Skip to main content
Erschienen in: Acta Neuropathologica 4/2017

Open Access 05.11.2016 | Original Paper

Differences in T cell cytotoxicity and cell death mechanisms between progressive multifocal leukoencephalopathy, herpes simplex virus encephalitis and cytomegalovirus encephalitis

verfasst von: Susanne Laukoter, Helmut Rauschka, Anna R. Tröscher, Ulrike Köck, Etsuji Saji, Kurt Jellinger, Hans Lassmann, Jan Bauer

Erschienen in: Acta Neuropathologica | Ausgabe 4/2017

Abstract

During the appearance of human immunodeficiency virus infection in the 1980 and the 1990s, progressive multifocal leukoencephalopathy (PML), a viral encephalitis induced by the JC virus, was the leading opportunistic brain infection. As a result of the use of modern immunomodulatory compounds such as Natalizumab and Rituximab, the number of patients with PML is once again increasing. Despite the presence of PML over decades, little is known regarding the mechanisms leading to death of infected cells and the role the immune system plays in this process. Here we compared the presence of inflammatory T cells and the targeting of infected cells by cytotoxic T cells in PML, herpes simplex virus encephalitis (HSVE) and cytomegalovirus encephalitis (CMVE). In addition, we analyzed cell death mechanisms in infected cells in these encephalitides. Our results show that large numbers of inflammatory cytotoxic T cells are present in PML lesions. Whereas in HSVE and CMVE, single or multiple appositions of CD8+ or granzyme-B+ T cells to infected cells are found, in PML such appositions are significantly less apparent. Analysis of apoptotic pathways by markers such as activated caspase-3, caspase-6, poly(ADP-ribose) polymerase-1 (PARP-1) and apoptosis-inducing factor (AIF) showed upregulation of caspase-3 and loss of caspase-6 from mitochondria in CMVE and HSVE infected cells. Infected oligodendrocytes in PML did not upregulate activated caspase-3 but instead showed translocation of PARP-1 from nucleus to cytoplasm and AIF from mitochondria to nucleus. These findings suggest that in HSVE and CMVE, cells die by caspase-mediated apoptosis induced by cytotoxic T cells. In PML, on the other hand, infected cells are not eliminated by the immune system but seem to die by virus-induced PARP and AIF translocation in a type of cell death defined as parthanatos.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00401-016-1642-1) contains supplementary material, which is available to authorized users.
S. Laukoter and H. Rauschka contributed equally.

Introduction

Progressive multifocal leukoencephalopathy (PML) is a demyelinating and often fatal disease caused by infection of oligodendrocytes and astrocytes in human brain [50]. It is thought to be initiated by immunosuppression-associated reactivation of latent JC virus and was first seen in immunosuppressed cancer patients and organ transplant recipients [2]. PML reached epidemic proportions as a major complication in acquired immune deficiency syndrome (AIDS) patients [9, 37]. In recent years, PML has been observed in patients treated with immunomodulatory compounds such as Natalizumab [26, 27, 29, 43] and Rituximab [17]. Generally, it is assumed that infected oligodendrocytes are eliminated by cytotoxic T lymphocytes. Indeed, JC-specific T cells can be detected in the blood of PML patients [31] and such cytotoxic CD8+ T cells have been found to enter the central nervous system (CNS) where they come into contact with JC-infected cells [48]. Nevertheless, at present there is no clear in situ evidence that these cytotoxic T cells are actively engaged in the elimination of infected cells.
Surprisingly little is known about the cause of oligodendrocyte cell death in PML. Some evidence of apoptosis of oligodendrocytes was shown in 2002 [41]. Others however demonstrated that infected oligodendrocytes start to express the anti-apoptotic factor surviving in order to delay apoptosis [40]. Since the first descriptions of programmed cell death mechanisms there has been a tendency to use only two categories; either cells die by apoptosis or by necrosis. At present it has become clear that there are many different pathways in which a large variety of molecules play a distinct role [24]. Besides the caspase-dependent apoptosis pathways, there is a caspase-independent pathway which involves the DNA damage-responsive enzyme poly(ADP-ribose) polymerase-1 (PARP-1). At the molecular level, expression of PARP-1 leads to formation and release of poly(ADP-ribose) (PAR) from the nucleus. In mitochondria, PAR induces release of apoptosis-inducing factor (AIF). AIF redirects from the mitochondria towards the nucleus and mediates chromatinolysis, resulting in so-called regulated necrosis [24]. This cell death pathway has been designated as parthanatos and has been suggested to play a role in a variety of pathophysiological conditions such as stroke and diabetes [18].
Here we analyzed cytotoxic T cell mechanisms and cell death pathways in (AIDS-associated) viral encephalitides PML and cytomegalovirus encephalitis (CMVE) as well as herpes simplex virus encephalitis (HSVE). Our findings suggest that in CMVE and HSVE, cells are eliminated by the immune system via a caspase-mediated pathway. In PML, infected oligodendrocytes, rather than being eliminated by cytotoxic T lymphocytes, undergo parthanatos possibly induced by the enhanced cellular activity involved in viral replication.

