throbber
Microglia activation in retinal degeneration
`Thomas Langmann1
`Institute of Human Genetics, University of Regensburg, Regensburg, Germany
`
`Abstract: Microglia cells are phagocytic sentinels
`in the CNS and in the retina required for neuronal
`homeostasis and innate immune defense. Accumu-
`lating experimental evidence suggests that chronic
`microglia activation is associated with various neu-
`rodegenerative diseases including retinal dystro-
`phies. Endogenous triggers alert microglia cells
`rapidly in the degenerating retina, leading to local
`proliferation, migration, enhanced phagocytosis,
`and secretion of cytokines, chemokines, and neu-
`rotoxins. This amplified, immunological cascade
`and the loss of limiting control mechanisms may
`contribute significantly to retinal tissue damage
`and proapoptotic events. This review summarizes
`the developmental and immune surveillance func-
`tions of microglia in the healthy retina and dis-
`cusses early signaling events and transcriptional
`networks of microglia activation in retinal degen-
`eration. The characterization of activation path-
`ways at the molecular level may lead to innovative,
`therapeutic options in degenerative retinal dis-
`eases based on a selective, pharmacological in-
`terference with the neurotoxic activities of micro-
`glia cells, without compromising their homeostastic
`functions. J. Leukoc. Biol. 81: 1345–1351; 2007.
`
`Key Words: neuronal homeostasis 䡠 Toll-like receptors 䡠 early
`growth response factor 1
`
`SCOPE OF THIS REVIEW
`
`The concept of microglia activation in neuropathological con-
`ditions has changed profoundly over the last few years. Initially
`thought as bystander cells with only marginal causal effects on
`neurodegeneration, an exaggerated microglia reaction is now
`thought to contribute significantly or even trigger neuronal
`apoptosis in several diseases including retinal dystrophies [1].
`Furthermore, physiological, neurotrophic microglia function
`and microglia-neuron cross-talk are now known as prerequi-
`sites for maintaining the immune privilege of the CNS and the
`retina. Loss of this regulation could also be a major cause for
`tissue damage. Based on these findings, microglia-targeted
`treatment could be envisioned to inhibit overactivation and
`support neuronal survival and tissue regeneration.
`Several excellent review articles provide a comprehensive
`overview over microglia physiology and pathology in the brain,
`especially related to Alzheimer’s disease, Parkinson’s disease,
`and multiple sclerosis [2–5]. In contrast, the homeostatic func-
`tions of retinal microglia and the contribution of overactivated
`
`0741-5400/07/0081-1345 © Society for Leukocyte Biology
`
`resident microglia to retinal neurodegeneration are less well
`covered. Therefore, this review will focus on the role of micro-
`glia-related mechanisms in the healthy retina and will empha-
`size the involvement of microglia in the process of retinal
`degeneration.
`
`MICROGLIA CELLS IN THE DEVELOPING CNS
`AND THE RETINA
`
`It is now generally accepted that microglia cells are part of the
`mononuclear phagocyte system in the parenchyma of the CNS
`including the retina [6]. As shown by bone marrow transplan-
`tation experiments and in vivo labeling assays, microglia pre-
`cursors migrate through immature blood vessels into the retinal
`tissue during late embryonic development and in the early
`postnatal period [7]. The colonization of microglia precursors is
`coordinated by a spatially and temporally regulated expression
`of
`the chemokines MCP-1 and RANTES [8]. Besides the
`remodeling of extracellular matrix (ECM) components during
`the formation of the neuron-glia network, the main functions of
`microglia in the developing retina are phagocytosis and elim-
`ination of cellular debris from apoptotic neurons in the gan-
`glion cell layer and inner nuclear layer.
