Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 ·...

18
REVIEW ARTICLE Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy, PhD and Eng-Ang Ling, PhD, DSc Abstract Microglia exist in different morphological forms in the develop- ing brain. They show a small cell body with scanty cytoplasm with many branching processes in the grey matter of the developing brain. However, in the white matter such as the corpus callosum where the unmyelinated axons are loosely organized, they appear in an amoeboid form having a round cell body endowed with copious cytoplasm rich in organelles. The amoeboid cells eventually trans- form into ramified microglia in the second postnatal week when the tissue becomes more compact with the onset of myelination. Micro- glia serve as immunocompetent macrophages that act as neuropath- ology sensors to detect and respond swiftly to subtle changes in the brain tissues in pathological conditions. Microglial functions are broadly considered as protective in the normal brain development as they phagocytose dead cells and sculpt neuronal connections by pruning excess axons and synapses. They also secrete a number of trophic factors such as insulin-like growth factor-1 and transforming growth factor-b among many others that are involved in neuronal and oligodendrocyte survival. On the other hand, microglial cells when activated produce a plethora of molecules such as proinflam- matory cytokines, chemokines, reactive oxygen species, and nitric oxide that are implicated in the pathogenesis of many pathological conditions such as epilepsy, cerebral palsy, autism, and perinatal hypoxic-ischemic brain injury. Although many studies have investi- gated the origin and functions of the microglia in the developing brain, in-depth in vivo studies along with analysis of their transcrip- tome and epigenetic changes need to be undertaken to elucidate their full potential be it protective or neurotoxic. This would lead to a bet- ter understanding of their roles in the healthy and diseased develop- ing brain and advancement of therapeutic strategies to target microglia-mediated neurotoxicity. Key Words: Antigen presentation, Developing brain, Hypoxia-is- chemia, Inflammatory cytokines, Microglia, Phagocytosis. INTRODUCTION Microglia are gaining tremendous recognition in recent years because they are implicated in different neurodegenera- tive diseases and neurological disorders (1, 2). In the mature rodent brain, they contribute 5%–12% of the total glial popula- tion, and in reference to other glial cells, the frequency appears to vary with regions, for example, in the cerebral cortex or cor- pus callosum (3). The prevailing view regarding roles of microglia is that they act as resident immunocompetent phago- cytic cells in the disease processes in infectious, traumatic, in- flammatory, ischemic, and degenerative conditions of the cen- tral nervous system (CNS) (4). As far as can be ascertained, the existence of microglia as a separate cellular entity in the normal brain or in pathological conditions was first docu- mented by Del Rio-Hortega (5) in the early part of the last cen- tury; yet compared with other glial types in the brain, it would appear that microglia had attracted little attention by the con- temporary authors until decades later (6, 7). It is possible that this might have been hindered by the lack of a reliable staining method for identification of microglia during that period. The weak silver carbonate staining method routinely used then for this purpose was described to be capricious, often yielding un- satisfactory results. Despite the technical limitations, some early authors had attempted to investigate the origin, develop- ment and roles of microglia. The study was later extended to the ultrastructural identification of microglia in which the sil- ver carbonate staining was adapted for transmission electron microscopy (6, 7). The seminal report on the characterization of microglia by electron microscopy was documented in a comprehensive review by Ling (8). Adding to this was the first ultrastructural evidence indicating that microglia shared fea- tures of phagocytes (9). Another step forward was a subsequent experimental study that demonstrated that microglia in the developing brain represent nascent brain macrophages; more importantly, they were derived from infiltrated blood monocytes (8, 10). The view that microglia in the developing brain were monocyte derived had since opened a new vista on the roles of microglia in the normal and diseased brain (11). This was followed by the immunophenotypic characterization of microglia using specific markers common to tissue macrophages (1215). Since then, studies on microglia specifically their roles in neuropathology have been extensively explored using differ- ent experimental models or paradigms (1618). The past 3 decades have seen an upsurge in microglia research and, in- deed, an exponential growth in our knowledge on microglia especially in connection with their roles in various From the Department of Anatomy, Yong Loo Lin School of Medicine, Na- tional University of Singapore, Singapore (CK, E-AL); and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin (GR) Send correspondence to: Charanjit Kaur, PhD, Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, Singapore 117594; E-mail: antkaurc@nus.edu.sg This study was supported by research grants R-181-000-148-750, R-181- 000-162-733, and R-181-000-173-112 from the National University Health System (NUHS), Singapore. The authors have no conflicts of interest to declare. 1 V C 2017 American Association of Neuropathologists, Inc. All rights reserved. J Neuropathol Exp Neurol Vol. 0, No. 0, 2017, pp. 1–18 doi: 10.1093/jnen/nlx056

Transcript of Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 ·...

Page 1: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

REVIEW ARTICLE

Biology of Microglia in the Developing Brain

Charanjit Kaur, PhD, Gurugirijha Rathnasamy, PhD and Eng-Ang Ling, PhD, DSc

AbstractMicroglia exist in different morphological forms in the develop-

ing brain. They show a small cell body with scanty cytoplasm with

many branching processes in the grey matter of the developing

brain. However, in the white matter such as the corpus callosum

where the unmyelinated axons are loosely organized, they appear in

an amoeboid form having a round cell body endowed with copious

cytoplasm rich in organelles. The amoeboid cells eventually trans-

form into ramified microglia in the second postnatal week when the

tissue becomes more compact with the onset of myelination. Micro-

glia serve as immunocompetent macrophages that act as neuropath-

ology sensors to detect and respond swiftly to subtle changes in the

brain tissues in pathological conditions. Microglial functions are

broadly considered as protective in the normal brain development as

they phagocytose dead cells and sculpt neuronal connections by

pruning excess axons and synapses. They also secrete a number of

trophic factors such as insulin-like growth factor-1 and transforming

growth factor-b among many others that are involved in neuronal

and oligodendrocyte survival. On the other hand, microglial cells

when activated produce a plethora of molecules such as proinflam-

matory cytokines, chemokines, reactive oxygen species, and nitric

oxide that are implicated in the pathogenesis of many pathological

conditions such as epilepsy, cerebral palsy, autism, and perinatal

hypoxic-ischemic brain injury. Although many studies have investi-

gated the origin and functions of the microglia in the developing

brain, in-depth in vivo studies along with analysis of their transcrip-

tome and epigenetic changes need to be undertaken to elucidate their

full potential be it protective or neurotoxic. This would lead to a bet-

ter understanding of their roles in the healthy and diseased develop-

ing brain and advancement of therapeutic strategies to target

microglia-mediated neurotoxicity.

Key Words: Antigen presentation, Developing brain, Hypoxia-is-

chemia, Inflammatory cytokines, Microglia, Phagocytosis.

INTRODUCTIONMicroglia are gaining tremendous recognition in recent

years because they are implicated in different neurodegenera-tive diseases and neurological disorders (1, 2). In the maturerodent brain, they contribute 5%–12% of the total glial popula-tion, and in reference to other glial cells, the frequency appearsto vary with regions, for example, in the cerebral cortex or cor-pus callosum (3). The prevailing view regarding roles ofmicroglia is that they act as resident immunocompetent phago-cytic cells in the disease processes in infectious, traumatic, in-flammatory, ischemic, and degenerative conditions of the cen-tral nervous system (CNS) (4). As far as can be ascertained,the existence of microglia as a separate cellular entity in thenormal brain or in pathological conditions was first docu-mented by Del Rio-Hortega (5) in the early part of the last cen-tury; yet compared with other glial types in the brain, it wouldappear that microglia had attracted little attention by the con-temporary authors until decades later (6, 7). It is possible thatthis might have been hindered by the lack of a reliable stainingmethod for identification of microglia during that period. Theweak silver carbonate staining method routinely used then forthis purpose was described to be capricious, often yielding un-satisfactory results. Despite the technical limitations, someearly authors had attempted to investigate the origin, develop-ment and roles of microglia. The study was later extended tothe ultrastructural identification of microglia in which the sil-ver carbonate staining was adapted for transmission electronmicroscopy (6, 7). The seminal report on the characterizationof microglia by electron microscopy was documented in acomprehensive review by Ling (8). Adding to this was the firstultrastructural evidence indicating that microglia shared fea-tures of phagocytes (9).

Another step forward was a subsequent experimentalstudy that demonstrated that microglia in the developing brainrepresent nascent brain macrophages; more importantly, theywere derived from infiltrated blood monocytes (8, 10). Theview that microglia in the developing brain were monocytederived had since opened a new vista on the roles of microgliain the normal and diseased brain (11). This was followed bythe immunophenotypic characterization of microglia usingspecific markers common to tissue macrophages (12–15).Since then, studies on microglia specifically their roles inneuropathology have been extensively explored using differ-ent experimental models or paradigms (16–18). The past 3decades have seen an upsurge in microglia research and, in-deed, an exponential growth in our knowledge on microgliaespecially in connection with their roles in various

From the Department of Anatomy, Yong Loo Lin School of Medicine, Na-tional University of Singapore, Singapore (CK, E-AL); and Departmentof Ophthalmology and Visual Sciences, School of Medicine and PublicHealth, University of Wisconsin, Madison, Wisconsin (GR)

Send correspondence to: Charanjit Kaur, PhD, Department of Anatomy,Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, NationalUniversity of Singapore, Singapore 117594; E-mail:[email protected]

This study was supported by research grants R-181-000-148-750, R-181-000-162-733, and R-181-000-173-112 from the National UniversityHealth System (NUHS), Singapore.

The authors have no conflicts of interest to declare.

1VC 2017 American Association of Neuropathologists, Inc. All rights reserved.

J Neuropathol Exp NeurolVol. 0, No. 0, 2017, pp. 1–18doi: 10.1093/jnen/nlx056

Page 2: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

neurodegenerative diseases and neurological disorders. Un-doubtedly, the roles of microglia are now better clarified andamplified, although they appear to be extremely complex ei-ther as neuroprotective or neurotoxic (4, 19).

ORIGIN OF MICROGLIAMicroglial cells have been reported to originate from

different mesodermal sources such as the embryonic mesen-chymal cells in the pia, fetal macrophages of the yolk sac orthe fetal liver depending on the embryonic age (20). The con-cept of mesodermal origin was first postulated by Del Rio-Hortega (5) based on congregation of pial cells stained withsilver carbonate in the superior tela choroidea, pia coveringthe cerebral peduncles and inferior tela choroidea in the em-bryonic brain. From these concentrations the cells theninvaded the brain and transformed into the round amoeboidmicroglial cells distributed throughout the brain. The pial(mesodermal) origin of microglial cells was supported bymany authors (21–25). Later studies supported further themesodermal origin of microglia using histochemical stainingwith isolectin Griffonia simplicifolia (GSA I-B4) and Ricinuscommunis agglutinin-1 (RCA-1) (26–28). Other studies havesuggested that precursor cells in the yolk sac give origin tomicroglia during the embryonic period (29). In the embryos ofchick and quail, macrophages in the CNS were reported to ori-ginate from the yolk sac before the circulation was established(30). It has been demonstrated that microglia in the avian em-bryonic brain were derived by invasion and proliferation ofmacrophages from the pial surface (31).

As opposed to the above views some authors, however,had argued in favor of a neuroectodermal lineage of microglia(32–35). Thus, the subependymal cells (glioblasts) in the sub-ventricular zone of lateral ventricles were thought to be theprecursors of microglia (32, 36, 37). With the use of RCA-1and monoclonal antibodies that recognize tissue macrophagesand microglia, it was proposed that microglia originate in thegerminal matrix rather than in the pial mesenchyme. However,others authors had contended that microglial precursors withinthe neuroepithelium were cells traversing the neuroepitheliumfrom the cerebral ventricles to enter the nervous parenchymaand settle as microglia (38).

The hypothesis that microglia are derived from circulat-ing monocytes was proposed by other workers including thepresent authors based on experimental animal studies (9, 10,39–42). However, the possibility that microglia are derivedfrom other sources, including the bone marrow or the myeloidtissue (43), cannot be excluded.

MICROGLIA IN THE DEVELOPING HUMANBRAIN

There is only a modicum of information addressing theissue of origin of microglia in the human brain. Almost allstudies regarding microglial origin and differentiation inhumans have been carried out in the fetal CNS tissue collectedat the time of elective or spontaneous pregnancy terminations(44–46). Microglia have been reported to colonize the humanbrain and spinal cord before 12th week of gestation (47, 48),

with limited numbers of amoeboid microglia being present inthese regions (45, 49). By using RCA-1 or antibodies such asCD 68 (EBM-11) that recognize human tissue macrophagesand microglia, it had been shown that the greatest number oflabeled cells between 13 and 18 weeks of gestation wasobserved in the germinal matrix (44) and in the white matter(50). During this period of development, the morphology ofmicroglia was of the amoeboid type in the germinal matrix,whereas they progressively differentiate into ramified cellswith the growth of the brain (44, 50) and disperse throughoutthe CNS. Andjelkovic et al had used immunoperoxidase,RCA-1, Lycopersicon esculentum (tomato lectin) and CD68 tolabel microglia in the brain tissue of human embryos andfetuses ranging from 4.5 to 13.5 gestational weeks of age (45).Based on the staining patterns, these authors believed that 2populations of microglia exist in the human brain during earlydevelopment and these may arise from 2 different sources—monocytes and the yolk sac—and progressively develop typicalmicroglia morphology. Ramified microglial cells have beenreported to be the predominant type in the infant brain (48).