Materials and methods

Patients

This study was performed on paraffin-embedded, formalin-fixed archival material that was collected between 1982 and 1998 in the Department of Neurology at the Hospital Lainz in Vienna, Austria. The samples consisted of eight cases of AIDS-associated PML, seven cases of AIDS-associated CMVE, five cases of HSVE and five control (no neurological disease) brains. The histopathological diagnosis of these cases was established by an experienced neuropathologist (KJ). Demographic data of the individual patients are presented in Table 1.
Table 1
Demographic data of the patients
Sample number
Disease
Age
Male/female
Disease duration (week)
Sample area
Cause of death
12–93
PML (AIDS-assoc.)
45
Male
118
Lenticular nucleus
Pulmonary edema
47–93
PML (AIDS-assoc.)
42
Male
20
Pons
Pneumonia
97–93
PML (AIDS-assoc.)
50
Male
20
Parietal lobe right
Pulmonary edema
103–93
PML (AIDS-assoc.)
39
Male
15
Parietal lobe left
Acute respiratory insufficiency
278–93
PML (AIDS-assoc.)
29
Male
19
Parietal lobe left
Cachexia
291–93
PML (AIDS-assoc.)
49
Female
5
Parietal lobe right
Pneumonia
358–93
PML (AIDS-assoc.)
39
Male
27
Parietal lobe left
Pneumonia
359–93
PML (AIDS-assoc.)
34
Male
<18
Pons
Pneumonia
330–82
HSV
9
Female
1.5
Hippocampus
Acute respiratory insufficiency
67–83
HSV
40
Male
3.5
Hippocampus
Necrotic hemorrhagic pneumonia
204–83
HSV
61
Female
1.5
Hippocampus
Pneumonia
529–84
HSV
37
Male
2.3
Hippocampus
Pneumonia
237–91
HSV
76
Male
3.15
Hippocampus
Pneumonia
364–92
CMV (AIDS-assoc.)
40
Male
unknown
Parahippocampal region
Acute cardiovascular failure
455–92
CMV (AIDS-assoc.)
48
Male
<5
Parahippocampal region
Pneumonia, sepsis
559–92
CMV (AIDS-assoc.)
33
Male
20
Cerebellum
Pneumonia
234–93
CMV (AIDS-assoc.)
28
Male
11
Medulla oblongata
Pneumonia, sepsis
430–93
CMV (AIDS-assoc.)
35
Male
15
Medulla oblongata
Pneumonia
62–94
CMV (AIDS-assoc.)
32
Male
>4
Medulla oblongata
Pneumonia, sepsis
118–94
CMV (AIDS-assoc.)
55
Male
9
Hippocampus
Pneumonia
Control brains
50 (37–83)
3 Female
2 Male
Cortex
Non-neurological

Histopathology and immunohistochemistry

4 μm sections of paraffin-embedded specimens were routinely stained with hematoxylin and eosin (H&E) and luxol fast blue/periodic acid-Schiff (LFB-PAS) for myelin. Immunohistochemical stainings were performed according to previously published protocols [5, 11] using primary antibodies against inflammatory T cells [CD3, CD8, CD4 and Granzyme-B (GrB)] and other inflammatory cells such as CD20+ B cells and CD138+ plasma cells. For CD3, CD8, CD4 and GrB stainings, biotinylated tyramine enhancement was used as described previously [10]. For analysis of cell death pathways we used antibodies against activated caspase-3 (CM1), caspase-6, Poly(ADP-ribose) (PAR) and apoptosis-inducing factor (AIF). For detection of virus-infected cells we used monoclonal Pab2003 [12] and monoclonal and polyclonal anti-SV40 [15], an antibody against CMV antigens and antibodies recognizing HSV-1. Light microscopical double-stainings for viral proteins and GrB or cell death markers (caspase-3) were done with an alkaline-phosphatase system (development with Fast Blue) and a peroxidase system (development with 3,3′-Diaminobenzidine (DAB) or amino ethyl carbazole (AEC) as described in detail [7]. Details on the individual antibodies are listed in Supplementary Table 1 (online resource 1).

Confocal laser fluorescence microscopy

Fluorescence immunohistochemistry was performed on paraffin sections as previously described in detail [7]. For confocal fluorescent double-labeling or triple labeling with primary antibodies from different species, antibodies were applied simultaneously at 4 °C overnight. After washing with DAKO washing buffer (DakoCytomation, Glostrup, Denmark), secondary antibodies consisting of donkey-anti-mouse Cy3 or donkey-anti-mouse Dylight 546 (Jackson ImmunoResearch, 1:200), biotinylated donkey-anti-rabbit (Amersham Pharmacia Biotech; 1:200) and (in case of triple labeling) Cy5-conjugated donkey-anti-goat, were applied simultaneously for 1 h at room temperature, followed by application of streptavidin-Cy2 (Jackson ImmunoResearch; 1:75) for 1 h at room temperature. Fluorescent preparations were then stained with 4′,6′-diamidino-2-phenylindole (DAPI, Sigma) or TO-PRO-3 (Thermo Scientific, Waltham, MA, USA) as nuclear counterstain, embedded and examined using a confocal laser scan microscope (Leica SP5, Leica Mannheim, Germany) equipped with lasers for 504, 488, 543 and 633 nm excitation. Scanning for DAPI (504 nm), Cy2 (488 nm), Cy3 (543 nm), Cy5 (633 nm) or TO-PRO-3 (633 nm) was performed sequentially to rule out fluorescence bleed through.

Quantification of cells

Quantification of CD3+, CD8+, CD4+, GrB+, CD20+ B cells, CD138+ plasma cells and virus-infected SV40+, HSV+ or CMV+ cells was performed in 5.8 mm2 of tissue regions exhibiting infected cells and inflammation. Typically, in PML, such areas were on the active border of large confluent chronic lesions or in small active lesions. In CMVE cases, infected cells were mostly found paraventricular (four cases) in nuclei in medulla oblongata (three cases) or in the cerebellar molecular layer (one case). In order to quantitate appositions of CD8+ or GrB+ cells with virus-infected cells we used light microscopically double-labeled sections as described above. In each case we screened 500 infected cells for the presence of single (one CD8+ or GrB+ cell attached to a virus-infected cell) or multiple (two or more CD8+ or GrB+ cells in direct contact with an infected cell) appositions. CD8+ cells were assessed at 200-fold magnification while GrB+ cells were assessed at a 400-fold magnification to be able to see the small granular signal.
Quantification of TPPP/p25+ oligodendrocytes in white matter of control brains, PML, HSVE and CMVE was performed using an ocular morphometric grid covering an area of 0.25 mm2 at 200-fold magnification in 2 mm2 of tissue regions exhibiting infected cells and inflammation (lesions) and in 2 mm of the surrounding (normal appearing) white matter.
Quantification of the percentage of virus-infected cells with caspase-3 immunoreactivity or with nuclear AIF reactivity was performed on light microscopically double-labeled sections. In each case we analyzed 500 infected cells at a 400-fold magnification.

Statistics

Statistical analysis was performed with Graphpad Prism 6.0. For metric data, a two-sided Mann–Whitney U test; and for categorical data, a two-sided chi square or Fisher exact test was used as applicable. A p value of <0.05 was considered significant.

Results

Basic neuropathology

Progressive multifocal leucoencephalopathy

16 sections from 8 cases of PML were stained with LFB-PAS for detection of demyelinating lesions. Three cases contained one large lesion with a demyelinated core and with active demyelination on the border. Another two cases contained both large demyelinated lesions as well as multiple smaller demyelinating lesions. The remaining three cases had multiple smaller lesions (Fig. 1a). Quantification of oligodendrocytes in white matter showed a significant loss in and outside of PML lesions as compared to white matter of normal control brain (online resource 1, Suppl. Fig. 1). All cases were stained with Pab2003, an antibody recognizing early JCV T proteins [12]. Double-staining of Pab2003 with SV40 showed that most cells (64.5%) were double-labeled (Fig. 1b) while 6.5% were only positive for Pab2003 and 29% were only positive for SV40. Since the anti-SV40 antibody recognized more infected cells, we proceeded with this marker. In small demyelinating lesions SV40+ oligodendrocytes were located on the border (Fig. 1c). Unlike the larger lesions which had many (bizarre) astrocytes in the core of the lesion (Fig. 1d, e), these small lesions did not contain bizarre astrocytes. SV40+ oligodendrocytes, double-labeled with carbonic anhydrase II (CAII), typically had a round swollen nucleus and enlarged cytoplasm (Fig. 1f). Basic inflammation was analyzed by H&E staining showing lymphocytes in the perivascular space of blood vessels as well as infiltration in the parenchyma of the CNS. Demyelinating lesions also contained macrophages (Fig. 1d) with LFB+ and/or PAS+ myelin degradation products.

HSV encephalitis

Five sections from five cases showed acute haemorrhagic and subacute necrotizing lesions in both white and gray matter areas (Fig. 1g). Lesional sites in HSVE contained large numbers of inflammatory infiltrates with T lymphocytes and macrophages in meninges, the perivascular space of blood vessels and the CNS parenchyma. Numerous infected cells could be recognized by the presence of nuclear inclusions. The full amount of infected cells however only became clear after staining for HSV1, which showed HSV antigens in the nuclei and cytoplasm (Fig. 1h) of large numbers of infected astrocytes (Figs. 1i, 5b), cortical neurons (Fig. 1j) and oligodendrocytes (Fig. 1k). Like in PML, loss of oligodendrocytes in lesions was shown by quantification of stainings for TPPP/p25 (online resource 1, Suppl. Fig. 1).

CMV encephalitis

H&E sections showed the presence of large, often necrotic lesions with moderate infiltration of inflammatory lymphocytes (Fig. 1l). CMV-infected cells were recognized by their enlarged cytoplasm and characteristic “owl’s eye” morphology. Smaller infected cells in H&E sections were more difficult to detect but were easily found in sections stained with anti-CMV antibodies (Fig. 1m). Double-staining for CMV and GFAP showed that most of these cells were astrocytes (Fig. 1n) while oligodendrocytes were all negative for CMV (Fig. 1o). Nine sections from seven cases were studied for the presence of CMV positive cells. Almost all infected areas were found in gray matter. In four cases, groups of infected cells were found in a paraventricular position near the hippocampus. In addition, in these cases we found incidental single cells in cortical regions. Often these cells were observed in the midst of a microglial nodule. In one of these four cases a large haemorrhagic lesion was found in the hippocampus itself. In this patient we observed foci of CMV+ cells in cortical subpial positions. In one case, infected cells were present in small lesions of the molecular layer of the cerebellum (Fig. 1m) and in a large haemorrhagic lesion in the cerebellum. Such a large haemorrhagic lesion also was found in the medulla oblongata of a third patient. In one patient we only found single CMV+ cells dispersed over the medulla oblongata. Again, such cells were often found to be surrounded by microglial cells as part of a microglial nodule. Like in PML and HSVE we also quantified oligodendrocytes in the white matter but did not find any loss (online resource 1, Suppl. Fig. 1).

