Microglia, the resident immune cells of the mammalian central nervous system (CNS), play a pivotal role in both physiological and pathological conditions such as the restoration of CNS integrity and the progression of neurodegenerative disorders. Extensive data have been published that describe neuroinflammation by microglial activation to have detrimental consequences on the developing and mature brain. On the other hand, a properly directed and limited inflammatory response is known to be a natural healing process after an insult in several other tissues. Thus, it is not surprising that research results illustrating benefits of neuroinflammation have been emerging over the past decade. Inflammation-mediated benefits for CNS outcomes include mechanisms such as neuroprotection, mobilization of neural precursors for repair, remyelination and axonal regeneration. Here, we review data that highlight the dual aspects of microglia with a focus on the developing brain, i.e. as aggressors potentiating damage and as helpers in the recovery process following CNS damage.

Microglia are the main cellular regulators of the brain’s innate immune response to both physiological and pathological conditions. Their phenotypic diversity was recognized already in the early 20th century when Río Hortega demonstrated the variety in microglial phenotypes in the healthy and the diseased brain. Although their physiological functions are only partially understood, their central role in the pathomechanisms of several neurological disorders – including acute and chronic neurodegenerative disorders of the immature and mature central nervous system (CNS) – are well established [1,2,3,4,5,6,7,8,9]. Clinical, epidemiological and experimental studies have shown that damage to the developing brain is caused by multiple factors and mechanisms, including inflammation, excitotoxicity and oxidative stress (see our recent review [1]). Depending on the injury as well as on the activating or inhibiting and microenvironmental conditions, microglia can aggravate an injury and subsequent neurodegeneration, but they can also play an important role in the protective mechanisms underlying tissue repair and regeneration. Thus, microglia are also a prime target for future therapeutic interventions in a wide variety of CNS disease conditions [10,11]. In this review, we summarize current results (without any claim of being exhaustive) concerning the ‘yin-and-yang’ function of microglia in physiological conditions and different neurological disorders with a focus on the developing brain.

Microglia, the resident macrophages of the CNS, represent approximately 5–10% of the adult brain cell population [12]. Although first described almost a century ago, the developmental origin of microglia is still under debate [13,14,15,16,17]. Several reports have delineated the existence of various types of microglia of different origins, especially under pathological conditions [12,18]. Resident parenchymal microglia originate from pial macrophages and mesenchymal progenitors as amoeboid microglia. They invade the brain in two waves during embryonic development and early postnatal life, first through the meninges, choroid plexus and ventricle, and later through the blood vessel walls [12,15,16,17,19,20]. Ultimately, these amoeboid microglia transform into ‘surveying microglia’ and concomitantly upregulate macrophage surface markers [21,22]. In the human brain, this event occurs in the first two trimesters of pregnancy [16,17,23], and in rodents shortly before and at the time of birth [23]. Surveying microglia – originally referred to as ‘resting microglia’ – are characterized by a small cell body with fine, long and ramified (branched) processes and a sparse expression of surface molecules associated with the monocyte-macrophage lineage [24]. These surveying microglia constantly screen the CNS and can be rapidly activated by various environmental changes [21].

These early microglial cell populations are not the only ones capable of colonizing the developing CNS. Indeed, bone marrow-derived myeloid cells have recently been shown to enter the immature and mature CNS and to differentiate into microglial cells [16,25,26,27,28]. This infiltration seems to play a central role in disease modulation in the adult brain affected by neurodegeneration or neurological insults [12] and is favored but not exclusively possible when the blood-brain barrier is compromised [12,13,29]. The immunological characteristics of microglia as the main immune cells of the CNS appear to differ between two major microglia subtypes of different origin [12]: parenchymal microglia express low levels of major histocompatibility complex (MHC) class II and demonstrate poor antigen-presenting cell function [12]. In contrast, bone marrow-derived microglia express MHC II at higher levels and show better antigen-presenting cell function [12].

The physiological role of microglia in brain development and the physiological function in the mature brain are still largely uncharacterized (fig. 1). Amoeboid microglia constitute specific clusters after they have penetrated the parenchyma [30]. These specific clusters at the junctions of the internal capsule with the thalamus, with the external capsule and with the cerebral peduncle, as well as at the junctions of the cerebral peduncle with the optic tract, the medial septum, the periventricular hypothalamic area and the corpus callosum, are transient in the developing brain [30]. In the cortical layers and white matter, microglial cells migrate from the ventricular zone to the deep cortical plate by radial and tangential migration [30,31]. At this time point at about 16–22 weeks of gestation, both the expression of monocyte chemoattractant protein 1 and chemokine macrophage inflammatory protein 1α can be detected in the upper layer of the human cerebral cortex [20,32]. At 19–30 weeks of gestation, microglia proliferate and accumulate in the semioval center [20,30]. In the mature brain, differences in microglial cell density still exist between different brain regions, with high densities being found in the telencephalon, especially in myelinated regions. This difference in microglial cell localization has been suggested to be associated with a functional difference, and phenotypic heterogeneity including receptor expression patterns has been observed within one anatomical region [33]. The mechanism underlying this phenotype diversity and its consequences are still a matter of speculation.

Fig. 1

Microglia in the developing brain. The physiological role of microglia in brain development is still largely uncharacterized. During development, microglia have been reported to be capable of phagocytosis and implemented in the regulation of apoptosis, cell proliferation, neuronal differentiation and angiogenesis. They also carry a dual function as mediators of inflammation potentially aggravating or causing damage, and as helpers in the recovery process following CNS damage.

Fig. 1

Microglia in the developing brain. The physiological role of microglia in brain development is still largely uncharacterized. During development, microglia have been reported to be capable of phagocytosis and implemented in the regulation of apoptosis, cell proliferation, neuronal differentiation and angiogenesis. They also carry a dual function as mediators of inflammation potentially aggravating or causing damage, and as helpers in the recovery process following CNS damage.

Close modal

Microglial cells are capable of phagocytosis during development [34]. During this period, microglia not only have key functions such as the clearance of dying or dead cells. Several studies are emerging that demonstrate a role in developmental processes such as apoptosis, elimination of excess axons, promotion of neuroaxonal growth, axonal guidance, neuronal differentiation, regulation of embryonic cortical precursor cell development, astrocyte proliferation and angiogenesis [20,30,34,35,36,37,38,39,40,41,42,43,44,45]. Especially the role of microglia in neurogenesis is intriguing as, within the framework of CNS inflammation, the impact of this feature on the developing brain can be enormous. Several receptors have been described on microglia, indicating extensive crosstalk with CNS cells such as neurons (see below). Microglia are also capable of secreting various factors such as those that can induce cell proliferation, e.g. brain-derived neurotrophic factor, basic fibroblast growth factor and insulin-like growth factor [46,47]. Thus, microglia might be more extensively involved in higher-order brain functions than currently believed.

