Regulation of CNS precursor function by neuronal chemokines

Regulation of CNS precursor function by neuronal chemokines

Journal Pre-proof Regulation of CNS precursor function by neuronal chemokines Adrianne Eve Scovil Watson, Kara Goodkey, Tim Footz, Anastassia Voronova...

5MB Sizes 1 Downloads 57 Views

Journal Pre-proof Regulation of CNS precursor function by neuronal chemokines Adrianne Eve Scovil Watson, Kara Goodkey, Tim Footz, Anastassia Voronova

PII:

S0304-3940(19)30636-6

DOI:

https://doi.org/10.1016/j.neulet.2019.134533

Reference:

NSL 134533

To appear in:

Neuroscience Letters

Received Date:

21 June 2019

Revised Date:

16 September 2019

Accepted Date:

1 October 2019

Please cite this article as: Scovil Watson AE, Goodkey K, Footz T, Voronova A, Regulation of CNS precursor function by neuronal chemokines, Neuroscience Letters (2019), doi: https://doi.org/10.1016/j.neulet.2019.134533

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. 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. © 2019 Published by Elsevier.

Title: Regulation of CNS precursor function by neuronal chemokines Authors: Adrianne Eve Scovil Watson1, Kara Goodkey1, Tim Footz1 and Anastassia Voronova1-4 # Affiliations: Department of Medical Genetics1, Neurosciences and Mental Health Institute2, MS Centre3, Women and Children’s Hospital Research Institute4, Faculty of

oo f

Medicine & Dentistry, University of Alberta, Edmonton, Canada

# Corresponding author ([email protected], Department of Medical Genetics,

pr

University of Alberta, 8-39 Medical Sciences Building, Edmonton, AB T6G2H7 Canada)

Highlights

Neurons communicate with OPCs via paracrine signalling and/or neuronal

e-



activity/synapses;

OPCs and NPCs express chemokine receptors and respond to chemokines;



Neuronally secreted chemokines regulate NPC and OPC migration, survival,

Pr



proliferation and differentiation;

Mutations in chemokine signalling genes are detected both in patients with MS

na l



and neurodevelopmental disorders, raising the possibility that aberrant neuronprecursor chemokine signalling may play a role in these disorders.

ur

Abstract

Oligodendrocyte and neural precursor cells (OPCs and NPCs, respectively) in the central

Jo

nervous system (CNS) have diverse roles in development and homeostasis. During development, precursors build the CNS. In adulthood, they maintain their ability to proliferate and generate differentiated progeny, indicating their tremendous potential to regenerate and repair injured or degenerated CNS. How can we utilize this capability? Cross-talk between neurons and OPCs may hold some clues. Neurons communicate with OPCs via two mechanisms: 1) paracrine secretion of ligands, and 2) neuronal activity and bona fide synapses with OPCs. Intriguingly, OPCs express receptors for chemokines,

which are small signalling molecules produced by various cells, including neurons. In addition to inducing chemotaxis, chemokines also regulate cell proliferation, survival and differentiation. In this review, we will summarize the roles of neuronally secreted chemokines and their documented ability to directly regulate the diverse functions of OPCs and NPCs in the developing as well as adult normal and injured CNS. We will focus on the following neuronal chemokines: CCL2, CCL3, CCL20, CCL21, CXCL1, CXCL8, CXCL9, CXCL10, CXCL11, CXCL12 and CX3CL1. We will discuss the implications for neuronal chemokine signalling in OPCs and NPCs not only in

oo f

developmental myelination and adult CNS regeneration, but also in cognition, behavior,

Jo

ur

na l

Pr

e-

pr

neuroinflammation and neuronal function.

2

Abbreviations APC - adenomatous polyposis coli ASD – autism spectrum disorder BBB – blood-brain barrier BDNF - brain derived neurotrophic factor CNS - central nervous system cOPC - committed OPC

oo f

CPM - counts per million E – embryonic day EAE - experimental autoimmune encephalomyelitis

pr

EDSS - expanded disability status scale FKN – fractalkine (CX3CL1)

e-

FKPM - fragments per kilobase million Gro-1 - growth stimulating activity, alpha (CXCL1)

IFN- - interferon gamma IL-1 - interleukin-1 beta

na l

IL-8 - interleukin-8 (CXCL8)

Pr

GW – gestational week

ImOLGs – immune oligodendrocyte lineage cells IP-10 - IFN-γ-induced protein 10 (CXCL10) IP-9 - IFN-γ-induced protein 9 (CXCL11)

ur

LARC - liver activation regulated chemokine (CCL20) MBP – myelin basic protein

Jo

MCP-1 - monocyte chemoattractant protein-1 (CCL2) MIG – monokine induced by IFN-γ (CXCL9) MIP-1α - macrophage inflammatory protein-1α (CCL3) MS – multiple sclerosis NF-κB - nuclear factor-kappa B NG2 - neural/glial antigen 2 NPC – neural precursor cell

3

OPC – oligodendrocyte precursor cell OL - oligodendrocyte P – postnatal day PDGF - platelet derived growth factor PDGFR - platelet derived growth factor alpha RFP – red fluorescent protein RRMS - relapse-remitting MS

SLC - secondary lymphoid-tissue chemokine (CCL21) SNP - single nucleotide polymorphism

TGF-β - transforming growth factor beta TNF- - tumor necrosis factor alpha VSMC - vascular smooth muscle cells

Pr

Keywords

e-

SVZ – subventricular zone

pr

SPMS - secondary progressive MS

oo f

SDF-1 - stromal cell–derived factor (CXCL12)

Chemokine, neuron-glia interactions, OPC, neural stem cell, oligodendrocyte,

1. Introduction

na l

neuroinflammation, multiple sclerosis, neurodevelopment

Chemokines (chemotactic cytokines) are secreted signalling molecules that initiate chemotaxis in nearby responsive cells. Chemokines comprise a family of small 8-10 kDa

ur

proteins that have Cys (cysteine) residues separated by 0-3 amino acids. The distance between Cys residues forms the basis of their nomenclature, ranging from CC, CXC and

Jo

XC to CX3C, where X represents amino acid(s) positioned between the Cys residues in their N-terminus. The CC group of chemokines is the largest, being comprised of 27 members, followed by the CXC group with 17 members, the XC group with 2 members and the CX3C group with just 1 member [1]. Secretion of chemokines is regulated at the level of transcription and translation as well as post-translationally in the endoplasmic reticulum, Golgi, and at or near the cell surface [2]. Chemokine release is also regulated by inflammatory or injurious stimuli [2-5]. In this light, chemokines are translated with a

4

~23 amino acid signal sequence, which is usually cleaved prior to secretion of the mature protein [4]. Moreover, chemokines can be truncated from the C- or N-terminus via proteases leading to change in their function (reviewed in [4] and described below in each chemokine section where appropriate). Transport of chemokines inside cells occurs via the canonical protein trafficking pathway associated with endoplasmic reticulum and Golgi [6]. Overall, the secretion of chemokines varies vastly between cell types and activation stimuli and can involve a myriad of secretory pathways, including secretory granules and vesicles [2, 5, 6].

oo f

Chemokines converge their function on ~20 chemokine receptors discovered to

date [7]. Chemokine receptors are G protein-coupled seven transmembrane receptors that were initially discovered in leukocytes and subsequently shown to be critical regulators

pr

of their recruitment to sites of inflammation [7]. Since their initial discovery in the

immune system, chemokines have been demonstrated to be important in the central

e-

nervous system (CNS) for microglia, neurons, glia and neural stem cells as well as infiltrating immune cells during neuroinflammation [7-10]. In addition, the role of

Pr

chemokines extends beyond chemotaxis. Chemokines are important in cellular communication, proliferation, survival and differentiation of various CNS cell types [7, 11, 12]. With regards to cellular communication, chemokines and chemokine receptors

na l

can be expressed by a variety of CNS cells, such as microglia, endothelial cells, astrocytes, oligodendrocytes and neurons [7, 9, 12-18]. This has been recently reinforced with bulk and single-cell RNA sequencing reports from various CNS cell types (e.g. [1924]). Notably, chemokines can have protective or degenerative effects on CNS cells [7,

ur

12, 13]. This review will focus on the neuronal chemokine regulation of oligodendrocyte

Jo

precursor cell (OPC) function in the developing and adult CNS.

2. Neuron-OPC interactions Oligodendrocytes (OLs) are responsible for the formation of myelin that wraps

around neuronal axons. Moreover, OLs were recently found to regulate presynaptic properties and neurotransmission [25, 26], which suggests OLs have versatile roles in neuron homeostasis and neurotransmission beyond myelin formation and maintenance. The role of OL lineage cells in neuronally driven processes, such as cognition, has been

5

recently exemplified through an elegant study, where conditional deletion of BDNF (brain derived neurotrophic factor) receptor TrkB in OPCs led to poor outcomes in cognitive tests [27]. The ability of OLs and myelin to alter neuronal function is further reinforced by a recent study, in which clemastine, an antihistamine drug that showed remyelinating capacity in MS patients [28], reversed neuronal and behavioural deficits in a mouse model of neurodevelopmental Williams syndrome [29]. Furthermore, clemastine rescues behavioral deficits in socially isolated mice [30]. In turn, neuronal activity is known to boost myelination [31-34]. It has been proposed that this activity dependent

oo f

myelination could explain, at least in part, why pianists and jugglers have increased

myelination in specific brain areas [35, 36], while schizophrenia patients, who have

abnormal function of parvalbumin-positive interneurons, have decreased myelination

pr

[37]. Finally, new OL genesis and myelination influenced by neuronal activity are

necessary for new motor skill learning [32]. Thus, neuron-oligodendroglia interactions

e-

are important in both developing and adult CNS.

OLs in the developing CNS are generated via a two-step process: 1) neural

Pr

precursor cell (NPC) commitment to OPC formation; and 2) OPC differentiation into OL [38]. In the adult CNS, there are two sources of cells that generate OLs: subventricular zone (SVZ) NPCs and parenchymal OPCs, both of which arise from embryonic NPCs

na l

and OPCs [23, 39-44]. Neuron-OPC cell-to-cell communication occurs both in the developing and adult CNS in at least two ways [45]: 1) paracrine regulation of OPC function by neuronally secreted ligands [46-48]; and 2) via neuronal activity and bona fide synapses with OPCs [32, 33, 49-52].

ur

Intriguingly, OPCs and OLs enhance the expression of immune genes, including

chemokine receptors and ligands, in demyelinating conditions in both rodents and

Jo

humans. This raises the possibility that OL lineage cells may have additional roles in modulating immune responses [21, 22, 53]. Similar to immune cells, OPCs are highly migratory and can respond to several chemokines, as discussed below. The roles of chemokine receptors in OPCs, however, extend beyond chemotaxis and include regulation of OPC proliferation, survival and differentiation (Fig. 1 and discussed below). Both inhibitory and excitatory neurons secrete various chemokines in the developing [46, 54] as well as adult healthy and injured CNS [3].

6

Chemokines such as CCL2, CCL3, CXCL1, CXCL10, CXCL12 and CX3CL1 affect synaptic responses, neuronal activity and/or neuronal ion channel function [55-60]. Moreover, some reports suggest neuronal activity can in turn trigger chemokine release. For example, CXCL12 is localized in both types of neuronally secreted vesicles, small and dense core vesicles. Additionally, potassium-induced depolarization of neurons leads to a large release of CXCL12 into the culture medium [61, 62]. Similarly, CCL2 is packaged into dense core vesicles and their release is induced by calcium-dependent depolarization from dorsal root ganglion neurons [63]. In contrast, while CCL21 is

oo f

present in the synaptic cleft, pre-synaptic structures and dense core vesicles [64, 65], its release is currently only known to depend on glutamate-induced neuron damage [64]. Pharmacogenetic or optogenetic stimulation of neuronal activity is known to

pr

increase proliferation and differentiation of OPCs, leading to increased axonal

myelination in murine CNS in vivo [32, 34]. Interestingly, blocking neuronal activity

e-

dependent vesicle release in cultured neurons or the developing zebrafish nervous system reduces axonal myelination [66-68]. Whether the dependence of neuronal activity-

Pr

induced myelination on vesicular release is linked to chemokine secretion remains to be addressed.

In this review, we will discuss the roles of the known repertoire of neuronal

na l

chemokines, which has been reviewed elsewhere [3], on OPC function in the developing as well as adult healthy and injured CNS. We will first focus on neuronal chemokines that have more extensive literature on their roles in OPC function (CXCL1, CXCL12 and CX3CL1) and then summarize the roles of the less described neuronal chemokine

ur

signalling axis in OPCs and NPCs (CXCL8, CXCL9, CXCL10, CXCL11, CCL2, CCL3,

Jo

CCL20 and CCL21).

3. CXCL1

CXCL1 (C-X-C motif ligand 1) or Gro-1α (Growth Stimulating Activity, Alpha)

plays an important role in mediating OPC and NPC migration and proliferation in the developing central nervous system [69, 70] (Figs. 1-2). In the murine brain, CXCL1 is expressed in neurons, astrocytes, and endothelial cells (Fig. 3A) and is increased after induced seizures in rats [71]. Human studies demonstrate CXCL1 is expressed in neurons

7

during gestation (27 to 36 GW) and disappears by 2 years of age [72], which suggests that CXCL1 is functional during CNS development. In agreement, murine neural/glial antigen 2 (NG2)-positive OPCs in the spinal cord white matter tracts express CXCR2, a cognate receptor for CXCL1 (Fig. 3D), and respond to CXCL1 signalling as early as embryonic day (E) 14 [69, 70]. A recent RNA-sequencing report confirms CXCR2 expression in developmental OPCs and demonstrates its highest expression in embryonic spinal cord OPCs (Fig. 4A). At this developmental stage OPCs are regulated by a number of growth factors,

oo f

one of which being platelet derived growth factor (PDGF) [69]. Through CXCR2,

CXCL1 acts synergistically with PDGF to increase OPC proliferation and reduce their migration in the white matter tracts of the developing spinal cord [69, 70]. While the

pr

mechanism is largely unknown, CXCL1-dependent migration of OPCs may be due to

CXCL1-mediated increase in cell-substrate adhesion [70], but is independent of calcium

e-

signalling and activation of the cell cycle [73]. In contrast, CXCL1 acts as a chemoattractant for rat SVZ NPCs and increases their migration in culture [74].

Pr

Interestingly, dysmyelinating jimpy mutant mice that harbour a point mutation in proteolipid protein (PLP) show elevated CXCL1 expression that is correlated with elevated OPC proliferation [75]. Moreover, estrogen receptor  (ER) is believed to

na l

promote axon myelination at least in part by inducing CXCL1 expression [76]. Human fetal OPCs also express CXCR2 and proliferate in response to CXCL1 [77, 78]. Rat ventral midbrain NPCs exhibit increased proliferation in response to CXCL1 [79]. Mouse SVZ NPCs increase differentiation into O4-positive OLs in the

ur

presence of CXCL1 [80]. In agreement, cultured postnatal CXCR2 knockout murine OPCs have reduced ability to differentiate into O1-positive OLs [81]. In vivo, CXCR2

Jo

knockout leads to decreased OPC proliferation and increased migration, resulting in myelin mis-localization, decreased myelin thickness and hypomyelination leading to impaired nerve conduction velocity [70, 81]. In adulthood, CXCL1 is expressed in astrocytes in white matter surrounding active multiple sclerosis (MS) lesions, but not in astrocytes from normal samples [77]. In vitro, CXCL1 expression in human fetal astrocytes is induced with IL-1 (Interleukin-1 beta), but not with TGF- (Transforming growth factor beta) or IFN- (Interferon

8

gamma) [77], suggesting that the inflammatory environment may mediate astrocytic expression and secretion of CXCL1. In an adult murine EAE (Experimental autoimmune encephalomyelitis) model of MS, constitutive overexpression of CXCL1 in astrocytes produced conflicting results. Omari et al. show that CXCL1 overexpression in astrocytes results in less inflammation and demyelination followed by a greater proliferation of OL lineage cells and more remyelination compared to wild type[82]. This protective effect of CXCL1 is supported by a different report showing CXCL1 reduces virally induced OL apoptosis in the spinal cord, while blocking receptor function increases the severity of the

oo f

viral infection [83]. In contrast, Grist et al. show expression of CXCL1 in astrocytes leads to increased severity of EAE that is associated with increased recruitment of neutrophils [84]. This is corroborated by another report, where injections of CXCR2-specific

pr

antibodies or CXCR2 antagonist were shown to enhance remyelination in lysolethicin-

induced spinal cord demyelination mouse model [85]. These differences in results may

e-

arise from different constructs used to overexpress CXCL1 that have a direct impact on protein stability [84] suggesting that the concentration and/or duration of CXCL1 may be

Pr

an important determinant in its ability to influence de- and remyelination. Single-nuclei RNA sequencing from MS patient brain lesions show CXCL1 is expressed in pericytes, endothelial cells, vascular smooth muscle cells (VSMCs) and macrophages (Fig. 3C)

addressed.

