Hadi Sasani1, Oytun Erbaş2

1Tekirdağ Namık Kemal University, Faculty of Medicine, Department of Radiology, Tekirdağ, Turkey
2ERBAS Institute of Experimental Medicine, Illinois, USA & Gebze, Turkey

Keywords: Central nervous system, magnetic resonance imaging, microglia, neuroinflammatory disorders, positron-emission tomography

Abstract

Microglia are important in the development, homeostasis, and disorders of the central nervous system. Neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis, as well as autism, severe depression, and schizophrenia, have all been related to microglia-derived neuroinflammation. Imaging techniques such as magnetic resonance imaging and positron emission tomography can detect neuroinflammation. The purpose of this review was to examine the role of microglia in neurodegenerative disorders, with a focus on imaging.

Neuroglia are cells that support neurons in the central nervous system (CNS), although they are not stimulated. These cells are usually smaller than neurons and are five to 10 times more numerous. They make up about half of the total volume of the brain and spinal cord. There are four types of neuroglial cells defined as astrocytes, oligodendrocytes, microglia, and ependymal cells.[1]

In 1856, Rudolf Virchow defined glial cells as a collection of cells that differed from other neurons.[2] Subsequently, in 1913, Ramón y Cajal described an apolar cell population.[3,4] In 1919, Pío del Río‐Hortega introduced the modern terminology describing glial cells.[5] With the work of Georg Kreutzberg’s group in 1968, microglia cells became the focus of neuroscience research.[6]

Microglia play important roles in brain development and the maintenance of homeostasis as well as neuroinflammation and neurodegeneration.[7] Many genetic and molecular studies conducted in the following years revealed the importance of microglia.[8-10] Thanks to the modern real-time imaging methods such as translocator protein-18 kDa (TSPO) positron emission tomography (PET), diffusion magnetic resonance imaging (MRI)[11,12], microglia have been found to be more active, frequently monitoring brain activity by extending and retracting their processes. In addition, microglia interact with all CNS components and have a significant effect on normal brain functioning and tissue integrity.[13-16] On the other hand, microglial cells play a role in a wide spectrum of diseases and disorders, including infectious diseases (acquired immunodeficiency syndrome (AIDS), human immunodeficiency virus (HIV)), inflammatory-neurodegenerative disorders (Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, etc.), spinal cord lesions, traumatic brain injury, and psychiatric disorders (schizophrenia).[1,17-21] Here, we aimed to review the effects of microglia on normal brain development and disorders, as well as the diagnosis of microglial cells as imaging, accompanied by the latest technological developments.

MICROGLIA

Embryology

Microglial cells differ from other embryological neuroglial cells and are derived from cells of mesenchymal origin (embryonic yolk sac precursors give rise to macrophages outside the nervous system) that invade the CNS late in fetal development.[1,22-24]

It is the smallest of the neuroglial cells and is distributed throughout the CNS. Morphologically, they have small cell bodies that yield numerous spine-like projections and are very similar to connective tissue macrophages. They migrate to the nervous system during fetal life.[1]

Microglia originate from a pool of primitive macrophages from the yolk sac at embryonic day 8.5 in the mouse. At embryonic day 13, microglial precursors can be seen at the base of the 4th ventricle. These cells completely differ from other hematopoietic stem cells and form an independent lineage. In humans, microglial-like cells can be detected at 13 weeks of gestation, while branched microglia can be seen at 21 weeks. Interleukin-34 and DNAX-activating protein of 12 kDa (DAP12) (known as TYRO protein tyrosine kinase-binding protein) and interferon regulatory factor (IRF)-8 deficiency also cause a decrease in microglial density. The colony-stimulating factor-1 (CSF-1) signaling is important for microglia development, and the number of tissue macrophages (including microglia), is strongly reduced in mice lacking the receptor.[25-27]

Histology

Microglial cells also have elongated nuclei forming thin, highly branched projections and relatively little cytoplasm. Immunohistochemical staining is the best method of visualizing microglia.[22]

Function and Role in Immunity

Since microglial cells in the normal brain and spinal cord are not active, they are sometimes called resting microglial cells. Microglia mediate a range of brain activities in healthy settings, including synaptic pruning and remodeling. Bidirectional communication between neurons and microglia is very important for neuronal circuits and brain connections.[28,29] However, an inflammatory disorder of the CNS, they become immune effector cells, migrate to the lesion site, proliferate and become antigen-presenting cells, and encounter invading organisms together with T lymphocytes, operate as a double-edged sword, either relieving or exacerbating the injury. It also performs an active phagocytic function, and its cytoplasm is filled with lipids and cell debris.[1]

Also, in response to tissue damage from different causes, microglia transform into large cells. They are amoeboid phagocytic cells and therefore CNS representatives of the macrophage-monocyte defense system.[22] When a pathogen or brain injury is encountered, upon activation, the morphology becomes more amoebic, and during the change of morphology, cell surface receptor expression, secretion of chemokines, and cytokines also change.[7] Monocytes from adjacent blood vessels combine microglial cells.[1]

Activation

Microglial activation is divided into pro-inflammatory activation (classic, M1) or anti-inflammatory activation (alternative, M2). M1 microglia cells produce pro-inflammatory cytokines such as interleukin (IL)-1 and tumor necrosis factor (TNF) and express nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, which produces superoxide and reactive oxygen species (ROS). M2 microglia cells support the release of anti-inflammatory factors, neurotrophic factors, and other growth factors as well as the healing process.[7] Microglia activation is frequently linked to neurodegeneration, a degenerative process that underpins the pathophysiology of neurodegenerative disorders.

