than neuronal death in the ischemic region (Pantoni et al., 1996). Oligodendrocyte progenitor cells (OPCs) derived from NPCs in the SVZ could proliferate and differentiate into oligodendrocytes in ischemic brain (Li et al., 2010; Zhang et al., 2001, 2012b). Oligodendrocytes are major components of white matter. Damage of oligodendrocytes and white matter is associated with function impairment after ischemia induced brain injury (Matute et al., 2013). Several studies demonstrated that microglia activation causes the impairment of oligodendrocytes/OPCs via producing pro-inflammatory cytokines such as TNF-aand IL-1b (Deng et al., 2008; Haynes et al., 2005; Moxon-Emre and Schlichter, 2010). For example, in a rat model of chronic cerebral ischemia, a study observed that increased loss of oligodendrocytes was coupled with increased microglia activation from 2 to 4 weeks after surgery. Interestingly, TNF-a was highly expressed at 2 weeks after surgery (Masumura et al., 2001). Another study using the same model found that activated microglia expressing TNF-a appeared at the white matter lesion, where oligodendrocytes died in a caspase3 dependent apoptotic death manner (Jalal et al., 2012). Minocycline treatment inhibited microglia activation, reduced pro-inflammatory cytokines expression, and attenuated oligodendrocytes/OPCs damage under hyperoxia and ischemia-hypoxia conditions in neonatal rats (Cai et al., 2006; Schmitz et al., 2014). These studies suggested that there is a link between microglia activation mediated neuro-inflammation and hyperoxia/ischemia induced oligodendrocytes loss, but the detailed molecular mechanism connecting the link is unclear. Recently, emerging evidence indicated that microglia exert beneficial function on oligodendrocytes/OPCs, and it is mainly M2 polarization of microglia that plays the protective role. In vitro, conditioned media from LPS induced M1 microglia or untreated microglia enhanced oligodendrocytes death after OGD treatment for 24 h, while conditioned media from IL-4 induced M2 microglia reduced oligodendrocytes apoptosis (Wang et al., 2013). Ina mouse model of MS, depletion of M2 microglia in mice impaired OPCs differentiation. Conditioned medium from M2 microglia promoted
cultured OPCs differentiation. Inhibiting activin-A released by M2 microglia suppressed OPCs differentiation after demyelination (Miron et al., 2013). These two studies shed new light on the potential role of microglia polarization in different pathological conditions. M2 microglia derived activin-A could be a target for the treatment of neurological diseases. Another study found that there is a temporal and spatial activation of microglia in the SVZ in neonatal rats. Inhibiting microglia activation via minocycline treatment reduced NPCs and OPCs proliferation in the SVZ, as well as the expression of pro-inflammatory cytokines IL-1b, IL-6, TNFa, and IFN-g. Co-culture of neurosphere with microglia enhanced NSCs differentiation into OPCs, which could be blocked by inhibiting microglia activation or blocking production of IL-1b, IL-6, TNF-a, and IFN-g. This study further supported the beneficial effect of microglia activation on oligodendrocytes/OPCs (Shigemoto-Mogami et al., 2014). Thus, it is imprecise to simply declare that microglia activation is detrimental or beneficial for oligodendrocytes/OPCs, as well as the role of pro-inflammatory cytokines released by microglia. Different polarization of microglia and different pathological conditions could influence the function of microglia. Understanding when and how microglia activation exert detrimental or beneficial role on oligodendrocytes/OPCs will be helpful for the regeneration of white matter injury under different pathological conditions including ischemic stroke.
1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405
7. Relationship between microglia and stem cells
1406
Stem cell transplantation is a promising strategy for the treatment of various neurological diseases including AD, PD, ALS, MS, spinal cord injury, and ischemic stroke (Abe et al., 2012; Boulis et al., 2011; Gogel et al., 2011; Liu et al., 2014b; Martino et al., 2010; Sahni and Kessler, 2010). The interaction of grafted cells with its surrounding environment and host cells are important for the neuroprotective effects (Darsalia et al., 2011; Gogel et al., 2011). Emerging evidence indicated that transplanted stem cells could interact with and influence brain resident microglia activation after ischemia. In a rat model of transient cerebral ischemia, transplantation of human NSCs, CTX0E03, promoted microglia proliferation in the striatum from 1 to 4 weeks after MCAO. CTX0E03 transplantation slowed the rate of decline of DCX+/Ki67+ neuroblasts after 4 weeks of ischemia (Hassani et al., 2012). These results suggested that transplanted NSCs could facilitate neuroblast proliferation, which is associated with enhanced microglia activation. Further studies to examine the phenotype of these activated microglia could help elucidate the mechanism of how transplanted NSCs affect microglia activation. Compared with NSCs, transplanted mesenchymal stem cells (MSCs) exert beneficial function on neurological recovery after ischemic brain injury partly via anti-inflammatory response, which is mainly mediated by brain resident microglia, macrophages, and lymphocytes (mainly T and B cells) (Kokaia et al., 2012; Sheikh et al., 2011; Tang et al., 2014). MSCs transplantation inhibited microglia activation following3 and 7 days of transient MCAO in rats. Fractalkine and IL-5 secreted by MSCs could reduce microglia response to LPS stimulation (Sheikh et al., 2011). However, another study showed that MSCs transplantation via tail vein improved motor behavior recovery after MCAO accompanied by enhanced microglia activation (Yang et al., 2010). The controversial results of transplanted MSCs on microglia activation might partly due to the timing and dose of MSCs transplantation, as well as the observation time during experiments. In a model of hypoxic-ischemia (HI) in nine-day-old neonatal mice, transplantation of MSCs reduced microglia response and induced microglia polarization to M2 phenotype, contributing to enhanced regeneration and improved neurological outcome at 18 or 21 days after ischemic brain injury (Donega et al., 2014; van Velthoven et al.,
1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457
2010). In vitro studies showed that MSCs could promote M2 polarization of microglia and affect microglia activity via releasing NO (Hegyi et al., 2014; Neubrand et al., 2014). Collectively, transplanted stem cells could either inhibit or promote microglia activation and modulate microglia polarization after ischemia. However, there is still no strong evidence to support that microglia activation could influence transplanted stem cells survival, migration, and differentiation after ischemic stroke. Endothelia progenitor cells (EPCs) is another effective stem cell type used for the treatment of ischemic disease. The number of EPCs in circulation is considered as a predictor for acute stroke (Chu et al., 2008). There is a transient elevation of circulating EPCs
17
after stroke (Zhou et al., 2009). The level of circulating EPCs negatively correlates with the severity of ischemia (Bogoslovsky et al., 2010). Recent studies demonstrated that EPCs transplantation reduces brain infarct volume and improves neurological outcome (Chen et al., 2012; Fan et al., 2010; Iskander et al., 2013; Zhang et al., 2002). Our previous study demonstrated that there is an interaction between microglia and EPCs. LPS activated microglia could release HMGB1, which promoted cultured EPCs to release IL8. Conditioned medium from EPCs enhanced the viability of human umbilical cord vein cells (HUVEC) under OGD condition and promoted tube formation. Administration of HMGB1 inhibitor glycyrrhizin or knocking down IL-8 in EPCs reversed the beneficial
Fig. 1. Microglia polarization after ischemic stroke. The figure illustrates the role of M1 or M2 microglia in ischemic brain injury. Ischemia causes neural death and tissue damage. Injured tissue release a variety of damage associated molecular pattern molecules, pro-inflammatory cytokines, and reactive oxygen species (ROS), causing microglia to polarize toward M1 or M2 phenotype. M1 microglia could exacerbate neural death, astrocyte apoptosis, and BBB disruption via releasing IL-6, TNF-a, IL-1b, MMPs, and iNOS. Conversely, M2 microglia exert a protective role after ischemia through releasing neurotrophic factors including BDGF, IGF-1, IL-4, and IL-10. M2 microglia could maintain BBB integrity, promote the proliferation and differentiation of neural stem cells (NSCs) and oligodendrocyte progenitor cells (OPCs), and facilitate myelin regeneration and tissue repair. However, the exact molecular mechanism mediating microglia polarization toward M1 or M2 phenotype after ischemic brain injury needs further investigation.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469
G Model
PRONEU 1417 1–25 18
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
effect of EPCs on HUVEC. Intravenous EPCs transplantation enhanced angiogenesis, reduced brain atrophy volume, and improved neurological behavior after transient focal cerebral ischemia. While administration of HMGB1 inhibitor glycyrrhizin compromised the therapeutic effects of EPCs transplantation. Therefore, microglia could modulate EPCs paracrine function via releasing HMGB1, contributing to enhanced therapeutic function of EPCs transplantation after ischemic stroke (Chen et al., 2014). Although microglia display a positive regulation on transplanted EPCs after ischemia, whether transplanted EPCs affect microglia activation and function is unknown. Studies exploring the interaction between microglia activation and EPCs transplantation are also scarce.
vehicle for gene therapy for glioma (Ribot et al., 2007). Collectively, these studies suggested that microglia transplantation manifests a potential for the treatment of neurological diseases including ischemic stroke. However, several questions remain to be addressed before microglia can be considered as an alternative choice of cell based therapy for ischemic diseases. What is the exact mechanism of microglia transplantation in influencing the neurological outcome after tissue injury? What is the advantages of microglia transplantation? More studies are needed to further the understanding of the role of transplanted microglia in neurological diseases.
1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519
9. Conclusion and prospect
1520
1483 8. Microglia as a potential candidate of cell based therapy for 1484 ischemic stroke
Microglia are resident macrophages in the brain. Unlike other neural cells such as neurons, astrocytes, and oligodendrocytes, which are of neuroectodermal origin, microglia are originated from primitive progenitor cells located in yolk sac (Ginhoux et al., 2010; Kierdorf et al., 2013). Microglia could sense even small imbalances of environmental homeostasis and are rapidly activated with a characterization of dynamic morphology and polarization (Prinz and Priller, 2014). The temporal and spatial course of microglia activation are detected both in human being and experimental animals (Darsalia et al., 2011; Gulyas et al., 2012; Perego et al., 2011; Price et al., 2006; Tomimoto et al., 1996; Zhang et al., 1997). Traditionally, microglia activation are considered to play a deleterious role during ischemic stroke, as inhibition of microglia activation attenuates ischemia induced brain injury (Franco et al., 2012; Machado et al., 2009; Yenari et al., 2006). Activated microglia could mediate BBB disruption and induce neuronal cell death via releasing a variety of pro-inflammatory cytokines and toxic substances (da Fonseca et al., 2014; Jolivel et al., 2015; Lai and Todd, 2006). However, increasing evidence showed that microglia activation could also be beneficial through attenuating neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after cerebral ischemia (Faustino et al., 2011; LalancetteHebert et al., 2007; Neumann et al., 2008; Thored et al., 2009). Different polarization of microglia could likely explain the biphasic role of microglia in different pathological conditions (Cherry et al., 2014; Hu et al., 2015) (Fig. 1). Many substances are involved in modulating microglia polarization, among which microRNAs show
1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547
1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482
1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508
As microglia are scavenger cells in the brain and could produce various neurotrophic factors to promote neurological recovery after ischemic brain injury, several studies directly transplanted microglia into the brain to explore whether microglia could be protective for ischemic stroke (Hanisch and Kettenmann, 2007; Kreutzberg, 1996). In a mouse model of transient MCAO, intracerebroventricular injection of microglia at 1 h after brain ischemia enhanced neuronal survival, reduced neurodegeneration, and improved neurological behaviors (Kitamura et al., 2004; Kitamura et al., 2005). In a rat model of transient MCAO, intravenous injection of microglia at 48 h after brain ischemia contributed to reducing neuronal apoptosis, increasing neurotrophic factors including GDNF, BDNF, VEGF, and BMP7 and antiinflammatory cytokinesIL-4 and IL-15 expression, and promoting neurological function recovery (Narantuya et al., 2010a). However, in a rat model of permanent MCAO, microglia transplantation via tail vein did not affect neurological outcome (Jiang et al., 2013). The controversial results are likely due to the different experimental animals, different time, routes, and dose of microglia transplantation. Besides ischemic stroke, microglia transplantation was also conducted in chronic ischemia model, spinal cord injury, AD, and PD, and showed a beneficial effect on tissue repair and functional recovery (Danielyan et al., 2014; Narantuya et al., 2010b; Takata et al., 2007; Yu et al., 2009). In addition, microglia were used as a
Fig. 2. Possible therapeutic strategies targeting microglia activation to reduce inflammatory response and promote tissue repair. Microglia could be either beneficial or detrimental for tissue repair after brain injury, which is depending on how microglia are activated. Clinical trial and basic research indicated that we could possibly modulate microglia activation via suppressing microglia to secrete pro-inflammatory cytokines or promoting microglia polarization to M2 phenotype. Receptors (e.g. TLRs, FcRs, and RAGE) located on the left microglia body mainly mediate inflammatory response and could be inhibited via knocking out these receptors or using antagonist and anti-inflammatory response drugs, for example Minocycline and Ginsenoside. Similarly, receptors (e.g. TREM-2 and CB2R) located on the right microglia body mainly mediate microglia protective function. Applying receptor agonist or neuroprotective drugs, for example Metformin, could dampen microglia mediated inflammatory response and promote microglia to secrete neuroprotective factors, such as IGF-1, TGF-b and IL-4. Overexpression of anti-inflammatory miRNAs or suppression of pro-inflammatory miRNAs in microglia could also reduce inflammatory cytokines release and promote microglia polarization to M2 phenotype. Therefore, these receptors and miRNAs in microglia could be potential therapeutic targets for modulating microglia activation and polarization, and eventually promote tissue repair.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570
a potential for microglia-targeted therapy (Ponomarev et al., 2013). More and more current studies demonstrated that there are biphasic communications between microglia and other neuronal cells including neurons, astrocytes, and oligodendrocytes/OPCs (Eyo et al., 2013; Inose et al., 2014; Ransohoff and Brown, 2012; Rosito et al., 2014; Schmitz et al., 2014; Wang et al., 2013; Xing et al., 2014). Exclusively suppressing microglia activation might compromise the protective effects of microglia and therefore not a suitable therapeutic strategy for ischemic brain injury. There is an urgent need to establish the best strategy to modulate microglia activation and drive microglia polarization to a protective phenotype at different stages of ischemic stroke (Fig. 2). Stem cell transplantation shows a promising future for cell-based therapy. Many studies showed that transplanted stem cells modulate microglia activation and contribute to reducing brain injury (Donega et al., 2014; Hassani et al., 2012; Sheikh et al., 2011; Tang et al., 2014). Whether microglia activation could in turn affect transplanted stem cell survival, migration, and differentiation is largely unknown. Our study indicated that microglia enhance EPC therapeutic effects after focal brain ischemia (Chen et al., 2014). Understanding the detailed mechanism of interaction between microglia and transplanted stem cells would facilitate clinical translation of stem cell therapy for ischemic stroke.