4. CXCL12

na l

[22]. The contribution of CXCL1 from these cells to OPC function remains to be

ur

CXCL12 (C-X-C motif chemokine 12), also known as stromal cell–derived factor

1 (SDF-1), is important for regulation of OPC survival, migration, proliferation and

Jo

differentiation (Fig. 1). Reporter mice expressing CXCL12-RFP (red fluorescent protein) fusion protein demonstrate that CXCL12 is first expressed in E12.5 spinal cord endothelial cells in meninges [86, 87]. Over the course of postnatal development, CXCL12 begins to be expressed in microglia, astrocytes and neurons; expression in these cell types persists throughout adulthood [88, 89]. RNA-sequencing data from murine adult CNS cell types confirms these findings (Fig. 3A) [19]. Interestingly, the expression of meningeal CXCL12 decreases in adulthood [88]. In adult rodent CNS, neuronal

9

CXCL12 expression is detected in multiple brain structures, including the cerebral cortex, basal ganglia, amygdala, hippocampus and thalamus amongst others [89]. Widespread neuronal expression of CXCL12 is upregulated in response to spinal nerve ligation, a model for murine neuropathic pain [90]. Similarly, CXCL12 can be detected in active and inactive lesions in MS patients, although the expression appears to be restricted to astrocytes and blood vessels [91]. Furthermore, the level of CXCL12 expression in MS patients is correlated with the degree of disease severity [92]. Brain damage is also associated with a cleaved form of CXCL12, CXCL12(5-67), produced by partial cleavage

oo f

of the N-terminus by the soluble factors matrix metalloproteinase 2 and 9 [93]. Since CXCL12 requires the first 8 amino acids in the N-terminus to bind to its canonical receptor CXCR4 (C-X-C chemokine receptor type 4) [94], partial cleavage in

pr

CXCL12(5-67) leads to impairment in CXCR4 affinity, inhibition of NPC migration and

induction of apoptosis via signalling through CXCR3 (C-X-C chemokine receptor type 3)

e-

[95]. The function of CXCR4 is described below and the function of CXCR3 is discussed in the “IFN-γ-inducible chemokines CXCL9, CXCL10 and CXCL11” section.

Pr

CXCL12 mRNA is spliced into various isoforms, such as alpha and beta isoforms [96]. While CXCL12β is exclusively expressed by endothelial cells, the CXCL12α isoform is predominantly expressed by neurons in the adult mouse brain [96]. Exogenous application of CXCL12α to primary mixed OL lineage cell cultures increases

na l

proliferation of PDGFRα-positive OPCs, as well as increases myelin basic protein (MBP)-positive OLs and myelin segment formation [97]. The major role of CXCL12 in regulating the immune system is believed to be through stimulating the migration and

ur

adhesion of leukocytes [96]. However, in injured CNS, such as ischemia mouse models, CXCL12 is involved in neurogenesis and angiogenesis (reviewed in [87]).

Jo

OPCs express two receptors for CXCL12: CXCR4 and CXCR7 (C-X-C

chemokine receptor type 7) [98-101]. During development, they are thought to have independent functions, as they have little spatial or temporal overlap of expression in OPCs [98, 99, 102, 103]. CXCR4 is highly expressed in murine OPCs during early embryonic and postnatal development (Fig. 4A) [98]. Interestingly, while CXCR4 expression is decreased during OPC differentiation, the expression of CXCR7 is upregulated during OPC differentiation and peaks in mature OLs [99, 100]. In agreement,

10

CXCL12 enhances OPC migration via the CXCR4 signalling axis [98] resulting in activation of the MEK/ERK and PI3K/AKT pathways [104], and inhibition of the MAPK pathway [100]. CXCL12 enhances OPC differentiation via CXCR7 signalling resulting in activation of the ERK1/2 pathway axis [99]. CXCL12 stimulation of fetal human OPCs leads to an increased differentiation into OLs, which display more mature morphology [105]. CXCL12 also enhances NPC migration and survival [98, 106, 107], decreases human fetal NPC proliferation in vitro and in adult human hippocampal slice ex vivo cultures [108], as well as promotes OPC

oo f

survival and proliferation [97, 100]. NPCs isolated from CXCR4 knockout mice have

decreased migration, but not differentiation into OLs [98, 106]. Furthermore, CXCR4

inhibition with function blocking antibodies or siRNA knockdown of CXCR4 in OPCs

pr

blocks CXCL12-induced proliferation and migration [97, 100]. Specifically, CXCR4

antagonism inhibits the proliferation effect of the neuronal isoform CXCL12α [97]. On

effect on proliferation/migration [100].

e-

the other hand, CXCR7 knockdown decreases MBP protein expression, but has no direct

Pr

Notably, CXCR4 protein levels are regulated by CXCR7 and Wnt signalling [109, 110]. siRNA knockdown of CXCR7 in OPCs in vitro leads to increased protein expression of CXCL12 and CXCR4 [100]. High activity of Wnt in mice lacking

na l

expression of Wnt repressor adenomatous polyposis coli (APC) in OL lineage cells increases expression of CXCR4 in corpus callosum and decreases OPC differentiation, resulting in hypomyelination [110]. These mice also display aberrant OPC clustering around blood vessels, which can be rescued with CXCR4/CXCL12 inhibitor AMD3100

ur

[110]. Moreover, conditional knockout of APC in OPCs leads to displacement of astrocyte end feet from blood vessels and a leaky blood-brain-barrier (BBB) [111]. Thus,

Jo

it is possible that as Wnt signalling decreases during normal development, OPC-specific expression of CXCR4 is downregulated, migration and proliferation decrease and differentiation occurs [110]. While the regulation of CXCR7 expression in OPCs is currently not known, it has been shown to require NF-κB (nuclear factor-kappa B) in rhabdomyosarcoma [112] and its expression is repressed by HIC1 (hypermethylated in cancer 1) in U2OS human osteosarcoma cells [113]. Notably, CXCR4 and CXCR7 were also proposed to form heterodimers to regulate CXCL12-mediated G protein signalling

11

([114] and reviewed in [87]), however, the role of these heterodimers in OPCs remains to be addressed. Please note that CXCL12-CXCR4-CXCR7 signalling axis in glioblastoma and ischemia has been extensively reviewed in [87, 115] and will not be discussed here. In contrast to the developing CNS, in which CXCR4 expression is downregulated over time, CXCR4 expression in demyelinated CNS is upregulated [116]. The EAE model shows that the level of CXCR4/CXCL12 expression in spinal cord is tightly linked to the induction of disease, but the levels remain high even after spontaneous recovery, underlining the importance of CXCL12 in the process of myelination [116]. Furthermore,

oo f

CXCL12-positive microglia and astrocytes are present in the corpus callosum of

cuprizone-demyelinated, but not in control mice, and decrease in remyelinating mice, but do not completely disappear [117]. Robust upregulation of CXCL12 in demyelinated

pr

corpus callosum is accompanied by a sharp increase in CXCR4-, NG2-positive activated OPCs [117]. Additionally, viral-induced demyelination in mice increases both CXLC12

e-

expression and the number of PDGFRα-, CXCR4-positive OPCs in the spinal cord [118]. Treatment of cuprizone-demyelinated mice with AMD3100, an inhibitor of CXCL12

Pr

binding to CXCR4, or siRNA against CXCR4 prevents OPC differentiation and remyelination, likely due to the inhibition of the migratory action of CXCL12/CXCR4 axis [117]. Additionally, CXCR4 inhibitor AMD3100 causes an increase in OPC

na l

proliferation in the SVZ, but not within the remyelinating corpus callosum area [117]. In contrast, short term treatment of a viral-induced demyelinated mice with AMD3100 followed by a 2 week recovery period increases the amount of mature OLs and remyelination, and improves clinical scores [118]. The apparent difference in effect of

ur

AMD3100 could be attributed to different demyelination mouse models or the area of CNS analyzed (brain vs. spinal cord).

Jo

Treatment of cuprizone-demyelinated mice with the CXCR7 antagonist CCX771

increases OPC proliferation and differentiation resulting in enhanced remyelination [119]. Additionally, CXCR7 antagonism has been shown to reduce T-cell infiltration and decrease axonal injury in the spinal cord of EAE mice [120]. While the results obtained in demyelination mouse models with CXCR7 antagonists seem to be contrary to the positive role of CXCR7 in developmental OL formation from OPCs, recent reports suggest that CXCR7 acts as a scavenger receptor internalizing CXCL12 [98, 121, 122],

12

which suggests CXCR7 may be limiting the amount of freely available CXCL12. Indeed, demyelinated mice treated with CXCR7 antagonist show increased CXCL12 levels and a slightly elevated number of OPCs positive for phosphorylated CXCR4 [119]. This suggests that inactivation of CXCR7 leads to activation of CXCR4 in OPCs [119]. This is supported by the report that shows CXCR7 knockdown in cultured OPCs increases CXCR4 expression [100]. Finally, when CXCR4 and CXCR7 antagonists are co-applied during remyelination, this leads to decreased OL numbers and myelin intensity in the corpus callosum compared to treatment with CXCR7 antagonist alone, suggesting that

oo f

CXCR7 restricts CXCR4-mediated OPC maturation [119]. Notably, CXCR4 is expressed in immune oligodendroglia (ImOLGs), in addition to immune cells, in brain lesions in MS patients (Fig. 4C) [22]. In the future it will be interesting to understand the role of

pr

CXCL12-CXCR4 signalling in this novel class of OL lineage cells in neuroinflammation

e-

and remyelination.

5. CX3CL1

Pr

The only member of the CX3C family, CX3CL1 (C-X3-C motif ligand 1) or fractalkine (FKN), has a unique transmembrane structure which can be cleaved to produce a soluble glycoprotein by metalloproteinases ADAM 10 and ADAM 17 or

na l

lysosomal cysteine protease and cathepsin S (CatS) [123]. While the cleaved form contains only the chemokine domain located in the N-terminus, the transmembrane form contains the chemokine domain, a mucin-like stalk, a transmembrane domain and an intracellular C-terminal domain [123]. Both the membrane bound and secreted forms of

ur

CX3CL1 are bioactive. The transmembrane form of CX3CL1 has been proposed to act as an adhesion molecule, and the soluble form to act as a chemoattractant [124]. Notably,

Jo

transmembrane and soluble CX3CL1 elicit different cytokine response from immune cells [125, 126]. Moreover, the CX3CL1-CX3CR1 signalling axis has been shown to have both deleterious and protective effects towards neurons in mouse models of neurological disorders [123]. In this light, neuron-specific expression of soluble CX3CL1, and not the transmembrane form lacking the ADAM10/17 cleavage site, rescues neuronal cell death in a synucleinopathy mouse model [127]. This is corroborated by additional reports showing that infusion of the soluble form of CX3CL1 into the CNS

13

has neuroprotective properties in a stroke mouse model [128]. Notably, in addition to widely-studied neuronal death in stroke and alpha-synucleinopathies, there is also extensive, but less studied oligodendroglial cell death and/or myelin deficiency [129132]. It is not currently known whether CX3CL1 has a protective effect on oligodendroglial lineage cells in these conditions. In the developing mouse brain, CX3CL1 is expressed by ventral forebrain inhibitory interneurons that migrate into the developing dorsal forebrain (cortex) and is secreted by interneurons in culture [46]. Ablation of these interneurons in vivo reduces

oo f

the formation of OPCs and decreases CX3CL1-positive cells in the late embryonic

cortical SVZ area, where dorsal OPCs are first generated [46]. Apart from interneurons, CX3CL1 is also expressed in excitatory neurons in the developing mouse cortex [54,

pr

133] and is regulated by the CXCL12 signalling pathway [133]. In the adult brain,

CX3CL1 is expressed in the hippocampus, olfactory blub, cerebral cortex, amygdala,

e-

globus pallidus and thalamus [134]. In adult healthy cerebral cortex and in brain lesions in MS patients, CX3CL1 is expressed at the highest level in neurons and at lower levels

Pr

in OL lineage and endothelial cells (Fig. 3A, C) [19, 22]. In healthy adult spinal cord, CX3CL1 expression is restricted to neurons [135]. Interestingly, astrocytes have little to no CX3CL1 expression in a normal CNS (Fig. 3A) [19], but show strong expression in

na l

response to inflammatory stimuli, such as TNF- (tumor necrosis factor alpha) stimulation or EAE disease [134, 136, 137]. This is corroborated by moderate expression level of CX3CL1 in human astrocytes in MS patient brain lesions (Fig. 3C) [22]. Finally, CX3CL1 is expressed in the choroid plexus in aging and EAE-induced mice [138, 139].

ur

CX3CR1 is the only murine receptor for CX3CL1 (Fig. 3D) [10]. While CX3CR1

has previously been believed to be expressed exclusively in microglia [140], recent

Jo

reports suggest CX3CR1 is also expressed in neurons [141], neural precursors [16, 46, 108] and OPCs [46] in vivo and in culture, albeit at a lower level. RNA-sequencing of murine embryonic and postnatal OPCs isolated from brain and spinal cord confirms CX3CR1 expression in developing OPCs and shows the highest expression in postnatal day 7 brain OPCs (Fig. 4A) [23]. This is in agreement with elevated expression of CX3CR1 in postnatal OPCs when compared to embryonic OPCs in the developing cortex [46]. While NPCs are not included in the murine adult CNS cell types RNA-sequencing

14

analysis [19], Fig. 4B demonstrates CX3CR1 is expressed at the highest level in microglia and at a lower level in OPCs. Finally, single-nuclei RNA sequencing analysis from MS patient brain lesions also shows CX3CR1 is expressed in committed OPCs (cOPCs), immune oligodendroglia (ImOLGs) and microglia/macrophages (Fig. 4C) [22]. In culture, soluble CX3CL1 acts directly on CX3CR1 expressed in murine NPCs and OPCs and enhances their differentiation into OLs without affecting their proliferation or survival [46]. Moreover, when CX3CR1 is specifically knocked down in embryonic NPCs through in utero electroporation, this leads to decreased production of OPCs and

oo f

OLs, but not astrocytes [46]. Intriguingly, mice constitutively lacking one or two copies of CX3CR1 also show decreased oligodendoglial differentiation in postnatal brain [46],

however, this mouse model does not allow for isolating the role of CX3CR1 signalling in

pr

different cell types. What is the contribution of neuronal CX3CL1 to developmental

oligodendrogenesis? Voronova et al. show that conditioned media from interneurons

e-

enhances oligodendrogenesis from NPCs in culture, and that this effect is abolished when CX3CL1 is neutralized with anti-CX3CL1 function blocking antibodies [46].

Pr

Interneuron-mediated increase in oligodendrogenesis is also abrogated when CX3CR1 on NPCs is blocked with anti-CX3CR1 function blocking antibodies [46]. In vivo, ablation of interneurons leads to reduced expression of CX3CL1 in the developing cortex and

na l

reduced OPC numbers [46]. Perinatal lethality of these mice did not allow evaluation of the production of mature OLs [46]. Notably, enhanced production of OL lineage cells is detected in human forebrain organoids that develop from both ventrally and dorsally derived precursors [142]. In these fused organoids, there is an extensive migration of

ur

ventrally derived cells, which leads to increased OL production when compared to organoids derived only from ventral or dorsal precursors [142]. Whether these effects are

Jo

due to CX3CL1 in migrating ventral interneurons in fused organoids remains to be addressed.