Microglia can be rapidly activated by various environmental changes [21]. The process of microglial activation is associated with proliferation and transformation into ‘reactive’ microglia with different response phenotypes [10,48,49,50,51,52]. During activation, microglia change from a ramified to a hyperramified phenotype and subsequently adopt an amoeboid morphology, a process which has been suggested to help microglia invade lesions [53]. Activated microglia not only change their phenotype, but also proliferate, migrate to the site of damage and secrete pro- and anti-inflammatory cytokines and chemokines, oxidative stress-inducing factors such as nitric oxide (NO) as well as growth factors [50,54]. In case of acute neuronal death, microglia can function as brain macrophages and phagocyte cell debris [54].

Microglia have the remarkable ability to recognize a wide range of signals that indicate a threat to the structural and functional integrity of the CNS through various receptors [50]. Injured neurons release adenosine-5′-triphosphate (ATP) and chemokine CXC motif ligand 10 (CXCL10), which attract microglia via activated purinoreceptors [55,56] and chemokine CXC motif receptor 3 (CXCR3), respectively [57]. Moreover, microglia may sense neuronal activity through neurotransmitter receptors present on the microglial membrane [58]. In this line, data from our laboratories and others demonstrate the presence of glutamate receptors on microglia as a link between inflammation and excitotoxic brain damage [20,58,59]. As the main cells of innate immunity of the CNS, microglia constitutively express the most important immune receptors (MHC I and II, chemokine receptors) at low levels [60]. During activation, the immunologically relevant molecules are upregulated, and the appropriate antigen is presented [54] via MHC II. Additionally, it has recently been shown that microglia are able to cross-present exogenous antigens on MHC I to CD8+ T cells [61]. In the course of recovery from injury, activated microglia can be eliminated by apoptosis [54,62]. Not only neurons, but also astrocytes can be critical for microglial activation, e.g. in the course of infections [63]. Astrocytes, moreover, regulate the trafficking of lymphocytes across the brain endothelial barriers [64].

Microglial activation can be acute or chronic, and it has been suggested that this depends not only on the duration of an external cue, but also on the specific factor (stress, infection, inflammation, signals from damaged neurons) responsible for the activation process [54]. It has been shown that microglial activation during stress differs from that during infection/inflammation [65]. Chronic microglial activation can lead to microglial overactivation followed by microglial degeneration, as has been demonstrated in several in vitro studies [66,67]. Because of the absence of microglial support, the degeneration of microglia will be followed by secondary neurodegeneration.

Microglia are already widely dispersed throughout the immature white matter by 22 weeks of gestation, and have complete functionality. After stimulation with lipopolysaccharides (LPS) and proinflammatory cytokines [IFN-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-1β], they are fully capable of producing proinflammatory cytokines, reactive oxygen intermediates and proteolytic enzymes, and they have phagocytic activity [1,24,68]. In hypoxic-ischemic neonatal brain injury, the presence of activated microglia inducing cell death in immature white matter, both in preoligodendrocytes and in astrocytes, has been widely confirmed [59,69]. It also seems that microglia and resident mononuclear phagocytes are the primary sources of proinflammatory cytokines in brains with periventricular leukomalacia [70].

Microglial activation has often been the first – or at least a significant – cellular event detected in and around a lesion in several animal models of developing brain injuries such as those induced by mechanical trauma, infection/inflammation, excitotoxic insults and hypoxia-ischemia [59,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86]. Moreover, microglial activation has been demonstrated in postmortem brain specimens of premature infants with periventricular leukomalacia [87,88,89]. The activation of microglia by LPS induces oligodendrocyte cell death and also greatly impairs oligodendrocyte development through cytokines and modulations of growth factor secretion [90]. Oligodendrocyte-microglial communication could be one of the mechanisms underlying selective white matter damage and hypomyelination in periventricular leukomalacia. Similarly, activated microglia-macrophages are seen in abundance following both neonatal hypoxia-ischemia [91,92] and focal stroke [93,94], producing inflammatory cytokines, high levels of NO, complement molecules and matrix metalloproteinases (MMP). By proteolytic cleavage, the MMP control the components of the extracellular matrix proteins, such as adhesion, membrane receptors and soluble proteins. The early postinjury macrophage population comprises resident microglia rather than invading monocytes [69,93]. The notion that microglia contribute to, rather than limit, acute ischemic injury in the immature brain comes from studies illustrating an association between the reduced extent of a lesion and reduced microglial activation/monocyte infiltration [69,95,96].

Abundant evidence shows that the microglial cell phenotype and function changes during the normal aging process. Several authors have shown in various species that during the physiological aging process, microglia adopt an ‘activated’ phenotype with short and plump processes [33]. These ‘primed’ or ‘sensitized’ microglia are characterized by chronic low-level inflammation and increased microglial reactivity [97]. Microglia from aged animals showed increased expression of MHC II, CD68, CD11b, CD80, CD86 and intercellular adhesion molecule 1 [97,98], increased secretion of proinflammatory cytokines IL-1β, IL-6 and TNF-α, and reduced secretion of anti-inflammatory cytokines IL-10 and IL-4 [97,98,99]. Functional alterations included increased microglial reactivity, leading to a more rapid and pronounced response to pathological stimuli. According to this, aged mice not only have higher basal levels of these inflammatory mediators, but also show a higher stress-induced increase in these mediators [100]. Several studies have shown an increased secretion of IL-1β or IL-6 [100,101,102] after stress or stimulation. Although the amount of TNF-α constitutively secreted by microglia from aged mice was greater as well, the secretion of TNF-α was less responsive to stimulation, probably because of the dramatically enhanced release under basal conditions [99]. Furthermore, microglia from aged animals showed reduced glutathione levels, suggesting that the insult from reactive oxygen species (ROS) may be greater in aged brains [99]. The decreased amount of anti-inflammatory cytokines may enhance age-related neuroinflammation [97]. Microglia from aged mice also internalize less amyloid-β peptide, suggesting a lack of amyloid elimination by parenchymal microglia in aged adults suffering from Alzheimer’s disease [99]. Gene expression profiles showed that expression of genes indicative of oxidative stress, inflammation and glial activation increases with age. In contrast, expression of genes associated with synaptic function/transport, growth factors and trophic factors decreases with age [97,103,104,105]. Moreover, microglial dystrophy, i.e. the adoption of aberrant features indicating microglial senescence, has been acknowledged [33]. This may change the capability of microglia to adopt a protective phenotype with aging.