Associating Conditions

The number of microglial cells increases in the presence of disorders such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and AIDS, after trauma or ischemic injury. Most of these new cells are monocytes that have migrated from the blood.[1,30]

Alzheimer’s disease is a progressive neurodegenerative disorder afflicting mainly the elderly and is the most common cause of dementia. There are accumulation of extracellular amyloid-beta (Aβ) peptides, derived from the cleavage of amyloid precursor protein (APP), and intracellular deposits of hyperphosphorylated tau. Astrocytes and microglia express apolipoprotein E (APOE) under the control of nuclear hormone receptors, and it plays an important role in Aβ phagocytosis by microglia.[31,32] In the brain, microglia mount an immediate immunological response to damaging stimuli such as misfolded proteins like Aβ. Microglia’s physiological and beneficial functions are diverted if the response does not resolve. In the brains of AD patients and AD mice models, activated microglia, immunoglobulins, and complement components are all linked to Aβ deposition. Microglial stimulation causes morphological changes such as process shortening and soma enlargement, changes in surface phenotype and secretory profile, and enhanced proliferative responses.[33,34]

Parkinson’s disease is the second most common neurodegenerative disorder, characterized by resting tremors, bradykinesia, postural instability, cognitive impairment, and autonomic dysfunction. There is an accumulation of intracellular inclusions (Lewy bodies or Lewy neurites) and the loss of dopaminergic neurons in the substantia nigra pars compacta. Genome-wide association analysis identified the human leukocyte antigen gene, which is expressed particularly in microglia, as a genetic risk factor for late-onset PD. It is also linked to the R47H variation of the microglial triggering receptor expressed on myeloid cells-2 (TREM-2).[35,36]

Amyotrophic lateral sclerosis is characterized by degeneration of both upper and lower motor neurons in bulbar and spinal regions leading to fasciculation, weakness, atrophy associated with hyperreflexia, and spasticity. It is the most common motor neuron disorder in adults and has a fatal progression. Microglial activation with increased expression of TSPO in the brain has been found in ALS. [30]

Multiple sclerosis is characterized by multiple focal lesions, continuing demyelination, and a lack of remyelination. Microglia, peripheral macrophages, T lymphocytes, and plasma cells infiltrate active MS lesions, which are frequently present in early relapsing-remitting MS. Depending on the existence of intracytoplasmic myelin breakdown products, these lesions might be demyelinating or post-demyelinating. Microglia/macrophages containing both minor and major myelin proteins (myelin/oligodendrocyte glycoprotein, 2'3'cyclic nucleotide 3'-phosphohydrolase protein, and myelin-associated glycoprotein) are seen in early demyelinating lesions (myelin basic protein, and proteolipid protein). Only significant myelin proteins are seen in late demyelinating lesions.[37] According to the marker transmembrane protein 119 (TMEM119), which is expressed exclusively on microglia and not on macrophages, the initial pool of phagocytic cells in an early MS lesion is made up of around 40% microglia. As the lesion advances, peripheral macrophages are more recruited. The presence of purinergic receptor P2Y12 (P2RY12), an ADP-responsive G-protein coupled receptor unique for the ramified processes of microglia observed in the resting state, indicates that almost none of the microglia in an active lesion are homeostatic. There are nodules of activated microglia even in the normal-appearing white matter of MS patients, but whether these microglia are homeostatic or activated is controversial because a study indicated a decrease of P2RY12 while another showed intact P2RY12 gene expression. [38-41]

PSYCHIATRIC DISORDERS

Autism spectrum disorder (ASD) is a developmental disability defined by persistent deficits in social interaction and the existence of confined, repetitive patterns of behaviors, interests, or activities. It is believed to affect 18.5 per 1,000 children under the age of eight, and males are 4.3 times more likely than females to be affected.[42] As shown in a recent study, an exaggerated translation in microglia causes autism-like behaviors in male mice. Although microglial eukaryotic translation initiation factor 4E (eIF4E) overexpression enhances translation in both sexes, it only increases microglial density and size in males, which is accompanied by a microglial shift from homeostatic to functional, with increased phagocytic capacity but decreased motility and synaptic engulfment. The disruption of male microglia function is a major contributor to sex-biased ASD.[43]

Major depressive disorder is a common mental disorder characterized by abnormalities of various brain cell types. Microglia, the major resident immune cells, have been shown to have a crucial role in the genesis and course of depression. Microglia-derived neuroinflammation-related characteristics have been highly linked to depression in patients. Pro-inflammatory cytokines are persistently increased.[44-47]