Intriguingly, infusion of soluble CX3CL1 into hippocampus of aged rats increases

proliferation and neurogenic differentiation from hippocampal NPCs, whereas acute block of CX3CR1 with anti-CX3CR1 function blocking antibodies decreases NPC proliferation and differentiation [143]. Currently, it is not clear whether this is due to a direct effect of CX3CL1 on hippocampal NPCs or indirect effect through microglia.

15

What is known is that human NPCs isolated from second trimester fetal CNS tissue show increased survival in response to CX3CL1 [108]. Moreover, adult mouse SVZ NPCs exhibit higher neurosphere formation in the presence of CX3CL1, which is indicative of their increased activation and proliferation [139]. CX3CR1 constitutive knockout mice challenged with cuprizone demyelination show decreased myelin debris phagocytosis by microglia/macrophages, aberrant myelin morphology as well as reduced numbers of OPCs, which has been attributed to reduced OPC migration and proliferation [144]. It is tempting to speculate that the deficiencies in

oo f

OPC functions in this mouse model may be attributed to the lack of CX3CR1 signalling

in OPCs, which otherwise express CX3CR1 in both healthy and diseased CNS (Fig. 4B, C) [19, 22]. Mice expressing CX3CR1 with a mutation detected in MS patients

pr

(CX3CR1I249/M280 [145], Table 1) that makes CX3CR1 less responsive to CX3CL1 [146], have more severe EAE, exhibit a decrease in calbindin-positive neurons and MBP

e-

staining, as well as an increase in CD45-positive cells in the cerebellum [147]. EAE induction in CX3CR1 constitutive knockout mice also leads to more severe EAE disease

Pr

progression and demyelination as well as increased neurological deficits [148]. Interestingly, some of the increased EAE disease severity in CX3CR1 knockout mice has been attributed to bone marrow and natural killer immune cells, which also

na l

express CX3CR1 and infiltrate CNS during EAE [148, 149]. When wild-type rats are treated with CNS impenetrant CX3CR1 inhibitor AZD8797, which blocks peripheral leukocyte infiltration, this reduces disease severity and neurological impairments [150]. The difference in the effect of enhanced or decreased CX3CR1 signalling in the outcome

ur

of de- and remyelination can be attributed to i) differences in CX3CR1 signalling in the peripheral immune system vs. the CNS; or ii) acute vs. chronic depletion of CX3CR1

Jo

signalling. In support of the former, injections of soluble CX3CL1 directly into the CNS lead to neuroprotection, increased neurogenesis as well as improved behavior and cognition in various mouse models of neurodegeneration, aging, schizophrenia and stress [128, 143, 151-154]. Moreover, exogenous CX3CL1 protects CNS cells from demyelination-induced cell death in ex vivo cerebellar slice cultures [155]. In support of the latter, Cipriani et al. show that in a stroke mouse model, constitutive CX3CL1 or CX3CR1 knockout reduces ischemic damage [128]. However, injection of CX3CL1

16

directly into the lateral ventricle in wild-type, but not in CX3CL1 or CX3CR1 knockout, mice also reduces ischemic damage and increases neuroprotection [128]. Thus, the authors hypothesize that the absence of the CX3CL1-CX3CR1 signalling axis from early embryonic development alters the microenvironment in the brain. They further suggest that acute vs. chronic manipulation of the CX3CL1-CX3CR1 signalling axis may lead to different outcomes [128]. Indeed, when microglia isolated from wild-type or CX3CL1 knockout mice are subjected to conditioned medium from oxygen-glucose deprived

oo f

neuronal cultures, they elicit different phagocytic activity and TNF- release in response to exogenous CX3CL1 [128]. The response of OPCs with chronic vs. acute manipulation of the CX3CL1-CX3CR1 signalling axis during neurodegeneration remains to be addressed.

pr

While these positive effects of soluble CX3CL1 in injury, neurodegeneration,

stress and schizophrenia mouse models have been attributed to microglia for the most

e-

part, a recent report shows overexpression of soluble CX3CL1 by cells lining the lateral ventricle leads to enhanced cognition in a mouse model of Alzheimer’s disease without

Pr

affecting microglial function [156]. Thus, it is tempting to speculate that at least some of the positive effects of CX3CL1 on functional outcomes in the aforementioned studies

6. CXCL8

na l

could be attributed to the direct role of CX3CL1 on NPCs and/or OPCs.

CXCL8 (C-X-C motif chemokine 8) or interleukin-8 (IL-8) is produced by a variety of CNS cells, including microglia, astrocytes, neural precursors, OLs and neurons

ur

[157-160] (Fig. 3B). While there are very few reports on neuronal expression of CXCL8, it is known that co-culture of neurons differentiated from human embryonal carcinoma

Jo

cell line with epithelial cells stimulated with TNF- and IFN- results in robust increase in neuronal CXCL8 expression [160]. Moreover, incubation of cultured human neurons with amyloid beta (A) peptide or TNF-, but not CCL2, robustly increases secretion of CXCL8 [158]. However, it should be noted that the total amount of secreted CXCL8 by A- or TNF--stimulated neurons is lower when compared to A-stimulated human microglia or astrocytes [158]. Thus, it is possible that CXCL8 production in neurons may be induced by inflammatory conditions.

17

CXCL8 functions via two cognate receptors: CXCR1 (C-X-C chemokine motif receptor 1) and CXCR2 (C-X-C chemokine motif receptor 2) (Fig. 3D). CXCR1 is expressed in NG2-positive human precursors, whereas CXCR2 is expressed in mouse PDGFR-positive OPCs in vivo and in vitro (Fig. 4A) [80] and in mouse and human immature O4-positive OLs [70, 157]. The functional significance of CXCR2 signalling in vivo has been discussed in the CXCL1 section. Exogenous CXCL8 induces CXCR1-dependent cell death of human embryonic

oo f

stem cell (hESC)-derived NPCs, but not purified OPCs or OLs [157]. Notably, CXCL8 enhances migration of human NPCs and OPCs via CXCR1 signalling [157]. In rat ventral midbrain- -derived NPC cultures, CXCL8 increases NPC proliferation [79]. Incubation of Oli-Neu cell line cultures (mouse OPC cell line) with CXCL8 increases OPC

pr

proliferation and MBP expression, which may indicate enhanced OL differentiation [97] (Figs. 1-2). Interestingly, human fetal exposure to CXCL8 is associated with aberrant

e-

brain structure formation and increased risk of schizophrenia [161]. It is therefore tempting to speculate that aberrant brain formation and function could be in part caused

Pr

by the effect of CXCL8 on NPCs and OPCs that build the developing brain.

7. IFN--inducible chemokines CXCL9, CXCL10 and CXCL11

na l

The CXCL9 (C-X-C motif chemokine 9), CXCL10 (C-X-C motif chemokine 10), and CXCL11 (C-X-C motif chemokine 11) genes are located in close proximity in the CXCL cluster on chromosome 4q21.1 indicating that these chemokines have evolved together and could explain, at least in part, why they share the same receptor, CXCR3 (C-

ur

X-C chemokine receptor 3) (reviewed in [162]). CXCL9, also known as Monokine induced by IFN- (MIG), and CXCL11, also known as IFN--induced protein 9 (IP-9), do

Jo

not appear to be expressed in normal adult CNS cells. However, the expression of CXCL9, CXCL10 (IP-10) and CXCL11 rises in so called ‘endangered’ neurons upon CNS infection in vivo [163, 164]. Moreover, they are in general elevated during CNS inflammation [163-165]. A unifying feature of these chemokines, apart from being induced in endangered neurons, is their induction in response to IFN- (reviewed in [162]). IFN- in turn is critical in mediating antiviral and antimicrobial immunity [166] and is significantly elevated during CNS infection and inflammation [167-169]. Hence,

18

CXCL9, CXCL10 and CXCL11, together with IFN-, are often referred to as inflammatory chemokines [162, 170]. In EAE, robust upregulation of CXCL9, CXCL10 and CXCL11 expression precedes upregulation of CXCR3 expression [170], and this expression upregulation is maximal during disease peak [165, 170, 171]. The role of CXCL9, CXCL10 and CXCL11 in EAE has not been completely elucidated. In vivo administration of exogenous CXCL11 fused to immunoglobulin protein, which is particularly useful in evaluating T-cell stimulation [172], suppresses

oo f

ongoing EAE and provides a prolonged state of disease resistance [173]. For CXCL10, there is conflicting evidence in the literature. Klein et al. show CXCL10 knockout mice display increased susceptibility to EAE, however, treatment of wild-type mice with

CXCL10-specific antibodies does not affect incidence or severity of the EAE [174]. In

pr

contrast, Narumi et al. demonstrate neutralization of CXCL10 with function blocking antibodies in rats exacerbates EAE severity [175]. Finally, conditional ablation of

e-

CXCL10 in astrocytes delays clinical onset of EAE, but does not impact axonal loss [176]. Skripuletz et al. show ablation of astrocytes diminishes, but does not completely

Pr

suppress CXCL10 expression in cuprizone-demyelinated mice [177]. This suggests that other cells, such as damaged or endangered neurons in demyelinated CNS, may express CXCL10. While the precise role of CXCL9, CXCL10 and CXCL11 in de- and

na l

remyelination remains elusive, it is commonly accepted that CXCL9, CXCL10 and CXCL11 act as chemoattractants for immune cells expressing CXCR3 to infiltrate the CNS, which contributes to the neuroinflammation-associated CNS pathology [162]. CXCR3 knockout mice develop more severe chronic EAE diseases with increased

ur

demyelination and axonal damage in both spinal cord and cerebellum at least in part due to aberrant distribution and reduction of regulatory T-cells [178]. When CXCR3

Jo

knockout mice are challenged with cuprizone-induced demyelination, however, they do not develop a more severe demyelination, but display less active microglia, astrocytes and reduced expression of inflammatory cytokines, such as CXCL9, CXCL10, CCL2, IFN- and others [179]. While the numbers of OPCs and OLs in cuprizone-demyelinated CXCR3 knockout animals were not measured, remyelination was not different from wildtype mice [179]. CXCR3 antagonists prevent, delay the onset of and/or reduce the severity of EAE disease [180, 181] at least in part due to reduction in immune cell

19

infiltration into the CNS without affecting T-cell ability to respond to MOG immunization or to transfer EAE [181]. With regards to OPCs, which express CXCR3 in the brain (Fig. 4A and [182, 183]), both CXCL10 and IFN-, through induction of CXCL10, induce apoptosis of mouse and human OPCs [182, 183]. In agreement, OPCs isolated from CXCL10 knockout mice have reduced cell death and are less susceptible to apoptosis with IFN- treatment [182]. Likewise, OPCs isolated from CXCR3 knockout mice show decreased

oo f

apoptosis when treated with CXCL10 or IFN- [182]. Interestingly, CXCL1 protects OPCs from CXCL10- or IFN--induced cell death via CXCR2 receptor signalling [183] providing an interesting crosstalk between these different chemokines that could be

important to both understand and potentially treat demyelinating disorders. Moreover,

pr

astrocyte-secreted CXCL10 inhibits OL differentiation from OPCs [184]. Exogenous

CXCL9 increases OL maturation from adult mouse SVZ NPCs without affecting NPC

e-

cell survival, proliferation or migration [80]. While the role of CXCL11 is not currently known in NPCs or OPCs, CXCR7, which can bind CXCL11 and CXCL12, has been

Pr

reported to act as a scavenger receptor internalizing both CXCL11 and CXCL12 [121, 122]. Considering CXCL11 is upregulated in inflamed CNS [162, 170], and OPCs were recently reported to express immune genes and internalize myelin debris in EAE mice

na l

[21], it would be interesting to investigate the role of the CXCL11-CXCR7 axis in OPCs in diseased or injured CNS for their ability to respond to and internalize this

ur

inflammatory chemokine.

8. CCL2

CCL2 (chemokine ligand 2), also known as MCP-1 (monocyte chemoattractant

Jo

protein-1), is expressed in dorsal root ganglion neurons in various rodent models of neuropathic pain (reviewed in [185]). Furthermore, it has been suggested that CCL2 gets packaged into neuronal vesicles for the delivery into spinal cord [186, 187], where it could potentially signal to microglia expressing the cognate receptor CCR2 (C-C chemokine receptor type 2) (reviewed in [185]). In the brain, CCL2 is expressed in astrocytes and neurons in various structures, such as the cerebral cortex, hippocampus, hypothalamus, substantia nigra and cerebellum amongst others [188]. Notably, in MS

20

patient brain lesions, CCL2 is predominantly expressed in astrocytes and endothelial cells (Fig. 3C) [22]. CCR2 is expressed in cultured OPCs [189]; however, exogenous CCL2 does not affect postnatal cortical OPC survival or differentiation [189]. There was a CCL2mediated decrease on OPC proliferation, however it was not statistically significant [189]. RNA-sequencing of purified PDGFR-positive OPCs demonstrates CCR2 is preferentially expressed in spinal cord and not brain postnatal OPCs (Fig. 4A) [23]. This

oo f

could explain why cortical OPCs had minimal to no response to CCL2 [189]. In the future, it would be interesting to analyze the response of CCR2-positive spinal cord OPCs to CCL2. Interestingly, activated cortical OPCs in cuprizone-demyelinated brains express both CCL2 and CCR2 [53]. Moreover, incubation of activated OPCs with CCL2

pr

increases their migration [53]. Similarly, CCR2 is also important for NPC migration

[190]. CCR2 deficient (knockout) NPCs fail to migrate towards the inflammatory site

e-

when transplanted onto TNF- and IFN- treated hippocampal slices [190].

Pr

9. CCL3

CCL3 (C-C motif ligand 3), also known as MIP-1 (macrophage inflammatory protein-1), is expressed in cultured cortical neurons of the developing mouse brain [54].

na l

In the adult human brain, CCL3 is expressed predominantly in neurons in Alzheimer’s disease patients [191]. Interestingly, elevated CCL3 levels are found in mouse models of epilepsy [192, 193], Alzheimer’s disease [194, 195] and MS (EAE) [196], raising the

ur

possibility that this chemokine is associated with pathological mechanisms of neurodegeneration.

The cognate receptor for CCL3, CCR1 (C-C chemokine receptor type 1, Fig. 3D),

Jo

is expressed in cultured OPCs [197]. RNA-sequencing data from purified OPCs suggest CCR1 and another CCL3 receptor, CCR5 (C-C chemokine receptor type 5, Fig. 3D), are expressed in vivo at the highest level in embryonic spinal cord and postnatal brain OPCs, respectively (Fig. 4A) [23]. Stimulation of purified rat OPCs with CCL3 leads to a decrease in migration of OPCs with no effect on proliferation [197]. In agreement, exogenous CCL3 has no effect on human NPC proliferation [108]. This is in contrast to

21

CCL3-mediated chemoattraction and increased migration of CCR1- and CCR5-positive mononuclear cells from the circulating pool into the CNS [198]. Importantly, CCR1 knockout is protective against EAE disease initiation, at least in part due to reduction in monocyte infiltration [199]. On the other hand, Tran et al. have demonstrated CCL3 or CCR1 knockout mice do not differ in EAE disease initiation or severity [200]. The differences between these studies may lie in the myelin protein peptide used to immunize the animals, which may represent different stages of MS [200]. It is also possible that acute vs. chronic depletion of CCL3 signalling may have different

oo f

effects on EAE. For example, mice with constitutive knockout of CX3CL1 or its cognate receptor CX3CR1 have less severe injury in a mouse model of stroke [128]. Yet, acute infusion of exogenous CX3CL1 is neuroprotective in wild-type mice subjected to the

pr

same stroke paradigm [128]. In agreement, wild-type rodents treated acutely with CCL3specific antibodies or CCR1 antagonist BX-471 show reduced EAE disease initiation and

e-

severity, respectively [201, 202]. Finally, CCR5 knockout mice develop less severe EAE with decreased immune cell infiltration and microglia activation [203]. Interestingly,

Pr

NG2- and O4-positive oligodendroglial cells are reduced in CCR5 knockout mice with EAE most probably due to decreased demyelination [203]. The direct role of CCL3 on

10. CCL20

na l

OPCs in MS mouse models remains to be addressed.

