Introduction
Inflammation plays a key role in highly prevalent diseases, including many cardiovascular, neurological, and rheumatological diseases. While an appropriate immune response can be beneficial and protective, aberrant activation of this response recruits excessive pro-inflammatory cells to cause damage (Figure 1). Neuroinflammation is inflammation within the central nervous system (CNS). Because the CNS is separated from the periphery by the blood-brain barrier that creates an immune-privileged site, it has its own unique immune cells and immune response. Moreover, neuroinflammation can compromise the blood-brain barrier (BBB) causing an influx of peripheral immune cells and factors. This chapter will provide a broad overview of neuroinflammation and describe its unique aspects. Key immune cells and factors in mediating neuroinflammation will be introduced followed by a discussion of neuroinflammation in different neurological diseases.
Figure 1:

Neuroinflammation. When the blood-brain barrier (BBB) is compromised, peripheral immune cells infiltrate into the CNS and interact with microglia and astrocytes. Depending on the stimuli, the resultant response can be either detrimental or protective to the CNS.
I. Key Players in Neuroinflammation
A. Neutrophils
Neutrophils form 50–70% of leukocytes and play a central role in neuroinflammation. As one of the major defenses, neutrophils are quickly activated in the bloodstream and are the first peripheral leukocytes to respond to stimuli. Neutrophils at the injured site will attempt to eliminate pathogens and initiate an inflammatory cascade to repair the damaged tissue. Neutrophils phagocytose pathogens to form a phagosome that undergoes lysosomal degradation to destroy the pathogen’s cell walls and proteins. After ingesting a pathogen neutrophil produces reactive oxygen species (ROS) such as superoxide (O2−), hypochlorite (HOCl), hydrogen peroxide (H2O2), hydroxyl radicals, and nitric oxide (NO)1. Neutrophils also secrete myeloperoxidase (MPO), an enzyme that produces HOCl and other reactive species. MPO is also involved in the formation of neutrophil extracellular traps (NETs) which limit pathogens from multiplying by entrapping bacteria, viruses, and fungi2. However, NETs can also cause a proinflammatory responses and lead to tissue damage via long-term production of proteolytic enzymes. Formation of NETs can eventually lead to apoptosis, known as NETosis.
The BBB serves as a significant immunological defense from foreign pathogens and toxic molecules. However, with injury and resultant neuroinflammation, the BBB can become compromised allowing peripheral immune cells to infiltrate into the CNS. The infiltration of neutrophils has been related to brain damage due to the appearance of pro-inflammatory cytokines, ROS, and NETs release. However, not all neutrophils are pro-inflammatory. Recently, other neutrophil phenotypes have been described that have roles in tissue repair, tumor killing, and immune regulation3. This has implications in therapy as strategies that indiscriminately deplete or inhibit neutrophils may not always be beneficial.
B. Macrophages
Activated macrophages regulate inflammatory responses and have diverse phenotypes and functions, allowing macrophage to change fluidly into different states depending on its environment4. An M1/M2 classification has been described from in vitro observations. Macrophages (M0) can be “classically” stimulated with pro-inflammatory mediators, i.e., LPS and IFN-y, to adopt an aggressive (M1) phenotype. Conversely, cells “alternatively” stimulated with anti-inflammatory cytokines (i.e., IL-4 & IL-13) will adopt an M2 phenotype, thought to have restorative and immunomodulating functions. When inflammation initially occurs, macrophages can polarize into M1-like phenotypes and generate pro-inflammatory cytokines and chemokines like TNF, IL-1β, and IL-125. These macrophages will amplify inflammation by generating large amounts of pro-inflammatory cytokines and ROS to remove pathogens or foreign molecules from the injured site, though prolonged M1 macrophage activity will result in tissue damage and chronic inflammation. M2-like phenotypes are thought to be anti-inflammatory and reduces inflammation to enable tissue repair by secreting anti-inflammatory cytokines, chemokines, and growth factors like IL-10, TGF-β, and CCL186. Underscoring the complexity and diversity of macrophages, the M2 phenotype has been found to have subsets known as M2a, M2b, M2c, and M2d. M2a amplifies cell growth and tissue repair, M2b plays a central role in inflammation, M2c is important for phagocytosis, and M2d stimulates tumor progression7. Beyond the M1/M2 phenotypes, other phenotypes are starting to be described, primarily based on advanced techniques such as RNAseq8.
C. Lymphocytes
Lymphocytes are classified into B-cells, T-cells, and natural killer cells (NK). The T- and B-cells participate in acquired or antigen-specific immune responses. B-cells can turn into plasmacytes to produce antibodies. While humoral immunity depends on B-Cells, cell immunity depends on T-cells9. T cells play major roles in adaptive immune response to tissue damage and infection. They are able to clone highly polymorphic antigen receptors which allows them to detect protein-based antigens10. Upon recognition of protein-associated antigens, T cells will undergo clonal amplification and progressively obtain the ability to react to stimuli. Major types of T-cells are CD4+ (helper T-cells) and CD8+ (cytotoxic T-cells)11. T-cell receptors and their related receptors, such as CD3 and CD4, form a complex between the major histocompatibility complex-2 (MHC-II) receptor and the target antigen. CD4+ cells generate cytokines to start an immune reaction and activate T-cell-associated humoral immunity. Once activated, CD8+ cells migrate to its target antigen to eventually kill it. Regulatory T-cells (Tregs) can help manage immune tolerance and suppress extreme inflammation, playing a protective role. Conversely, effector T-cells like Th1 and Th17 cells can play a part in neuroinflammation and tissue damage. NK cells play a versatile role by serving as a rapid defense mechanism against neural damage and helping immune modulation within the CNS.