Immune system changes, as well as neuroinflammation, contribute to progressive brain changes. Inflammatory cytokines released by activated microglia induce indoleamine 2,3-dioxygenase (IDO) activity and deplete CNS tryptophan, resulting in reduced serotonin levels and changes in glutamate, dopamine, and downstream ROS. In double-blind, randomized trials of patients with recent-onset schizophrenia, minocycline added to an antipsychotic demonstrated a significant reduction in symptoms and additive cognitive benefit when compared to antipsychotic monotherapy.[48,49] Recent research has found that microglia play a substantial role in excessive synaptic destruction in schizophrenia pathogenesis. In postmortem brain tissue from schizophrenia patients, synapse density was shown to be reduced. In patient-derived neuronal cells and isolated synaptosomes, synapse removal was higher. Physiologically and pathologically, several molecules such as CX3C chemokine ligand 1/CX3C chemokine receptor 1 (CX3CL1/CX3CR1), cluster of differentiation 47/signal-regulatory protein (CD47/SIRP), and lectins are implicated. Also linked to enhanced neuronal complement deposition and synapse uptake are schizophrenia risk-associated polymorphisms in the human complement component four locus. In this regard, the antibiotic minocycline inhibits microglia-mediated synapse uptake in vitro and is linked to a slight reduction in incident schizophrenia risk when compared to other antibiotics, suggesting that excessive pruning could be a target for delaying or preventing the onset of schizophrenia in high-risk individuals.[50,51]

MOLECULAR IMAGING

Microglia cells play a role in neuroinflammation caused by many underlying disorders such as stroke, trauma, AD, schizophrenia, major depression, ALS, and some methods such as MRI and PET, which are used to image these cells, have been described in the literature.[11,12,17-19,21] While diffusion-weighted imaging (DWI) as an MRI technique is used to measure cellular changes associated with neuroinflammation and microglial activation, multi-compartment DWI methods such as neurite orientation dispersion and density imaging (NODDI) can detect water diffusion from different tissue compartments, including the extra neurite compartment.[52] In the NODDI method, diffusivity in the extra-neurite compartment is measured by orientation dispersion index (ODI). The ODI was developed to quantify how changes in neurite distribution affect water diffusion in the extraneurite space without accounting for the potential contribution of cells such as microglia to quantitative measures of ODI. Since microglia constitute 5-15% of all glial cells and glial cells in the extra-neurite compartment a large percentage of non-neuronal cells in mice (35%) and human brain (50%), they undergo significant changes in both morphology and density in response to inflammation. Therefore, Microglial activation and microglial-mediated neuroinflammation can be evaluated in the DWI sequence, as the result of these changes significantly alters the degree of diffusion restriction in the extra-neurite compartment.[23, 53-56]

The neurite orientation dispersion and density imaging is sensitive to capturing changes in microglial density, increased fullness of the extraneurite space is associated with more restricted diffusion, and there is a significant statistical correlation between quantitative measures of ODI. Therefore, MRI is important in detecting cellular changes associated with microglial activation during neuroinflammation.[12] Therefore, monitoring microglial activation through changes in microglial density during the stages of neuroinflammation is of great importance in terms of clinical diagnosis of NODDI, treatment of neuroinflammation, patient risk classification, and clinical care and research of neuropsychiatric disorders.[57,58] For example, in a recent study, the changes in ODI or neurite density index (NDI) were investigated to predict the later emergence of interferon-alpha (IFN-α)-induced fatigue in 18 patients receiving IFN-α based treatment for hepatitis-C using NODDI. An acute increase in NDI was observed in patients administered IFN-α and predicted the development of long-term fatigue. This indicates that NODDI may be useful as a potential in vivo biomarker for detecting central effects of peripheral inflammation.[56]

Although TSPO has been a traditional method for evaluating microglial activation (for example for monitoring AD progression and susceptibility to anti-inflammatory treatments), it has some limitations such as high non-specific binding, low brain uptake, genotypic variation, complex tracer kinetics, plasma variability, and TSPO polymorphism which causes large differences in binding affinity between patients.[59-62]

Although upregulation of TSPO has been associated with the M1 activation status of microglia (less commonly M2 activation), a decrease in TSPO protein expression has been observed in human adult microglia and monocyte-derived macrophages in pro-inflammatory conditions.[63,64]

Therefore, in order to detect microglial activation in PET imaging, it is necessary to monitor different microglial phenotypes and to know the changes in the expression of some other receptors and enzymes during microglial activation. For example, cannabinoid type 2 (CB2) receptor, cyclooxygenase-2 (COX-2), purinergic receptor P2X7, and ROS are the recently developed PET tracers for imaging in neuroinflammation.[65,66]

PET TRACERS TARGETING

Cannabinoid Type 2 (CB2) Receptor

Cannabinoid receptors are a kind of G-protein-coupled receptors. While the CB1 receptor is expressed in the CNS, CB2 is predominantly expressed in peripheral organs as well as microglia and neurons, increasing significantly in neuroinflammatory conditions. [11C] A-836339, [11C] NE40, [18F]29 (18F-labeled analog of A-836339), [11C] RS-016, [11C]RS-056, [18F]RS-126 and [11C]RS-028 are examples for radiotracers targeting the CB2 receptor. Among these [11C]RS-016, [18F]RS-126, and [11C] RS-028 have more than 10,000-fold selectivity for CB1.[56-65,67-72] Thus, while CB2 may only be useful in the earliest stages of neurodegenerative disorders, high selectivity for CB2 rather than CB1 of radiotracers is required to eliminate a non-specific PET signal due to the abundance of CB1 in the CNS.[65]