CCL20 (C-C motif ligand 20) is also known as LARC (liver activation regulated chemokine) or MIP3A (macrophage inflammatory protein-3). CCL20 is not expressed in

ur

healthy adult CNS [204], but is elevated in vivo in hippocampal neurons after traumatic brain injury [205] and in astrocytes in EAE mice [204]. CCL20 acts through CCR6 (C-C

Jo

chemokine receptor 6, Fig. 3D). CCR6 knockout mice display more severe EAE disease progression at least in part due to decrease in splenic dendritic cells [206]. Intriguingly, CCR6 is not required by T-cells to induce EAE as shown by similar EAE disease onset when either wild-type and CCR6 knockout antigen re-stimulated T-cells were transferred to WT or CCR6 knockout mice [206]. RNA-sequencing analysis of purified murine adult CNS cells shows CCR6 is also expressed in microglia/macrophages and to a lower extent in OPCs (Fig. 4B) [19]. Moreover, postnatal spinal cord OPCs express the highest level

22

of CCR6 (Fig. 4A) [23]. While the direct role of the CCL20-CCR6 signalling axis in OPCs is currently not known, exogenous CCL20 is known to increase OL cell death in culture [205]. Furthermore, human NPCs express CCR6 and exhibit higher migration in response to CCL20 [8] raising the possibility that CCL20 may have a direct effect on OPC function as well.

11. CCL21 CCL21 (Chemokine [C-C motif] ligand 20), also known as secondary lymphoid-

oo f

tissue chemokine (SLC), is rapidly expressed and secreted from dissociated cortical

excitatory neurons in culture and hippocampal neurons in ex vivo slices when treated with neurotoxic glutamate [64]. It has also been shown that CCL21 is expressed in cortical

pr

neurons after ischemia [207]. Its vesicular secretion from neurons occurs via the trans-

Golgi apparatus and is dependent on the presence of an intact N-terminus [65]. Cognate

e-

receptors for CCL21, CCR7 (C-C motif chemokine receptor 7) and CXCR3 (Fig. 3D), are expressed in developing OPCs in vivo (Fig. 4A) [23] and in human NPCs in vitro [8]. and CXCL11” section.

Pr

The role of CXCR3 was discussed in “IFN-γ-inducible chemokines CXCL9, CXCL10

Similarly to microglia/macrophages [64], human NPCs stimulated with CCL21

na l

exhibit increased migration [8]. Interestingly, CCL21 promotes neuronal, but not astrocyte or OL differentiation of mouse SVZ NPCs [80]. The role of CCL21-CXCR3 or CCL21-CCR7 in OPCs in the developing and remyelinating CNS remains to be

ur

addressed.

12. Discussion and conclusions

Jo

While the name “chemokine” implies chemotaxis, it is clear that chemokines play

multiple roles in CNS precursor function, including proliferation, survival, differentiation and migration (Figs. 1-2). Notably, each chemokine can affect multiple cell functions. For example, CXCL1 reduces OPC death and migration, but increases proliferation and differentiation (Fig. 1). Thus, it is not surprising that chemokine receptors have been implicated in OPC and NPC function both in CNS development and regeneration.

23

The identification of a novel immune class of OL lineage cells, including OPCs, in neurodegenerative conditions [21, 22] and the dynamic expression of chemokines in so called “endangered” neurons in injured or infected CNS [3] suggest that there may be novel chemokine-mediated mechanisms of neuron-oligodendroglia communication in demyelinating conditions. Moreover, this poses a new question about the role of these immune OL lineage cells in neuroinflammation as well as CNS regeneration and remyelination. Finally, microglia and OPCs share common immune genes, including chemokine receptors, in demyelinating conditions [21, 22, 53]. Miron et al. elegantly

oo f

showed alternate state microglia/macrophages associated with anti-inflammatory

cytokine production (formerly known as M2), secrete paracrine ligands that promote OPC differentiation [208]. In the future, it will be important to address how immune

pr

ligands, including chemokines, affect microglia-OPC cell-to-cell communication.

Chemokines and their receptors are often mutated in MS patients and have been

e-

proposed to have protective, detrimental or disease attenuating function (Table 1). Notably, while chemokine ligands display mutations in non-coding regions, such as the

Pr

promoter and 3’-UTR (untranslated region), chemokine receptors mutations are primarily detected in protein coding regions (Table 1). This could indicate that mutations in chemokines can potentially affect their expression or mRNA stability ultimately resulting

na l

in the availability of the chemokine ligand, while mutations in receptors may primarily affect their function. Indeed, some of the single nucleotide polymorphisms (SNPs) in the CCL2 gene have been associated with increased levels of CCL2 protein expression [209], whereas mutations in CX3CR1 affect its ability to respond to CX3CL1 and/or activate

ur

downstream Akt signalling [146, 210]. It is tempting to speculate that mutated chemokine receptors in OPCs could contribute to MS onset and/or progression. An interesting

Jo

observation that OPCs express functional chemokine receptors and other immune genes [21, 53], as well as their ability to perform myelin debris phagocytosis [21] and present antigens to cytotoxic T-cells [211], have led Jakel et al. to propose that OL lineage cells could actively participate in the “inside-out” mechanism of neurodegeneration in MS [22]. Chemokine signalling network members are also mutated in neurodevelopmental disorders, such as Autism Spectrum Disorder (ASD) and Schizophrenia (Table 2).

24

Mutations in chemokine ligands and receptors and their signalling pathways are enriched in analysis of schizophrenia risk genes [212]. Intriguingly, OPCs have been recently shown to directly contribute to cognition through BDNF-TrkB signalling [27]. Moreover, hippocampal NPCs have long been known to participate in behavioural and cognitive processes through postnatal and adult neurogenesis [213]. Finally, ASD-risk genes and environmental factors are known to play an important role in cortical NPCs, ultimately impacting neural circuitry development [15, 214-217]. Could chemokine signalling in OPCs and/or NPCs contribute to these physiological processes? Moreover, could mutated

oo f

chemokine ligands and receptors in OPCs and/or NPCs contribute to the development of

neurodevelopmental disorders? This is a particularly interesting hypothesis in light of the fact that CX3CR1 constitutive knockout mice have reduced sociability in juveniles and

pr

adults, as well as increased self-grooming in response to stress in adults. This is

accompanied by reduced functional connectivity in the prefrontal cortex and aberrant

e-

synapse formation [218]. Moreover, infusion or overexpression of CX3CL1 in the CNS leads to behavioural deficit rescue in the BDNFVal66Met mouse model [153] and cognitive

Pr

improvement in the tauopathy mouse model [156]. It is tempting to speculate that chemokine signalling in CNS precursor cells, such as the CX3CL1-CX3CR1 signalling axis, may play an important role in cognition and/or behaviour. While this question is

na l

important to address in the field of neurodevelopment, the generated information could also be useful for understanding and treating cognitive decline and depression in patients with MS and other immune disorders [219].

ur

13. Acknowledgements.

A.W., K.G., T.F. and A.V. co-wrote the manuscript and K.G. created the figures. A.W. is

Jo

supported by the University of Alberta, Faculty of Medicine & Dentistry 75th Anniversary Award, K.G. by University of Alberta Research Initiative summer studentship and A.V. by Canada Research Chair Tier II in Neural Stem Cell Biology. This work was funded by grants to A.V. from CIHR (#161466), MS Society of Canada (#3573) and University of Alberta Hospital Foundation. We thank Dr. Jason Plemel for helpful discussions and Dr. Plemel and Jessica Li for reading the manuscript.

25

References

[6] [7]

[8]

[9]

[10]

oo f

Jo

[11]

pr

[5]

e-

[4]

Pr

[3]

na l

[2]

Nomiyama, Osada, Yoshie, Systematic classification of vertebrate chemokines based on conserved synteny and evolutionary history. Genes Cells 18 (2013) 1-16. https://doi.org/10.1111/gtc.12013. Murray, Stow, Cytokine secretion in macrophages: Snares, rabs, and membrane trafficking. Front Immunol 5 (2014) 538. https://doi.org/10.3389/fimmu.2014.00538. de Haas, van Weering, de Jong, Boddeke, Biber, Neuronal chemokines: Versatile messengers in central nervous system cell interaction. Mol Neurobiol 36 (2007) 137-151. https://doi.org/10.1007/s12035-007-0036-8. Stone, Hayward, Huang, Z, Sanchez, Mechanisms of regulation of the chemokinereceptor network. Int J Mol Sci 18 (2017). https://doi.org/10.3390/ijms18020342. Arango Duque, Descoteaux, Macrophage cytokines: Involvement in immunity and infectious diseases. Front Immunol 5 (2014) 491. https://doi.org/10.3389/fimmu.2014.00491. Lacy, Editorial: Secretion of cytokines and chemokines by innate immune cells. Front Immunol 6 (2015) 190. https://doi.org/10.3389/fimmu.2015.00190. Ambrosini, Aloisi, Chemokines and glial cells: A complex network in the central nervous system. Neurochem Res 29 (2004) 1017-1038. https://doi.org/10.1023/B:NERE.0000021246.96864.89. Pluchino, Gritti, Blezer, Amadio, Brambilla, Borsellino, et al., Human neural stem cells ameliorate autoimmune encephalomyelitis in non-human primates. Ann Neurol 66 (2009) 343-354. https://doi.org/10.1002/ana.21745. Storer, Gallagher, Fatt, Simonetta, Kaplan, Miller, Interleukin-6 regulates adult neural stem cell numbers during normal and abnormal post-natal development. Stem Cell Reports 10 (2018) 1464-1480. https://doi.org/10.1016/j.stemcr.2018.03.008. Sheridan, Murphy, Neuron-glia crosstalk in health and disease: Fractalkine and cx3cr1 take centre stage. Open Biol 3 (2014) 130181. https://doi.org/10.1098/rsob.130181. Deverman, Patterson, Cytokines and cns development. Neuron 64 (2009) 61-78. https://doi.org/10.1016/j.neuron.2009.09.002. Tran, Miller, Chemokine receptors: Signposts to brain development and disease. Nat Rev Neurosci 4 (2003) 444-455. https://doi.org/10.1038/nrn1116. Stuart, Singhal, Baune, Systematic review of the neurobiological relevance of chemokines to psychiatric disorders. Front Cell Neurosci 9 (2015) 357. https://doi.org/10.3389/fncel.2015.00357. Kim, Kim, Cui, Jang, Kim, Lee, et al., Secretome analysis of human oligodendrocytes derived from neural stem cells. PLoS One 9 (2014) e84292. https://doi.org/10.1371/journal.pone.0084292.

ur

[1]

[12] [13]

[14]

26

[20]

[21]

[22]

[23]

[24]

oo f

Jo

[25]

pr

[19]

e-

[18]

Pr

[17]

na l

[16]

Gallagher, Norman, Woodard, Yang, Gauthier-Fisher, Fujitani, et al., Transient maternal il-6 mediates long-lasting changes in neural stem cell pools by deregulating an endogenous self-renewal pathway. Cell Stem Cell 13 (2013) 564576. https://doi.org/10.1016/j.stem.2013.10.002. Ji, He, Dheen, Tay, Expression of chemokine receptors cxcr4, ccr2, ccr5 and cx3cr1 in neural progenitor cells isolated from the subventricular zone of the adult rat brain. Neurosci Lett 355 (2004) 236-240. https://doi.org/10.1016/j.neulet.2003.11.024. Omari, John, Sealfon, Raine, Cxc chemokine receptors on human oligodendrocytes: Implications for multiple sclerosis. Brain 128 (2005) 10031015. https://doi.org/10.1093/brain/awh479. Williams, Holman, Klein, Chemokines in the balance: Maintenance of homeostasis and protection at cns barriers. Front Cell Neurosci 8 (2014) 154. https://doi.org/10.3389/fncel.2014.00154. Zhang, Chen, Sloan, Bennett, Scholze, O'Keeffe, et al., An rna-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci 34 (2014) 11929-11947. https://doi.org/10.1523/jneurosci.1860-14.2014. Zhang, Sloan, Clarke, Caneda, Plaza, Blumenthal, et al., Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89 (2016) 37-53. https://doi.org/10.1016/j.neuron.2015.11.013. Falcao, van Bruggen, Marques, Meijer, Jakel, Agirre, et al., Disease-specific oligodendrocyte lineage cells arise in multiple sclerosis. Nat Med 24 (2018) 18371844. https://doi.org/10.1038/s41591-018-0236-y. Jakel, Agirre, Mendanha Falcao, van Bruggen, Lee, Knuesel, et al., Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature 566 (2019) 543-547. https://doi.org/10.1038/s41586-019-0903-2. Marques, van Bruggen, Vanichkina, Floriddia, Munguba, Varemo, et al., Transcriptional convergence of oligodendrocyte lineage progenitors during development. Dev Cell 46 (2018) 504-517.e507. https://doi.org/10.1016/j.devcel.2018.07.005. Marques, Zeisel, Codeluppi, van Bruggen, Mendanha Falcao, Xiao, et al., Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352 (2016) 1326-1329. https://doi.org/10.1126/science.aaf6463. Jang, Gould, Xu, Kim, Kim, Oligodendrocytes regulate presynaptic properties and neurotransmission through bdnf signaling in the mouse brainstem. Elife 8 (2019). https://doi.org/10.7554/eLife.42156. Xin, Mironova, Shen, Marino, Waisman, Lamers, et al., Oligodendrocytes support neuronal glutamatergic transmission via expression of glutamine synthetase. Cell Rep 27 (2019) 2262-2271.e2265. https://doi.org/10.1016/j.celrep.2019.04.094. Geraghty, Gibson, Ghanem, Greene, Ocampo, Goldstein, et al., Loss of adaptive myelination contributes to methotrexate chemotherapy-related cognitive

ur

[15]

[26]

[27]

27

[33]

[34]

[35]

[36]

[37]

oo f

Jo

[38]

pr

[32]

e-

[31]

Pr

[30]

na l

[29]

ur

[28]

impairment. Neuron 103 (2019) 250-265. https://doi.org/10.1016/j.neuron.2019.04.032. Green, Gelfand, Cree, Bevan, Boscardin, Mei, et al., Clemastine fumarate as a remyelinating therapy for multiple sclerosis (rebuild): A randomised, controlled, double-blind, crossover trial. Lancet 390 (2017) 2481-2489. https://doi.org/10.1016/s0140-6736(17)32346-2. Barak, Zhang, Liu, Nir, Trangle, Ennis, et al., Neuronal deletion of gtf2i, associated with williams syndrome, causes behavioral and myelin alterations rescuable by a remyelinating drug. Nat Neurosci 22 (2019) 700-708. https://doi.org/10.1038/s41593-019-0380-9. Liu, Dupree, Gacias, Frawley, Sikder, Naik, et al., Clemastine enhances myelination in the prefrontal cortex and rescues behavioral changes in socially isolated mice. J Neurosci 36 (2016) 957-962. https://doi.org/10.1523/jneurosci.3608-15.2016. Stedehouder, Brizee, Shpak, Kushner, Activity-dependent myelination of parvalbumin interneurons mediated by axonal morphological plasticity. J Neurosci 38 (2018) 3631-3642. https://doi.org/10.1523/jneurosci.007418.2018. Gibson, Purger, Mount, Goldstein, Lin, Wood, et al., Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science 344 (2014) 1252304. https://doi.org/10.1126/science.1252304. Ortiz, Habermacher, Graciarena, Houry, Nishiyama, Oumesmar, et al., Neuronal activity in vivo enhances functional myelin repair. JCI Insight 5 (2019). https://doi.org/10.1172/jci.insight.123434. Mitew, Gobius, Fenlon, McDougall, Hawkes, Xing, et al., Pharmacogenetic stimulation of neuronal activity increases myelination in an axon-specific manner. Nat Commun 9 (2018) 306. https://doi.org/10.1038/s41467-017-02719-2. Bengtsson, Nagy, Skare, Forsman, Forssberg, Ullen, Extensive piano practicing has regionally specific effects on white matter development. Nat Neurosci 8 (2005) 1148-1150. https://doi.org/10.1038/nn1516. Scholz, Klein, Behrens, Johansen-Berg, Training induces changes in white-matter architecture. Nat Neurosci 12 (2009) 1370-1371. https://doi.org/10.1038/nn.2412. Stedehouder, Kushner, Myelination of parvalbumin interneurons: A parsimonious locus of pathophysiological convergence in schizophrenia. Mol Psychiatry 22 (2017) 4-12. https://doi.org/10.1038/mp.2016.147. Goldman, Kuypers, How to make an oligodendrocyte. Development 142 (2015) 3983-3995. https://doi.org/10.1242/dev.126409. Yuzwa, Borrett, Innes, Voronova, Ketela, Kaplan, et al., Developmental emergence of adult neural stem cells as revealed by single-cell transcriptional profiling. Cell Rep 21 (2017) 3970-3986. https://doi.org/10.1016/j.celrep.2017.12.017. Fuentealba, Rompani, Parraguez, Obernier, Romero, Cepko, et al., Embryonic origin of postnatal neural stem cells. Cell 161 (2015) 1644-1655. https://doi.org/10.1016/j.cell.2015.05.041.