AMOEBOID MICROGLIA AND RAMIFIEDMICROGLIA

In the search of the origin and mode of formation ofmicroglia in the corpus callosum in the mature brain (32), inwhich the cells were first identified and characterized ultra-structurally (6, 7), attention was first drawn to the investiga-tion of microglia in the same region of the pre (41) andpostnatal (9, 33, 51) rat brain (Fig. 1). Interestingly, microgliaas identified by their small cell body and scanty cytoplasmwith branching processes in the mature brain and termed“ramified microglia” were absent in the corpus callosum; in-stead, the same area composed of loosely organized andunmyelinated axons was occupied by a large number of roundand amoeboid cells that were also stained by the weak silvercarbonate staining (Fig. 2) (33). These cells termed“amoeboid microglia” exhibited cytochemical characteristicssuch as staining with acid phosphatase and nonspecific ester-ase shared by tissue macrophages (51, 52), thus alluding tothe possibility of them being monocytic in nature as in othertissue macrophages. With the progress of brain maturation,the amoeboid microglia transformed into ramified microglia(Fig. 2) within 2 weeks postnatally as evidenced by using afluorescent tracer, rhodamine isothiocynate (53), and specificmicroglia markers such as the antibody OX42 (which recog-nizes complement type 3 receptors) (14) and lectins (GSA I-B4, RCA-1, and tomato lectin) (54, 55) (Fig. 2). The trans-formation of amoeboid microglia into ramified microglia is aprocess that is coincident with the onset of myelination ofaxons in the corpus callosum (11, 56, 57). Electron micro-scopic studies have shown that amoeboid cells in 1- to 5-day-old animals possess a round nucleus with marginal chromatinclumps and abundant cytoplasm displaying lysosomal densegranules, vacuoles and a well-developed Golgi apparatus(58). Most of the cells in older animals were elongated andbranched, showing a flattened nucleus and scanty cytoplasmcontaining a few lysosomal granules (58) (Fig. 3). These cellswere immunoreactive with the antibody OX42 and showed

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

2

Page 3: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

labeling with lectin (Fig. 4) It is to be noted that most of thein vitro studies of primary microglia in later years by us aswell as by others have been based on microglia harvestedfrom neonatal rats, which presumably would represent pre-dominantly the amoeboid microglia (59, 60). Notwithstand-ing, in view of their developmental relationship of having thesame cell lineage and the existence of transitional forms, theyare regarded as and simply referred to as microglia in the pre-sent description.

MICROGLIA AS A NEUROPATHOLOGY SENSORIn the developing brain, microglia notably those prepon-

derant in the white matter, respond vigorously to lipopolysac-charide (LPS) and interferon-c (IFN-c) (61–63) injected intra-peritoneally into postnatal rats. They were induced to expressmajor histocompatibility complex II (MHC II) antigens withsome of the cells closely associated with the blood capillaries,indicating that they serve as immunocompetent cells in thedeveloping brain, which might interact with the circulatinglymphocytes. Microglia in the postnatal brain are also ex-tremely sensitive to hypoxic exposure. It was argued that theymay contribute to the periventricular white matter damage inthe developing brain in hypoxia (64). Either in the developingor mature brain, microglia act as a neuropathology sensor thatcan detect and respond swiftly to subtle changes in the braintissues, be it acute or chronic (11).

FUNCTIONS OF MICROGLIAAccumulating evidence in recent years support that

microglia are immune-regulatory cells that play importantroles in the healthy and diseased CNS. They help to maintainthe homoestasis of the brain environment under normal condi-tions but display robust reaction in response to inflammatorystimuli, injury, hypoxia-ischemia and other adverse conditionsaffecting the normal function of the brain. The activatedmicroglia assume an amoeboid phenotype, proliferate and mi-grate to the site of injury or damage where they execute pro-tective function involved in removal of dead cells and cellulardebris (65, 66). On the other hand, overactivation of microgliawith excess production of inflammatory mediators may resultin neurotoxic consequences (67, 68).

Protective RolesPhagocytosis

Microgia are active phagocytes that help to eliminatedegenerating axons and cells during early CNS developmentas well as during infections, injuries and other pathologicalconditions. They share many common features of peripheraltissue macrophages. The phagocytic nature microglia espe-cially on activation was evidenced in the developing brain asmany of these cells were found to contain phagosomes at theultrastructural level (Fig. 3). Additional evidence of their mac-rophagic nature was provided by the demonstration of hydro-lytic enzymes including acid phosphatase, aryl phosphatase,nonspecific esterase and 50-nucleotidase localized in them (52,69). Remarkably, microglia in the developing brain exhibitnonspecific esterase known to be specific to blood monocytes(52), thus alluding to for the first time their cytochemical linkto monocytes. The above-mentioned enzymes were found tobe localized in the lysosomes. These cells avidly engulfed ex-ogenous substances such as rhodamine isothiocyanate andhorseradish peroxidase (HRP) that leaked into the brain tissuewhen administered intraperitoneally or intravenously (70, 71)and biotinylated dextran administered directly into the brain(72). Ingestion of HRP by the microglial cells occurred fol-lowing injection of HRP in the lumbosacral region of the spi-

FIGURE 1. (A) A schematic diagram showing a representativecoronal section of the postnatal rat brain at the level of theoptic chiasma. Note the concentration of amoeboid microglia(circled area) in the corpus callosum (cc) above the lateralventricle (LV). Ramified microglia (squared area) aredistributed mainly in the cerebral cortex (cx). (B) A confocalimage of a coronal section of the brain of a 3-day-old ratstained with tomato lectin. The amoeboid microglia in thecorpus callosum (cc) are stained (green). Blood vessels in thecerebral cortex (cx) and other parts of the brain are alsostained (green). LV, lateral ventricle.

J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017 Microglia in the Developing Brain

3

Page 4: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

nal cord (73). E coli injected intracerebrally into the neonatalbrain were internalized by the microglial cells in<3 hours fol-lowing the injection (74). Recent investigations in the devel-oping cerebellum have provided further evidence for thephagocytic nature of these cells. Microglial cells were foundto engulf varying amounts of cellular debris in the neonatalcerebellum until postnatal day 17 (75). Microglial cells wereshown to limit the production of cortical neurons in the cere-bral cortex of prenatal and postnatal macaques and rats byphagocytosing the neural precursor cells (76). In our own stud-ies, we have encountered phagocytosis of dead cells and non-myelinated axons in the normal developing corpus callosumby electron microscopy (33, 58).

In fetal and postnatal rat brains, the microglial cellswere found to phagocytose necrotic and apoptotic cells as wellas degenerating axons following a hypoxic insult (77, 78).They tend to accumulate near dead neurons and clear dead ordying cells from the neonatal hippocampus following injury,thereby helping to limit secondary injury during the criticalearly time point following excision of hippocampal slices (79,80). In inflammatory lesions and neonatal stroke, microglialcells were found to be engaged in phagocytosis (81).

Microglial cells in the developing brain express com-plement type 3 receptors (CR3), which are known to beinvolved in endocytosis (14), similar to tissue macrophages.CR3-expressing microglial cells appeared to phagocytose a

FIGURE 2. (A) Amoeboid microglia in the corpus callosum of a 1-day-old rat are evidently labeled by the weak silver carbonatestaining. The cells are rich in cytoplasm, which appears to be vacuolated. The cells appear round or amoeboidic bearing shortprocesses. (B) Two ramified microglia in the cerebral cortex of a 5-day-old rat brain stained by weak silver carbonate stain. Notethe long branching processes. (C) OX42-labeled round amoeboid microglial cells in the corpus callosum of a 2-day old rat. (D)OX 42-labeled ramified cells in the cerebral cortex of a 2-day-old rat. (E) Lectin-labeled round, amoeboid microglial cells in thecorpus callosum of a 1-day-old rat. Blood vessels (asterisks) are also labeled. (F) Lectin-labeled ramified microglial cells in thecerebral cortex of 7-day-old rat.

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

4

Page 5: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

large number of apoptotic cells following an X-ray inducedinjury in the neonatal rat brain (82). Upregulation of CR3was observed on the microglial cells following intracerebralE. coli administration in postnatal rats (74). Besides CR3,microglia are known to express triggering receptorexpressed on myeloid cells-2 (TREM2), which is thought tobe involved in phagocytosis (83, 84). Recent studies in the

developing brain have reported that TREM2 was expressedon a subpopulation of microglial cells during first postnatalweek in grey and white matter (85), suggesting that it maybe related to phagocytosis of apoptotic cells. In the whitematter of the developing cerebellum, microglia express gan-glioside GD3 (86), which has been reported to play a partin phagocytosis of oligodendrocytes during CNS develop-ment. Toll-like receptors (TLRs) have also been shown tobe involved in microglial clearance of axonal debris ofdegenerating axons in microglial-axon coculture (87), and

FIGURE 3. (A) Electron micrograph of an amoeboid microgliain the loosely structured corpus callosum of a 5-day-old rat.Note the widely spaced unmyelinated axons. The abundantcytoplasm shows profiles of rough endoplasmic reticulum(rER), Golgi apparatus (G), lysosomes (Ly), a large phagosome(P) and some vacuoles (V). (B) Electron micrograph of aramified microglia in the corpus callosum of a 20-day-old rat.Note the closely packed axons some of them are evidentlymyelinated (Ax). The cell shows a small amount of cytoplasmwith a paucity of organelles and inclusions, which include afew lysosomes (Ly), Golgi apparatus (g) and profiles of roughendoplasmic reticulum (arrow).

FIGURE 4. (A) Electron micrograph of an amoeboid microglialcell showing OX42 immunostaining reaction products on theplasma membrane (arrows) in the corpus callosum of a 5-day-old rat. The cell shows a reniform nucleus (N), Golgi apparatus(G) and a phagosome (P). (B) Electron micrograph shows anelongated microglial cell whose plasma membrane is outlinedby the lectin GSA I-B4 staining (arrows) in the corpus callosumof a 10-day old rat. The cell contains a nucleus (N) bearingcoarse chromatin masses and some lysosomes (Ly).

J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017 Microglia in the Developing Brain

5

Page 6: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

their deficiency impairs phagocytosis of degenerating axonsby microglial cells. We have reported enhanced TLR4 ex-pression in microglia in neonatal rat brain following a hyp-oxic injury (88). Although the expression was related toneuroinflammation in response to hypoxia, it may also beinvolved in phagocytosis of degenerating axons and apop-totic cells which are seen frequently in different parts ofthe developing brain such as the hippocampus, cerebellum,and the corpus callosum (78, 89) (Fig. 5).

Antigen Presentation

Macrophages, dendritic, and other cells belonging to theimmune system capture foreign antigens, process them andpresent them to T lymphocytes during initial stages of an im-mune response. Major histocompatibility complex I (MHC I)and MHC II molecules on the surface of macrophages andother cells (90–92) mediate antigen presentation.

The CNS has been considered historically as an im-munologically privileged site for a long time based on thepresence of blood-brain barrier (BBB), lack of lymphatics andan absence of antigen presenting cells. This concept has beenchanged in recent years in view of the expression of MHC Iantigens, involved in presentation of foreign antigens to cyto-toxic T lymphocytes, in microglial cells in the developingbrain (15). Expression of MHC II antigens responsible forpresentation of a foreign antigen to helper T lymphocytes,however, was not found on these cells under normal condi-tions. When stimulated with LPS, IFN- c, or E-coli, the ex-pression of MHC II antigens was induced on microglial cellsin the developing brain (61, 62, 74). Expression of MHC II hasalso been reported on microglia in the normal and pathologicalhuman fetal spinal cord (93). MHC I/II antigen expression onmicroglial cells has also been reported following perinatalhypoxia (94). The expression of these molecules providesevidence for the protective role of microglial cells in thedeveloping brain as BBB is immature and leaky and, indeed,vulnerability to a potential immune threat is feasible. Disrup-tion of the BBB has been reported in neonatal meningitis (95),hypoxic-ischemic injuries (96) and in excitotoxic lesions (97),and increased numbers of CD4-positive T cells in the brainhave been noted in the white matter of brains of mice sufferingfrom chronic perinatal hypoxia (98).

Synaptic Pruning and Remodeling

It has been suggested that microglia play an importantrole in regulating synaptic function through elimination ofsynaptic connections and their involvement in maturation ofsynapses in the developing brain. Studies have shown thatmicroglia are involved in synaptic pruning and remodelingduring postnatal brain development (99, 100) and impairedmicroglia function results in delayed maturation of synapticcircuits in certain regions of the brain such as the hippocampus(99). Complement 3 (C3) molecules are localized on thedeveloping synapses and microglia specific phagocytic path-way, CR3/C3-dependent signaling, is believed to be one of theunderlying mechanisms in synaptic pruning and remodeling

of developing synapses by microglia (100, 101). In vitro stud-ies suggested that interleukin-10 signaling may be involved inpromoting synapse formation by microglia (102). Depletion ofmicroglia from the mouse brain had resulted in alterations insynaptic protein levels and glutamatergic synaptic function(103). Besides CR3/C3 cascade, MHC class I molecules mayalso be involved in developmental microglia-mediated prun-ing or refinement of synapses (104–107).

Role in Development of Various Cells in the Brain

Microglia can regulate the number of neurons in thedeveloping brain by phagocytosis of dead or dying cells andprovide trophic support to the neural progenitor cells for theirproliferation and maturation (108–110). In vitro studies have

FIGURE 5. (A) Electron micrograph of an apoptotic cell (Ap) inthe hippocampus of a 2-day-old hypoxic rat showing crescentshaped chromatin condensation at the periphery of thenucleus. (B) An amoeboid microglial (Am) cell in the corpuscallosum of a 2-day-old hypoxic rat showing lysosomes (Ly)and some vacuoles (asterisks) in the cytoplasm. The amoeboidmicroglial cell is in the process of phagocytosing an apoptotic(Ap) cell with electron dense nuclear chromatin condensation.Reproduced from Kaur and You (89), with permission fromElsevier.

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

6

Page 7: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

shown that depletion of microglia or a deficiency of the che-mokine receptor, CX3C chemokine receptor 1 (CX3CR1),also known as fractalkine receptor, in microglia increases thenumber of apoptotic neurons in the cerebral cortex in normalbrain (111). Microglia-derived insulin-like growth factor-1(IGF)-1 was also identified as a trophic factor involved inmaintenance of neuronal survival. Production of factors suchas IGF-1, nuclear factor-kappaB (NF-jB), interleukin (IL)-1b,and IL-6 by microglia is thought to play a part in the survival,differentiation and maturation of oligodendrocytes (112–114).Using cultures of neural progenitor/stem cells obtained fromrat embryonic day 16 subventricular zone and microglial cellsfrom I day old rat cortex, Nakanishi et al (115) have shownthat microglia-derived IL-6 and leukemia inhibitory factor areessential molecules for differentiation of neural progenitorcells into astrocytes.

In addition to the above, microglial cells in the develop-ing CNS have also been suggested to regulate vascularization(110, 116) and influence myelination (117).

Angiotensin and Angiotensin Receptors

The renin angiotensin system (RAS) regulates bloodpressure and electrolyte homeostasis.