Cyclooxygenase-2 (COX)

Cyclooxygenases play a role in the arachidonic acid cascade and activation of inflammatory pathways. Although COX-1 and COX-2 are expressed in the brain, COX-2 is rapidly overexpressed in the case of neuroinflammation.[73] COX-2 targeting tracers have some limitations including high blood pool retention, limited uptake in target organs, high amounts of non-specific binding, relatively low affinities (>50 nM), and rapid metabolism or, substantial defluorination in the case of 18F-labeled compounds.[74,75] [11C] MC1 and [18F]1 (18F-labeled analog of celecoxib) are examples for COX-2 selective radiotracers.[76,77] [11C] MC1 was used in LPS-induced neuroinflammation and upregulation of COX-2 was observed [78,79] [18F]1 may be a valuable COX-2 radiotracer in vivo due to its good in vitro affinity, high metabolic stability, and high brain uptake.

P2X7 Receptor (P2X7R)

It is expressed in multiple cell types of the myeloid cell line and mainly in microglia in the CNS. Adenosine triphosphate (ATP) is the natural agonist of this receptor, and although its affinity for the receptor is low, it is activated only at high ATP concentrations (mM). Activation of P2X7R plays a key role in triggering neuroinflammation and leads to the pro-inflammatory release of cytokines like interleukin 1 beta (IL-1β). P2X7R is associated with the pro-inflammatory phenotype of microglia and its functional upregulated expression in CNS disorders.[80-82] [3H]A-740003, [18F] EFB, [11C]JNJ-54173717 have good affinity toward P2X7R.[83-85] P2X7R may be a promising alternative for TSPO-PET in the imaging of microglial activation.[82,84]

Reactive Oxygen Species

Nitric oxide (NO) and superoxide are formed as a result of increased inducible nitric oxide synthase (iNOS) expression and high levels of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity and activation of pro-inflammatory microglia and astrocytes in the oxidative stress state. Under normal conditions, superoxide is removed from cells by the action of superoxide dismutase (SOD). However, under high levels of NADPH oxidase activity, superoxide can react with NO to form peroxynitrite (ONOO-). Peroxynitrite can damage macromolecules in the cytoplasm and nucleus, including lipid peroxidation, DNA strand breaks, and oxidation of sulfur groups in proteins.[86,87] The development of PET radiotracers that can monitor superoxide levels in the CNS has led to the monitoring of pro-inflammatory neuroinflammation in neurodegenerative disorders and also the development of inhibitors of this enzyme for therapeutic purposes in neurodegenerative disorders also due to the key role of NADPH oxidase in oxidative stress.[75] Although [18F] FDMT (18F-labeled analog of fluorescent probe dihydroethidium) has been shown to be sensitive in measuring superoxide levels in cells and tissues using microscopy and optical imaging methods, it is not sufficient for imaging superoxide levels during neuroinflammation due to not crossing the blood-brain barrier (BBB).[88,89] [3H] Dihydroethidium is oxidized by both superoxide and hydroxyl radicals, having high brain uptake in microPET imaging studies.[65,90] [18F] ROStrace is a suitable radiotracer for PET imaging of superoxide levels in the CNS because it rapidly crosses the BBB and is retained in the brain of animals treated with lipopolysaccharide. However, [18F] ox-ROStrace (the oxidized form of [18F] ROStrace) does not cross the BBB.[88]

11C-labeled dihydroquinoline derivative ([11C] DHQ1) is an analog of NADH/NADPH that can cross the BBB and become trapped in the brain, which can be used in imaging oxidative stress.

DISEASE-MODIFYING THERAPIES

Interferon-beta (IFN-β), glatiramer acetate, fingolimod, teriflunomide, alemtuzumab, and minocycline are agents that can be used in reducing the microglial activation. IFN-β and glatiramer acetate have an indirect effect on microglia by producing a T-helper 2 (Th2) shift in the lymphocyte profile, which reduces their pro-inflammatory phenotype. In microglia, glatiramer acetate produced an alternatively activated phenotype. Teriflunomide has been shown in vitro to inhibit microglial proliferation without affecting the microglial phenotype. Fingolimod can enter the CNS and binds to sphingosine 1‐phosphate (S1P) receptors on microglia, causing TNF-α, IL-1β, and interleukin-6 (IL-6) to be downregulated. Alemtuzumab appears to have an indirect effect on microglia by causing reconstituting lymphocytes to produce more brain-derived neurotrophic factors, platelet-derived growth factors, and ciliary neurotrophic factors. The antibiotic minocycline reduces the severity of sickness in an experimental autoimmune encephalomyelitis model by inhibiting microglial activation.[91-97]

In conclusion, since microglia play an important role in the normal physiology of the brain (brain development, maintenance of homeostasis) as well as in neuroinflammation and neurodegeneration, their detection is very important, especially in disease states. Multi-compartment DWI methods such as NODDI and PET imaging can be used for imaging microglial activation. Although TSPO-PET has conventionally been used in imaging of neuroinflammation, due to being highly dynamic microglia activation in neuroinflammation and protein expression is dependent on both microglial phenotypes, more specific PET radiotracers targeting (such as CB2, COX-2, P2X7R, and ROS) have been developed. Although these are involved in the pro-inflammatory phenotype (M1) of microglial activation, there is also a need for radiotracers such as the P2Y12 receptor to be involved in the anti-inflammatory phenotype (M2), which is G-protein-coupled and overexpressed in anti-inflammatory phenotype.