`Using an antibody against the macrophage marker F4/80,
`Hume et al. [9] could demonstrate the migration of amoeboid-
`shaped microglia cells with short and broad processes and their
`colocalization with pyknotic nuclei of dying cells shortly after
`birth. Studies in embryonic chicken retina demonstrated that
`the developmental neuronal cell death is reduced strongly in
`retinal explants lacking microglia cells and that microglia-
`derived nerve growth factor (NGF) is required to induce apo-
`ptosis [10]. Furthermore, microglia are able to initiate a cell
`death program in vascular cells of the developing eye by
`activating the canonical wingless pathway [11] and also to
`regulate synaptogenesis in early CNS development [12]. The
`signaling adaptor protein, DNAX-activating protein of 12 kDa
`(DAP12)/killer cell-activating receptor-associated protein/ty-
`rosine kinase-binding protein,
`is expressed specifically on
`amoeboid microglia, and its loss of function impairs the brain-
`derived neurotrophic factor (BDNF)/tyrosine kinase receptor B
`synaptic pathway, leading to developmental defects in synap-
`togenesis [13]. It
`is interesting that DAP12 together with
`
`1 Correspondence: Institute of Human Genetics, University of Regensburg,
`Franz-Josef-Strauss-Allee
`11,
`93053 Regensburg, Germany. E-mail:
`thomas.langmann@klinik.uni-regensburg.de
`Received February 15, 2007; accepted March 25, 2007.
`doi: 10.1189/jlb.0207114
`
`Journal of Leukocyte Biology Volume 81, June 2007 1345
`CYAN EXHIBIT 1067
`
`

`

`triggering receptor expressed on myeloid cells 2 are also re-
`quired to potentiate phagocytosis, to clear apoptotic neurons,
`and to block an exaggerated response to TLR signaling after
`microbial challenge or CNS injury [14, 15]. The scavenger
`receptors CD36, SR-A, and SR-BI are further microglia recep-
`tors involved in the engulfment and uptake of cell corpses and
`membrane debris from developmentally dying neurons [16].
`As development is completed, these activated phagocytes
`are transformed into resting microglia cells, constituting
`5–20% of the overall glial population. Because of their unique
`neuronal microenvironment, microglia differ from other tissue
`macrophages by their high capability of proliferation in situ
`and their unique, characteristic ramified morphology. Specific
`interactions with astrocytes, their ECM components, and their
`receptor-mediated responses to astrocyte-secreted factors in-
`cluding GM-CSF and M-CSF favor microglia proliferation,
`survival, and differentiation [17, 18].
`In the adult retina, ramified microglia are found in regular,
`ordered structures in inner and outer plexiform layers as shown
`by F4/80 immunohistochemistry [9] and cellular tracing exper-
`iments with fluorescent dyes [19]. When retinal ganglion cells
`(RGCs) were labeled retrogradely with the lipophilic dye 4Di-
`10ASP, microglia cells containing fluorescent RGC debris
`could still be observed after 10 months, indicating a long
`period of survival of the local microglia population [19 –21].
`Further support for microglial stability and local repopulation
`in the retina comes from bone marrow transplantation experi-
`ments using chimeric (Y3 X) Lewis rats. In this model, a
`rapid, chimeric repopulation of hematopoietic cells in the
`spleen and the lung of lethally irradiated and transplanted
`female recipient rats has been observed, whereas no chimerism
`could be detected in the brain parenchyma and the retina until
`52 weeks of transplantation [22]. Similarly, engraftment studies
`with lacZ-expressing murine bone marrow cells indicate a slow
`turnover of microglia cells compared with other resident tissue
`macrophages with local proliferation capacity such as alveloar
`macrophages and Langerhans cells [23, 24]. These studies also
`show that bone marrow-derived monocytes do not typically
`cross the blood-retina barrier in the healthy retina.
`
`CROSS-TALK BETWEEN RESTING MICROGLIA
`AND RETINAL CELLS
`
`Different types of glia cells with neuro-supporting function are
`present in the CNS, including oligodendrocytes, astrocytes,
`Mu¨ller cells, and microglia cells, which are cells with a role in
`immunocompetent defense and also play an active part in the
`support of surrounding cells [3]. The immunological potential
`of microglia is comparable with blood monocytes and other
`tissue macrophages in terms of secretory functions. However,
`resting microglia express lower levels of costimulatory mole-
`cules and possess relatively low phagocytic activity [25]. The
`maintenance of the normal retinal immune regulation seems to
`actively involve cytokines from the retinal pigment epithelium
`such as TGF-␤ (Fig. 1). This cytokine contributes to the
`immune privilege by predisposing microglia to the preferential
`production of IL-10, which in turn down-regulates antigen-
`presenting molecules including MHC-II, CD80, and CD86
`
`[26]. TGF-␤ also directs TNF/IFN-␥-stimulated microglia cells
`to an anti-inflammatory phenotype by broadly blocking inflam-
`matory gene expression [27].