D. Microglia
Microglia, originating from the yolk sac12, are the primary immune cells of the CNS, constituting 5–10% of the adult brain cell population. They are the resident macrophages in the CNS and first responders to insults13. These cells have highly motile processes allowing them to scan their environment constantly. The three primary functions of microglia are 1) surveying their environments for any changes using their sensomes, which encode various proteins to allow microglia to sense endogenous ligands and microbes14; 2) physiological housekeeping, which includes migrating to sites of injury15, synapse pruning and remodeling16, myelin homeostasis17, and interacting with astrocytes18; and 3) protecting the organism against harmful stimuli, which include pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) recognized by the various receptors expressed on microglia, including toll-like receptors, nuclear oligomerization domain-like receptors, and more (Figure 2)19. As the immune cells of the CNS, microglia protect the brain from harmful antigens, such as apoptotic cells, microbes, protein aggregates, and lipoprotein particles.
Figure 2:

Microglia. Under normal physiological circumstances, “resting” microglia are ramified, consisting of many receptors. Receptor and cytokine interactions maintain the ramified state. Upon activation, microglia can shift toward an amoeboid “M1” phenotype that upregulates a pro-inflammatory response. Activated microglial cells can also shift to the M2-phenotype that is more reparative and anti-inflammatory.
Microglia are also capable of contributing to oxidative stress and have a role in producing ROS that lead to tissue damage when produced in excess. These phagocytes can mediate respiratory burst to produce powerful microbicidal agents downstream of the initial superoxide production20. Activated microglia produce nitric oxide (NO) via inducible nitric oxide synthase (iNOS), and the release of NO leads to neuronal cell death by inhibiting neurons from undergoing cellular respiration21. Contributing to oxidative stress is MPO22. Normal brain microglia rarely express MPO23,24, but activated microglia are positive for MPO25. Elevated MPO activity can be detrimental to tissues and is implicated in many diseases26. In vivo MPO activity can be imaged by molecular imaging agents for MRI27–31 and PET32.
E. Astrocytes
Astrocytes are the brain’s most common glial cells and help regulate neuronal activity and maintain brain homeostasis. They regulate blood flow, maintain BBB integrity, supply energy metabolites to neurons, modulate synaptic activity, monitor the secretion of neurotrophins, remove dead cells from their surrounding environment, manage the extracellular balance of ions, fluids, and neurotransmitters, and form scars in response to brain injury or damage13. Furthermore, astrocytes can phagocytose protein aggregates like amyloid plaques33. In contrast to microglia, astrocytes tend to engulf distal processes and diffuse neuritic debris34.
Astrocytes respond to insults by a process called reactive astrogliosis. Activation of astrocytes changes its molecular expression and morphology such as increasing the number and size of astrocytes expressing glial fibrillary acidic protein (GFAP)35. When astrocytes are activated, they can take one of two phenotypes: the A1-phenotype or A2-phenotype, which are neurotoxic or neuroprotective, respectively13. Many intercellular signaling molecules can also trigger reactive astrogliosis, e.g., reactive oxygen species, neurotransmitters, cytokines (FGF2, IL-10, CNTF, IFNγ), and molecules associated with systemic metabolic toxicity36. Activated astrocytes become not only neurotoxic but also lose many of their vital functions pertinent to homeostasis and maintenance of neuronal health. A1-reactive astrocytes negatively affect synaptic activity by disassembling synapses and inducing the formation of fewer synapses. Additionally, A1 astrocytes almost completely lose their ability to phagocytose myelin debris from their environment. Other detrimental functions include production of neurotoxic levels of reactive oxygen species37, releasing potentially excitotoxic glutamate38, and compromising the blood-brain barrier via vascular endothelial growth factor (VEGF) production39.
Not all reactive astrocytes are detrimental. Astrocytes also contribute to normal physiological inflammatory responses to protect the CNS. Some beneficial functions of reactive astrogliosis include uptake of glutamate40, protection against oxidative stress by producing glutathione41, degradation of β-amyloid plaques42, facilitating repair of the BBB43, and restricting the spread of inflammatory cells or infectious agents into healthy CNS parenchyma44.