Cite this article as: Sasani H, Erbaş O. Microglial Effects on Psychiatric Disorders. JEB Med Sci 2022;3(1):26-34.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

References

  1. Splittgerber R. Snell’s Clinical neuroanatomy. 8th ed. Philadelphia: Wolters Kluwer; 2019. p.54-59.
  2. Virchow R. Gesammelte Abhandlungen zur wissenschaftlichen Medicin. Frankfurt: Verlag von Meidinger Sohn & Comp;1856.
  3. Cajal RY. Algo sobre la significaci ´on fisiol ´ogica de la neurogl´ıa. Rev. Trimest. Microgr 1897;2:33-47.
  4. Pérez-Cerdá F, Sánchez-Gómez MV, Matute C. Pío del Río Hortega and the discovery of the oligodendrocytes. Front Neuroanat. 2015;9:92.
  5. Sierra A, de Castro F, Del Río-Hortega J, Rafael Iglesias-Rozas J, Garrosa M, Kettenmann H. The "Big-Bang" for modern glial biology: Translation and comments on Pío del Río-Hortega 1919 series of papers on microglia. Glia 2016;64:1801-40.
  6. Blinzinger K, Kreutzberg G. Displacement of synaptic terminals from regenerating motoneurons by microglial cells. Z Zellforsch Mikrosk Anat 1968;85:145-57.
  7. Colonna M, Butovsky O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu Rev Immunol 2017;35:441-68.
  8. Crotti A, Ransohoff RM. Microglial Physiology and Pathophysiology: Insights from Genome-wide Transcriptional Profiling. Immunity 2016;44:505-15.
  9. Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, et al. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat Neurosci 2014;17:131-43.
  10. Gautier EL, Shay T, Miller J, Greter M, Jakubzick C, Ivanov S, et al; Immunological Genome Consortium. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat Immunol 2012;13:1118-28.
  11. Casellas P, Galiegue S, Basile AS. Peripheral benzodiazepine receptors and mitochondrial function. Neurochem Int 2002;40:475-86.
  12. Yi SY, Barnett BR, Torres-Velázquez M, Zhang Y, Hurley SA, Rowley PA, et al. Detecting Microglial Density With Quantitative Multi-Compartment Diffusion MRI. Front Neurosci 2019;13:81.
  13. Tremblay MÈ, Lowery RL, Majewska AK. Microglial interactions with synapses are modulated by visual experience. PLoS Biol 2010;8:e1000527.
  14. Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J Neurosci 2009;29:3974-80.
  15. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005;308:1314-8.
  16. Frost JL, Schafer DP. Microglia: Architects of the Developing Nervous System. Trends Cell Biol 2016;26:587-97.
  17. Chen Z, Zhong D, Li G. The role of microglia in viral encephalitis: a review. J Neuroinflammation 2019;16:76.
  18. Lehnardt S. Innate immunity and neuroinflammation in the CNS: the role of microglia in Toll-like receptor-mediated neuronal injury. Glia 2010;58:253-63.
  19. Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer's disease. J Cell Biol 2018;217:459-72.
  20. Ho MS. Microglia in Parkinson's Disease. Adv Exp Med Biol 2019;1175:335-53.
  21. Inta D, Lang UE, Borgwardt S, Meyer-Lindenberg A, Gass P. Microglia Activation and Schizophrenia: Lessons From the Effects of Minocycline on Postnatal Neurogenesis, Neuronal Survival and Synaptic Pruning. Schizophr Bull 2017;43:493-6.
  22. Young B, Woodford P, O'Dowd G. Wheater’s Functional Histology A Text and Colour Atlas. 6th ed. Philadelphia: Elsevier Ltd; 2014. p. 384-99.
  23. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010;330:841-5.
  24. Ginhoux F, Guilliams M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity 2016;44:439-49.
  25. Hoeffel G, Ginhoux F. Ontogeny of Tissue-Resident Macrophages. Front Immunol 2015;6:486.
  26. Ginhoux F, Prinz M. Origin of microglia: current concepts and past controversies. Cold Spring Harb Perspect Biol 2015;7:a020537.
  27. Nayak D, Roth TL, McGavern DB. Microglia development and function. Annu Rev Immunol 2014;32:367-402.
  28. Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011;333:1456-8.
  29. Zhan Y, Paolicelli RC, Sforazzini F, Weinhard L, Bolasco G, Pagani F, et al. Deficient neuron-microglia signaling results in impaired functional brain connectivity and social behavior. Nat Neurosci 2014;17:400-6.
  30. Corcia P, Tauber C, Vercoullie J, Arlicot N, Prunier C, Praline J, et al. Molecular imaging of microglial activation in amyotrophic lateral sclerosis. PLoS One 2012;7:e52941.