[39]

[40]

28

[47]

[48]

[49]

[50]

[51]

Jo

[52]

oo f

[46]

pr

[45]

e-

[44]

Pr

[43]

na l

[42]

Furutachi, Miya, Watanabe, Kawai, Yamasaki, Harada, et al., Slowly dividing neural progenitors are an embryonic origin of adult neural stem cells. Nat Neurosci 18 (2015) 657-665. https://doi.org/10.1038/nn.3989. Kessaris, Fogarty, Iannarelli, Grist, Wegner, Richardson, Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage. Nat Neurosci 9 (2006) 173-179. https://doi.org/10.1038/nn1620. Winkler, Franco, Loss of shh signaling in the neocortex reveals heterogeneous cell recovery responses from distinct oligodendrocyte populations. Dev Biol (2019). https://doi.org/10.1016/j.ydbio.2019.04.016. Menn, Garcia-Verdugo, Yaschine, Gonzalez-Perez, Rowitch, Alvarez-Buylla, Origin of oligodendrocytes in the subventricular zone of the adult brain. J Neurosci 26 (2006) 7907-7918. https://doi.org/10.1523/jneurosci.129906.2006. Maldonado, Angulo, Multiple modes of communication between neurons and oligodendrocyte precursor cells. Neuroscientist 21 (2015) 266-276. https://doi.org/10.1177/1073858414530784. Voronova, Yuzwa, Wang, Zahr, Syal, Wang, et al., Migrating interneurons secrete fractalkine to promote oligodendrocyte formation in the developing mammalian brain. Neuron 94 (2017) 1-17. https://doi.org/http://dx.doi.org/10.1016/j.neuron.2017.04.018. Venkatesh, Johung, Caretti, Noll, Tang, Nagaraja, et al., Neuronal activity promotes glioma growth through neuroligin-3 secretion. Cell 161 (2015) 803-816. https://doi.org/10.1016/j.cell.2015.04.012. Hardy, Reynolds, Neuron-oligodendroglial interactions during central nervous system development. J Neurosci Res 36 (1993) 121-126. https://doi.org/10.1002/jnr.490360202 [doi]. Bergles, Roberts, Somogyi, Jahr, Glutamatergic synapses on oligodendrocyte precursor cells in the hippocampus. Nature 405 (2000) 187-191. https://doi.org/10.1038/35012083. Orduz, Maldonado, Balia, Velez-Fort, de Sars, Yanagawa, et al., Interneurons and oligodendrocyte progenitors form a structured synaptic network in the developing neocortex. Elife 4 (2015). https://doi.org/10.7554/eLife.06953 [doi]. Balia, Benamer, Angulo, A specific gabaergic synapse onto oligodendrocyte precursors does not regulate cortical oligodendrogenesis. Glia 65 (2017) 18211832. https://doi.org/10.1002/glia.23197. Lin, Bergles, Synaptic signaling between gabaergic interneurons and oligodendrocyte precursor cells in the hippocampus. Nat Neurosci 7 (2004) 24-32. https://doi.org/10.1038/nn1162. Moyon, Dubessy, Aigrot, Trotter, Huang, Dauphinot, et al., Demyelination causes adult cns progenitors to revert to an immature state and express immune cues that support their migration. J Neurosci 35 (2015) 4-20. https://doi.org/10.1523/jneurosci.0849-14.2015. Yuzwa, Yang, Borrett, Clarke, Cancino, Zahr, et al., Proneurogenic ligands defined by modeling developing cortex growth factor communication networks. Neuron 91 (2016) 988-1004. https://doi.org/10.1016/j.neuron.2016.07.037.

ur

[41]

[53]

[54]

29

[61]

[62]

[63]

[64]

Jo

[65]

oo f

[60]

pr

[59]

e-

[58]

Pr

[57]

na l

[56]

Wang, Strong, Xie, Yang, Coyle, Wick, et al., The chemokine cxcl1/growth related oncogene increases sodium currents and neuronal excitability in small diameter sensory neurons. Mol Pain 4 (2008) 38. https://doi.org/10.1186/1744-8069-4-38. Bertollini, Ragozzino, Gross, Limatola, Eusebi, Fractalkine/cx3cl1 depresses central synaptic transmission in mouse hippocampal slices. Neuropharmacology 51 (2006) 816-821. https://doi.org/10.1016/j.neuropharm.2006.05.027. Kodangattil, Möddel, Müller, Weber, Gorji, The inflammatory chemokine cxcl10 modulates synaptic plasticity and neuronal activity in the hippocampus. European Journal of Inflammation 10 (2012) 311-328. https://doi.org/10.1177/1721727x1201000307. Marciniak, Faivre, Dutar, Alves Pires, Demeyer, Caillierez, et al., The chemokine mip-1alpha/ccl3 impairs mouse hippocampal synaptic transmission, plasticity and memory. Sci Rep 5 (2015) 15862. https://doi.org/10.1038/srep15862. Gao, Ji, Chemokines, neuronal-glial interactions, and central processing of neuropathic pain. Pharmacol Ther 126 (2010) 56-68. https://doi.org/10.1016/j.pharmthera.2010.01.002. Guyon, Cxcl12 chemokine and its receptors as major players in the interactions between immune and nervous systems. Frontiers in Cellular Neuroscience 8 (2014). https://doi.org/10.3389/fncel.2014.00065. Bhattacharyya, Banisadr, Jung, Ren, Cronshaw, Zou, et al., The chemokine stromal cell-derived factor-1 regulates gabaergic inputs to neural progenitors in the postnatal dentate gyrus. J Neurosci 28 (2008) 6720-6730. https://doi.org/10.1523/jneurosci.1677-08.2008. Reaux-Le Goazigo, Rivat, Kitabgi, Pohl, Melik Parsadaniantz, Cellular and subcellular localization of cxcl12 and cxcr4 in rat nociceptive structures: Physiological relevance. Eur J Neurosci 36 (2012) 2619-2631. https://doi.org/10.1111/j.1460-9568.2012.08179.x. Jung, Toth, White, Miller, Monocyte chemoattractant protein-1 functions as a neuromodulator in dorsal root ganglia neurons. J Neurochem 104 (2008) 254-263. https://doi.org/10.1111/j.1471-4159.2007.04969.x. de Jong, Dijkstra, Hensens, Brouwer, van Amerongen, Liem, et al., Vesiclemediated transport and release of ccl21 in endangered neurons: A possible explanation for microglia activation remote from a primary lesion. J Neurosci 25 (2005) 7548-7557. https://doi.org/10.1523/jneurosci.1019-05.2005. de Jong, Vinet, Stanulovic, Meijer, Wesseling, Sjollema, et al., Expression, transport, and axonal sorting of neuronal ccl21 in large dense-core vesicles. Faseb j 22 (2008) 4136-4145. https://doi.org/10.1096/fj.07-101907. Wake, Ortiz, Woo, Lee, Angulo, Fields, Nonsynaptic junctions on myelinating glia promote preferential myelination of electrically active axons. Nat Commun 6 (2015) 7844. https://doi.org/10.1038/ncomms8844. Mensch, Baraban, Almeida, Czopka, Ausborn, El Manira, et al., Synaptic vesicle release regulates myelin sheath number of individual oligodendrocytes in vivo. Nat Neurosci 18 (2015) 628-630. https://doi.org/10.1038/nn.3991.

ur

[55]

[66]

[67]

30

[73]

[74]

[75]

[76]

[77]

oo f

Jo

[78]

pr

[72]

e-

[71]

Pr

[70]

na l

[69]

Hines, Ravanelli, Schwindt, Scott, Appel, Neuronal activity biases axon selection for myelination in vivo. Nat Neurosci 18 (2015) 683-689. https://doi.org/10.1038/nn.3992. Robinson, Tani, Strieter, Ransohoff, Miller, The chemokine growth-regulated oncogene-alpha promotes spinal cord oligodendrocyte precursor proliferation. J Neurosci 18 (1998) 10457-10463. https://doi.org/https://doi.org/10.1523/JNEUROSCI.18-24-10457.1998 Tsai, Frost, To, Robinson, Ffrench-Constant, Geertman, et al., The chemokine receptor cxcr2 controls positioning of oligodendrocyte precursors in developing spinal cord by arresting their migration. Cell 110 (2002) 373-383. https://doi.org/https://doi.org/10.1016/S0092-8674(02)00838-3. Johnson, Dao, Guignet, Geddes, Koemeter-Cox, Kan, Increased expression of the chemokines cxcl1 and mip-1alpha by resident brain cells precedes neutrophil infiltration in the brain following prolonged soman-induced status epilepticus in rats. J Neuroinflammation 8 (2011) 41. https://doi.org/10.1186/1742-2094-841. Meng, Oka, Takashima, Developmental expression of monocyte chemoattractant protein-1 in the human cerebellum and brainstem. Brain Dev 21 (1999) 30-35. https://doi.org/10.1016/S0387-7604(98)00065-5. Vora, Pillai, Mustapha, Kowal, Shaffer, Bose, et al., Cxcl1 regulation of oligodendrocyte progenitor cell migration is independent of calcium signaling. Exp Neurol 236 (2012) 259-267. https://doi.org/10.1016/j.expneurol.2012.04.012. Gordon, McGregor, Connor, Chemokines direct neural progenitor cell migration following striatal cell loss. Mol Cell Neurosci 41 (2009) 219-232. https://doi.org/10.1016/j.mcn.2009.03.001. Wu, Miller, Ransohoff, Robinson, Bu, Nishiyama, Elevated levels of the chemokine gro-1 correlate with elevated oligodendrocyte progenitor proliferation in the jimpy mutant. J Neurosci 20 (2000) 2609-2617. https://doi.org/0.1523/JNEUROSCI.20-07-02609.2000 Karim, Kim, Lapato, Yasui, Katzenellenbogen, Tiwari-Woodruff, Increase in chemokine cxcl1 by erbeta ligand treatment is a key mediator in promoting axon myelination. Proc Natl Acad Sci U S A 115 (2018) 6291-6296. https://doi.org/10.1073/pnas.1721732115. Omari, John, Lango, Raine, Role for cxcr2 and cxcl1 on glia in multiple sclerosis. Glia 53 (2006) 24-31. https://doi.org/10.1002/glia.20246. Filipovic, Zecevic, The effect of cxcl1 on human fetal oligodendrocyte progenitor cells. Glia 56 (2008) 1-15. https://doi.org/10.1002/glia.20582. Edman, Mira, Erices, Malmersjo, Andersson, Uhlen, et al., Alpha-chemokines regulate proliferation, neurogenesis, and dopaminergic differentiation of ventral midbrain precursors and neurospheres. Stem Cells 26 (2008) 1891-1900. https://doi.org/10.1634/stemcells.2007-0753. Turbic, Leong, Turnley, Chemokines and inflammatory mediators interact to regulate adult murine neural precursor cell proliferation, survival and differentiation. PLoS One 6 (2011) e25406. https://doi.org/10.1371/journal.pone.0025406.

ur

[68]

[79]

[80]

31

[87]

[88]

[89]

[90]

Jo

[91]

oo f

[86]

pr

[85]

e-

[84]

Pr

[83]

na l

[82]

Padovani-Claudio, Liu, Ransohoff, Miller, Alterations in the oligodendrocyte lineage, myelin, and white matter in adult mice lacking the chemokine receptor cxcr2. Glia 54 (2006) 471-483. https://doi.org/10.1002/glia.20383. Omari, Lutz, Santambrogio, Lira, Raine, Neuroprotection and remyelination after autoimmune demyelination in mice that inducibly overexpress cxcl1. Am J Pathol 174 (2009) 164-176. https://doi.org/10.2353/ajpath.2009.080350. Hosking, Tirotta, Ransohoff, Lane, Cxcr2 signaling protects oligodendrocytes and restricts demyelination in a mouse model of viral-induced demyelination. PLoS One 5 (2010) e11340. https://doi.org/10.1371/journal.pone.0011340. Grist, Marro, Skinner, Syage, Worne, Doty, et al., Induced cns expression of cxcl1 augments neurologic disease in a murine model of multiple sclerosis via enhanced neutrophil recruitment. Eur J Immunol 48 (2018) 1199-1210. https://doi.org/10.1002/eji.201747442. Kerstetter, Padovani-Claudio, Bai, Miller, Inhibition of cxcr2 signaling promotes recovery in models of multiple sclerosis. Exp Neurol 220 (2009) 44-56. https://doi.org/10.1016/j.expneurol.2009.07.010. Mithal, Ren, Miller, Cxcr4 signaling regulates radial glial morphology and cell fate during embryonic spinal cord development. Glia 61 (2013) 1288-1305. https://doi.org/10.1002/glia.22515. Cheng, Wang, Zhang, Zhao, Zhou, Mu, et al., The role of sdf-1/cxcr4/cxcr7 in neuronal regeneration after cerebral ischemia. Front Neurosci 11 (2017) 590. https://doi.org/10.3389/fnins.2017.00590. Tham, Lazarini, Franceschini, Lachapelle, Amara, Dubois-Dalcq, Developmental pattern of expression of the alpha chemokine stromal cell-derived factor 1 in the rat central nervous system. Eur J Neurosci 13 (2001) 845-856. https://doi.org/ https://doi.org/10.1046/j.0953-816x.2000.01451.x. Banisadr, Skrzydelski, Kitabgi, Rostene, Parsadaniantz, Highly regionalized distribution of stromal cell-derived factor-1/cxcl12 in adult rat brain: Constitutive expression in cholinergic, dopaminergic and vasopressinergic neurons. Eur J Neurosci 18 (2003) 1593-1606. https://doi.org/10.1046/j.0953816x.2000.01451.x. Liu, Song, Guo, He, Wang, Zhu, et al., Cxcl12/cxcr4 signaling contributes to neuropathic pain via central sensitization mechanisms in a rat spinal nerve ligation model. CNS Neurosci Ther (2019). https://doi.org/10.1111/cns.13128. Krumbholz, Theil, Cepok, Hemmer, Kivisakk, Ransohoff, et al., Chemokines in multiple sclerosis: Cxcl12 and cxcl13 up-regulation is differentially linked to cns immune cell recruitment. Brain 129 (2006) 200-211. https://doi.org/10.1093/brain/awh680. McCandless, Piccio, Woerner, Schmidt, Rubin, Cross, et al., Pathological expression of cxcl12 at the blood-brain barrier correlates with severity of multiple sclerosis. Am J Pathol 172 (2008) 799-808. https://doi.org/10.2353/ajpath.2008.070918. McQuibban, Butler, Gong, Bendall, Power, Clark-Lewis, et al., Matrix metalloproteinase activity inactivates the cxc chemokine stromal cell-derived factor-1. J Biol Chem 276 (2001) 43503-43508. https://doi.org/10.1074/jbc.M107736200.

ur

[81]

[92]

[93]

32

[99]

[100]

[101]

[102]

[103]

oo f

Jo

[104]

pr

[98]

e-

[97]

Pr

[96]

na l

[95]