1571
Conflict of interest statement
1572 1573
None. Acknowledgements
1574 Q6 This work was supported by China 973 Program 2011CB504405 1575 Q7 (GYY, WY), NSFC 81070939 (GYY) and U1232205 (GYY). 1576
References
1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612 1613 1614 1615 1616
Aarum, J., Sandberg, K., Haeberlein, S.L., Persson, M.A., 2003. Migration and differentiation of neural precursor cells can be directed by microglia. Proc. Natl. Acad. Sci. U.S.A. 100, 15983–15988. Abbracchio, M.P., Burnstock, G., Verkhratsky, A., Zimmermann, H., 2009. Purinergic signalling in the nervous system: an overview. Trends Neurosci. 32, 19–29. Abe, K., Yamashita, T., Takizawa, S., Kuroda, S., Kinouchi, H., Kawahara, N., 2012. Stem cell therapy for cerebral ischemia: from basic science to clinical applications. J. Cereb. Blood Flow Metab. 32, 1317–1331. Adachi, M., Abe, M., Sasaki, T., Kato, H., Kasahara, J., Araki, T., 2010. Role of inducible or neuronal nitric oxide synthase in neurogenesis of the dentate gyrus in aged mice. Metab. Brain Dis. 25, 419–424. Adams, R.A., Bauer, J., Flick, M.J., Sikorski, S.L., Nuriel, T., Lassmann, H., Degen, J.L., Akassoglou, K., 2007. The fibrin-derived gamma377-395 peptide inhibits microglia activation and suppresses relapsing paralysis in central nervous system autoimmune disease. J. Exp. Med. 204, 571–582. Ajami, B., Bennett, J.L., Krieger, C., McNagny, K.M., Rossi, F.M., 2011. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat. Neurosci. 14, 1142–1149. Akerblom, M., Sachdeva, R., Quintino, L., Wettergren, E.E., Chapman, K.Z., Manfre, G., Lindvall, O., Lundberg, C., Jakobsson, J., 2013. Visualization and genetic modification of resident brain microglia using lentiviral vectors regulated by microRNA-9. Nat. Commun. 4, 1770. Aktas, O., Ullrich, O., Infante-Duarte, C., Nitsch, R., Zipp, F., 2007. Neuronal damage in brain inflammation. Arch. Neurol. 64, 185–189. Alliot, F., Godin, I., Pessac, B., 1999. Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Brain Res. Dev. Brain Res. 117, 145–152. Arno, B., Grassivaro, F., Rossi, C., Bergamaschi, A., Castiglioni, V., Furlan, R., Greter, M., Favaro, R., Comi, G., Becher, B., Martino, G., Muzio, L., 2014. Neural progenitor cells orchestrate microglia migration and positioning into the developing cortex. Nat. Commun. 5, 5611. Arvidsson, A., Collin, T., Kirik, D., Kokaia, Z., Lindvall, O., 2002. Neuronal replacement from endogenous precursors in the adult brain after stroke. Nat. Med. 8, 963–970. Back, S.A., Han, B.H., Luo, N.L., Chricton, C.A., Xanthoudakis, S., Tam, J., Arvin, K.L., Holtzman, D.M., 2002. Selective vulnerability of late oligodendrocyte progenitors to hypoxia-ischemia. J. Neurosci. 22, 455–463. Bala, S., Marcos, M., Kodys, K., Csak, T., Catalano, D., Mandrekar, P., Szabo, G., 2011. Up-regulation of microRNA-155 in macrophages contributes to increased tumor necrosis factor {alpha} (TNF{alpha}) production via increased mRNA halflife in alcoholic liver disease. J. Biol. Chem. 286, 1436–1444.
19
Bard, F., Cannon, C., Barbour, R., Burke, R.L., Games, D., Grajeda, H., Guido, T., Hu, K., Huang, J., Johnson-Wood, K., Khan, K., Kholodenko, D., Lee, M., Lieberburg, I., Motter, R., Nguyen, M., Soriano, F., Vasquez, N., Weiss, K., Welch, B., Seubert, P., Schenk, D., Yednock, T., 2000. Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat. Med. 6, 916–919. Biber, K., Neumann, H., Inoue, K., Boddeke, H.W., 2007. Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci. 30, 596–602. Binder, M.D., Xiao, J., Kemper, D., Ma, G.Z., Murray, S.S., Kilpatrick, T.J., 2011. Gas6 increases myelination by oligodendrocytes and its deficiency delays recovery following cuprizone-induced demyelination. PLoS ONE 6, e17727. Biswas, S.K., Mantovani, A., 2010. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat. Immunol. 11, 889–896. Blobe, G.C., Schiemann, W.P., Lodish, H.F., 2000. Role of transforming growth factor beta in human disease. N. Engl. J. Med. 342, 1350–1358. Block, F., Dihne, M., Loos, M., 2005. Inflammation in areas of remote changes following focal brain lesion. Prog. Neurobiol. 75, 342–365. Block, M.L., Zecca, L., Hong, J.S., 2007. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat. Rev. Neurosci. 8, 57–69. Bogoslovsky, T., Chaudhry, A., Latour, L., Maric, D., Luby, M., Spatz, M., Frank, J., Warach, S., 2010. Endothelial progenitor cells correlate with lesion volume and growth in acute stroke. Neurology 75, 2059–2062. Boison, D., Chen, J.F., Fredholm, B.B., 2010. Adenosine signaling and function in glial cells. Cell Death Differ. 17, 1071–1082. Boulis, N.M., Federici, T., Glass, J.D., Lunn, J.S., Sakowski, S.A., Feldman, E.L., 2011. Translational stem cell therapy for amyotrophic lateral sclerosis. Nat. Rev. Neurol. 8, 172–176. Brifault, C., Gras, M., Liot, D., May, V., Vaudry, D., Wurtz, O., 2015. Delayed pituitary adenylate cyclase-activating polypeptide delivery after brain stroke improves functional recovery by inducing m2 microglia/macrophage polarization. Stroke 46, 520–528. Broderick, C., Hoek, R.M., Forrester, J.V., Liversidge, J., Sedgwick, J.D., Dick, A.D., 2002. Constitutive retinal CD200 expression regulates resident microglia and activation state of inflammatory cells during experimental autoimmune uveoretinitis. Am. J. Pathol. 161, 1669–1677. Brown, G.C., Neher, J.J., 2014. Microglial phagocytosis of live neurons. Nat. Rev. Neurosci. 15, 209–216. Burnstock, G., 2007. Physiology and pathophysiology of purinergic neurotransmission. Physiol. Rev. 87, 659–797. Burnstock, G., 2008. Purinergic signalling and disorders of the central nervous system. Nat. Rev. Drug Discovery 7, 575–590. Burnstock, G., Fredholm, B.B., Verkhratsky, A., 2011. Adenosine and ATP receptors in the brain. Curr. Top. Med. Chem. 11, 973–1011. Butovsky, O., Jedrychowski, M.P., Cialic, R., Krasemann, S., Murugaiyan, G., Fanek, Z., Greco, D.J., Wu, P.M., Doykan, C.E., Kiner, O., Lawson, R.J., Frosch, M.P., Pochet, N., Fatimy, R.E., Krichevsky, A.M., Gygi, S.P., Lassmann, H., Berry, J., Cudkowicz, M.E., Weiner, H.L., 2015. Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann. Neurol. 77, 75–99. Butovsky, O., Jedrychowski, M.P., Moore, C.S., Cialic, R., Lanser, A.J., Gabriely, G., Koeglsperger, T., Dake, B., Wu, P.M., Doykan, C.E., Fanek, Z., Liu, L., Chen, Z., Rothstein, J.D., Ransohoff, R.M., Gygi, S.P., Antel, J.P., Weiner, H.L., 2014. Identification of a unique TGF-beta-dependent molecular and functional signature in microglia. Nat. Neurosci. 17, 131–143. Butovsky, O., Siddiqui, S., Gabriely, G., Lanser, A.J., Dake, B., Murugaiyan, G., Doykan, C.E., Wu, P.M., Gali, R.R., Iyer, L.K., Lawson, R., Berry, J., Krichevsky, A.M., Cudkowicz, M.E., Weiner, H.L., 2012. Modulating inflammatory monocytes with a unique microRNA gene signature ameliorates murine ALS. J. Clin. Invest. 122, 3063–3087. Butovsky, O., Ziv, Y., Schwartz, A., Landa, G., Talpalar, A.E., Pluchino, S., Martino, G., Schwartz, M., 2006. Microglia activated by IL-4 or IFN-gamma differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells. Mol. Cell. Neurosci. 31, 149–160. Cai, Z., Lin, S., Fan, L.W., Pang, Y., Rhodes, P.G., 2006. Minocycline alleviates hypoxic– ischemic injury to developing oligodendrocytes in the neonatal rat brain. Neuroscience 137, 425–435. Campanella, M., Sciorati, C., Tarozzo, G., Beltramo, M., 2002. Flow cytometric analysis of inflammatory cells in ischemic rat brain. Stroke 33, 586–592. Cardona, A.E., Pioro, E.P., Sasse, M.E., Kostenko, V., Cardona, S.M., Dijkstra, I.M., Huang, D., Kidd, G., Dombrowski, S., Dutta, R., Lee, J.C., Cook, D.N., Jung, S., Lira, S.A., Littman, D.R., Ransohoff, R.M., 2006. Control of microglial neurotoxicity by the fractalkine receptor. Nat. Neurosci. 9, 917–924. Cardoso, A.L., Guedes, J.R., Pereira de Almeida, L., Pedroso de Lima, M.C., 2012. miR155 modulates microglia-mediated immune response by down-regulating SOCS-1 and promoting cytokine and nitric oxide production. Immunology 135, 73–88. Carreira, B.P., Morte, M.I., Santos, A.I., Lourenco, A.S., Ambrosio, A.F., Carvalho, C.M., Araujo, I.M., 2014. Nitric oxide from inflammatory origin impairs neural stem cell proliferation by inhibiting epidermal growth factor receptor signaling. Front. Cell. Neurosci. 8, 343. Cartier, N., Lewis, C.A., Zhang, R., Rossi, F.M., 2014. The role of microglia in human disease: therapeutic tool or target? Acta Neuropathol. 128, 363–380. Caso, J.R., Pradillo, J.M., Hurtado, O., Lorenzo, P., Moro, M.A., Lizasoain, I., 2007. Tolllike receptor 4 is involved in brain damage and inflammation after experimental stroke. Circulation 115, 1599–1608. Cavaliere, F., Florenzano, F., Amadio, S., Fusco, F.R., Viscomi, M.T., D’Ambrosi, N., Vacca, F., Sancesario, G., Bernardi, G., Molinari, M., Volonte, C., 2003. Up-regulation of
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
1617 1618 1619 1620 1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663 1664 1665 1666 1667 1668 1669 1670 1671 1672 1673 1674 1675 1676 1677 1678 1679 1680 1681 1682 1683 1684 1685 1686 1687 1688 1689 1690 1691 1692 1693 1694 1695 1696 1697 1698 1699 1700 1701 1702
G Model
PRONEU 1417 1–25 20
1703 1704 1705 1706 1707 1708 1709 1710 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723 1724 1725 1726 1727 1728 1729 1730 1731 1732 1733 1734 1735 1736 1737 1738 1739 1740 1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752 1753 1754 1755 1756 1757 1758 1759 1760 1761 1762 1763 1764 1765 1766 1767 1768 1769 1770 1771 1772 1773 1774 1775 1776 1777 1778 1779 1780 1781 1782 1783 1784 1785 1786
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
P2X2, P2X4 receptor and ischemic cell death: prevention by P2 antagonists. Neuroscience 120, 85–98. Chen, C., Lin, X., Wang, J., Tang, G., Mu, Z., Chen, X., Xu, J., Wang, Y., Zhang, Z., Yang, G.Y., 2014. Effect of HMGB1 on the paracrine action of EPC promotes postischemic neovascularization in mice. Stem Cells 32, 2679–2689. Chen, J., Chen, J., Chen, S., Zhang, C., Zhang, L., Xiao, X., Das, A., Zhao, Y., Yuan, B., Morris, M., Zhao, B., Chen, Y., 2012. Transfusion of CXCR4-primed endothelial progenitor cells reduces cerebral ischemic damage and promotes repair in db/ db diabetic mice. PLoS ONE 7, e50105. Cherry, J.D., Olschowka, J.A., O’Banion, M.K., 2014. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J. Neuroinflammation 11, 98. Chhor, V., Le Charpentier, T., Lebon, S., Ore, M.V., Celador, I.L., Josserand, J., Degos, V., Jacotot, E., Hagberg, H., Savman, K., Mallard, C., Gressens, P., Fleiss, B., 2013. Characterization of phenotype markers and neuronotoxic potential of polarised primary microglia in vitro. Brain Behav. Immun. 32, 70–85. Cho, K.J., Song, J., Oh, Y., Lee, J.E., 2015. MicroRNA-Let-7a regulates the function of microglia in inflammation. Mol. Cell. Neurosci. 68, 167–176. Chu, H.X., Kim, H.A., Lee, S., Moore, J.P., Chan, C.T., Vinh, A., Gelderblom, M., Arumugam, T.V., Broughton, B.R., Drummond, G.R., Sobey, C.G., 2014. Immune cell infiltration in malignant middle cerebral artery infarction: comparison with transient cerebral ischemia. J. Cereb. Blood Flow Metab. 34, 450–459. Chu, K., Jung, K.H., Lee, S.T., Park, H.K., Sinn, D.I., Kim, J.M., Kim, D.H., Kim, J.H., Kim, S.J., Song, E.C., Kim, M., Lee, S.K., Roh, J.K., 2008. Circulating endothelial progenitor cells as a new marker of endothelial dysfunction or repair in acute stroke. Stroke 39, 1441–1447. Chu, K., Yin, B., Wang, J., Peng, G., Liang, H., Xu, Z., Du, Y., Fang, M., Xia, Q., Luo, B., 2012. Inhibition of P2X7 receptor ameliorates transient global cerebral ischemia/reperfusion injury via modulating inflammatory responses in the rat hippocampus. J. Neuroinflammation 9, 69. Cipriani, R., Villa, P., Chece, G., Lauro, C., Paladini, A., Micotti, E., Perego, C., De Simoni, M.G., Fredholm, B.B., Eusebi, F., Limatola, C., 2011. CX3CL1 is neuroprotective in permanent focal cerebral ischemia in rodents. J. Neurosci. 31, 16327–16335. Clark, A.K., Yip, P.K., Grist, J., Gentry, C., Staniland, A.A., Marchand, F., Dehvari, M., Wotherspoon, G., Winter, J., Ullah, J., Bevan, S., Malcangio, M., 2007. Inhibition of spinal microglial cathepsin S for the reversal of neuropathic pain. Proc. Natl. Acad. Sci. U.S.A. 104, 10655–10660. Clausen, B.H., Lambertsen, K.L., Babcock, A.A., Holm, T.H., Dagnaes-Hansen, F., Finsen, B., 2008. Interleukin-1beta and tumor necrosis factor-alpha are expressed by different subsets of microglia and macrophages after ischemic stroke in mice. J. Neuroinflammation 5, 46. Correa, F.G., Hernangomez, M., Guaza, C., 2013. Understanding microglia-neuron cross talk: relevance of the microglia-neuron cocultures. Methods Mol. Biol. 1041, 215–229. Cuadros, M.A., Martin, C., Coltey, P., Almendros, A., Navascues, J., 1993. First appearance, distribution, and origin of macrophages in the early development of the avian central nervous system. J. Comp. Neurol. 330, 113–129. da Fonseca, A.C., Matias, D., Garcia, C., Amaral, R., Geraldo, L.H., Freitas, C., Lima, F.R., 2014. The impact of microglial activation on blood–brain barrier in brain diseases. Front. Cell. Neurosci. 8, 362. Danielyan, L., Beer-Hammer, S., Stolzing, A., Schafer, R., Siegel, G., Fabian, C., Kahle, P., Biedermann, T., Lourhmati, A., Buadze, M., Novakovic, A., Proksch, B., Gleiter, C.H., Frey, W.H., Schwab, M., 2014. Intranasal delivery of bone marrow-derived mesenchymal stem cells, macrophages, and microglia to the brain in mouse models of Alzheimer’s and Parkinson’s disease. Cell Transplant. 23 (Suppl. 1), 123–139. Darsalia, V., Allison, S.J., Cusulin, C., Monni, E., Kuzdas, D., Kallur, T., Lindvall, O., Kokaia, Z., 2011. Cell number and timing of transplantation determine survival of human neural stem cell grafts in stroke-damaged rat brain. J. Cereb. Blood Flow Metab. 31, 235–242. David, S., Kroner, A., 2011. Repertoire of microglial and macrophage responses after spinal cord injury. Nat. Rev. Neurosci. 12, 388–399. de Rivero Vaccari, J.P., Bastien, D., Yurcisin, G., Pineau, I., Dietrich, W.D., De Koninck, Y., Keane, R.W., Lacroix, S., 2012. P2X4 receptors influence inflammasome activation after spinal cord injury. J. Neurosci. 32, 3058–3066. Deckert, M., Sedgwick, J.D., Fischer, E., Schluter, D., 2006. Regulation of microglial cell responses in murine Toxoplasma encephalitis by CD200/CD200 receptor interaction. Acta Neuropathol. 111, 548–558. DeKoter, R.P., Walsh, J.C., Singh, H., 1998. PU.1 regulates both cytokine-dependent proliferation and differentiation of granulocyte/macrophage progenitors. EMBO J. 17, 4456–4468. Del Puerto, A., Wandosell, F., Garrido, J.J., 2013. Neuronal and glial purinergic receptors functions in neuron development and brain disease. Front. Cell. Neurosci. 7, 197. Denes, A., Ferenczi, S., Halasz, J., Kornyei, Z., Kovacs, K.J., 2008. Role of CX3CR1 (fractalkine receptor) in brain damage and inflammation induced by focal cerebral ischemia in mouse. J. Cereb. Blood Flow Metab. 28, 1707–1721. Deng, Y., Lu, J., Sivakumar, V., Ling, E.A., Kaur, C., 2008. Amoeboid microglia in the periventricular white matter induce oligodendrocyte damage through expression of proinflammatory cytokines via MAP kinase signaling pathway in hypoxic neonatal rats. Brain Pathol. 18, 387–400. Deng, Y.Y., Lu, J., Ling, E.A., Kaur, C., 2009. Monocyte chemoattractant protein-1 (MCP-1) produced via NF-kappaB signaling pathway mediates migration of amoeboid microglia in the periventricular white matter in hypoxic neonatal rats. Glia 57, 604–621.