`Another important control mechanism to limit microglia
`activation in the healthy retina relies on the Ig superfamily
`domain-containing molecule CD200, which is recognized by
`the well-known mAb OX2 and its receptor on microglia [28]
`(Fig. 1). CD200 is a transmembrane glycoprotein with a short
`cytoplasmic tail without signaling motifs and is expressed on
`many different cell types including neurons, endothelial cells,
`lymphocytes, and dendritic cells [29, 30]. The CD200R is
`expressed exclusively on myeloid cells, and ligand-binding via
`cellular contact triggers signaling events, sustaining the basal,
`deactivated state with mainly homeostatic functions of macro-
`phages and microglia cells [31, 32]. Strong evidence for potent
`inhibition of microglia activity comes from CD200 knockout
`mice, which display an expansion of the myeloid population in
`several
`tissues and increased expression of
`the activation
`markers DAP12, CD11b, CD45, CD68, and inducible NO
`synthase (iNOS) on brain microglia [31]. A broad, constitutive
`expression of CD200 on retinal vessel endothelium and neu-
`rons and the induction of CD200R expression during retinal
`inflammation also strongly implicate CD200/CD200R interac-
`tions in the retina [33]. Furthermore, the potential of microglia
`to migrate and to secrete the anti-inflammatory cytokine IL-10
`following LPS/IFN-␥ stimulation requires CD200R, as shown
`by the potent inhibitory activity of CD200-Fc fusion proteins
`when added to retinal explants [34].
`In the quiescent state and following activation, microglia
`cells secrete several polypeptide neurotrophic factors, which
`impact the physiology and survival of neurons. Among these
`factors, BDNF, ciliary neurotrophic factor (CNTF), glial cell
`line-derived neurotrophic factor (GDNF), NGF, neurotrophin-3
`(NT3), and basic fibroblast growth factor (bFGF) have been
`shown to protect and regulate the survival of photoreceptors
`[35] (see Table 1). It is important that a functional microglia-
`Mu¨ller glia cell interaction is required for these trophic factors
`to work in an autocrine and paracrine manner [36]. Additional
`microglia-derived factors that stimulate the survival and regen-
`eration of retinal ganglion cells after nerve injury have been
`identified using coculture of neurons with microglia-condi-
`tioned medium [37]. One of
`these neuronal growth factor
`proteins secreted by microglia is the small Ca2⫹-binding pro-
`tein oncomodulin, which acts through a Ca2⫹/calmodulin ki-
`nase-dependent signaling pathway to stimulate neurite regen-
`eration of RGCs [38].
`
`IMMUNE SURVEILLANCE FUNCTION OF
`RESTING MICROGLIA
`
`In contrast to an initially prevailing view that mature microglia
`cells are in a dormant state when deactivated in the healthy
`CNS, these “resting” cells monitor their environment with
`highly motile protrusions to clear metabolic products and tis-
`sue debris [39]. Microglia can sense their microenvironment
`through various surface proteins including receptors for im-
`mune components such as complement, cytokines, chemo-
`kines, antibodies, and adhesion molecules [3] (see Table 1). It
`
`1346 Journal of Leukocyte Biology Volume 81, June 2007
`
`http://www.jleukbio.org
`
`

`

`Fig. 1. Mechanisms of microglia quiescence and activation related to neuronal degeneration. Ramified microglia are kept in a resting, stand-by mode to regulate
`retinal homeostasis by intraretinal cell contacts and soluble factors from neurons, astrocytes, and the retinal pigment epithelium. Several triggers from retinal
`degeneration can initiate TLR signaling leading to microglia activation, proliferation, and migration. Transformed amoeboid microglia secrete various bioactive
`molecules, initially active in tissue repair. Chronic activation may lead to exaggerated microglia responses, leading to retinal damage and neuronal apoptosis. Egr1,
`Early growth response factor 1; CD220R, CD200 receptor; ROS, reactive oxygen species.
`
`is important that the cells also possess receptors for the che-
`mokine fractalkine (CX3CL1) and for purine nucleotides [4]. A
`constitutive release of fractalkine from healthy neurons, which
`bind to CX3CR1 on microglia cells, seems to regulate micro-
`glia homeostasis, and loss of CX3CR1 leads to neurotoxicity
`and degeneration [40]. Fractalkine may also serve as a neuro-
`nal stress signal to induce intimate neuron-glia cross-talk,
`resulting in the release of trophic molecules such as TGF-␤1,
`which facilitates neuronal tissue regeneration [41]. As fractal-
`kine also exists as a membrane-bound molecule on neurons,
`direct cell-cell contact and interaction with the ECM could
`additionally restrain microglia activation via this pathway [42].
