• No results found

The multifaceted role(s) of astrocytes in the pathology of multiple sclerosis Kamermans, A.

N/A
N/A
Protected

Academic year: 2021

Share "The multifaceted role(s) of astrocytes in the pathology of multiple sclerosis Kamermans, A."

Copied!
19
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

The multifaceted role(s) of astrocytes in the pathology of multiple sclerosis Kamermans, A.

2020

document version

Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)

Kamermans, A. (2020). The multifaceted role(s) of astrocytes in the pathology of multiple sclerosis.

General rights

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights.

• Users may download and print one copy of any publication from the public portal for the purpose of private study or research.

• You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal ?

Take down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address:

vuresearchportal.ub@vu.nl

(2)

2

CHAPTER CHAPTER

2

(3)

THE ASTROCYTE PERIVASCULAR BARRIER RESTRICTS LEUKOCYTE INFLUX INTO THE CNS VIA CLDN4-INTEGRIN INTERACTION IN MULTIPLE SCLEROSIS

Alwin Kamermans | Bert van het Hof | Susanne van der Pol | Sandra J. van Vliet Jack van Horssen | Merel Rijnsburger* | Helga E. de Vries*

*Authors contributed equally

Department of Molecular Cell Biology and Immunology, MS center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Centers, Amsterdam, the Netherlands

(4)

GRAPHICAL ABSTRACT

ABSTRACT

Trafficking of autoreactive T-cells into the brain is a crucial event in the onset and progression of multiple sclerosis (MS). In order to invade the central nervous system (CNS), immune cells need to cross the barrier endothelial cells layers of the blood-brain barrier (BBB), Underneath the barrier endothelium lies the perivascular space, delineated by the endothelial basement membrane and a second barrier, termed the glial limitans, formed by the astroglia basement membrane and astrocytic processes that surround the brain capillaries.

Although astrocytes are known to play an important role in the control of BBB function, less is known about the involvement of the glial limitans and the role of this second barrier in leukocyte trafficking into the CNS. Here we provide evidence that under inflammatory conditions, astrocytes interact with T cells within perivascular cuffs. Using in vitro models consisting of astrocytes and T cells, we show that the tight junction molecule claudin-4, expressed on astrocytes, and CD18 integrin, expressed on T cells, are important for this interaction. Furthermore, we show that interfering with this interaction decreases the binding of T cells to astrocytes. Together these findings suggest that astrocytes are actively involved in preventing the trafficking of T cells into the brain parenchyma.

(5)

Chapter 2 INTRODUCTION

A crucial step in the onset and progression of the neuro-inflammatory disorder multiple sclerosis (MS) is the trafficking of immune cells into the brain. To enter the central nervous system (CNS), immune cells have to cross multiple barriers. First, immune cells encounter the blood-brain barrier (BBB), a physical barrier that is formed by specialized endothelial cells that express adherens junction and tight junction molecules that tightly interconnect the cells, preventing ions and small molecules from passing through [1]. During inflammation, circulating immune cells employ a physical interaction with the endothelium mediated by cell surface molecules. Consequently, immune cells tether to and roll along the vascular wall. Next, chemokines, released during a neuroinflammatory event, activate immune cell integrins, thereby causing increased affinity to firmly adhere to the cell adhesion molecules on the endothelium.

During this phase, immune cells scan the endothelial surface for a site where they can transmigrate the endothelium [2]. Once inside the perivascular space, immune cells need to cross a second barrier directly adjacent to the CNS parenchyma, the glial limitans, a barrier formed by the interconnected endfeet of astrocytes and is associated with a basement membrane [3,4]. Accumulation of leukocytes in the perivascular space in itself does not result in clinical symptoms in the mouse model for MS (experimental autoimmune encephalomyelitis (EAE)). Instead, leukocyte transmigration over the glial limitans, which leads to the infiltration of the CNS parenchyma, is responsible for the clinical symptoms [5–7]. It is not yet entirely clear which mechanisms are employed by immune cells to migrate over the glial limitans, but it is apparent that they differ from leukocyte transmigration over the endothelium. One of the proposed mechanisms is by the production of metalloproteinases (MMPs). MMP-2 and -9, produced by immune cells, can cleave a protein called dystroglycan. A protein involved in the anchoring of the astrocytic endfeet to the basement membrane of the glial limitans [8]. Disruption of this interaction impairs the glial limitans structure. In addition, MS lesions are characterized by neuroinflammation and this inflammatory micromilieu induces a reactive profile of astrocytes. A recently published study showed that cytokines interleukin 1 beta (IL1-β) and transforming growth factor beta (TGF-β) induced expression of tight junction proteins claudin-1 and claudin-4 in primary human astrocytes in vitro [9]. They provide evidence that in vitro and in vivo astrocytes induce the clustering and migration of activated T cells using claudin-4 in particular. The proposed hypothesis is that astrocytes upregulate tight junction proteins at the astrocyte-astrocyte junction to tighten the barrier, which further prevents transmigration of T cells over the glial limitans. However, the precise mechanism on how astrocytes regulate immune cell migration into the brain is not yet fully understood. Increased knowledge about the functional role of the glial limitans and astrocytic tight junction proteins can lead to a better understanding of immune cell infiltration into the CNS. Here we provide immunohistochemical evidence

(6)

that astrocytes are in direct contact to perivascular immune cells. We further show that this interaction, mediated via tight junction molecule claudin-4 expressed on astrocytic endfeet and CD18 integrin receptor expressed on (activated) T cells, is a novel mechanism by which astrocytes might control leukocyte influx into the CNS.