Renin produced by the juxtaglomerular cells of the kid-ney converts angiotensinogen to angiotensin I, which is thenconverted to angiotensin II (Ang II) by the angiotensin-converting enzyme. Ang II is the most active peptide of theRAS and binds to 2 receptors, Ang II type 1 (AT1R) and type2 (AT2R). Most of the physiological or pathophysiologicalfunctions of Ang II are carried out through its binding withthese receptors (118, 119). Several studies have reported theexistence of various components of the RAS in the brain (120)and have also reported that this system is involved in the regu-lation of fetal cardiovascular responses, body fluid balance,and neuroendocrine regulation (121). We have reported thatAng II, AT1R, and AT2R are expressed on microglial cells inthe neonatal rat and the expression is sustained in the maturebrain (122). The expression of Ang II and AT1R was reducedfollowing a hypoxic injury whereas the expression of AT2Rwas increased (122). Since hypoxic insults to the brain areknown to reduce the blood flow to the various regions of thebrain and induce BBB dysfunction (123, 124), decreased AngII and AT1R expression may be helpful in restoring the bloodflow in such conditions. AT1R has also been reported to playa part in in apoptosis, oxidative stress, and neuroinflammation,and its suppression with AT1R blockers reduces inflammation(125) and suppresses apoptosis (126). An enhanced expressionof AT2R in the microglial cells following a hypoxic injurywas suggested to be neuroprotective (122).

Production of Neuroprotective FactorsEndothelins

The family of endothelins (ETs), considered tradition-ally as potent vasoconstrictors, consists of 3 isopeptides, ieendothelin-1 (ET-1), endothelin-2, and endothelin-3 (127,128), which bind to 2 specific G-protein-coupled receptors

subtypes, ET-A and ET-B receptors (129, 130), to carry outtheir actions. Endothelial cells, monocytes and macrophagesare important sources of circulating ET-1 (131). In addition toits vasoconstrictor action, ET-1 exerts mitogenic and anti-apoptotic actions (132, 133). Microglia in the periventricularwhite matter in the developing brain express ET-1 (134),which was suggested to be involved in the proliferation anddifferentiation of glial precursors under normal conditions.Hypoxic injury resulted in a decreased production of ET-1 bythe microglial cells with implications that its reduction mayadversely affect the development of glial cells (134). How-ever, ET-1 appears to be a double-edged sword with severalstudies reporting its beneficial effects as mentioned abovewhereas others argued that its increased expression iscorrelated with vasogenic edema formation via BBBdisruption (135).

Insulin-Like Growth Factors

Microglial cells produce IGF-1 and -2 (136, 137) thatare known to regulate the development of the nervous system(138) by promoting cell proliferation and differentiation (139)especially oligodendrocyte survival and myelination (140–142). In a coculture of primary microglia and neurons,microglia-derived IGF-1 favored the survival of neurons(111). IGF-1 is known to play a significant role in recoveryfrom hypoxic-ischemic insults (143) by blocking tumor necro-sis factor (TNF)-a-induced apoptosis and promoting prolifer-ation/differentiation and survival of oligodendrocyte precur-sors (144, 145). However, when proinflammatory cytokinesare overexpressed the secretion of IGF-1 by microgliadecreases, which could cause deleterious effects (146).

Transforming Growth Factor-b

Transforming growth factor-b (TGF-b) family plays animportant role in embryogenesis determining the right-left or-ganization, axis formation and tissue patterning. All 3 iso-forms of TGF-b, (TGF-b1-3), are expressed in the brain andare essential for neuronal and glial differentiation (147, 148).Following hypoxic exposure, TGF-b1 mRNA expression wasincreased in the periventricular white matter of neonatal rats.In these animals, TGF-b1 expression was localized in themicroglial cells (149). In TGF-b1 knockout mice there wasincreased neuronal loss and microglial activation (150), sug-gesting a role for TGF-b1 in regulating microglial activationstatus. Interestingly the microglial cells also expressed recep-tors for TGF-b (149), which supports the idea of TGF-b-medi-ated regulation of microglial functions. This could include theability of TGF-b to decrease the expression of MHCII, inter-cellular cell adhesion molecule 1, vascular cell adhesion mol-ecule and TNF-a (151), which are known to be expressed inactivated microglia. TGF-b is known to induce the productionof nerve growth factors by acting through its receptorsexpressed by the brain parenchymal cells (152). It is highlypossible that TGF-b could do the same in microglia by actingin an autocrine manner. Moreover, TGF-b1 was recently dem-

J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017 Microglia in the Developing Brain

7

Page 8: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

onstrated to have a role in the synapse formation in developingcerebellum (153).

Neurotrophins

Microglia also serve as a source for neurotrophins suchas nerve growth factor (NGF), brain-derived neurotrophic fac-tor (BDNF) and neurotrophin-3 (NT3) (154–157). While un-stimulated cultured microglia could express mRNAs for allthese neurotrophic factors mentioned above, stimulation withLPS up-regulates the mRNA expression of these factors (158).These neurotrophins are essential for the survival, differenti-ation and proliferation of the brain parenchymal cells duringpostnatal development (159), and they exhibit their action byeither activating the tyrosine kinase B (TrkB) receptors or thep75 neurotrophin receptors (160–162). While activation ofTrkB receptors could result in enhanced survival, activation ofp75 neurotrophin receptor has been linked to neuronal apop-tosis (163). In cocultures of microglia and dorsal root ganglionneurons, microglia-derived BDNF promotes neurite outgrowthand terminal contacts of dorsal root ganglion neurons (164).BDNF derived from ethanol treated microglia was shown toprevent apoptosis of hypothalamic neurons through mecha-nisms modulating reactive oxygen species (ROS) and cAMPresponsive element binding protein pathway (165). Addition ofconditioned medium from neurons to the microglial culturesfavors the production of neurotrophins in microglia (157). Inmicroglia cell lines, BV2 and N9, BDNF and NGF enhancetheir proliferation in a concentration dependent manner (166).Consistent with this, under inflammatory conditions, intranasaladministration of BDNF was found to increase the number ofactivated and phagocytic microglia at the site of injury (167).Similar to TGF-b, these neurotrophins could inhibit the expres-sion of MHC II in the activated microglial cells via the p75 neu-rotrophin receptor (160). Taken together these studies putforward the premise that brain injury could trigger neurotrophinproduction in microglia in order to enhance the survival of neu-rons. In turn, the neurotrophins could act in an autocrine fashionon microglia and inhibit inflammation providing a conduciveenvironment for the survival of neurons at the injured site.

Neurotoxic RolesBesides their neuroprotective functions, microglial cells

are also known to play detrimental roles under various patho-logical conditions when they produce a plethora of moleculessuch as proinflammatory cytokines, chemokines, reactive oxy-gen species (ROS), and nitric oxide (NO), thus causingneurotoxicity.

Role in Neuroinflammation

Proinflammatory molecules are expressed at higher lev-els in the developing than in the mature brain in the absence ofany pathology. Thus, it is suggested that they may have an im-portant role in CNS development. Involvement of cytokinessuch as TNF-a and interleukin (IL)-1b in developmental proc-esses such as neural cell migration, proliferation, differenti-ation, and death has been reported (168), whereas IL-6 has

been shown to contribute to development of the vasculature(169, 170). In CNS injury, microglial cells release augmentedamounts of proinflammatory cytokines which when sustainedover a long period can cause damage to the developing braincells. TNF-a and IL-1b are released in varying amounts bymicroglial cells in different areas of the developing brain fol-lowing a hypoxic injury (67, 68, 171, 172). Upregulation ofTNF-a and IL-1b in microglial cells was coupled with expres-sion of their respective receptors TNF-R (1) and IL-1R (1) onoligodendrocytes in the periventricular white matter and thePurkinje neurons in the developing cerebellum (67, 68). Thebinding of these cytokines to their respective receptors wassuggested as a mechanism leading to cell death. In neonatalrats subjected to a hypoxic exposure iron accumulation wasincreased in the microglial cells and this was shown to mediateaugmented production of TNF-a, IL-1b, and ROS (172, 173).In addition to the above, cyclooxygenase-1 (COX-1),cyclooxygenase-2 (COX-2), microsomal prostaglandin-E syn-thase, E-prostanoid receptor 2, and prostaglandin E2 (PGE2)expression was increased in microglia in the hypoxic develop-ing brain (174). In connection with this, PGE2 was found to beinvolved in the regulation of TNF-a and IL-1b production.Increased accumulation of microglial cells occurred in theperiventricular white matter in hypoxic neonatal rats throughexpression of monocyte chemoattractant protein (MCP)-1 andaugmented the inflammatory response (175). Macrophagecolony-stimulating factor (M-CSF) is another cytokinederived from microglia that promoted proinflammatory cyto-kine production by other glial cells such as the astrocytes inthe periventricular white matter following hypoxic exposure,which added to the white matter damage induced bymicroglia-derived inflammatory cytokines (176). TLRs havebeen reported to play a role in immune responses. Microgliaalso express TLR4 whose expression is enhanced in hypoxicinjuries and mediates neuroinflammation via NF-jB signalingpathway through production of TNF-a, IL-1b, inducible nitricoxide synthase (iNOS), ROS, and NO (88).

Microglia in the developing brain have also been dem-onstrated to be activated by alcohol in conditions such as thefetal alcohol syndrome. In this regard, ethanol triggers produc-tion of ROS and inflammatory cytokines as well as phagocyt-osis through activation of TLR2 and TLR4 signaling inmicroglia (177–179). Although alcohol-induced abnormalitiesin glial cells have been implicated in the adverse effects ofalcohol on the developing brain (180), more work is clearlydesirable to unravel the underlying molecular mechanisms.

Following a maternal injection of the teratogen cyclo-phosphamide to induce neural tube defects, the microglialcells in the fetal brain showed a marked increase in the levelsof TNF-a and TGF-b expression (181, 182). It was suggestedthat upregulation of proinflammatory cytokines caused bycyclophosphamide may be the underlying cause of increasedrate of neural tube defects. Increased expression of M-CSF bythe microglia and a concomitant increased expression of M-CSF receptor in these cells following the teratogen-inducedneuronal injury suggested that microglia are capable ofresponding to self-derived M-CSF in an autocrine fashion thatresults in cell proliferation and a proinflammatory response(183). This observation is supported by several studies that

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

8

Page 9: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

have reported that M-CSF receptor expression induces micro-glial proliferation, cytokine expression, and a paracrine in-flammatory response in brain pathologies (184, 185).

Accumulation of glutamate in hypoxic conditions in thedeveloping brain results in activation of N-methyl D-aspartatereceptor (NMDAR) and a-amino-3-hydroxy-5-methyl-4-iso-xazolepropionic acid receptors (AMPAR) on microglial cells(137, 186). Increased accumulation of glutamate enhanced theproduction of TNF-a and IL-1b by microglia (137). Further-more, NMDAR expression in microglia resulted in TNF-a andIL-1b release via NF-kB signaling pathway in the periven-tricular white matter of neonatal rats following hypoxia (186).In addition, activation of glutamate receptors also led to theproduction of excessive amounts of NO through the iNOS iso-form in the microglial cells (186). Activation of the microglialNMDAR triggers inflammation and plays a pivotal role inneuronal death in the perinatal and mature brain (187)

Notch signaling pathway is a key regulator of neurogen-esis in the CNS (188) and administration of Notch ligands inthe brain after ischemic injury has been reported to expandstem cell numbers and improve motor skills (189, 190). On theother hand, activation of Notch signaling and Notch pathwayhas been reported to be associated with exaggerated inflamma-tion in many tissues (191), and inhibition of this pathway hasbeen shown to ameliorate the severity of conditions such asautoimmune encephalomyelitis (192) and experimental auto-immune uveoretinitis (193). We have shown that microglia inthe developing brain express Notch-1 receptor, which wascolocalized with its ligands, Jagged-1, and Delta-1 (194).Notch-1 receptor expression was increased in the microglialcells in postnatal rats challenged with LPS and its suppressionresulted in increased levels of TNF-a released from microglialcells, suggesting that Notch signaling pathway plays an im-portant role in neuroinflamnation (194).

Apart from cytokines, activated microglia also secretechemokines that mediate the infiltration of leukocytes to thesite of injury. LPS treatment increases the levels of chemo-kines such as interferon-c-inducible protein-10, regulated onactivation normal T expressed and secreted (RANTES), MCP-1, macrophage inflammatory protein (MIP)-1a (MIP-1a, alsocalled CCL3), and MIP-1b in human fetal microglia (195,196). In neonatal rat, following hypoxic ischemic injury, therewas increased expression of MIP-1a/CCL3 in microglia(197). Hypoxic microglia in the presence of syndecan-2released excessive amounts of CCL2/MCP-1, CXCL12 (alsocalled stromal derived factor-1 [SDF-1]) (198). Infection ofthe prenatal rodent brains in utero with cytomegalovirus alsoupregulated the expression of chemokines such as CCL2/MCP1, CCL3, CCL4/MIP-1b, CCL7/MCP-3, and CCL12/MCP-5 in microglia (199), and all of the above-mentionedchemokines have been demonstrated to attract peripheralmonocytes into the brain and exacerbate the injury. In additionto secreting chemokines, microglia also express chemokinereceptors that influence their activation. One of the chemokinereceptors expressed by microglia during development includeCX3CR1, a receptor for neuronal chemokine fractalkine(200). The fractalkine-CX3CR1 signaling is implicated in thedevelopmental events carried out by microglia, such as phago-cytosis and synaptic pruning (201). Similarly, microglial

migration during cortical development is also dependent onthe chemokine CXCL12/SDF-1. This is facilitated in micro-glia through the expression of SDF-1 receptor, CXCR4. Che-mokine signaling in an injured brain could also be detrimental.For instance, following hypoxic-ischemic injury in neonatalrodents, the neurons at the site of injury express MCP-1 that isreported to facilitate the migration of microglial cells to theinjury location (202), which might exaggerate the injury byproducing inflammatory mediators. In these animals ,the ex-pression of MIP-1a and MIP-1b, in the infarcted region of thebrain, preceded the microglia/monocyte accumulation (203).

Role in Oxidative Stress

Microglia-derived free radicals or ROS is a common ob-servance following an injury to the brain. Free radicals such assuperoxide, hydrogen peroxide, etc. are unstable moleculesthat could initiate oxidative stress. Under physiological condi-tions, they are neutralized by the antioxidant defense system.However, in the immature brains there is a lack of antioxidantsystem to combat the excessive oxidants (204, 205). Hence ex-cessive generation of free radicals in the developing braincould cause damage to the susceptible cells (ie oligodendro-cytes [206] and neurons) by causing lipid peroxidation (207)and by damaging the myelin sheath (208). As aforementioned,microglial activation could also lead to excess production ofiNOS and subsequent NO, which is also a free radical. NO hasmultiple roles depending on the cellular source of origin, andNO produced by iNOS is considered detrimental. The mostharmful effect of NO is observed when it reacts with super-oxide to produce the highly reactive peroxynitrite, which couldresult in DNA strand breaks, lipid peroxidation, and protein ni-tration. In developing periventricular white matter, followinghypoxic exposure the expression of iNOS was predominant inthe microglia, suggesting them to be the source of NO (78). Incultured microglia, hypoxic exposure increases the generationof reactive oxygen as well as reactive nitrogen species (172),including NO (186), and their subsequent release into the cul-ture medium. In primary oligodendrocytes treated with condi-tioned medium from hypoxic microglia there was increasedlipid peroxidation with a parallel reduction in the glutathionecontent (172). However, antioxidant drugs such as edaravonecould inhibit free radical production by microglia and renderprotection to the immature brains (198). Taken together, all ofthese studies provide converging evidence for the damagingrole of activated microglia in an injured immature brain.