`The Gi-coupled P2Y12 purinergic receptor is highly ex-
`pressed on resting microglia in the brain but not other tissue
`macrophages. Membrane-anchored P2Y12 allows detection of
`ATP and ADP release from damaged neurons and is required
`for subsequent early activation mechanisms [43]. Mice lacking
`P2Y12 show normal prevalence, distribution, and morphology
`of resting microglia; however, no membrane ruffling and filo-
`podia extension occur upon stimulation with ADP or ATP.
`P2Y12 is down-regulated actively during microglial transfor-
`mation from the resting to the amoeboid state, and thus, the
`P2Y12 receptor may be a novel molecular marker for visual-
`izing microglia in their ramified state [43]. These mechanisms
`observed in the brain likely occur also in retinal microglia, as
`several P2Y receptor subtypes have been detected in the retina
`at the mRNA and protein level [44].
`
`MICROGLIA ACTIVATION IN RETINAL
`DEGENERATION MODELS
`
`Early microglia activation in the retina is a common response
`to ocular infections, autoimmune mechanisms, neuronal injury,
`ischemia, and metabolic as well as hereditary retinopathies,
`which are all associated with progressive neurodegeneration
`[1]. Activated microglia exhibit strongly enhanced prolifera-
`tion, migration, phagocytosis, and production of many different
`bioactive molecules (Table 1 and Fig. 1). The morphological
`change from ramified cells to amoeboid phagocytes is accom-
`panied by the expression of several surface markers such as
`F4/80, complement receptor 3 (CD11b/CD18, OX42), MHC-II
`(OX6), CD68, and Griffonia simplicifolia isolectin B4, which
`have been used classically to detect microglia activity by
`immunohistochemistry and immunofluorescence-staining pro-
`cedures [2, 46].
`There are now several studies [47, 48], including work from
`my own group [49], demonstrating early microglia activation in
`animal models of inherited photoreceptor degeneration. Zeiss
`and Johnson [47] could show prominent microglia migration
`and proliferation in the outer nuclear layer of retinal degener-
`ation mice. Furthermore, increased expression of the micro-
`glia-activating chemokines MCP-1, MCP-3, and RANTES as
`well as high levels of microglia-secreted TNF were observed in
`the retina of these mice well before the onset of photoreceptor
`apoptosis [48]. A recent investigation of our group, reported on
`
`Langmann Microglia quiescence and activation in the retina 1347
`
`

`

`TABLE 1. Selection of Microglia-Expressed Receptors and Secreted Factors
`
`Molecule class
`
`Scavenger receptors
`Ig receptors
`Ig-like receptors
`
`Complement receptors
`
`Growth factor receptors
`Chemokine and cytokine receptors
`
`Purinergic receptors
`
`TLRs
`Antigen-presenting proteins
`Cytokines and chemokines
`
`Neurotransmitters and neurotoxins
`Neurotrophins
`
`Molecules
`
`Functions in microglia
`
`SR-A, SR-BI, CD36
`Fc␥RI, Fc␥RII, Fc␥RIII, Fc⑀RI
`CD200R
`
`C3bi-R (CR3, CD11b, CD18), C1q-R,
`C3a-R, C5a-R
`M-CSF-R, GM-CSF-R
`CX3CR1 (Fractalkine-R), TGF-␤-R,
`CCR2 (MCP-1-R), CCR5 (RANTES-R)
`Receptors for P2X4, P2X7, P2Y1, P2Y2,
`P2Y4, P2Y6, P2Y12
`TLR1–9
`MHC-II, CD80, CD86
`TNF, TGF-␤, IL-1␣/␤, IL-3, IL-6, IL-8,
`IL-10, IL-12, IL-15, IL-18, GRO␣,
`IP-10, MCP-1, MDC, M-CSF, MIP-1␣/
`␤, MIP-2, MIP-3␤, RANTES
`Glutamate, NO, ROS
`BDNF, CNTF, GDNF, NGF, NT3, bFGF,
`oncomodulin
`
`Uptake of cell debris and apoptotic corpses
`Uptake of Ig-opsonized particles
`Deactivation of resting cells upon ligation
`with CD200
`Uptake of complement components and
`opsonized particles
`Proliferation and survival
`Neuron-microglia signaling and immune
`signaling
`Purinergic sensing and early activation
`
`Pathogen and damage-associated activation
`Presentation of phagocytosed material
`Immunostimulation
`
`Cellular toxicity
`Neurotrophic signals
`
`Summarized and adapted from refs. [3, 4, 28, 45]. GRO␣, Growth-related oncogene ␣; IP-10, IFN-inducible protein 10; MDC, macrophage-derived chemokine.