Crump, Gong, Loetscher, Rajarathnam, Amara, Arenzana-Seisdedos, et al., Solution structure and basis for functional activity of stromal cell-derived factor1; dissociation of cxcr4 activation from binding and inhibition of hiv-1. Embo j 16 (1997) 6996-7007. https://doi.org/10.1093/emboj/16.23.6996. Adelita, Stilhano, Han, Justo, Porcionatto, Proteolytic processed form of cxcl12 abolishes migration and induces apoptosis in neural stem cells in vitro. Stem Cell Res 22 (2017) 61-69. https://doi.org/10.1016/j.scr.2017.05.013. Stumm, Rummel, Junker, Culmsee, Pfeiffer, Krieglstein, et al., A dual role for the sdf-1/cxcr4 chemokine receptor system in adult brain: Isoform-selective regulation of sdf-1 expression modulates cxcr4-dependent neuronal plasticity and cerebral leukocyte recruitment after focal ischemia. J Neurosci 22 (2002) 58655878. https://doi.org/20026609. Kadi, Selvaraju, de Lys, Proudfoot, Wells, Boschert, Differential effects of chemokines on oligodendrocyte precursor proliferation and myelin formation in vitro. J Neuroimmunol 174 (2006) 133-146. https://doi.org/10.1016/j.jneuroim.2006.01.011. Dziembowska, Tham, Lau, Vitry, Lazarini, Dubois-Dalcq, A role for cxcr4 signaling in survival and migration of neural and oligodendrocyte precursors. Glia 50 (2005) 258-269. https://doi.org/doi:10.1002/glia.20170. Gottle, Kremer, Jander, Odemis, Engele, Hartung, et al., Activation of cxcr7 receptor promotes oligodendroglial cell maturation. Ann Neurol 68 (2010) 915924. https://doi.org/10.1002/ana.22214. Yuan, Chang, Luo, Li, Wang, Song, et al., Cxcl12 gene engineered endothelial progenitor cells further improve the functions of oligodendrocyte precursor cells. Experimental Cell Research 367 (2018) 222-231. https://doi.org/10.1016/j.yexcr.2018.03.040. Maysami, Nguyen, Zobel, Pitz, Heine, Höpfner, et al., Modulation of rat oligodendrocyte precursor cells by the chemokine cxcl12. NeuroReport 17 (2006) 1187-1190. https://doi.org/10.1097/01.wnr.0000227985.92551.9a. Banisadr, Podojil, Miller, Miller, Pattern of cxcr7 gene expression in mouse brain under normal and inflammatory conditions. J Neuroimmune Pharmacol 11 (2016) 26-35. https://doi.org/10.1007/s11481-015-9616-y. Schönemeier, Kolodziej, Schulz, Jacobs, Hoellt, Stumm, Regional and cellular localization of the cxcl12/sdf-1 chemokine receptor cxcr7 in the developing and adult rat brain. Journal of Comparative Neurology 510 (2008) 207-220. https://doi.org/doi:10.1002/cne.21780. Tian, Yin, Deng, Tang, Ren, Jiang, Cxcl12 induces migration of oligodendrocyte precursor cells through the cxcr4‑activated mek/erk and pi3k/akt pathways, Vol. 18, 2018. https://doi.org/10.3892/mmr.2018.9444. Kremer, Cui, Göttle, Kuhlmann, Hartung, Antel, et al., Cxcr7 is involved in human oligodendroglial precursor cell maturation. PLOS ONE 11 (2016) e0146503. https://doi.org/10.1371/journal.pone.0146503. Chen, Zhang, Li, Xu, Wang, Song, et al., Cxcr7 mediates neural progenitor cells migration to cxcl12 independent of cxcr4. Stem Cells 33 (2015) 2574-2585. https://doi.org/10.1002/stem.2022.

ur

[94]

[105]

[106]

33

Jo

ur

na l

Pr

e-

pr

oo f

[107] Zamproni, Mundim, Porcionatto, des Rieux, Injection of sdf-1 loaded nanoparticles following traumatic brain injury stimulates neural stem cell recruitment. Int J Pharm 519 (2017) 323-331. https://doi.org/10.1016/j.ijpharm.2017.01.036. [108] Krathwohl, Kaiser, Chemokines promote quiescence and survival of human neural progenitor cells. Stem Cells 22 (2004) 109-118. https://doi.org/10.1634/stemcells.22-1-109 [doi]. [109] Sanchez-Alcaniz, Haege, Mueller, Pla, Mackay, Schulz, et al., Cxcr7 controls neuronal migration by regulating chemokine responsiveness. Neuron 69 (2011) 77-90. https://doi.org/10.1016/j.neuron.2010.12.006. [110] Tsai, Niu, Munji, Davalos, Chang, Zhang, et al., Oligodendrocyte precursors migrate along vasculature in the developing nervous system. Science 351 (2016) 379-384. https://doi.org/10.1126/science.aad3839. [111] Niu, Tsai, Hoi, Huang, Yu, Kim, et al., Aberrant oligodendroglial-vascular interactions disrupt the blood-brain barrier, triggering cns inflammation. Nat Neurosci 22 (2019) 709-718. https://doi.org/10.1038/s41593-019-0369-4. [112] Tarnowski, Grymula, Reca, Jankowski, Maksym, Tarnowska, et al., Regulation of expression of stromal-derived factor-1 receptors: Cxcr4 and cxcr7 in human rhabdomyosarcomas. Mol Cancer Res 8 (2010) 1-14. https://doi.org/10.1158/1541-7786.Mcr-09-0259. [113] Van Rechem, Rood, Touka, Pinte, Jenal, Guerardel, et al., Scavenger chemokine (cxc motif) receptor 7 (cxcr7) is a direct target gene of hic1 (hypermethylated in cancer 1). J Biol Chem 284 (2009) 20927-20935. https://doi.org/10.1074/jbc.M109.022350. [114] Levoye, Balabanian, Baleux, Bachelerie, Lagane, Cxcr7 heterodimerizes with cxcr4 and regulates cxcl12-mediated g protein signaling. Blood 113 (2009) 60856093. https://doi.org/10.1182/blood-2008-12-196618. [115] Wurth, Bajetto, Harrison, Barbieri, Florio, Cxcl12 modulation of cxcr4 and cxcr7 activity in human glioblastoma stem-like cells and regulation of the tumor microenvironment. Front Cell Neurosci 8 (2014) 144. https://doi.org/10.3389/fncel.2014.00144. [116] Zilkha-Falb, Kaushansky, Kawakami, Ben-Nun, Post-cns-inflammation expression of cxcl12 promotes the endogenous myelin/neuronal repair capacity following spontaneous recovery from multiple sclerosis-like disease. Journal of neuroinflammation 13 (2016) 7-7. https://doi.org/10.1186/s12974-015-04684. [117] Patel, McCandless, Dorsey, Klein, Cxcr4 promotes differentiation of oligodendrocyte progenitors and remyelination. Proc Natl Acad Sci U S A 107 (2010) 11062-11067. https://doi.org/10.1073/pnas.1006301107. [118] Carbajal, Miranda, Tsukamoto, Lane, Cxcr4 signaling regulates remyelination by endogenous oligodendrocyte progenitor cells in a viral model of demyelination. Glia 59 (2011) 1813-1821. https://doi.org/10.1002/glia.21225. [119] Williams, Patel, Daniels, Klein, Targeting cxcr7/ackr3 as a therapeutic strategy to promote remyelination in the adult central nervous system. J Exp Med 211 (2014) 791-799. https://doi.org/10.1084/jem.20131224.

34

Jo

ur

na l

Pr

e-

pr

oo f

[120] Cruz-Orengo, Chen, Kim, Dorsey, Song, Klein, Cxcr7 antagonism prevents axonal injury during experimental autoimmune encephalomyelitis as revealed by in vivo axial diffusivity. J Neuroinflammation 8 (2011) 170. https://doi.org/10.1186/1742-2094-8-170. [121] Naumann, Cameroni, Pruenster, Mahabaleshwar, Raz, Zerwes, et al., Cxcr7 functions as a scavenger for cxcl12 and cxcl11. PLoS One 5 (2010) e9175. https://doi.org/10.1371/journal.pone.0009175. [122] Boldajipour, Mahabaleshwar, Kardash, Reichman-Fried, Blaser, Minina, et al., Control of chemokine-guided cell migration by ligand sequestration. Cell 132 (2008) 463-473. https://doi.org/10.1016/j.cell.2007.12.034. [123] Luo, Chu, Zhang, Xia, Chen, Fractalkine/cx3cr1 is involved in the cross-talk between neuron and glia in neurological diseases. Brain Res Bull 146 (2019) 1221. https://doi.org/10.1016/j.brainresbull.2018.11.017. [124] Paolicelli, Bisht, Tremblay, Fractalkine regulation of microglial physiology and consequences on the brain and behavior. Front Cell Neurosci 8 (2014) 129. https://doi.org/10.3389/fncel.2014.00129. [125] Kim, Vallon-Eberhard, Zigmond, Farache, Shezen, Shakhar, et al., In vivo structure/function and expression analysis of the cx3c chemokine fractalkine. Blood 118 (2011) e156-167. https://doi.org/10.1182/blood-2011-04-348946. [126] Yoneda, Imai, Nishimura, Miyaji, Mimori, Okazaki, et al., Membrane-bound form of fractalkine induces ifn-gamma production by nk cells. Eur J Immunol 33 (2003) 53-58. https://doi.org/10.1002/immu.200390007. [127] Nash, Moran, Finneran, Hudson, Robinson, Morgan, et al., Fractalkine over expression suppresses alpha-synuclein-mediated neurodegeneration. Mol Ther 23 (2015) 17-23. https://doi.org/10.1038/mt.2014.175. [128] Cipriani, Villa, Chece, Lauro, Paladini, Micotti, et al., Cx3cl1 is neuroprotective in permanent focal cerebral ischemia in rodents. J Neurosci 31 (2011) 1632716335. https://doi.org/10.1523/jneurosci.3611-11.2011. [129] Ettle, Schlachetzki, Winkler, Oligodendroglia and myelin in neurodegenerative diseases: More than just bystanders? , Mol Neurobiol 53 (2016) 3046-3062. https://doi.org/10.1007/s12035-015-9205-3. [130] McTigue, Tripathi, The life, death, and replacement of oligodendrocytes in the adult cns. J Neurochem 107 (2008) 1-19. https://doi.org/10.1111/j.14714159.2008.05570.x. [131] Dewar, Underhill, Goldberg, Oligodendrocytes and ischemic brain injury. J Cereb Blood Flow Metab 23 (2003) 263-274. https://doi.org/10.1097/01.Wcb.0000053472.41007.F9. [132] Lakhani, Hayward, Boyd, Hemispheric asymmetry in myelin after stroke is related to motor impairment and function. Neuroimage Clin 14 (2017) 344-353. https://doi.org/10.1016/j.nicl.2017.01.009. [133] Cook, Hippensteel, Shimizu, Nicolai, Fatatis, Meucci, Interactions between chemokines: Regulation of fractalkine/cx3cl1 homeostasis by sdf/cxcl12 in cortical neurons. J Biol Chem 285 (2010) 10563-10571. https://doi.org/10.1074/jbc.M109.035477. [134] Sunnemark, Eltayeb, Nilsson, Wallstrom, Lassmann, Olsson, et al., Cx3cl1 (fractalkine) and cx3cr1 expression in myelin oligodendrocyte glycoprotein-

35

[140]

[141]

[142]

oo f

Jo

[143]

pr

[139]

e-

[138]

Pr

[137]

na l

[136]

ur

[135]

induced experimental autoimmune encephalomyelitis: Kinetics and cellular origin. J Neuroinflammation 2 (2005) 17. https://doi.org/10.1186/1742-20942-17. Verge, Milligan, Maier, Watkins, Naeve, Foster, Fractalkine (cx3cl1) and fractalkine receptor (cx3cr1) distribution in spinal cord and dorsal root ganglia under basal and neuropathic pain conditions. Eur J Neurosci 20 (2004) 11501160. https://doi.org/10.1111/j.1460-9568.2004.03593.x. Maciejewski-Lenoir, Chen, Feng, Maki, Bacon, Characterization of fractalkine in rat brain cells: Migratory and activation signals for cx3cr-1-expressing microglia. J Immunol 163 (1999) 1628-1635. Zhu, Acosta, MacNeil, Cortes, Intrater, Gong, et al., Elevated expression of fractalkine (cx3cl1) and fractalkine receptor (cx3cr1) in the dorsal root ganglia and spinal cord in experimental autoimmune encephalomyelitis: Implications in multiple sclerosis-induced neuropathic pain. Biomed Res Int 2013 (2013) 480702. https://doi.org/10.1155/2013/480702. Mills, Alabanza, Mahamed, Bynoe, Extracellular adenosine signaling induces cx3cl1 expression in the brain to promote experimental autoimmune encephalomyelitis. J Neuroinflammation 9 (2012) 193. https://doi.org/10.1186/1742-2094-9-193. Silva-Vargas, Maldonado-Soto, Mizrak, Codega, Doetsch, Age-dependent niche signals from the choroid plexus regulate adult neural stem cells. Cell Stem Cell 19 (2016) 643-652. https://doi.org/S1934-5909(16)30163-1. Jung, Aliberti, Graemmel, Sunshine, Kreutzberg, Sher, et al., Analysis of fractalkine receptor cx(3)cr1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol 20 (2000) 4106-4114. https://doi.org/10.1128/MCB.20.11.4106-4114.2000. Dworzak, Renvoise, Habchi, Yates, Combadiere, Knowles, et al., Neuronal cx3cr1 deficiency protects against amyloid beta-induced neurotoxicity. PLoS One 10 (2015) e0127730. https://doi.org/10.1371/journal.pone.0127730. Kim, Xu, Padmashri, Dunaevsky, Liu, Dreyfus, et al., Pluripotent stem cellderived cerebral organoids reveal human oligodendrogenesis with dorsal and ventral origins. Stem Cell Reports 12 (2019) 890-905. https://doi.org/10.1016/j.stemcr.2019.04.011. Bachstetter, Morganti, Jernberg, Schlunk, Mitchell, Brewster, et al., Fractalkine and cx 3 cr1 regulate hippocampal neurogenesis in adult and aged rats. Neurobiol Aging 32 (2011) 2030-2044. https://doi.org/10.1016/j.neurobiolaging.2009.11.022. Lampron, Larochelle, Laflamme, Prefontaine, Plante, Sanchez, et al., Inefficient clearance of myelin debris by microglia impairs remyelinating processes. J Exp Med 212 (2015) 481-495. https://doi.org/10.1084/jem.20141656. Stojkovic, Djuric, Stankovic, Dincic, Stancic, Veljkovic, et al., The association of v249i and t280m fractalkine receptor haplotypes with disease course of multiple sclerosis. J Neuroimmunol 245 (2012) 87-92. https://doi.org/10.1016/j.jneuroim.2011.12.028. McDermott, Fong, Yang, Sechler, Cupples, Merrell, et al., Chemokine receptor mutant cx3cr1-m280 has impaired adhesive function and correlates with

[144]

[145]

[146]

36

[152]

[153]

[154]

[155]

oo f

Jo

[156]

pr

[151]

e-

[150]

Pr

[149]

na l

[148]

ur

[147]

protection from cardiovascular disease in humans. J Clin Invest 111 (2003) 12411250. https://doi.org/10.1172/jci16790. Cardona, Kim, Church, Torres, Cleary, Mendiola, et al., Role of the fractalkine receptor in cns autoimmune inflammation: New approach utilizing a mouse model expressing the human cx3cr1(i249/m280) variant. Front Cell Neurosci 12 (2018) 365. https://doi.org/10.3389/fncel.2018.00365. Garcia, Pino, Mizutani, Cardona, Charo, Ransohoff, et al., Regulation of adaptive immunity by the fractalkine receptor during autoimmune inflammation. J Immunol 191 (2013) 1063-1072. https://doi.org/10.4049/jimmunol.1300040. Huang, Shi, Jung, Pien, Wang, Salazar-Mather, et al., The neuronal chemokine cx3cl1/fractalkine selectively recruits nk cells that modify experimental autoimmune encephalomyelitis within the central nervous system. Faseb j 20 (2006) 896-905. https://doi.org/10.1096/fj.05-5465com. Ridderstad Wollberg, Ericsson-Dahlstrand, Jureus, Ekerot, Simon, Nilsson, et al., Pharmacological inhibition of the chemokine receptor cx3cr1 attenuates disease in a chronic-relapsing rat model for multiple sclerosis. Proc Natl Acad Sci U S A 111 (2014) 5409-5414. https://doi.org/10.1073/pnas.1316510111. Pabon, Bachstetter, Hudson, Gemma, Bickford, Cx3cl1 reduces neurotoxicity and microglial activation in a rat model of parkinson's disease. J Neuroinflammation 8 (2011) 9. https://doi.org/10.1186/1742-2094-8-9. Lyons, Lynch, Downer, Hanley, O'Sullivan, Smith, et al., Fractalkine-induced activation of the phosphatidylinositol-3 kinase pathway attentuates microglial activation in vivo and in vitro. J Neurochem 110 (2009) 1547-1556. https://doi.org/10.1111/j.1471-4159.2009.06253.x. Wang, Tian, Dong, Xu, Yu, Wang, et al., Transcriptome profiling analysis of the mechanisms underlying the bdnf val66met polymorphism induced dysfunctions of the central nervous system. Hippocampus 24 (2014) 65-78. https://doi.org/10.1002/hipo.22204 [doi]. Slusarczyk, Trojan, Wydra, Glombik, Chamera, Kucharczyk, et al., Beneficial impact of intracerebroventricular fractalkine administration on behavioral and biochemical changes induced by prenatal stress in adult rats: Possible role of nlrp3 inflammasome pathway. Biochem Pharmacol 113 (2016) 45-56. https://doi.org/10.1016/j.bcp.2016.05.008. O'Sullivan, Dev, The chemokine fractalkine (cx3cl1) attenuates h2o2-induced demyelination in cerebellar slices. J Neuroinflammation 14 (2017) 159. https://doi.org/10.1186/s12974-017-0932-4. Finneran, Morgan, Gordon, Nash, Cns-wide over expression of fractalkine improves cognitive functioning in a tauopathy model. J Neuroimmune Pharmacol 14 (2019) 312-325. https://doi.org/10.1007/s11481-018-9822-5. Kelland, Gilmore, Weiner, Lund, The dual role of cxcl8 in human cns stem cell function: Multipotent neural stem cell death and oligodendrocyte progenitor cell chemotaxis. Glia 59 (2011) 1864-1878. https://doi.org/doi:10.1002/glia.21230. Ashutosh, Kou, Cotter, Borgmann, Wu, Persidsky, et al., Cxcl8 protects human neurons from amyloid-β-induced neurotoxicity: Relevance to alzheimer’s disease.

[157]

[158]

37

[164]

[165]

[166] [167]

[168]

oo f

Jo

[169]

pr

[163]

e-

[162]

Pr

[161]

na l

[160]

ur

[159]

Biochemical and Biophysical Research Communications 412 (2011) 565-571. https://doi.org/10.1016/j.bbrc.2011.07.127. Bakhiet, Tjernlund, Mousa, Gad, Stromblad, Kuziel, et al., Rantes promotes growth and survival of human first-trimester forebrain astrocytes. Nat Cell Biol 3 (2001) 150-157. https://doi.org/10.1038/35055057. Tixier, Galmiche, Neunlist, Intestinal neuro-epithelial interactions modulate neuronal chemokines production. Biochem Biophys Res Commun 344 (2006) 554-561. https://doi.org/10.1016/j.bbrc.2006.03.159. Ellman, Deicken, Vinogradov, Kremen, Poole, Kern, et al., Structural brain alterations in schizophrenia following fetal exposure to the inflammatory cytokine interleukin-8. Schizophr Res 121 (2010) 46-54. https://doi.org/10.1016/j.schres.2010.05.014. Michlmayr, McKimmie, Role of cxcl10 in central nervous system inflammation. International Journal of Interferon, Cytokine and Mediator Research 6 (2014) 118. https://doi.org/10.2147/IJICMR.S35953. Campanella, Tager, El Khoury, Thomas, Abrazinski, Manice, et al., Chemokine receptor cxcr3 and its ligands cxcl9 and cxcl10 are required for the development of murine cerebral malaria. Proc Natl Acad Sci U S A 105 (2008) 4814-4819. https://doi.org/10.1073/pnas.0801544105. Klein, Lin, Zhang, Luster, Tollett, Samuel, et al., Neuronal cxcl10 directs cd8+ tcell recruitment and control of west nile virus encephalitis. J Virol 79 (2005) 11457-11466. https://doi.org/10.1128/jvi.79.17.11457-11466.2005. Ransohoff, Hamilton, Tani, Stoler, Shick, Major, et al., Astrocyte expression of mrna encoding cytokines ip-10 and je/mcp-1 in experimental autoimmune encephalomyelitis. Faseb j 7 (1993) 592-600. https://doi.org/10.1096/fasebj.7.6.8472896. Tau, Rothman, Biologic functions of the ifn-gamma receptors. Allergy 54 (1999) 1233-1251. https://doi.org/10.1034/j.1398-9995.1999.00099.x. Shapshak, Duncan, Minagar, Rodriguez de la Vega, Stewart, Goodkin, Elevated expression of ifn-gamma in the hiv-1 infected brain. Front Biosci 9 (2004) 10731081. https://doi.org/10.2741/1271. Sa, Ochiai, Tiwari, Perkins, Mullins, Gehman, et al., Cutting edge: Ifn-gamma produced by brain-resident cells is crucial to control cerebral infection with toxoplasma gondii. J Immunol 195 (2015) 796-800. https://doi.org/10.4049/jimmunol.1500814. Arellano, Ottum, Reyes, Burgos, Naves, Stage-specific role of interferon-gamma in experimental autoimmune encephalomyelitis and multiple sclerosis. Front Immunol 6 (2015) 492. https://doi.org/10.3389/fimmu.2015.00492. Borjini, Fernandez, Giardino, Calza, Cytokine and chemokine alterations in tissue, csf, and plasma in early presymptomatic phase of experimental allergic encephalomyelitis (eae), in a rat model of multiple sclerosis. J Neuroinflammation 13 (2016) 291. https://doi.org/10.1186/s12974-016-0757-6. Carter, Muller, Manders, Campbell, Induction of the genes for cxcl9 and cxcl10 is dependent on ifn-gamma but shows differential cellular expression in experimental autoimmune encephalomyelitis and by astrocytes and microglia in vitro. Glia 55 (2007) 1728-1739. https://doi.org/10.1002/glia.20587.

[170]

[171]

38

Jo

ur

na l

Pr

e-

pr

oo f

[172] Chapoval, Zhu, Chen, Immunoglobulin fusion proteins as a tool for evaluation of t-cell costimulatory molecules. Mol Biotechnol 21 (2002) 259-264. https://doi.org/10.1385/mb:21:3:259. [173] Zohar, Wildbaum, Novak, Salzman, Thelen, Alon, et al., Cxcl11-dependent induction of foxp3-negative regulatory t cells suppresses autoimmune encephalomyelitis. J Clin Invest 124 (2014) 2009-2022. https://doi.org/10.1172/jci71951. [174] Klein, Izikson, Means, Gibson, Lin, Sobel, et al., Ifn-inducible protein 10/cxc chemokine ligand 10-independent induction of experimental autoimmune encephalomyelitis. J Immunol 172 (2004) 550-559. https://doi.org/https://doi.org/10.4049/jimmunol.172.1.550 [175] Narumi, Kaburaki, Yoneyama, Iwamura, Kobayashi, Matsushima, Neutralization of ifn-inducible protein 10/cxcl10 exacerbates experimental autoimmune encephalomyelitis. Eur J Immunol 32 (2002) 1784-1791. https://doi.org/10.1002/1521-4141(200206)32:6<1784::Aidimmu1784>3.0.Co;2-r. [176] Mills Ko, Ma, Guo, Miers, Lee, Bannerman, et al., Deletion of astroglial cxcl10 delays clinical onset but does not affect progressive axon loss in a murine autoimmune multiple sclerosis model. J Neuroinflammation 11 (2014) 105. https://doi.org/10.1186/1742-2094-11-105. [177] Skripuletz, Hackstette, Bauer, Gudi, Pul, Voss, et al., Astrocytes regulate myelin clearance through recruitment of microglia during cuprizone-induced demyelination. Brain 136 (2013) 147-167. https://doi.org/10.1093/brain/aws262. [178] Muller, Carter, Hofer, Manders, Getts, Getts, et al., Cxcr3 signaling reduces the severity of experimental autoimmune encephalomyelitis by controlling the parenchymal distribution of effector and regulatory t cells in the central nervous system. J Immunol 179 (2007) 2774-2786. https://doi.org/https://doi.org/10.4049/jimmunol.179.5.2774 [179] Krauthausen, Saxe, Zimmermann, Emrich, Heneka, Muller, Cxcr3 modulates glial accumulation and activation in cuprizone-induced demyelination of the central nervous system. J Neuroinflammation 11 (2014) 109. https://doi.org/10.1186/1742-2094-11-109. [180] Jenh, Cox, Cui, Reich, Sullivan, Chen, et al., A selective and potent cxcr3 antagonist sch 546738 attenuates the development of autoimmune diseases and delays graft rejection. BMC Immunol 13 (2012) 2. https://doi.org/10.1186/1471-2172-13-2. [181] Ni, Zhu, Zhong, Ding, Hou, Tong, et al., The chemokine receptor antagonist, tak779, decreased experimental autoimmune encephalomyelitis by reducing inflammatory cell migration into the central nervous system, without affecting t cell function. Br J Pharmacol 158 (2009) 2046-2056. https://doi.org/10.1111/j.1476-5381.2009.00528.x. [182] Tirotta, Ransohoff, Lane, Cxcr2 signaling protects oligodendrocyte progenitor cells from ifn-γ/cxcl10-mediated apoptosis. Glia 59 (2011) 1518-1528. https://doi.org/doi:10.1002/glia.21195.

39

Jo

ur

na l

Pr

e-

pr

oo f

[183] Tirotta, Kirby, Hatch, Lane, Ifn-γ-induced apoptosis of human embryonic stem cell derived oligodendrocyte progenitor cells is restricted by cxcr2 signaling. Stem Cell Research 9 (2012) 208-217. https://doi.org/10.1016/j.scr.2012.06.005. [184] Moore, Cui, Warsi, Durafourt, Zorko, Owen, et al., Direct and indirect effects of immune and central nervous system-resident cells on human oligodendrocyte progenitor cell differentiation. J Immunol 194 (2015) 761-772. https://doi.org/10.4049/jimmunol.1401156. [185] Biber, Boddeke, Neuronal cc chemokines: The distinct roles of ccl21 and ccl2 in neuropathic pain. Front Cell Neurosci 8 (2014) 210. https://doi.org/10.3389/fncel.2014.00210. [186] Jung, Bhangoo, Banisadr, Freitag, Ren, White, et al., Visualization of chemokine receptor activation in transgenic mice reveals peripheral activation of ccr2 receptors in states of neuropathic pain. J Neurosci 29 (2009) 8051-8062. https://doi.org/10.1523/jneurosci.0485-09.2009. [187] Van Steenwinckel, Reaux-Le Goazigo, Pommier, Mauborgne, Dansereau, Kitabgi, et al., Ccl2 released from neuronal synaptic vesicles in the spinal cord is a major mediator of local inflammation and pain after peripheral nerve injury. J Neurosci 31 (2011) 5865-5875. https://doi.org/10.1523/jneurosci.598610.2011. [188] Banisadr, Gosselin, Mechighel, Kitabgi, Rostene, Parsadaniantz, Highly regionalized neuronal expression of monocyte chemoattractant protein-1 (mcp1/ccl2) in rat brain: Evidence for its colocalization with neurotransmitters and neuropeptides. J Comp Neurol 489 (2005) 275-292. https://doi.org/10.1002/cne.20598. [189] Gaupp, Arezzo, Dutta, John, Raine, On the occurrence of hypomyelination in a transgenic mouse model: A consequence of the myelin basic protein promoter? , J Neuropathol Exp Neurol 70 (2011) 1138-1150. https://doi.org/10.1097/NEN.0b013e31823b188b. [190] Belmadani, Tran, Ren, Miller, Chemokines regulate the migration of neural progenitors to sites of neuroinflammation. J Neurosci 26 (2006) 3182-3191. https://doi.org/10.1523/jneurosci.0156-06.2006. [191] Xia, Qin, Wu, Mackay, Hyman, Immunohistochemical study of the betachemokine receptors ccr3 and ccr5 and their ligands in normal and alzheimer's disease brains. Am J Pathol 153 (1998) 31-37. https://doi.org/10.1016/s00029440(10)65542-3. [192] Xu, Long, Tang, Zhang, Hut, Tang, Ccr3, ccr2a and macrophage inflammatory protein (mip)-1a, monocyte chemotactic protein-1 (mcp-1) in the mouse hippocampus during and after pilocarpine-induced status epilepticus (pise). Neuropathol Appl Neurobiol 35 (2009) 496-514. https://doi.org/10.1111/j.1365-2990.2009.01022.x. [193] Guzik-Kornacka, Sliwa, Plucinska, Lukasiuk, Status epilepticus evokes prolonged increase in the expression of ccl3 and ccl4 mrna and protein in the rat brain. Acta Neurobiol Exp (Wars) 71 (2011) 193-207. [194] Song, Jin, Lim, Kou, Pattanayak, Rehman, et al., Tlr4 mutation reduces microglial activation, increases abeta deposits and exacerbates cognitive deficits in a mouse

40

[200]

[201]

[202]

oo f

Jo

[203]

pr

[199]

e-

[198]

Pr

[197]

na l

[196]

ur

[195]

model of alzheimer's disease. J Neuroinflammation 8 (2011) 92. https://doi.org/10.1186/1742-2094-8-92. Garwood, Cooper, Hanger, Noble, Anti-inflammatory impact of minocycline in a mouse model of tauopathy. Front Psychiatry 1 (2010) 136. https://doi.org/10.3389/fpsyt.2010.00136. Glabinski, Tuohy, Ransohoff, Expression of chemokines rantes, mip-1alpha and gro-alpha correlates with inflammation in acute experimental autoimmune encephalomyelitis. Neuroimmunomodulation 5 (1998) 166-171. https://doi.org/10.1159/000026333. Nguyen, Höpfner, Zobel, Henke, Scherübl, Stangel, Rat oligodendroglial cell lines express a functional receptor for the chemokine ccl3 (macrophage inflammatory protein-1alpha). Neuroscience Letters 351 (2003) 71-74. https://doi.org/10.1016/j.neulet.2003.07.014. Zang, Samanta, Halder, Hong, Tejada-Simon, Rivera, et al., Aberrant t cell migration toward rantes and mip-1 alpha in patients with multiple sclerosis. Overexpression of chemokine receptor ccr5. Brain 123 ( Pt 9) (2000) 1874-1882. https://doi.org/10.1093/brain/123.9.1874. Rottman, Slavin, Silva, Weiner, Gerard, Hancock, Leukocyte recruitment during onset of experimental allergic encephalomyelitis is ccr1 dependent. Eur J Immunol 30 (2000) 2372-2377. https://doi.org/10.1002/15214141(2000)30:8<2372::Aid-immu2372>3.0.Co;2-d. Tran, Kuziel, Owens, Induction of experimental autoimmune encephalomyelitis in c57bl/6 mice deficient in either the chemokine macrophage inflammatory protein1alpha or its ccr5 receptor. Eur J Immunol 30 (2000) 1410-1415. https://doi.org/10.1002/(sici)1521-4141(200005)30:5<1410::Aidimmu1410>3.0.Co;2-l. Karpus, Lukacs, McRae, Strieter, Kunkel, Miller, An important role for the chemokine macrophage inflammatory protein-1 alpha in the pathogenesis of the t cell-mediated autoimmune disease, experimental autoimmune encephalomyelitis. J Immunol 155 (1995) 5003-5010. Liang, Mallari, Rosser, Ng, May, Monahan, et al., Identification and characterization of a potent, selective, and orally active antagonist of the cc chemokine receptor-1. J Biol Chem 275 (2000) 19000-19008. https://doi.org/10.1074/jbc.M001222200. Gu, Park, Yun, Han, Oh, Son, et al., Ccr5 knockout suppresses experimental autoimmune encephalomyelitis in c57bl/6 mice. Oncotarget 7 (2016) 1538215393. https://doi.org/10.18632/oncotarget.8097. Ambrosini, Columba-Cabezas, Serafini, Muscella, Aloisi, Astrocytes are the major intracerebral source of macrophage inflammatory protein-3α/ccl20 in relapsing experimental autoimmune encephalomyelitis and in vitro. Glia 41 (2003) 290-300. https://doi.org/doi:10.1002/glia.10193. Leonardo, Musso, Das, Rowe, Collier, Mohapatra, et al., Ccl20 is associated with neurodegeneration following experimental traumatic brain injury and promotes cellular toxicity in vitro. Translational Stroke Research 3 (2012) 357-363. https://doi.org/10.1007/s12975-012-0203-8.

[204]

[205]

41

Jo

ur

na l

Pr

e-

pr

oo f

[206] Elhofy, Depaolo, Lira, Lukacs, Karpus, Mice deficient for ccr6 fail to control chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 213 (2009) 91-99. https://doi.org/10.1016/j.jneuroim.2009.05.011. [207] Biber, Sauter, Brouwer, Copray, Boddeke, Ischemia-induced neuronal expression of the microglia attracting chemokine secondary lymphoid-tissue chemokine (slc). Glia 34 (2001) 121-133. https://doi.org/ https://doi.org/10.1002/glia.1047. [208] Miron, Boyd, Zhao, Yuen, Ruckh, Shadrach, et al., M2 microglia and macrophages drive oligodendrocyte differentiation during cns remyelination. Nat Neurosci 16 (2013) 1211-1218. https://doi.org/10.1038/nn.3469. [209] Navratilova, Polymorphisms in ccl2&ccl5 chemokines/chemokine receptors genes and their association with diseases. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 150 (2006) 191-204. https://doi.org/10.5507/bp.2006.028. [210] Ishizuka, Fujita, Kawabata, Kimura, Iwayama, Inada, et al., Rare genetic variants in cx3cr1 and their contribution to the increased risk of schizophrenia and autism spectrum disorders. Transl Psychiatry 7 (2017) e1184. https://doi.org/10.1038/tp.2017.173. [211] Kirby, Jin, Cardona, Smith, Martin, Wang, et al., Oligodendrocyte precursor cells present antigen and are cytotoxic targets in inflammatory demyelination. Nat Commun 10 (2019) 3887. https://doi.org/10.1038/s41467-019-11638-3. [212] Lin, Cai, Zhang, Zhang, Nogales-Cadenas, Zhang, Integrated post-gwas analysis sheds new light on the disease mechanisms of schizophrenia. Genetics 204 (2016) 1587-1600. https://doi.org/10.1534/genetics.116.187195. [213] Anacker, Hen, Adult hippocampal neurogenesis and cognitive flexibility - linking memory and mood. Nat Rev Neurosci 18 (2017) 335-346. https://doi.org/10.1038/nrn.2017.45. [214] Gallagher, Voronova, Zander, Cancino, Bramall, Krause, et al., Ankrd11 is a chromatin regulator involved in autism that is essential for neural development. Dev Cell 32 (2015) 31-42. https://doi.org/10.1016/j.devcel.2014.11.031. [215] Zander, Burns, Yang, Kaplan, Miller, Snail coordinately regulates downstream pathways to control multiple aspects of mammalian neural precursor development. J Neurosci 34 (2014) 5164-5175. https://doi.org/10.1523/JNEUROSCI.0370-14.2014. [216] Yang, Cancino, Zahr, Guskjolen, Voronova, Gallagher, et al., A glo1methylglyoxal pathway that is perturbed in maternal diabetes regulates embryonic and adult neural stem cell pools in murine offspring. Cell Rep 17 (2016) 10221036. https://doi.org/10.1016/j.celrep.2016.09.067. [217] Yang, Smibert, Kaplan, Miller, An eif4e1/4e-t complex determines the genesis of neurons from precursors by translationally repressing a proneurogenic transcription program. Neuron 84 (2014) 723-739. https://doi.org/10.1016/j.neuron.2014.10.022. [218] Zhan, Paolicelli, Sforazzini, Weinhard, Bolasco, Pagani, et al., Deficient neuronmicroglia signaling results in impaired functional brain connectivity and social behavior. Nat Neurosci 17 (2014) 400-406. https://doi.org/10.1038/nn.3641. [219] Whitehouse, Fisk, Bernstein, Berrigan, Bolton, Graff, et al., Comorbid anxiety, depression, and cognition in ms and other immune-mediated disorders. Neurology (2019). https://doi.org/10.1212/wnl.0000000000006854.

42

Jo

ur

na l

Pr

e-

pr

oo f

[220] Karrer, Bosserhoff, Weiderer, Distler, Landthaler, Szeimies, et al., The -2518 promotor polymorphism in the mcp-1 gene is associated with systemic sclerosis. J Invest Dermatol 124 (2005) 92-98. https://doi.org/10.1111/j.0022202X.2004.23512.x. [221] El Sharkawi, Ali, Hegazy, Atya, The combined effect of il-17f and ccl20 gene polymorphism in susceptibility to multiple sclerosis in egypt. Gene 685 (2019) 164-169. https://doi.org/10.1016/j.gene.2018.11.006. [222] Kamali-Sarvestani, Nikseresht, Aliparasti, Vessal, Il-8 (-251 a/t) and cxcr2 (+1208 c/t) gene polymorphisms and risk of multiple sclerosis in iranian patients. Neurosci Lett 404 (2006) 159-162. https://doi.org/10.1016/j.neulet.2006.05.033. [223] Galimberti, Scalabrini, Fenoglio, Comi, De Riz, Venturelli, et al., Cxcl10 haplotypes and multiple sclerosis: Association and correlation with clinical course. Eur J Neurol 14 (2007) 162-167. https://doi.org/10.1111/j.14681331.2006.01629.x. [224] Miyagishi, Niino, Fukazawa, Yabe, Kikuchi, Tashiro, C-c chemokine receptor 2 gene polymorphism in japanese patients with multiple sclerosis. J Neuroimmunol 145 (2003) 135-138. [225] Kantor, Bakhanashvili, Achiron, A mutated ccr5 gene may have favorable prognostic implications in ms. Neurology 61 (2003) 238-240. https://doi.org/10.1212/01.wnl.0000069921.20347.9e. [226] Barcellos, Schito, Rimmler, Vittinghoff, Shih, Lincoln, et al., Cc-chemokine receptor 5 polymorphism and age of onset in familial multiple sclerosis. Multiple sclerosis genetics group. Immunogenetics 51 (2000) 281-288. https://doi.org/10.1007/s002510050621. [227] Kallaur, Kaimen-Maciel, Morimoto, Watanabe, Georgeto, Reiche, Genetic polymorphisms associated with the development and clinical course of multiple sclerosis (review). Int J Mol Med 28 (2011) 467-479. https://doi.org/10.3892/ijmm.2011.731. [228] Arli, Irkec, Menevse, Yilmaz, Alp, Fractalkine gene receptor polymorphism in patients with multiple sclerosis. Int J Neurosci 123 (2013) 31-37. https://doi.org/10.3109/00207454.2012.723079 [doi]. [229] Dasdemir, Kucukali, Bireller, Tuzun, Cakmakoglu, Chemokine gene variants in schizophrenia. Nord J Psychiatry 70 (2016) 407-412. https://doi.org/10.3109/08039488.2016.1141981. [230] Rasmussen, Timm, Wang, Soeby, Lublin, Fenger, et al., Association between the ccr5 32-bp deletion allele and late onset of schizophrenia. Am J Psychiatry 163 (2006) 507-511. https://doi.org/10.1176/appi.ajp.163.3.507. [231] Butler, Rafi, Manzardo, High-resolution chromosome ideogram representation of currently recognized genes for autism spectrum disorders. Int J Mol Sci 16 (2015) 6464-6495. https://doi.org/10.3390/ijms16036464. [232] Piton, Gauthier, Hamdan, Lafreniere, Yang, Henrion, et al., Systematic resequencing of x-chromosome synaptic genes in autism spectrum disorder and schizophrenia. Mol Psychiatry 16 (2011) 867-880. https://doi.org/10.1038/mp.2010.54.

43

Jo

ur

na l

Pr

e-

pr

oo f

[233] Lim, Raychaudhuri, Sanders, Stevens, Sabo, MacArthur, et al., Rare complete knockouts in humans: Population distribution and significant role in autism spectrum disorders. Neuron 77 (2013) 235-242. https://doi.org/10.1016/j.neuron.2012.12.029.

44

Figure legends.

Fig. 1. Summary of direct effect of neuronal chemokines [3] on OPC survival, proliferation, differentiation and migration. A positive (enhancing) effect on OPC function is displayed with chemokines and upward arrows in blue. A negative (inhibitory) effect is displayed with chemokines and downward arrows in red. Please note CCL20 has an effect on oligodendrocyte (OLs) cell death. Please see text for details and references.

na l

Pr

e-

pr

oo f

This figure was generated using BioRender and Adobe Illustrator.

Fig. 2. Summary of direct effect of neuronal chemokines [3] on NPC survival,

ur

proliferation, differentiation and migration. A positive (enhancing) effect on NPC function is displayed with chemokines and upward arrows in blue. A negative (inhibitory)

Jo

effect is displayed with chemokines and downward arrows or lines in red. Please note CCL21 increases neurogenic differentiation (marked with an asterisk). Please see text for details and references. This figure was generated using BioRender and Adobe Illustrator.

45

oo f pr e-

Fig. 3. Expression of chemokines in CNS cell types. (A): Expression of CCL2, CCL3,

Pr

CXCL1, CXCL10, CXCL12 and CX3CL1 mRNA was extracted from bulk RNAsequencing analysis of purified cells from murine adult cerebral cortex tissue using the BrainRnaSeq.org database [19]; (B): Expression of CXCL8 mRNA was extracted from

na l

bulk RNA-sequencing analysis of purified cells from human CNS tissue using the BrainRnaSeq.org database [20]. Error bars are SEM. Please note the logarithimic y-axis scale. FKPM = Fragments Per Kilobase Million; h = human; OPC = oligodendrocyte precursor cell. (C): Expression of CCL2, CXCL1, CXCL8 and CX3CL1 mRNA from

ur

single-nuclei RNA sequencing analysis of human MS patient brain lesion tissue using the Ki.se/en/mbb/oligointernode database [22]. Data is presented as a heatmap generated in

Jo

Prism8, where purple has the lowest level of expression and red has the highest level of expression. 23 cell types and the expression levels of chemokines are presented across individual rows. Please note that other chemokines discussed in this review were below the detection limit or not expressed. ImOLGs = Immune oligodendroglia; VSMCs = vascular smooth muscle cells; Oligo = Oligodendrocyte cluster; cOPC = committed OPC. (D): Diagram of chemokine ligand – receptor cognate pairs. Chemokine ligands are listed using conventional and official gene symbols. Please see text for details.

46

oo f pr ePr na l ur

Jo

Fig. 4. Expression of chemokine receptors in CNS cell types. (A-B): Expression of CCR1-2, CCR5-7, CXCR2-4, CXCR7 and CX3CR1 mRNA was extracted from bulk RNA-sequencing analysis of purified PDGFR-positive OPCs in murine embryonic and postnatal brain and spinal cord tissue [23] (A) or purified cell types from murine adult cerebral cortex tissue [19] (B) using the Ki.se/en/mbb/oligointernode or BrainRnaSeq.org databases, respectively. Error bars are SEM. Please note the logarithmic y-axis scale in (B). CPM = Counts Per Million; FKPM = Fragments Per Kilobase Million; OPC =

47

oligodendrocyte precursor cell; E = embryonic day; P = postnatal day. N.D. = not detected. Please note that receptors that showed an expression level below log1 were considered not expressed (N.E.). (C): Expression of CXCR4 and CX3CR1 mRNA from single-nuclei RNA sequencing analysis of human MS patient brain lesion tissue using the Ki.se/en/mbb/oligointernode database [22]. Data is presented as a heatmap generated in Prism8, where purple has the lowest level of expression and red has the highest level of expression. 23 cell types and the expression level of chemokine receptors are presented across individual rows. Please note that other chemokine receptors discussed in this paper

oo f

were below the detection limit or were not expressed. ImOLGs = Immune

oligodendroglia; VSMCs = vascular smooth muscle cells; Oligo = Oligodendrocyte

Jo

ur

na l

Pr

e-

pr

cluster; cOPC = committed OPC.

48

49

na l

ur

Jo

oo f

pr

e-

Pr

50

na l

ur

Jo

oo f

pr

e-

Pr

Tables.

Table 1. Summary of mutations in neuronal chemokine ligands and cognate receptors in Multiple and Systemic Sclerosis patients. § = Non-coding single nucleotide polymorphism (SNP); # = missense mutation; * = nonsense or frameshift mutation. Coordinates (e.g. -2518) indicate the location of polymorphism as it relates to the transcriptional start site. MS = Multiple Sclerosis; SPMS = secondary progressive

oo f

MS; RRMS = relapse-remitting MS; EDSS = Expanded Disability Status Scale. Mutation

Description (note protective or susceptible)

Reference

CCL2 (MCP-1)

-2518 (A/G) § Susceptible to Systemic Sclerosis  Individuals homozygous for the rs1024611 mutant allele were found in a higher percentage in patients with Systemic Sclerosis (28%) than in controls (6%)

[209, 220]

CCL20

-786 (C/T) § rs6749704

CXCL8 (IL-8)

-251 (A/T) § rs4073

Susceptible  TT genotype is more prevalent in MS patients

[222]

CXCL10 (IP-10)

rs3921 (G/C) Protective §  Progression index was lower in MS patients carrying G/G;T/T (wild-type) rs8878 (T/C) § haplotype as compared with G/C;T/C (Polymorphis or C/C;C/C carriers ms are in 3’untranslated region)

[223]

CCR2

V64I # rs1799864

[224]

e-

pr

Gene

Jo

ur

na l

Pr

Susceptible [221]  Frequency of C/T genotype in CCL20 together with C/T genotype in IL-17F (rs76378) was significantly higher in female MS patients when compared to male MS patients  EDSS was significantly higher in MS patients carrying C/T genotype in CCL20 when compared to MS patients carrying a TT genotype in CCL20

Protective  I64 allele is less frequent in MS

51



patients than controls Not associated with clinical course or severity

CCR5

Δ32 * rs333

Susceptible and protective  MS patients carrying the Δ32 have prolonged time to develop disability and have overall slower disability progression;  Delayed onset of MS with the Δ32 mutation (conflicting reports)  Higher mortality rate in MS patients with the Δ32 mutation

CX3CR1

“V249I” = V322I = V354I # rs3732379

Susceptible and protective [145, 147, 228]  SPMS was significantly higher in V249 homozygous carriers when compared to heterozygous carriers  RRMS was significantly higher in I249 homozygous carriers when compared to heterozygous carriers  I249 homozygous carriers had higher EDSS scores when compared to heterozygous carriers  I249:T280 haplotype was significantly lower in SPMS when compared to RRMS patients  Mice expressing human CX3CR1I249:M280 have more severe EAE, but milder in comparison to CX3CR1 knockout mice

oo f

pr

e-

Jo

ur

na l

Pr

“T280M” = T312M # rs3732378

[225-227]

52

Table 2. Summary of mutations in OPC chemokine receptors in neurodevelopmental disorders, such as Autism Spectrum Disorder (ASD) and Schizophrenia. § = Non-coding single nucleotide polymorphism (SNP); # = missense mutation; * = nonsense or frameshift mutation. Coordinates indicate the location of mutated amino acid (e.g. V64I) or a location of polymorphism as it relates to the transcriptional start site. Name

Mutation

CCR2

V64I # rs1799864



I64 allele was overrepresented in Schizophrenia patients

CCR5

“Δ32” * rs333



[229, 230] Δ32 heterozygous or homozygous genotype was overrepresented in late onset Schizophrenia patients (age at first admission 40 years and older) rs1799987_A/G and G/G genotypes were overrepresented in patients with Schizophrenia CCR2WT:CCR5rs1799987_A and CCR2I64:CCR5rs1799987_A haplotypes were overrepresented in patients with Schizophrenia

V115L = V162L # rs760864060



oo f

V115L or Q25X mutation identified in ASD patients

na l

CXCR3

Pr



[229]

pr



Reference

e-

“55029” (A/G) § rs1799987

Description

[231-233]

A55T = A87T # rs750585901 M138I = M170I # rs758302878

ur

CX3CR1

Jo

Q25X * rs188959001

  

A55T and G112A rare mutations were detected in both ASD and Schizophrenia patients; M138I was detected only in schizophrenia; A55T mutant does not activate Akt signalling when ovexexpressed in HEK293 cells in the presence of CX3CL1 as compared to wild-type CX3CR1.

[210]

53