  31. Terwel D, Steffensen KR, Verghese PB, Kummer MP, Gustafsson JÅ, Holtzman DM, et al. Critical role of astroglial apolipoprotein E and liver X receptor-α expression for microglial Aβ phagocytosis. J Neurosci 2011;31:7049-59.
  32. Evren V, Apaydin M, Khalilnezhad A, Erbas O, Taskiran D. Protective effect of edaravone against manganese-induced toxicity in cultured rat astrocytes. Environ Toxicol Pharmacol 2015;40:563-7.
  33. Kamphuis W, Orre M, Kooijman L, Dahmen M, Hol EM. Differential cell proliferation in the cortex of the APPswePS1dE9 Alzheimer’s disease mouse model. Glia 2012;60:615-29.
  34. Streit WJ, Xue QS, Tischer J, Bechmann I. Microglial pathology. Acta Neuropathol Commun 2014;2:142.
  35. Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson's disease. Nat Genet 2010;42:781-5.
  36. Rayaprolu S, Mullen B, Baker M, Lynch T, Finger E, Seeley WW, et al. TREM2 in neurodegeneration: evidence for association of the p.R47H variant with frontotemporal dementia and Parkinson's disease. Mol Neurodegener 2013;8:19.
  37. Kuhlmann T, Ludwin S, Prat A, Antel J, Brück W, Lassmann H. An updated histological classification system for multiple sclerosis lesions. Acta Neuropathol 2017;133:13-24.
  38. Satoh J, Kino Y, Asahina N, Takitani M, Miyoshi J, Ishida T, et al. TMEM119 marks a subset of microglia in the human brain. Neuropathology 2016;36:39-49.
  39. Zrzavy T, Hametner S, Wimmer I, Butovsky O, Weiner HL, Lassmann H. Loss of 'homeostatic' microglia and patterns of their activation in active multiple sclerosis. Brain 2017;140:1900-13.
  40. Moore CS, Ase AR, Kinsara A, Rao VT, Michell-Robinson M, Leong SY, et al. P2Y12 expression and function in alternatively activated human microglia. Neurol Neuroimmunol Neuroinflamm 2015;2:e80.
  41. van der Poel M, Ulas T, Mizee MR, Hsiao CC, Miedema SSM, Adelia, Schuurman KG, et al. Transcriptional profiling of human microglia reveals grey-white matter heterogeneity and multiple sclerosis-associated changes. Nat Commun 2019;10:1139.
  42. Baio J, Wiggins L, Christensen DL, Maenner MJ, Daniels J, Warren Z, et al. Prevalence of Autism Spectrum Disorder Among Children Aged 8 Years - Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2014. MMWR Surveill Summ 2018;67:1-23.
  43. Xu ZX, Kim GH, Tan JW, Riso AE, Sun Y, Xu EY, et al. Elevated protein synthesis in microglia causes autism-like synaptic and behavioral aberrations. Nat Commun 2020;11:1797.
  44. Jia X, Gao Z, Hu H. Microglia in depression: current perspectives. Sci China Life Sci 2021;64:911-25.
  45. Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatry 2009;65:732-41.
  46. Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry 2010;67:446-57.
  47. Solmaz V, Çınar BP, Yiğittürk G, Çavuşoğlu T, Taşkıran D, Erbaş O. Exenatide reduces TNF-α expression and improves hippocampal neuron numbers and memory in streptozotocin treated rats. Eur J Pharmacol 2015;765:482-7.
  48. Leza JC, García-Bueno B, Bioque M, Arango C, Parellada M, Do K, et al. Inflammation in schizophrenia: A question of balance. Neurosci Biobehav Rev 2015;55:612-26.
  49. Watkins DC, Assari S, Johnson-Lawrence V. Race and Ethnic Group Differences in Comorbid Major Depressive Disorder, Generalized Anxiety Disorder, and Chronic Medical Conditions. J Racial Ethn Health Disparities 2015;2:385-94.
  50. Zuo N, Nitta A. New Insights Regarding Diagnosis and Medication for Schizophrenia Based on Neuronal Synapse-Microglia Interaction. J Pers Med 2021;11:371.
  51. Sellgren CM, Gracias J, Watmuff B, Biag JD, Thanos JM, Whittredge PB, et al. Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat Neurosci 2019;22:374-85.
  52. Zhang H, Schneider T, Wheeler-Kingshott CA, Alexander DC. NODDI: practical in vivo neurite orientation dispersion and density imaging of the human brain. Neuroimage 2012;61:1000-16.
  53. Mota B, Herculano-Houzel S. All brains are made of this: a fundamental building block of brain matter with matching neuronal and glial masses. Front Neuroanat 2014;8:127.
  54. Von Bartheld CS, Bahney J, Herculano-Houzel S. The search for true numbers of neurons and glial cells in the human brain: A review of 150 years of cell counting. J Comp Neurol 2016;524:3865-95.
  55. Herculano-Houzel S. The glia/neuron ratio: how it varies uniformly across brain structures and species and what that means for brain physiology and evolution. Glia 2014;62:1377-91.