`
`the retinal gene expression profile in the murine model of
`X-linked juvenile retinoschisis, was analyzed [49, 50]. A hall-
`mark of the retinoschisin-deficient murine retina is the pro-
`gressive loss of cone and rod photoreceptor cells [50] as a
`result of apoptotic events peaking at approximately Postnatal
`Day 18 [51]. Using DNA-microarray analyses, we could iden-
`tify several transcripts from activated microglia cells preceding
`gene expression patterns related to apoptosis in the diseased
`retina. Besides detrimental proinflammatory cytokines, chemo-
`kines, complement components, and scavenger receptors, we
`have also identified self-regulatory mechanisms by prominent
`expression of the macrophage deactivation gene DAP12 (see
`above) and caspase 11, which have been shown to mediate
`overactivation-induced microglial cell death [52, 53]. Related
`to our study, partially overlapping gene expression patterns
`reflecting activated microglia and indicating common tran-
`scriptional mechanisms have been identified in light-induced
`retinal degeneration [54], glaucoma models [55, 56], retinal
`needle injury [57], and neurodegeneration in the brain of
`Sandhoff disease [58] and mucopolysaccharidoses [59].
`Based on these data of early microglia responses, it can be
`hypothesized that activated microglia cells are not simply
`bystanders of neurodegeneration but instead, may be in-
`volved significantly in the initiation and perpetuation of the
`degenerative process.
`As an important tool to analyze ex vivo microglia character-
`istics in early stages of retinal dystrophies, Roque and Cald-
`well [60] developed an isolation and culture model of retinal
`microglia cells from Royal College of Surgeons (RCS) rats, an
`established model of genetic photoreceptor dystrophy. The
`phagocytic potential of the isolated cells, monitored by the
`ingestion of latex beads and the ability to proliferate in vitro,
`was dependent on the presence of recombinant M-CSF in the
`culture medium [60]. Our group has adapted this protocol for
`the isolation of early postnatal microglia cells from retinoschi-
`
`is interesting that we
`sin-deficient and wild-type mice. It
`observed the preservation of the activated microglia phenotype
`defined by amoeboid morphology and specific gene expression
`patterns only in growth medium supplemented with M-CSF
`(unpublished observations). Thus, M-CSF, which acts via the
`Fms tyrosine kinase receptor on myeloid cells, seems to rep-
`resent a key factor for the initiation and maintenance of mi-
`croglia activation, independent of the initial trigger. Supporting
`this view, overexpression of M-CSF-R in microglia cell lines
`results in proliferation and strongly increased expression of
`iNOS, IL-1␤, MIP-1␣, IL-6, and M-CSF itself [61]. As another
`consequence of M-CSF-R ligation, the myeloid-specific down-
`stream signaling molecule Iba1 causes Rac-dependent, dy-
`namic remodeling of the actin cytoskeleton, resulting in mem-
`brane ruffling, which is required for efficient phagocytosis [62].
`To study whether retinal microglia cells from RCS rats can
`induce photoreceptor apoptosis in vitro, coculture experiments
`with microglia-conditioned medium and the 661w photorecep-
`tor cell line were performed by Roque et al. [63]. In contrast to
`medium from isolated Mu¨ller cells, addition of cell culture
`supernatant from activated microglia cells resulted in a signif-
`icant increase of apoptosis-related cell death in 661w cells
`[63]. This effect was dependent on microglia-derived NGF and
`p75 neurotrophin receptors (p75NTR) on photoreceptor cells.
`It is noteworthy that augmented NGF and p75NTR mRNA
`levels were also detected in dystrophic compared with normal
`retinae [64]. These data clearly show that microglia activation
`can trigger neuronal cell death. It remains to be determined,
`however, whether the same factors are also involved in apo-
`ptosis induction in other retinal dystrophies. Furthermore, as
`immortalized 661w cells only express a subset of cone-specific
`photoreceptor markers [65], the observed in vitro effects need
`to be verified in vivo with cultures of purified, primary rods and
`cones.