F. Identification, Polarization, and Imaging
1. Macrophages and Microglia
To differentiate macrophages from microglia, human and murine microglia specifically express TMEM119, by which they may be identified45. Among the putative M1-macrophages/microglia markers are iNOS, CD38, CD80, CD86, and FPPR2 as well as pro-inflammatory cytokines such as TNF, IL-6, IL-12A, IL-23A and IL-27. Putative markers of M2-macrophages/microglia are Arginase-1, CD206, CD163, RELMα, CHI3L3, ALOX15, EGR2, and c-MYC46. However, transcriptomic experiments suggest that the M1/M2 nomenclature falls short of the complexity of activation states in vivo. A study comparing homeostatic microglia to those in different diseases found ten transcriptomic patterns during development, which reappeared under disease conditions, and three other distinct clusters associated with neurodegeneration, demyelination, and remyelination, hinting at a highly diverse inflammatory milieu47.
Molecular imaging studies aim to unravel some aspects of this convoluted network utilizing established disease models to address different aspects of neuroinflammation. One example is imaging agents targeting MPO. Paramagnetic substrates that are activated and retained at sites of MPO allow for visualization of MPO activity with MRI27,31,48. Activated innate immune cells phagocytose and store Fe2+ which can be visualized by iron oxide nanoparticles49. MPO and iron oxide nanoparticle MRI have been used in combination to identify and map M1-dominant and M2-dominant lesions in the CNS (Figure 3)50. PET imaging requires significantly less agent to be detectable, which increases sensitivity. Imaging probes targeting the translocator protein (TSPO), which is expressed by activated microglia and to a lesser extent reactive macrophages and astrocytes, are widely used in the research setting51. A radioligand targeting MPO has been recently developed32. Targets for PET-agents beyond TSPO and MPO include endocannabinoid receptor CB252, cyclooxygenase 153 and 254, matrix metalloproteinases55, and P2RY1256.
Figure 3:

Differentiating different macrophage/microglia phenotypes by MRI. MPO activity (MPO-Gd) and iron oxide nanoparticle (CLIO) MR imaging in a murine model of experimental autoimmune encephalomyelitis. In (A), MPO-Gd+ lesions with matched CLIO+ lesions (area 2) represent M1-like cells, corroborated by immunohistochemistry in (B) (red = MPO, yellow = CD206, blue = DAPI). Lesions that were CLIO+ only without corresponding MPO-Gd+ signal (area 1) represent M2-like phagocytes that expressed CD206 but virtually no MPO on immunohistochemistry. Modified from50.
2. Astrocytes
Even though GFAP has been commonly used to characterize reactive astrogliosis, many caveats exist. One of the main limitations includes regional differences within the brain regarding GFAP expression. Most importantly, GFAP does not allow for the differentiation between the A1- and A2-phenotypes. Transcriptomic studies found 57 genes, such as C3 and guanine nucleotide-binding protein 2 (GBP2), that exhibited preferential expression in A1-astrocytes, as well as 150 genes, such as the S100 calcium-binding protein A10 (S100a10), that were preferentially expressed in A2-astrocytes57. Of these genes, the most used marker for A1-astrocytes is C3, while the most common markers for distinguishing A2-astrocytes include S100a10 and pentraxin-3 (PTX3)58. Imaging astrocytes includes PET agents targeting monoamine oxidase B, acetate metabolism, and imidazoline2 binding sites59, although none differentiate between the different phenotypes.
II. Neuroinflammation in Diseases
A. Multiple Sclerosis
People living with MS (PwMS) develop neurodegeneration due to neuroinflammation throughout the brain and spinal cord, leading to accelerated atrophy. Neuroinflammation in MS results from a complex interplay of adaptive and innate immune cells60. Traditionally, MS has been viewed as a T-cell-mediated disease. Autoreactive CD4+ and CD8+ T-cells migrate across the broken-down blood-brain barrier and are present in active demyelinating lesions61. Dysregulated regulatory T-cells may also contribute to the development of autoreactivity in MS62.
While T-cells play a central role in MS neuroinflammation, the critical importance of B-cells has recently been recognized. B-cells act through interaction and antigen presentation to T-cells, the release of inflammatory cytokines and chemokines, and the production of pathogenic antibodies63. PwMS have immune cell infiltrates and ectopic lymphoid follicles in the meninges64. Innate immune cells are also important in MS. Microglia and macrophages are abundant in active demyelinating lesions65. Active demyelinating lesions predominate early in the disease course but become less frequent later in the disease66. These later lesions slowly expand, called smoldering plaques, and have iron-containing microglia and macrophages at the periphery67 that may play an important role in the gradual progression of disability in PwMS68.
B. Neurodegenerative diseases
Alzheimer’s disease (AD) is the most frequent cause of dementia. A substantial unmet need exists to elucidate the mechanisms underlying AD and find robust non-invasive biomarkers to aid the diagnosis and earlier detection of AD, but also the development and assessment of new therapeutic strategies. In AD, there are multiple potential triggers of inflammation, among which protein aggregates seem to be playing a pivotal role69, and crosstalk of protein misfolding and inflammation are a constant part of the neurodegenerative processes70. Persistent activation of glial cells that compromises neuronal functionality can result from continuous exposure to an inflammatory stimulus or impairment of resolution71. Recent studies reveal a significant number of novel AD-risk single nucleotide polymorphisms are exclusively or dominantly expressed in microglia, emphasizing the critical roles these cells play in AD72. Different phenotypes of microglia have been found in AD, including activated response microglia (ARMs), interferon-responsive microglia (IRMs), human AD microglia (HAMs)73, disease-associated microglia (DAM)74, microglial neurodegenerative (MGnD)75, and dark microglia76. Research is ongoing to elucidate the roles these different types of microglia play in AD and other neurodegenerative diseases.