  56. Yang TT, Lin C, Hsu CT, Wang TF, Ke FY, Kuo YM. Differential distribution and activation of microglia in the brain of male C57BL/6J mice. Brain Struct Funct 2013;218:1051-60.
  57. Dheen ST, Kaur C, Ling EA. Microglial activation and its implications in the brain diseases. Curr Med Chem 2007;14:1189-97.
  58. Dowell NG, Bouyagoub S, Tibble J, Voon V, Cercignani M, Harrison NA. Interferon-alpha-Induced Changes in NODDI Predispose to the Development of Fatigue. Neuroscience 2019;403:111-7.
  59. Owen DR, Gunn RN, Rabiner EA, Bennacef I, Fujita M, Kreisl WC, et al. Mixed-affinity binding in humans with 18-kDa translocator protein ligands. J Nucl Med 2011;52:24-32.
  60. Turkheimer FE, Rizzo G, Bloomfield PS, Howes O, Zanotti-Fregonara P, Bertoldo A, et al. The methodology of TSPO imaging with positron emission tomography. Biochem Soc Trans 2015;43:586-92.
  61. Guo Q, Colasanti A, Owen DR, Onega M, Kamalakaran A, Bennacef I, et al. Quantification of the specific translocator protein signal of 18F-PBR111 in healthy humans: a genetic polymorphism effect on in vivo binding. J Nucl Med 2013;54:1915-23.
  62. Kreisl WC, Jenko KJ, Hines CS, Lyoo CH, Corona W, Morse CL, et al. Biomarkers Consortium PET Radioligand Project Team. A genetic polymorphism for translocator protein 18 kDa affects both in vitro and in vivo radioligand binding in human brain to this putative biomarker of neuroinflammation. J Cereb Blood Flow Metab 2013;33:53-8.
  63. Bonsack F 4th, Alleyne CH Jr, Sukumari-Ramesh S. Augmented expression of TSPO after intracerebral hemorrhage: a role in inflammation? J Neuroinflammation 2016;13:151.
  64. Owen DR, Narayan N, Wells L, Healy L, Smyth E, Rabiner EA, et al. Pro-inflammatory activation of primary microglia and macrophages increases 18kDa translocator protein expression in rodents but not humans. J Cereb Blood Flow Metab 2017;37:2679-90.
  65. Janssen B, Vugts DJ, Windhorst AD, Mach RH. PET Imaging of Microglial Activation-Beyond Targeting TSPO. Molecules 2018;23:607.
  66. Soydan S, Arda B, Altuntaş I, Erbaş O. Beneficial Effects of Cannabis Sativa Extract on Oxidative Stress. JEB Med Sci 2021;2:336-42.
  67. Horti AG, Gao Y, Ravert HT, Finley P, Valentine H, Wong DF, et al. Synthesis and biodistribution of [11C]A-836339, a new potential radioligand for PET imaging of cannabinoid type 2 receptors (CB2). Bioorg Med Chem 2010;18:5202-7.
  68. Slavik R, Grether U, Müller Herde A, Gobbi L, Fingerle J, Ullmer C, et al. Discovery of a high affinity and selective pyridine analog as a potential positron emission tomography imaging agent for cannabinoid type 2 receptor. J Med Chem 2015;58:4266-77.
  69. Evens N, Vandeputte C, Coolen C, Janssen P, Sciot R, Baekelandt V, et al Preclinical evaluation of [11C]NE40, a type 2 cannabinoid receptor PET tracer. Nucl Med Biol 2012;39:389-99.
  70. Moldovan RP, Teodoro R, Gao Y, Deuther-Conrad W, Kranz M, Wang Y, et al. Development of a High-Affinity PET Radioligand for Imaging Cannabinoid Subtype 2 Receptor. J Med Chem 2016;59:7840-55.
  71. Haider A, Spinelli F, Herde AM, Mu B, Keller C, Margelisch M, et al. Evaluation of 4-oxo-quinoline-based CB2 PET radioligands in R6/2 chorea huntington mouse model and human ALS spinal cord tissue. Eur J Med Chem 2018;145:746-59.
  72. Slavik R, Müller Herde A, Haider A, Krämer SD, Weber M, Schibli R, et al. Discovery of a fluorinated 4-oxo-quinoline derivative as a potential positron emission tomography radiotracer for imaging cannabinoid receptor type 2. J Neurochem 2016;138:874-86.
  73. Aïd S, Bosetti F. Targeting cyclooxygenases-1 and -2 in neuroinflammation: Therapeutic implications. Biochimie 2011;93:46-51.
  74. Tietz O, Marshall A, Wuest M, Wang M, Wuest F. Radiotracers for molecular imaging of cyclooxygenase-2 (COX-2) enzyme. Curr Med Chem 2013;20:4350-69.
  75. Pacelli A, Greenman J, Cawthorne C, Smith G. Imaging COX-2 expression in cancer using PET/SPECT radioligands: current status and future directions. J Labelled Comp Radiopharm 2014;57:317-22.
  76. Cortes M, Singh P, Morse C, Kowalski A, Jenko K, Shrestha S, et al. Synthesis of a candidate brain-penetrant COX-2 PET radioligand as a potential probe for neuroinflammation. J. Label. Compd. Radiopharm 2015;58:S312.