Deo, M., Yu, J.Y., Chung, K.H., Tippens, M., Turner, D.L., 2006. Detection of mammalian microRNA expression by in situ hybridization with RNA oligonucleotides. Dev. Dyn. 235, 2538–2548. Dewar, D., Underhill, S.M., Goldberg, M.P., 2003. Oligodendrocytes and ischemic brain injury. J. Cereb. Blood Flow Metab. 23, 263–274. Dimitrijevic, O.B., Stamatovic, S.M., Keep, R.F., Andjelkovic, A.V., 2007. Absence of the chemokine receptor CCR2 protects against cerebral ischemia/reperfusion injury in mice. Stroke 38, 1345–1353. Doeppner, T.R., Doehring, M., Bretschneider, E., Zechariah, A., Kaltwasser, B., Muller, B., Koch, J.C., Bahr, M., Hermann, D.M., Michel, U., 2013. MicroRNA-124 protects against focal cerebral ischemia via mechanisms involving Usp14-dependent REST degradation. Acta Neuropathol. 126, 251–265. Donega, V., Nijboer, C.H., van Tilborg, G., Dijkhuizen, R.M., Kavelaars, A., Heijnen, C.J., 2014. Intranasally administered mesenchymal stem cells promote a regenerative niche for repair of neonatal ischemic brain injury. Exp. Neurol. 261, 53–64. Dringen, R., 2005. Oxidative and antioxidative potential of brain microglial cells. Antioxid. Redox Signal. 7, 1223–1233. Dziewulska, D., Mossakowski, M.J., 2003. Cellular expression of tumor necrosis factor a and its receptors in human ischemic stroke. Clin. Neuropathol. 22, 35– 40. Egashira, Y., Suzuki, Y., Azuma, Y., Takagi, T., Mishiro, K., Sugitani, S., Tsuruma, K., Shimazawa, M., Yoshimura, S., Kashimata, M., Iwama, T., Hara, H., 2013. The growth factor progranulin attenuates neuronal injury induced by cerebral ischemia-reperfusion through the suppression of neutrophil recruitment. J. Neuroinflammation 10, 105. Ekdahl, C.T., 2012. Microglial activation—tuning and pruning adult neurogenesis. Front. Pharmacol. 3, 41. Ekdahl, C.T., Claasen, J.H., Bonde, S., Kokaia, Z., Lindvall, O., 2003. Inflammation is detrimental for neurogenesis in adult brain. Proc. Natl. Acad. Sci. U.S.A. 100, 13632–13637. Ekdahl, C.T., Kokaia, Z., Lindvall, O., 2009. Brain inflammation and adult neurogenesis: the dual role of microglia. Neuroscience 158, 1021–1029. Elkabes, S., DiCicco-Bloom, E.M., Black, I.B., 1996. Brain microglia/macrophages express neurotrophins that selectively regulate microglial proliferation and function. J. Neurosci. 16, 2508–2521. Ernst, A., Alkass, K., Bernard, S., Salehpour, M., Perl, S., Tisdale, J., Possnert, G., Druid, H., Frisen, J., 2014. Neurogenesis in the striatum of the adult human brain. Cell 156, 1072–1083. Eyo, U.B., Miner, S.A., Ahlers, K.E., Wu, L.J., Dailey, M.E., 2013. P2X7 receptor activation regulates microglial cell death during oxygen–glucose deprivation. Neuropharmacology 73, 311–319. Eyo, U.B., Wu, L.J., 2013. Bidirectional microglia-neuron communication in the healthy brain. Neural Plast. 2013, 456857. Fagan, S.C., Waller, J.L., Nichols, F.T., Edwards, D.J., Pettigrew, L.C., Clark, W.M., Hall, C.E., Switzer, J.A., Ergul, A., Hess, D.C., 2010. Minocycline to improve neurologic outcome in stroke (MINOS): a dose-finding study. Stroke 41, 2283–2287. Fan, Y., Shen, F., Frenzel, T., Zhu, W., Ye, J., Liu, J., Chen, Y., Su, H., Young, W.L., Yang, G.Y., 2010. Endothelial progenitor cell transplantation improves long-term stroke outcome in mice. Ann. Neurol. 67, 488–497. Fang, F., Lue, L.F., Yan, S., Xu, H., Luddy, J.S., Chen, D., Walker, D.G., Stern, D.M., Yan, S., Schmidt, A.M., Chen, J.X., Yan, S.S., 2010. RAGE-dependent signaling in microglia contributes to neuroinflammation, Abeta accumulation, and impaired learning/ memory in a mouse model of Alzheimer’s disease. FASEB J. 24, 1043–1055. Faustino, J.V., Wang, X., Johnson, C.E., Klibanov, A., Derugin, N., Wendland, M.F., Vexler, Z.S., 2011. Microglial cells contribute to endogenous brain defenses after acute neonatal focal stroke. J. Neurosci. 31, 12992–13001. Franco, E.C., Cardoso, M.M., Gouveia, A., Pereira, A., Gomes-Leal, W., 2012. Modulation of microglial activation enhances neuroprotection and functional recovery derived from bone marrow mononuclear cell transplantation after cortical ischemia. Neurosci. Res. 73, 122–132. Fuhrmann, M., Bittner, T., Jung, C.K., Burgold, S., Page, R.M., Mitteregger, G., Haass, C., LaFerla, F.M., Kretzschmar, H., Herms, J., 2010. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer’s disease. Nat. Neurosci. 13, 411–413. Gay, F.W., Drye, T.J., Dick, G.W., Esiri, M.M., 1997. The application of multifactorial cluster analysis in the staging of plaques in early multiple sclerosis. Identification and characterization of the primary demyelinating lesion. Brain 120 (Pt 8), 1461–1483. Gehrmann, J., Bonnekoh, P., Miyazawa, T., Hossmann, K.A., Kreutzberg, G.W., 1992. Immunocytochemical study of an early microglial activation in ischemia. J. Cereb. Blood Flow Metab. 12, 257–269. Gelderblom, M., Leypoldt, F., Steinbach, K., Behrens, D., Choe, C.U., Siler, D.A., Arumugam, T.V., Orthey, E., Gerloff, C., Tolosa, E., Magnus, T., 2009. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke 40, 1849–1857. Gelderblom, M., Weymar, A., Bernreuther, C., Velden, J., Arunachalam, P., Steinbach, K., Orthey, E., Arumugam, T.V., Leypoldt, F., Simova, O., Thom, V., Friese, M.A., Prinz, I., Holscher, C., Glatzel, M., Korn, T., Gerloff, C., Tolosa, E., Magnus, T., 2012. Neutralization of the IL-17 axis diminishes neutrophil invasion and protects from ischemic stroke. Blood 120, 3793–3802. Gendron, F.P., Chalimoniuk, M., Strosznajder, J., Shen, S., Gonzalez, F.A., Weisman, G.A., Sun, G.Y., 2003. P2X7 nucleotide receptor activation enhances IFN gammainduced type II nitric oxide synthase activity in BV-2 microglial cells. J. Neurochem. 87, 344–352.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
1787 1788 1789 1790 1791 1792 1793 1794 1795 1796 1797 1798 1799 1800 1801 1802 1803 1804 1805 1806 1807 1808 1809 1810 1811 1812 1813 1814 1815 1816 1817 1818 1819 1820 1821 1822 1823 1824 1825 1826 1827 1828 1829 1830 1831 1832 1833 1834 1835 1836 1837 1838 1839 1840 1841 1842 1843 1844 1845 1846 1847 1848 1849 1850 1851 1852 1853 1854 1855 1856 1857 1858 1859 1860 1861 1862 1863 1864 1865 1866 1867 1868 1869 1870
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
1871 1872 1873 1874 1875 1876 1877 1878 1879 1880 1881 1882 1883 1884 1885 1886 1887 1888 1889 1890 1891 1892 1893 1894 1895 1896 1897 1898 1899 1900 1901 1902 1903 1904 1905 1906 1907 1908 1909 1910 1911 1912 1913 1914 1915 1916 1917 1918 1919 1920 1921 1922 1923 1924 1925 1926 1927 1928 1929 1930 1931 1932 1933 1934 1935 1936 1937 1938 1939 1940 1941 1942 1943 1944 1945 1946 1947 1948 1949 1950 1951 1952 1953 1954 1955 1956
Gerhard, A., Schwarz, J., Myers, R., Wise, R., Banati, R.B., 2005. Evolution of microglial activation in patients after ischemic stroke: a [11C](R)-PK11195 PET study. NeuroImage 24, 591–595. Gibson, C.L., Coughlan, T.C., Murphy, S.P., 2005. Glial nitric oxide and ischemia. Glia 50, 417–426. Gilje, P., Gidlof, O., Rundgren, M., Cronberg, T., Al-Mashat, M., Olde, B., Friberg, H., Erlinge, D., 2014. The brain-enriched microRNA miR-124 in plasma predicts neurological outcome after cardiac arrest. Crit. Care 18, R40. Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S., Mehler, M.F., Conway, S.J., Ng, L.G., Stanley, E.R., Samokhvalov, I.M., Merad, M., 2010. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845. Ginhoux, F., Lim, S., Hoeffel, G., Low, D., Huber, T., 2013. Origin and differentiation of microglia. Front. Cell. Neurosci. 7, 45. Ginis, I., Jaiswal, R., Klimanis, D., Liu, J., Greenspon, J., Hallenbeck, J.M., 2002. TNFalpha-induced tolerance to ischemic injury involves differential control of NFkappaB transactivation: the role of NF-kappaB association with p300 adaptor. J. Cereb. Blood Flow Metab. 22, 142–152. Girard, S., Brough, D., Lopez-Castejon, G., Giles, J., Rothwell, N.J., Allan, S.M., 2013. Microglia and macrophages differentially modulate cell death after brain injury caused by oxygen–glucose deprivation in organotypic brain slices. Glia 61, 813– 824. Gogel, S., Gubernator, M., Minger, S.L., 2011. Progress and prospects: stem cells and neurological diseases. Gene Ther. 18, 1–6. Gomes, C., Ferreira, R., George, J., Sanches, R., Rodrigues, D.I., Goncalves, N., Cunha, R.A., 2013. Activation of microglial cells triggers a release of brain-derived neurotrophic factor (BDNF) inducing their proliferation in an adenosine A2A receptor-dependent manner: A2A receptor blockade prevents BDNF release and proliferation of microglia. J. Neuroinflammation 10, 16. Goodman, Y., Mattson, M.P., 1996. Ceramide protects hippocampal neurons against excitotoxic and oxidative insults, and amyloid beta-peptide toxicity. J. Neurochem. 66, 869–872. Gordon, S., 2003. Alternative activation of macrophages. Nat. Rev. Immunol. 3, 23–35. Gordon, S., Taylor, P.R., 2005. Monocyte and macrophage heterogeneity. Nat. Rev. Immunol. 5, 953–964. Gregersen, R., Lambertsen, K., Finsen, B., 2000. Microglia and macrophages are the major source of tumor necrosis factor in permanent middle cerebral artery occlusion in mice. J. Cereb. Blood Flow Metab. 20, 53–65. Guedes, J., Cardoso, A.L., Pedroso de Lima, M.C., 2013. Involvement of microRNA in microglia-mediated immune response. Clin. Dev. Immunol. 2013, 186872. Gulyas, B., Toth, M., Schain, M., Airaksinen, A., Vas, A., Kostulas, K., Lindstrom, P., Hillert, J., Halldin, C., 2012. Evolution of microglial activation in ischaemic core and peri-infarct regions after stroke: a PET study with the TSPO molecular imaging biomarker [((11))C]vinpocetine. J. Neurol. Sci. 320, 110–117. Hanisch, U.K., Kettenmann, H., 2007. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 10, 1387–1394. Hassani, Z., O’Reilly, J., Pearse, Y., Stroemer, P., Tang, E., Sinden, J., Price, J., Thuret, S., 2012. Human neural progenitor cell engraftment increases neurogenesis and microglial recruitment in the brain of rats with stroke. PLoS ONE 7, e50444. Haynes, R.L., Baud, O., Li, J., Kinney, H.C., Volpe, J.J., Folkerth, D.R., 2005. Oxidative and nitrative injury in periventricular leukomalacia: a review. Brain Pathol. 15, 225–233. Haynes, S.E., Hollopeter, G., Yang, G., Kurpius, D., Dailey, M.E., Gan, W.B., Julius, D., 2006. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 9, 1512–1519. Hegyi, B., Kornyei, Z., Ferenczi, S., Fekete, R., Kudlik, G., Kovacs, K.J., Madarasz, E., Uher, F., 2014. Regulation of mouse microglia activation and effector functions by bone marrow-derived mesenchymal stem cells. Stem Cells Dev. 23, 2600– 2612. Heldmann, U., Mine, Y., Kokaia, Z., Ekdahl, C.T., Lindvall, O., 2011. Selective depletion of Mac-1-expressing microglia in rat subventricular zone does not alter neurogenic response early after stroke. Exp. Neurol. 229, 391–398. Herrera-Molina, R., von Bernhardi, R., 2005. Transforming growth factor-beta 1 produced by hippocampal cells modulates microglial reactivity in culture. Neurobiol. Dis. 19, 229–236. Hide, I., Tanaka, M., Inoue, A., Nakajima, K., Kohsaka, S., Inoue, K., Nakata, Y., 2000. Extracellular ATP triggers tumor necrosis factor-alpha release from rat microglia. J. Neurochem. 75, 965–972. Hirasawa, T., Ohsawa, K., Imai, Y., Ondo, Y., Akazawa, C., Uchino, S., Kohsaka, S., 2005. Visualization of microglia in living tissues using Iba1-EGFP transgenic mice. J. Neurosci. Res. 81, 357–362. Hoehn, B.D., Palmer, T.D., Steinberg, G.K., 2005. Neurogenesis in rats after focal cerebral ischemia is enhanced by indomethacin. Stroke 36, 2718–2724. Hoek, R.M., Ruuls, S.R., Murphy, C.A., Wright, G.J., Goddard, R., Zurawski, S.M., Blom, B., Homola, M.E., Streit, W.J., Brown, M.H., Barclay, A.N., Sedgwick, J.D., 2000. Down-regulation of the macrophage lineage through interaction with OX2 (CD200). Science 290, 1768–1771. Hu, X., Leak, R.K., Shi, Y., Suenaga, J., Gao, Y., Zheng, P., Chen, J., 2015. Microglial and macrophage polarization-new prospects for brain repair. Nat. Rev. Neurol. 11, 56–64. Hu, X., Li, P., Guo, Y., Wang, H., Leak, R.K., Chen, S., Gao, Y., Chen, J., 2012. Microglia/ macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke 43, 3063–3070. Hu, X., Liou, A.K., Leak, R.K., Xu, M., An, C., Suenaga, J., Shi, Y., Gao, Y., Zheng, P., Chen, J., 2014. Neurobiology of microglial action in CNS injuries: receptor-mediated signaling mechanisms and functional roles. Prog. Neurobiol. 119–120, 60–84.