C. Stroke
Stroke induces a complex cascade of changes dominated by the innate immune response, with activation of local microglia, influx of leukocytes into the brain, and production of ROS that elevate oxidative stress and contribute to stroke progression77. Whereas neutrophils become scarce after 3 days, activated microglia and macrophages are elevated within hours and can last for weeks to months after stroke29,78. Neutrophils arrive in the ischemic brain within 24 hours after stroke, with macrophages peaking about 3 days after79. Pro-inflammatory molecules and cytokines such as TNF and IL-1β are elevated in the blood of patients. The levels of pro-inflammatory molecules and leukocytes correlate with outcomes after stroke80. Similarly, decreasing leukocyte recruitment and pro-inflammatory and oxidizing molecules, such as MPO, have decreased infarct size by 30–60%81, demonstrating that inflammation extends ischemic injury. Another example is Rho-associated kinase (ROCK), which is elevated in patients after stroke and is an independent predictor for recurrent stroke.27,28 ROCK inhibition reduced neutrophil accumulation in the ischemic region and resulted in reduction of infarction volume.30,31
D. Glioma
The tumor microenvironment (TME) in glioma is characterized by a complex interplay of various cell types, including tumor cells, immune cells, and endothelial cells, along with the secretion of various pro and anti-inflammatory cytokines82. Glioma-associated macrophages/microglia (GAMs) can manipulate immune response and tumor development. M1-like GAMs exhibit an anti-tumor effect, while M2-like GAMs have an immunosuppressive impact. The presence of macrophages in low-grade gliomas decrease survival and they are more abundant in higher-grade gliomas compared to microglia83. Similarly, tumor-infiltrated neutrophils (TINs) can modulate other immune cells and promote tumor infiltration. T-lymphocytes also have a distinct role in glioma progression and suppression but at lower fractions than in brain metastasis84. T-cells constitute 1–5% of total glioma cellular content. The percentages of both CD4+ and CD8+ tumor-infiltrating T-cells increase with tumor grade85.
Conclusion
Neuroinflammation is a complex interplay between different peripheral and central immune cells that can have damaging and beneficial effects depending on the context. Imaging of these cells and their functions is still at a relatively early stage and represents opportunities in imaging and clinical research to advance our knowledge and improve patient care.
Key Points.
Neuroinflammation is a key immune response in the central nervous system (CNS) observed in many neurological diseases.
The CNS is separated from the periphery by the blood-brain barrier that creates an immune-privileged site with its own unique immune cells and immune response.
BBB compromise causes an influx of peripheral immune cells and factors that interact with resident immune cells in the CNS.
Neuroinflammation can exert damaging as well as beneficial effects depending on the context.
Synopsis:
Neuroinflammation is a key immune response observed in many neurological diseases. While an appropriate immune response can be beneficial, aberrant activation of this response recruits excessive proinflammatory cells to cause damage. Because the central nervous system is separated from the periphery by the blood-brain barrier that creates an immune-privileged site, it has its own unique immune cells and immune response. Moreover, neuroinflammation can compromise the blood-brain barrier causing an influx of peripheral immune cells and factors. Recent advances have brought a deeper understanding of neuroinflammation that can be leveraged to develop more potent therapies and improve patient selection.
Abbreviation:
- CNS
central nervous system
- BBB
blood-brain barrier
- IL
interleukin
- ROS
reactive oxygen species
- MPO
myeloperoxidase
- NET
neutrophil extracellular trap
- LPS
lipopolysaccharide
- IFN
interferon
- TNF
tumor necrosis factor
- TGF
transforming growth factor
- CCL
chemokine ligand
- NK
natural killer cell
- CD
cluster of differentiation
- MHC
major histocompatibility complex
- PAMP
pathogen-associated molecular patterns
- DAMP
damage-associated molecular patterns
- GFAP
glial fibrillary acidic protein
- FGF2
fibroblast growth factor 2
- CNTF
ciliary neutrotrophic factor
- FPPR2
formyl peptide receptor 2
- RELMα
Retnla resistin like alpha
- CHI3L3
Chitinase-3-like-3
- ALOX15
arachidonate 15-lipoxygenase
- EGR2
early growth response 2
- TSPO
translocator protein
- P2RY12
purinergic receptor P2Y12
- ROCK
Rho-associated kinase
- GAM
glioma-associated microglia/macrophage
- TIN
tumor infiltrated-neutrophil
- iNOS
inducible nitric oxide synthase
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosures: John W. Chen has a financial interest in Silverier LLC, a company that founds biotech start‐up companies, and its subsidiary, Einsenca, Inc., a company developing PET and MR imaging agents that target myeloperoxidase. Dr. Chen’s interests were reviewed and are managed by Massachusetts General Hospital and Mass General Brigham in accordance with their conflict-of-interest policies. The other authors declare no conflict of interest.