MATERIAL AND METHODS Immunohistochemistry

Blocks of formalin-fixed paraffin-embedded brains tissues from 8 MS patients were obtained from the VUmc MS Centrum Amsterdam and the Netherlands Brain Bank. All parties received permission to perform autopsies, for the use of tissue and for access to medical records for research purposes from the Netherlands Brain Bank. Detailed clinical data are summarized in table 1. Sections (5μm) from each block were cut with a microtome and mounted on superfrost plus slides. Sections were deparaffinized in xylene and rehydrated through graded ethanol into distilled water. Antigen retrieval was performed using citrate buffer pH 6.0 (0.01M) at 100°C for 10 minutes. Sections were incubated overnight with appropriate antibodies (see table 2) in phosphate buffered saline (PBS) supplemented with 1% bovine serum albumin (BSA) and subsequently stained with the EnVision horseradish peroxidase kit (Dako, Belgium) for 30 min at room temperature and followed by 3,3’diaminobenzidine-tetrachloridedihydrate (DAB). Between incubation steps, sections were thoroughly washed with PBS. After a short rinse in tap water the sections were incubated with haematoxylin for 1 min and intensely washed with tap water for 10 min. Finally, the sections were dehydrated with ethanol followed by xylene and mounted with Entellan (Merck, Germany). For fluorescence stainings, sections were incubated overnight with antibodies at 4oC. After washing with PBS, secondary antibodies consisting of donkey-anti- mouse Alexa Fluor 488 (1:200, Abcam, UK), rabbit-anti-goat Alexa Fluor 647 (1:200, Abcam, UK), or goat-anti-rabbit Alexa Fluor 647 (1:200, Abcam, UK) were applied for 1 hour at room temperature.

Fluorescent preparations were embedded and analysis was performed with a Leica TCS SP8 confocal laser-scanning microscope (Leica Microsystems, Germany).

TABLE 1. Clinical data of MS patients and non-neurological controls

Case Age

(years) Type of MS Gender

Post- mortem

delay (h:min)

Disease duration

(years) Cause of death

MS 1 41 PP M 07:23 14 Urosepsis and pneumonia

MS 2 41 SP F 8:25 11 Deterioration due to MS

MS 3 61 PP F 10:55 11 Sepsis

MS 4 39 RR / SP F 8:30 8 Ileus

MS 5 47 SP F 8:45 20 Legal euthanasia

MS 6 49 PP F 8:30 25 Legal euthanasia

MS 7 48 SP F 9:20 24 Pneunomia

MS 8 48 RR M 6:35 Dehydration, ileus

MS, multiple sclerosis; SP, secondary progressive MS; RR, relapse remitting MS; PP, primary progressive MS; M, male; F, female

(7)

Chapter 2

Cell culture and treatments

Human astrocytoma cells (U373) were cultured in Dulbecco’s modified Eagle’s medium (DMEM)/

F12 (Life Technologies, USA) containing 10% fetal bovine serum (FBS; Biowest, USA), and penicillin/

streptomycin (50 mg/mL; Life Technologies) in 5% CO2 at 37oC. Astrocytes were grown to semi- confluence in 24-well plates and incubated with human recombinant IL-1β (10ng/ml, PeproTech, Rocky Hill, NJ, USA) and human recombinant TGF-β1 (10 ng/ml, R&D System, Minneapolis, MN, USA) for 24 hours.

Human peripheral blood lymphocytes (PBLs) were retrieved after monocyte depletion from buffy coats of healthy donors (Sanquin Blood Bank, the Netherlands). 2.0*106 cells per mL were cultured in RPMI + 0.5% BSA + 25 mM HEPES and subsequently activated with 1 mg/mL phytohaemagglutinin (PHA-L, Sigma-Aldrich, the Netherlands) and 10 ng/mL IL-2 (MACS Milteny Biotech, Germany) for 24 hrs.

Primary rat astrocytes were obtained from mixed glial cells. Brains of 0-1 days old postnatal rats of either sex were placed in cold dissection medium consisting of HBSS (Invitrogen, USA) 1mM sodium pyruvate (Sigma-Aldrich), 0.1% w/v D-glucose (Riedel-de Haën, Germany) and 10mM HEPES (Invitrogen). After the removal of the meninges, cortices were dissociated using a digestion mix consisting of MEM (Invitrogen), 2.5mg/ml Papain (Sigma-Aldrich), 40µg/ml DNAse (Sigma- Aldrich) 240 µg/ml I-Cysteine (Fluka, Romania) for 60 minutes at 37oC. Dissociated tissue was passed through a 70 µm cell strainer and subsequently centrifuged at 500 g for 5 min. Pellet was resuspended in plating medium consisting of DMEM containing pyruvate, glucose and glutamine (Invitrogen), penicillin (100U/ml, Lonza, Switzerland) and 10% FCS (Invitrogen). Cells were cultured for 7 to 10 days. Microglia were removed from the mixed glial culture using an orbital shaker at 230 rpm for 3 hrs.

