Skip to main content
Erschienen in: Acta Neuropathologica 6/2015

Open Access 01.12.2015 | Original Paper

Highly encephalitogenic aquaporin 4-specific T cells and NMO-IgG jointly orchestrate lesion location and tissue damage in the CNS

verfasst von: Bleranda Zeka, Maria Hastermann, Sonja Hochmeister, Nikolaus Kögl, Nathalie Kaufmann, Kathrin Schanda, Simone Mader, Tatsuro Misu, Paulus Rommer, Kazuo Fujihara, Zsolt Illes, Fritz Leutmezer, Douglas Kazutoshi Sato, Ichiro Nakashima, Markus Reindl, Hans Lassmann, Monika Bradl

Erschienen in: Acta Neuropathologica | Ausgabe 6/2015

Abstract

In neuromyelitis optica (NMO), astrocytes become targets for pathogenic aquaporin 4 (AQP4)-specific antibodies which gain access to the central nervous system (CNS) in the course of inflammatory processes. Since these antibodies belong to a T cell-dependent subgroup of immunoglobulins, and since NMO lesions contain activated CD4+ T cells, the question arose whether AQP4-specific T cells might not only provide T cell help for antibody production, but also play an important role in the induction of NMO lesions. We show here that highly pathogenic, AQP4-peptide-specific T cells exist in Lewis rats, which recognize AQP4268–285 as their specific antigen and cause severe panencephalitis. These T cells are re-activated behind the blood–brain barrier and deeply infiltrate the CNS parenchyma of the optic nerves, the brain, and the spinal cord, while T cells with other AQP4-peptide specificities are essentially confined to the meninges. Although AQP4268–285-specific T cells are found throughout the entire neuraxis, they have NMO-typical “hotspots” for infiltration, i.e. periventricular and periaqueductal regions, hypothalamus, medulla, the dorsal horns of spinal cord, and the optic nerves. Most remarkably, together with NMO-IgG, they initiate large astrocyte-destructive lesions which are located predominantly in spinal cord gray matter. We conclude that the processing of AQP4 by antigen presenting cells in Lewis rats produces a highly encephalitogenic AQP4 epitope (AQP4268–285), that T cells specific for this epitope are found in the immune repertoire of normal Lewis rats and can be readily expanded, and that AQP4268–285-specific T cells produce NMO-like lesions in the presence of NMO-IgG.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00401-015-1501-5) contains supplementary material, which is available to authorized users.

Introduction

Neuromyelitis optica (NMO) is an inflammatory, astrocytopathic disease of the central nervous system (CNS) [8, 36]. Ever since the hallmark of this disease—the presence of pathogenic autoantibodies in the serum of most NMO patients [19, 20]—has been recognized, a lot of effort has been made to study the role of antibodies and T cells in lesion formation and expansion. Based on these studies we know (1) that in most patients, the pathogenic autoantibodies are directed against aquaporin 4 (AQP4), a water channel enriched on astrocytic endfeet at the perivascular and subpial glia limitans [20, 27], (2) that these antibodies belong to the IgG1 subgroup of immunoglobulins which need T cell help in their formation [20], and (3) that these antibodies have additional requirements for T cells in lesion formation: They need them for opening of the blood–brain barrier to gain access to the CNS parenchyma [6, 7, 14], and they need them to create a CNS environment facilitating antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytotoxicity (CDC) against their astrocytic targets [15, 33]. Such an environment is only created by T cells which are activated within the CNS [33], which is in line with the presence of Ox40+ CD4+ T cells in early lesions of human NMO patients and their experimental counterparts [33]. In order to become activated within the CNS, CD4+ T cells must encounter “their” specific, CNS-intrinsic antigen in the context of MHC class II products [13]. Considering the facts that AQP4-specific T cells provide help to the formation of AQP4-specific antibodies, and that AQP4-specific T cells are clonally expanded in NMO patients [23, 38], it is tempting to speculate that the antigen leading to T cell activation within the CNS of NMO patients is AQP4 as well. However, T cell responses against AQP4 target a surprisingly large number of epitopes in humans [22, 37] and even in single patients [23], which is reflected in Lewis rats [32] and in C57BL/6 or SJL/J mice [12, 25] (Fig. 1). Moreover, until recently, only weakly pathogenic AQP4 peptide-specific T lymphocytes could be derived from Lewis rats [32]. These T cells essentially pile up in the meninges, but hardly infiltrate the CNS parenchyma, suggestive of a limited activation of these cells [13, 32, 33]. AQP4-reactive T cells could also be obtained after immunization of C57BL/6 aquaporin-4 null mice with a combination of the human AQP4 extracellular loop peptides AQP456–69, AQP4135–153, and AQP4212–230 in complete Freund’s adjuvans and subsequent in vitro polarization of the peptide-specific T cells towards a TH17 phenotype [10]. These T cells were encephalitogenic, as evidenced by the induction of inflammatory lesions in spinal cords and optic nerves and by the induction of clinical signs of CNS inflammation, but derived from animals which did not have to overcome immunological tolerance, due to the absence of AQP4. In addition, the animals have been challenged with human instead of murine AQP4 [10]. Cumulatively, the findings obtained from both animal models raised the questions whether strongly encephalitogenic AQP4-specific T cells exist at all in the normal immune repertoire, and whether these cells can guide astrocyte-destructive lesions to NMO-typical sites. These questions were addressed in the current study, using Lewis rats as a model organism.

Materials and methods

Animals

Lewis rats (7–8 weeks old) were obtained from Charles River Wiga (Sulzfeld, Germany). They were housed in the Decentral Facilities of the Institute for Biomedical Research (Medical University Vienna) under standardized conditions. The experiments were approved by the Ethic Commission of the Medical University Vienna and performed with the license of the Austrian Ministry for Science and Research.

Characterization of the immunoglobulins used in transfer experiments

The NMO-IgG preparations containing pathogenic AQP4-specific antibodies derived from therapeutic plasmapheresates/sera of two different patients (“NMO-IgG9” and “pt1”; both NMO-IgGs worked equally well). The NMO-IgGs were essentially prepared and purified as described [7], and adjusted to an IgG concentration of 10 mg/ml. The use of it for research was approved by the Ethics Committee of Tohoku University School of Medicine (No. 2007-327) and by the Regional and National Ethical Committee of Hungary (3893.316-12464/KK4/2010 and 42341-2/2013/EKU). The normal human IgG preparation used as a negative control was commercially available (Subcuvia™, Baxter, Vienna), and was also diluted with phosphate-buffered saline (PBS) to an IgG concentration of 10 mg/ml prior to use.