SIGNALLING PATHWAYS IN MICROGLIAAssociated with the production of various proinflamma-

tory mediators, activated microglia showed upregulated ex-pression of JNK and p38 MAPK pathways (209). Moreover,increased NF-jB/p65 expression was a consistent feature(210), which was to be expected given the fact that the cellsproduce significant amount of iNOS and NO. JAK-STATpathway is also implicated in the production of pro-inflammatory molecules, iNOS and NO in N9 microglialcells (211). In addition, activation of JAK/STAT1 pathwaywas reported to favor migration of microglia towards the

J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017 Microglia in the Developing Brain

9

Page 10: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

injury site in a stab wound mouse model (212). Hence, it ispossible that depending upon the STAT subunit that is acti-vated, it might have an influence on the functions of microglia.The most unexpected finding was the detection of Notch-1signaling in activated amoeboid microglia including its down-stream elements when challenged with LPS both in vivo andin vitro (88, 193, 213, 214). Even more striking was the dem-onstration of a reciprocal transactivation between Notch-1 andNF-jB in activated amoeboid microglia in regulation of pro-duction of proinflammatory mediators (209, 215). Microgliaalso produce pro-inflammatory molecules through activationof receptors for recognizing molecular patterns that are associ-ated with pathogens or any type of danger. TLRs are one ofsuch receptors expressed by amoeboid microglia and activa-tion of TLRs might lead to inflammation either via the NF-jBpathway (88) or the interferon regulatory-3 (IRF3) signalingpathway (216). The other pattern recognition receptors thatare not well studied in amoeboid microglia include thenucleotide-binding oligomerization domain (nod)-like recep-tors, and the retinoic acid-inducible gene-1-like receptors.Similar to TLR, activation of these pathways could also leadto inflammation by converging at either the NF-jB or IRF3signaling pathways; however, further studies are required toconfirm this in microglia in the developing brain. TREM2 sig-naling pathway is a pathway through which microglial innateimmune response is controlled in the postnatal brain (85) anda favorable environment is ensured for myelination and nor-mal development (217). Suffice it to say that the above mayrepresent only a few of the many signaling pathways that regu-late microglial activation (4).

RELEASE OF GLUTAMATE AND GLUTAMATERECEPTOR EXPRESSION

Glutamate is the most predominant and vital excitatoryneurotransmitter in the immature brain, that exerts its actionby binding through its receptors, which are either ionotropic(AMPAR, NMDAR, kainate receptors) or metabotropic recep-tors (mGluR) (218–220). In the developing brain, it aids in theearly development by signaling for proliferation, differenti-ation and migration of neurons. However, in the injured imma-ture brains, excessive levels of glutamate are detrimental andactivated microglia seem to be one of the sources of glutamate(137, 221–223). For instance, production of glutamate was ap-parently increased in cultured microglia treated with eitherLPS or sodium arsenite or infected with virus (222, 224). Thesame was observed even in microglial cultures subjected tohypoxia (137). In addition to releasing glutamate, microglialcells express receptors for glutamate (185, 225). In the whitematter of neonatal rat brain following hypoxic injury, micro-glial cells were found to concomitantly overexpress bothAMPAR (GluR2-4) and NMDAR subunits (NR1, NR2A-D)(137, 186). Overactivation of these receptors instigates theproduction of pro-inflammatory cytokines such as TNF-a andIL-1b along with other toxic factors such as NO and Fas L,which are known to cause oligodendrocyte and neuronal apop-tosis. Addition of glutamate or kainate to the microglialcultures enhances the release of TNF-a through activation ofeither AMPAR or kainate receptors (226, 227). Along with

TNF-a and IL-1b, hypoxia-induced activation of microglialNMDAR increases the expression of iNOS and subsequentproduction of NO (186). Even activation of mGluR2expressed in microglia could enhance the release of cytokinesand Fas ligand. All of these toxic factors act upon the highlysusceptible cells of the brain, such as the oligodendrocytes andneurons, and cause their death by activating caspase-3 (172,186, 228). While activated microglia could also produceneurotrophic factors such as IGF-I and IGF-II, excess glutam-ate suppressed IGF-I levels and delayed the repair mecha-nisms (137). The proinflammatory cytokines released frommicroglia could inhibit the glutamate reuptake by astrocytes(229), further complicating the entire injury mechanism. Thusexcess glutamate released by activated microglia could be det-rimental to oligodendrocytes and neurons, either by directbinding to their receptors expressed on these cells, or indir-ectly by enhancing the release of toxic factors and suppressingthe release of neurotrophic factors from microglia.

MICROGLIAL ACTIVATION IN VARIOUSDISORDERS OF THE DEVELOPING BRAIN

Periventricular White Matter DamageMicroglia in the amoeboid form accumulate preferen-

tially in the periventricular region in the corpus callosumabove the lateral ventricles and the subependyma (33, 53),cavum septum pellucidum (230, 231), and also the subpendy-mal cysts associated with the ventricular system (231, 232).The significance of this remains speculative, but it was sug-gested that microglia in these areas may be involved in clear-ance of cellular debris resulting from spontaneous cell deathor remodeling of callosal axons (33) or early physical expan-sion of the developing brain (231). In view of their closeassociation with the fiber tracts, it is conceivable that whenactivated by hypoxia (64), ischemia or septicemia (233), ex-cess amounts of proinflammatory mediators released by themwould affect the structural and functional integrity of the fibertracts. There is evidence supporting the involvement of acti-vated microglia in aggravating death of immature oligoden-drocytes and degeneration of axons in the periventricularwhite matter in neonatal rats following a hypoxic insult (64,78). We have demonstrated that the pathogenesis of periven-tricular white matter damage is multifactorial involving fac-tors such as inflammation, excitotoxicity, excess production ofNO, iron related oxidative stress, and vascular changes amongmany others (64, 67, 78, 172, 175, 186). The involvement ofsome of the above factors in periventricular white matter dam-age in hypoxic injuries has received support from other studies(223, 234–238).

Cerebral PalsyCerebral palsy is a motor disorder due to brain white

matter damage that appears in infancy or in early childhoodand affects movement, muscle tone, coordination, and bal-ance. Most cases are congenital as the damage to thebrain white matter occurs before birth. Intrauterine infection/inflammation, premature birth, and low birth weight have beensuggested as the underlying causes leading to cerebral palsy. Pre-

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

10

Page 11: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

clinical studies subjecting animals to hypoxia–ischemia or LPShave shown axonal damage and oligodendrocyte death in thecerebral white matter as well as neuronal damage in many regionsof the developing brain (78, 239–241). Microglia are present inlarge numbers in the developing white matter tracts and arebelieved to regulate the development of the white matter byremoving excessive axons in the developing brain (242). Postmor-tem studies in the human newborns have implicated cytokinessuch as TNF-a and its receptors TNFR1 and TNFR2 as the majorplayers in causing damage (243, 244). Activation of microglialcells in the white matter and other regions of the brain andenhanced release of TNF-a, IL-1b, and other proinflammatorymolecules such as release of NO through iNOS by them has beenreported in various animal models (67, 172, 186, 245, 246). Be-sides their inflammatory role, microglia may also have a protect-ive role in early stages by phagocytizing dead cells and degenerat-ing axons (78). Microglial activation and accompanying COX-1enzyme expression have also been implicated in overproductionof proinflammatory mediators resulting in white matter damageand development of cerebral palsy (247). Excitotoxic mechanismshave also been suggested to play a role in neuronal death (248)and in axonal damage in the developing brain and this is sup-ported by expression of NMDAR on premyelinated axons inhuman mid-term fetuses (249) and on the microglial cells in theneonatal rat brains (186).

EpilepsyEpilepsy is a disorder in which seizures develop due to an

imbalance between cerebral excitability and inhibition resultingin uncontrolled excitability. Seizures have been reported to de-velop more readily in the immature brain than in the maturebrain. Many developmental processes such as cell division, mi-gration, expression of receptors, formation, and stabilization ofsynapses are affected by seizures (250, 251). Temporal andfrontal lobes are frequently involved. Premature birth or lowbirth weight, birth trauma, fever or infections are some of theunderlying causes of epilepsy in newborns and infants. Glial ac-tivation and neuronal damage occurring in seizures has beenreported in brain regions such as the hippocampus (252). Severalstudies have shown that proinflammatory molecules influencesusceptibility to seizures (253–255). Enhanced levels of TNF-aand IL-1b were reported to play a role in the pathophysiology ofepilepsy (256). Microglial activation in the postnatal mousehippocampus was linked to heightened seizure susceptibility(257). Upregulation of TNF-a and IL-1b in microglia was read-ily detected after the induction of acute seizures (257, 258).Binding of IL-1b to its receptor in the hippocampus has beensuggested to result in increased synaptic excitability (259). TNF-a affects neural circuits by preventing refinement and elimin-ation of synapses occurring during normal brain developmentthat may lead to increased connectivity and epilepsy (260).

Autism Spectrum DisordersAutism spectrum disorders (ASD) are neurodevelop-

mental disorders with disabilities in communication and socialskills and repetitive behaviors. Genetic factors, prenatal drugs,or chemical exposures are some of the risk factors for develop-

ment of autism. Involvement of microglia due to their role insynaptic refinement during development has been suggestedin these disorders (261). Deficient synaptic pruning due to areduction in microglial numbers has been associated withweak synaptic transmission and decreased functional brainconnectivity (262) that may be involved in autism and otherneuropsychiatric disorders. Cerebellar dysfunction and subse-quent thalamic hyperactivation in early childhood may berelated to the development of ASD (263). Thalamic hyperacti-vation is thought to be induced by microglia-mediated neuro-inflammation (263), and neuroinflammation is considered asan important factor in the pathogenesis of neuropsychiatricdisorders (264, 265). Microglia activation and proliferation,along with increased expression of TNF-a, IL-1b, IL-6, andIL-17 in the brain and cerebrospinal fluid of ASD patients, hasbeen observed (266, 267). Focal inflammation due to abnor-mal microglial activation and proliferation seems to affectnormal synaptic activity in brains of patients with ASD (268–270). Along with increased microglia activation, decreasedneuronal activity was observed in human brain cortical tissuesamples from ASD patients (267). The above findings pointtowards the important role that microglia play in the pathogen-esis of ASD (271, 272). Interactions between mast cells andmicroglia in the brain have been reported to occur as evi-denced by induction of microglia activation and proliferationby mast cell-derived histamine (273, 274) that amelioratedwith suppression of mast cells (275).

Perinatal StrokePerinatal stroke occurs in newborns in the first few days

after birth due to a disturbance in the blood supply to the brainresulting in hypoxia and leads to significant morbidity andlong-term neurological and cognitive deficits. Blood clottingdisorders, maternal infection, preeclampsia, maternal diabetesand smoking are some of the risk factors that can lead to aperinatal stroke. In the rodent model of neonatal stroke aftertransient middle cerebral artery occlusion, microglial cellshave been reported to have a neuroprotective function. Deple-tion of microglia by intracerebral injection of liposome-encapsulated clodronate in rats at postnatal day 5 has beenshown to trigger hemorrhages at 24 hours after transient mid-dle cerebral artery occlusion (276). Along with this, increasedlevels of several cytokines and chemokines already elevatedby ischemia-reperfusion and increase in the severity and vol-ume of injury were demonstrated in animal models of neonatalstroke following depletion of microglia (81). These findingspoint towards a protective role exerted by microglia during thesubacute injury phase. However, other studies have reportedthat infiltration and activation of microglia in the brain of post-natal rats after neonatal stroke occurs over several days result-ing in neuroinflammation and cell death (277).

EPIGENETICS AND TRANSCRIPTOME ANALYSISA recent search for the interacting noncoding RNAs,

genes, and their epigenetic changes, (epigenetics refers to theheritable changes in the mechanisms that regulate the gene ex-pression without altering the underlying genetic code), has

J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017 Microglia in the Developing Brain

11

Page 12: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

brought into light several unknown aspects of microglia,including their involvement in disorders that were thought tobe microglia independent. By cDNA microarray analysis,amoeboid microglia were found to express proliferation- anddifferentiation-related genes, Sox4, Sox11, and Runx1t1 andalso those genes involved in cell cycle process and migration(278). Interestingly, with the ramification of the cells withage, the expression of various genes was altered and this maybe related to the specific functions of the 2 phenotypes. Micro-glia also express a neuroprotective and inflammatory pheno-type, which depends on the environmental cues. For instance,transcriptomic analysis in postmortem brain samples from aut-istic children revealed the transformation of microglia towardsthe inflammatory phenotype (279). Genes of immune path-way such as C1qA, C3, CR3, and TNF-a, which are expressedby microglia and transcription factors such as IRF8 andSPI1, that are essential for development of microglia werefound to be methylated in autistic brains (280). Additionalevidence for the epigenetic regulation in microglia and its in-fluence in the childhood disorders comes from microglia spe-cific methyl cytosine binding protein 2 (MeCP2) knockoutmice. Of note, MeCP2 regulates gene transcription by bind-ing to the methylated cytosine and guanine rich sites (CpGislands), and its reduced expression has a role in the etiologyof autism (281). Deletion of MeCP2 in mice contributed tothe dysregulation of extracellular glutamate levels and aber-rant neuronal dendrites (282). In primary cultures of micro-glia from neonatal rodents, hypoxia mediated reduction inthe microRNA21 expression accounted for the increased pro-duction of Fas ligand that induces neuronal injury (283).microRNA124 is specifically expressed in microglia and itsuppresses the expression of MHCII antigens. Loss of micro-RNA124 precedes the onset of experimental autoimmune en-cephalitis (284), which explains the enhanced autoimmunityobserved. Prenatal stress has been shown to increase themethylation of genes such as those for glucocorticoid recep-tors elsewhere (285); however, at this juncture there is noclear evidence if the same is seen in microglia of prenatalbrains, which could be linked to the increased inflammatoryphenotype that is observed under injury conditions. All thesestudies converge to a point, that epigenetic regulations occur-ring in microglia could have a greater impact on the normalbrain development as well as various disorders of the devel-oping brain.