Retroviral-induced overexpression of claudin-4

U373 cells stably overexpressing claudin-4 and empty vector control were generated by retroviral transduction (as described before, [10]). Expression vector encoding for human claudin-4 (pLenti- GIII-CMV-GFP-2A-Puro) was obtained from Applied Biological Materials (ABM, USA). HEK293FT

TABLE 2. Antibody details

Antigen Species Dilution Manufacturer Cat.

number Application

α-tubulin Mouse 1:1000 Cedarlane CLT9002 WB

CD3 Rabbit 1:200 Dako A0452 IHC / ICC

Claudin-4 Mouse 1:200 ThermoFisher 32-9400 IHC / ICC / WB

Fibronectin Rabbit 1:250 Dako Q0149 ICC

GFAP-cy3 Mouse 1:300 Sigma C9205 IHC / ICC

HLA-DR Mouse 1:1000 eBioscience 14-9956-82 IHC

Laminin Rabbit 1:500 Novus NB300-144 ICC

PLP Mouse 1:3000 Rio-rad MCA839G IHC

HLA-DR, human leukocyte antigen-DR; GFAP, glial fibrillary acidic protein; PLP, proteolipid protein; CD3 cluster of differentiation 3; IHC, immunohistochemistry, ICC;

immunocytochemistry; WB, westernblot

(8)

cells were cultured in DMEM containing 10% FCS, 1% penicillin/streptomycin (50 mg/mL; Life Technologies) in 5% CO2 at 37oC. HEK293FT cells were transfected with calcium phosphate as a transfection reagent. Medium was refreshed 6 hr post transfection. Supernatant containing virus was collected and virus was concentrated by centrifugation and stored at -80oC. For transduction of U373 cells, virus-containing supernatant was added dropwise to U373 cells in a 6 wells multiwell plate. Virus supernatant was replaced with appropriate medium after 24 hr incubation.

Transduced cells were selected using puromycin (2 µg/ml).

RNA isolation and real-time quantitative PCR

RNA was isolated using Trizol (Invitrogen) according to manufacturer’s protocol. mRNA concentration and quality (OD 280/260 ratios of 1.8 or higher) was measured using Nanodrop (Nanodrop Technologies, USA). cDNA syntheses was performed using the Reverse Transcription System kit (Applied Biosystems, USA) following manufacture’s guidelines. Expression was assessed by quantitative RT-PCR using SYBR Green Power mix (Applied Biosystems). All primer sequences are listed in table 3. qPCR reaction was performed using the Step-one (Applied Bioscience) Real- Time PCR System with the following program: 2 minutes at 50°C, 10 minutes at 95°C and then 40 cycles of 15 seconds at 95°C and 1 minute at 60°C. mRNA expression levels were quantified using the 2-ΔΔCT method as described in [11].

Westernblot

Cells were grown to semi-confluence in a 6 wells plate and lysed using RIPA buffer (150mM NaCl, 1% NP-40, 0.5% sodium doxycholate, 0.1% SDS, 25mM Tris, 1× phosphostop and 1× protease inhibitor (Roche, the Netherlands)). Protein concentrations were measured with BCA assay according to manufacturer’s protocol. A total of 10-15 µg protein was loaded onto SDS-PAGE gels (10%) along with a pre-stained protein marker (Precision plus, Bio-Rad Laboratories, the Netherlands). Proteins were subsequently transferred onto a nitrocellulose membrane (Bio-Rad, USA, pore size 0.45 µm). The membrane was incubated with anti-Claudin4 (1:1000, ThermoFisher Scientific) and anti-α-tubulin as a loading control (1:1000, Cedarlane, Canada) in Odyssey blocking buffer (LI-COR, USA) diluted 1:1 in TBS, after initial blocking with blocking buffer for 1 hr at RT.

Primary antibodies were detected by incubation with corresponding IRDye secondary antibodies (1:15000) for 1 hr at RT in blocking buffer and the Odyssey infrared imaging system (LI-COR, USA).

Intensity measurements of immunoreactivity were obtained using ImageJ software.

TABLE 3. Primer details

Target gene Species Forward primer Reverse primer

VCAM-1 Human TGA AGG ATG CGG GAG TAT ATG A TTA AGG AGG ATG CAA AAT AGA GCA CLDN4 Human GGC CTT ATG GTG ATA GTG CCG AGG CCA CCA GCG GAT TGT A 18SrRNA Rat TAC CAC ATC CAA GGA AGG CAG CA TGG AAT TAC CGC GGC TGC TGG CA

CCL2 Rat TGA TCC CAA TGA GTC GGC TG TGG ACC CAT TCC TTA TTG GGG Fibronectin1 Human ACT GTA CAT GCT TCG GTC AG AGT CTC TGA ATC CTG GCA TTG GAPDH Human CCA TGT TCG TGG GTG TG GGT GCT AAG CAG TTG GTG GTG GAPDH Rat AGG TTG TCT CCT GTG ACT TC CTG TTG CTG TAG CCA TAT TC ICAM-1 Human TAG CAG CCG CAG TCA TAA TGG G AGG CGT GGC TTG TGT GTT CG Serping1 Rat TGG CTC AGA GGC TAA CTG GC GAA TCT GAG AAG GCT CTA TCC CCA

(9)

Chapter 2

Migration assay

U373 astrocytes (either wild-type or claudin-4 overexpressing cells) were grown to confluence in a 24-well plate. Subsequently, either activated or resting human T cells (7.5*10^5 cells/ml) were added to the U373 monolayers. Co-cultures were placed in an inverted phase-contrast microscope (Zeiss Axiovert 200M) housed in a temperature-controlled (37oC), 5% CO2 chamber.