Which intra- and/or extracellular factors are responsible for transforming microglia into aggressors potentiating damage, or then helpers in the recovery following CNS damage? Whether microglia have a beneficial or harmful function depends on several factors: the kind of stress and damage signals, the duration/timing of an impact, the microenvironment, the interaction with other cell types and, interestingly, even the age of an organism [60]. Macrophages/microglia can be classified into at least two subsets with distinct molecular phenotypes and effector functions depending on the activation pathway. The ‘classically activated’ proinflammatory M1 macrophages, activated by LPS and by the proinflammatory cytokine IFN-γ, express CD86 and CD16/32 and produce high amounts of oxidative metabolites (NO and superoxide), proteases and proinflammatory cytokines. They play a central role in host defense against pathogens and tumor cells, and they can also damage healthy cells such as neurons and glial cells. In contrast, M2 macrophages are ‘alternatively activated’, anti-inflammatory macrophages induced by IL-4 and IL-13, and they express CD206 and arginase 1. The latter downregulate inflammation and promote tissue remodeling/repair and angiogenesis (fig. 2). The M1/M2 classification is, of course, a simplification of matters, and for macrophages, further intermediate phenotypes have been described [106]. In a mouse model of spinal cord injury, it has been shown that the M1/M2 ratio is markedly enhanced, leading to secondary neurodegeneration, in contrast to wound healing and healing of the myocardium with a shift from M1 to M2 macrophages during the healing process. Hypothetically, this delayed phase depends on the activation of an alternative set of transcription factors [107,108,109]. In a model of LPS-stimulated chronic systemic inflammation, it was shown that microglia in young mice protect dopamine-producing neurons against the toxin; however, microglia in old mice promoted the death of these cells [110]. Another study also illustrated a dual role: microglia induced both axon regeneration and neurotoxicity in neurons transplanted into rat spinal cord [111]. One of the most important functions of the microglia, phagocytosis, can also be both beneficial (pathogens, brain injury) and harmful (autoimmune disease). Activation of Toll-like receptor (TLR)4, the best-analyzed TLR in microglia, which plays a central role in many bacterial infections, also can be both neuroprotective and neurodestructive [60]. It can induce neurodegeneration by release of proinflammatory molecules, but it can also promote remyelination by recruiting oligodendrocyte progenitor cells [60,112]. Concerning cytokine production in the CNS, microglia can both produce and respond to cytokines, and these can be either neuroprotective [e.g. IL-10, tumor growth factor (TGF)-β, TNF-α] and/or neurotoxic (e.g. IL-1, TNF-α, IFN-γ) [60]. In addition, secreted factors such as one of the classical proinflammatory cytokines of neurodegeneration, TNF-α, can act both neurotoxically and neuroprotectively. Although TNF-α is neurotoxic at high levels, it may be neuroprotective at low levels, as has been demonstrated in a study using TNF receptor (TNFR)-deficient mice, for instance [113]. Among several factors, this seems to be dependent on the targeted receptor: TNFR1 activation can induce neurodegeneration, whereas TNFR2 activation promotes neuroprotection [114]. MMP-3 and -9 are important mediators during stroke and are released by microglia [115]. They play an important role in neurodegeneration after stroke, as was shown by (i) a reduction of infarct size following MMP inhibition and (ii) reduced ischemic injury in mice deficient in MMP-9 or -3 [116,117]. However, MMP also play a critical role in neurovascular remodeling after stroke [118].

Fig. 2

Microglial phenotypes – the yin and yang. Macrophages/microglia can be classified in a simplified manner into two subsets of phenotypes and effector functions depending on the activation pathway. Further intermediate phenotypes have been described.

Fig. 2

Microglial phenotypes – the yin and yang. Macrophages/microglia can be classified in a simplified manner into two subsets of phenotypes and effector functions depending on the activation pathway. Further intermediate phenotypes have been described.

Close modal

Microglial contact can be a ‘kiss of death’, and such contact between microglia and neurons plays a pivotal role in the pathogenesis of neurodegenerative disorders [50]. The presence of activated microglia and their ability to induce cell death in the immature white matter, both in preoligodendrocytes and in astrocytes, has been confirmed by several authors [59,69]. Also, microglia have been reported to be the primary sources of proinflammatory cytokines detected in brains with periventricular leukomalacia [70].

The exact mechanism leading to microglial overactivation is still not fully understood, but glial-neuronal crosstalk seems to be central [60]. Moreover, microglia and astrocyte interaction seems important: proinflammatory cytokines secreted by activated microglia inhibit astrocyte gap junction communication, which influences the role of astrocytes in providing neuronal support [60,119]. Microglial activation is one of the first steps in the inflammatory processes within the CNS, and it is often followed – depending on the activating mechanisms – by an infiltration of neutrophils, T lymphocytes and reactive astrocytosis [120]. Activated microglia release several cytotoxic substances such as ROS and the proinflammatory cytokines IL-1β [121,122], TNF-α [123], MMP and glutamate [60]. The ROS (superoxide, hydrogen peroxide, NO) cannot only kill invading microbes, but they also induce neuronal damage and reactive gliosis [60]. NO synthase [inducible (i)NOS, NOS2] is not expressed at high levels in the healthy brain, but in microglia and some astrocytes during inflammation [60] and stroke [124]. In vitro, iNOS is induced in microglia by IFN-γ, TNF-α and IL-1β, but not by TGF-β [125]. iNOS-deficient mice have smaller infarcts [126]. Superoxide is produced by microglia via NADPH oxidase [127]. Glucose availability can be rate limiting for NADPH production. This explains why ischemic or inflammatory injury can be exacerbated by hyperglycemia [128,129]. It has also been shown that mice deficient in the gp91 subunit of NADPH oxidase 2 have smaller infarcts than their wild-type littermates [130,131]. Such a role of NADPH detected in the adult brain has been shown not to contribute significantly to perinatal brain damage [132]. Microglia can release glutamate, a neurotransmitter that is well known to trigger excitotoxic neurodegeneration and cell death of astrocytes and oligodendrocytes. Especially the glutamate release from chronically activated microglia in the postischemic period could play an important role in brain injury [124]. Other processes such as activation of the prostaglandin E2 receptor of the prostaglandin E2 pathway can lead to microglia-induced paracrine neurotoxicity [133].

Blocking overactivated microglia in neurodegeneration settings seems to be an attractive therapeutic strategy for various neurodegenerative disorders. The tetracycline antibiotic minocycline is a drug that has been used in adults to treat infectious diseases without major side effects, but it has not been administered to children. It effectively crosses the blood-brain barrier, targets microglia by inhibiting the production of proinflammatory cytokines and NO and reduces their migration to injured neurons [134]. An inhibition of the MMP-9 pathway was described for both minocycline and doxycycline [135,136]. Minocycline has been shown to be neuroprotective in several adult animal models of neurological disorders [137,138,139,140,141,142,143,144]. Similarly, minocycline has been reported to be neuroprotective in developing brain damage models such as hypoxia-ischemia in neonatal or juvenile rats [145,146,147,148,149,150] and excitotoxic perinatal brain damage in mice [69]. On the other hand, minocycline worsened hypoxic-ischemic brain injury in a model of neonatal brain damage [151]. The reason for the discrepancy is not clear but may depend on dosage, application time with respect to brain damage induction, developmental age and genetic background. This finding, however, highlights that microglial inhibition may not be protective at all times.