`
`1348 Journal of Leukocyte Biology Volume 81, June 2007
`
`http://www.jleukbio.org
`
`

`

`EARLY SIGNALING EVENTS AND
`TRANSCRIPTIONAL NETWORKS IN
`MICROGLIA ACTIVATION
`
`To identify triggers in early microglia activation, mainly in
`vitro culture models with primary cells and several immortal-
`ized cell lines such as BV-2, HMO6, and CHME were used
`[66]. After stimulation, the read-out parameters for enhanced
`microglia activation were cell proliferation, phagocytosis, mi-
`gration, expression of surface receptors, and the release of
`cytokines, chemokines, prostaglandins, NO, superoxide an-
`ions, and glutamate [53]. The majority of these microglia-
`secreted molecules can cause progressive neurodegeneration
`upon chronic exposition. The most potent natural triggers for
`microglia activation include LPS, proinflammatory cytokines
`including IFN-␥ and TNF, complement components, thrombin,
`and aggregated, insoluble peptides [1]. It is important that LPS
`seems to be the strongest inducer of microglia cells, and LPS
`signaling can be initiated, even in the absence of exogenous,
`infectious agents.
`It is becoming increasingly recognized that the pattern rec-
`ognition receptors of the TLR family, which are expressed
`broadly on microglia, can react to aberrant endogenous ligands
`in neuronal tissues [67]. Gangliosides, hyaluronic acid, hepa-
`ran sulfate, and heat shock proteins carry damage-associated
`molecular patterns and thereby can elicit microglia activation
`[68]. It is thus tempting to speculate that early alarm signals
`from degenerating neuronal tissues might initiate TLR-depen-
`dent activation and hyperactivation of microglia, which may
`even lead to attacks against healthy neurons (Fig. 1). In line
`with this hypothesis, we have detected in the retina of retinos-
`chisin-deficient mice early microglial TLR4 induction and a
`strong up-regulation of activation-related transcripts, likely
`leading to neurotoxicity and consecutively, photoreceptor ap-
`optosis [49]. It is interesting that TLR4-dependent pathways in
`microglia activation have also been connected to other models
`of neuronal injury including spinal nerve transection and pain-
`ful neuropathy [69, 70]. It is noteworthy that TLR4 and TLR2
`also control autoregulatory mechanisms by triggering the in-
`duction of microglia apoptosis in vitro [71]; however, the con-
`tribution of this pathway to prevent latent microglia overacti-
`vation remains to be determined in vivo.
`A novel candidate for downstream signaling of TLR4 in
`overactivated microglia is Egr1, which is also required for
`ocular tissue development [72, 73]. Egr1 is strongly up-regu-
`lated in several retinal dystrophies including retinoschisin
`deficiency [49]. Egr1 expression is LPS-responsive in the mi-
`croglia cell line BV-2 and ex vivo-isolated retinal primary
`microglia (own unpublished observations). Furthermore, sev-
`eral microglia genes induced upon activation contain predicted
`binding sites for Egr1 in their promoter regions. To study the in
`vivo effects of Egr1 in the retina, mice lacking retinoschisin
`and Egr1 were generated [49]. It is unexpected that no major
`differences in retinal microglia activation could be detected in
`these animals compared with retinoschisin-deficient mice [49].
`A likely explanation for this finding is the functional redun-
`dance of the Egr family members. Indeed, a recent study by
`Laslo et al. [74] showed that Egr1 and Egr2 are exchangeable
`in activating macrophage genes and that only Egr1⫺/⫺Egr2-/⫹
`
`hematopoietic progenitors are defective in M-CSF-dependent
`macrophage differentiation.
`
`CONCLUSIONS AND PERSPECTIVES
`
`Similar to the CNS, microglia cells in the retina are sensors for
`disturbances in their neuronal environment. Their balanced
`activities initially contribute to neuronal protection and tissue
`regeneration. Continuous stimulation with alarm signals from
`exogenous and endogenous sources can lead to chronic over-
`activation and loss of autoregulatory mechanisms, which are
`detrimental for neurons. In vitro culture systems and animal
`models of retinal degeneration have provided a first insight into
`specific gene expression profiles associated with microglia
`activation. To understand further the processes and triggers of
`microglia activation, molecular characterization of distinct mi-
`croglia populations using cell isolation and sorting techniques
`combined with large-scale gene expression studies might be
`useful. This should enhance the identification of novel biolog-
`ical markers specific for resting, activated, or overactivated
`cells and may allow unraveling of genetic networks and key
`transcription factors controlling the different steps of microglia
`transformation. Furthermore, these studies may provide novel
`treatment options for selective inhibition of overshooting mi-
`croglia activity and preserving the trophic and homeostatic
`functions of these important immune cells.