Concluding RemarksMicroglia in the developing brain exist in 2 different

phenotypes: amoeboid microglia and ramified microglia.Amoeboid microglia are monocyte-derived brain macro-phages, that evolve to become the ramified microglia in thecourse of brain development. Microglia play an indispensablerole in building the normal brain histoarchitecture includingphagocytosis of apoptotic neurons and axons, and pruning ofunwanted synapses in brain remodeling. They secrete neuro-trophic factors such as IGF-1, NT-3, BDNF, and NGF; henceare neuroprotective. Overt microglia activation due to perturb-ation of the microenvironment can lead to increased release ofglutamate and proinflammatory mediators including TNF-a,

IL-1b, NO, ROS, etc., which in turn would exacerbate braindamage. Microglia-mediated neuroinflammation is implicatedin different neurological diseases and disorders such as peri-ventricular white matter damage, cerebral palsy, autism spec-trum disorders, epilepsy, and perinatal stroke. Microglia acti-vation in the developing brain is controlled through a complexregulatory mechanism and multiple signaling pathways suchas NF-kB, Notch-1, JNK, and p38 MARK that are linked toproduction of proinflammatory mediators. While the func-tional roles of microglia in the developing brain are greatlyamplified in recent years, there remain many issues to be fullyexplored such as transcriptome and epigenetics changes inmicroglial activation. This may pave way for designing appro-priate therapeutic strategies in which the neurotoxic effects ofmicroglial activation may be eliminated to mitigate the injuryin the developing brain.

ACKNOWLEDGMENTThe technical assistance provided by Ms. Suat Hoon

Tan, Ms. Yee Gek Chan, Mrs. Eng Siang Yong, Mrs. GeokLan Ng, Mr. Tuck Yong Yick, Dr. Qiong Cao, and Dr. YajunWu is gratefully acknowledged.

REFERENCES1. Yuan Y, Rangarajan P, Kan EM, et al. Scutellarin regulates the

Notch pathway and affects the migration and morphological trans-formation of activated microglia in experimentally induced cerebralischemia in rats and in activated BV-2 microglia. J Neuroinflamma-tion 2015;12:11

2. Fang M, Yuan Y, Lu J, et al. Scutellarin promotes microglia-mediatedastrogliosis coupled with improved behavioral function in cerebral is-chemia. Neurochem Int 2016;97:154–71

3. Ling EA, Leblond CP. Investigation of glial cells in semithin sections.II. Variation with age in the numbers of the various glial cell types in ratcortex and corpus callosum. J Comp Neurol 1973;149:73–81

4. Dheen ST, Kaur C, Ling EA. Microglial activation and its implicationsin the brain diseases. Curr Med Chem 2007;14:1189–97

5. Del Rio-Hortega P, Penfield W, Microglia In: Penfield W, ed. Cytologyand Cellular Pathology of the Nervous System. New York: Hoeber1932:481–534

6. Mori S, Leblond CP. Identification of microglia in light and electronmicroscopy. J Comp Neurol 1969;135:57–79

7. Ling EA, Paterson JA, Privat A, et al. Investigation of glial cells insemithin sections. I. Identification of glial cells in the brain of youngrats. J Comp Neurol 1973;149:43–71

8. Ling EA, The origin and nature of microglia. In: Fedoroff S, Hertz L,eds. Advances in Cellular Neurobiology, Vol II. New York: AcademicPress, 1981:33–82

9. Ling EA, Tan CK. Amoeboid microglial cells in the corpus callosum ofneonatal rats. Arch Histol Jpn 1974;36:265–80

10. Ling EA, Penney D, Leblond CP. Use of carbon labeling to demonstratethe role of blood monocytes as precursors of the ‘ameboid cells’ presentin the corpus callosum of postnatal rats. J Comp Neurol 1980;193:631–57

11. Ling EA, Ng YK, Wu CH, et al. Microglia: its development and role asa neuropathology sensor. Prog Brain Res 2001;132:61–79

12. Valentino K, Jones E. Morphological and immunocytochemical identi-fication of macrophages in the developing corpus callosum. AnatEmbryol 1981;163:157–72

13. Perry VH, Hume DA, Gordon S. Immunohistochemical localization ofmacrophages and microglia in the adult and developing mouse brain.Neuroscience 1985;15:313–26

14. Ling EA, Kaur LC, Yick TY, et al. Immunocytochemical localizationof CR3 complement receptors with OX-42 in amoeboid microglia inpostnatal rats. Anat Embryol 1990;182:481–6

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

12

Page 13: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

15. Ling EA, Kaur C, Wong WC. Expression of major histocompatibilitycomplex and leukocyte common antigens in amoeboid microglia inpostnatal rats. J Anat 1991;177:117–26

16. Giulian D, Baker TJ. Characterization of ameboid microglia isolatedfrom developing mammalian brain. J Neurosci 1986;6:2163–78

17. Streit WJ, Graeber MB. Heterogeneity of microglial and perivascularcell populations: insights gained from the facial nucleus paradigm. Glia1993;7:68–74

18. Banati RB, Gehrmann J, Schubert P, et al. Cytotoxicity of microglia.Glia 1993;7:111–8

19. Yuan Y, Fang M, Wu C-Y, et al. Scutellarin as a potential therapeuticagent for microglia-mediated neuroinflammation in cerebral ischemia.Neuromolecular Med 2016;18:264–73

20. Dalmau I, Vela JM, Gonzalez B, et al. Expression of LFA-1alpha andICAM-1 in the developing rat brain: a potential mechanism for the re-cruitment of microglial cell precursors. Dev Brain Res 1997;103:163–70

21. Penfield W, Neuroglia and microglia. The interstitial tissue of the cen-tral nervous system. In: Cowdry EV, ed., Special Cytology, 2nd edn,Vol. III. New York: Hoeber, 1932:1445–82.

22. Kershman J. Genesis of microglia in the human brain. Arch Neurol Psy-chiatr 1939; 41:24–50

23. Dougherty TF. Studies on the cytogenesis of microglia and their relationto the cells of the reticulo-endothelial system. Am J Anat 1944;74:61–95

24. Cammermeyer J. The life history of the microglial cell: a light micro-scopic study. Neurosci Res 1970;3:43–129

25. Boya J, Calvo J, Prado A. The origin of microglial cells. J Anat 1979;129:177–86

26. Boya J, Carbonell AL, Calvo J, et al. Ultrastructural study on the originof rat microglia cells. Acta Anat 1987;130:329–35

27. Boya J, Calvo JL, Carbonell AL, et al. A lectin histochemistrystudy on the development of rat microglial cells. J Anat 1991;175:229–36

28. Kaur C, Hao AJ, Wu CH, et al. Origin of microglia. Microsc Res Tech2001;54:2–9

29. Alliot F, Godin I, Pessac B. Microglia derive from progenitors, originat-ing from the yolk sac, and which proliferate in the brain. Dev Brain Res1999; 117:145–52

30. Cuadros MA, Martin C, Coltey P, et al. First appearance, distributionand origin of macrophages in the early development of the avian centralnervous system. J Comp Neurol 1993;330:113–29

31. Kurz H, Christ B. Embryonic CNS macrophages and microglia do notstem from circulating, but from extravascular precursors. Glia 1998;22:98–102

32. Paterson JA, Privat A, Ling EA, et al. Investigation of glial cells insemithin sections. 3. Transformation of subependymal cells into glialcells, as shown by radioautography after 3 H-thymidine injection intothe lateral ventricle of the brain of young rats. J Comp Neurol 1973;149:83–102

33. Ling EA. Some aspects of amoeboid microglia in the corpus callosumand neighbouring regions of neonatal rats. J Anat 1976;121:29–45

34. Fedoroff S, Zhai R, Novak JP. Microglia and astroglia have a commonprogenitor cell. J Neurosci Res 1997; 50:477–86

35. McKanna JA. Primitive glial compartments in the floor plate of mam-malian embryos: distinct progenitors of adult astrocytes and microgliasupport the notoplate hypothesis. Perspect Dev Neurobiol 1993;1:245–55

36. Fujita S, Kitamura T. Origin of brain macrophages and the nature of theso-called microglia. Acta Neuropathol Suppl (Berl) 1975;6:291–6

37. Fujita S, Tsuchihashi Y, Kitamura T. Origin, morphology and functionof the microglia. Prog Clin Biol Res 1981;59:141–69

38. Cuadros MA, Moujahid A, Quesada A, et al. Development of microgliain the quail optic tectum. J Comp Neurol 1994;348:207–24

39. Imamoto K, Leblond CP. Radioautographic investigation of gliogenesisin the corpus callosum of young rats. II. Origin of microglial cells.J Comp Neurol 1978;180:139–63

40. Imamoto K, Fujiwara R, Nagai T, et al. Distribution and fate of macro-phagic ameboid cells in the rat brain. Arch Histol Jpn 1982;45:505–18

41. Tseng C, Ling EA, Wong WC. Light and electron microscopic andcytochemical identification of amoeboid microglial cells in the brain ofprenatal rats. J Anat 1983;136:837

42. Chugani DC, Kedersha NL, Rome LH. Vault immunofluorescence inthe brain: new insights regarding the origin of microglia. J Neurosci1991;11:256–68

43. Hickey WF, Kimura H. Perivascular microglial cells of the CNS arebone marrow-derived and present antigen in vivo. Science 1988; 239:290–2

44. Hutchins KD, Dickson DW, Rashbaum WK, et al. Localization of mor-phologically distinct microglial populations in the developing humanfetal brain: implications for ontogeny. Brain Res Dev Brain Res 1990;55:95–102

45. Andjelkovic AV, Nikolic B, Pachter JS, et al. Macrophages/microglialcells in human central nervous system during development: an immuno-histochemical study. Brain Res 1998;814:13–25

46. Rezaie P, Dean A, Male D, et al. Microglia in the cerebral wall of thehuman telencephalon at second trimester. Cereb Cortex 2005;15:938–49

47. Rezaie P, Male D. Colonisation of the developing human brain and spi-nal cord by microglia: a review. Microsc Res Tech 1999;45:359–82

48. Rezaie P. Microglia in the human nervous system during development.Neuroembryology 2003;2:18–3

49. Fujimoto E, Miki A, Mizuguti H. Histochemical study of the differenti-ation of microglial cells in developing human cerebral hemispheres.J Anat 1989;166:253–64

50. Monier A, Evrard P, Gressens P, Verney C. Distribution and differenti-ation of microglia in the human encephalon during the first two trimes-ters of gestation. J Comp Neurol 2006;499:565–82

51. Ling EA. Light and electron microscopic demonstration of some lyso-somal enzymes in the amoeboid microglia in neonatal rat brain. J Anat1977;123:637–48

52. Ling EA, Kaur C, Wong WC. Light and electron microscopic demon-stration of non-specific esterase in amoeboid microglial cells in the cor-pus callosum in postnatal rats: a cytochemical link to monocytes. J Anat1982;135:385–94

53. Leong SK, Ling EA. Amoeboid and ramified microglia: their interrela-tionship and response to brain injury. Glia 1992;6:39–47

54. Kaur C, Ling EA. Study of the transformation of amoeboid microglialcells into microglia labelled with the isolectin Griffonia simplicifolia inpostnatal rats. Acta Anat (Basel) 1991;142:118–25

55. Villacampa N, Almolda B, Gonz�alez B, et al. Tomato lectin histochem-istry for microglial visualization. Methods Mol Biol 2013;1041:261–79

56. Wu CH, Wen C, Shieh J, et al. A quantitative and morphometric studyof the transformation of amoeboid microglia into ramified microglia inthe developing corpus callosum in rats. J Anat 1992;181:423

57. Wu CH, Wen C, Shieh J, et al. Down-regulation of membrane glycopro-tein in amoeboid microglia transforming into ramified microglia inpostnatal rat brain. J Neurocytol 1994;23:258–69

58. Kaur C, Ling EA, Wong WC. Transformation of amoeboid microglialcells into microglia in the corpus callosum of the postnatal rat brain. Anelectron microscopical study. Arch Histol Jpn 1985;48:17–25

59. Ling EA, Tseng CY, Voon FC, et al. Isolation and culture of amoeboidmicroglial cells from the corpus callosum and cavum septum pelluci-dum in postnatal rats. J Anat 1983;137:223–33

60. Wu C, Yeh S, Ling EA. Studies of lectin receptors of rat microglia inculture: receptor distribution and internalization. Exp Brain Res 1999;124:89–99

61. Xu J, Ling EA. Expression of major histocompatibility complex class IIantigen on amoeboid microglial cells in early postnatal rat brain follow-ing intraperitoneal injections of lipopolysaccharide. Exp Brain Res1994;100:287–92

62. Xu J, Ling EA. Upregulation and induction of surface antigens with spe-cial reference to MHC class II expression in microglia in postnatal ratbrain following intravenous or intraperitoneal injections of lipopolysac-charide. J Anat 1994;184:285

63. Xu J, Ling E. Upregulation and induction of major histocompatibilitycomplex class I and II antigens on microglial cells in early postnatal ratbrain following intraperitoneal injections of recombinant interferon-gamma. Neuroscience 1994;60:959–67

64. Kaur C, Ling EA. Periventricular white matter damage in the hypoxicneonatal brain: role of microglial cells. Prog Neurobiol 2009;87:264–80

65. Brockhaus J, Moller T, Kettenmann H. Phagocytozing ameboid micro-glial cells studied in a mouse corpus callosum slice preparation. Glia1996;16:81–90

J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017 Microglia in the Developing Brain

13

Page 14: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

66. Petersen MA, Dailey ME. Diverse microglial motility behaviors duringclearance of dead cells in hippocampal slices. Glia 2004;46:195–206

67. Deng Y, Lu J, Sivakumar V, et al. Amoeboid microglia in the periven-tricular white matter induce oligodendrocyte damage through expres-sion of proinflammatory cytokines via MAP kinase signaling pathwayin hypoxic neonatal rats. Brain Pathol 2008;18:387–400

68. Kaur C, Sivakumar V, Zou Z, et al. Microglia-derived proinflammatorycytokines tumor necrosis factor-alpha and interleukin-1beta induce Pur-kinje neuronal apoptosis via their receptors in hypoxic neonatal ratbrain. Brain Struct Funct 2014;219:151–70