In parallel with their negative or neurotoxic effects, microglia also play an important role in the maintenance of neuronal wellbeing [52,152]. Based on their phagocytic function as the ‘professional’ phagocytes of the CNS, microglia can enter damaged brain regions and remove toxic byproducts, invading pathogens and cell debris. In case of pathogens, stimulation of TLR induces a proinflammatory cascade. In case of cell debris due to brain injury, the recognition of phosphatidylserine of the apoptotic cell membrane induces an anti-inflammatory response [60]. Microglia have an important protective function in brain injury by removal of damaged cells, promoting neurogenesis, inducing the reestablishment of a functional neuronal environment by restoration of the myelin sheath, and by releasing neurotrophic factors and anti-inflammatory molecules [60,153,154]. Insufficient removal of myelin by microglia impairs the recruitment of oligodendrocyte precursor cells and induces an arrest of oligodendrocyte differentiation [155].

The neuroprotective role of microglia has not been well studied in models of developmental brain damage. Though in the immature and mature brain, microglia appear to have differing properties, studies performed on the adult brain may offer more insight. Neuroprotective properties of microglia have been described for adult neurological diseases such as Alzheimer’s. Here, microglia promote protection via the secretion of proteolytic enzymes that degrade amyloid-β, and by the phagocytotic clearance of amyloid-β plaques [60]. Furthermore, a specific macropinocytic mechanism, different from that known for phagocytosis, has been described both in vitro and in vivo [156]. Chemokine (C-C motif) receptor 2 seems to play an important role in the protection of microglia in Alzheimer’s disease since mice deficient in chemokine (C-C motif) receptor 2 exhibited increased amyloid-β deposits and died prematurely [157]. In experimental models of Alzheimer’s disease, two microglial subtypes have been suggested to play different neuroimmunomodulating roles [158]. Bone marrow-derived microglia were able to eliminate amyloid deposits by phagocytosis [159]. Microglia can also be protective via the production of cytokines: (i) IL-6 may act on astrocytes to induce brain tissue repair [160]; (ii) IL-10 can inhibit apoptosis of microglia [161], and (iii) TGF-β can be neuroprotective. Inhibition of TGF-β activity in a rodent model of prion disease induced cerebral inflammation [162]. In a model of Alzheimer’s disease, it reduced the plaque load [163].

Microglia seem to be much more integrated into neuronal function than was thought in the past, and recent findings indicate neuronal-microglial crosstalk [164]. Recent findings suggest the intriguing hypothesis that microglia can sense neuronal activity based on local neurotransmitter levels [164]. This is at least in part maintained by neurotransmitter receptors identified on microglial cells, including metabotropic and ionotropic glutamate [165,166,167,168,169,170,171] as well as γ-aminobutyric acid B receptors [172].

Apart from the well-known, defense-oriented reactions of microglia [173], there is accumulating evidence of their role in physiologic brain development and normal function of the mature and immature nervous system [50,174]. Future studies will need to illustrate more and in detail the differences in microglial function at various stages of development as well as the protective role of microglia in the immature brain. Deciphering the factors that influence the transformation of microglia into aggressors potentiating damage or into kind helpers in recovery following CNS damage will be a challenge for the future. These ‘factors’ may be targets for a highly effective pharmaceutical therapy for neurodegenerative diseases such as perinatal brain damage.

Our research is supported by the German Research Foundation (DFG; SFB665 and IB of the Bundesministerium für Bildung und Forschung), the Sonnenfeld Stiftung, the Berliner Krebsgesellschaft e.V., Inserm, Université Paris Diderot, PremUP and the Fondation Leducq.

1.
Kaindl AM, Favrais G, Gressens P: Molecular mechanisms involved in injury to the preterm brain. J Child Neurol 2009;24:1112–1118.
2.
Weinstein JR, Koerner IP, Möller T: Microglia in ischemic brain injury. Future Neurol 2010;5:227–246.
3.
Yadav A, Collman RG: CNS inflammation and macrophage/microglial biology associated with HIV-1 infection. J Neuroimmune Pharmacol 2009;4:430–447.
4.
Watters JJ, Schartner JM, Badie B: Microglia function in brain tumors. J Neurosci Res 2005;81:447–455.
5.
Jack C, Ruffini F, Bar-Or A, Antel JP: Microglia and multiple sclerosis. J Neurosci Res 2005;81:363–373.
6.
Moisse K, Strong MJ: Innate immunity in amyotrophic lateral sclerosis. Biochim Biophys Acta 2006;1762:1083–1093.
7.
Morgan D: The role of microglia in antibody-mediated clearance of amyloid-β from the brain. CNS Neurol Disord Drug Targets 2009;8:7–15.
8.
Teismann P, Tieu K, Cohen O, et al: Pathogenic role of glial cells in Parkinson’s disease. Mov Disord 2003;18:121–129.
9.
Pavese N, Gerhard A, Tai YF, et al: Microglial activation correlates with severity in Huntington disease: a clinical and PET study. Neurology 2006;66:1638–1643.
10.
Garden GA, Möller T: Microglia biology in health and disease. J Neuroimmune Pharmacol 2006;1:127–137.
11.
Heppner FL, Greter M, Marino D, et al: Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat Med 2005;11:146–152.
12.
Turrin NP, Rivest S: Molecular and cellular immune mediators of neuroprotection. Mol Neurobiol 2006;34:221–242.
13.
Prinz M, Mildner A: Microglia in the CNS: immigrants from another world. Glia 2011;59:177–187.
14.
Ginhoux F, Greter M, Leboeuf M, et al: Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010;330:841–845.
15.
Kaur C, Hao AJ, Wu CH, Ling EA: Origin of microglia. Microsc Res Tech 2001;54:2–9.
16.
Monier A, Adle-Biassette H, Delezoide AL, et al: Entry and distribution of microglial cells in human embryonic and fetal cerebral cortex. J Neuropathol Exp Neurol 2007;66:372–382.
17.
Monier A, Evrard P, Gressens P, Verney C: Distribution and differentiation of microglia in the human encephalon during the first two trimesters of gestation. J Comp Neurol 2006;499:565–582.
18.
Clausen BH, Lambertsen KL, Babcock AA, et al: Interleukin-1β and tumor necrosis factor-α are expressed by different subsets of microglia and macrophages after ischemic stroke in mice. J Neuroinflammation 2008;5:46.
19.
Barron KD: Microglia: history, cytology, and reactions. J Neurol Sci 2003;207:98.
20.
Verney C, Monier A, Fallet-Bianco C, Gressens P: Early microglial colonization of the human forebrain and possible involvement in periventricular white-matter injury of preterm infants. J Anat 2010;217:436–448.
21.
Nimmerjahn A, Kirchhoff F, Helmchen F: Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005;308:1314–1318.
22.
Streit WJ, Kreutzberg GW: Response of endogenous glial cells to motor neuron degeneration induced by toxic ricin. J Comp Neurol 1988;268:248–263.
23.
Esiri MM, al Izzi MS, Reading MC: Macrophages, microglial cells, and HLA-DR antigens in fetal and infant brain. J Clin Pathol 1991;44:102–106.
24.
Rezaie P, Cairns NJ, Male DK: Expression of adhesion molecules on human fetal cerebral vessels: relationship to microglial colonisation during development. Brain Res Dev Brain Res 1997;104:175–189.
25.
Simard AR, Rivest S: Bone marrow stem cells have the ability to populate the entire central nervous system into fully differentiated parenchymal microglia. FASEB J 2004;18:998–1000.
26.
Massengale M, Wagers AJ, Vogel H, Weissman IL: Hematopoietic cells maintain hematopoietic fates upon entering the brain. J Exp Med 2005;201:1579–1589.
27.
Tomita M, Mori T, Maruyama K, et al: A comparison of neural differentiation and retinal transplantation with bone marrow-derived cells and retinal progenitor cells. Stem Cells 2006;24:2270–2278.
28.
Opydo-Chanek M, Dabrowski Z: Response of astrocytes and microglia/macrophages to brain injury after bone marrow stromal cell transplantation: a quantitative study. Neurosci Lett 2011;487:163–168.
29.
Priller J, Flügel A, Wehner T, et al: Targeting gene-modified hematopoietic cells to the central nervous system: use of green fluorescent protein uncovers microglial engraftment. Nat Med 2001;7:1356–1361.
30.
Rezaie P: Microglia in the human nervous system during development. Neuroembryology 2003;2:18–31.
31.
Rezaie P, Male D: Colonisation of the developing human brain and spinal cord by microglia: a review. Microsc Res Tech 1999;45:359–382.
32.
Rezaie P, Patel K, Male DK: Microglia in the human fetal spinal cord: patterns of distribution, morphology and phenotype. Brain Res Dev Brain Res 1999;115:71–81.
33.
Olah M, Biber K, Vinet J, Boddeke HW: Microglia phenotype diversity. CNS Neurol Disord Drug Targets 2011;10:108–118.
34.
Ferrer I, Bernet E, Soriano E, et al: Naturally occurring cell death in the cerebral cortex of the rat and removal of dead cells by transitory phagocytes. Neuroscience 1990;39:451–458.
35.
Antony JM, Paquin A, Nutt SL, et al: Endogenous microglia regulate development of embryonic cortical precursor cells. J Neurosci Res 2011;89:286–298.
36.
Welser JV, Li L, Milner R: Microglial activation state exerts a biphasic influence on brain endothelial cell proliferation by regulating the balance of TNF and TGF-β1. J Neuroinflammation 2010;7:89.
37.
Miller FD, Gauthier-Fisher A: Home at last: neural stem cell niches defined. Cell Stem Cell 2009;4:507–510.
38.
Ekdahl CT, Kokaia Z, Lindvall O: Brain inflammation and adult neurogenesis: the dual role of microglia. Neuroscience 2009;158:1021–1029.
39.
Sonnenfeld MJ, Jacobs JR: Macrophages and glia participate in the removal of apoptotic neurons from the Drosophila embryonic nervous system. J Comp Neurol 1995;359:644–652.
40.
Marin-Teva JL, Dusart I, Colin C, et al: Microglia promote the death of developing Purkinje cells. Neuron 2004;41:535–547.
41.
Aarum J, Sandberg K, Haeberlein SL, Persson MA: Migration and differentiation of neural precursor cells can be directed by microglia. Proc Natl Acad Sci USA 2003;100:15983–15988.
42.
Jonakait GM, Luskin MB, Wei R, et al: Conditioned medium from activated microglia promotes cholinergic differentiation in the basal forebrain in vitro. Dev Biol 1996;177:85–95.
43.
Jonakait GM, Wen Y, Wan Y, Ni L: Macrophage cell-conditioned medium promotes cholinergic differentiation of undifferentiated progenitors and synergizes with nerve growth factor action in the developing basal forebrain. Exp Neurol 2000;161:285–296.
44.
Nakanishi M, Niidome T, Matsuda S, et al: Microglia-derived interleukin-6 and leukaemia inhibitory factor promote astrocytic differentiation of neural stem/progenitor cells. Eur J Neurosci 2007;25:649–658.
45.
Zhu P, Hata R, Cao F, et al: Ramified microglial cells promote astrogliogenesis and maintenance of neural stem cells through activation of Stat3 function. FASEB J 2008;22:3866–3877.
46.
Dougherty KD, Dreyfus CF, Black IB: Brain-derived neurotrophic factor in astrocytes, oligodendrocytes, and microglia/macrophages after spinal cord injury. Neurobiol Dis 2000;7:574–585.
47.
O’Donnell SL, Frederick TJ, Krady JK, et al: IGF-I and microglia/macrophage proliferation in the ischemic mouse brain. Glia 2002;39:85–97.
48.
Banati RB, Graeber MB: Surveillance, intervention and cytotoxicity: is there a protective role of microglia? Dev Neurosci 1994;16:114–127.
49.
Kreutzberg GW: Microglia: a sensor for pathological events in the CNS. Trends Neurosci 1996;19:312–318.
50.
Hanisch UK, Kettenmann H: Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci 2007;10:1387–1394.
51.
Lobsiger CS, Cleveland DW: Glial cells as intrinsic components of non-cell-autonomous neurodegenerative disease. Nat Neurosci 2007;10:1355–1360.
52.
Streit WJ: Microglia as neuroprotective, immunocompetent cells of the CNS. Glia 2002;40:133–139.
53.
Raivich G: Like cops on the beat: the active role of resting microglia. Trends Neurosci 2005;28:571–573.
54.
Graeber MB, Streit WJ: Microglia: biology and pathology. Acta Neuropathol 2010;119:89–105.
55.
Davalos D, Grutzendler J, Yang G, et al: ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci 2005;8:752–758.
56.
Haynes SE, Hollopeter G, Yang G, et al: The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci 2006;9:1512–1519.
57.
Rappert A, Bechmann I, Pivneva T, et al: CXCR3-dependent microglial recruitment is essential for dendrite loss after brain lesion. J Neurosci 2004;24:8500–8509.
58.
Pocock JM, Kettenmann H: Neurotransmitter receptors on microglia. Trends Neurosci 2007;30:527–535.
59.
Tahraoui SL, Marret S, Bodenant C, et al: Central role of microglia in neonatal excitotoxic lesions of the murine periventricular white matter. Brain Pathol 2001;11:56–71.
60.
Walter L, Neumann H: Role of microglia in neuronal degeneration and regeneration. Semin Immunopathol 2009;31:513–525.
61.
Beauvillain C, Donnou S, Jarry U, et al: Neonatal and adult microglia cross-present exogenous antigens. Glia 2008;56:69–77.
62.
Jones LL, Banati RB, Graeber MB, et al: Population control of microglia: does apoptosis play a role? J Neurocytol 1997;26:755–770.
63.
Ovanesov MV, Ayhan Y, Wolbert C, et al: Astrocytes play a key role in activation of microglia by persistent Borna disease virus infection. J Neuroinflammation 2008;5:50.
64.
Hudson LC, Bragg DC, Tompkins MB, Meeker RB: Astrocytes and microglia differentially regulate trafficking of lymphocyte subsets across brain endothelial cells. Brain Res 2005;1058:148–160.
65.
Sugama S, Takenouchi T, Fujita M, et al: Differential microglial activation between acute stress and lipopolysaccharide treatment. J Neuroimmunol 2009;207:24–31.
66.
Liu B, Wang K, Gao HM, et al: Molecular consequences of activated microglia in the brain: overactivation induces apoptosis. J Neurochem 2001;77:182–189.
67.
Polazzi E, Contestabile A: Overactivation of LPS-stimulated microglial cells by co-cultured neurons or neuron-conditioned medium. J Neuroimmunol 2006;172:104–111.
68.
Smith ME, van der Maesen K, Somera FP: Macrophage and microglial responses to cytokines in vitro: phagocytic activity, proteolytic enzyme release, and free radical production. J Neurosci Res 1998;54:68–78.
69.
Dommergues MA, Plaisant F, Verney C, Gressens P: Early microglial activation following neonatal excitotoxic brain damage in mice: a potential target for neuroprotection. Neuroscience 2003;121:619–628.
70.
Kadhim H, Tabarki B, Verellen G, et al: Inflammatory cytokines in the pathogenesis of periventricular leukomalacia. Neurology 2001;56:1278–1284.
71.
Ide CF, Scripter JL, Coltman BW, et al: Cellular and molecular correlates to plasticity during recovery from injury in the developing mammalian brain. Prog Brain Res 1996;108:365–377.
72.
Hornig M, Weissenböck H, Horscroft N, Lipkin WI: An infection-based model of neurodevelopmental damage. Proc Natl Acad Sci USA 1999;96:12102–12107.
73.
Hagberg H, Peebles D, Mallard C: Models of white matter injury: comparison of infectious, hypoxic-ischemic, and excitotoxic insults. Ment Retard Dev Disabil Res Rev 2002;8:30–38.
74.
Jantzie LL, Cheung PY, Todd KG: Doxycycline reduces cleaved caspase-3 and microglial activation in an animal model of neonatal hypoxia-ischemia. J Cereb Blood Flow Metab 2005;25:314–324.
75.
Svedin P, Kjellmer I, Welin AK, et al: Maturational effects of lipopolysaccharide on white-matter injury in fetal sheep. J Child Neurol 2005;20:960–964.
76.
Chew LJ, Takanohashi A, Bell M: Microglia and inflammation: impact on developmental brain injuries. Ment Retard Dev Disabil Res Rev 2006;12:105–112.
77.
Kannan S, Saadani-Makki F, Muzik O, et al: Microglial activation in perinatal rabbit brain induced by intrauterine inflammation: detection with 11C-(R)-PK11195 and small-animal PET. J Nucl Med 2007;48:946–954.
78.
Jin Y, Silverman AJ, Vannucci SJ: Mast cell stabilization limits hypoxic-ischemic brain damage in the immature rat. Dev Neurosci 2007;29:373–384.
79.
Hutton LC, Castillo-Melendez M, Smythe GA, Walker DW: Microglial activation, macrophage infiltration, and evidence of cell death in the fetal brain after uteroplacental administration of lipopolysaccharide in sheep in late gestation. Am J Obstet Gynecol 2008;198:117e1–e11.
80.
Keller M, Griesmaier E, Auer M, et al: Dextromethorphan is protective against sensitized N-methyl-D-aspartate receptor-mediated excitotoxic brain damage in the developing mouse brain. Eur J Neurosci 2008;27:874–883.
81.
Jin Y, Silverman AJ, Vannucci SJ: Mast cells are early responders after hypoxia-ischemia in immature rat brain. Stroke 2009;40:3107–3112.
82.
Fan LW, Mitchell HJ, Tien LT, et al: Interleukin-1β-induced brain injury in the neonatal rat can be ameliorated by α-phenyl-n-tert-butyl-nitrone. Exp Neurol 2009;220:143–153.
83.
Baud O, Daire JL, Dalmaz Y, et al: Gestational hypoxia induces white matter damage in neonatal rats: a new model of periventricular leukomalacia. Brain Pathol 2004;14:1–10.
84.
Mallard C, Welin AK, Peebles D, et al: White matter injury following systemic endotoxemia or asphyxia in the fetal sheep. Neurochem Res 2003;28:215–223.
85.
Olivier P, Baud O, Evrard P, et al: Prenatal ischemia and white matter damage in rats. J Neuropathol Exp Neurol 2005;64:998–1006.
86.
Deng Y, Lu J, Sivakumar V, et al: 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 2008;18:387–400.
87.
Hirayama A, Okoshi Y, Hachiya Y, et al: Early immunohistochemical detection of axonal damage and glial activation in extremely immature brains with periventricular leukomalacia. Clin Neuropathol 2001;20:87–91.
88.
Deguchi K, Oguchi K, Takashima S: Characteristic neuropathology of leukomalacia in extremely low birth weight infants. Pediatr Neurol 1997;16:296–300.
89.
Rezaie P, Dean A: Periventricular leukomalacia, inflammation and white matter lesions within the developing nervous system. Neuropathology 2002;22:106–132.
90.
Pang Y, Campbell L, Zheng B, et al: Lipopolysaccharide-activated microglia induce death of oligodendrocyte progenitor cells and impede their development. Neuroscience 2010;166:464–475.
91.