Antigens

For immunization and T cell isolation/propagation, large peptides or fusion proteins containing predicted epitopes [32] were used (Table 1). These peptides were synthesized by Centic Biotec (Heidelberg, Germany), or, in the case of the human AQP-4 peptide AQP-4278–323, were expressed in E. coli using the pBAD/TOPO ThioFusion Expression System (Invitrogen, Carlsbad, CA, USA) and purified as described [32].
Table 1
Peptide and epitope sequences
Peptide
Amino acid sequence
Epitopes contained
AQP427–69
KGVWTQAFWKAVTAEFLAMLIFVLLSVGSTINWGGSENPLPVD
p33–41: AFWKAVTAE
p55–63: STINWGGSE
AQP4171–190
VFTIFASCDSKRTDVTGSVA
p176–184: ASCDSKRTD
AQP4207–232
YTGASMNPARSFGPAVIMGNWENHWI
p220–228: PAVIMGNWE
AQP4237–277
PIIGAVLAGALYEYVFCPDVELKRRLKEAFSKAAQQTKGSY
p241–249: AVLAGALYE
p249–257: EYVFCPDVE
AQP4268–285
KAAQQTKGSYMEVEDNRS
p271–279: QQTKGSYME
p273–281: TKGSYMEVE
AQP4 278–323 a
MEVEDNRSQVETDDLILKPGVVHVIDVDRGEEKKGKDQSGEVLSSV
p297–305: GVVHVIDVD
p300–308: HVIDVDRGE
Unless otherwise stated, the peptides/epitopes and amino acid sequences derive from the rat AQP4 sequence (GI:5019990)
aThis peptide and the corresponding amino acid sequences and epitopes derive from the human AQP4 sequence (GI:4502181)
For specificity tests (see below), also full-length human M23 AQP4 (gene bank accession number: NP-004019) was used, which has 100 % identity to the rat epitopes contained in AQP4207–232 (PAVIMGNWE) and AQP4268–285 (QQTKGSYME and TKGSYMEVE), and contains the human sequence of AQP4 recognized by AQP4278–323-specific T cells (GVVHVIDVD and HVIDVRGE, Table 1). For the preparation of this protein, HEK293A cells were transiently transfected with pcDNA3.1(M23)AQP4, allowing the production of AQP4 as a 6-HIS-tagged protein. 72 h later, the cells were washed with sterile phosphate-buffered saline (PBS) and exposed to lysis buffer (10 mM Tris buffer pH7.5, 100 mM NaCl, 1 mM EDTA, 1 % Triton X-100 and complete protease inhibitor cocktail tablet) for 1 h at 4 °C. The lysate was thoroughly mixed by pipetting, subjected to repeated rounds of freezing and thawing, sonicated using a Sonopuls GM70 (Bandelin, Berlin, Germany), and finally passaged through a 23 gauge needle. Ni NTA-Agarose Superflow (Qiagen) was then used for the purification of AQP4 following the instructions of the manufacturer. Briefly, Ni NTA beads were gently applied to a column, washed with 5 volumes of wash buffer (20 mM Tris/125 mM NaCl) prior to applying the lysate mix diluted 1:1 in wash buffer containing 1 % Triton X-100. Following washing steps with 10 volumes of wash buffer containing 1 % Triton X-100, we were washing the column with 10 volumes of washing buffer containing 0.1 % Triton X-100. Subsequently, we eluted AQP4 in 20 mM Tris/125 mM NaCl/0.1 % Triton X-100/600 mM Imidiazole. Following dialyses against PBS we used AQP4 in a concentration of 1 mg/ml in PBS/0.1 % Triton X-100. The eluted AQP4 protein was confirmed by Western blot (data not shown). In specificity tests, also recombinant human MOG1–125 was used, which was essentially produced and purified as described [3]. The MOG35–55-specific T cells used were raised against rat/mouse MOG35–55 (Sigma).

Immunization and T cell line preparation

The animals were subcutaneously immunized with 100 µl of a 1:1 mixture of the relevant antigen (stock 2 mg/ml) in Freund’s incomplete adjuvans supplemented with 4 mg/ml mycobacterium tuberculosis H37Ra. 9–11 days after the immunization, the animals were killed. At this point, they were all clinically healthy and did not show any evidence for inflammation of the CNS or of peripheral organs. The lymph nodes draining the immunization site were removed, and peptide-specific T cell lines were established as described [32, 33].

Isolation of naïve T cells

The naïve T cells tested derived from the spleen of an adult Lewis rat housed under specific pathogen-free conditions. The spleen was processed to a single cell suspension, and contaminating erythrocytes were removed by incubation of the cells for 5 min in hypotonic salt solution (0.15 M NH4Cl, 1 mM KHCO3, 0.1 mM Na2EDTA) pH 7.4.

Preparation of T cells for immunocytochemistry

T cells were embedded in HistoGel (Thermoscientific, Cheshire, UK) according to the manufacturer’s instructions and subsequently fixed with 4 % paraformaldehyde for 24 h. The HistoGel blocks were then processed for immunohistochemical analysis as detailed [2, 7].

Characterization of T cell lines

Specificity tests

Specificity was determined in T cell proliferation assays, using 96 well plates. 5 × 105 AQP4-peptide-specific T cells were cocultured in triplicates with 1 × 106 thymic antigen presenting cells in the absence of externally added antigen or in the presence of irrelevant CNS antigens (i.e. myelin basic protein or unrelated AQP4-peptides; 10 µg/ml final) as negative controls, of the peptide against which the cell line was established as specific antigen (10 µg/ml final), or of concanavalin A (2.5 µg/ml final) as positive control [9, 11].
The human M23 AQP4 preparation had a protein concentration of 1 mg/ml in phoshate-buffered saline/0.1 % Triton X-100. To avoid the toxic effects of Triton X-100, we diluted the antigen preparation 1:100 with PBS and coated it in 100 µl aliquots over night at 4 °C onto flat-bottom 96-well plates. On the next morning, the coating solution was drained from the plates, and each well was washed gently and briefly with 100 µl culture medium containing 1 % rat serum. Then, 1 × 106 thymic antigen presenting cells in 100 µl medium were cultured in these plates for 8 h at 37 °C prior to the addition of T cells. Exactly the same procedure was applied to recombinant human MOG1–125.
The cells were cultured for 48–72 h. For the final 18–24 h in culture, [3H]-thymidine was added to reveal de novo DNA synthesis during the S-phase of the cell cycle of activated T cells.

Analysis of surface marker expression by flow cytometry

For staining, the cells were incubated for 30 min at room temperature with antibodies against rat CD4 (W3/25, mouse monoclonal, Serotec) or mouse IgG1 (Dako, Glostrup, Denmark; isotype matched control antibody). After washing, the cells were incubated for 30 min at room temperature for 30 min with polyclonal Alexa488-labeled goat anti-mouse IgG (Jackson ImmunoResearch). For staining of the αβ-T cell receptor (TCR), PE-labeled mouse anti-αβTCR (eBioscience, San Diego, CA) was used. All antibodies were used in a dilution of 1:100 in stain buffer (PBS/10 % fetal calf serum/1 mM EDTA).

Quantitative real-time polymerase chain reaction (qPCR) for the detection of transcripts for IFN-γ and IL-17

RNA was purified from freshly activated T cell blasts using the RNeasy Plus Mini Kit (Qiagen GMbH, Hilden, Germany). Genomic DNA was removed and the RNA transcribed to cDNA as described [16]. qPCR was performed using the SsoAdvanced Universal SYBR Green Supermix (Bio-Rad) according to the manufacturer’s instructions, using a 10 µl reaction mixture [5 µl SsoAdvanced Universal SYBR Green Supermix, 0.2 µl forward primer (10 pmol/µl), 0.2 µl reverse primer (10 pmol/µl), 3.6 µl double-distilled H2O, 1 µl DNA template] in a StepOnePlus system (applied biosystems). The following primers were used: IL-17 forward: 5′-TACCAGCTGATCAGGACGAG-3′; IL-17 reverse: 5′-CATCAGGCACATGGATGGAA-3′; IFN-γ forward: 5′-ATTCATGAGCATCGCCAAGTTC-3′; IFN-γ reverse: 5′-TGACAGCTGGTGAATCACTCTGAT-3′; GAPDH forward: 5′-CCGAGGGCCCACTAAAGG-3′; GAPDH reverse: 5′-ATGGGAGTTGCTGTTGAAGTCA-3′. For qPCR, an initial denaturation step (95 °C, 30 s) was followed by 40 cycles of denaturation (95 °C, 15 s) and annealing/extension (60 °C, 1 min). The absence of unspecific amplification was determined by melt curve analysis. All reactions were run in triplicates.

Induction of experimental autoimmune encephalomyelitis (EAE) and experimental autoimmune neuromyelitis optica (ENMO)

EAE was induced in Lewis rats by intraperitoneal transfer of activated AQP4-peptide-specific T cells. The numbers of T cells transferred for each T cell line is shown in Fig. 2. The animals were monitored daily for weight loss and clinical signs of EAE, which was scored according to the following scheme: 0: healthy; 0.5: partial loss of tail tonus; 1: complete loss of tail tonus; 2: hind limb weakness, unsteady gait; 3: hind limb paralysis (this was for ethical reasons set as endpoint for EAE experiments). The animals were killed by an overdose of CO2 on days 5/6 after T cell-transfer, and perfused with 4 % phosphate-buffered paraformaldehyde (PFA). Brains and spinal cords were dissected, immersed for another 18–24 h in PFA and embedded in paraffin for histological analysis. For the induction of ENMO, AQP4-peptide-specific T cells were injected intraperitoneally on day 0, followed by an intraperitoneal injection with 1 ml phosphate-buffered saline (PBS) containing either 10 mg NMO-IgG or 10 mg normal human IgG on day 5. Also these animals were killed on day 7 after T cell-transfer with CO2, and perfused with 4 % PFA.