69. Kaur C, Ling EA, Wong WC. Cytochemical localisation of 5’-nucleoti-dase in amoeboid microglial cells in postnatal rats. J Anat 1984;139:1–7

70. Kaur C, Ling EA, Wong WC. Labelling of amoeboid microglial cells inrats of various ages following an intravenous injection of horseradishperoxidase. Acta Anat (Basel) 1986;125:132–7

71. Xu J, Kaur C, Ling E. Variation with age in the labelling of amoeboidmicroglial cells in rats following intraperitoneal or intravenous injectionof a fluorescent dye. J Anat 1993;182:55

72. Earle KL, Mitrofanis J. Identification of transient microglial cell colo-nies in the forebrain white matter of developing rats. J Comp Neurol1997;387:371–84

73. Leong SK, Shieh JY, Ling EA, et al. Labelling of amoeboid microglialcells in the supraventricular corpus callosum following the applicationof horseradish peroxidase in the cerebrum and spinal cord in rats. J Anat1983;136:367–77

74. Kaur C, Too HF, Ling EA. Phagocytosis of Escherichia coli by amoeb-oid microglial cells in the developing brain. Acta Neuropathol 2004;107:204–8

75. Perez-Pouchoulen M, VanRyzin JW, McCarthy MM. Morphologicaland phagocytic profile of microglia in the developing rat cerebel-lum(1,2,3). eNeuro 2015;2. doi: 10.1523/ENEURO.0036-15.2015

76. Cunningham CL, Mart�ınez-Cerde~no V, Noctor SC. Microglia regulatethe number of neural precursor cells in the developing cerebral cortex.J Neurosci 2013;33:4216–33

77. Li YB, Kaur C, Ling EA. Neuronal degeneration and microglial reac-tion in the fetal and postnatal rat brain after transient maternal hypoxia.Neurosci Res 1998;32:137–48

78. Kaur C, Sivakumar V, Ang LS, et al. Hypoxic damage to the periven-tricular white matter in neonatal brain: role of vascular endothelialgrowth factor, nitric oxide and excitotoxicity. J Neurochem 2006;98:1200–16

79. Kurpius D, Wilson N, Fuller L, et al. Early activation, motility, andhoming of neonatal microglia to injured neurons does not require pro-tein synthesis. Glia 2006;54:58–70

80. Kurpius D, Nolley EP, Dailey ME. Purines induce directed migrationand rapid homing of microglia to injured pyramidal neurons in develop-ing hippocampus. Glia 2007;55:873–84

81. Faustino JV, Wang X, Johnson CE, et al. Microglial cells contribute toendogenous brain defenses after acute neonatal focal stroke. J Neurosci2011;31:12992

82. Ferrer I, Olive M, Blanco R, et al. Amoeboid microglial response fol-lowing X-ray-induced apoptosis in the neonatal rat brain. Neurosci Lett1995;193:109–12

83. Takahashi K, Rochford CD, Neumann H. Clearance of apoptotic neu-rons without inflammation by microglial triggering receptor expressedon myeloid cells-2. J Exp Med 2005;201:647–57

84. Kawabori M, Kacimi R, Kauppinen T, et al. Triggering receptorexpressed on myeloid cells 2 (TREM2) deficiency attenuates phago-cytic activities of microglia and exacerbates ischemic damage in experi-mental stroke. J Neurosci 2015;35:3384–96

85. Chertoff M, Shrivastava K, Gonzalez B, et al. Differential modulationof TREM2 protein during postnatal brain development in mice. PLoSOne 2013;8:e72083

86. Wolswijk G. Strongly GD3þ cells in the developing and adult rat cere-bellum belong to the microglial lineage rather than to the oligodendro-cyte lineage. Glia 1995;13:13–26

87. Rajbhandari L, Tegenge MA, Shrestha S, et al. Toll-like receptor 4 defi-ciency impairs microglial phagocytosis of degenerating axons. Glia2014;62:1982–91

88. Yao L, Kan EM, Lu J, et al. Toll-like receptor 4 mediates microglial ac-tivation and production of inflammatory mediators in neonatal rat brain

following hypoxia: role of TLR4 in hypoxic microglia. J Neuroinflam-mation 2013;10:23

89. Kaur C, You Y. Ultrastructure and function of the amoeboid microglialcells in the periventricular white matter in postnatal rat brain followinga hypoxic exposure. Neurosci Lett 2000;290:17–20

90. Rocha N, Neefjes J. MHC class II molecules on the move for successfulantigen presentation. EMBO J 2008;27:1–5

91. English L, Chemali M, Duron J, et al. Autophagy enhances the presenta-tion of endogenous viral antigens on MHC class I molecules duringHSV-1 infection. Nat Immunol 2009;10:480–7

92. Roche PA, Furuta K. The ins and outs of MHC class II-mediated anti-gen processing and presentation. Nat Rev Immunol 2015;15:203–16

93. Wierzba-Bobrowicz T, Kosno-Kruszewska E, Lewandowska E, et al.Major histocompatibility complex class II (MHC II) expression duringdevelopment of human fetal brain and haemopoietic organs. Adv ExpMed Biol 2001;495:93–102

94. McRae A, Gilland E, Bona E, et al. Microglia activation after neonatalhypoxic-ischemia. Brain Res Dev Brain Res 1995;84:245–52

95. Krishnan S, Chang AC, Stoltz BM, et al. Escherichia coli K1 modulatesperoxisome proliferator–activated receptor c and glucose transporter 1at the blood-brain barrier in neonatal meningitis. J Infect Dis 2016;214:1092–104

96. Zhang W, Zhang H, Mu H, et al. Omega-3 polyunsaturated fatty acidsmitigate blood–brain barrier disruption after hypoxic–ischemic brain in-jury. Neurobiol Dis 2016;91:37–46

97. Moretti R, Zanin A, Pansiot J, et al. Melatonin reduces excitotoxicblood-brain barrier breakdown in neonatal rats. Neuroscience 2015;311:382–97

98. Ortega SB, Kong X, Venkataraman R, et al. Perinatal chronic hypoxiainduces cortical inflammation, hypomyelination, and peripheralmyelin-specific T cell autoreactivity. J Leukoc Biol 2016;99:21–9

99. Paolicelli RC, Bolasco G, Pagani F, et al. Synaptic pruning bymicroglia is necessary for normal brain development. Science 2011;333:1456–8

100. Schafer DP, Lehrman EK, Kautzman AG, et al. Microglia sculpt postna-tal neural circuits in an activity and complement-dependent manner.Neuron 2012;74:691–705

101. Stevens B, Allen NJ, Vazquez LE, et al. The classical complement cas-cade mediates CNS synapse elimination. Cell 2007;131:1164–78

102. Lim S-H, Park E, You B, et al. Neuronal synapse formation induced bymicroglia and interleukin 10. PLoS One 2013;8:e81218

103. Parkhurst CN, Yang G, Ninan I, et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic fac-tor. Cell 2013;155:1596–609

104. Corriveau RA, Huh GS, Shatz CJ. Regulation of class I MHC gene ex-pression in the developing and mature CNS by neural activity. Neuron1998;21:505–20

105. Datwani A, McConnell MJ, Kanold PO, et al. Classical MHCI mole-cules regulate retinogeniculate refinement and limit ocular dominanceplasticity. Neuron 2009;64:463–70

106. Goddard CA, Butts DA, Shatz CJ. Regulation of CNS synapses by neur-onal MHC class I. Proc Natl Acad Sci USA 2007;104:6828–33

107. Huh GS, Boulanger LM, Du H, et al. Functional requirement for class IMHC in CNS development and plasticity. Science 2000;290:2155–9

108. Chamak B, Morandi V, Mallat M. Brain macrophages stimulate neuritegrowth and regeneration by secreting thrombospondin. J Neurosci Res1994;38:221–33

109. Morgan SC, Taylor DL, Pocock JM. Microglia release activators ofneuronal proliferation mediated by activation of mitogen-activated pro-tein kinase, phosphatidylinositol-3-kinase/Akt and delta-Notch signal-ling cascades. J Neurochem 2004;90:89–101

110. Frost JL, Schafer DP. Microglia: architects of the developing nervoussystem. Trends Cell Biol 2016;26:587–97

111. Ueno M, Fujita Y, Tanaka T, et al. Layer V cortical neurons requiremicroglial support for survival during postnatal development. Nat Neu-rosci 2013;16:543–51

112. Shigemoto-Mogami Y, Hoshikawa K, Goldman JE, et al. Microglia en-hance neurogenesis and oligodendrogenesis in the early postnatal sub-ventricular zone. J Neurosci 2014;34:2231–43

113. O’Kusky J, Ye P. Neurodevelopmental effects of insulin-like growthfactor signaling. Front Neuroendocrinol 2012;33:230–51

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

14

Page 15: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

114. Nicholas RS, Wing MG, Compston A. Nonactivated microglia promoteoligodendrocyte precursor survival and maturation through the tran-scription factor NF-jB. Eur J Neurosci 2001;13:959–67

115. Nakanishi M, Niidome T, Matsuda S, et al. Microglia-derivedinterleukin-6 and leukaemia inhibitory factor promote astrocytic differ-entiation of neural stem/progenitor cells. Eur J Neurosci 2007;25:649–58

116. Arnold T, Betsholtz C. The importance of microglia in the developmentof the vasculature in the central nervous system. Vasc Cell 2013; 5:4

117. Cheepsunthorn P, Palmer C, Connor JR. Cellular distribution of ferritinsubunits in postnatal rat brain. J Comp Neurol 1998; 400:73–86

118. Fitzsimons JT. Angiotensin, thirst, and sodium appetite. Physiol Rev1998;78:583–686

119. Paul M, Poyan Mehr A, Kreutz R. Physiology of local renin-angiotensinsystems. Physiol Rev 2006;86:747–803

120. Saavedra JM. Brain and pituitary angiotensin. Endocr Rev 1992;13:329–80

121. Mao C, Shi L, Xu F, et al. Development of fetal brain renin-angiotensinsystem and hypertension programmed in fetal origins. Prog Neurobiol2009;87:252–63

122. Li JJ, Lu J, Kaur C, et al. Expression of angiotensin II and its receptorsin the normal and hypoxic amoeboid microglial cells and murine BV-2cells. Neuroscience 2009;158:1488–99

123. Baburamani AA, Ek CJ, Walker DW, et al. Vulnerability of the devel-oping brain to hypoxic-ischemic damage: contribution of the cerebralvasculature to injury and repair? Front Physiol 2012;3:424

124. Ek CJ, D’Angelo B, Baburamani AA, et al. Brain barrier properties andcerebral blood flow in neonatal mice exposed to cerebral hypoxia–is-chemia. J Cereb Blood Flow Metab 2015;35:818–27

125. Ando H, Zhou J, Macova M, et al. Angiotensin II AT1 receptor block-ade reverses pathological hypertrophy and inflammation in brain micro-vessels of spontaneously hypertensive rats. Stroke 2004;35:1726–31

126. Jung K-H, Chu K, Lee S-T, et al. Blockade of AT1 receptor reducesapoptosis, inflammation, and oxidative stress in normotensive rats withintracerebral hemorrhage. J Pharmacol Exp Ther 2007;322:1051

127. Yanagisawa M, Inoue A, Ishikawa T, et al. Primary structure, synthesis,and biological activity of rat endothelin, an endothelium-derived vaso-constrictor peptide. Proc Natl Acad Sci USA 1988;85:6964–7

128. Inoue A, Yanagisawa M, Kimura S, et al. The human endothelin family:three structurally and pharmacologically distinct isopeptides predictedby three separate genes. Proc Natl Acad Sci USA 1989;86:2863–7

129. Arai H, Hori S, Aramori I, et al. Cloning and expression of a cDNAencoding an endothelin receptor. Nature 1990;348:730–2

130. Sakurai T, Yanagisawa M, Takuwa Y, et al. Cloning of a cDNA encod-ing a non-isopeptide-selective subtype of the endothelin receptor. Na-ture 1990;348:732

131. McMillen MA, Cunningham ME. Origin of endothelin in sepsis. CritCare Med 1996;24:721–2

132. MacCumber MW, Ross CA, Snyder SH. Endothelin in brain: receptors,mitogenesis, and biosynthesis in glial cells. Proc Natl Acad Sci USA1990;87:2359–63

133. Wu-Wong JR, Chiou JW, Dickinson R, et al. Endothelin attenuatesapoptosis in human smooth muscle cells. Biochem J 1997;328:733–7

134. Wu CY, Kaur C, Lu J, et al. Transient expression of endothelins in theamoeboid microglial cells in the developing rat brain. Glia 2006;54:513–25

135. Jo SM, Ryu HJ, Kim JE, et al. Up-regulation of endothelial endothelin-1 expression prior to vasogenic edema formation in the rat piriform cor-tex following status epilepticus. Neurosci Lett 2011;501:25–30

136. Kaur C, Sivakumar V, Dheen ST, et al. (2006) Insulin-like growth fac-tor I and II expression and modulation in amoeboid microglial cells bylipopolysaccharide and retinoic acid. Neuroscience 2006;138:1233–44.

137. Sivakumar V, Ling EA, Lu J, et al. Role of glutamate and its receptorsand insulin-like growth factors in hypoxia induced periventricular whitematter injury. Glia 2010;58:507–23.