21
Iadecola, C., Anrather, J., 2011. The immunology of stroke: from mechanisms to 1957 translation. Nat. Med. 17, 796–808. 1958 Iadecola, C., Ross, M.E., 1997. Molecular pathology of cerebral ischemia: delayed 1959 gene expression and strategies for neuroprotection. Ann. N.Y. Acad. Sci. 835, 1960 203–217. 1961 Inose, Y., Kato, Y., Kitagawa, K., Uchiyama, S., Shibata, N., 2014. Activated microglia 1962 in ischemic stroke penumbra upregulate MCP-1 and CCR2 expression in re1963 sponse to lysophosphatidylcholine derived from adjacent neurons and astro1964 cytes. Neuropathology. Q81965 Iskander, A., Knight, R.A., Zhang, Z.G., Ewing, J.R., Shankar, A., Varma, N.R., Bagher1966 Ebadian, H., Ali, M.M., Arbab, A.S., Janic, B., 2013. Intravenous administration of 1967 human umbilical cord blood-derived AC133+ endothelial progenitor cells in rat 1968 stroke model reduces infarct volume: magnetic resonance imaging and histo1969 logical findings. Stem Cells Transl. Med. 2, 703–714. 1970 Ito, D., Tanaka, K., Suzuki, S., Dembo, T., Fukuuchi, Y., 2001. Enhanced expression of 1971 Iba1, ionized calcium-binding adapter molecule 1, after transient focal cerebral 1972 ischemia in rat brain. Stroke 32, 1208–1215. 1973 Jackman, K., Kahles, T., Lane, D., Garcia-Bonilla, L., Abe, T., Capone, C., Hochrainer, K., 1974 Voss, H., Zhou, P., Ding, A., Anrather, J., Iadecola, C., 2013. Progranulin deficiency 1975 promotes post-ischemic blood–brain barrier disruption. J. Neurosci. 33, 19579– 1976 19589. 1977 Jalal, F.Y., Yang, Y., Thompson, J., Lopez, A.C., Rosenberg, G.A., 2012. Myelin 1978 loss associated with neuroinflammation in hypertensive rats. Stroke 43, 1979 1115–1122. 1980 Jiang, C., Wang, J., Yu, L., Ou, C., Liu, X., Zhao, X., Wang, J., 2013. Comparison of the 1981 therapeutic effects of bone marrow mononuclear cells and microglia for per1982 manent cerebral ischemia. Behav. Brain Res. 250, 222–229. 1983 Jin, K., Minami, M., Lan, J.Q., Mao, X.O., Batteur, S., Simon, R.P., Greenberg, D.A., 2001. 1984 Neurogenesis in dentate subgranular zone and rostral subventricular zone 1985 after focal cerebral ischemia in the rat. Proc. Natl. Acad. Sci. U.S.A. 98, 4710– 1986 4715. 1987 Jin, Q., Cheng, J., Liu, Y., Wu, J., Wang, X., Wei, S., Zhou, X., Qin, Z., Jia, J., Zhen, X., 2014. 1988 Improvement of functional recovery by chronic metformin treatment is asso1989 ciated with enhanced alternative activation of microglia/macrophages and 1990 increased angiogenesis and neurogenesis following experimental stroke. Brain 1991 Behav. Immun. 40, 131–142. 1992 Jin, R., Yang, G., Li, G., 2010. Inflammatory mechanisms in ischemic stroke: role of 1993 inflammatory cells. J. Leukoc. Biol. 87, 779–789. 1994 Jinno, S., Fleischer, F., Eckel, S., Schmidt, V., Kosaka, T., 2007. Spatial arrangement of 1995 microglia in the mouse hippocampus: a stereological study in comparison with 1996 astrocytes. Glia 55, 1334–1347. 1997 Jolivel, V., Bicker, F., Biname, F., Ploen, R., Keller, S., Gollan, R., Jurek, B., Birkenstock, 1998 J., Poisa-Beiro, L., Bruttger, J., Opitz, V., Thal, S.C., Waisman, A., Bauerle, T., 1999 Schafer, M.K., Zipp, F., Schmidt, M.H., 2015. Perivascular microglia promote 2000 blood vessel disintegration in the ischemic penumbra. Acta Neuropathol. 129, 2001 279–295. 2002 Junn, E., Mouradian, M.M., 2012. MicroRNAs in neurodegenerative diseases and 2003 their therapeutic potential. Pharmacol. Ther. 133, 142–150. 2004 Kacimi, R., Giffard, R.G., Yenari, M.A., 2011. Endotoxin-activated microglia injure 2005 brain derived endothelial cells via NF-kappaB, JAK-STAT and JNK stress kinase 2006 pathways. J. Inflamm. (Lond.) 8, 7. 2007 Kanazawa, M., Kawamura, K., Takahashi, T., Miura, M., Tanaka, Y., Koyama, M., 2008 Toriyabe, M., Igarashi, H., Nakada, T., Nishihara, M., Nishizawa, M., Shimohata, 2009 T., 2015. Multiple therapeutic effects of progranulin on experimental acute 2010 ischaemic stroke. Brain 138, 1932–1948. 2011 Kataoka, A., Tozaki-Saitoh, H., Koga, Y., Tsuda, M., Inoue, K., 2009. Activation of P2X7 2012 receptors induces CCL3 production in microglial cells through transcription 2013 factor NFAT. J. Neurochem. 108, 115–125. 2014 Kawanokuchi, J., Shimizu, K., Nitta, A., Yamada, K., Mizuno, T., Takeuchi, H., 2015 Suzumura, A., 2008. Production and functions of IL-17 in microglia. 2016 J. Neuroimmunol. 194, 54–61. 2017 Kettenmann, H., Hanisch, U.K., Noda, M., Verkhratsky, A., 2011. Physiology of 2018 microglia. Physiol. Rev. 91, 461–553. 2019 Kierdorf, K., Erny, D., Goldmann, T., Sander, V., Schulz, C., Perdiguero, E.G., Wie2020 ghofer, P., Heinrich, A., Riemke, P., Holscher, C., Muller, D.N., Luckow, B., Brocker, 2021 T., Debowski, K., Fritz, G., Opdenakker, G., Diefenbach, A., Biber, K., Heiken2022 walder, M., Geissmann, F., Rosenbauer, F., Prinz, M., 2013. Microglia emerge 2023 from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. 2024 Nat. Neurosci. 16, 273–280. 2025 Kigerl, K.A., Gensel, J.C., Ankeny, D.P., Alexander, J.K., Donnelly, D.J., Popovich, P.G., 2026 2009. Identification of two distinct macrophage subsets with divergent effects 2027 causing either neurotoxicity or regeneration in the injured mouse spinal cord. 2028 J. Neurosci. 29, 13435–13444. 2029 Kilic, U., Kilic, E., Matter, C.M., Bassetti, C.L., Hermann, D.M., 2008. TLR-4 deficiency 2030 protects against focal cerebral ischemia and axotomy-induced neurodegenera2031 tion. Neurobiol. Dis. 31, 33–40. 2032 Kim, B.J., Kim, M.J., Park, J.M., Lee, S.H., Kim, Y.J., Ryu, S., Kim, Y.H., Yoon, B.W., 2009. 2033 Reduced neurogenesis after suppressed inflammation by minocycline in tran2034 sient cerebral ischemia in rat. J. Neurol. Sci. 279, 70–75. 2035 Kim, J., Krichevsky, A., Grad, Y., Hayes, G.D., Kosik, K.S., Church, G.M., Ruvkun, G., 2036 2004. Identification of many microRNAs that copurify with polyribosomes in 2037 mammalian neurons. Proc. Natl. Acad. Sci. U.S.A. 101, 360–365. 2038 Kim, J.Y., Kim, N., Yenari, M.A., 2014. Mechanisms and potential therapeutic 2039 applications of microglial activation after brain injury. CNS Neurosci. Ther.. 2040 Kim, S.U., de Vellis, J., 2005. Microglia in health and disease. J. Neurosci. Res. 81, 2041 302–313. 2042
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
G Model
PRONEU 1417 1–25 22
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
2043 Kitamura, Y., Takata, K., Inden, M., Tsuchiya, D., Yanagisawa, D., Nakata, J., Taniguchi, 2044 T., 2004. Intracerebroventricular injection of microglia protects against focal 2045 brain ischemia. J. Pharmacol. Sci. 94, 203–206. 2046 Kitamura, Y., Yanagisawa, D., Inden, M., Takata, K., Tsuchiya, D., Kawasaki, T., 2047 Taniguchi, T., Shimohama, S., 2005. Recovery of focal brain ischemia-induced 2048 behavioral dysfunction by intracerebroventricular injection of microglia. 2049 J. Pharmacol. Sci. 97, 289–293. 2050 Kleinschnitz, C., Grund, H., Wingler, K., Armitage, M.E., Jones, E., Mittal, M., Barit, D., 2051 Schwarz, T., Geis, C., Kraft, P., Barthel, K., Schuhmann, M.K., Herrmann, A.M., 2052 Meuth, S.G., Stoll, G., Meurer, S., Schrewe, A., Becker, L., Gailus-Durner, V., Fuchs, 2053 H., Klopstock, T., de Angelis, M.H., Jandeleit-Dahm, K., Shah, A.M., Weissmann, 2054 N., Schmidt, H.H., 2010. Post-stroke inhibition of induced NADPH oxidase type 2055Q9 4 prevents oxidative stress and neurodegeneration. PLoS Biol. 8. 2056 Kobayashi, K., Imagama, S., Ohgomori, T., Hirano, K., Uchimura, K., Sakamoto, K., 2057 Hirakawa, A., Takeuchi, H., Suzumura, A., Ishiguro, N., Kadomatsu, K., 2013. 2058 Minocycline selectively inhibits M1 polarization of microglia. Cell Death Dis. 4, 2059 e525. 2060 Kokaia, Z., Martino, G., Schwartz, M., Lindvall, O., 2012. Cross-talk between neural 2061 stem cells and immune cells: the key to better brain repair? Nat. Neurosci. 15, 2062 1078–1087. 2063 Komine-Kobayashi, M., Chou, N., Mochizuki, H., Nakao, A., Mizuno, Y., Urabe, T., 2064 2004. Dual role of Fcgamma receptor in transient focal cerebral ischemia in 2065 mice. Stroke 35, 958–963. 2066 Korematsu, K., Goto, S., Nagahiro, S., Ushio, Y., 1994. Microglial response to transient 2067 focal cerebral ischemia: an immunocytochemical study on the rat cerebral 2068 cortex using anti-phosphotyrosine antibody. J. Cereb. Blood Flow Metab. 14, 2069 825–830. 2070 Korzhevskii, D.E., Lentsman, M.V., Kirik, O.V., Otellin, V.A., 2012. Morphological 2071 types of activated microglia in the hippocampus observed following transient 2072 total brain ischemia. Morfologiia 142, 30–33. 2073 Kreutzberg, G.W., 1996. Microglia: a sensor for pathological events in the CNS. 2074 Trends Neurosci. 19, 312–318. 2075 Krupinski, J., Kaluza, J., Kumar, P., Kumar, S., 1996. Immunocytochemical studies of 2076 cellular reaction in human ischemic brain stroke, MAB anti-CD68 stains macro2077 phages, astrocytes and microglial cells in infarcted area. Folia Neuropathol. 34, 2078 17–24. 2079 Lai, A.Y., Todd, K.G., 2006. Microglia in cerebral ischemia: molecular actions and 2080 interactions. Can. J. Physiol. Pharmacol. 84, 49–59. 2081 Lalancette-Hebert, M., Gowing, G., Simard, A., Weng, Y.C., Kriz, J., 2007. Selective 2082 ablation of proliferating microglial cells exacerbates ischemic injury in the 2083 brain. J. Neurosci. 27, 2596–2605. 2084 Lalancette-Hebert, M., Swarup, V., Beaulieu, J.M., Bohacek, I., Abdelhamid, E., Weng, 2085 Y.C., Sato, S., Kriz, J., 2012. Galectin-3 is required for resident microglia activa2086 tion and proliferation in response to ischemic injury. J. Neurosci. 32, 10383– 2087 10395. 2088 Lambertsen, K.L., Clausen, B.H., Babcock, A.A., Gregersen, R., Fenger, C., Nielsen, H.H., 2089 Haugaard, L.S., Wirenfeldt, M., Nielsen, M., Dagnaes-Hansen, F., Bluethmann, H., 2090 Faergeman, N.J., Meldgaard, M., Deierborg, T., Finsen, B., 2009. Microglia protect 2091 neurons against ischemia by synthesis of tumor necrosis factor. J. Neurosci. 29, 2092 1319–1330. 2093 Lampl, Y., Boaz, M., Gilad, R., Lorberboym, M., Dabby, R., Rapoport, A., Anca2094 Hershkowitz, M., Sadeh, M., 2007. Minocycline treatment in acute stroke: an 2095 open-label, evaluator-blinded study. Neurology 69, 1404–1410. 2096 Landgraf, P., Rusu, M., Sheridan, R., Sewer, A., Iovino, N., Aravin, A., Pfeffer, S., Rice, A., 2097 Kamphorst, A.O., Landthaler, M., Lin, C., Socci, N.D., Hermida, L., Fulci, V., 2098 Chiaretti, S., Foa, R., Schliwka, J., Fuchs, U., Novosel, A., Muller, R.U., Schermer, 2099 B., Bissels, U., Inman, J., Phan, Q., Chien, M., Weir, D.B., Choksi, R., De Vita, G., 2100 Frezzetti, D., Trompeter, H.I., Hornung, V., Teng, G., Hartmann, G., Palkovits, M., 2101 Di Lauro, R., Wernet, P., Macino, G., Rogler, C.E., Nagle, J.W., Ju, J., Papavasiliou, 2102 F.N., Benzing, T., Lichter, P., Tam, W., Brownstein, M.J., Bosio, A., Borkhardt, A., 2103 Russo, J.J., Sander, C., Zavolan, M., Tuschl, T., 2007. A mammalian 2104 microRNA expression atlas based on small RNA library sequencing. Cell 129, 2105 1401–1414. 2106 Latta, C.H., Sudduth, T.L., Weekman, E.M., Brothers, H.M., Abner, E.L., Popa, G.J., 2107 Mendenhall, M.D., Gonzalez-Oregon, F., Braun, K., Wilcock, D.M., 2015. Deter2108 mining the role of IL-4 induced neuroinflammation in microglial activity and 2109 amyloid-beta using BV2 microglial cells and APP/PS1 transgenic mice. 2110 J. Neuroinflammation 12, 41. 2111 Lawson, L.J., Perry, V.H., Dri, P., Gordon, S., 1990. Heterogeneity in the distribution 2112 and morphology of microglia in the normal adult mouse brain. Neuroscience 39, 2113 151–170. 2114 Le, W., Rowe, D., Xie, W., Ortiz, I., He, Y., Appel, S.H., 2001. Microglial activation and 2115 dopaminergic cell injury: an in vitro model relevant to Parkinson’s disease. 2116 J. Neurosci. 21, 8447–8455. 2117 Lee, Y., Lee, S.R., Choi, S.S., Yeo, H.G., 2014. Therapeutically targeting neuroinflam2118 mation and microglia after acute ischemic stroke. 2014, 297241. 2119 Lehnardt, S., Lehmann, S., Kaul, D., Tschimmel, K., Hoffmann, O., Cho, S., Krueger, C., 2120 Nitsch, R., Meisel, A., Weber, J.R., 2007. Toll-like receptor 2 mediates CNS injury 2121 in focal cerebral ischemia. J. Neuroimmunol. 190, 28–33. 2122 Lehnardt, S., Massillon, L., Follett, P., Jensen, F.E., Ratan, R., Rosenberg, P.A., Volpe, J.J., 2123 Vartanian, T., 2003. Activation of innate immunity in the CNS triggers neuro2124 degeneration through a Toll-like receptor 4-dependent pathway. Proc. Natl. 2125 Acad. Sci. U.S.A. 100, 8514–8519. 2126 Li, F., Wang, L., Li, J.W., Gong, M., He, L., Feng, R., Dai, Z., Li, S.Q., 2011. Hypoxia 2127 induced amoeboid microglial cell activation in postnatal rat brain is mediated 2128 by ATP receptor P2X4. BMC Neurosci. 12, 111.