Antigen injections into the cisterna magna

On day 5 after EAE induction by T cell transfer, the animals were anesthetized with Ketanest S/Rompun. Afterwards, the neck of the animals was flexed, and the skin was cut open to expose the atlanto-occipital membrane, which was then punctured with a thin glass capillary. When the entry of clear cerebrospinal fluid into the capillary indicated the accuracy of the injection site, 5 µl of AQP4 peptides (dissolved in a concentration of 2 mg/ml in RPMI), or of RPMI alone were injected slowly. Afterwards, the capillary was withdrawn and the skin was closed with wound clips. 24 h later, the animals were killed with CO2 and perfused with 4 % PFA. Brains and spinal cords were processed for immunohistochemical analysis as detailed [2, 7].

Immunohistochemistry

All stainings were done essentially as described [2, 7] using the mouse monoclonal antibody ED1 (to stain macrophages and activated microglia; Serotec, Germany), rabbit polyclonal antibodies against CD3 (to stain T cells; NeoMarkers, Fremont, USA), rabbit polyclonal antibodies against AQP4 (to stain astrocytes; Sigma, Germany), rabbit polyclonal or mouse monoclonal antibodies against glial fibrillary acidic protein (GFAP; from Dako, Denmark, or NeoMarkers, respectively), anti-human immunoglobulin (biotinylated donkey; polyclonal; Amersham, UK), anti-rat immunoglobulin (biotinylated donkey; polyclonal; Jackson ImmunoResearch), and anti-complement C9 (rabbit polyclonal [29]).
For double immunostainings of proliferating cell nuclear antigen (PCNA, mouse, clone PC10, DAKO) and CD3 (rabbit polyclonal; NeoMarkers, Fremont, USA), sections were steamed in citrate buffer for 30 min. After incubation with mouse anti-proliferating cell nuclear antigen (PCNA, clone PC10, DAKO, 1:10 000) over night at 4 °C, the sections were incubated with alkaline-phosphatase-labeled anti-mouse antibodies (Jackson ImmunoResearch, West Grove PA, USA, 1:200) for 1 h at RT and developed with Fast Blue (FB, Sigma, Germany) substrate. Then the sections were incubated with rabbit anti-CD3 (polyclonal; NeoMarkers, Fremont, USA, 1:2000) over night at 4 °C. After incubation with biotinylated anti-rabbit antibodies (1:2000) for 1 h at RT and enhancement with CSA (1:1000) for 20 min, avidin-peroxidase (Sigma) was applied and the sections were developed with aminoethyl carbazole (AEC, Sigma).

Quantitative evaluation of immunostained sections

Quantification was done by using a morphometric grid. To determine the extent of T cell infiltration, the number of CD3+ cells was determined in 3 different areas: within in the meninges, within the superficial parenchyma (<100 µm distance from the meninges) and within the deep parenchyma (>100 µm distance from the meninges).

Statistical evaluation

Statistics were calculated with the IBM SPSS Statistics 21. The Mann–Whitney (Wilcoxon) W test (comparison of medians) was used in all cases. For multiple comparisons, Bonferroni correction was used.

Results

All AQP4 epitopes suitable for binding to Lewis rat MHC class II (RT1.BL) give rise to antigen-specific T cell responses

We used peptides spanning AQP4 epitopes previously identified as potential binders to RT1.BL of Lewis rats [32] for the immunization of Lewis rats. Although all animals remained clinically healthy upon immunization, they mounted T cell responses against all of the peptides used. Consequently, different peptide-specific T cell lines could be established by alternating cycles of antigen-specific T cell activation and IL-2-driven T cell propagation (Table 1) which constrains the generation of TH17 cells [18]. These cells were CD4+, and expressed the αβ T cell receptor (suppl. fig  1). All of these cell lines showed a dominant expression of IFN-γ over IL-17 (suppl. fig 2), and therefore belonged to the TH1 subset of cells. They were all responsive to their specific peptides, but the AQP427–69- and AQP4278–232-specific T cell lines did not reach a stimulation index >2 (suppl fig. 3).
We tested the encephalitogenic potential of these cells, i.e. their ability to induce CNS inflammation, by their transfer into naïve Lewis rats.

AQP4-peptide-specific T cell lines vary in encephalitogenicity and ability for parenchymal infiltration

The different AQP4-peptide-specific CD4+ T cells varied in their ability to induce CNS inflammation and could be grouped into weak, medium, and strongly encephalitogenic lines. Weak encephalitogenicity was observed upon transfer of T cells with specificity for AQP4171–190, AQP427–69, AQP4278–323, and AQP4237–277. Even after transfer of as many as 10 × 107 T cells/animal, these lines were just able to yield an average of less than 2 lesions per rat. These lesions were essentially located within the meninges, and only very few T cells entered the CNS parenchyma (Fig. 2). The recipient animals did not show any symptoms of clinical disease, in line with this weak histological evidence of CNS inflammation. Medium encephalitogenicity was observed with T cells specific for AQP4207–232 containing the epitope PAVIMGNWE. On average, transfer of these cells lead to ~10 lesions per animal, distributed along the entire neuraxis. However, these cells only barely infiltrated the CNS parenchyma. Instead, they piled up in the meninges, resulting in up to 10 layers of T cells on top of each other (Fig. 2). Although there was more histological evidence of CNS inflammation than seen with the AQP4 peptide-specific T cells described above, we still did not see signs of clinical disease. Strong encephalitogenicity was observed in T cells specific for AQP4268–285.

AQP4268–285-specific T cells are highly encephalitogenic

While for all other cell lines, 3.5-10 × 107 T cells had to be transferred to produce inflammatory CNS lesions, as few as 0.03 × 107 AQP4268–285-specific T cells/animals sufficed to do so. For all analysis described below, 3 × 106 AQP4268–285-specific T cells/animals were used. These cells caused CNS inflammation along the entire neuraxis, i.e. in all levels of the spinal cord, throughout the optic nerve, and in the entire brain (Figs. 3, 4). “Hotspots” for inflammatory lesions were areas around the 3rd and 4th ventricle, in the hippocampus, in the periaqueductal gray, in cerebellum and medulla, and in the spinal cord in dorsal horns and central gray matter (Figs. 3, 4). Lesions at similar sites in the brain were also seen after transfer of T cells with other CNS antigen specificities like S100β or MBP (suppl fig. 4). However, the massive dominance of T cell infiltrates in spinal cord gray matter seen with AQP4268–285-specific T cells was unique for these cells (suppl. fig 4). The CNS lesions provoked by AQP4268–285-specific T cells were essentially T cell-dominated with little microglia activation/macrophage recruitment (Figs. 3, 4), and caused weight loss with partial loss of tail tonus as sole clinical symptom. Some AQP4268–285-specific T cells were found in the meninges, but a much larger proportion of these cells was able to infiltrate the CNS gray and white matter parenchyma than seen with any other AQP4-peptide-specific T cell line before (Figs. 3, 4).