  77. Penning TD, Talley JJ, Bertenshaw SR, Carter JS, Collins PW, Docter S, et al. Synthesis and biological evaluation of the 1,5-diarylpyrazole class of cyclooxygenase-2 inhibitors: identification of 4-[5-(4-methylphenyl)-3-(trifluoromethyl)- 1H-pyrazol-1-yl] benzenesulfonamide (SC-58635, celecoxib). J Med Chem 1997;40:1347-65.
  78. Kim MJ, Shrestha S, Eldridge M, Cortes M, Singh P, Liow JS, et al. Novel pet radioligands show that, in rhesus monkeys, cox-1 is constitutively expressed and cox-2 is induced by inflammation. J. Nucl. Med 2017;58:2.
  79. Inanir S, Copoglu US, Kokacya H, Dokuyucu R, Erbas O, Inanir A. Agomelatine Protection in an LPS-Induced Psychosis-Relevant Behavior Model. Med Sci Monit 2015;21:3834-9.
  80. Bhattacharya A, Biber K. The microglial ATP-gated ion channel P2X7 as a CNS drug target. Glia 2016;64:1772-87.
  81. Bartlett R, Stokes L, Sluyter R. The P2X7 receptor channel: recent developments and the use of P2X7 antagonists in models of disease. Pharmacol Rev 2014;66:638-75.
  82. Beaino W, Janssen B, Kooij G, van der Pol SMA, van Het Hof B, van Horssen J, et al. Purinergic receptors P2Y12R and P2X7R: potential targets for PET imaging of microglia phenotypes in multiple sclerosis. J Neuroinflammation 2017;14:259.
  83. Honore P, Donnelly-Roberts D, Namovic MT, Hsieh G, Zhu CZ, Mikusa JP, et al. A-740003 [n-(1-{[(cyanoimino) (5-quinolinylamino) methyl]amino}-2,2-dimethylpropyl)- 2-(3,4-dimethoxyphenyl)acetamide], a novel and selective P2X7 receptor antagonist, dose-dependently reduces neuropathic pain in the rat. J. Pharmacol. Exp. Ther 2006;319:1376-85.
  84. Fantoni ER, Dal Ben D, Falzoni S, Di Virgilio F, Lovestone S, Gee A. Design, synthesis and evaluation in an LPS rodent model of neuroinflammation of a novel 18F-labelled PET tracer targeting P2X7. EJNMMI Res 2017;7:31.
  85. Wilkinson SM, Barron ML, O‘Brien-Brown J, Janssen B, Stokes L, Werry EL, et al. Pharmacological evaluation of novel bioisosteres of an adamantanyl benzamide P2X7 receptor antagonist. ACS Chem. Neurosci 2017;8:2374-80.
  86. Makvandi M, Sellmyer MA, Mach RH. Inflammation and DNA damage: Probing pathways to cancer and neurodegeneration. Drug Discov Today Technol 2017;25:37-43.
  87. Raina R, Sen D. Can crosstalk between DOR and PARP reduce oxidative stress mediated neurodegeneration? Neurochem Int 2018;112:206-18.
  88. Hou C, Hsieh CJ, Li S, Lee H, Graham TJ, Xu K, et al. Development of a Positron Emission Tomography Radiotracer for Imaging Elevated Levels of Superoxide in Neuroinflammation. ACS Chem Neurosci 2018;9:578-86.
  89. Dugan LL, Ali SS, Shekhtman G, Roberts AJ, Lucero J, Quick KL, et al. IL-6 mediated degeneration of forebrain GABAergic interneurons and cognitive impairment in aged mice through activation of neuronal NADPH oxidase. PLoS One 2009;4:e5518.
  90. Takai N, Abe K, Tonomura M, Imamoto N, Fukumoto K, Ito M, et al. Imaging of reactive oxygen species using [(3)H] hydromethidine in mice with cisplatin-induced nephrotoxicity. EJNMMI Res 2015;5:116.
  91. Healy LM, Michell-Robinson MA, Antel JP. Regulation of human glia by multiple sclerosis disease modifying therapies. Semin Immunopathol 2015;37:639-49.
  92. Kawanokuchi J, Mizuno T, Kato H, Mitsuma N, Suzumura A. Effects of interferon-beta on microglial functions as inflammatory and antigen presenting cells in the central nervous system. Neuropharmacology 2004;46:734-42.
  93. Kim HJ, Ifergan I, Antel JP, Seguin R, Duddy M, Lapierre Y, et al. Type 2 monocyte and microglia differentiation mediated by glatiramer acetate therapy in patients with multiple sclerosis. J Immunol 2004;172:7144-53.
  94. Noda H, Takeuchi H, Mizuno T, Suzumura A. Fingolimod phosphate promotes the neuroprotective effects of microglia. J Neuroimmunol 2013;256:13-8.
  95. Jones JL, Anderson JM, Phuah CL, Fox EJ, Selmaj K, Margolin D, et al. Improvement in disability after alemtuzumab treatment of multiple sclerosis is associated with neuroprotective autoimmunity. Brain 2010;133:2232-47.
  96. Bogie JF, Stinissen P, Hendriks JJ. Macrophage subsets and microglia in multiple sclerosis. Acta Neuropathol 2014;128:191-213.
  97. Wostradowski T, Prajeeth CK, Gudi V, Kronenberg J, Witte S, Brieskorn M, et al. In vitro evaluation of physiologically relevant concentrations of teriflunomide on activation and proliferation of primary rodent microglia. J Neuroinflammation 2016;13:250.