T cell migration was followed for 4 hours with an interval of 5 minutes and analysed using ImageJ analysis software. After, the plates were extensively washed to remove non-adherent cells, cells were fixed (4% PFA) and stained (DAPI) adherent T-cells were counted. Where indicated, cells were incubated in the presence of either blocking or stimulating antibodies. Anti-β2 mAb KIM185 was used to activate CD18 integrins [12], anti-LFA-1mAb NKI-L15 was used to block LFA-1 [13], NKI-L7 was used to block CD11A, NKI-L19 was used to block β2 integrins, TS2/16 was used to activate β1 integrins.

RESULTS

To investigate the role of astrocytes in immune cell infiltration into the CNS, sections of human brain tissue containing active MS lesions were immuno-stained and analysed by high resolution 3D fluorescence microscopy. Active lesions were selected based on the loss of myelin, as shown by staining for myelin proteolipid protein (PLP), and the presence of infiltrating or activated myeloid cells, indicated by MHC class II, throughout the lesional area (Supplemental Figure S1). Immunostaining revealed the classical neurovascular unit structure in normal appearing white matter (NAWM) brain tissue, with endothelial cells, surrounded by a glial fibrillary acidic protein (GFAP) immunoreactive astrocytic layer (Figure 1A). Interestingly, this structure seems to be altered in MS lesions. We observed that astrocytes in active MS lesions appeared to penetrate the basement membrane with fine processes. Our 3D analysis shows that the astrocytic processes are in direct contact with T cells, marked by anti-CD3 (Figure 1B). Importantly, we observed that not the entire CD3 positive T cell is covered with processes of the astrocytes and that the different astrocyte processes do not interact with each other.

The tight junction molecule claudin-4 is expressed by astrocytes under inflammatory conditions forms a physical barrier which prevents entry of immune cells into the CNS [9]. We here observed that IL1-β- and TGF-β-stimulated primary rat astrocytes show a reactive phenotype based on the expression of CCL2 and Serping1 (Supplemental figure S2) and indeed increased mRNA expression of claudin-4 (Figure 1C). Based on these findings, we hypothesized that claudin-4 is expressed on the astrocytic processes in MS tissue and that astrocyte processes cross through the basement membrane to interact with immune cells. This interaction could prevent immune cells from entering the CNS. Immunofluorescence staining confirmed that claudin-4 localized specifically to GFAP positive astrocytes (Figure 1D).

(10)

FIGURE 1. Fluorescence staining of astrocytes (GFAP), T-cells (CD3) and nuclei (DAPI) in the neurovascular unit in normal appearing white matter and an active MS lesion. (A&B) Dense CD3-immunolabeling (green) was observed in the perivascular space in active MS lesions, while being non detectable in NAWM. Astrocytes extend their endfeet through the basement membrane to interact with CD3+ T cells (Scale-bar = 5 µm). (C) Induction of CLDN4 mRNA on primary rat astrocytes after treatment with IL1-β and TGF-β. (D) Double immunofluorescence labelling shows co-localization of CLDN4 (green) with GFAP-positive astrocytes (red) in active MS lesions (scale-bar = 50 µm).

(11)

Chapter 2

FIGURE 2. Integrin β2 dependent cell migration and adhesion to claudin-4 expressing astrocytes. (A) Migration of activated and resting T cells on either a monolayer of U373/EC cells (black bar) and on a monolayer of U373/claudin-4 cells (grey bars). (B) Migration of activated T cells on a monolayer of U373/claudin-4 cells in the presence of integrin blocking antibodies. (C) Migration of resting T cells on a monolayer of U373/claudin-4 cells in the presence of integrin activating antibodies. (D) Adhesion of T cells on U373/EV and U373/claudin-4 cells, cells stained with DAPI. T cells were identified by their small nuclear appearance. Scale bar = 5µm (E) Quantification of adherent cells on U373/EV and U373/claudin-4 astrocytes.

The extracellular loop of tight junction proteins is known to interact with their counterpart on adjacent cells, thereby sealing cells together. However, claudin proteins are also capable of interacting with other molecules [14,15]. To address whether astrocytic claudin-4 functionally contributes to the physical interaction with T cells observed in MS lesions, we assessed whether claudin-4 overexpression on astrocytes promotes adhesion of T cells. Hereto, two astrocyte cell-lines were generated that either overexpress human claudin-4 (U373/claudin-4) or an empty vector (EV) control (U373/EV) (Supplementary figure S3A). PHA-L / IL-2 activated T cells plated on a

(12)

confluent monolayer of U373/claudin-4 showed reduced cell migration compared to T cells plated on U373/EV astrocytes. In addition, similar as described by Sam Horng et al., resting T cells showed reduced cell migration compared to activated T cells [9]

(Figure 2A). In addition, the number of attached CD3+ T cells, was significantly higher in U373/claudin-4 astrocytes compared to U373/EV (Figure 2D&E). These data confirm that activation of T cells is required for the reduced migration and increased adhesion of T cells and importantly, show that claudin-4 plays an important role in this process.