138. Fushimi S, Shirabe T. Expression of insulin-like growth factors inremyelination following ethidium bromide-induced demyelination inthe mouse spinal cord. Neuropathology 2004;24:208–18

139. Guan J, Bennet L, Gluckman PD, et al. Insulin-like growth factor-1 andpost-ischemic brain injury. Prog Neurobiol 2003;70:443–62

140. Logan A, Gonzalez AM, Hill DJ, et al. Coordinated pattern of expres-sion and localization of insulin-like growth factor-II (IGF-II) and IGF-

binding protein-2 in the adult rat brain. Endocrinology 1994;135:2255–64

141. Dubois-Dalcq M, Murray K. Why are growth factors important in oligo-dendrocyte physiology? Pathol Biol 2000;48:80–6

142. Zeger M, Popken G, Zhang J, et al. Insulin-like growth factor type 1 re-ceptor signaling in the cells of oligodendrocyte lineage is required fornormal in vivo oligodendrocyte development and myelination. Glia2007;55:400–11

143. Beilharz EJ, Russo VC, Butler G, et al. Co-ordinated and cellular spe-cific induction of the components of the IGF/IGFBP axis in the rat brainfollowing hypoxic–ischemic injury. Brain Res Mol Brain Res 1998;59:119–34

144. Ye P, D’ercole A. Insulin-like growth factor i protects oligodendrocytesfrom tumor necrosis factor-a-induced injury 1. Endocrinology 1999;140:3063–72

145. Pang Y, Zheng B, Fan L-W, et al. IGF-1 protects oligodendrocyte pro-genitors against TNFa-induced damage by activation of PI3K/Akt andinterruption of the mitochondrial apoptotic pathway. Glia 2007;55:1099–107

146. Suh H-S, Zhao M-L, Derico L, et al. Insulin-like growth factor 1 and 2(IGF1, IGF2) expression in human microglia: differential regulation byinflammatory mediators. J Neuroinflammation 2013;10:3

147. Stipursky J, Gomes FCA. TGF-b1/SMAD signaling induces astrocytefate commitment in vitro: Implications for radial glia development. Glia2007;55:1023–33

148. Yi JJ, Barnes AP, Hand R, et al. TGF-b signaling specifies axons duringbrain development. Cell 2010;142:144–57

149. Li JJ, Lu J, Kaur C, et al. Effects of hypoxia on expression of transform-ing growth factor-beta1 and its receptors I and II in the amoeboid micro-glial cells and murine BV-2 cells. Neuroscience 2008;156:662–72

150. Brionne TC, Tesseur I, Masliah E, et al. Loss of TGF-b1 leads toincreased neuronal cell death and microgliosis in mouse brain. Neuron2003;40:1133–45

151. O’Keefe GM, Nguyen VT, Benveniste EN. Regulation and function ofclass II major histocompatibility complex, CD40, and B7 expression inmacrophages and microglia: implications in neurological diseases.J Neurovirol 2002;8:496–512

152. Lindholm D, Castren E, Kiefer R, et al. Transforming growth factor-beta 1 in the rat brain: increase after injury and inhibition of astrocyteproliferation. J Cell Biol 1992;117:395–400

153. Araujo AP, Diniz LP, Eller CM, et al. Effects of transforming growthfactor beta 1 in cerebellar development: role in synapse formation.Front Cell Neurosci 2016;10:104

154. Heese K, Fiebich BL, Bauer J, et al. NF-jB modulateslipopolysaccharide-induced microglial nerve growth factor expression.Glia 1998;22:401–7

155. Nakajima K, Honda S, Tohyama Y, et al. Neurotrophin secretion fromcultured microglia. J Neurosci Res 2001;65:322–3

156. Elkabes S, Peng L, Black IB. Lipopolysaccharide differentially regu-lates microglial trk receptor and neurotrophin expression. J NeurosciRes 1998;54:117–22

157. Nakajima K, Tohyama Y, Maeda S, et al. Neuronal regulation by whichmicroglia enhance the production of neurotrophic factors for GABAer-gic, catecholaminergic, and cholinergic neurons. Neurochem Int 2007;50:807–20

158. Miwa T, Furukawa S, Nakajima K, et al. Lipopolysaccharide enhancessynthesis of brain-derived neurotrophic factor in cultured rat microglia.J Neurosci Res 1997;50:1023–9

159. Chao MV. Neurotrophins and their receptors: a convergence point formany signalling pathways. Nat Rev Neurosci 2003;4:299–309

160. Neumann H, Misgeld T, Matsumuro K, et al. Neurotrophins inhibitmajor histocompatibility class II inducibility of microglia: involvementof the p75 neurotrophin receptor. Proc Natl Acad Sci USA 1998;95:5779–84

161. Davis MI. Ethanol-BDNF interactions: still more questions thananswers. Pharmacol Ther 2008;118:36–57

162. Zucca S, Valenzuela CF. Low concentrations of alcohol inhibit BDNF-dependent GABAergic plasticity via L-type Ca2þ channel inhibition indeveloping CA3 hippocampal pyramidal neurons. J Neurosci 2010;30:6776–81

163. Kenchappa RS, Tep C, Korade Z, et al. p75 Neurotrophin receptor-mediated apoptosis in sympathetic neurons involves a biphasic

J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017 Microglia in the Developing Brain

15

Page 16: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

activation of JNK and up-regulation of tumor necrosis factor-a-convert-ing enzyme/ADAM17. J Biol Chem 2010;285:20358–68

164. Yang H, Feng GD, Liang Z, et al. In vitro beneficial activation of micro-glial cells by mechanically-injured astrocytes enhances the synthesisand secretion of BDNF through p38MAPK. Neurochem Int 2012;61:175–86

165. Boyadjieva NI, Sarkar DK. Cyclic adenosine monophosphate andbrain-derived neurotrophic factor decreased oxidative stress and apop-tosis in developing hypothalamic neuronal cells: role of microglia. Al-cohol Clin Exp Res 2013;37:1370–9

166. Zhang J, Geula C, Lu C, et al. Neurotrophins regulate proliferation andsurvival of two microglial cell lines in vitro. Exp Neurol 2003;183:469–81

167. Jiang Y, Wei N, Lu T, et al. Intranasal brain-derived neurotrophic factorprotects brain from ischemic insult via modulating local inflammationin rats. Neuroscience 2011;172:398–405

168. Merrill JE. Tumor necrosis factor alpha, Interleukin 1 and related cyto-kines in brain development: normal and pathological. Dev Neurosci1992;14:1–10

169. Fee D, Grzybicki D, Dobbs M, et al. Interleukin 6 promotes vasculogen-esis of murine brain microvessel endothelial cells. Cytokine 2000;12:655–65

170. Gadient RA, Otten U. Expression of interleukin-6 (IL-6) andinterleukin-6 receptor (IL-6R) mRNAs in rat brain during postnatal de-velopment. Brain Res 1994;637:10–4

171. Li F, Lu J, Wu CY, et al. Expression of Kv1.2 in microglia and its puta-tive roles in modulating production of proinflammatory cytokines andreactive oxygen species. J Neurochem 2008; 106:2093–105

172. Rathnasamy G, Ling EA, Kaur C. Iron and iron regulatory proteins inamoeboid microglial cells are linked to oligodendrocyte death in hyp-oxic neonatal rat periventricular white matter through production ofproinflammatory cytokines and reactive oxygen/nitrogen species.J Neurosci 2011;31:17982–95

173. Rathnasamy G, Ling EA, Kaur C. Hypoxia inducible factor-1alphamediates iron uptake which induces inflammatory response in amoeb-oid microglial cells in developing periventricular white matter throughMAP kinase pathway. Neuropharmacology 2014;77:428–40

174. Li P, Lu J, Kaur C, et al. Expression of cyclooxygenase-1/-2, micro-somal prostaglandin-E synthase-1 and E-prostanoid receptor 2 andregulation of inflammatory mediators by PGE(2) in the amoeboidmicroglia in hypoxic postnatal rats and murine BV-2 cells. Neurosci-ence 2009; 164:948–62

175. Deng YY, Lu J, Ling EA, et al. Monocyte chemoattractant protein-1(MCP-1) produced via NF-kappaB signaling pathway mediates migra-tion of amoeboid microglia in the periventricular white matter in hyp-oxic neonatal rats. Glia. 2009; 57:604–21.

176. Deng YY, Lu J, Ling, et al. Microglia-derived macrophage colony stim-ulating factor promotes generation of proinflammatory cytokines byastrocytes in the periventricular white matter in the hypoxic neonatalbrain. Brain Pathol 2010;20:909–25

177. Fernandez-Lizarbe S, Pascual M, Guerri C. Critical role of TLR4 re-sponse in the activation of microglia induced by ethanol. J Immunol2009;183:4733–44

178. Fernandez-Lizarbe S, Montesinos J, Guerri C. Ethanol induces TLR4/TLR2 association, triggering an inflammatory response in microglialcells. J Neurochem 2013;126:261–73

179. Guizzetti M, Zhang X, Goeke C, et al. Glia and neurodevelopment:focus on fetal alcohol spectrum disorders. Front Pediatr 2014;2:123

180. Wilhelm CJ, Guizzetti M. Fetal alcohol spectrum disorders: an over-view from the glia perspective. Front Integr Neurosci 2015;9:65

181. Hao A-J, Dheen ST, Ling E-A. Response of amoeboid microglia/brainmacrophages in fetal rat brain exposed to a teratogen. J Neurosci Res2001;64:79–93

182. Lian Q, Dheen ST, Liao D, et al. Enhanced inflammatory response inneural tubes of embryos derived from diabetic mice exposed to a terato-gen. J Neurosci Res 2004;75:554–64

183. Hao AJ, Dheen ST, Ling EA. Expression of macrophage colony-stimulating factor and its receptor in microglia activation is linked toteratogen-induced neuronal damage. Neuroscience 2002;112:889–900

184. Mitrasinovic OM, Perez GV, Zhao F, et al. Overexpression of macro-phage colony-stimulating factor receptor on microglial cells induces aninflammatory response. J Biol Chem 2001;276:30142–9

185. Mitrasinovic OM, Robinson CC, Tenen DG, et al. Biolistic expressionof the macrophage colony stimulating factor receptor in organotypiccultures induces an inflammatory response. J Neurosci Res 2004;77:420–9

186. Murugan M, Sivakumar V, Lu J, et al. Expression of N-methyl D-aspar-tate receptor subunits in amoeboid microglia mediates production of ni-tric oxide via NF-jB signaling pathway and oligodendrocyte cell deathin hypoxic postnatal rats. Glia 2011;59:521–39

187. Kaindl AM, Degos V, Peineau S, et al. Activation of microglial N-me-thyl-D-aspartate receptors triggers inflammation and neuronal celldeath in the developing and mature brain. Ann Neurol 2012;72:536–49

188. Ratie L, Ware M, Barloy-Hubler F, et al. Novel genes upregulated whenNOTCH signalling is disrupted during hypothalamic development.Neural Dev 2013;8:25

189. Androutsellis-Theotokis A, Leker RR, Soldner F, et al. Notch signallingregulates stem cell numbers in vitro and in vivo. Nature 2006;442:823–6

190. Androutsellis-Theotokis A, Rueger MA, Mkhikian H, et al. Signalingpathways controlling neural stem cells slow progressive brain disease.Cold Spring Harb Symp Quant Biol 2008;73:403–10

191. Yang H, Zheng S, Mao Y, et al. Modulating of ocular inflammationwith macrophage migration inhibitory factor is associated with notchsignalling in experimental autoimmune uveitis. Clin Exp Immunol2016;183:280–93

192. Bassil R, Zhu B, Lahoud Y, et al. Notch ligand delta-like 4 blockadealleviates experimental autoimmune encephalomyelitis by promotingregulatory T cell development. J Immunol 2011;187:2322–8

193. Ishida W, Fukuda K, Sakamoto S, et al. Regulation of experimentalautoimmune uveoretinitis by anti-delta-like ligand 4 monoclonal anti-body. Invest Ophthalmol Vis Sci 2011;52:8224–30

194. Cao Q, Lu J, Kaur C, et al. Expression of Notch-1 receptor and itsligands Jagged-1 and Delta-1 in amoeboid microglia in postnatal ratbrain and murine BV-2 cells. Glia 2008;56:1224–37

195. McManus CM, Brosnan CF, Berman JW. Cytokine induction of MIP-1a and MIP-1b in human fetal microglia. J Immunol 1998;160:1449–55

196. Goldman D, Song X, Kitai R, et al. Cryptococcus neoformans inducesmacrophage inflammatory protein 1alpha (MIP-1alpha) and MIP-1betain human microglia: role of specific antibody and soluble capsular poly-saccharide. Infect Immun 2001;69:1808–15

197. Cowell RM, Xu H, Galasso JM, et al. Hypoxic-ischemic injury inducesmacrophage inflammatory protein-1alpha expression in immature ratbrain. Stroke 2002;33:795–801

198. Kaur C, Sivakumar V, Yip GW, et al. Expression of syndecan-2 in theamoeboid microglial cells and its involvement in inflammation in thehypoxic developing brain. Glia 2009;57:336–49

199. Cloarec R, Bauer S, Luche H, et al. Cytomegalovirus infection of the ratdeveloping brain in utero prominently targets immune cells and pro-motes early microglial activation. PLoS One 2016;11:e0160176

200. Mizutani M, Pino PA, Saederup N, et al. The fractalkine receptor butnot CCR2 is present on microglia from embryonic developmentthroughout adulthood. J Immunol 2012;188:29–36

201. Limatola C, Ransohoff RM. Modulating neurotoxicity throughCX3CL1/CX3CR1 signaling. Front Cell Neurosci 2014;8:229

202. Ivacko J, Szaflarski J, Malinak C, et al. Hypoxic-ischemic injury indu-ces monocyte chemoattractant protein-1 expression in neonatal ratbrain. J Cereb Blood Flow Metab 1997;17:759–70

203. Bona E, Andersson A-L, Blomgren K, et al. Chemokine and inflamma-tory cell response to hypoxia–ischemia in immature rats. Pediatr Res1999;45:500–9

204. Buonocore G, Perrone S, Bracci R. Free radicals and brain damage inthe newborn. Biol Neonate 2001; 79:180–6

205. Welin A-K, Sandberg M, Lindblom A, et al. White matter injury fol-lowing prolonged free radical formation in the 0.65 gestation fetal sheepbrain. Pediatric Res 2005;58:100–5

206. Haynes RL, Baud O, Li J, et al. Oxidative and nitrative injury in peri-ventricular leukomalacia: a review. Brain Pathol 2005;15:225–33

207. Griot C, Vandevelde M, Richard A, et al. Selective degeneration of oli-godendrocytes mediated by reactive oxygen species. Free Radic ResCommun 1990;11:181–93

208. van der Goes A, Brouwer J, Hoekstra K, et al. Reactive oxygen speciesare required for the phagocytosis of myelin by macrophages. J Neuro-immunol 1998;92:67–75

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

16

Page 17: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

209. Zhou Y, Ling EA, Dheen ST. Dexamethasone suppresses monocytechemoattractant protein-1 production via mitogen activated protein kin-ase phosphatase-1 dependent inhibition of Jun N-terminal kinase andp38 mitogen-activated protein kinase in activated rat microglia. J Neu-rochem 2007;102:667–78

210. Cao Q, Li P, Lu J, et al. Nuclear factor-kappaB/p65 responds to changesin the Notch signaling pathway in murine BV-2 cells and in amoeboidmicroglia in postnatal rats treated with the gamma-secretase complexblocker DAPT. J Neurosci Res 2010;88:2701–14