Li, L., Harms, K.M., Ventura, P.B., Lagace, D.C., Eisch, A.J., Cunningham, L.A., 2010. Focal cerebral ischemia induces a multilineage cytogenic response from adult subventricular zone that is predominantly gliogenic. Glia 58, 1610–1619. Li, Z., Li, K., Zhu, L., Kan, Q., Yan, Y., Kumar, P., Xu, H., Rostami, A., Zhang, G.X., 2013. Inhibitory effect of IL-17 on neural stem cell proliferation and neural cell differentiation. BMC Immunol. 14, 20. Liang, K.J., Lee, J.E., Wang, Y.D., Ma, W., Fontainhas, A.M., Fariss, R.N., Wong, W.T., 2009. Regulation of dynamic behavior of retinal microglia by CX3CR1 signaling. Invest. Ophthalmol. Vis. Sci. 50, 4444–4451. Liao, B., Zhao, W., Beers, D.R., Henkel, J.S., Appel, S.H., 2012. Transformation from a neuroprotective to a neurotoxic microglial phenotype in a mouse model of ALS. Exp. Neurol. 237, 147–152. Lichanska, A.M., Hume, D.A., 2000. Origins and functions of phagocytes in the embryo. Exp. Hematol. 28, 601–611. Liu, D.Z., Tian, Y., Ander, B.P., Xu, H., Stamova, B.S., Zhan, X., Turner, R.J., Jickling, G., Sharp, F.R., 2010. Brain and blood microRNA expression profiling of ischemic stroke, intracerebral hemorrhage, and kainate seizures. J. Cereb. Blood Flow Metab. 30, 92–101. Liu, G., Guo, J., Liu, J., Wang, Z., Liang, D., 2014a. Toll-like receptor signaling directly increases functional IL-17RA expression in neuroglial cells. Clin. Immunol. 154, 127–140. Liu, X., Li, F., Zhao, S., Luo, Y., Kang, J., Zhao, H., Yan, F., Li, S., Ji, X., 2013. MicroRNA124-mediated regulation of inhibitory member of apoptosis-stimulating protein of p53 family in experimental stroke. Stroke 44, 1973–1980. Liu, X., Ye, R., Yan, T., Yu, S.P., Wei, L., Xu, G., Fan, X., Jiang, Y., Stetler, R.A., Liu, G., Chen, J., 2014b. Cell based therapies for ischemic stroke: from basic science to bedside. Prog. Neurobiol. 115, 92–115. Liu, Z., Fan, Y., Won, S.J., Neumann, M., Hu, D., Zhou, L., Weinstein, P.R., Liu, J., 2007. Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke 38, 146–152. Lue, L.F., Walker, D.G., Brachova, L., Beach, T.G., Rogers, J., Schmidt, A.M., Stern, D.M., Yan, S.D., 2001. Involvement of microglial receptor for advanced glycation endproducts (RAGE) in Alzheimer’s disease: identification of a cellular activation mechanism. Exp. Neurol. 171, 29–45. Lv, M., Liu, Y., Zhang, J., Sun, L., Liu, Z., Zhang, S., Wang, B., Su, D., Su, Z., 2011. Roles of inflammation response in microglia cell through Toll-like receptors 2/interleukin-23/interleukin-17 pathway in cerebral ischemia/reperfusion injury. Neuroscience 176, 162–172. Lynch, M.A., 2009. The multifaceted profile of activated microglia. Mol. Neurobiol. 40, 139–156. Machado, L.S., Sazonova, I.Y., Kozak, A., Wiley, D.C., El-Remessy, A.B., Ergul, A., Hess, D.C., Waller, J.L., Fagan, S.C., 2009. Minocycline and tissue-type plasminogen activator for stroke: assessment of interaction potential. Stroke 40, 3028–3033. MacMicking, J., Xie, Q.W., Nathan, C., 1997. Nitric oxide and macrophage function. Annu. Rev. Immunol. 15, 323–350. Madinier, A., Bertrand, N., Mossiat, C., Prigent-Tessier, A., Beley, A., Marie, C., Garnier, P., 2009. Microglial involvement in neuroplastic changes following focal brain ischemia in rats. PLoS ONE 4, e8101. Maiorino, C., Khorooshi, R., Ruffini, F., Lobner, M., Bergami, A., Garzetti, L., Martino, G., Owens, T., Furlan, R., 2013. Lentiviral-mediated administration of IL-25 in the CNS induces alternative activation of microglia. Gene Ther. 20, 487–496. Mantovani, A., Sica, A., Sozzani, S., Allavena, P., Vecchi, A., Locati, M., 2004. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 25, 677–686. Martino, G., Franklin, R.J., Baron Van Evercooren, A., Kerr, D.A., 2010. Stem cell transplantation in multiple sclerosis: current status and future prospects. Nat. Rev. Neurol. 6, 247–255. Masumura, M., Hata, R., Nagai, Y., Sawada, T., 2001. Oligodendroglial cell death with DNA fragmentation in the white matter under chronic cerebral hypoperfusion: comparison between normotensive and spontaneously hypertensive rats. Neurosci. Res. 39, 401–412. Matute, C., Domercq, M., Perez-Samartin, A., Ransom, B.R., 2013. Protecting white matter from stroke injury. Stroke 44, 1204–1211. McCann, S.K., Dusting, G.J., Roulston, C.L., 2014. Nox2 knockout delays infarct progression and increases vascular recovery through angiogenesis in mice following ischaemic stroke with reperfusion. PLoS ONE 9, e110602. McKercher, S.R., Torbett, B.E., Anderson, K.L., Henkel, G.W., Vestal, D.J., Baribault, H., Klemsz, M., Feeney, A.J., Wu, G.E., Paige, C.J., Maki, R.A., 1996. Targeted disruption of the PU.1 gene results in multiple hematopoietic abnormalities. EMBO J. 15, 5647–5658. Mecha, M., Feliu, A., Carrillo-Salinas, F.J., Rueda-Zubiaurre, A., Ortega-Gutierrez, S., de Sola, R.G., Guaza, C., 2015. Endocannabinoids drive the acquisition of an alternative phenotype in microglia. Brain Behav. Immun. 49, 233–245. Menini, T., Ikeda, H., Kimura, S., Gugliucci, A., 2014. Circulating soluble RAGE increase after a cerebrovascular event. Clin. Chem. Lab. Med. 52, 109–116. Mildner, A., Schmidt, H., Nitsche, M., Merkler, D., Hanisch, U.K., Mack, M., Heikenwalder, M., Bruck, W., Priller, J., Prinz, M., 2007. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat. Neurosci. 10, 1544–1553. Miron, V.E., Boyd, A., Zhao, J.W., Yuen, T.J., Ruckh, J.M., Shadrach, J.L., van Wijngaarden, P., Wagers, A.J., Williams, A., Franklin, R.J., ffrench-Constant, C., 2013. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat. Neurosci. 16, 1211–1218. Mittelbronn, M., Dietz, K., Schluesener, H.J., Meyermann, R., 2001. Local distribution of microglia in the normal adult human central nervous system differs by up to one order of magnitude. Acta Neuropathol. 101, 249–255.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
2129 2130 2131 2132 2133 2134 2135 2136 2137 2138 2139 2140 2141 2142 2143 2144 2145 2146 2147 2148 2149 2150 2151 2152 2153 2154 2155 2156 2157 2158 2159 2160 2161 2162 2163 2164 2165 2166 2167 2168 2169 2170 2171 2172 2173 2174 2175 2176 2177 2178 2179 2180 2181 2182 2183 2184 2185 2186 2187 2188 2189 2190 2191 2192 2193 2194 2195 2196 2197 2198 2199 2200 2201 2202 2203 2204 2205 2206 2207 2208 2209 2210 2211 2212 2213 2214
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
2215 2216 2217 2218 2219 2220 2221 2222 2223 2224 2225 2226 2227 2228 2229 2230 2231 2232 2233 2234 2235 2236 2237 2238 2239 2240 2241 2242 2243 2244 2245 2246 2247 2248 2249 2250 2251 2252 2253 2254 2255 2256 2257 2258 2259 2260 2261 2262 2263 2264 2265 2266 2267 2268 2269 2270 2271 2272 2273 2274 2275 2276 2277 2278 2279 2280 2281 2282 2283 2284 2285 2286 2287 2288 2289 2290 2291 2292 2293 2294 2295 2296 2297 2298 2299 2300
Moore, C.S., Rao, V.T., Durafourt, B.A., Bedell, B.J., Ludwin, S.K., Bar-Or, A., Antel, J.P., 2013. miR-155 as a multiple sclerosis-relevant regulator of myeloid cell polarization. Ann. Neurol. 74, 709–720. Morioka, T., Kalehua, A.N., Streit, W.J., 1993. Characterization of microglial reaction after middle cerebral artery occlusion in rat brain. J. Comp. Neurol. 327, 123– 132. Morrison, H.W., Filosa, J.A., 2013. A quantitative spatiotemporal analysis of microglia morphology during ischemic stroke and reperfusion. J. Neuroinflammation 10, 4. Mosher, K.I., Andres, R.H., Fukuhara, T., Bieri, G., Hasegawa-Moriyama, M., He, Y., Guzman, R., Wyss-Coray, T., 2012. Neural progenitor cells regulate microglia functions and activity. Nat. Neurosci. 15, 1485–1487. Moskowitz, M.A., Lo, E.H., Iadecola, C., 2010. The science of stroke: mechanisms in search of treatments. Neuron 67, 181–198. Moxon-Emre, I., Schlichter, L.C., 2010. Evolution of inflammation and white matter injury in a model of transient focal ischemia. J. Neuropathol. Exp. Neurol. 69, 1– 15. Mucenski, M.L., McLain, K., Kier, A.B., Swerdlow, S.H., Schreiner, C.M., Miller, T.A., Pietryga, D.W., Scott Jr., W.J., Potter, S.S., 1991. A functional c-myb gene is required for normal murine fetal hepatic hematopoiesis. Cell 65, 677–689. Muhammad, S., Barakat, W., Stoyanov, S., Murikinati, S., Yang, H., Tracey, K.J., Bendszus, M., Rossetti, G., Nawroth, P.P., Bierhaus, A., Schwaninger, M., 2008. The HMGB1 receptor RAGE mediates ischemic brain damage. J. Neurosci. 28, 12023–12031. Nakagawa, Y., Chiba, K., 2014. Role of microglial m1/m2 polarization in relapse and remission of psychiatric disorders and diseases. Pharmaceuticals (Basel) 7, 1028–1048. Nakajima, K., Kohsaka, S., 2004. Microglia: neuroprotective and neurotrophic cells in the central nervous system. Curr. Drug Targets Cardiovasc. Haematol. Disord. 4, 65–84. Nakashima, M.N., Yamashita, K., Kataoka, Y., Yamashita, Y.S., Niwa, M., 1995. Time course of nitric oxide synthase activity in neuronal, glial, and endothelial cells of rat striatum following focal cerebral ischemia. Cell. Mol. Neurobiol. 15, 341– 349. Narantuya, D., Nagai, A., Sheikh, A.M., Masuda, J., Kobayashi, S., Yamaguchi, S., Kim, S.U., 2010a. Human microglia transplanted in rat focal ischemia brain induce neuroprotection and behavioral improvement. PLoS ONE 5, e11746. Narantuya, D., Nagai, A., Sheikh, A.M., Wakabayashi, K., Shiota, Y., Watanabe, T., Masuda, J., Kobayashi, S., Kim, S.U., Yamaguchi, S., 2010b. Microglia transplantation attenuates white matter injury in rat chronic ischemia model via matrix metalloproteinase-2 inhibition. Brain Res. 1316, 145–152. Nath, N., Khan, M., Paintlia, M.K., Singh, I., Hoda, M.N., Giri, S., 2009. Metformin attenuated the autoimmune disease of the central nervous system in animal models of multiple sclerosis. J. Immunol. 