References
- 1.Healy LM, Perron G, Won S-Y, et al. MerTK is a functional regulator of myelin phagocytosis by human myeloid cells. The Journal of Immunology. 2016;196(8):3375–3384. [DOI] [PubMed] [Google Scholar]
- 2.Hidalgo A, Libby P, Soehnlein O, Aramburu IV, Papayannopoulos V, Silvestre-Roig C. Neutrophil extracellular traps: from physiology to pathology. Cardiovasc Res. Oct 21 2022;118(13):2737–2753. doi: 10.1093/cvr/cvab329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hellebrekers P, Vrisekoop N, Koenderman L. Neutrophil phenotypes in health and disease. Eur J Clin Invest. Nov 2018;48 Suppl 2(Suppl Suppl 2):e12943. doi: 10.1111/eci.12943 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Butterfield TA, Best TM, Merrick MA. The dual roles of neutrophils and macrophages in inflammation: a critical balance between tissue damage and repair. J Athl Train. 2006 2006;41(4):457–65. [PMC free article] [PubMed] [Google Scholar]
- 5.Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. European Journal of Pharmacology. 2020 2020;877:173090. [DOI] [PubMed] [Google Scholar]
- 6.Novak ML, Koh TJ. Macrophage phenotypes during tissue repair. J Leukoc Biol. 2013/06//undefined 2013;93(6):875–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Yao Y, Xu XH, Jin L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front Immunol. 2019 2019;10:792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sreejit G, Fleetwood AJ, Murphy AJ, Nagareddy PR. Origins and diversity of macrophages in health and disease. Clin Transl Immunology. 2020;9(12):e1222. doi: 10.1002/cti2.1222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cano R, Lopera H. Introduction to T and B lymphocytes. In: Anaya J, Shoenfeld Y, A R-V, eds. Autoimmunity: From Bench to Bedside. El Rosario University Press; 2013:chap 5. [PubMed] [Google Scholar]
- 10.Fabbri M, Smart C, Pardi R. T lymphocytes. The International Journal of Biochemistry & Cell Biology. 2003 2003;35(7):1004–1008. [DOI] [PubMed] [Google Scholar]
- 11.Bauer ME, Teixeira AL. Neuroinflammation in Mood Disorders: Role of Regulatory Immune Cells. Neuroimmunomodulation. 2021 2021;28(3):99–107. [DOI] [PubMed] [Google Scholar]
- 12.Saijo K, Glass CK. Microglial cell origin and phenotypes in health and disease. Nat Rev Immunol. Oct 25 2011;11(11):775–87. doi: 10.1038/nri3086 [DOI] [PubMed] [Google Scholar]
- 13.Kwon HS, Koh S-H. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Translational neurodegeneration. 2020;9:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hickman SE, Kingery ND, Ohsumi TK, et al. The microglial sensome revealed by direct RNA sequencing. Nature neuroscience. 2013;16(12):1896–1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sieger D, Moritz C, Ziegenhals T, Prykhozhij S, Peri F. Long-range Ca2+ waves transmit brain-damage signals to microglia. Developmental cell. 2012;22(6):1138–1148. [DOI] [PubMed] [Google Scholar]
- 16.Vasek MJ, Garber C, Dorsey D, et al. A complement–microglial axis drives synapse loss during virus-induced memory impairment. Nature. 2016;534(7608):538–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pluvinage JV, Haney MS, Smith BA, et al. CD22 blockade restores homeostatic microglial phagocytosis in ageing brains. Nature. 2019;568(7751):187–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Rothhammer V, Borucki DM, Tjon EC, et al. Microglial control of astrocytes in response to microbial metabolites. Nature. 2018;557(7707):724–728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Stephenson J, Nutma E, van der Valk P, Amor S. Inflammation in CNS neurodegenerative diseases. Immunology. 2018;154(2):204–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Thomas DC. The phagocyte respiratory burst: Historical perspectives and recent advances. Immunology letters. 2017;192:88–96. [DOI] [PubMed] [Google Scholar]
- 21.Bal-Price A, Brown GC. Inflammatory neurodegeneration mediated by nitric oxide from activated glia-inhibiting neuronal respiration, causing glutamate release and excitotoxicity. Journal of Neuroscience. 2001;21(17):6480–6491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yap YW, Whiteman M, Cheung NS. Chlorinative stress: an under appreciated mediator of neurodegeneration? Cellular signalling. 2007;19(2):219–228. [DOI] [PubMed] [Google Scholar]
- 23.Reynolds WF, Rhees J, Maciejewski D, et al. Myeloperoxidase polymorphism is associated with gender specific risk for Alzheimer’s disease. Experimental neurology. 1999;155(1):31–41. [DOI] [PubMed] [Google Scholar]
- 24.Nagra RM, Becher B, Tourtellotte WW, et al. Immunohistochemical and genetic evidence of myeloperoxidase involvement in multiple sclerosis. Journal of neuroimmunology. 1997;78(1–2):97–107. [DOI] [PubMed] [Google Scholar]