Since claudin-4 itself is not expressed by CD3+ T cells (data not shown), other molecules on the T cells are needed to interact with claudin-4. As lymphocyte function-associated antigen 1 (LFA-1, αLβ2 integrin) and very late antigen-4 (VLA-4, α4β1 integrin) are the predominant integrins expressed on CD3+ T cells, we hypothesized that these integrins might interact with astrocytic claudin-4. To this end, we used several integrin blocking or activating antibodies in the migration and cell adhesion assays.

Surprisingly, we observed that cell migration was most effectively reduced by anti-β2 blocking antibodies in the case of activated T cells (Figure 2B) and by anti-β2 stimulating antibodies in the case of resting T cells (Figure 2C). To rule out that induction of the expression of multiple ligands in astrocytes overexpressing claudin-4 mediated the interaction with β2 integrin on the T cells, we assessed the expression cell adhesion molecules by qPCR. Analysis on U373/EV and U373/claudin-4 cells revealed no change in vascular cell adhesion molecule 1 (VCAM-1), fibronectin and Intercellular Adhesion Molecule 1 (ICAM-1) (Supplementary figure S4A). In addition, immunocytochemistry analyses of fibronectin and laminin on U373/EV and U373/claudin-4 cells also did not differ in intensity or cellular location (Supplementary figure S4B). This suggests that the increased interaction between the claudin-4 expressing astrocytes and T cells is not due to changes in expression of astrocytic adhesion molecules.

DISCUSSION

In this study we show that astrocytes, under inflammatory conditions, interact with T cells in the perivascular space. Using in vitro models in which we cultured astrocytes together with T cells, we were able to show that the tight junction molecule claudin-4, expressed on astrocytes, and CD18 integrins expressed on T cells, are important in this interaction. Furthermore, we demonstrate that interfering with this interaction leads to a reduced binding of T cells to astrocytes. Taken together, this shows that astrocytes are actively involved in preventing T cell infiltration of the brain parenchyma

The importance of astrocytes in the regulation of immune cell influx into the brain has recently been demonstrated using in vivo models [16,17]. These studies show that astrocytes can both cause the barrier to strengthen but also that astrocytes can reduce

(13)

Chapter 2 the barrier properties. In addition to the formation of the glial limitans, astrocytes are actively involved in the regulation of BBB permeability through multiple pathways [18–20], and are a crucial source of chemoattractants when activated [21]. Reactive astrocytes are important drivers of BBB breakdown in MS lesions as they produce pro- inflammatory cytokines and pro-inflammatory lipids [22]. Together, these mediators induce BBB dysfunction by modulating the endothelial tight junction proteins [17]. Importantly, reactive astrocytes also serve a protective role in inflammatory conditions, as their conditional elimination or suppression results in strikingly larger and more aggressive lesions with increased infiltration of CD45+ immune cells in a CNS injury mouse model [23]. These studies highlight the significant role of astrocytes in the maintenance and establishment of the neurovascular unit and subsequently the trafficking of immune cells into the brain. However, the precise mechanism on how astrocytes regulate immune cell transmigration over the BBB and subsequently the glial limitans are not yet fully understood. Our results show that reactive astrocytes in active MS lesions employ a direct interaction with perivascular T cells via astrocytic processes that extend through the glial limitans. Furthermore, we and others show that astrocytes express tight junction molecules, claudin-4, claudin-1 and JAM-A on these processes [9]. Using in vitro co-culture models, it was previously shown that by employing these tight junction proteins, astrocytes corral activated T cells into distinct clusters. The authors hypothesised that the astrocytic processes are interconnected and form an impermeable barrier. Interestingly, the authors showed that the corralling of T cells by astrocytes only occurred when T cells were activated. This suggests that expression of tight junction proteins on astrocytes alone is not enough to enclose T cells, and also points out that there is probably a more complex mechanism behind T cell clustering than just a physical barrier. We show, using a similar in vitro co- culture model, that activated T-cells are able to interact with and bind to astrocytes.

Importantly, we show that astrocytes that overexpress tight junction protein claudin-4 bind more T cells compared to empty vector astrocytes. In addition, we also observed that this interaction is lost when T cells were not activated.

As activated T cells increase the surface expression of active integrins in contrast to resting T cells, we hypothesised that the interaction between T cells and astrocytes might be mediated by integrins expressed on the cell surface of activated T cells together with the tight junction proteins of the astrocytes. To this extent, we inhibited specific integrins on activated T cells with blocking antibodies, and vice versa, active specific integrins on resting T cells with activating antibodies. With both approaches, we observed that CD18 was important for the interaction between T cells and astrocytes and conclude that claudin-4 is able to interact with a CD18 integrin expressed on activated T cells. Whether similar interactions are employed between T cell integrins and the tight junction proteins claudin-1 and JAM-A remain to be investigated.

(14)

Astrocytes express MHC-I and MHC-II molecules, at least in vitro, upon exposure to inflammatory stimuli [24,25], that can interact with the T-cell receptor on T cells.

Furthermore, astrocytes are able to increase the expression of co-stimulatory molecules CD80 and CD86 after an pro-inflammatory insult [24]. Interestingly, both these co-stimulatory molecules were found to be expressed by astrocytes in chronic MS lesions [26]. However, we show that overexpression of claudin-4 leads to increased interaction with the T cells, and inhibiting CD18 prevented this interaction, indicating that claudin-4 is necessary. Whether a possible interaction between MHC-I and T-cell receptors is also important for the described process remains to be determined.

Astrocytes also express cell adhesion molecules such as ICAM-1 and VCAM-1. These adhesion molecules might facilitate adhesion between T cells and astrocytes, although we found that these molecules were not affected by overexpression of claudin-4, an interaction with the t cells could lead to further activation of the astrocytes, resulting in higher expression of other astrocytic adhesion molecules which could strengthen the T cell astrocyte interaction.

It is interesting to note that Clostridium perfringens enterotoxin (CPE), a potent ligand of tight junction claudin-3 and claudin-4 [27] is more than 10 times as prevalent in patients suffering from MS compared to healthy controls [28]. In addition, people suffering from MS have higher levels of antibodies against the toxin [29]. Although to date it is not completely understood how the CPE is involved in the pathophysiology of MS, there are some indications that the toxin can induce demyelination [30]. As our data show that astrocytes in MS lesions upregulate claudin-4, thereby interfering with T cell migration into the CNS, it is tempting to speculate whether CPE could possibly interfere with the interaction between T cells and astrocytes. Binding of CPE to claudin-3 or claudin-4 expressing prostate cancer epithelium cells leads to rapid lysis of the cells [31], so the presence of CPE could possibly lead to lysis of claudin-4 expressing astrocytes. This in turn could cause increased migration of CD3+ T-cells into the CNS, resulting in a more severe inflammatory response.

In many cases, binding of integrins with their ligands induces intracellular signalling pathways, leading to cellular activation. Indeed, it is thought that perivascular T cells need to be reactivated in order to cross the glial limitans and leave the perivascular space [4,6]. Whether the observed interaction between astrocytes and T cells is involved in the reactivation of T cells at the glial limitans is a subject for further research.

Our findings show that astrocytes interact with perivascular T cells in MS lesions. This interaction seems to be mediated via tight junction molecule claudin-4 expressed on astrocytes, and CD18 integrin expressed on the surface of activated T cells. We hypothesise that this interaction allows astrocytes to trap T cells within the perivascular space thereby, preventing T cell invasion into the brain.

(15)

Chapter 2 AUTHOR CONTRIBUTIONS

AK, MR, EV, JH and SV designed and interpreted the experiments. AK with the help of BH and SP performed the experiments. AK with the help of JH, MR and EV wrote the manuscript, which was reviewed by all authors.

ACKNOWLEDGEMENTS

We would like to thank Yvette van Kooyk for providing the KIM185, anti-LFA-1mAb NKI-L15, NKI-L7, NKI-L19 and TS2/16 antibodies. Expert technical support by the AO|2M facility (Advanced Optical Microscopy facility in O|2, VU Medical Center, Amsterdam) was highly appreciated.

FUNDING

This work was supported by the Dutch MS research foundation (MS-14-358e, AK), which had no role in study design, data collection and analyses, decision to publish, or preparation of the manuscript.

ETHICAL APPROVAL

VUmc MS Centrum Amsterdam and the Netherlands Brain Bank received permission to perform autopsies for the use of tissue and for access to medical records for research purposes from the Ethical Committee of the VU University Medical Center, Amsterdam, The Netherlands. All patients and controls, or their next of kin, had given informed consent for autopsy and use of brain tissue for research purposes. Buffy coats were obtained from volunteer donors at Sanquin after written informed consent was obtained.

REFERENCES

1. Reese TS, Karnovsky MJ. Fine structural localization of a blood-brain barrier to exogenous peroxidase. J Cell Biol 1967; 34:207–

17.

2. Lopes Pinheiro MA, Kooij G, Mizee MR, Kamermans A, Enzmann G, Lyck R, Schwaninger M, Engelhardt B, de Vries HE. Immune cell trafficking across the barriers of the central nervous system in multiple sclerosis and

stroke. Biochim Biophys Acta 2016; 1862:461–

71.

3. Engelhardt B, Coisne C. Fluids and barriers of the CNS establish immune privilege by confining immune surveillance to a two- walled castle moat surrounding the CNS castle.

Fluids Barriers CNS 2011; 8:4.

4. Owens T, Bechmann I, Engelhardt B.

Perivascular spaces and the two steps to

(16)

neuroinflammation. J Neuropathol Exp Neurol 2008; 67:1113–21.

5. Tran EH, Hoekstra K, van Rooijen N, Dijkstra CD, Owens T. Immune invasion of the central nervous system parenchyma and experimental allergic encephalomyelitis, but not leukocyte extravasation from blood, are prevented in macrophage-depleted mice. J Immunol 1998; 161:3767–75.

6. Körner H, Riminton DS, Strickland DH, Lemckert FA, Pollard JD, Sedgwick JD. C r i t i c a l points of tumor necrosis factor action in central nervous system autoimmune inflammation defined by gene targeting. J Exp Med 1997;

186:1585–90.

7. Grewal IS, Foellmer HG, Grewal KD, Wang H, Lee WP, Tumas D, Janeway CA, Flavell RA.

CD62L is required on effector cells for local interactions in the CNS to cause myelin damage in experimental allergic encephalomyelitis.

Immunity 2001; 14:291–302.

8. Agrawal S, Anderson P, Durbeej M, Van Rooijen N, Ivars F, Opdenakker G, Sorokin LM. Dystroglycan is selectively cleaved at the parenchymal basement membrane at sites of leukocyte extravasation in experimental autoimmune encephalomyelitis. J Cell Biol 2006; 173:1007–1019.

9. Horng S, Therattil A, Moyon S, Gordon A, Kim K, Argaw AT, Hara Y, Mariani JN, Sawai S, Flodby P, Crandall ED, Borok Z, Sofroniew M V., et al. Astrocytic tight junctions control inflammatory CNS lesion pathogenesis. J Clin Invest 2017; 127:3136–3151.

10. Chakraborty A, Kamermans A, van Het Hof B, Castricum K, Aanhane E, van Horssen J, Thijssen VL, Scheltens P, Teunissen CE, Fontijn RD, van der Flier WM, de Vries HE.

Angiopoietin like-4 as a novel vascular mediator in capillary cerebral amyloid angiopathy. Brain 2018; 141:3377–3388.

11. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real- time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001; 25:402–8.

12. Andrew D, Shock A, Ball E, Ortlepp S, Bell J, Robinson M. KIM185, a monoclonal antibody to CD18 which induces a change in the conformation of CD18 and promotes both LFA-1- and CR3-dependent adhesion. Eur J Immunol 1993; 23:2217–22.

13. Grabovsky V, Dwir O, Bleijs DA, Alon R, Figdor CG, van Kooyk Y, van Vliet SJ, Sigal A.

The LFA-1 Integrin Supports Rolling Adhesions on ICAM-1 Under Physiological Shear Flow in a Permissive Cellular Environment. J Immunol 2014; 165:442–452.

14. Ludwig RJ, Hardt K, Hatting M, Bistrian R, Diehl S, Radeke HH, Podda M, Schön MP, Kaufmann R, Henschler R, Pfeilschifter JM, Santoso S, Boehncke W-H. J u n c t i o n a l adhesion molecule (JAM)-B supports lymphocyte rolling and adhesion through interaction with alpha4beta1 integrin.

Immunology 2009; 128:196–205.

15. Van Itallie CM, Anderson JM. Claudin interactions in and out of the tight junction.

Tissue Barriers 2013; 1:e25247.

16. Yang JF, Tao HQ, Liu YM, Zhan XX, Liu Y, Wang XY, Wang JH, Mu LL, Yang LL, Gao ZM, Kong QF, Wang GY, Han JH, et al.

Characterization of the interaction between astrocytes and encephalitogenic lymphocytes during the development of experimental autoimmune encephalitomyelitis (EAE) in mice.

Clin Exp Immunol 2012; 170:254–265.

17. Nair A, Frederick TJ, Miller SD.

Astrocytes in multiple sclerosis: a product of their environment. Cell Mol Life Sci 2008;

65:2702–20.

18. Podjaski C, Alvarez JI, Bourbonniere L, Larouche S, Terouz S, Bin JM, L??cuyer MA, Saint-Laurent O, Larochelle C, Darlington PJ, Arbour N, Antel JP, Kennedy TE, et al.

Netrin 1 regulates blood-brain barrier function and neuroinflammation. Brain 2015; 138:1598–

1612.

19. Mizee MR, Nijland PG, van der Pol SMA a, Drexhage JARR, van Het Hof B, Mebius R, van der Valk P, van Horssen J, Reijerkerk A, de Vries HE. Astrocyte-derived retinoic acid: a novel regulator of blood-brain barrier function in multiple sclerosis. Acta Neuropathol 2014;

128:691–703.

20. Minagar A, Alexander JS. B l o o d - brain barrier disruption in multiple sclerosis.

Mult Scler 2003; 9:540–9.

21. Li C, Zhao R, Gao K, Wei Z, Yin MY, Lau LT, Chui D, Hoi Yu AC. Astrocytes: implications for neuroinflammatory pathogenesis of Alzheimer’s disease. Curr Alzheimer Res 2011;

8:67–80.

(17)

Chapter 2

22. van Doorn R, Nijland PG, Dekker N, Witte ME, Lopes-Pinheiro MA, van het Hof B, Kooij G, Reijerkerk A, Dijkstra C, Van Van Der Valk P, van Horssen J, De Vries HE.

Fingolimod attenuates ceramide-induced blood-brain barrier dysfunction in multiple sclerosis by targeting reactive astrocytes. Acta Neuropathol 2012; 124:397–410.

23. Bush TG, Puvanachandra N, Horner CH, Polito A, Ostenfeld T, Svendsen CN, Mucke L, Johnson MH, Sofroniew M V.

Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar- forming, reactive astrocytes in adult transgenic mice. Neuron 1999; 23:297–308.

24. Cornet A, Bettelli E, Oukka M, Cambouris C, Avellana-Adalid V, Kosmatopoulos K, Liblau RS. Role of astrocytes in antigen presentation and naive T-cell activation. J Neuroimmunol 2000; 106:69–77.

25. Zeinstra E, Wilczak N, Chesik D, Glazenburg L, Kroese FGM, De Keyser J.

Simvastatin inhibits interferon-γ-induced MHC class II up-regulation in cultured astrocytes. J Neuroinflammation 2006; 3:7–10.

26. Zeinstra E, Wilczak N, De Keyser J.

Reactive astrocytes in chronic active lesions of multiple sclerosis express co-stimulatory molecules B7-1 and B7-2. J Neuroimmunol 2003; 135:166–171.

27. Hewitt KJ, Agarwal R, Morin PJ. T h e claudin gene family: expression in normal and neoplastic tissues. BMC Cancer 2006; 6:186.

28. Rumah KR, Linden J, Fischetti VA, Vartanian T. Isolation of Clostridium perfringens type B in an individual at first clinical presentation of multiple sclerosis provides clues for environmental triggers of the disease. PLoS One 2013; 8:e76359.

29. Wagley S, Bokori-Brown M, Morcrette H, Malaspina A, D’Arcy C, Gnanapavan S, Lewis N, Popoff MR, Raciborska D, Nicholas R, Turner B, Titball RW. Evidence of Clostridium perfringens epsilon toxin associated with multiple sclerosis. Mult Scler 2018:1352458518767327.

30. Harris JM, Zhao B, Ma Y, Linden JR, Vartanian T, Schaeren-Wiemers N, Rumah KR.

Clostridium perfringens Epsilon Toxin Causes Selective Death of Mature Oligodendrocytes and Central Nervous System Demyelination.

MBio 2015; 6:1–13.

31. Long H, Crean CD, Lee WH, Cummings OW, Gabig TG. Expression of Clostridium perfringens enterotoxin receptors claudin-3 and claudin-4 in prostate cancer epithelium.

Cancer Res 2001; 61:7878–81.

(18)

SUPPLEMENTAL FIGURE 1. MS lesions are characterized by loss of myelin (PLP) and abundant presence of HLA-DR (MHC-II)-positive cells compared to normal appearing white matter (NAWM).

Scale bar = 5mm (overview) and 50µm (higher magnifications).

SUPPLEMENTAL FIGURE 2. IL1-β and TGF-β treatment induced expression of CCL2 and Serping1, two reactive astrocyte markers. N=4 independent experiments. ***p <0.002

(19)

Chapter 2

SUPPLEMENTAL FIGURE 3. Claudin-4 expression in U373/empty vector and U373/claudin-4 cell lines. (A) Gene expression levels of claudin-4 in U373/claudin-4 cells compared to U373/EV (N = 2) (B) Representative western blot of U373/EV and U373/claudin-4 protein lysates developed with anti-claudin-4 and anti-α tubulin (loading control) antibodies. (C) protein expression of claudin-4 detected using immunocytochemistry. Scale bar = 25 µm

SUPPLEMENTAL FIGURE 4. No effect of claudin-4 expression on adhesion molecules. (A) mRNA expression of adhesion molecules on U373/EV and U373/claudin-4 cells (B) Protein expression of adhesion molecules on U373/EV and U373/claudin-4 cells as measured by microscopy.

Referenties

GERELATEERDE DOCUMENTEN

Echter door het huidige gebruik (met name de verdieping) is de kwaliteit van het systeem hard achteruitgegaan: het water is troebel, de waterkwaliteit is slecht, de vloedstroom is

— Bij inzet van middelen kiezen voor minst milieubelastende stof (door middel van milieumeetlat).. De groepen &#34;Natuursla in Noord-Brabant en

Het is te verwachten dat het in figuur 3.6 gepresenteerde raamwerk waarin de achtergronden van vraag en aanbod worden belicht vanuit beleids- optiek een bijdrage zal leveren aan

anti H-Y CTL line was incubated for 4 h with bone marrow mononuclear cells, a dose-dependent inhibition of the growth of CFU-GM, BFU-E and CFU-GEMM from HLA-A2 or -B7 positive

Here, we investigated the expression of five mH antigens, designated HA- oictic progenitor cells, using a cell-mediated cytotoxicity assay (32). We report the expression of the mH

Here, we investigated the expression pattern of DAY- SLEEPER, assessed functional complementation of the daysleeper phenotype with different deletions of the DAYSLEEPER coding

Based on the similarity between the yeast mating pathway and mammalian GPCR signaling pathways, human GPCRs have been expressed and further coupled to a reporter gene

Dependent variable Household expectations Scaled to actual inflation Perceived inflation scaled to lagged inflation Perceived inflation scaled to mean inflation of past