AQP4207–232- and AQP4268–285-specific T cells differ in their extent of activation within the CNS

Why are AQP4268–285-specific T cells able to deeply immigrate into the CNS parenchyma, while all other AQP4peptide-specific T cells studied so far pile up in the meninges? To address this question, we made T cell proliferation assays to analyze whether antigen presenting cells can process full-length AQP4 to produce and present T cell epitopes to AQP4207–232-, and AQP4268–285-specific T cells. We observed that these T cells became activated and proliferated in the presence of AQP4 and syngenic antigen presenting cells (Fig. 5; suppl fig. 5), indicating that the epitopes recognized arise from naturally processed AQP4. We next focused on AQP4268–285-specific T cells and on AQP4207–232-specific T cells as “prototype” for all other AQP4peptide-specific T cells and studied the activation of AQP4268–285- and AQP4207–232-specific T cells in the CNS. It was not possible to re-isolate and characterize these cells by FACS analysis, since AQP4207–232-specific T cells had to be used in extremely high cell numbers for EAE induction (Fig. 2) and caused only few lesions of small size. Instead, we studied T cell activation by immunohistochemistry, using double stainings with CD3 as T cell marker, and with the proliferating cell nuclear antigen PCNA as activation marker. To ensure that PCNA is a reliable marker for T cell activation, we first tested the anti-PCNA antibody in vitro, using T cells cultured for different lengths of time after their antigen-specific activation. We observed, that PCNA was detectable in 90 % of the T cells on day 0 after a 48 h-lasting T cell activation by antigen in the context of RT1.BL, in 80 % of the T cells on day 2, in 50 % of T cells on day 4, and became essentially undetectable (<1 %) in T cells on day 6 (Fig. 6). We next evaluated the percentage of activated PCNA+ AQP4268–285- specific T cells in the total CD3+ T cell pool, depending on the depth of parenchymal infiltration. In medians, 14 % PCNA+ CD3+ T cells were found in the meninges, 32 % located in the superficial, and 44 % in the deep parenchyma. Cumulatively, these data showed that AQP4268–285-specific T cells are much better activated in the CNS than AQP4207–232-specific T cells. What restricts the activation of AQP4207–232-specific T cells?

The amount of available AQP4207–232-peptide limits CNS infiltration by AQP4207–232-peptide-specific T cells

To test whether low amounts of AQP4207–232 preclude efficient T cell activation and subsequent parenchyma infiltration, we increased its concentration by injecting AQP4207–232 into the cisterna magna at the very onset of AQP4207–232-peptide-specific T cell-mediated EAE [1, 42]. Intracisternal injections of an irrelevant AQP4 peptide (AQP4236–277) and of vehicle (=RPMI) into AQP4207–232-peptide-specific T cell-challenged rats, and of peptides AQP4236–277 and AQP4207–232 into naïve rats served as controls. The animals were killed 24 h later for immunohistochemical analyses. When AQP4207–232 was injected into the cisterna magna, 33 % of the T cells migrated into the deep parenchyma, while this was the case for only 7.5 % of the T cells when the irrelevant AQP4236–277 peptide has been used (data not shown). Moreover, 25 % of the CD3+ T cells found in the deep parenchyma had an activated phenotype, as evidenced by the expression of PCNA (Fig. 7). The most likely source of these activated T cells are the AQP4207–232-specific T cells which had been injected into these animals to induce EAE, since we did not see PCNA expression in naïve T cells cultured without any antigen, or cultured in the presence of AQP4207–232 and splenic antigen presenting cells (Fig. 7).
Hence, T cell activation and subsequent parenchymal infiltration by AQP4207–232-peptide-specific T cells can be increased by injections of AQP4207–232 into the cisterna magna.

In the presence of NMO-IgG, the numbers of pathogenic T cells determine location and size of astrocyte-destructive lesions

We have shown above that AQP4268–285-specific T cells are sufficiently activated to immigrate the CNS parenchyma. Are they also sufficiently activated to allow the formation of astrocyte-destructive lesions in the presence of NMO-IgG? To address this question, we challenged Lewis rats with AQP4268–285-specific T cells and NMO-IgG, and studied their spinal cords, optic nerves, and brains by immunohistochemistry. When 3 × 106 AQP4268–285-specific T cells were used in combination with 10 mg NMO-IgG, the animals showed loss of tail tonus (EAE score 1) at the day of sacrifice (suppl. fig 6), and lesions exhibiting loss of both AQP4 and GFAP reactivity almost exclusively confined to the spinal cords (54/55 lesions). These lesions were most frequently found in thoracic cord gray matter (51.8 %) than in cervical (22.2 %) or lumbar/sacral cord gray matter (18.5 %) (Fig. 8), reaching sizes up to 47,105 µm2. Astrocyte-destructive lesions were not seen in the optic nerves, and were only detected once in the dorsal medulla (1/55 lesions) (Fig. 8, suppl. fig 8). The outcome was different, when 2 × 107 AQP4268–285-specific T cells were used together with 10 mg NMO-IgG. Then, lesions with AQP4 loss were not only seen in the spinal cord, reaching sizes up to 26,274 µm2, but also in the brain (Fig. 9). The optic nerves contained T cell infiltrates and activated microglia/macrophages, but did not show any evidence of AQP4 loss (suppl. fig 8). Lesions with AQP4 loss were also absent from optic nerves of 3-week-old rats injected with AQP4268–285-specific T cells and NMO-IgG (data not shown).
Hence, in the presence of NMO-IgG, low numbers of AQP4268–285-specific T cells initiate astrocyte-destructive lesions almost exclusively in spinal cord gray matter, while higher numbers of AQP4268–285-specific T cells trigger additional lesions with AQP4 and GFAP loss in the brain.

Discussion

Since the discoveries of T cells in NMO lesions [21], and of NMO-IgG containing pathogenic AQP4-specific antibodies with the T cell-dependent immunoglobulin subclass IgG1 [19, 20], the question arose whether AQP4 specific T cells are only responsible for T cell help in antibody formation, or whether they can also participate in directing lesions to NMO-typical sites, and induce there astrocyte-destructive lesions in the presence of NMO-IgG. And indeed, all AQP4-specific T cells established so far from wildtype animals were only weakly encephalitogenic, targeted the wrong sites, and were essentially confined to the meninges, but only poorly infiltrated the CNS parenchyma [32]. To finally answer these questions, we used one model organism—Lewis rats—to raise T cell lines against all AQP4 epitopes predicted to bind to the MHC class II products (RT1.BL) of these animals [40], and to test the encephalitogenic potential of these cells in vivo. While all of these peptides could serve as antigens to produce stable CD4+ TH1 cell lines and were fully able to activate T cells in vitro, the vast majority of the resulting AQP4 peptide-specific T cells were only weakly or moderately encephalitogenic. Such cells had to be transferred in very high numbers into naïve animals, did not yield any clinical symptoms, and barely infiltrated the CNS parenchyma. However, one of the peptides used, i.e. AQP4268–285, was clearly different. It contains two overlapping epitopes (AQP4271–279 with the amino acid sequence QQTKGSYME, and AQP4273–281 with the amino acid sequence TKGSYMEVE), is the autoantigen of highly encephalitogenic T cell responses in Lewis rats, and gives rise to T cells which induce clinical symptoms, deeply immigrate the CNS parenchyma, and initiate large astrocyte-destructive lesions in the presence of NMO-IgG.
What did we learn from these AQP4268–285-specific T cells, and how close do we come to an animal model for NMO when we use them to induce CNS inflammation in combination with pathogenic NMO-IgG in Lewis rats?
1.
AQP4268–285-specific CD4+ T cells are found in the normal healthy immune repertoire, can be readily activated upon immunization, and induce severe panencephalitis upon injection into naïve rats. Hence, AQP4268–285 is a true self-antigen in Lewis rats.
 
2.
AQP4268–285-specific T cells can immigrate into the CNS parenchyma throughout the entire neuraxis, but are particularly frequent at sites described to be typical for NMO [31]: AQP4268–285-specific T cells cause myelitis with a strong involvement of the dorsal horns and central gray matter, optic neuritis, and encephalitis with profound infiltration around the 3rd and 4th ventricle and in the hippocampus, in the periaqueductal gray, in the cerebellum and in the medulla. These sites are also lesion sites in NMO patients with brain involvement [30, 43], which has initially been ascribed to the high AQP4 expression at these sites [31]. However, since we also see inflammation at these sites, when animals have been challenged with other CNS-antigen-specific T cells, this distribution of brain lesions is more likely to reflect the local action of other regulatory mechanisms for T cells and antibody-mediated processes.
 
3.
AQP4268–285-specific T cells yield inflammatory lesions in which ~44 % of the deeply infiltrating T cells express PCNA as a sign of recent activation. Again, this is a crucial point, since T cell activation within the CNS is an important prerequisite for the formation of astrocyte-destructive lesions in the presence of NMO-IgG lesions [33], and since activated CD4+ T cells are found in NMO lesions [33]. It is tempting to speculate that this high level of PCNA expression might be due to reactivation within the CNS and not due to activation in vitro before transfer. A supportive, but certainly not definitive evidence for this speculation is the low number of proliferating AQP4207–232-specific T cells in the CNS, which is massively increased by additional injection of the respective peptide into the CSF. Unfortunately, however, we cannot formally prove whether or not antigen presenting cells in the CNS process antigens in a similar way as their counterparts in vitro do, and whether full-length AQP4 is cleaved by these cells to AQP4207–232 as efficiently in the CNS as it is in vitro.
 