`
`ACKNOWLEDGMENT
`
`The author thanks Prof. Bernhard Weber for discussions and
`critical reading of the manuscript.
`
`REFERENCES
`
`1. Schuetz, E., Thanos, S. (2004) Microglia-targeted pharmacotherapy in
`retinal neurodegenerative diseases. Curr. Drug Targets 5, 619 – 627.
`2. Kreutzberg, G. W. (1996) Microglia: a sensor for pathological events in the
`CNS. Trends Neurosci. 19, 312–318.
`3. Streit, W. J. (2002) Microglia as neuroprotective, immunocompetent cells
`of the CNS. Glia 40, 133–139.
`4. van Rossum, D., Hanisch, U. K. (2004) Microglia. Metab. Brain Dis. 19,
`393– 411.
`5. Kim, Y. S., Joh, T. H. (2006) Microglia, major player in the brain
`inflammation: their roles in the pathogenesis of Parkinson’s disease. Exp.
`Mol. Med. 38, 333–347.
`6. Oehmichen, M.
`(1982) Are resting and/or reactive microglia macro-
`phages? Immunobiology 161, 246 –254.
`7. Barron, K. D. (1995) The microglial cell. A historical review. J. Neurol.
`Sci. 134 (Suppl.), 57– 68.
`8. Rezaie, P., Male, D. (1999) Colonization of the developing human brain
`and spinal cord by microglia: a review. Microsc. Res. Tech. 45, 359 –382.
`9. Hume, D. A., Perry, V. H., Gordon, S. (1983) Immunohistochemical
`localization of a macrophage-specific antigen in developing mouse retina:
`phagocytosis of dying neurons and differentiation of microglial cells to
`form a regular array in the plexiform layers. J. Cell Biol. 97, 253–257.
`10. Frade, J. M., Barde, Y. A. (1998) Microglia-derived nerve growth factor
`causes cell death in the developing retina. Neuron 20, 35– 41.
`11. Lobov, I. B., Rao, S., Carroll, T. J., Vallance, J. E., Ito, M., Ondr, J. K.,
`Kurup, S., Glass, D. A., Patel, M. S., Shu, W., Morrisey, E. E., McMahon,
`A. P., Karsenty, G., Lang, R. A. (2005) WNT7b mediates macrophage-
`induced programmed cell death in patterning of the vasculature. Nature
`437, 417– 421.
`
`Langmann Microglia quiescence and activation in the retina 1349
`
`

`

`12. Bessis, A., Bechade, C., Bernard, D., Roumier, A. (2007) Microglial
`control of neuronal death and synaptic properties. Glia 55, 233–238.
`13. Roumier, A., Bechade, C., Poncer, J. C., Smalla, K. H., Tomasello, E.,
`Vivier, E., Gundelfinger, E. D., Triller, A., Bessis, A. (2004) Impaired
`synaptic function in the microglial KARAP/DAP12-deficient mouse.
`J. Neurosci. 24, 11421–11428.
`14. Takahashi, K., Rochford, C. D., Neumann, H. (2005) Clearance of apo-
`ptotic neurons without inflammation by microglial triggering receptor
`expressed on myeloid cells-2. J. Exp. Med. 201, 647– 657.
`15. Klesney-Tait, J., Turnbull, I. R., Colonna, M. (2006) The TREM receptor
`family and signal integration. Nat. Immunol. 7, 1266 –1273.
`16. Husemann, J., Loike, J. D., Anankov, R., Febbraio, M., Silverstein, S. C.
`(2002) Scavenger receptors in neurobiology and neuropathology: their role
`on microglia and other cells of the nervous system. Glia 40, 195–205.
`17. Perry, V. H., Gordon, S. (1988) Macrophages and microglia in the nervous
`system. Trends Neurosci. 11, 273–277.
`18. Schmidtmayer, J., Jacobsen, C., Miksch, G., Sievers, J. (1994) Blood
`monocytes and spleen macrophages differentiate into microglia-like cells
`on monolayers of astrocytes: membrane currents. Glia 12, 259 –267.