- 25.Gellhaar S, Sunnemark D, Eriksson H, Olson L, Galter D. Myeloperoxidase-immunoreactive cells are significantly increased in brain areas affected by neurodegeneration in Parkinson’s and Alzheimer’s disease. Cell and tissue research. 2017;369:445–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pravalika K, Sarmah D, Kaur H, et al. Myeloperoxidase and neurological disorder: a crosstalk. ACS chemical neuroscience. 2018;9(3):421–430. [DOI] [PubMed] [Google Scholar]
- 27.Chen JW, Breckwoldt MO, Aikawa E, Chiang G, Weissleder R. Myeloperoxidase-targeted imaging of active inflammatory lesions in murine experimental autoimmune encephalomyelitis. Brain. 2008;131(4):1123–1133. doi: 10.1093/brain/awn004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Chen JW, Querol Sans M, Bogdanov A Jr., Weissleder R. Imaging of myeloperoxidase in mice by using novel amplifiable paramagnetic substrates. Radiology. Aug 2006;240(2):473–81. doi: 10.1148/radiol.2402050994 [DOI] [PubMed] [Google Scholar]
- 29.Breckwoldt MO, Chen JW, Stangenberg L, et al. Tracking the inflammatory response in stroke in vivo by sensing the enzyme myeloperoxidase. Proc Natl Acad Sci U S A. Nov 25 2008;105(47):18584–9. doi: 10.1073/pnas.0803945105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang C, Pulli B, Jalali Motlagh N, et al. A versatile imaging platform with fluorescence and CT imaging capabilities that detects myeloperoxidase activity and inflammation at different scales. Theranostics. 2019;9(25):7525–7536. doi: 10.7150/thno.36264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wang C, Cheng D, Jalali Motlagh N, et al. Highly Efficient Activatable MRI Probe to Sense Myeloperoxidase Activity. Journal of Medicinal Chemistry. 2021;64(9):5874–5885. doi: 10.1021/acs.jmedchem.1c00038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wang C, Keliher E, Zeller MWG, et al. An activatable PET imaging radioprobe is a dynamic reporter of myeloperoxidase activity in vivo. Proceedings of the National Academy of Sciences. 2019;116(24):11966–11971. doi: 10.1073/pnas.1818434116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Fujita Y, Maeda T, Sato C, et al. Engulfment of Toxic Amyloid β-protein in Neurons and Astrocytes Mediated by MEGF10. Neuroscience. 2020;443:1–7. [DOI] [PubMed] [Google Scholar]
- 34.Damisah EC, Hill RA, Rai A, et al. Astrocytes and microglia play orchestrated roles and respect phagocytic territories during neuronal corpse removal in vivo. Science Advances. 2020;6(26):eaba3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Eddleston M, Mucke L. Molecular profile of reactive astrocytes—implications for their role in neurologic disease. Neuroscience. 1993;54(1):15–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta neuropathologica. 2010;119:7–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Šimić G, Lucassen PJ, Krsnik Ž, et al. nNOS expression in reactive astrocytes correlates with increased cell death related DNA damage in the hippocampus and entorhinal cortex in Alzheimer’s disease. Experimental neurology. 2000;165(1):12–26. [DOI] [PubMed] [Google Scholar]
- 38.Talantova M, Sanz-Blasco S, Zhang X, et al. Aβ induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss. Proceedings of the National Academy of Sciences. 2013;110(27):E2518–E2527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Argaw AT, Gurfein BT, Zhang Y, Zameer A, John GR. VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier breakdown. Proceedings of the National Academy of Sciences. 2009;106(6):1977–1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mahmoud S, Gharagozloo M, Simard C, Gris D. Astrocytes maintain glutamate homeostasis in the CNS by controlling the balance between glutamate uptake and release. Cells. 2019;8(2):184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Franco R, Cidlowski J. Apoptosis and glutathione: beyond an antioxidant. Cell Death & Differentiation. 2009;16(10):1303–1314. [DOI] [PubMed] [Google Scholar]
- 42.Li M-Z, Zheng L-J, Shen J, et al. SIRT1 facilitates amyloid beta peptide degradation by upregulating lysosome number in primary astrocytes. Neural regeneration research. 2018;13(11):2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Heithoff BP, George KK, Phares AN, Zuidhoek IA, Munoz-Ballester C, Robel S. Astrocytes are necessary for blood–brain barrier maintenance in the adult mouse brain. Glia. 2021;69(2):436–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Toft‐Hansen H, Füchtbauer L, Owens T. Inhibition of reactive astrocytosis in established experimental autoimmune encephalomyelitis favors infiltration by myeloid cells over T cells and enhances severity of disease. Glia. 2011;59(1):166–176. [DOI] [PubMed] [Google Scholar]
- 45.Bennett ML, Bennett FC, Liddelow SA, et al. New tools for studying microglia in the mouse and human CNS. Proceedings of the National Academy of Sciences. 2016;113(12):E1738–E1746. doi: 10.1073/pnas.1525528113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Amici SA, Dong J, Guerau-de-Arellano M. Molecular Mechanisms Modulating the Phenotype of Macrophages and Microglia. Frontiers in Immunology. 2017;8(NOV):1–18. doi: 10.3389/fimmu.2017.01520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jordão MJC, Sankowski R, Brendecke SM, et al. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science. 2019;363(6425)doi: 10.1126/science.aat7554 [DOI] [PubMed] [Google Scholar]
- 48.Li A, Wu Y, Pulli B, et al. Myeloperoxidase Molecular MRI Reveals Synergistic Combination Therapy in Murine Experimental Autoimmune Neuroinflammation. Radiology. 2019;293(1):158–165. doi: 10.1148/radiol.2019182492 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kirschbaum K, Sonner JK, Zeller MW, et al. In vivo nanoparticle imaging of innate immune cells can serve as a marker of disease severity in a model of multiple sclerosis. Proc Natl Acad Sci U S A. Nov 15 2016;113(46):13227–13232. doi: 10.1073/pnas.1609397113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wang J, Jalali Motlagh N, Wang C, et al. d-mannose suppresses oxidative response and blocks phagocytosis in experimental neuroinflammation. Proc Natl Acad Sci U S A. Nov 2 2021;118(44)doi: 10.1073/pnas.2107663118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Alam MM, Lee J, Lee SY. Recent Progress in the Development of TSPO PET Ligands for Neuroinflammation Imaging in Neurological Diseases. Nuclear Medicine and Molecular Imaging. 2017;51(4):283–296. doi: 10.1007/s13139-017-0475-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Evens N, Muccioli GG, Houbrechts N, et al. Synthesis and biological evaluation of carbon-11- and fluorine-18-labeled 2-oxoquinoline derivatives for type 2 cannabinoid receptor positron emission tomography imaging. Nuclear Medicine and Biology. 2009;36(4):455–465. doi: 10.1016/j.nucmedbio.2009.01.009 [DOI] [PubMed] [Google Scholar]
- 53.Shukuri M, Mawatari A, Takatani S, et al. Synthesis and Preclinical Evaluation of 18F-Labeled Ketoprofen Methyl Esters for Cyclooxygenase-1 Imaging in Neuroinflammation. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2022;63(11):1761–1767. doi: 10.2967/jnumed.121.263713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.de Vries EFJ, Doorduin J, Dierckx RA, van Waarde A. Evaluation of [11C]rofecoxib as PET tracer for cyclooxygenase 2 overexpression in rat models of inflammation. Nuclear Medicine and Biology. 2008;35(1):35–42. doi: 10.1016/j.nucmedbio.2007.07.015 [DOI] [PubMed] [Google Scholar]
- 55.Gerwien H, Hermann S, Zhang X, et al. Imaging matrix metalloproteinase activity in multiple sclerosis as a specific marker of leukocyte penetration of the blood-brain barrier. Science Translational Medicine. 2016;8(364):1–13. doi: 10.1126/scitranslmed.aaf8020 [DOI] [PubMed] [Google Scholar]
- 56.Baez-Pagan CA, Martinez-Ortiz Y, Otero-Cruz JD, et al. Potential role of caveolin-1-positive domains in the regulation of the acetylcholine receptor’s activatable pool: implications in the pathogenesis of a novel congenital myasthenic syndrome. Channels (Austin). May-Jun 2008;2(3):180–90. doi: 10.4161/chan.2.3.6155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zamanian JL, Xu L, Foo LC, et al. Genomic analysis of reactive astrogliosis. Journal of neuroscience. 2012;32(18):6391–6410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lopez-Sanchez C, Poejo J, Garcia-Lopez V, Salazar J, Garcia-Martinez V, Gutierrez-Merino C. Kaempferol prevents the activation of complement C3 protein and the generation of reactive A1 astrocytes that mediate rat brain degeneration induced by 3-nitropropionic acid. Food and Chemical Toxicology. 2022;164:113017. [DOI] [PubMed] [Google Scholar]
- 59.Liu Y, Jiang H, Qin X, Tian M, Zhang H. PET imaging of reactive astrocytes in neurological disorders. Eur J Nucl Med Mol Imaging. Mar 2022;49(4):1275–1287. doi: 10.1007/s00259-021-05640-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Attfield KE, Jensen LT, Kaufmann M, Friese MA, Fugger L. The immunology of multiple sclerosis. Nature Reviews Immunology. 2022;22(12):734–750. doi: 10.1038/s41577-022-00718-z [DOI] [PubMed] [Google Scholar]
- 61.Fransen NL, Hsiao C-C, Poel Mvd, et al. Tissue-resident memory T cells invade the brain parenchyma in multiple sclerosis white matter lesions. Brain. 2020;143(6):1714–1730. doi: 10.1093/brain/awaa117 [DOI] [PubMed] [Google Scholar]
- 62.Benallegue N, Nicol B, Lasselin J, et al. Patients With Severe Multiple Sclerosis Exhibit Functionally Altered CD8+ Regulatory T Cells. Neurology - Neuroimmunology Neuroinflammation. 2022;9(6):e200016. doi: 10.1212/nxi.0000000000200016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Comi G, Bar‐Or A, Lassmann H, et al. Role of B Cells in Multiple Sclerosis and Related Disorders. Annals of Neurology. 2021;89(1):13–23. doi: 10.1002/ana.25927 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Magliozzi R, Howell OW, Calabrese M, Reynolds R. Meningeal inflammation as a driver of cortical grey matter pathology and clinical progression in multiple sclerosis. Nature Reviews Neurology. 2023;19(8):461–476. doi: 10.1038/s41582-023-00838-7 [DOI] [PubMed] [Google Scholar]