182, 8005–8014. Nayak, D., Roth, T.L., McGavern, D.B., 2014. Microglia development and function. Annu. Rev. Immunol. 32, 367–402. Nedergaard, M., Dirnagl, U., 2005. Role of glial cells in cerebral ischemia. Glia 50, 281–286. Neher, J.J., Emmrich, J.V., Fricker, M., Mander, P.K., Thery, C., Brown, G.C., 2013. Phagocytosis executes delayed neuronal death after focal brain ischemia. Proc. Natl. Acad. Sci. U.S.A. 110, E4098–E4107. Neher, J.J., Neniskyte, U., Zhao, J.W., Bal-Price, A., Tolkovsky, A.M., Brown, G.C., 2011. Inhibition of microglial phagocytosis is sufficient to prevent inflammatory neuronal death. J. Immunol. 186, 4973–4983. Neubrand, V.E., Pedreno, M., Caro, M., Forte-Lago, I., Delgado, M., Gonzalez-Rey, E., 2014. Mesenchymal stem cells induce the ramification of microglia via the small RhoGTPases Cdc42 and Rac1. Glia 62, 1932–1942. Neumann, H., Kotter, M.R., Franklin, R.J., 2009. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain 132, 288–295. Neumann, J., Gunzer, M., Gutzeit, H.O., Ullrich, O., Reymann, K.G., Dinkel, K., 2006. Microglia provide neuroprotection after ischemia. FASEB J. 20, 714–716. Neumann, J., Sauerzweig, S., Ronicke, R., Gunzer, F., Dinkel, K., Ullrich, O., Gunzer, M., Reymann, K.G., 2008. Microglia cells protect neurons by direct engulfment of invading neutrophil granulocytes: a new mechanism of CNS immune privilege. J. Neurosci. 28, 5965–5975. Ni, J., Wang, X., Chen, S., Liu, H., Wang, Y., Xu, X., Cheng, J., Jia, J., Zhen, X., 2015. MicroRNA let-7c-5p protects against cerebral ischemia injury via mechanisms involving the inhibition of microglia activation. Brain Behav. Immun.. Nimmerjahn, A., Kirchhoff, F., Helmchen, F., 2005. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 308, 1314–1318. Noda, M., Doi, Y., Liang, J., Kawanokuchi, J., Sonobe, Y., Takeuchi, H., Mizuno, T., Suzumura, A., 2011. Fractalkine attenuates excito-neurotoxicity via microglial clearance of damaged neurons and antioxidant enzyme heme oxygenase-1 expression. J. Biol. Chem. 286, 2308–2319. Norden, D.M., Godbout, J.P., 2013. Review: microglia of the aged brain: primed to be activated and resistant to regulation. Neuropathol. Appl. Neurobiol. 39, 19–34. Nunan, R., Sivasathiaseelan, H., Khan, D., Zaben, M., Gray, W., 2014. Microglial VPAC1R mediates a novel mechanism of neuroimmune-modulation of hippocampal precursor cells via IL-4 release. Glia 62, 1313–1327. O’Neill, L.A., Hardie, D.G., 2013. Metabolism of inflammation limited by AMPK and pseudo-starvation. Nature 493, 346–355. Obermeier, B., Daneman, R., Ransohoff, R.M., 2013. Development, maintenance and disruption of the blood–brain barrier. Nat. Med. 19, 1584–1596. Okuno, T., Nakatsuji, Y., Moriya, M., Takamatsu, H., Nojima, S., Takegahara, N., Toyofuku, T., Nakagawa, Y., Kang, S., Friedel, R.H., Sakoda, S., Kikutani, H., Kumanogoh, A., 2010. Roles of Sema4D-plexin-B1 interactions in the central
23
nervous system for pathogenesis of experimental autoimmune encephalomyelitis. J. Immunol. 184, 1499–1506. Orkin, S.H., Zon, L.I., 2008. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644. Pantoni, L., Garcia, J.H., Gutierrez, J.A., 1996. Cerebral white matter is highly vulnerable to ischemia. Stroke 27, 1641–1646, discussion 1647. Parisi, C., Arisi, I., D’Ambrosi, N., Storti, A.E., Brandi, R., D’Onofrio, M., Volonte, C., 2013. Dysregulated microRNAs in amyotrophic lateral sclerosis microglia modulate genes linked to neuroinflammation. Cell Death Dis. 4, e959. Pascual, O., Ben Achour, S., Rostaing, P., Triller, A., Bessis, A., 2012. Microglia activation triggers astrocyte-mediated modulation of excitatory neurotransmission. Proc. Natl. Acad. Sci. U.S.A. 109, E197–E205. Perego, C., Fumagalli, S., De Simoni, M.G., 2011. Temporal pattern of expression and colocalization of microglia/macrophage phenotype markers following brain ischemic injury in mice. J. Neuroinflammation 8, 174. Perez-de Puig, I., Miro, F., Salas-Perdomo, A., Bonfill-Teixidor, E., Ferrer-Ferrer, M., Marquez-Kisinousky, L., Planas, A.M., 2013. IL-10 deficiency exacerbates the brain inflammatory response to permanent ischemia without preventing resolution of the lesion. J. Cereb. Blood Flow Metab. 33, 1955–1966. Pocock, J.M., Kettenmann, H., 2007. Neurotransmitter receptors on microglia. Trends Neurosci. 30, 527–535. Ponomarev, E.D., Maresz, K., Tan, Y., Dittel, B.N., 2007. CNS-derived interleukin-4 is essential for the regulation of autoimmune inflammation and induces a state of alternative activation in microglial cells. J. Neurosci. 27, 10714–10721. Ponomarev, E.D., Shriver, L.P., Maresz, K., Dittel, B.N., 2005. Microglial cell activation and proliferation precedes the onset of CNS autoimmunity. J. Neurosci. Res. 81, 374–389. Ponomarev, E.D., Veremeyko, T., Barteneva, N., Krichevsky, A.M., Weiner, H.L., 2011. MicroRNA-124 promotes microglia quiescence and suppresses EAE by deactivating macrophages via the C/EBP-alpha-PU.1 pathway. Nat. Med. 17, 64–70. Ponomarev, E.D., Veremeyko, T., Weiner, H.L., 2013. MicroRNAs are universal regulators of differentiation, activation, and polarization of microglia and macrophages in normal and diseased CNS. Glia 61, 91–103. Pradillo, J.M., Fernandez-Lopez, D., Garcia-Yebenes, I., Sobrado, M., Hurtado, O., Moro, M.A., Lizasoain, I., 2009. Toll-like receptor 4 is involved in neuroprotection afforded by ischemic preconditioning. J. Neurochem. 109, 287–294. Prajeeth, C.K., Lohr, K., Floess, S., Zimmermann, J., Ulrich, R., Gudi, V., Beineke, A., Baumgartner, W., Muller, M., Huehn, J., Stangel, M., 2014. Effector molecules released by Th1 but not Th17 cells drive an M1 response in microglia. Brain Behav. Immun. 37, 248–259. Price, C.J., Wang, D., Menon, D.K., Guadagno, J.V., Cleij, M., Fryer, T., Aigbirhio, F., Baron, J.C., Warburton, E.A., 2006. Intrinsic activated microglia map to the periinfarct zone in the subacute phase of ischemic stroke. Stroke 37, 1749–1753. Prinz, M., Priller, J., 2014. Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat. Rev. Neurosci. 15, 300–312. Prinz, M., Priller, J., Sisodia, S.S., Ransohoff, R.M., 2011. Heterogeneity of CNS myeloid cells and their roles in neurodegeneration. Nat. Neurosci. 14, 1227– 1235. Ramasamy, R., Yan, S.F., Schmidt, A.M., 2009. RAGE: therapeutic target and biomarker of the inflammatory response–the evidence mounts. J. Leukoc. Biol. 86, 505–512. Ransohoff, R.M., Brown, M.A., 2012. Innate immunity in the central nervous system. J. Clin. Invest. 122, 1164–1171. Ransohoff, R.M., Perry, V.H., 2009. Microglial physiology: unique stimuli, specialized responses. Annu. Rev. Immunol. 27, 119–145. Ravetch, J.V., 1994. Fc receptors: rubor redux. Cell 78, 553–560. Ribot, E., Bouzier-Sore, A.K., Bouchaud, V., Miraux, S., Delville, M.H., Franconi, J.M., Voisin, P., 2007. Microglia used as vehicles for both inducible thymidine kinase gene therapy and MRI contrast agents for glioma therapy. Cancer Gene Ther. 14, 724–737. Rivera-Oliver, M., Diaz-Rios, M., 2014. Using caffeine and other adenosine receptor antagonists and agonists as therapeutic tools against neurodegenerative diseases: a review. Life Sci. 101, 1–9. Rosenberger, K., Derkow, K., Dembny, P., Kruger, C., Schott, E., Lehnardt, S., 2014. The impact of single and pairwise Toll-like receptor activation on neuroinflammation and neurodegeneration. J. Neuroinflammation 11, 166. Rosito, M., Lauro, C., Chece, G., Porzia, A., Monaco, L., Mainiero, F., Catalano, M., Limatola, C., Trettel, F., 2014. Trasmembrane chemokines CX3CL1 and CXCL16 drive interplay between neurons, microglia and astrocytes to counteract pMCAO and excitotoxic neuronal death. Front. Cell. Neurosci. 8, 193. Rouach, N., Avignone, E., Meme, W., Koulakoff, A., Venance, L., Blomstrand, F., Giaume, C., 2002a. Gap junctions and connexin expression in the normal and pathological central nervous system. Biol Cell 94, 457–475. Rouach, N., Calvo, C.F., Glowinski, J., Giaume, C., 2002b. Brain macrophages inhibit gap junctional communication and downregulate connexin 43 expression in cultured astrocytes. Eur J. Neurosci. 15, 403–407. Roy, S., Zhang, B., Lee, V.M., Trojanowski, J.Q., 2005. Axonal transport defects: a common theme in neurodegenerative diseases. Acta Neuropathol. 109, 5–13. Sahni, V., Kessler, J.A., 2010. Stem cell therapies for spinal cord injury. Nat. Rev. Neurol. 6, 363–372. Saijo, K., Glass, C.K., 2011. Microglial cell origin and phenotypes in health and disease. Nat. Rev. Immunol. 11, 775–787. Samokhvalov, I.M., Samokhvalova, N.I., Nishikawa, S., 2007. Cell tracing shows the contribution of the yolk sac to adult haematopoiesis. Nature 446, 1056–1061. Savarin-Vuaillat, C., Ransohoff, R.M., 2007. Chemokines and chemokine receptors in neurological disease: raise, retain, or reduce? Neurotherapeutics 4, 590–601.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
2301 2302 2303 2304 2305 2306 2307 2308 2309 2310 2311 2312 2313 2314 2315 2316 2317 2318 2319 2320 2321 2322 2323 2324 2325 2326 2327 2328 2329 2330 2331 2332 2333 2334 2335 2336 2337 2338 2339 2340 2341 2342 2343 2344 2345 2346 2347 2348 2349 2350 2351 2352 2353 2354 2355 2356 2357 2358 2359 2360 2361 2362 2363 2364 2365 2366 2367 2368 2369 2370 2371 2372 2373 2374 2375 2376 2377 2378 2379 2380 2381 2382 2383 2384 2385 2386
G Model
PRONEU 1417 1–25 24
2387 2388 2389 2390 2391 2392 2393 2394 2395 2396 2397 2398 2399 2400 2401 2402 2403 2404 2405 2406 2407 2408 2409 2410 2411 2412 2413 2414 2415 2416 2417 2418 2419 2420 2421 2422 2423 2424 2425 2426 2427 2428 2429 2430 2431 2432 2433 2434 2435 2436 2437 2438 2439 2440 2441 2442 2443 2444 2445 2446 2447 2448 2449 2450 2451 2452 2453 2454 2455 2456 2457 2458 2459 2460 2461 2462 2463 2464 2465 2466 2467 2468 2469 2470 2471 2472
Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
Sawano, T., Watanabe, F., Ishiguchi, M., Doe, N., Furuyama, T., Inagaki, S., 2015. Effect of Sema4D on microglial function in middle cerebral artery occlusion mice. Glia. Schilling, M., Besselmann, M., Leonhard, C., Mueller, M., Ringelstein, E.B., Kiefer, R., 2003. Microglial activation precedes and predominates over macrophage infiltration in transient focal cerebral ischemia: a study in green fluorescent protein transgenic bone marrow chimeric mice. Exp. Neurol. 183, 25–33. Schilling, M., Strecker, J.K., Ringelstein, E.B., Schabitz, W.R., Kiefer, R., 2009. The role of CC chemokine receptor 2 on microglia activation and blood-borne cell recruitment after transient focal cerebral ischemia in mice. Brain Res. 1289, 79–84. Schmid, C.D., Sautkulis, L.N., Danielson, P.E., Cooper, J., Hasel, K.W., Hilbush, B.S., Sutcliffe, J.G., Carson, M.J., 2002. Heterogeneous expression of the triggering receptor expressed on myeloid cells-2 on adult murine microglia. J. Neurochem. 83, 1309–1320. Schmitz, T., Krabbe, G., Weikert, G., Scheuer, T., Matheus, F., Wang, Y., Mueller, S., Kettenmann, H., Matyash, V., Buhrer, C., Endesfelder, S., 2014. Minocycline protects the immature white matter against hyperoxia. Exp. Neurol. 254, 153–165. Schroeter, M., Jander, S., Huitinga, I., Witte, O.W., Stoll, G., 1997. Phagocytic response in photochemically induced infarction of rat cerebral cortex. The role of resident microglia. Stroke 28, 382–386. Schulz, C., Gomez Perdiguero, E., Chorro, L., Szabo-Rogers, H., Cagnard, N., Kierdorf, K., Prinz, M., Wu, B., Jacobsen, S.E., Pollard, J.W., Frampton, J., Liu, K.J., Geissmann, F., 2012. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86–90. Sedgwick, J.D., Ford, A.L., Foulcher, E., Airriess, R., 1998. Central nervous system microglial cell activation and proliferation follows direct interaction with tissue-infiltrating T cell blasts. J. Immunol. 160, 5320–5330. Seifert, G., Schilling, K., Steinhauser, C., 2006. Astrocyte dysfunction in neurological disorders: a molecular perspective. Nat. Rev. Neurosci. 7, 194–206. Sheikh, A.M., Nagai, A., Wakabayashi, K., Narantuya, D., Kobayashi, S., Yamaguchi, S., Kim, S.U., 2011. Mesenchymal stem cell transplantation modulates neuroinflammation in focal cerebral ischemia: contribution of fractalkine and IL-5. Neurobiol. Dis. 41, 717–724. Shi, C., Pamer, E.G., 2011. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 11, 762–774. Shi, Q.J., Wang, H., Liu, Z.X., Fang, S.H., Song, X.M., Lu, Y.B., Zhang, W.P., Sa, X.Y., Ying, H.Z., Wei, E.Q., 2015. HAMI 3379, a CysLT2R antagonist, dose- and timedependently attenuates brain injury and inhibits microglial inflammation after focal cerebral ischemia in rats. Neuroscience 291, 53–69. Shi, Q.J., Xiao, L., Zhao, B., Zhang, X.Y., Wang, X.R., Xu, D.M., Yu, S.Y., Fang, S.H., Lu, Y.B., Zhang, W.P., Sa, X.Y., Wei, E.Q., 2012. Intracerebroventricular injection of HAMI 3379, a selective cysteinyl leukotriene receptor 2 antagonist, protects against acute brain injury after focal cerebral ischemia in rats. Brain Res. 1484, 57–67. Shigemoto-Mogami, Y., Hoshikawa, K., Goldman, J.E., Sekino, Y., Sato, K., 2014. Microglia enhance neurogenesis and oligodendrogenesis in the early postnatal subventricular zone. J. Neurosci. 34, 2231–2243. Shinozaki, Y., Nomura, M., Iwatsuki, K., Moriyama, Y., Gachet, C., Koizumi, S., 2014. Microglia trigger astrocyte-mediated neuroprotection via purinergic gliotransmission. Sci. Rep. 4, 4329. Shiratori, M., Tozaki-Saitoh, H., Yoshitake, M., Tsuda, M., Inoue, K., 2010. P2X7 receptor activation induces CXCL2 production in microglia through NFAT and PKC/MAPK pathways. J. Neurochem. 114, 810–819. Shors, T.J., Miesegaes, G., Beylin, A., Zhao, M., Rydel, T., Gould, E., 2001. Neurogenesis in the adult is involved in the formation of trace memories. Nature 410, 372– 376. Sierra, A., Abiega, O., Shahraz, A., Neumann, H., 2013. Janus-faced microglia: beneficial and detrimental consequences of microglial phagocytosis. Front. Cell. Neurosci. 7, 6. Sierra, A., Encinas, J.M., Deudero, J.J., Chancey, J.H., Enikolopov, G., OverstreetWadiche, L.S., Tsirka, S.E., Maletic-Savatic, M., 2010. Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem Cell 7, 483–495. Song, J., Oh, Y., Lee, J.E., 2015. miR-Let7A modulates autophagy induction in LPSactivated microglia. Exp. Neurobiol. 24, 117–125. Sonobe, Y., Liang, J., Jin, S., Zhang, G., Takeuchi, H., Mizuno, T., Suzumura, A., 2008. Microglia express a functional receptor for interleukin-23. Biochem. Biophys. Res. Commun. 370, 129–133. Soriano, S.G., Amaravadi, L.S., Wang, Y.F., Zhou, H., Yu, G.X., Tonra, J.R., FairchildHuntress, V., Fang, Q., Dunmore, J.H., Huszar, D., Pan, Y., 2002. Mice deficient in fractalkine are less susceptible to cerebral ischemia-reperfusion injury. J. Neuroimmunol. 125, 59–65. Stoll, G., Jander, S., Schroeter, M., 1998. Inflammation and glial responses in ischemic brain lesions. Prog. Neurobiol. 56, 149–171. Stoll, G., Schroeter, M., Jander, S., Siebert, H., Wollrath, A., Kleinschnitz, C., Bruck, W., 2004. Lesion-associated expression of transforming growth factor-beta-2 in the rat nervous system: evidence for down-regulating the phagocytic activity of microglia and macrophages. Brain Pathol. 14, 51–58. Stubbe, T., Ebner, F., Richter, D., Engel, O., Klehmet, J., Royl, G., Meisel, A., Nitsch, R., Meisel, C., Brandt, C., 2013. Regulatory T cells accumulate and proliferate in the ischemic hemisphere for up to 30 days after MCAO. J. Cereb. Blood Flow Metab. 33, 37–47. Su, W., Aloi, M.S., Garden, G.A., 2015. MicroRNAs mediating CNS inflammation: Small regulators with powerful potential. Brain Behav. Immun.. Su, W., Hopkins, S., Nesser, N.K., Sopher, B., Silvestroni, A., Ammanuel, S., Jayadev, S., Moller, T., Weinstein, J., Garden, G.A., 2014. The p53 transcription factor
modulates microglia behavior through microRNA-dependent regulation of cMaf. J. Immunol. 192, 358–366. Sudduth, T.L., Schmitt, F.A., Nelson, P.T., Wilcock, D.M., 2013. Neuroinflammatory phenotype in early Alzheimer’s disease. Neurobiol. Aging 34, 1051–1059. Sugimoto, K., Nishioka, R., Ikeda, A., Mise, A., Takahashi, H., Yano, H., Kumon, Y., Ohnishi, T., Tanaka, J., 2014. Activated microglia in a rat stroke model express NG2 proteoglycan in peri-infarct tissue through the involvement of TGF-beta1. Glia 62, 185–198. Sun, Y., Gui, H., Li, Q., Luo, Z.M., Zheng, M.J., Duan, J.L., Liu, X., 2013. MicroRNA-124 protects neurons against apoptosis in cerebral ischemic stroke. CNS Neurosci. Ther. 19, 813–819. Svahn, A.J., Giacomotto, J., Graeber, M.B., Rinkwitz, S., Becker, T.S., 2015. miR-124 Contributes to the functional maturity of microglia. Dev. Neurobiol.. Sylvestre, D.L., Ravetch, J.V., 1994. Fc receptors initiate the Arthus reaction: redefining the inflammatory cascade. Science 265, 1095–1098. Takahashi, K., Rochford, C.D., Neumann, H., 2005. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. J. Exp. Med. 201, 647–657. Takata, K., Kitamura, Y., Yanagisawa, D., Morikawa, S., Morita, M., Inubushi, T., Tsuchiya, D., Chishiro, S., Saeki, M., Taniguchi, T., Shimohama, S., Tooyama, I., 2007. Microglial transplantation increases amyloid-beta clearance in Alzheimer model rats. FEBS Lett. 581, 475–478. Tang, G., Liu, Y., Zhang, Z., Lu, Y., Wang, Y., Huang, J., Li, Y., Chen, X., Gu, X., Wang, Y., Yang, G.Y., 2014. Mesenchymal stem cells maintain blood–brain barrier integrity by inhibiting aquaporin-4 upregulation after cerebral ischemia. Stem Cells 32, 3150–3162. Tang, S.C., Arumugam, T.V., Xu, X., Cheng, A., Mughal, M.R., Jo, D.G., Lathia, J.D., Siler, D.A., Chigurupati, S., Ouyang, X., Magnus, T., Camandola, S., Mattson, M.P., 2007. Pivotal role for neuronal Toll-like receptors in ischemic brain injury and functional deficits. Proc. Natl. Acad. Sci. U.S.A. 104, 13798–13803. Taniguchi, Y., Amazaki, M., Furuyama, T., Yamaguchi, W., Takahara, M., Saino, O., Wada, T., Niwa, H., Tashiro, F., Miyazaki, J., Kogo, M., Matsuyama, T., Inagaki, S., 2009. Sema4D deficiency results in an increase in the number of oligodendrocytes in healthy and injured mouse brains. J. Neurosci. Res. 87, 2833–2841. Thiel, A., Heiss, W.D., 2011. Imaging of microglia activation in stroke. Stroke 42, 507–512. Thiel, A., Radlinska, B.A., Paquette, C., Sidel, M., Soucy, J.P., Schirrmacher, R., Minuk, J., 2010. The temporal dynamics of poststroke neuroinflammation: a longitudinal diffusion tensor imaging-guided PET study with 11C-PK11195 in acute subcortical stroke. J. Nucl. Med. 51, 1404–1412. Thored, P., Heldmann, U., Gomes-Leal, W., Gisler, R., Darsalia, V., Taneera, J., Nygren, J.M., Jacobsen, S.E., Ekdahl, C.T., Kokaia, Z., Lindvall, O., 2009. Long-term accumulation of microglia with proneurogenic phenotype concomitant with persistent neurogenesis in adult subventricular zone after stroke. Glia 57, 835–849. Tikka, T., Fiebich, B.L., Goldsteins, G., Keinanen, R., Koistinaho, J., 2001. Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J. Neurosci. 21, 2580–2588. Tobin, M.K., Bonds, J.A., Minshall, R.D., Pelligrino, D.A., Testai, F.D., Lazarov, O., 2014. Neurogenesis and inflammation after ischemic stroke: what is known and where we go from here. J. Cereb. Blood Flow Metab. 34, 1573–1584. Tomimoto, H., Akiguchi, I., Wakita, H., Kinoshita, A., Ikemoto, A., Nakamura, S., Kimura, J., 1996. Glial expression of cytokines in the brains of cerebrovascular disease patients. Acta Neuropathol. 92, 281–287. Tonchev, A.B., 2011. Brain ischemia, neurogenesis, and neurotrophic receptor expression in primates. Arch Ital Biol 149, 225–231. Tremblay, M.E., Stevens, B., Sierra, A., Wake, H., Bessis, A., Nimmerjahn, A., 2011. The role of microglia in the healthy brain. J. Neurosci. 31, 16064–16069. Trincavelli, M.L., Melani, A., Guidi, S., Cuboni, S., Cipriani, S., Pedata, F., Martini, C., 2008. Regulation of A(2A) adenosine receptor expression and functioning following permanent focal ischemia in rat brain. J. Neurochem. 104, 479–490. Tsai, H.H., Li, H., Fuentealba, L.C., Molofsky, A.V., Taveira-Marques, R., Zhuang, H., Tenney, A., Murnen, A.T., Fancy, S.P., Merkle, F., Kessaris, N., Alvarez-Buylla, A., Richardson, W.D., Rowitch, D.H., 2012. Regional astrocyte allocation regulates CNS synaptogenesis and repair. Science 337, 358–362. Tsai, Y.W., Yang, Y.R., Wang, P.S., Wang, R.Y., 2011. Intermittent hypoxia after transient focal ischemia induces hippocampal neurogenesis and c-Fos expression and reverses spatial memory deficits in rats. PLoS ONE 6, e24001. Tsiperson, V., Li, X., Schwartz, G.J., Raine, C.S., Shafit-Zagardo, B., 2010. GAS6 enhances repair following cuprizone-induced demyelination. PLoS ONE 5, e15748. Tsoyi, K., Jang, H.J., Nizamutdinova, I.T., Kim, Y.M., Lee, Y.S., Kim, H.J., Seo, H.G., Lee, J.H., Chang, K.C., 2011. Metformin inhibits HMGB1 release in LPS-treated RAW 264.7 cells and increases survival rate of endotoxaemic mice. Br. J. Pharmacol. 162, 1498–1508. Tureyen, K., Vemuganti, R., Sailor, K.A., Bowen, K.K., Dempsey, R.J., 2004. Transient focal cerebral ischemia-induced neurogenesis in the dentate gyrus of the adult mouse. J. Neurosurg. 101, 799–805. van Velthoven, C.T., Kavelaars, A., van Bel, F., Heijnen, C.J., 2010. Mesenchymal stem cell treatment after neonatal hypoxic-ischemic brain injury improves behavioral outcome and induces neuronal and oligodendrocyte regeneration. Brain Behav. Immun. 24, 387–393. Varnum, M.M., Ikezu, T., 2012. The classification of microglial activation phenotypes on neurodegeneration and regeneration in Alzheimer’s disease brain. Arch. Immunol. Ther. Exp. (Warsz) 60, 251–266. Varvel, N.H., Grathwohl, S.A., Baumann, F., Liebig, C., Bosch, A., Brawek, B., Thal, D.R., Charo, I.F., Heppner, F.L., Aguzzi, A., Garaschuk, O., Ransohoff, R.M., Jucker, M.,
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