Ivacko JA, Sun R, Silverstein FS: Hypoxic-ischemic brain injury induces an acute microglial reaction in perinatal rats. Pediatr Res 1996;39:39–47.
92.
McRae A, Gilland E, Bona E, Hagberg H: Microglia activation after neonatal hypoxic-ischemia. Brain Res Dev Brain Res 1995;84:245–252.
93.
Denker SP, Ji S, Dingman A, et al: Macrophages are comprised of resident brain microglia not infiltrating peripheral monocytes acutely after neonatal stroke. J Neurochem 2007;100:893–904.
94.
Dingman A, Lee SY, Derugin N, et al: Aminoguanidine inhibits caspase-3 and calpain activation without affecting microglial activation following neonatal transient cerebral ischemia. J Neurochem 2006;96:1467–1479.
95.
Arvin KL, Han BH, Du Y, et al: Minocycline markedly protects the neonatal brain against hypoxic-ischemic injury. Ann Neurol 2002;52:54–61.
96.
Fox C, Dingman A, Derugin N, et al: Minocycline confers early but transient protection in the immature brain following focal cerebral ischemia-reperfusion. J Cereb Blood Flow Metab 2005;25:1138–1149.
97.
Jurgens HA, Johnson RW: Dysregulated neuronal-microglial cross-talk during aging, stress and inflammation. Exp Neurol 2010, E-pub ahead of print.
98.
Frank MG, Wieseler-Frank JL, Watkins LR, Maier SF: Rapid isolation of highly enriched and quiescent microglia from adult rat hippocampus: immunophenotypic and functional characteristics. J Neurosci Methods 2006;151:121–130.
99.
Njie EG, Boelen E, Stassen FR, et al: Ex vivo cultures of microglia from young and aged rodent brain reveal age-related changes in microglial function. Neurobiol Aging 2010, E-pub ahead of print.
100.
Buchanan JB, Sparkman NL, Chen J, Johnson RW: Cognitive and neuroinflammatory consequences of mild repeated stress are exacerbated in aged mice. Psychoneuroendocrinology 2008;33:755–765.
101.
Frank MG, Barrientos RM, Watkins LR, Maier SF: Aging sensitizes rapidly isolated hippocampal microglia to LPS ex vivo. J Neuroimmunol 2010;226:181–184.
102.
Henry CJ, Huang Y, Wynne AM, Godbout JP: Peripheral lipopolysaccharide (LPS) challenge promotes microglial hyperactivity in aged mice that is associated with exaggerated induction of both pro-inflammatory IL-1β and anti-inflammatory IL-10 cytokines. Brain Behav Immun 2009;23:309–317.
103.
Bishop NA, Lu T, Yankner BA: Neural mechanisms of ageing and cognitive decline. Nature 2010;464:529–535.
104.
Blalock EM, Chen KC, Sharrow K, et al: Gene microarrays in hippocampal aging: statistical profiling identifies novel processes correlated with cognitive impairment. J Neurosci 2003;23:3807–3819.
105.
Lee CK, Weindruch R, Prolla TA: Gene-expression profile of the ageing brain in mice. Nat Genet 2000;25:294–297.
106.
Mosser DM, Edwards JP: Exploring the full spectrum of macrophage activation. Nat Rev Immunol 2008;8:958–969.
107.
Kigerl KA, Gensel JC, Ankeny DP, et al: Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci 2009;29:13435–13444.
108.
Gordon S: Alternative activation of macrophages. Nat Rev Immunol 2003;3:23–35.
109.
Mantovani A, Sica A, Sozzani S, et al: The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 2004;25:677–686.
110.
Sawada M, Sawada H, Nagatsu T: Effects of aging on neuroprotective and neurotoxic properties of microglia in neurodegenerative diseases. Neurodegener Dis 2008;5:254–256.
111.
Gensel JC, Nakamura S, Guan Z, et al: Macrophages promote axon regeneration with concurrent neurotoxicity. J Neurosci 2009;29:3956–3968.
112.
Glezer I, Lapointe A, Rivest S: Innate immunity triggers oligodendrocyte progenitor reactivity and confines damages to brain injuries. FASEB J 2006;20:750–752.
113.
Bruce AJ, Boling W, Kindy MS, et al: Altered neuronal and microglial responses to excitotoxic and ischemic brain injury in mice lacking TNF receptors. Nat Med 1996;2:788–794.
114.
Fontaine V, Mohand-Said S, Hanoteau N, et al: Neurodegenerative and neuroprotective effects of tumor necrosis factor (TNF) in retinal ischemia: opposite roles of TNF receptor 1 and TNF receptor 2. J Neurosci 2002;22:RC216.
115.
del Zoppo GJ, Milner R, Mabuchi T, et al: Microglial activation and matrix protease generation during focal cerebral ischemia. Stroke 2007;38:646–651.
116.
Asahi M, Asahi K, Jung JC, et al: Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab 2000;20:1681–1689.
117.
Walker EJ, Rosenberg GA: TIMP-3 and MMP-3 contribute to delayed inflammation and hippocampal neuronal death following global ischemia. Exp Neurol 2009;216:122–131.
118.
Zhao BQ, Wang S, Kim HY, et al: Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med 2006;12:441–445.
119.
Retamal MA, Froger N, Palacios-Prado N, et al: Cx43 hemichannels and gap junction channels in astrocytes are regulated oppositely by proinflammatory cytokines released from activated microglia. J Neurosci 2007;27:13781–13792.
120.
Zheng Z, Yenari MA: Post-ischemic inflammation: molecular mechanisms and therapeutic implications. Neurol Res 2004;26:884–892.
121.
Lee SC, Liu W, Dickson DW, et al: Cytokine production by human fetal microglia and astrocytes: differential induction by lipopolysaccharide and IL-1β. J Immunol 1993;150:2659–2667.
122.
Davies CA, Loddick SA, Toulmond S, et al: The progression and topographic distribution of interleukin-1β expression after permanent middle cerebral artery occlusion in the rat. J Cereb Blood Flow Metab 1999;19:87–98.
123.
Sawada M, Kondo N, Suzumura A, Marunouchi T: Production of tumor necrosis factor-α by microglia and astrocytes in culture. Brain Res 1989;491:394–397.
124.
Yenari MA, Kauppinen TM, Swanson RA: Microglial activation in stroke: therapeutic targets. Neurotherapeutics 2010;7:378–391.
125.
Vodovotz Y, Bogdan C: Control of nitric oxide synthase expression by transforming growth factor-β: implications for homeostasis. Prog Growth Factor Res 1994;5:341–351.
126.
Zhao X, Haensel C, Araki E, et al: Gene-dosing effect and persistence of reduction in ischemic brain injury in mice lacking inducible nitric oxide synthase. Brain Res 2000;872:215–218.
127.
Groemping Y, Rittinger K: Activation and assembly of the NADPH oxidase: a structural perspective. Biochem J 2005;386:401–416.
128.
Suh SW, Shin BS, Ma H, et al: Glucose and NADPH oxidase drive neuronal superoxide formation in stroke. Ann Neurol 2008;64:654–663.
129.
Decoursey TE, Ligeti E: Regulation and termination of NADPH oxidase activity. Cell Mol Life Sci 2005;62:2173–2193.
130.
Kahles T, Luedike P, Endres M, et al: NADPH oxidase plays a central role in blood-brain barrier damage in experimental stroke. Stroke 2007;38:3000–3006.
131.
Chen H, Song YS, Chan PH: Inhibition of NADPH oxidase is neuroprotective after ischemia-reperfusion. J Cereb Blood Flow Metab 2009;29:1262–1272.
132.
Doverhag C, Keller M, Karlsson A, et al: Pharmacological and genetic inhibition of NADPH oxidase does not reduce brain damage in different models of perinatal brain injury in newborn mice. Neurobiol Dis 2008;31:133–144.
133.
Cimino PJ, Keene CD, Breyer RM, et al: Therapeutic targets in prostaglandin E2 signaling for neurologic disease. Curr Med Chem 2008;15:1863–1869.
134.