4.
High numbers of AQP4268–285-specific T cells target astrocyte-destructive lesions throughout the entire spinal cord, and also to the brain in NMO-IgG seropositive hosts. However, when present in low numbers, AQP4268–285-specific T cells target 98 % of all astrocyte-destructive lesions to cervical/thoracic spinal cord gray matter in NMO-IgG seropositive hosts. The preference of spinal cord is important, since NMO often presents with episodes of myelitis. Targeting of this site could result from higher levels of expression of AQP4 mRNA, protein, and large supramolecular aggregates in spinal cord and optic nerve relative to other regions of the brain [22], and in gray relative to white matter cord [7]. Higher antigen concentrations might then translate to better binding of NMO-IgG, to an enhanced availability of this antigen for local antigen presenting cells and subsequent T cell activation [24], and to an increased astrocytotoxicity of microglia/macrophages via complement- and antibody-mediated cellular mechanisms [33, 34].
In combination with NMO-IgG, AQP4268–285-specific T cells also show a predilection for cervical/thoracic spinal cord, which are sites most often affected in NMO patients. Since this area is also most frequently targeted in the spinal cords of Lewis (LEW), LEW.1 N and LEW.1A rats with EAE provoked by the action of myelin oligodendrocyte glycoprotein (MOG)-specific antibodies and T cells [41], this might point to a gateway for autoreactive TH1 cells and antibodies to cross the blood–brain barrier at this site, possibly defined by regional neural activation [5].
We do not know yet why in our current NMO model the optic nerves are spared from astrocyte-destructive lesions, although they contain numerous inflammatory T cells and activated microglia/macrophages. Most trivially, the formation of NMO-like lesions in the optic nerve could just simply be a rare event in ENMO provoked by AQP4268–285-specific T cells and NMO-IgG, and could become visible when higher numbers of animals are examined. Alternatively, also the genetic background of our animals might play a role, since there is, again, a striking resemblance to the MOG-induced EAE model described above. In the MOG-model, demyelinating spinal cord lesions formed in LEW.1 N, LEW.1A, LEW.1AV1, BN, and DA rats, but additionally in the optic nerves only in BN and DA rats [35]. This contribution of genetic background to disease phenotype could find its human correlate in the different, ethnicity-dependent frequencies of longitudinally extensive transverse myelitis seen at onset attack in 53 % of Caucasian vs. 33 % of Afro-Caribbean patients in an UK cohort of NMO patients [16].
 
5.
Also low numbers of AQP4268–285-specific T cells can initiate large lesions with AQP4 loss. These findings recapitulate observations which have been made earlier in an EAE model using myelin basic protein-specific T cells with demyelinating anti-MOG antibodies [17], and suggest that the large, astrocyte-destructive lesions in NMO-IgG seropositive NMO patients could be provoked by the action of very few AQP4-specific T cells.
In Lewis rats, AQP4268–285 is highly encephalitogenic. For the time being, we do not know yet whether intracellular AQP4 fragments also contain highly encephalitogenic antigens in humans, since their MHC could select different epitopes. Does this mean that AQP4-specific T cells recognizing weakly encephalitogenic AQP4 epitopes are irrelevant for NMO in Lewis rats or NMO patients? Probably not. While certain antigenic fragments might not be present in sufficient amounts to warrant T cell infiltration into the intact CNS, i.e. to trigger the very first NMO lesion, they might play a crucial role in the propagation of relapses, for example when antigens are released from necrotic, astrocyte-destructive lesions. Then, the liberated antigens might become available for local antigen presenting cells [13, 26], and provide the basis for the activation of naive T cells within the CNS in a process called epitope spreading [17]. At least in EAE and MS, this process underlies the shift of autoreactivity from primary initiating self-determinants, which invariably regress with time and might even become undetectable during periods of disease progression, to sustained secondary autoreactivity [37]. Considering the fact that AQP4 contains a large number of potential T cell epitopes, not only in mice and rats [12, 25, 32], but also in humans [6, 23] (Fig. 1), it is tempting to speculate that this might be a strong argument in favor of very early T cell vaccination, and a strong counter-argument for later T cell vaccination as a therapeutic option for NMO patients.
 