211. Yang X, He G, Hao Y, et al. The role of the JAK2-STAT3 pathway inpro-inflammatory responses of EMF-stimulated N9 microglial cells.J Neuroinflammation 2010;7:54

212. Jeon H, Kim JH, Kim JH, et al. Plasminogen activator inhibitor type 1regulates microglial motility and phagocytic activity. J Neuroinflamma-tion 2012;9:149

213. Yao L, Cao Q, Wu C, et al. Notch signaling in the central nervous sys-tem with special reference to its expression in microglia. CNS NeurolDisord Drug Targets 2013;12:807–14

214. Yao L, Kan EM, Kaur C, et al. Notch-1 signaling regulates microgliaactivation via NF-jB pathway after hypoxic exposure in vivo andin vitro. PLoS One 2013;8:e78439

215. Cao Q, Kaur C, Wu CY, et al. Nuclear factor-kappa beta regulatesNotch signaling in production of proinflammatory cytokines and nitricoxide in murine BV-2 microglial cells. Neuroscience 2011;192:140–54

216. Stridh L, Mottahedin A, Johansson ME, et al. Toll-like receptor-3 acti-vation increases the vulnerability of the neonatal brain to hypoxia–is-chemia. J Neurosci 2013;33:12041–51

217. Thrash JC, Torbett BE, Carson MJ. Developmental regulation ofTREM2 and DAP12 expression in the murine CNS: implications forNasu-Hakola disease. Neurochem Res 2009;34:38–45

218. Follett PL, Deng W, Dai W, et al. Glutamate receptor-mediated oligo-dendrocyte toxicity in periventricular leukomalacia: a protective rolefor topiramate. J Neurosci 2004;24:4412

219. Pitt D, Werner P, Raine CS. Glutamate excitotoxicity in a model of mul-tiple sclerosis. Nature Med 2000;6:67–70

220. Grunder T, Kohler K, Guenther E. Alterations in NMDA receptor ex-pression during retinal degeneration in the RCS rat. Vis Neurosci 2001;18:781–7

221. Hagberg H. Hypoxic-ischemic damage in the neonatal brain: excitatoryamino acids. Dev Pharmacol Ther 1992;18:139–44

222. Patrizio M, Levi G. Glutamate production by cultured microglia: differ-ences between rat and mouse, enhancement by lipopolysaccharide andlack effect of HIV coat protein gp120 and depolarizing agents. NeurosciLett 1994;178:184–9

223. Domercq M, S�anchez-G�omez MV, Sherwin C, et al. System xc- andglutamate transporter inhibition mediates microglial toxicity to oligo-dendrocytes. J Immunol 2007;178:6549–56

224. Singh V, Gera R, Kushwaha R, et al. Hijacking microglial glutathioneby inorganic arsenic impels bystander death of immature neuronsthrough extracellular cystine/glutamate imbalance. Sci Rep 2016;6:30601

225. Noda M, Nakanishi H, Nabekura J, et al. AMPA-kainate subtypes ofglutamate receptor in rat cerebral microglia. J Neurosci 2000;20:251–8

226. Volpe JJ. Perinatal brain injury: from pathogenesis to neuroprotection.Ment Retard Dev Disabil Res Rev 2001;7:56–64

227. Hagino Y, Kariura Y, Manago Y, et al. Heterogeneity and potentiationof AMPA type of glutamate receptors in rat cultured microglia. Glia2004;47:68–77

228. Guadagno J, Xu X, Karajgikar M, et al. Microglia-derived TNFa indu-ces apoptosis in neural precursor cells via transcriptional activation ofthe Bcl-2 family member Puma. Cell Death Dis 2013;4:e538

229. Hu S, Sheng WS, Ehrlich LC, et al. Cytokine effects on glutamate up-take by human astrocytes. Neuroimmunomodulation 2000;7:153–9

230. Tseng C, Ling E, Wong W. Scanning electron microscopy of amoeboidmicroglial cells in the transient cavum septum pellucidum in pre-andpostnatal rats. J Anat 1983;136:251

231. Kaur C, Ling EA. Transitory cystic cavities in the developing mamma-lian brain–normal or anomalous? J Anat 2017; 230:197–202

232. Kaur C, Ling EA, Wong WC. Scanning electron microscopy of transi-tory subependymal cysts in the developing midbrain of postnatal rats.Arch Histol Cytol 1989;52:311–7

233. Xie D, Shen F, He S, et al. IL-1b induces hypomyelination in the peri-ventricular white matter through inhibition of oligodendrocyte progeni-tor cell maturation via FYN/MEK/ERK signaling pathway in septicneonatal rats. Glia 2016;64:583–602.

234. Selmaj KW, Raine CS. Tumor necrosis factor mediates myelin andoligodendrocyte damage in vitro. Ann Neurol 1988;23:339–46

235. Louis JC, Magal E, Takayama S, et al. CNTF protection of oligodendro-cytes against natural and tumor necrosis factor-induced death. Science1993;259:689–92

236. Rezaie P, Dean A. Periventricular leukomalacia, inflammation andwhite matter lesions within the developing nervous system. Neuropath-ology 2002;22:106–32

237. Haynes RL, Folkerth RD, Keefe RJ, et al. Nitrosative and oxidative in-jury to premyelinating oligodendrocytes in periventricular leukomala-cia. J Neuropathol Exp Neurol 2003;62:441–50

238. Li J, Baud O, Vartanian T, et al. Peroxynitrite generated by inducible ni-tric oxide synthase and NADPH oxidase mediates microglial toxicity tooligodendrocytes. Proc Natl Acad Sci USA 2005;102:9936–41

239. Volpe JJ. Brain injury in premature infants: a complex amalgam of de-structive and developmental disturbances. Lancet Neurol 2009;8:110–24

240. Buser JR, Maire J, Riddle A, et al. Arrested preoligodendrocyte matur-ation contributes to myelination failure in premature infants. Ann Neu-rol 2012;71:93–109

241. Strasser K, Lueckemann L, Kluever V, et al. Dose-dependent effects oflevetiracetam after hypoxia and hypothermia in the neonatal mousebrain. Brain Res 2016;1646:116–24

242. Rochefort N, Quenech’du N, Watroba L, et al. Microglia and astrocytesmay participate in the shaping of visual callosal projections during post-natal development. J Physiol 2002;96:183–92

243. Kadhim H, Tabarki B, Verellen G, et al. Inflammatory cytokines in thepathogenesis of periventricular leukomalacia. Neurology 2001;56:1278–84

244. Kadhim H, Khalifa M, Deltenre P, et al. Molecular mechanisms of celldeath in periventricular leukomalacia. Neurology 2006;67:293–9

245. Dean JM, van de Looij Y, Sizonenko SV, et al. Delayed cortical impair-ment following lipopolysaccharide exposure in preterm fetal sheep.Ann Neurol 2011;70:846–56

246. Mathai S, Booth LC, Davidson JO, et al. Acute on chronic exposure toendotoxin in preterm fetal sheep. Am J Physiol Regul Integr CompPhysiol 2013;304:R189R97

247. Kapitanovi�c VH, Catela IT, VZ, et al. COX-1 and COX-2 polymor-phisms in susceptibility to cerebral palsy in very preterm infants. MolNeurobiol 2017;54:930–8

248. Johnston MV, Hoon AH. Cerebral palsy. Neuromolecular Med 2006;8:435–50

249. Huria T, Beeraka NM, Al-Ghamdi B, et al. Premyelinated central axonsexpress neurotoxic NMDA receptors: relevance to early developingwhite-matter injury. J Cereb Blood Flow Metab 2015;35:543–53

250. Holmes GL, Ben-Ari Y. Seizures in the developing brain: perhaps notso benign after all. Neuron 1998;21:1231–4

251. Holmes GL, Ben-Ari Y. The neurobiology and consequences of epi-lepsy in the developing brain. Pediatr Res 2001;49:320–5

252. Ravizza T, Rizzi M, Perego C, et al. Inflammatory response and glia ac-tivation in developing rat hippocampus after status epilepticus. Epilep-sia 2005;46:113–7

253. Fukuda M, Suzuki Y, Ishizaki Y, et al. Interleukin-1b enhances suscep-tibility to hyperthermia-induced seizures in developing rats. Seizure2009;18:211–4

254. Galic MA, Riazi K, Pittman QJ. Cytokines and brain excitability. FrontNeuroendocrinol 2012;33:116–25

255. Choy M, Dube CM, Ehrengruber M, et al. Inflammatory processes, fe-brile seizures, and subsequent epileptogenesis. Epilepsy Curr 2014;14:15–22

256. Vezzani A, Balosso S, Ravizza T. The role of cytokines in the patho-physiology of epilepsy. Brain Behav Immun 2008;22:797–803

257. Kim I, Mlsna LM, Yoon S, et al. A postnatal peak in microglial devel-opment in the mouse hippocampus is correlated with heightened sensi-tivity to seizure triggers. Brain Behav 2015;5:e0040

258. Vezzani A, Auvin S, Ravizza T, et al. Glia–neuron interactions in epi-lepsy: inflammatory mediators. Epilepsia 2010;51:55

J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017 Microglia in the Developing Brain

17

Page 18: Biology of Microglia in the Developing Brain › 9188 › 1c529130e23f419351... · 2017-10-09 · Biology of Microglia in the Developing Brain Charanjit Kaur, PhD, Gurugirijha Rathnasamy,

259. Rossi S, Furlan R, De Chiara V, et al. Interleukin-1b causes synaptichyperexcitability in multiple sclerosis. Ann Neurol 2012;71:76–83.

260. Lee RH, Mills EA, Schwartz N, et al. Neurodevelopmental effects ofchronic exposure to elevated levels of pro-inflammatory cytokines in adeveloping visual system. Neural Dev 2010;5:2

261. Kim HJ, Cho MH, Shim WH, et al. Deficient autophagy in microgliaimpairs synaptic pruning and causes social behavioral defects. MolPsychiatry 2016. doi: 10.1038/mp.2016.103

262. Zhan Y, Paolicelli RC, Sforazzini F, et al. Deficient neuron-microgliasignaling results in impaired functional brain connectivity and social be-havior. Nat Neurosci 2014;17:400–6

263. Nakagawa Y, Chiba K. Involvement of neuroinflammation duringbrain development in social cognitive deficits in autism spectrumdisorder and schizophrenia. J Pharmacol Exp Ther 2016;358:504–15

264. Theoharides TC, Zhang B. Neuro-inflammation, blood-brain barrier,seizures and autism. J Neuroinflammation 2011;8:168

265. Hagberg H, Gressens P, Mallard C. Inflammation during fetal and neo-natal life: Implications for neurologic and neuropsychiatric disease inchildren and adults. Ann Neurol 2012;71:444–57

266. Theoharides TC, Asadi S, Patel AB. Focal brain inflammation and aut-ism. J Neuroinflammation 2013;10:46

267. Theoharides T, Tsilioni I, Patel A, et al. Atopic diseases and inflamma-tion of the brain in the pathogenesis of autism spectrum disorders.Transl Psychiatry 2016;6:e844

268. Vargas DL, Nascimbene C, Krishnan C, et al. Neuroglial activation andneuroinflammation in the brain of patients with autism. Ann Neurol2005;57:67–81

269. Morgan JT, Chana G, Pardo CA, et al. Microglial activation andincreased microglial density observed in the dorsolateral prefrontal cor-tex in autism. Biol Psychiatry 2010;68:368–76

270. Rodriguez JI, Kern JK. Evidence of microglial activation in autism andits possible role in brain underconnectivity. Neuron Glia Biol 2011;7:205–13

271. Takano T. Role of microglia in autism: recent advances. Dev Neurosci2015;37:195–202

272. Koyama R, Ikegaya Y. Microglia in the pathogenesis of autism spec-trum disorders. Neurosci Res 2015;100:1–5

273. Dong H, Zhang W, Zeng X, et al. Histamine induces upregulated ex-pression of histamine receptors and increases release of inflammatorymediators from microglia. Mol Neurobiol 2014;49:1487–500

274. Skaper S, Facci L, Giusti P. Neuroinflammation, microglia and mastcells in the pathophysiology of neurocognitive disorders: a review. CNSNeurol Disord Drug Targets 2014;13:1654–66

275. Dong H, Zhang X, Wang Y, et al. Suppression of brain mast cells de-granulation inhibits microglial activation and central nervous system in-flammation. Mol Neurobiol 2016;54:997–1007

276. Fern�andez-L�opez D, Faustino J, Klibanov AL, et al. Microglial cellsprevent hemorrhage in neonatal focal arterial stroke. J Neurosci 2016;36:2881

277. Biran V, Joly LM, Heron A, et al. Glial activation in white matter follow-ing ischemia in the neonatal P7 rat brain. Exp Neurol 2006;199:103–12

278. Parakalan R, Jiang B, Nimmi B, et al. Transcriptome analysis of amoeb-oid and ramified microglia isolated from the corpus callosum of ratbrain. BMC Neurosci 2012;13:64

279. Gupta S, Ellis SE, Ashar FN, et al. Transcriptome analysis reveals dys-regulation of innate immune response genes and neuronal activity-dependent genes in autism. Nat Commun 2014;5:5748

280. Nardone S, Sharan Sams D, Reuveni E, et al. DNA methylation analysisof the autistic brain reveals multiple dysregulated biological pathways.Transl Psychiatry 2014;4:e433

281. Nagarajan RP, Hogart AR, Gwye Y, et al. Reduced MeCP2 expressionis frequent in autism frontal cortex and correlates with aberrant MECP2promoter methylation. Epigenetics 2006;1:e1

282. Maezawa I, Jin LW. Rett syndrome microglia damage dendrites andsynapses by the elevated release of glutamate. J Neurosci 2010;30:5346

283. Zhang L, Dong LY, Li YJ, et al. miR-21 represses FasL in microgliaand protects against microglia-mediated neuronal cell death followinghypoxia/ischemia. Glia 2012;60:1888–95

284. Ponomarev ED, Veremeyko T, Barteneva N, et al. MicroRNA-124 pro-motes microglia quiescence and suppresses EAE by deactivating mac-rophages via the C/EBP-alpha-PU.1 pathway. Nat Med 2011;17:64–70

285. Oberlander TF, Weinberg J, Papsdorf M, et al. Prenatal exposure to ma-ternal depression, neonatal methylation of human glucocorticoid recep-tor gene (NR3C1) and infant cortisol stress responses. Epigenetics2008;3:97–106

Kaur et al. J Neuropathol Exp Neurol • Volume 0, Number 0, Month 2017

18