`19. Bodeutsch, N., Thanos, S. (2000) Migration of phagocytotic cells and
`development of the murine intraretinal microglial network: an in vivo
`study using fluorescent dyes. Glia 32, 91–101.
`20. Thanos, S. (1991) The relationship of microglial cells to dying neurons
`during natural neuronal cell death and axotomy-induced degeneration of
`the rat retina. Eur. J. Neurosci. 3, 1189 –1207.
`21. Thanos, S., Kacza, J., Seeger, J., Mey, J. (1994) Old dyes for new scopes:
`the phagocytosis-dependent long-term fluorescence labeling of microglial
`cells in vivo. Trends Neurosci. 17, 177–182.
`22. Albini, T. A., Wang, R. C., Reiser, B., Zamir, E., Wu, G. S., Rao, N. A.
`(2005) Microglial stability and repopulation in the retina. Br. J. Ophthal-
`mol. 89, 901–903.
`23. Kennedy, D. W., Abkowitz, J. L. (1997) Kinetics of central nervous system
`microglial and macrophage engraftment: analysis using a transgenic bone
`marrow transplantation model. Blood 90, 986 –993.
`24. Kennedy, D. W., Abkowitz, J. L. (1998) Mature monocytic cells enter
`tissues and engraft. Proc. Natl. Acad. Sci. USA 95, 14944 –14949.
`25. Dick, A. D., Carter, D., Robertson, M., Broderick, C., Hughes, E., For-
`rester, J. V., Liversidge, J. (2003) Control of myeloid activity during retinal
`inflammation. J. Leukoc. Biol. 74, 161–166.
`26. D’Orazio, T. J., Niederkorn, J. Y. (1998) A novel role for TGF-␤ and IL-10
`in the induction of immune privilege. J. Immunol. 160, 2089 –2098.
`27. Paglinawan, R., Malipiero, U., Schlapbach, R., Frei, K., Reith, W.,
`Fontana, A. (2003) TGF␤ directs gene expression of activated microglia to
`an anti-inflammatory phenotype strongly focusing on chemokine genes and
`cell migratory genes. Glia 44, 219 –231.
`28. Nathan, C., Muller, W. A. (2001) Putting the brakes on innate immunity:
`a regulatory role for CD200? Nat. Immunol. 2, 17–19.
`29. Clark, M. J., Gagnon, J., Williams, A. F., Barclay, A. N. (1985) MRC OX-2
`antigen: a lymphoid/neuronal membrane glycoprotein with a structure like
`a single immunoglobulin light chain. EMBO J. 4, 113–118.
`30. McCaughan, G. W., Clark, M. J., Barclay, A. N. (1987) Characterization of
`the human homolog of the rat MRC OX-2 membrane glycoprotein. Immu-
`nogenetics 25, 329 –335.
`31. Hoek, R. M., Ruuls, S. R., Murphy, C. A., Wright, G. J., Goddard, R.,
`Zurawski, S. M., Blom, B., Homola, M. E., Streit, W. J., Brown, M. H.,
`Barclay, A. N., Sedgwick, J. D. (2000) Down-regulation of the macrophage
`lineage through interaction with OX2 (CD200). Science 290, 1768 –1771.
`32. Wright, G. J., Puklavec, M. J., Willis, A. C., Hoek, R. M., Sedgwick, J. D.,
`Brown, M. H., Barclay, A. N. (2000) Lymphoid/neuronal cell surface OX2
`glycoprotein recognizes a novel receptor on macrophages implicated in the
`control of their function. Immunity 13, 233–242.
`33. Dick, A. D., Broderick, C., Forrester, J. V., Wright, G. J. (2001) Distri-
`bution of OX2 antigen and OX2 receptor within retina. Invest. Ophthalmol.
`Vis. Sci. 42, 170 –176.
`34. Carter, D. A., Dick, A. D. (2004) CD200 maintains microglial potential to
`migrate in adult human retinal explant model. Curr. Eye Res. 28, 427–
`436.
`35. Carwile, M. E., Culbert, R. B., Sturdivant, R. L., Kraft, T. W. (1998) Rod
`outer segment maintenance is enhanced in the presence of bFGF, CNTF
`and GDNF. Exp. Eye Res. 66, 791– 805.
`36. Harada, C., Harada, T., Quah, H.

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket