CXCR2 antagonism promotes oligodendrocyte precursor cell differentiation and enhances remyelination in a mouse model of multiple sclerosis

CXCR2 antagonism promotes oligodendrocyte precursor cell differentiation and enhances remyelination in a mouse model of multiple sclerosis

Journal Pre-proof CXCR2 antagonism promotes oligodendrocyte precursor cell differentiation and enhances remyelination in a mouse model of multiple scl...

2MB Sizes 0 Downloads 44 Views

Journal Pre-proof CXCR2 antagonism promotes oligodendrocyte precursor cell differentiation and enhances remyelination in a mouse model of multiple sclerosis

Lu Wang, Hanyu Yang, Caixia Zang, Yi Dong, Junmei Shang, Jiajing Chen, Yue Wang, Hui Liu, Zihong Zhang, Heng Xu, Xiuqi Bao, Dan Zhang PII:

S0969-9961(19)30305-5

DOI:

https://doi.org/10.1016/j.nbd.2019.104630

Reference:

YNBDI 104630

To appear in:

Neurobiology of Disease

Received date:

31 May 2019

Revised date:

30 August 2019

Accepted date:

1 October 2019

Please cite this article as: L. Wang, H. Yang, C. Zang, et al., CXCR2 antagonism promotes oligodendrocyte precursor cell differentiation and enhances remyelination in a mouse model of multiple sclerosis, Neurobiology of Disease(2019), https://doi.org/10.1016/ j.nbd.2019.104630

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.

Journal Pre-proof

CXCR2 antagonism promotes oligodendrocyte precursor cell differentiation and enhances remyelination in a mouse model of multiple sclerosis Lu Wang, Hanyu Yang, Caixia Zang, Yi Dong, Junmei Shang, Jiajing Chen, Yue Wang, Hui Liu, Zihong Zhang, Heng Xu* [email protected], Xiuqi Bao* [email protected], Dan Zhang* [email protected]

of

State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union

Corresponding authors at: State Key Laboratory of Bioactive Substrate and

-p

*

ro

Medical College, 1 Xian Nong Tan Street, Beijing 100050, China

re

Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of

Abstract

na

Beijing 100050, China.

lP

Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street,

Jo ur

Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease characterized by the autoimmune attack of oligodendrocytes, leading to demyelination and progressive functional deficits. CXC chemokine receptor 2 (CXCR2) is recently reported to orchestrate the migration, proliferation and differentiation of oligodendrocyte precursor cells (OPCs), which implies its possible involvement in the demyelinating process. Here, we used a CXCR2 antagonist, compound 2, as a tool to investigate the role of CXCR2 in demyelination and the underlying mechanism. The primary cultured oligodendrocytes and cuprizone (CPZ)-intoxicated mice were applied in the present study. The results showed that compound 2 significantly

Journal Pre-proof

promoted OPC proliferation and differentiation. In the demyelinated lesions of CPZ-intoxicated mice, vigorous OPC proliferation and myelin repair was observed after compound 2 treatment. Subsequent investigation of the underlying mechanisms identified that upon inhibition of CXCR2, compound 2 treatment upregulated Ki67, transcription factor 2 (Olig2) and Caspr expression, activated PI3K/AKT/mTOR

of

signaling, ultimately promoted OPCs differentiation and enhanced remyelination. In conclusion, our results demonstrated that CXCR2 antagonism efficiently promoted

ro

OPC differentiation and enhanced remyelination in CPZ-intoxicated mice, supporting

diseases such as MS. demyelination;

differentiation

chemokine

receptor

2;

oligodendrocytes;

na

Introduction

CXC

lP

Keywords:

re

-p

CXCR2 as a promising therapeutic target for the treatment of chronic demyelinating

Jo ur

Multiple sclerosis (MS) is a chronic autoimmune demyelination disease of the central nervous system (CNS) characterized by sensory deficits and deteriorated motor coordination. The pathological hallmarks of MS are inflammation, immunization, demyelination and axonal injury, which appear throughout the CNS and are most easily recognized in white matter[1]. Although the etiology of MS remains unclear, myelin destruction is an acknowledged consequence of disease progression that results from the progressive impairment and ultimate death of oligodendrocytes[2, 3]. Current drugs that modify the course of MS, such as interferon beta, glatiramer acetate, mitoxantrone and fingolimod, can only improve symptoms after the attack[4],

Journal Pre-proof but failed as treatments against the progressive neurological deterioration of MS[5, 6]. Oligodendrocytes are myelin-forming cells in the CNS that play critical roles in brain development, neuron function and axon survival[7, 8]. In the early stage of MS, oligodendrocytes are recruited to injured areas, where they engage axons, form myelin and contribute to remyelination[9]. However, this process frequently stops or

of

fails in lesions during the later period of MS[10]. Therefore, regulating oligodendrocyte precursor cell (OPC) proliferation and differentiation is crucial for demyelinating

ro

disorders. Drugs targeting OPC proliferation and differentiation have been identified

-p

as important therapeutic strategies and might ameliorate the devastating consequences

re

of persistent demyelination[11].

lP

CXC chemokine receptor 2 (CXCR2), which is expressed by both inflammatory

na

myeloid cells and oligodendrocytes in the CNS[12, 13], is a member of the chemokine receptor family and primarily modulates immune coordination and inflammation.

Jo ur

CXCR2 also affects the intracellular processes that modulate a number of cellular events of oligodendrocytes, including migration, proliferation, and differentiation[14-16]. In addition, recent evidence has shown that a high level of CXCR2 is present in the chronic active lesions of MS patients[17], indicating that CXCR2 might be involved in MS pathogenesis. It has been reported that inhibition of CXCR2 could reduce regional recruitment of leukocytes to the spinal cord and attenuate inflammation and immune response in experimental autoimmune encephalomyelitis (EAE) and cuprizone (CPZ) -intoxicated mice[18][19]. Despite all the above-mentioned reports, the precise mechanisms of how CXCR2 antagonism contributes to MS treatment is still

Journal Pre-proof

not fully elucidated and needs further investigation. To date, over a dozen distinct classes of small molecules were identified as potent CXCR2 antagonists and several were advanced into clinical trials mainly toward developing new therapeutics for neutrophil-mediated inflammatory diseases[20]. However, most of these studies were focused on inhibition of CXCR2 in periphery. Recently, the GSK research team reported novel biarylurea-based brain penetrant [21, 22]

of

CXCR2 antagonists for the potential treatment of CNS diseases

. Among them,

ro

compound 2 exhibited nanomolar CXCR2 activity and was able to penetrate into the

-p

brain (brain/blood ratio = 0.62)[22]. In the present study, we synthesized compound 2

re

and profiled it in a β-arrestin assay with an IC50 value of 8.5nM against CXCR2

lP

(Fig. S1). Therefore, we employed compound 2 as a CXCR2 antagonist and designed

na

a set of experiments to elaborate the role of CXCR2 on demyelination and the underlying mechanisms. We found that CXCR2 antagonism significantly improved

Jo ur

motor deficits and remyelination by promoting OPC proliferation and differentiation. Mechanistic studies illustrated that the remyelination promoted by CXCR2 antagonism was closely related to the activation of the PI3K/AKT/mTOR signaling pathway. Our results clearly showed by directly affecting myelin repair and remyelination, that CXCR2 is a novel therapeutic target for the treatment of demyelination disease such as MS. Materials and Methods 1. Reagents Compound 2 was synthesized in-house according to a published procedure

[21]

.

Journal Pre-proof

Briefly,

2-(tert-butyl)-6-chlorobenzo[d]oxazole-7-sulfonyl

chloride

successively

underwent substitution by using 2-bromopropane under base conditions, oxidation by m-CPBA

and

hydrolysis

by

concentrated

H2SO4

to

generate

6-amino-3-chloro-2-(isopropylsulfonyl)phenol, and followed by a reaction with 1,2-dichloro-3-isocyanatobenzene to afford the target compound 2 as a white solid. 1H NMR (400 MHz, CDCl3) δ 11.00 (s, 1H), 8.35 (d, J = 8.8 Hz, 1H), 8.06 (dd, J =

of

7.2, 2.6 Hz, 1H), 7.35 (s, 1H), 7.22 – 7.17 (m, 2H), 7.14 (s, 1H), 7.03 (d, J = 8.8 Hz,

ro

1H), 3.89 (dt, J = 13.7, 6.9 Hz, 1H), 1.37 (d, J = 6.8 Hz, 6H); HRMS (ESI): m/z (M +

-p

H+) calcd for C16H16O4N2Cl3S, 436.9891, found: 436.9890; Purity > 95%.

re

2. Primary oligodendrocytes culture and treatments

lP

Mixed glial cultures were prepared from the cortex of postnatal day 1 Sprague

na

Dawley rats (Hua Fu Kang Bioscience, China). The brain tissue was cleared of the meninges and blood vessels and dissociated using mechanical trituration. Then, the

Jo ur

tissue was digested with 0.05 mg/mL trypsin, pipetted into PBS, and filtered through a 70 μm filter screen. Glia were plated on poly-lysine (Sigma, USA)-coated 75 cm2 flasks and cultured in DMEM (HyClone, USA) supplemented with 20% fetal bovine serum (Gibco), 100 U/mL penicillin and 100 μg/mL streptomycin (Pulilai, China) at 37°C in a humidified 5% CO2/95% air incubator. After 9 days of mixed culture, OPCs were sandwiched between microglia and astrocytes. The flasks were incubated on a shaker for 1 h at 200 r.p.m. at 37℃ to remove microglial cells. Then, the flasks were incubated with agitation overnight (18–20 h), and the cell suspension was collected to obtain OPCs. For all in vitro experiments, only cultures with ≥ 95% purity were used.

Journal Pre-proof OPCs were plated on poly-L-ornithine (Sigma, USA)-coated 75 cm2 flasks (2*104 cells/cm2). OPCs were first grown in proliferation media consisting of DMEM, 10 ng/mL platelet-derived growth factor AA (PDGFAA, Peprotech, USA), 10 ng/mL fibroblast growth factor (bFGF, Peprotech, USA), 1% fetal bovine serum, and N2 supplement (Invitrogen, USA). After two days of OPCs culture, the proliferation

of

medium was replaced with the differentiation medium, which did not contain PDGFAA and bFGF. Additionally, OPCs were treated for 5 days with compound 2 at

ro

concentrations of 0.1 μM and 1 μM, and triiodothyronine (T3, Sigma, USA) at 10 μM

-p

was used as a positive control.

re

3. Western blots

lP

Corpus callosum tissue, hippocampus tissue and primary cultured OLs were lysed

na

in lysis buffer (Applygen, China). Samples containing 30 μg of protein per lane were separated by 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–

Jo ur

PAGE) and transferred to PVDF membranes. The membranes were blocked in 5% skim milk-TBST (20 mM Tris–HCl at pH 7.5, 500 mM NaCl, 0.1% Tween 20) for 2 h. Then, the GAPDH, PDGFRα, Caspr, Ki67 (Abclonal, China), Olig2, mammalian target of rapamycin (mTOR), p-mTOR (Abcam, USA), MBP (BD Bioscience, USA), phosphoinositide 3-kinase (PI3K), p-PI3K, AKT and p-AKT (Santa Cruz, USA) primary antibodies were added in the same milk and incubated overnight at 4°C. Next, the blots were incubated with horseradish peroxidase-conjugated secondary antibody (Abclonal, China) in TBST for 2 h at room temperature. The blots were developed with an LAS3000 chemiluminescence system (Fujifilm, Tokyo, Japan), and the

Journal Pre-proof

densities of the bands were determined using Gel-Pro Analyzer 4.0 software. 4. Immunofluorescence The cells were fixed with 4% paraformaldehyde for 30 min and penetrated with 0.1% Triton X-100 for 10 min. Then, they were incubated with blocking solution containing 3% normal goat serum for 2 h. Samples were incubated overnight at 4℃

of

with Caspr, Ki67 (Abclonal, China), CXCR2, Olig2 (Abcam, USA), MBP and A2B5 (BD Bioscience, USA) antibodies (1:50). Afterward, the cells were washed three

ro

times in PBS and incubated for 1 h with FITC- or rhodamine-labeled secondary

-p

antibodies (Abcam, USA) (1:500). The cultures were washed in PBS and mounted

re

with DAPI-Fluoromount-G (Gentihold, China) for 5 min to stain the nuclei. All

lP

washing and incubation procedures were performed at room temperature, unless

na

stated otherwise, with 24-well plates and gentle shaking. Images were collected using a Leica DM5000B microscope (Leica, USA) and Leica Applications Suite software.

Jo ur

5. Animals and treatments

Male C57/BL mice of 6-8 weeks old and weighing 22–25 g were supplied by Hua Fu Kang Bioscience. Mice were maintained in a 12-h light/dark cycle at 24°C in a room with a 60% relative humidity and received food and water ad libitum. Mice were adapted for 1 week to the conditions described above before experimentation. Mouse demyelination was induced by adding 0.2% CPZ (Sigma, USA) to the standard rodent chow for 6 weeks, with 10 mice in each group. Then, the mice were orally administered compound 2 at doses of 25, 50, and 100 mg/kg or the positive control drug cyclosporin A (CyA, Neocyspin, China) 70 mg/kg twice per day for 9

Journal Pre-proof

consecutive days without CPZ. Control and CPZ-treated mice were administered 0.5% carboxymethylcellulose (CMC)-Na. All experimental procedures were performed in accordance with the guidelines of the Beijing Municipal Ethics Committee for the care and use of laboratory animals. 6. Assessment of neurological deficits

of

Neurological deficits were evaluated by a classical 5-degree scale: score 0, mouse tail is tilted upward at an angle greater than 45 degrees above the ground; score 0.5,

ro

mouse tail is tilted upward at an angle less than 45 degrees above the ground; score 1,

-p

mouse tail is parallel to the ground; score 1.5, the root of the mouse tail is parallel to

re

the ground and the tip of the tail hangs down; and score 2, the root of the mouse tail

design. 7. Rotarod test

na

lP

hang down. All evaluations were done by a researcher blind to the experimental

Jo ur

The rotarod test, which requires mice to balance and walk on a rotating cylinder, was used to measure coordinated motor skills. The rotarod test was performed as previously described[23]. Briefly, mice were positioned on the rotarod, which then revolved at 30 rpm for up to 120 sec. The rotarod automatically recorded the time point when the animal fell off the rod, which was designated as the latency. The mice were tested three times, and the latency was recorded for each test. Between each trial, the animals rested for 1 h. All evaluations were done by a researcher blind to the experimental design. 8. Luxol fast blue (LFB) staining

Journal Pre-proof

Mice were anesthetized with pentobarbital and transcardially perfused with saline and then with cold 4% paraformaldehyde in 0.1 M phosphate buffer at pH 7.4. After the brains were fixed overnight, the brains were frozen, coronally sectioned to 8 μm thickness and mounted onto slides. The extent of the demyelination of the corpus callosum was evaluated using Luxol fast blue (LFB) staining. Briefly, silane-coated

of

slides were rehydrated in a graded series of alcohol to 95% and incubated in LFB solution (0.01%) overnight at 60°C. Thereafter, sections were differentiated in a

ro

lithium carbonate solution (0.05%). Afterward, the sections stained with LFB were

-p

observed using a light microscope (NIKON 600, Japan).

re

9. Statistical analyses

lP

Data are expressed as the mean ± S. D. and were analyzed by one-way ANOVA

statistically significant.

Jo ur

Results

na

followed by Dunnett’s post hoc test with SPSS 18.0. P < 0.05 was considered

1. Compound 2 treatment promoted primary cultured OPC differentiation. In chronic demyelinating diseases, the failure of remyelination is usually caused by a decrease in the efficiency of OPC differentiation. PDGFRα is a receptor located on the surface of OPCs in the precursor stage, making it a negative marker of remyelination[9]. A2B5 is another marker of immature oligodendrocytes. In the present experiment, the incubation of compound 2 with primary cultured OPCs remarkably decreased the expression of PDGFRα in a dose-dependent manner (Fig. 1A, B). We then employed confocal microscopy to further validate the effects of compound 2 on OPC differentiation by observing A2B5 expression. Consistent with

Journal Pre-proof the above findings, the proportion of A2B5+ cells dramatically decreased with the presence of compound 2 (Fig. 1D). MBP is a protein that expresses only in mature oligodendrocytes and is frequently used as a marker for OPC maturation. The Western blot results showed that compound 2 treatment significantly increased the expression of MBP by approximately 1.5-fold relative to the control cells (Fig. 1A, C). The

of

proportion of MBP+ cells was also increased, which was detected by immunofluorescence. Consistently, compound 2 treatment induced an increased cell

ro

body size, and denser networks of MBP+ cells compared with control cells (Fig. 1E).

-p

The promotion of OPC differentiation induced by compound 2 was equivalent to that

25]

. It promotes the expression of differentiation gene and

lP

differentiation[24,

re

of T3, an agent that was required in vitro for the normal timing of oligodendrocyte

na

myelination of primary cultured OPCs [26]. Therefore, T3 has become the most widely used positive control in oligodendrocyte differentiation research in vitro. These

Jo ur

findings suggested that CXCR2 antagonism could promote the differentiation of OPCs to oligodendrocytes.

2. Compound 2 treatment upregulated Ki67, Olig2 and Caspr expression in primary cultured oligodendrocytes. There are many proteins that reported to regulate the proliferation and differentiation of OPCs, of which Ki67[27], Olig2[9] and Caspr[28] are known as critical regulators. We therefore investigated whether these proteins contributed to OPC maturation induced by CXCR2 antagonism. As expected, treatment with compound 2 markedly upregulated Ki67, Olig2 and Caspr expression detected by Western blot and

Journal Pre-proof

immunofluorescence in primary cultured oligodendrocytes (Fig. 2). These data provided evidences that inhibition of CXCR2 activity could promote the proliferation and differentiation of OPCs, and the formation of myelin. 3. Compound 2 treatment improved motor behavior and alleviated the neurological impairment of CPZ-intoxicated mice.

of

As CXCR2 was involved in OPC proliferation and differentiation, we therefore further validated whether CXCR2 antagonism could improve the demyelination in a

ro

CPZ mouse model. Mice challenged with CPZ for 6 weeks exhibited dysfunction of

-p

motor behavior and neurological deficits (Fig. S2). Based on previous experiments[21],

re

compound 2 was intragastrical administered at 25, 50, 100mg/kg to mice for 9

lP

consecutive days. The motor behavior of the mice was measured at the 5th and 9th day

na

of compound 2 treatment. The results showed that the time of mice treated with compound 2 staying on the rod significantly increased in a dose-dependent manner at

Jo ur

the 5th day (Fig. 3A). Administration of compound 2 to mice for 9 days further alleviated the symptoms of CNS injury, as indicated by markedly improved motor behavior and neurological function (Fig. 3C, D). These in vivo data strongly suggested that CXCR2 antagonism is effective in improving the pathological changes caused by demyelination. 4. Compound 2 treatment promoted the remyelination in the corpus callosum and hippocampus of CPZ-intoxicated mice. We next investigated the remyelination properties of compound 2 in the CPZ-induced remyelination mouse model. The results showed that there was robust

Journal Pre-proof

demyelination in the corpus callosum upon CPZ challenge, detected by the staining of myelin with LFB. When CXCR2 was inhibited by compound 2 treatment, the amount of LFB-stained myelin was significantly increased in the corpus callosum (Fig. 4A, B). The remyelination effects were further evaluated in Western blot assay by analyzing the proliferation and differentiation markers, PDGFRα and MBP. As shown in Figure

of

4, PDGFRα expression in the corpus callosum and hippocampus in compound 2 treated mice was significantly reduced in a dose-dependent manner. The same results

ro

were obtained in detection of MBP expression in the corpus callosum and

-p

hippocampus. It was worth noting that the capacity of compound 2 to promote

re

remyelination was more potent than that of the positive control drug CyA, which is an

lP

immunosuppressive agent and commonly used for treatment of acute exacerbations in

na

early multiple sclerosis[29]. It has also demonstrated inhibition of demyelination in a study of demyelinated animal models

[30]

. Combined with the above data, these

Jo ur

observations indicated that CXCR2 antagonism could promote remyelination and myelin repair in the mouse model of demyelination. 5. Compound 2 treatment upregulated Ki67, Olig2 and Caspr expression in the corpus callosum and hippocampus of CPZ-intoxicated mice. We also evaluated the effects of compound 2 on the expression of Ki67, Olig2 and Caspr in CPZ-induced mouse demyelination model. Consistent with the in vitro results, mice in compound 2 group exhibited higher expression of Ki67, Olig2 and Caspr in both the corpus callosum and hippocampus (Fig. 5). Similarly, the efficacy of compound 2 on Ki67, Olig2 and Caspr expression was better than that of the positive

Journal Pre-proof

drug CyA. These results suggested that CXCR2 antagonism effectively promoted the proliferation and differentiation of OPCs in CPZ mice. 6. Compound 2 treatment affected PI3K/AKT gene transcription in CPZ-intoxicated mice. To clarify the mechanisms of CXCR2 antagonism in remyelination, we performed a

of

transcriptomics study to examine the differentially expressed genes after CPZ intoxication and compound 2 treatment. As shown in Figure 6A, red represents high

ro

expression of the gene and green represents low expression of the gene. Comparing

-p

the color of each group on the same plane could reflect the expression of the same

re

gene under different treatments. The results showed that compared to control mice,

lP

the expression of each gene was greatly impacted by CPZ challenge, which was partly

na

reversed by compound 2 treatment. The results of the heat map might shed a light on the mechanism of compound 2 on CPZ-intoxicated mice. The Venn diagram

Jo ur

illustrated that 39 genes were affected by both CPZ challenge and compound 2 treatment (Fig. 6B). The transcriptomics data were further analyzed and revealed that the PI3K/AKT signaling was one of the most affected pathways after CPZ challenge and compound 2 treatment (Fig. 6C, D). 7. Compound 2 treatment promoted remyelination by activating the PI3K/AKT/mTOR signaling pathway. We then validated the effect of compound 2 on the activity of PI3K and AKT in primary cultured oligodendrocytes and CPZ-intoxicated mice. Treatment with compound 2 significantly increased the phosphorylation of PI3K and AKT both in

Journal Pre-proof

vitro and in vivo, indicating that the PI3K/AKT signaling pathway was activated by CXCR2 antagonism (Fig 7A, B, D, E, G, H). In addition, we examined the activation of mTOR, a critical downstream signaling of AKT activation and is closely related to cell proliferation and differentiation. As shown in Figure 7 C, F and I, treatment with compound 2 significantly increased the phosphorylation of mTOR in primary cultured

of

oligodendrocytes, as well as in the corpus callosum and the hippocampus of CPZ-intoxicated mice, indicating that mTOR was activated upon inhibition of

ro

CXCR2. These findings suggested that the CXCR2 antagonism could activate the

-p

PI3K/AKT/mTOR signaling pathway. This pathway may be involved in the OPC

re

proliferation and differentiation induced by CXCR2 antagonism.

lP

Discussion

Currently, the therapeutic strategies for MS mainly focus on modifying the disease

na

course by anti-inflammatory and immune-based therapies. Due to the critical role of

Jo ur

demyelination in the pathogenesis of MS, more researchers are trying to find new treatments which lead to potential remyelination. In the present study, we found that CXCR2 antagonism could promote the proliferation and differentiation of OPCs. In addition, CXCR2 antagonism significantly improved motor behavior and neurological function in CPZ-induced demyelinated mouse model through myelin repair and remyelination. CXCR2 is a receptor of CXC chemokines and belongs to the G-protein-coupled receptor (GPCR) superfamily. Studies have shown that the expression of CXCR2 increases in the brains of rodents and humans with chronic demyelinating disease, indicating that CXCR2 is somehow related to the pathology of demyelination. It was

Journal Pre-proof

reported that mice lacking CXCR2 had impaired autoimmune system and were resistant

to

demyelination[12,31].

In

focal

lysophosphatidylcholine-induced

demyelinating lesions, the localized inhibition of CXCR2 was found to reduce the release of cytokines and enhance remyelination[16]. Although the important role that CXCR2 plays on remyelination have been revealed by several reports, current studies

of

did not fully elucidate the precise mechanisms of how CXCR2 antagonism promotes remyelination. Compound 2 is a CNS penetrant CXCR2 antagonist, making it a good

ro

tool to study the influence of CXCR2 on the demyelination in CNS. And in our

-p

experiments, we found that compound 2 can inhibit the expression of CXCR2 both in

re

vivo and in vitro (Fig. S3). Our present study gave evidences that CXCR2 antagonism

lP

by compound 2 treatment markedly promoted the maturation of primary cultured

na

OPCs to oligodendrocytes and thus enhanced myelin repair in CPZ-intoxicated mouse model, further verifying that CXCR2 antagonism contributes to remyelination. We

Jo ur

also clarified the underlying mechanism that PI3K/AKT/mTOR pathway plays a critical role in the proliferation and differentiation of OPCs following CXCR2 antagonism.

In the present study, we employed primary cultured OPCs and demyelinating mouse model to investigate the remyelination effect of CXCR2. OPCs, a subtype of glial cells in the CNS, are precursors to oligodendrocytes. They proliferate and migrate throughout the CNS during late embryonic development, and then differentiate into mature myelinating oligodendrocytes[32]. Oligodendrocytes are well known as myelin-forming cells with crucial role in facilitating the rapid conduction of

Journal Pre-proof neuronal action potentials[33] and supporting axonal survival[34]. Primary culture of OPCs provides powerful means to characterize their differentiation, properties and potential for myelin repair. Our present study showed that compound 2 treatment could effectively promote the differentiation and mature of primary cultured OPCs to oligodendrocytes. The CPZ mouse model is a well-characterized model of demyelination that closely resembles the chronic and progressive clinical form of

of

demyelination disease[35]. CPZ-challenged mice exhibit several clinical deficits, such

ro

as progressive impaired motor coordination, paralysis associated with axonal loss and

-p

demyelination[36]. CPZ could also induce oxidative/nitrative stress, causing

re

mitochondrial impairment, inflammation, oligodendrocytes death and myelin loss.

lP

Our in vivo data demonstrated that compound 2 treatment significantly ameliorated

na

the clinical symptoms and repaired nerve damage of CPZ-intoxicated mice. As CXCR2 is closely correlated with immunity and inflammation, we also examined the

Jo ur

capacity of compound 2 to suppress neuroinflammation in CPZ-intoxicated mice. We found that compound 2 treatment significantly inhibited the activation of microglia and astrocytes, indicating its potent anti-neuroinflammatory capacity (Fig. S4). Our results indicated that inhibition of CXCR2 not only promoted OPC differentiation, but also attenuated inflammation and immune response, which might further have an influence on OPC differentiation. To further validate the effect of CXCR2 antagonism on remyelination, we investigated the change of proteins including Ki67, Olig2 and Caspr both in vitro and in vivo. In MS lesions, myelin repair requires the proliferation of OPCs to provide

Journal Pre-proof

enough cells to form myelin. Ki67 is a nuclear protein associated with cellular proliferation[27]. Olig2 is a transcription factor that is well known for determining motor neuron differentiation[28, 37]. Caspr is a membrane protein found in the neuronal membrane and is mainly expressed in the paranodal section of the axon during myelination[28]. Therefore, promoting the expression of Ki67, Olig2 and Caspr is very

of

important in remyelination. The present data illustrated that compound 2 treatment upregulated the expression of these proteins both in vivo and in vitro, which ensured

ro

that CXCR2 antagonism could promote the regeneration of myelin.

-p

To elucidate how CXCR2 antagonism affected proliferation and differentiation

re

of OPCs, we performed a transcriptomic analysis in CPZ-challenged mice and found

lP

that the PI3K/AKT signaling was the most affected pathway by compound 2

na

treatment. The transcriptomic results were further validated in oligodendrocytes and CPZ-intoxicated mice. The PI3K/AKT pathway could target many signaling

Jo ur

components that control almost every aspect of physiological and pathological cellular functions. mTOR is a direct substrate of the AKT kinase that promotes protein translation, growth, angiogenesis, and metabolism. In developing oligodendrocytes, mTOR signaling is required for radial sorting of axons, lipid biosynthesis, and myelin growth, which is activated before the onset of myelination and declines as myelination starts[38]. Consistent with these studies, the present data demonstrated that mTOR activation contributed to OPC maturation upon inhibition of CXCR2. Therefore, our in vivo and in vitro investigation further validated PI3K/AKT/mTOR signaling as a functionally important intracellular signaling cascade that regulates

Journal Pre-proof

OPCs proliferation and differentiation and promotes CNS remyelination after CXCR2 antagonism[39, 40]. In conclusion, the present study demonstrated that the CXCR2 antagonism could improve the motor behavior and alleviate neurological impairment in CPZ-intoxicated mice. This protective effect may be related to its ability to promote OPC proliferation

of

and differentiation. Further mechanistic studies showed that CXCR2 antagonism promoted remyelination through the activation of PI3K/AKT/mTOR signaling

ro

pathway. Collectively, our data support CXCR2 as a promising therapeutic target for

-p

chronic demyelinating diseases by directly targeting OPC maturation.

re

Competing interests

lP

The authors declare that there are no competing interests. Ethical approval

na

All experimental procedures were performed in accordance with the guidelines

Jo ur

of the Beijing Municipal Ethics Committee for the care and use of laboratory animals. Abbreviations

MS, Multiple sclerosis; CXCR2, CXC chemokine receptor 2; OPCs, oligodendrocyte precursor cells; CPZ, cuprizone; PDGFRα, platelet-derived growth factor receptor α; MBP, myelin basic protein; Olig2, transcription factor 2; CNS, central nervous system; EAE, experimental autoimmune encephalomyelitis; LPC, lysophosphatidylcholine;

mTOR,

mammalian

target

of

rapamycin;

PI3K,

phosphoinositide 3-kinase; PDGFAA, platelet-derived growth factor AA; bFGF, fibroblast growth factor; CyA, cyclosporin A; CMC, carboxymethylcellulose; LFB, Luxol fast blue; GPCR, G-protein-coupled receptor.

Journal Pre-proof

Acknowledgements This work was supported by grants from National Sciences Foundation of China (81630097, 81773718, 21772235), CAMS Innovation Fund for Medical Sciences (No. 2016-I2M-3-011), National Major Scientific and Technological Special Project for “Significant New Drugs Development” during the Thirteen Five-year Plan Period

CAMS

The

Fundamental

Research

Funds

for

the

ro

(2018RC350002).

of

(2018ZX09711001-008-005, 2018ZX09711001-003-005, 2018ZX09711001-003-020),

Jo ur

na

lP

re

-p

Figures

Central

Universities

Journal Pre-proof Figure 1. Effects of CXCR2 antagonism on the expression of PDGFRα, A2B5 and MBP in primary cultured oligodendrocytes. Primary cultured OPCs were incubated with Compound 2 at 0.1, 1 μM or T3 at 10 μM for five consecutive days. (A)Western blot bands image of PDGFRα and MBP. (B) Statistical analysis of PDGFRα expression. (C) Statistical analysis of MBP expression. A representative immunoblot

of

from four experiments was shown. (D)Confocal analysis of A2B5 expression. (E) Confocal analysis of MBP expression. A representative image from four experiments

ro

was shown. A2B5 (green), MBP (green). DAPI (blue). Results were expressed as

Jo ur

na

lP

re

-p

mean ± SD. *P < 0.05, **P < 0.01, *** P < 0.001 vs. Control.

Jo ur

na

lP

re

-p

ro

of

Journal Pre-proof

Figure 2. CXCR2 antagonism up-regulated the expression of Ki67, Olig2 and Caspr in primary cultured oligodendrocytes. Primary cultured OPCs were incubated with Compound 2 at 0.1 and 1 μM or T3 at 10 μM for five consecutive days. (A) Western blot analysis of Ki67 expression. (B) Confocal analysis of Ki67 expression. (C) Western blot analysis of Olig2 expression. (D) Confocal analysis of Olig2 expression. (E) Western blot analysis of Caspr expression. (F) Confocal analysis of Caspr expression. A representative image from four experiments was shown. MBP (green). Ki67 (red), Olig2 (red), Caspr (red). DAPI (blue). Results were expressed as mean ±

Journal Pre-proof

lP

re

-p

ro

of

SD. *P < 0.05, **P < 0.01, *** P < 0.001 vs. Control.

na

Figure 3. CXCR2 antagonism treatment improved motor behavior and alleviated neurological impairment in MS mice. Mice were intoxicated with CPZ for 6 weeks

Jo ur

and then administrated with Compound 2 for 9 days. (A) Motor behavior of mice treated with Compound 2 for 5 days. (B) Neurological function score of mice treated with Compound 2 for 5 days. (C) Motor behavior of mice treated with Compound 2 for 9 days. (D) Neurological function score of mice treated with Compound 2 for 9 days. Results were expressed as mean ± SD. n = 10. *

###

P < 0.001 vs. Control mice.

P < 0.05, **P < 0.01, ***P < 0.001 vs. CPZ-intoxicated mice.

na

lP

re

-p

ro

of

Journal Pre-proof

Jo ur

Figure 4. CXCR2 antagonism promoted remyelination in CPZ intoxicated mice. Mice were intoxicated with CPZ for 6 weeks, then treated with Compound 2 at doses of 25, 50 and 100 mg/kg for 9 consecutive days. (A) LFB staining of corpus callosum. Top, bar=125 μm, Bottom, bar=50 μm. (B) Quantification of myelinated area percentage in corpus callosum. (C, D, E) Western blot bands and analysis of PDGFRα and MBP expression in the corpus callosum of mice. (F, G, H) Western blot bands and analysis of PDGFRα and MBP expression in the hippocampus of mice. A representative image from four experiments was shown. Results were expressed as mean ± SD. # P < 0.05,

###

P < 0.001 vs. Control mice. *P < 0.05,

CPZ-intoxicated mice.

**

P < 0.01,

***

P < 0.001 vs.

-p

ro

of

Journal Pre-proof

re

Figure 5. CXCR2 antagonism up-regulated the expression of Ki67, Olig2 and Caspr

lP

expression in corpus callosum and hippocampus of CPZ-intoxicated mice. Mice received CPZ intoxication for 6 weeks and were administrated with 25, 50 and 100

na

mg/kg compound 2 or 70mg/kg CyA for 9 consecutive days.

Jo ur

(A, B, C) Western blot assay of Ki67, Olig2, Caspr expression in corpus callosum of mice. (D, E, F) Western blot assay of Ki67, Olig2, Caspr expression in hippocampus of mice. A representative image from four experiments was shown. Results were expressed as mean ± SD. # P < 0.05, ###P < 0.001 vs. Control mice. *P < 0.05, **P < 0.01 vs. CPZ-intoxicated mice.

lP

re

-p

ro

of

Journal Pre-proof

Figure 6. Transcriptomics analysis of CPZ intoxicated mice after CXCR2 antagonism

na

treatment. Mice received CPZ intoxication for 6 weeks and then treated with

Jo ur

100mg/kg compound 2 for 9 consecutive days. (A) Heat map of control group mice, CPZ-intoxicated mice and compound 2 treated mice. (B) Venn diagram of CPZ-intoxicated mice vs. control mice and CPZ-intoxicated mice vs. Compound 2-treated mice. (C) Changed signaling pathways between CPZ-intoxicated mice and control mice. (D) Changed signaling pathways between CPZ-intoxicated mice and Compound 2-treated mice.

Jo ur

na

lP

re

-p

ro

of

Journal Pre-proof

Figure 7. CXCR2 antagonism activated PI3K/AKT/mTOR signaling pathway both in vitro and in vivo. Primary cultured OPCs were incubated with compound 2 at 0.1 and 1 μM for 5 consecutive days. (A, B, C) Western blot analysis of p-PI3K, p-AKT, p-mTOR expression. Mice were intoxicated by CPZ for 6 weeks and treated with 25, 50 and 100 mg/kg compound 2 or 70mg/kg CyA for 9 consecutive days. (D, E, F) Western blot analysis of p-PI3K, p-AKT, p-mTOR expression in corpus callosum of

Journal Pre-proof

mice. (G, H, I) Western blot analysis of p-PI3K, p-AKT, p-mTOR expression in hippocampus of mice. A representative image from four experiments was shown. Results were expressed as mean ± SD. #P < 0.05, ##P < 0.01 vs. Control mice. *P < 0.05, *** P < 0.001 vs. CPZ-intoxicated mice.

References [1]

Nylander A, Hafler DA. Multiple sclerosis. J Clin Invest. 2012. 122(4): 1180-8.

[2]

Bjartmar C, Wujek JR, Trapp BD. Axonal loss in the pathology of MS: consequences for understanding the

[3]

of

progressive phase of the disease. J Neurol Sci. 2003. 206(2): 165-71.

Mestre L, Redondo M, Carrillo-Salinas FJ, et al. PDE7 inhibitor TC3.6 ameliorates symptomatology in a

[4]

ro

model of primary progressive multiple sclerosis. Br J Pharmacol. 2015. 172(17): 4277-90. McFarland HF, Martin R. Multiple sclerosis: a complicated picture of autoimmunity. Nat Immunol. 2007.

[5]

-p

8(9): 913-9.

Trapp BD, Nave KA. Multiple sclerosis: an immune or neurodegenerative disorder. Annu Rev Neurosci.

[6]

re

2008. 31: 247-69.

Curtin F, Hartung HP. Novel therapeutic options for multiple sclerosis. Expert Rev Clin Pharmacol. 2014. 7(1): 91-104.

Li JS, Yao ZX. MicroRNAs: novel regulators of oligodendrocyte differentiation and potential therapeutic

lP

[7]

targets in demyelination-related diseases. Mol Neurobiol. 2012. 45(1): 200-12. Nave KA, Werner HB. Myelination of the nervous system: mechanisms and functions. Annu Rev Cell Dev Biol. 2014. 30: 503-33. [9]

na

[8]

Ye F, Chen Y, Hoang T, et al. HDAC1 and HDAC2 regulate oligodendrocyte differentiation by disrupting

Jo ur

the beta-catenin-TCF interaction. Nat Neurosci. 2009. 12(7): 829-38. [10]

Chari DM. Remyelination in multiple sclerosis. Int Rev Neurobiol. 2007. 79: 589-620.

[11]

Skaper SD. Oligodendrocyte precursor cells as a therapeutic target for demyelinating diseases. Prog Brain Res, 2019,245:119-144.

[12]

Liu L, Darnall L, Hu T, Choi K, Lane TE, Ransohoff RM. Myelin repair is accelerated by inactivating CXCR2 on nonhematopoietic cells. J Neurosci. 2010. 30(27): 9074-83.

[13]

Ha H, Debnath B, Neamati N. Role of the CXCL8-CXCR1/2 Axis in Cancer and Inflammatory Diseases. Theranostics. 2017. 7(6): 1543-1588.

[14]

Bajetto A, Bonavia R, Barbero S, Florio T, Schettini G. Chemokines and their receptors in the central nervous system. Front Neuroendocrinol. 2001. 22(3): 147-84.

[15]

Miller RH. Regulation of oligodendrocyte development in the vertebrate CNS. Prog Neurobiol. 2002. 67(6): 451-67.

[16]

Kerstetter AE, Padovani-Claudio DA, Bai L, Miller RH. Inhibition of CXCR2 signaling promotes recovery in models of multiple sclerosis. Exp Neurol. 2009. 220(1): 44-56.

[17]

Omari KM, John G, Lango R, Raine CS. Role for CXCR2 and CXCL1 on glia in multiple sclerosis. Glia. 2006. 53(1): 24-31.

[18]

Stoolman JS, Duncker PC, Huber AK, et al. An IFNγ/CXCL2 regulatory pathway determines lesion localization during EAE. J Neuroinflammation. 2018. 15(1): 208.

Journal Pre-proof [19]

Liu L, Belkadi A, Darnall L, et al. CXCR2-positive neutrophils are essential for cuprizone-induced demyelination: relevance to multiple sclerosis. Nat Neurosci. 2010. 13(3): 319-26.

[20]

Dwyer MP, Yu Y. CXCR2 receptor antagonists: a medicinal chemistry perspective. Curr Top Med Chem. 2014. 14(13): 1590-605.

[21]

Xu H, Lu H, Xu Z, et al. Discovery of CNS Penetrant CXCR2 Antagonists for the Potential Treatment of CNS Demyelinating Disorders. ACS Med Chem Lett. 2016. 7(4): 397-402.

[22]

Lu H, Yang T, Xu Z, et al. Discovery of Novel 1-Cyclopentenyl-3-phenylureas as Selective, Brain Penetrant, and Orally Bioavailable CXCR2 Antagonists. J Med Chem. 2018. 61(6): 2518-2532.

[23]

Bao XQ, Kong XC, Qian C, Zhang D. FLZ protects dopaminergic neuron through activating protein kinase B/mammalian target of rapamycin pathway and inhibiting RTP801 expression in Parkinson's disease models. Neuroscience. 2012. 202: 396-404.

[24]

Yusta B, Besnard F, Ortiz-Caro J, Pascual A, Aranda A, Sarliève L. Evidence for the presence of nuclear

of

3,5,3'-triiodothyronine receptors in secondary cultures of pure rat oligodendrocytes. Endocrinology. 1988. 122(5): 2278-84.

Franco PG, Silvestroff L, Soto EF, Pasquini JM. Thyroid hormones promote differentiation of

ro

[25]

oligodendrocyte progenitor cells and improve remyelination after cuprizone-induced demyelination. Exp

[26]

-p

Neurol. 2008. 212(2): 458-67.

Younes-Rapozo V, Berendonk J, Savignon T, Manhães AC, Barradas PC. Thyroid hormone deficiency

Int J Dev Neurosci. 2006. 24(7): 445-53. [27]

re

changes the distribution of oligodendrocyte/myelin markers during oligodendroglial differentiation in vitro.

Patel JR, Williams JL, Muccigrosso MM, et al. Astrocyte TNFR2 is required for CXCL12-mediated

lP

regulation of oligodendrocyte progenitor proliferation and differentiation within the adult CNS. Acta Neuropathol. 2012. 124(6): 847-60. [28]

Yang HJ, Vainshtein A, Maik-Rachline G, Peles E. G protein-coupled receptor 37 is a negative regulator of

[29]

na

oligodendrocyte differentiation and myelination. Nat Commun. 2016. 7: 10884. Ruutiainen J, Salonen R, Halonen P, Panelius M, Eskola J, Salmi A. Treatment of acute exacerbations in

[30]

Jo ur

early multiple sclerosis: cyclosporin A or prednisolone. Acta Neurol Scand. 1991. 83(1): 52-4. Mazzanti CM, Spanevello R, Ahmed M, et al. Cyclosporine A inhibits acetylcholinesterase activity in rats experimentally demyelinated with ethidium bromide. Int J Dev Neurosci. 2007. 25(4): 259-64. [31]

Semple BD, Kossmann T, Morganti-Kossmann MC. Role of chemokines in CNS health and pathology: a focus on the CCL2/CCR2 and CXCL8/CXCR2 networks. J Cereb Blood Flow Metab. 2010. 30(3): 459-73.

[32]

Pfeiffer SE, Warrington AE, Bansal R. The oligodendrocyte and its many cellular processes. Trends Cell Biol. 1993. 3(6): 191-7.

[33]

Miller G. Neuroscience. The dark side of glia. Science. 2005. 308(5723): 778-81.

[34]

Lappe-Siefke C, Goebbels S, Gravel M, et al. Disruption of Cnp1 uncouples oligodendroglial functions in axonal support and myelination. Nat Genet. 2003. 33(3): 366-74.

[35]

Zimmermann J, Emrich M, Krauthausen M, et al. IL-17A Promotes Granulocyte Infiltration, Myelin Loss, Microglia Activation, and Behavioral Deficits During Cuprizone-Induced Demyelination. Mol Neurobiol. 2018. 55(2): 946-957.

[36]

Zhen W, Liu A, Lu J, Zhang W, Tattersall D, Wang J. An Alternative Cuprizone-Induced Demyelination and Remyelination Mouse Model. ASN Neuro. 2017. 9(4): 1759091417725174.

[37]

Yu Y, Chen Y, Kim B, et al. Olig2 targets chromatin remodelers to enhancers to initiate oligodendrocyte differentiation. Cell. 2013. 152(1-2): 248-61.

[38]

Marsh DR, Flemming JM. Inhibition of CXCR1 and CXCR2 chemokine receptors attenuates acute

Journal Pre-proof inflammation, preserves gray matter and diminishes autonomic dysreflexia after spinal cord injury. Spinal Cord. 2011. 49(3): 337-44. [39]

Yu JS, Ramasamy TS, Murphy N, et al. PI3K/mTORC2 regulates TGF-β/Activin signalling by modulating Smad2/3 activity via linker phosphorylation. Nat Commun. 2015. 6: 7212.

[40]

Mammana S, Bramanti P, Mazzon E, et al. Preclinical evaluation of the PI3K/Akt/mTOR pathway in animal models of multiple sclerosis. Oncotarget. 2018. 9(9): 8263-8277.

Highlights 1 Compound 2 promotes OPCs proliferation and differentiation. 2 Compound 2 enhances remyelination in a MS mouse model.

Jo ur

na

lP

re

-p

ro

of

3 Inhibition of CXCR2 activates PI3K/AKT/mTOR signaling pathway.