Acknowledgments

This work was supported by the Austrian Science Fund [Grant Numbers P25240-B24 to MB and I916-B13 (International Programme, Eugène Devic European Network) to HL and MR], by the Else Kröner-Fresenius-Stiftung (Grant Number 2013_A283 to MB), by the Austrian Ministery of Science, Research and Economy (BIGWIG-MS to HL and MR), and by Grants-in-aid for Scientific Research of the Ministry of Education, Culture, Sports, Science and Technology of Japan to KF and TM. We thank Dr. Dagoberto Callegaro for sample collection, Marianne Leisser, Ulrike Köck and Angela Kury for excellent technical assistance, Verena Berg and Madhura Modak for assistance at the β-counter, and the Core Facility Flow Cytometry, Medical University of Vienna.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Abbott NJ (2004) Evidence for bulk flow of brain interstitial fluid: significance for physiology and pathology. Neurochem Int 45:545–552CrossRefPubMed Abbott NJ (2004) Evidence for bulk flow of brain interstitial fluid: significance for physiology and pathology. Neurochem Int 45:545–552CrossRefPubMed
2.
Zurück zum Zitat Aboul-Enein F, Bauer J, Klein M, Schubart A, Flugel A, Ritter T, Kawakami N, Siedler F, Linington C, Wekerle H, Lassmann H, Bradl M (2004) Selective and antigen-dependent effects of myelin degeneration on central nervous system inflammation. J Neuropathol Exp Neurol 63:1284–1296PubMed Aboul-Enein F, Bauer J, Klein M, Schubart A, Flugel A, Ritter T, Kawakami N, Siedler F, Linington C, Wekerle H, Lassmann H, Bradl M (2004) Selective and antigen-dependent effects of myelin degeneration on central nervous system inflammation. J Neuropathol Exp Neurol 63:1284–1296PubMed
3.
Zurück zum Zitat Amor S, Groome N, Linington C, Morris MM, Dornmair K, Gardinier MV, Matthieu J-M, Baker D (1994) Identification of epitopes of myelin oligodendrocyte glycoprotein for the induction of experimental allergic encephalomyelitis in SJL and Biozzi AB/H mice. J Immunol 153:4349–4356PubMed Amor S, Groome N, Linington C, Morris MM, Dornmair K, Gardinier MV, Matthieu J-M, Baker D (1994) Identification of epitopes of myelin oligodendrocyte glycoprotein for the induction of experimental allergic encephalomyelitis in SJL and Biozzi AB/H mice. J Immunol 153:4349–4356PubMed
4.
Zurück zum Zitat Arellano B, Hussain R, Zacharias T, Yoon J, David C, Zein S, Steinman L, Forsthuber T, Greenberg BM, Lambracht-Washington D, Ritchie AM, Bennett JL, Stuve O (2012) Human aquaporin 4281–300 is the immunodominant linear determinant in the context of HLA-DRB1*03:01: relevance for diagnosing and monitoring patients with neuromyelitis optica. Arch Neurol 69:1125–1131CrossRefPubMed Arellano B, Hussain R, Zacharias T, Yoon J, David C, Zein S, Steinman L, Forsthuber T, Greenberg BM, Lambracht-Washington D, Ritchie AM, Bennett JL, Stuve O (2012) Human aquaporin 4281–300 is the immunodominant linear determinant in the context of HLA-DRB1*03:01: relevance for diagnosing and monitoring patients with neuromyelitis optica. Arch Neurol 69:1125–1131CrossRefPubMed
5.
Zurück zum Zitat Arima Y, Harada M, Kamimura D, Park JH, Kawano F, Yull FE, Kawamoto T, Iwakura Y, Betz UA, Marquez G, Blackwell TS, Ohira Y, Hirano T, Murakami M (2012) Regional neural activation defines a gateway for autoreactive T cells to cross the blood–brain barrier. Cell 148:447–457CrossRefPubMed Arima Y, Harada M, Kamimura D, Park JH, Kawano F, Yull FE, Kawamoto T, Iwakura Y, Betz UA, Marquez G, Blackwell TS, Ohira Y, Hirano T, Murakami M (2012) Regional neural activation defines a gateway for autoreactive T cells to cross the blood–brain barrier. Cell 148:447–457CrossRefPubMed
6.
Zurück zum Zitat Bennett JL, Lam C, Kalluri SR, Saikali P, Bautista K, Dupree C, Glogowska M, Case D, Antel JP, Owens GP, Gilden D, Nessler S, Stadelmann C, Hemmer B (2009) Intrathecal pathogenic anti-aquaporin-4 antibodies in early neuromyelitis optica. Ann Neurol 66:617–629PubMedCentralCrossRefPubMed Bennett JL, Lam C, Kalluri SR, Saikali P, Bautista K, Dupree C, Glogowska M, Case D, Antel JP, Owens GP, Gilden D, Nessler S, Stadelmann C, Hemmer B (2009) Intrathecal pathogenic anti-aquaporin-4 antibodies in early neuromyelitis optica. Ann Neurol 66:617–629PubMedCentralCrossRefPubMed
7.
Zurück zum Zitat Bradl M, Misu T, Takahashi T, Watanabe M, Mader S, Reindl M, Adzemovic M, Bauer J, Berger T, Fujihara K, Itoyama Y, Lassmann H (2009) Neuromyelitis optica: pathogenicity of patient immunoglobulin in vivo. Ann Neurol 66:630–643CrossRefPubMed Bradl M, Misu T, Takahashi T, Watanabe M, Mader S, Reindl M, Adzemovic M, Bauer J, Berger T, Fujihara K, Itoyama Y, Lassmann H (2009) Neuromyelitis optica: pathogenicity of patient immunoglobulin in vivo. Ann Neurol 66:630–643CrossRefPubMed
8.
Zurück zum Zitat Fujihara K, Misu T, Nakashima I, Takahashi T, Bradl M, Lassmann H, Takano R, Nishiyama S, Takai Y, Suzuki C, Sato D, Kuroda H, Nakamura M, Fujimori J, Narikawa K, Sato S, Itoyama Y, Aoki M (2012) Neuromyelitis optica should be classified as an astrocytopathic disease rather than a demyelinating disease. Clin Exp Neuroimmunol 3:58–73CrossRef Fujihara K, Misu T, Nakashima I, Takahashi T, Bradl M, Lassmann H, Takano R, Nishiyama S, Takai Y, Suzuki C, Sato D, Kuroda H, Nakamura M, Fujimori J, Narikawa K, Sato S, Itoyama Y, Aoki M (2012) Neuromyelitis optica should be classified as an astrocytopathic disease rather than a demyelinating disease. Clin Exp Neuroimmunol 3:58–73CrossRef
9.
Zurück zum Zitat Hochmeister S, Zeitelhofer M, Bauer J, Nicolussi EM, Fischer MT, Heinke B, Selzer E, Lassmann H, Bradl M (2008) After injection into the striatum, in vitro-differentiated microglia- and bone marrow-derived dendritic cells can leave the central nervous system via the blood stream. Am J Pathol 173:1669–1681PubMedCentralCrossRefPubMed Hochmeister S, Zeitelhofer M, Bauer J, Nicolussi EM, Fischer MT, Heinke B, Selzer E, Lassmann H, Bradl M (2008) After injection into the striatum, in vitro-differentiated microglia- and bone marrow-derived dendritic cells can leave the central nervous system via the blood stream. Am J Pathol 173:1669–1681PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Jones MV, Huang H, Calabresi PA (2015) Levy M (2015) Pathogenic aquaporin-4 reactive T cells are sufficient to induce mouse model of neuromyelitis optica. Acta Neuropathol Comm 3:28CrossRef Jones MV, Huang H, Calabresi PA (2015) Levy M (2015) Pathogenic aquaporin-4 reactive T cells are sufficient to induce mouse model of neuromyelitis optica. Acta Neuropathol Comm 3:28CrossRef
11.
Zurück zum Zitat Kaab G, Brandl G, Marx A, Wekerle H, Bradl M (1996) The myelin basic protein-specific t cell repertoire in (transgenic) lewis rat/scid mouse chimeras: preferential v beta 8.2 T cell receptor usage depends on an intact lewis thymic microenvironment. Eur J Immunol 26:981–988CrossRefPubMed Kaab G, Brandl G, Marx A, Wekerle H, Bradl M (1996) The myelin basic protein-specific t cell repertoire in (transgenic) lewis rat/scid mouse chimeras: preferential v beta 8.2 T cell receptor usage depends on an intact lewis thymic microenvironment. Eur J Immunol 26:981–988CrossRefPubMed
12.
Zurück zum Zitat Kalluri SR, Rothhammer V, Staszewski O, Srivastava R, Petermann F, Prinz M, Hemmer B, Korn T (2011) Functional characterization of aquaporin-4 specific t cells: towards a model for neuromyelitis optica. PLoS ONE 6:e16083PubMedCentralCrossRefPubMed Kalluri SR, Rothhammer V, Staszewski O, Srivastava R, Petermann F, Prinz M, Hemmer B, Korn T (2011) Functional characterization of aquaporin-4 specific t cells: towards a model for neuromyelitis optica. PLoS ONE 6:e16083PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Kawakami N, Lassmann S, Li Z, Odoardi F, Ritter T, Ziemssen T, Klinkert WE, Ellwart JW, Bradl M, Krivacic K, Lassmann H, Ransohoff RM, Volk HD, Wekerle H, Linington C, Flugel A (2004) The activation status of neuroantigen-specific t cells in the target organ determines the clinical outcome of autoimmune encephalomyelitis. J Exp Med 199:185–197PubMedCentralCrossRefPubMed Kawakami N, Lassmann S, Li Z, Odoardi F, Ritter T, Ziemssen T, Klinkert WE, Ellwart JW, Bradl M, Krivacic K, Lassmann H, Ransohoff RM, Volk HD, Wekerle H, Linington C, Flugel A (2004) The activation status of neuroantigen-specific t cells in the target organ determines the clinical outcome of autoimmune encephalomyelitis. J Exp Med 199:185–197PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Kinoshita M, Nakatsuji Y, Kimura T, Moriya M, Takata K, Okuno T, Kumanogoh A, Kajiyama K, Yoshikawa H, Sakoda S (2010) Anti-aquaporin-4 antibody induces astrocytic cytotoxicity in the absence of cns antigen-specific t cells. Biochem Biophys Res Comm 394:205–210CrossRefPubMed Kinoshita M, Nakatsuji Y, Kimura T, Moriya M, Takata K, Okuno T, Kumanogoh A, Kajiyama K, Yoshikawa H, Sakoda S (2010) Anti-aquaporin-4 antibody induces astrocytic cytotoxicity in the absence of cns antigen-specific t cells. Biochem Biophys Res Comm 394:205–210CrossRefPubMed
15.
Zurück zum Zitat Kitic M, Hochmeister S, Wimmer I, Bauer J, Misu T, Mader S, Reindl M, Fujihara K, Lassmann H, Bradl M (2013) Intrastriatal injection of interleukin 1 beta triggers the formation of neuromyelitis optica-like lesions in nmo-igg seropositive rats. Acta Neuropathol Comm 1:1CrossRef Kitic M, Hochmeister S, Wimmer I, Bauer J, Misu T, Mader S, Reindl M, Fujihara K, Lassmann H, Bradl M (2013) Intrastriatal injection of interleukin 1 beta triggers the formation of neuromyelitis optica-like lesions in nmo-igg seropositive rats. Acta Neuropathol Comm 1:1CrossRef
16.
Zurück zum Zitat Kitley J, Leite MI, Nakashima I, Waters P, McNeillis B, Brown R, Takai Y, Takahashi T, Misu T, Elsone L, Woodhall M, George J, Boggild M, Vincent A, Jacob A, Fujihara K, Palace J (2012) Prognostic factors and disease course in aquaporin-4 antibody-positive patients with neuromyelitis optica spectrum disorder from the United Kingdom and Japan. Brain 135:1834–1849CrossRefPubMed Kitley J, Leite MI, Nakashima I, Waters P, McNeillis B, Brown R, Takai Y, Takahashi T, Misu T, Elsone L, Woodhall M, George J, Boggild M, Vincent A, Jacob A, Fujihara K, Palace J (2012) Prognostic factors and disease course in aquaporin-4 antibody-positive patients with neuromyelitis optica spectrum disorder from the United Kingdom and Japan. Brain 135:1834–1849CrossRefPubMed
17.
Zurück zum Zitat Lassmann H, Brunner C, Bradl M, Linington C (1988) Experimental allergic encephalomyelitis: the balance between encephalitogenic T lymphocytes and demyelinating antibodies determines size and structure of demyelinated lesions. Acta Neuropathol 75:566–576CrossRefPubMed Lassmann H, Brunner C, Bradl M, Linington C (1988) Experimental allergic encephalomyelitis: the balance between encephalitogenic T lymphocytes and demyelinating antibodies determines size and structure of demyelinated lesions. Acta Neuropathol 75:566–576CrossRefPubMed
18.
Zurück zum Zitat Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, Blank RB, Melan F, Siegel R, Hennighausen L, Shevach EM, O´Shea JJ (2007) Interleukin-2 signaling via STAT5 Constrains T helper 17 cell generaration. Immunity 26:371–381CrossRefPubMed Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, Blank RB, Melan F, Siegel R, Hennighausen L, Shevach EM, O´Shea JJ (2007) Interleukin-2 signaling via STAT5 Constrains T helper 17 cell generaration. Immunity 26:371–381CrossRefPubMed
19.
Zurück zum Zitat Lennon VA, Wingerchuk DM, Kryzer TJ, Pittock SJ, Lucchinetti CF, Fujihara K, Nakashima I, Weinshenker BG (2004) A serum autoantibody marker of neuromyelitis optica: distinction from multiple sclerosis. Lancet 364:2106–2112CrossRefPubMed Lennon VA, Wingerchuk DM, Kryzer TJ, Pittock SJ, Lucchinetti CF, Fujihara K, Nakashima I, Weinshenker BG (2004) A serum autoantibody marker of neuromyelitis optica: distinction from multiple sclerosis. Lancet 364:2106–2112CrossRefPubMed
20.
Zurück zum Zitat Lennon VA, Kryzer TJ, Pittock SJ, Verkman AS, Hinson SR (2005) IgG marker of optic-spinal multiple sclerosis binds to the aquaporin-4 water channel. J Exp Med 202:473–477PubMedCentralCrossRefPubMed Lennon VA, Kryzer TJ, Pittock SJ, Verkman AS, Hinson SR (2005) IgG marker of optic-spinal multiple sclerosis binds to the aquaporin-4 water channel. J Exp Med 202:473–477PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Lucchinetti CF, Mandler RN, McGavern D, Bruck W, Gleich G, Ransohoff RM, Trebst C, Weinshenker B, Wingerchuk D, Parisi JE, Lassmann H (2002) A role for humoral mechanisms in the pathogenesis of Devic’s neuromyelitis optica. Brain 125:1450–1461CrossRefPubMed Lucchinetti CF, Mandler RN, McGavern D, Bruck W, Gleich G, Ransohoff RM, Trebst C, Weinshenker B, Wingerchuk D, Parisi JE, Lassmann H (2002) A role for humoral mechanisms in the pathogenesis of Devic’s neuromyelitis optica. Brain 125:1450–1461CrossRefPubMed
22.
Zurück zum Zitat Matiello M, Schaefer-Klein J, Sun D, Weinshenker BG (2013) Aquaporin 4 expression and tissue susceptibility to neuromyelitis optica. JAMA Neurology 70:1118–1125CrossRefPubMed Matiello M, Schaefer-Klein J, Sun D, Weinshenker BG (2013) Aquaporin 4 expression and tissue susceptibility to neuromyelitis optica. JAMA Neurology 70:1118–1125CrossRefPubMed
23.
Zurück zum Zitat Matsuya N, Komori M, Nomura K, Nakane S, Fukudome T, Goto H, Shiraishi H, Wandinger KP, Matsuo H, Kondo T (2011) Increased T-cell immunity against aquaporin-4 and proteolipid protein in neuromyelitis optica. Int Immunol 23:565–573CrossRefPubMed Matsuya N, Komori M, Nomura K, Nakane S, Fukudome T, Goto H, Shiraishi H, Wandinger KP, Matsuo H, Kondo T (2011) Increased T-cell immunity against aquaporin-4 and proteolipid protein in neuromyelitis optica. Int Immunol 23:565–573CrossRefPubMed
24.
Zurück zum Zitat Mues M, Bartholomaus I, Thestrup T, Griesbeck O, Wekerle H, Kawakami N, Krishnamoorthy G (2013) Real-time in vivo analysis of t cell activation in the central nervous system using a genetically encoded calcium indicator. Nat Med 19:778–783CrossRefPubMed Mues M, Bartholomaus I, Thestrup T, Griesbeck O, Wekerle H, Kawakami N, Krishnamoorthy G (2013) Real-time in vivo analysis of t cell activation in the central nervous system using a genetically encoded calcium indicator. Nat Med 19:778–783CrossRefPubMed
25.
Zurück zum Zitat Nelson PA, Khodadoust M, Prodhomme T, Spencer C, Patarroyo JC, Varrin-Doyer M, Ho JD, Stroud RM, Zamvil SS (2010) Immunodominant t cell determinants of aquaporin-4, the autoantigen associated with neuromyelitis optica. PLoS ONE 5:e15050PubMedCentralCrossRefPubMed Nelson PA, Khodadoust M, Prodhomme T, Spencer C, Patarroyo JC, Varrin-Doyer M, Ho JD, Stroud RM, Zamvil SS (2010) Immunodominant t cell determinants of aquaporin-4, the autoantigen associated with neuromyelitis optica. PLoS ONE 5:e15050PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Odoardi F, Kawakami N, Klinkert WE, Wekerle H, Flugel A (2007) Blood-borne soluble protein antigen intensifies T cell activation in autoimmune cns lesions and exacerbates clinical disease. Proc Natl Acad Sci (USA) 104:18625–18630CrossRef Odoardi F, Kawakami N, Klinkert WE, Wekerle H, Flugel A (2007) Blood-borne soluble protein antigen intensifies T cell activation in autoimmune cns lesions and exacerbates clinical disease. Proc Natl Acad Sci (USA) 104:18625–18630CrossRef
28.
Zurück zum Zitat Paxinos G, Watson C (1998) The rat brain in stereotaxic coordinates, 4th edn. Academic Press, San Diego Paxinos G, Watson C (1998) The rat brain in stereotaxic coordinates, 4th edn. Academic Press, San Diego
29.
Zurück zum Zitat Piddlesden SJ, Lassmann H, Zimprich F, Morgan BP, Linington C (1993) The demyelinating potential of antibodies to myelin oligodendrocyte glycoprotein is related to their ability to fix complement. Am J Pathol 143:555–564PubMedCentralPubMed Piddlesden SJ, Lassmann H, Zimprich F, Morgan BP, Linington C (1993) The demyelinating potential of antibodies to myelin oligodendrocyte glycoprotein is related to their ability to fix complement. Am J Pathol 143:555–564PubMedCentralPubMed
30.
Zurück zum Zitat Pittock SJ, Lennon VA, Krecke K, Wingerchuk DM, Lucchinetti CF, Weinshenker BG (2006) Brain abnormalities in neuromyelitis optica. Arch Neurol 63:390–396CrossRefPubMed Pittock SJ, Lennon VA, Krecke K, Wingerchuk DM, Lucchinetti CF, Weinshenker BG (2006) Brain abnormalities in neuromyelitis optica. Arch Neurol 63:390–396CrossRefPubMed
31.
Zurück zum Zitat Pittock SJ, Weinshenker BG, Lucchinetti CF, Wingerchuk DM, Corboy JR, Lennon VA (2006) Neuromyelitis optica brain lesions localized at sites of high aquaporin 4 expression. Arch Neurol 63:964–968CrossRefPubMed Pittock SJ, Weinshenker BG, Lucchinetti CF, Wingerchuk DM, Corboy JR, Lennon VA (2006) Neuromyelitis optica brain lesions localized at sites of high aquaporin 4 expression. Arch Neurol 63:964–968CrossRefPubMed
32.
Zurück zum Zitat Pohl M, Fischer MT, Mader S, Schanda K, Kitic M, Sharma R, Wimmer I, Misu T, Fujihara K, Reindl M, Lassmann H, Bradl M (2011) Pathogenic T cell responses against aquaporin 4. Acta Neuropathol 122:21–34PubMedCentralCrossRefPubMed Pohl M, Fischer MT, Mader S, Schanda K, Kitic M, Sharma R, Wimmer I, Misu T, Fujihara K, Reindl M, Lassmann H, Bradl M (2011) Pathogenic T cell responses against aquaporin 4. Acta Neuropathol 122:21–34PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Pohl M, Kawakami N, Kitic M, Bauer J, Martins R, Fischer MT, Machado-Santos J, Mader S, Ellwart JW, Misu T, Fujihara K, Wekerle H, Reindl M, Lassmann H, Bradl M (2013) T cell-activation in neuromyelitis optica lesions plays a role in their formation. Acta Neuropathol Commun 1:85PubMedCentralCrossRefPubMed Pohl M, Kawakami N, Kitic M, Bauer J, Martins R, Fischer MT, Machado-Santos J, Mader S, Ellwart JW, Misu T, Fujihara K, Wekerle H, Reindl M, Lassmann H, Bradl M (2013) T cell-activation in neuromyelitis optica lesions plays a role in their formation. Acta Neuropathol Commun 1:85PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Ratelade J, Asavapanumas N, Ritchie AM, Wemlinger S, Bennett JL, Verkman AS (2013) Involvement of antibody-dependent cell-mediated cytotoxicity in inflammatory demyelination in a mouse model of neuromyelitis optica. Acta Neuropathol 126:699–709PubMedCentralCrossRefPubMed Ratelade J, Asavapanumas N, Ritchie AM, Wemlinger S, Bennett JL, Verkman AS (2013) Involvement of antibody-dependent cell-mediated cytotoxicity in inflammatory demyelination in a mouse model of neuromyelitis optica. Acta Neuropathol 126:699–709PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Storch MK, Stefferl A, Brehm U, Weissert R, Wallstrom E, Kerschensteiner M, Olsson T, Linington C, Lassmann H (1998) Autoimmunity to myelin oligodendrocyte glycoprotein in rats mimics the spectrum of multiple sclerosis pathology. Brain Pathol 8:681–694CrossRefPubMed Storch MK, Stefferl A, Brehm U, Weissert R, Wallstrom E, Kerschensteiner M, Olsson T, Linington C, Lassmann H (1998) Autoimmunity to myelin oligodendrocyte glycoprotein in rats mimics the spectrum of multiple sclerosis pathology. Brain Pathol 8:681–694CrossRefPubMed
36.
Zurück zum Zitat Takano R, Misu T, Takahashi T, Sato S, Fujihara K, Itoyama Y (2010) Astrocytic damage is far more severe than demyelination in nmo: a clinical csf biomarker study. Neurology 75:208–216CrossRefPubMed Takano R, Misu T, Takahashi T, Sato S, Fujihara K, Itoyama Y (2010) Astrocytic damage is far more severe than demyelination in nmo: a clinical csf biomarker study. Neurology 75:208–216CrossRefPubMed
37.
Zurück zum Zitat Tuohy VK, Yu M, Yin L, Kawczak JA, Kinkel RP (1999) Spontaneous regression of primary autoreactivity during chronic progression of experimental autoimmune encephalomyelitis and multiple sclerosis. J Exp Med 189:1033–1042PubMedCentralCrossRefPubMed Tuohy VK, Yu M, Yin L, Kawczak JA, Kinkel RP (1999) Spontaneous regression of primary autoreactivity during chronic progression of experimental autoimmune encephalomyelitis and multiple sclerosis. J Exp Med 189:1033–1042PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Varrin-Doyer M, Spencer CM, Schulze-Topphoff U, Nelson PA, Stroud RM, Cree BA, Zamvil SS (2012) Aquaporin 4-specific t cells in neuromyelitis optica exhibit a th17 bias and recognize clostridium abc transporter. Ann Neurol 72:53–64PubMedCentralCrossRefPubMed Varrin-Doyer M, Spencer CM, Schulze-Topphoff U, Nelson PA, Stroud RM, Cree BA, Zamvil SS (2012) Aquaporin 4-specific t cells in neuromyelitis optica exhibit a th17 bias and recognize clostridium abc transporter. Ann Neurol 72:53–64PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Verkman AS, Ratelade J, Rossi A, Zhang H, Tradtrantip L (2011) Aquaporin-4: orthogonal array assembly, CNS functions, and role in neuromyelitis optica. Acta Pharmacol Sin 32:702–710PubMedCentralCrossRefPubMed Verkman AS, Ratelade J, Rossi A, Zhang H, Tradtrantip L (2011) Aquaporin-4: orthogonal array assembly, CNS functions, and role in neuromyelitis optica. Acta Pharmacol Sin 32:702–710PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Wauben MH, van der Kraan M, Grosfeld-Stulemeyer MC, Joosten I (1997) Definition of an extended mhc class ii-peptide binding motif for the autoimmune disease-associated lewis rat rt1.Bl molecule. Int Immunol 9:281–290CrossRefPubMed Wauben MH, van der Kraan M, Grosfeld-Stulemeyer MC, Joosten I (1997) Definition of an extended mhc class ii-peptide binding motif for the autoimmune disease-associated lewis rat rt1.Bl molecule. Int Immunol 9:281–290CrossRefPubMed
41.
Zurück zum Zitat Weissert R, Wallstrom E, Storch MK, Stefferl A, Lorentzen J, Lassmann H, Linington C, Olsson T (1998) MHC haplotype-dependent regulation of Mog-induced EAE in rats. J Clin Invest 102:1265–1273PubMedCentralCrossRefPubMed Weissert R, Wallstrom E, Storch MK, Stefferl A, Lorentzen J, Lassmann H, Linington C, Olsson T (1998) MHC haplotype-dependent regulation of Mog-induced EAE in rats. J Clin Invest 102:1265–1273PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Weller RO (1998) Pathology of cerebrospinal fluid and interstitial fluid of the CNS: significance for alzheimer disease, prion disorders and multiple sclerosis. J Neuropathol Exp Neurol 57:885–894CrossRefPubMed Weller RO (1998) Pathology of cerebrospinal fluid and interstitial fluid of the CNS: significance for alzheimer disease, prion disorders and multiple sclerosis. J Neuropathol Exp Neurol 57:885–894CrossRefPubMed
43.
Zurück zum Zitat Wingerchuk DM, Lennon VA, Lucchinetti CF, Pittock SJ, Weinshenker BG (2007) The spectrum of neuromyelitis optica. Lancet Neurol 6:805–815CrossRefPubMed Wingerchuk DM, Lennon VA, Lucchinetti CF, Pittock SJ, Weinshenker BG (2007) The spectrum of neuromyelitis optica. Lancet Neurol 6:805–815CrossRefPubMed
Metadaten
Titel
Highly encephalitogenic aquaporin 4-specific T cells and NMO-IgG jointly orchestrate lesion location and tissue damage in the CNS
verfasst von
Bleranda Zeka
Maria Hastermann
Sonja Hochmeister
Nikolaus Kögl
Nathalie Kaufmann
Kathrin Schanda
Simone Mader
Tatsuro Misu
Paulus Rommer
Kazuo Fujihara
Zsolt Illes
Fritz Leutmezer
Douglas Kazutoshi Sato
Ichiro Nakashima
Markus Reindl
Hans Lassmann
Monika Bradl
Publikationsdatum
01.12.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Acta Neuropathologica / Ausgabe 6/2015
Print ISSN: 0001-6322
Elektronische ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-015-1501-5

Weitere Artikel der Ausgabe 6/2015

Acta Neuropathologica 6/2015 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.