2473 2474 2475 2476 2477 2478 2479 2480 2481 2482 2483 2484 2485 2486 2487 2488 2489 2490 2491 2492 2493 2494 2495 2496 2497 2498 2499 2500 2501 2502 2503 2504 2505 2506 2507 2508 2509 2510 2511 2512 2513 2514 2515 2516 2517 2518 2519 2520 2521 2522 2523 2524 2525 2526 2527 2528 2529 2530 2531 2532 2533 2534 2535 2536 2537 2538 2539 2540 2541 2542 2543 2544 2545 2546 2547 2548 2549 2550 2551 2552 2553 2554 2555 2556 2557 2558
G Model
PRONEU 1417 1–25 Y. Ma et al. / Progress in Neurobiology xxx (2016) xxx–xxx
2559 2012. Microglial repopulation model reveals a robust homeostatic process for 2560 replacing CNS myeloid cells. Proc. Natl. Acad. Sci. U.S.A. 109, 18150–18155. 2561 Vexler, Z.S., Tang, X.N., Yenari, M.A., 2006. Inflammation in adult and neonatal 2562 stroke. Clin. Neurosci. Res. 6, 293–313. 2563 Vukovic, J., Colditz, M.J., Blackmore, D.G., Ruitenberg, M.J., Bartlett, P.F., 2012. 2564 Microglia modulate hippocampal neural precursor activity in response to 2565 exercise and aging. J. Neurosci. 32, 6435–6443. 2566 Wake, H., Moorhouse, A.J., Jinno, S., Kohsaka, S., Nabekura, J., 2009. Resting microglia 2567 directly monitor the functional state of synapses in vivo and determine the fate 2568 of ischemic terminals. J. Neurosci. 29, 3974–3980. 2569 Wake, H., Moorhouse, A.J., Miyamoto, A., Nabekura, J., 2013. Microglia: actively 2570 surveying and shaping neuronal circuit structure and function. Trends Neurosci. 2571 36, 209–217. 2572 Walter, J., Honsek, S.D., Illes, S., Wellen, J.M., Hartung, H.P., Rose, C.R., Dihne, M., 2573 2011. A new role for interferon gamma in neural stem/precursor cell dysre2574 gulation. Mol. Neurodegener 6, 18. 2575 Walton, N.M., Sutter, B.M., Laywell, E.D., Levkoff, L.H., Kearns, S.M., Marshall 2nd, 2576 G.P., Scheffler, B., Steindler, D.A., 2006. Microglia instruct subventricular zone 2577 neurogenesis. Glia 54, 815–825. 2578 Wang, D.D., Zhao, Y.F., Wang, G.Y., Sun, B., Kong, Q.F., Zhao, K., Zhang, Y., Wang, J.H., 2579 Liu, Y.M., Mu, L.L., Wang, D.S., Li, H.L., 2009. IL-17 potentiates neuronal injury 2580 induced by oxygen–glucose deprivation and affects neuronal IL-17 receptor 2581 expression. J. Neuroimmunol. 212, 17–25. 2582 Wang, G., Zhang, J., Hu, X., Zhang, L., Mao, L., Jiang, X., Liou, A.K., Leak, R.K., Gao, Y., 2583 Chen, J., 2013. Microglia/macrophage polarization dynamics in white matter 2584 after traumatic brain injury. J. Cereb. Blood Flow Metab. 33, 1864–1874. 2585 Wang, P., Hou, J., Lin, L., Wang, C., Liu, X., Li, D., Ma, F., Wang, Z., Cao, X., 2010. 2586 Inducible microRNA-155 feedback promotes type I IFN signaling in antiviral 2587 innate immunity by targeting suppressor of cytokine signaling 1. J. Immunol. 2588 185, 6226–6233. 2589 Wang, Y., Ge, P., Yang, L., Wu, C., Zha, H., Luo, T., Zhu, Y., 2014. Protection of ischemic 2590 post conditioning against transient focal ischemia-induced brain damage is 2591 associated with inhibition of neuroinflammation via modulation of TLR2 and 2592 TLR4 pathways. J. Neuroinflammation 11, 15. 2593 Webster, C.M., Hokari, M., McManus, A., Tang, X.N., Ma, H., Kacimi, R., Yenari, M.A., 2594 2013. Microglial P2Y12 deficiency/inhibition protects against brain ischemia. 2595 PLoS ONE 8, e70927. 2596 Weng, H., Shen, C., Hirokawa, G., Ji, X., Takahashi, R., Shimada, K., Kishimoto, C., Iwai, 2597 N., 2011. Plasma miR-124 as a biomarker for cerebral infarction. Biomed. Res. 2598 32, 135–141. 2599 Wieghofer, P., Knobeloch, K.P., Prinz, M., 2015. Genetic targeting of microglia. Glia 2600 63, 1–22. 2601 Wixey, J.A., Reinebrant, H.E., Carty, M.L., Buller, K.M., 2009. Delayed P2X4R expres2602 sion after hypoxia-ischemia is associated with microglia in the immature rat 2603 brain. J. Neuroimmunol. 212, 35–43. 2604 Woo, M.S., Wang, X., Faustino, J.V., Derugin, N., Wendland, M.F., Zhou, P., Iadecola, C., 2605 Vexler, Z.S., 2012. Genetic deletion of CD36 enhances injury after acute neonatal 2606 stroke. Ann. Neurol. 72, 961–970. 2607 Woodbury, M.E., Freilich, R.W., Cheng, C.J., Asai, H., Ikezu, S., Boucher, J.D., Slack, F., 2608 Ikezu, T., 2015. miR-155 Is Essential for Inflammation-Induced Hippocampal 2609 Q10 Neurogenic Dysfunction, vol. 35. , pp. 9764–9781. 2610 Wu, L.J., Wu, G., Akhavan Sharif, M.R., Baker, A., Jia, Y., Fahey, F.H., Luo, H.R., Feener, 2611 E.P., Clapham, D.E., 2012. The voltage-gated proton channel Hv1 enhances brain 2612 damage from ischemic stroke. Nat. Neurosci. 15, 565–573. 2613 Xing, C., Wang, X., Cheng, C., Montaner, J., Mandeville, E., Leung, W., van Leyen, K., 2614 Lok, J., Wang, X., Lo, E.H., 2014. Neuronal production of lipocalin-2 as a help-me 2615 signal for glial activation. Stroke 45, 2085–2092. 2616 Xu, Y., Qian, L., Zong, G., Ma, K., Zhu, X., Zhang, H., Li, N., Yang, Q., Bai, H., Ben, J., Li, X., 2617 Xu, Y., Chen, Q., 2012. Class A scavenger receptor promotes cerebral ischemic 2618 injury by pivoting microglia/macrophage polarization. Neuroscience 218, 35– 2619 48. 2620 Yamagishi, S., Matsui, T., 2010. Soluble form of a receptor for advanced glycation 2621 end products (sRAGE) as a biomarker. Front. Biosci. (Elite Ed) 2, 1184–1195. 2622 Yang, G.Y., Gong, C., Qin, Z., Ye, W., Mao, Y., Bertz, A.L., 1998. Inhibition of TNFalpha 2623 attenuates infarct volume and ICAM-1 expression in ischemic mouse brain. 2624 NeuroReport 9, 2131–2134. 2625 Yang, M., Wei, X., Li, J., Heine, L.A., Rosenwasser, R., Iacovitti, L., 2010. Changes in 2626 host blood factors and brain glia accompanying the functional recovery after 2627 systemic administration of bone marrow stem cells in ischemic stroke rats. Cell 2628 Transplant. 19, 1073–1084. 2629 Yang, Y., Rosenberg, G.A., 2011. Blood–brain barrier breakdown in acute and chronic 2630 cerebrovascular disease. Stroke 42, 3323–3328.
25
Yang, Y., Salayandia, V.M., Thompson, J.F., Yang, L.Y., Estrada, E.Y., Yang, Y., 2015. Attenuation of acute stroke injury in rat brain by minocycline promotes blood– brain barrier remodeling and alternative microglia/macrophage activation during recovery. J. Neuroinflammation 12, 26. Yang, Z.J., Wang, B., Kwansa, H., Heitmiller, K.D., Hong, G., Carter, E.L., Jamrogowicz, J.L., Larson, A.C., Martin, L.J., Koehler, R.C., 2013. Adenosine A2A receptor contributes to ischemic brain damage in newborn piglet. J. Cereb. Blood Flow Metab. 33, 1612–1620. Yao, H., Ma, R., Yang, L., Hu, G., Chen, X., Duan, M., Kook, Y., Niu, F., Liao, K., Fu, M., Hu, G., Kolattukudy, P., Buch, S., 2014. MiR-9 promotes microglial activation by targeting MCPIP1. Nat. Commun. 5, 4386. Yenari, M.A., Kauppinen, T.M., Swanson, R.A., 2010. Microglial activation in stroke: therapeutic targets. Neurotherapeutics 7, 378–391. Yenari, M.A., Xu, L., Tang, X.N., Qiao, Y., Giffard, R.G., 2006. Microglia potentiate damage to blood–brain barrier constituents: improvement by minocycline in vivo and in vitro. Stroke 37, 1087–1093. Yu, T.B., Cheng, Y.S., Zhao, P., Kou, D.W., Sun, K., Chen, B.H., Wang, A.M., 2009. Immune therapy with cultured microglia grafting into the injured spinal cord promoting the recovery of rat’s hind limb motor function. Chin. J. Traumatol. 12, 291–295. Zarruk, J.G., Fernandez-Lopez, D., Garcia-Yebenes, I., Garcia-Gutierrez, M.S., Vivancos, J., Nombela, F., Torres, M., Burguete, M.C., Manzanares, J., Lizasoain, I., Moro, M.A., 2012. Cannabinoid type 2 receptor activation downregulates strokeinduced classic and alternative brain macrophage/microglial activation concomitant to neuroprotection. Stroke 43, 211–219. Zhai, D.X., Kong, Q.F., Xu, W.S., Bai, S.S., Peng, H.S., Zhao, K., Li, G.Z., Wang, D.D., Sun, B., Wang, J.H., Wang, G.Y., Li, H.L., 2008. RAGE expression is up-regulated in human cerebral ischemia and pMCAO rats. Neurosci. Lett. 445, 117–121. Zhang, G., Xia, F., Zhang, Y., Zhang, X., Cao, Y., Wang, L., Liu, X., Zhao, G., Shi, M., 2015. Ginsenoside Rd is efficacious against acute ischemic stroke by suppressing microglial proteasome-mediated inflammation. Mol. Neurobiol.. Zhang, L., Dong, L.Y., Li, Y.J., Hong, Z., Wei, W.S., 2012a. The microRNA miR-181c controls microglia-mediated neuronal apoptosis by suppressing tumor necrosis factor. J. Neuroinflammation 9, 211. Zhang, R.L., Chopp, M., Roberts, C., Wei, M., Wang, X., Liu, X., Lu, M., Zhang, Z.G., 2012b. Sildenafil enhances neurogenesis and oligodendrogenesis in ischemic brain of middle-aged mouse. PLoS ONE 7, e48141. Zhang, R.L., Zhang, Z.G., Zhang, L., Chopp, M., 2001. Proliferation and differentiation of progenitor cells in the cortex and the subventricular zone in the adult rat after focal cerebral ischemia. Neuroscience 105, 33–41. Zhang, Z., Chopp, M., Powers, C., 1997. Temporal profile of microglial response following transient (2 h) middle cerebral artery occlusion. Brain Res. 744, 189– 198. Zhang, Z.G., Zhang, L., Jiang, Q., Chopp, M., 2002. Bone marrow-derived endothelial progenitor cells participate in cerebral neovascularization after focal cerebral ischemia in the adult mouse. Circ. Res. 90, 284–288. Zhao, C.Z., Zhao, B., Zhang, X.Y., Huang, X.Q., Shi, W.Z., Liu, H.L., Fang, S.H., Lu, Y.B., Zhang, W.P., Tang, F.D., Wei, E.Q., 2011. Cysteinyl leukotriene receptor 2 is spatiotemporally involved in neuron injury, astrocytosis and microgliosis after focal cerebral ischemia in rats. Neuroscience 189, 1–11. Zhao, H., Wang, J., Gao, L., Wang, R., Liu, X., Gao, Z., Tao, Z., Xu, C., Song, J., Ji, X., Luo, Y., 2013. MiRNA-424 protects against permanent focal cerebral ischemia injury in mice involving suppressing microglia activation. Stroke 44, 1706–1713. Zhao, X., Wang, H., Sun, G., Zhang, J., Edwards, N.J., Aronowski, J., 2015. Neuronal Interleukin-4 as a Modulator of Microglial Pathways and Ischemic Brain Damage, 35. , pp. 11281–11291. Zhou, G., Myers, R., Li, Y., Chen, Y., Shen, X., Fenyk-Melody, J., Wu, M., Ventre, J., Doebber, T., Fujii, N., Musi, N., Hirshman, M.F., Goodyear, L.J., Moller, D.E., 2001. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest. 108, 1167–1174. Zhou, W., Liesz, A., Bauer, H., Sommer, C., Lahrmann, B., Valous, N., Grabe, N., Veltkamp, R., 2013. Postischemic brain infiltration of leukocyte subpopulations differs among murine permanent and transient focal cerebral ischemia models. Brain Pathol. 23, 34–44. Zhou, X., Spittau, B., Krieglstein, K., 2012. TGFbeta signalling plays an important role in IL4-induced alternative activation of microglia. J. Neuroinflammation 9, 210. Zhu, F., Liu, J.L., Li, J.P., Xiao, F., Zhang, Z.X., Zhang, L., 2014. MicroRNA-124 (miR-124) regulates Ku70 expression and is correlated with neuronal death induced by ischemia/reperfusion. J. Mol. Neurosci. 52, 148–155.
Please cite this article in press as: Ma, Y., et al., The biphasic function of microglia in ischemic stroke. Prog. Neurobiol. (2016), http:// dx.doi.org/10.1016/j.pneurobio.2016.01.005
2631 2632 2633 2634 2635 2636 2637 2638 2639 2640 2641 2642 2643 2644 2645 2646 2647 2648 2649 2650 2651 2652 2653 2654 2655 2656 2657 2658 2659 2660 2661 2662 2663 2664 2665 2666 2667 2668 2669 2670 2671 2672 2673 2674 2675 2676 2677 2678 2679 2680 2681 2682 2683 2684 2685 2686 2687 2688 2689 2690 2691 2692 2693 2694 2695 2696 2697 2698 2699