- 65.Kuhlmann T, Ludwin S, Prat A, Antel J, Brück W, Lassmann H. An updated histological classification system for multiple sclerosis lesions. Acta Neuropathologica. 2017;133(1):13–24. doi: 10.1007/s00401-016-1653-y [DOI] [PubMed] [Google Scholar]
- 66.Frischer JM, Weigand SD, Guo Y, et al. Clinical and pathological insights into the dynamic nature of the white matter multiple sclerosis plaque. Annals of Neurology. 2015;78(5):710–721. doi: 10.1002/ana.24497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Dal-Bianco A, Grabner G, Kronnerwetter C, et al. Slow expansion of multiple sclerosis iron rim lesions: pathology and 7 T magnetic resonance imaging. Acta Neuropathologica. 2017;133(1):25–42. doi: 10.1007/s00401-016-1636-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Calvi A, Carrasco FP, Tur C, et al. Association of Slowly Expanding Lesions on MRI With Disability in People With Secondary Progressive Multiple Sclerosis. Neurology. 2022;98(17):e1783–e1793. doi: 10.1212/wnl.0000000000200144 [DOI] [PubMed] [Google Scholar]
- 69.Michalska P, León R. When it comes to an end: Oxidative stress crosstalk with protein aggregation and neuroinflammation induce neurodegeneration. Antioxidants 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Forloni G, La Vitola P, Balducci C. Oligomeropathies, inflammation and prion protein binding. Frontiers in Neuroscience 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Chamani S, Bianconi V, Tasbandi A, et al. Resolution of Inflammation in Neurodegenerative Diseases: The Role of Resolvins. Mediators of Inflammation. 2020;2020 doi: 10.1155/2020/3267172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wightman DP, Jansen IE, Savage JE, et al. A genome-wide association study with 1,126,563 individuals identifies new risk loci for Alzheimer’s disease. Nature Genetics. 2021;53(9)doi: 10.1038/s41588-021-00921-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Srinivasan K, Friedman BA, Etxeberria A, et al. Alzheimer’s Patient Microglia Exhibit Enhanced Aging and Unique Transcriptional Activation. Cell Reports. 2020/6//2020;31(13):107843–107843. doi: 10.1016/J.CELREP.2020.107843 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Rangaraju S, Dammer EB, Raza SA, et al. Identification and therapeutic modulation of a pro-inflammatory subset of disease-associated-microglia in Alzheimer’s disease. Molecular Neurodegeneration. 2018;13(1)doi: 10.1186/s13024-018-0254-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Krasemann S, Madore C, Cialic R, et al. The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity. 2017/9// 2017;47(3):566–581.e9. doi: 10.1016/j.immuni.2017.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bisht K, Sharma KP, Lecours C, et al. Dark microglia: A new phenotype predominantly associated with pathological states. GLIA. 2016;64(5)doi: 10.1002/glia.22966 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Rayasam A, Hsu M, Kijak JA, et al. Immune responses in stroke: how the immune system contributes to damage and healing after stroke and how this knowledge could be translated to better cures? Immunology. Jul 2018;154(3):363–376. doi: 10.1111/imm.12918 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. May 2010;87(5):779–89. doi: 10.1189/jlb.1109766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Tarkowski E, Rosengren L, Blomstrand C, et al. Intrathecal release of pro- and anti-inflammatory cytokines during stroke. Clin Exp Immunol. Dec 1997;110(3):492–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Boehme AK, Esenwa C, Elkind MS. Stroke Risk Factors, Genetics, and Prevention. Circ Res. Feb 3 2017;120(3):472–495. doi: 10.1161/CIRCRESAHA.116.308398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Forghani R, Kim HJ, Wojtkiewicz GR, et al. Myeloperoxidase propagates damage and is a potential therapeutic target for subacute stroke. J Cereb Blood Flow Metab. Mar 2015;35(3):485–93. doi: 10.1038/jcbfm.2014.222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Anderson NM, Simon MC. The tumor microenvironment. Curr Biol. Aug 17 2020;30(16):R921–R925. doi: 10.1016/j.cub.2020.06.081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Venteicher AS, Tirosh I, Hebert C, et al. Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science. Mar 31 2017;355(6332)doi: 10.1126/science.aai8478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Mitsdoerffer M, Aly L, Barz M, et al. The glioblastoma multiforme tumor site promotes the commitment of tumor-infiltrating lymphocytes to the T(H)17 lineage in humans. Proc Natl Acad Sci U S A. Aug 23 2022;119(34):e2206208119. doi: 10.1073/pnas.2206208119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, et al. Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas. Clin Cancer Res. Aug 15 2008;14(16):5166–72. doi: 10.1158/1078-0432.CCR-08-0320 [DOI] [PubMed] [Google Scholar]