Romero-Sandoval EA, Horvath RJ, DeLeo JA: Neuroimmune interactions and pain: focus on glial-modulating targets. Curr Opin Investig Drugs 2008;9:726–734.
135.
Lee H, Park JW, Kim SP, et al: Doxycycline inhibits matrix metalloproteinase-9 and laminin degradation after transient global cerebral ischemia. Neurobiol Dis 2009;34:189–198.
136.
Machado LS, Kozak A, Ergul A, et al: Delayed minocycline inhibits ischemia-activated matrix metalloproteinases 2 and 9 after experimental stroke. BMC Neurosci 2006;7:56.
137.
Yrjänheikki J, Keinänen R, Pellikka M, et al: Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc Natl Acad Sci USA 1998;95:15769–15774.
138.
Yrjänheikki J, Tikka T, Keinänen R, et al: A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci USA 1999;96:13496–13500.
139.
Du Y, Ma Z, Lin S, et al: Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinson’s disease. Proc Natl Acad Sci USA 2001;98:14669–14674.
140.
Chen M, Ona VO, Li M, et al: Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington disease. Nat Med 2000;6:797–801.
141.
Popovic N, Schubart A, Goetz BD, et al: Inhibition of autoimmune encephalomyelitis by a tetracycline. Ann Neurol 2002;51:215–223.
142.
Zhu S, Stavrovskaya IG, Drozda M, et al: Minocycline inhibits cytochrome c release and delays progression of amyotrophic lateral sclerosis in mice. Nature 2002;417:74–78.
143.
Carty ML, Wixey JA, Colditz PB, Buller KM: Post-insult minocycline treatment attenuates hypoxia-ischemia-induced neuroinflammation and white matter injury in the neonatal rat: a comparison of two different dose regimens. Int J Dev Neurosci 2008;26:477–485.
144.
Clark WM, Calcagno FA, Gabler WL, et al: Reduction of central nervous system reperfusion injury in rabbits using doxycycline treatment. Stroke 1994;25:1411–1415, discussion 1416.
145.
Bukowski L: Synthesis of 2-(2′-thiazolyl) alkanebenzimidazoles: potential anthelmintics (in Polish). Acta Pol Pharm 1975;32:651–656.
146.
Lechpammer M, Manning SM, Samonte F, et al: Minocycline treatment following hypoxic/ischaemic injury attenuates white matter injury in a rodent model of periventricular leucomalacia. Neuropathol Appl Neurobiol 2008;34:379–393.
147.
Fan LW, Lin S, Pang Y, et al: Minocycline attenuates hypoxia-ischemia-induced neurological dysfunction and brain injury in the juvenile rat. Eur J Neurosci 2006;24:341–350.
148.
Tang M, Alexander H, Clark RS, et al: Minocycline reduces neuronal death and attenuates microglial response after pediatric asphyxial cardiac arrest. J Cereb Blood Flow Metab 2010;30:119–129.
149.
Buller KM, Carty ML, Reinebrant HE, Wixey JA: Minocycline: a neuroprotective agent for hypoxic-ischemic brain injury in the neonate? J Neurosci Res 2009;87:599–608.
150.
Cai Z, Lin S, Fan LW, et al: Minocycline alleviates hypoxic-ischemic injury to developing oligodendrocytes in the neonatal rat brain. Neuroscience 2006;137:425–435.
151.
Tsuji M, Wilson MA, Lange MS, Johnston MV: Minocycline worsens hypoxic-ischemic brain injury in a neonatal mouse model. Exp Neurol 2004;189:58–65.
152.
Harry GJ, McPherson CA, Wine RN, et al: Trimethyltin-induced neurogenesis in the murine hippocampus. Neurotox Res 2004;5:623–627.
153.
Ziv Y, Ron N, Butovsky O, et al: Immune cells contribute to the maintenance of neurogenesis and spatial learning abilities in adulthood. Nat Neurosci 2006;9:268–275.
154.
Franklin RJ, Kotter MR: The biology of CNS remyelination: the key to therapeutic advances. J Neurol 2008;255(suppl 1):19–25.
155.
Kotter MR, Li WW, Zhao C, Franklin RJ: Myelin impairs CNS remyelination by inhibiting oligodendrocyte precursor cell differentiation. J Neurosci 2006;26:328–332.
156.
Mandrekar S, Jiang Q, Lee CY, et al: Microglia mediate the clearance of soluble Aβ through fluid phase macropinocytosis. J Neurosci 2009;29:4252–4262.
157.
El Khoury J, Toft M, Hickman SE, et al: Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nat Med 2007;13:432–438.
158.
Shimizu E, Kawahara K, Kajizono M, et al: IL-4-induced selective clearance of oligomeric β-amyloid peptide1–42 by rat primary type 2 microglia. J Immunol 2008;181:6503–6513.
159.
Simard AR, Soulet D, Gowing G, et al: Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer’s disease. Neuron 2006;49:489–502.
160.
Streit WJ, Hurley SD, McGraw TS, Semple-Rowland SL: Comparative evaluation of cytokine profiles and reactive gliosis supports a critical role for interleukin-6 in neuron-glia signaling during regeneration. J Neurosci Res 2000;61:10–20.
161.
Strle K, Zhou JH, Broussard SR, et al: IL-10 promotes survival of microglia without activating Akt. J Neuroimmunol 2002;122:9–19.
162.
Boche D, Cunningham C, Docagne F, et al: TGFβ1 regulates the inflammatory response during chronic neurodegeneration. Neurobiol Dis 2006;22:638–650.
163.
Wyss-Coray T, Lin C, Yan F, et al: TGF-β1 promotes microglial amyloid-β clearance and reduces plaque burden in transgenic mice. Nat Med 2001;7:612–618.
164.
Biber K, Neumann H, Inoue K, Boddeke HW: Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci 2007;30:596–602.
165.
Biber K, Laurie DJ, Berthele A, et al: Expression and signaling of group I metabotropic glutamate receptors in astrocytes and microglia. J Neurochem 1999;72:1671–1680.
166.
Taylor DL, Diemel LT, Cuzner ML, Pocock JM: Activation of group II metabotropic glutamate receptors underlies microglial reactivity and neurotoxicity following stimulation with chromogranin A, a peptide up-regulated in Alzheimer’s disease. J Neurochem 2002;82:1179–1191.
167.
Taylor DL, Diemel LT, Pocock JM: Activation of microglial group III metabotropic glutamate receptors protects neurons against microglial neurotoxicity. J Neurosci 2003;23:2150–2160.
168.
Taylor DL, Jones F, Kubota ES, Pocock JM: Stimulation of microglial metabotropic glutamate receptor mGlu2 triggers tumor necrosis factor α-induced neurotoxicity in concert with microglial-derived Fas ligand. J Neurosci 2005;25:2952–2964.
169.
Noda M, Nakanishi H, Nabekura J, Akaike N: AMPA-kainate subtypes of glutamate receptor in rat cerebral microglia. J Neurosci 2000;20:251–258.
170.
Hagino Y, Kariura Y, Manago Y, et al: Heterogeneity and potentiation of AMPA type of glutamate receptors in rat cultured microglia. Glia 2004;47:68–77.
171.
Gottlieb M, Matute C: Expression of ionotropic glutamate receptor subunits in glial cells of the hippocampal CA1 area following transient forebrain ischemia. J Cereb Blood Flow Metab 1997;17:290–300.
172.
Kuhn SA, van Landeghem FK, Zacharias R, et al: Microglia express GABAB receptors to modulate interleukin release. Mol Cell Neurosci 2004;25:312–322.
173.
Kim HJ, Ifergan I, Antel JP, et al: Type 2 monocyte and microglia differentiation mediated by glatiramer acetate therapy in patients with multiple sclerosis. J Immunol 2004;172:7144–7153.
174.
Butovsky O, Landa G, Kunis G, et al: Induction and blockage of oligodendrogenesis by differently activated microglia in an animal model of multiple sclerosis. J Clin Invest 2006;116:905–915.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.