T cell cytotoxicity in virus-infected brains

To gain information on the role of inflammation in these encephalitides, we investigated these brains with various markers for inflammatory cells. Stainings for CD57 in PML, HSVE and CMVE demonstrated that NK cells were very rare and were mostly found in the perivascular space (online resource 1, Suppl. Fig. 2). CD20+ B cells and CD138+ plasma cells were more abundant than NK cells but less abundant than T cells (Fig. 3a). These B cells and plasma cells were mostly present in the perivascular space (Suppl. Fig. 2f and 2g). Stainings for CD3 (Fig. 2a–c) showed that T cells were present in meninges, perivascular space and deeply infiltrated in the parenchyma. Double-staining for CD8 and GrB showed that more than 95% of all GrB reactivity could be found in the CD8+ cells (online resource 1, Suppl. Fig. 2a). In all three forms of encephalitis we selected areas with prominent viral presence and quantified various T cell subsets in the parenchyma. Quantification revealed that in CMVE less CD3+ T cells were present than in PML and HSVE (Fig. 3a). In addition, quantification of CD4+ T Helper cells showed that their numbers were lower in CMVE and PML than in HSVE (Fig. 3a and Supp. Fig. 2D and E). The latter probably is a direct result from the elimination of CD4+ T cells by HIV in these AIDS-associated encephalitides. In all cases most of the infiltrating CD3+T cells also were CD8+ cells (Fig. 2d–f). In PML we found that 67% of the CD3 T cells were of the CD8 phenotype. In HSVE 58% of the CD3 T cells were CD8+, while in CMVE this was 89% (Fig. 3a). Granzyme-B+ T lymphocytes were much less frequent. In PML, 18% of CD3+ T cells were GrB+, while in HSVE and CMVE these cells reached 19 and 31% respectively (Fig. 3a). Previously, in Rasmussen Encephalitis and paraneoplastic encephalitis we showed the presence of CD8+ GrB+ cytotoxic T cells targeting neurons [10, 11]. Here we investigated whether targeting of virus-infected cells could be found. First, we quantified and calculated the ratio between virus-infected cells and CD8+ as well as the ratio between virus-infected cells and GrB+ cells. These ratios were about four times lower in CMVE than in PML, but, probably due to large interindividual variety, these differences were not significant (Fig. 3b, c). To reveal objective differences in the targeting of infected cells we quantified the appositions of single or multiple (2 or more) CD8+ (Fig. 3b) or GrB+ cells (Fig. 3c) to virus-infected cells. Surprisingly, we found that in PML both single (Fig. 2g) as well as multiple appositions of CD8+ (Figs. 2g, 3b) or GrB+ cells (Figs. 2j, 3c) were significantly less frequent than in CMVE. In addition, we also found that CMVE (Fig. 2i) contains more multiple CD8+ T cell appositions than HSVE (Figs. 2h, 3b). The frequency of single and multiple GrB+ appositions in HSVE were in between those of PML and CMVE (Fig. 3c). A specific step in active killing of target cells by cytotoxic T cells is polarization of the cytoplasm and upregulation and redirection of GrB granules towards the target cell as described in in vitro cytotoxicity [47]. In addition to the above-mentioned quantitative findings, we did observe polarization of GrB towards the virus-infected cell in HSVE (Fig. 2k) and CMVE (Fig. 2l) but not in PML (Fig. 2j).

Cell death mechanisms in virus-infected cells

PML

Activated caspase-3 and caspase-6 play a central role in caspase-mediated apoptosis which can be induced by CD8+ cytotoxic T cells through release of GrB. We hypothesized that if cytotoxic GrB+ T cells eliminate virus-infected cells in the various encephalitides, we should be able to detect apoptotic virus-infected cells with upregulated activated caspase-3, or loss of caspase-6 from mitochondria [23]. Previously, we have shown that upregulation of activated caspase-3 is found in the cytoplasm in apoptotic cells, [35, 13]. Here, in normal control brain, activated caspase-3 upregulation was not present. In PML, staining for activated caspase-3 showed the presence of small apoptotic cells (Fig. 4a) that co-localized with CD3 (inset, Fig. 4a) and therefore were characterized as lymphocytes. Analysis of infected oligodendrocytes at the border of demyelinating lesions showed that none of these cells were reactive for caspase-3 (Fig. 4a; Table 2). In control brain (Fig. 4b) and in uninfected cells (Fig. 5g–h), caspase-6 was present in mitochondria. In PML, caspase-6 immunoreactivity was strongly increased. In infected oligodendrocytes, caspase-6 was seen in a granular mitochondrial pattern (Fig. 4c), with no detectable translocation to the cytoplasm being observed.
Table 2
Percentage of infected cells with caspase-3 or nuclear AIF expression
 
PML
HSV
CMV
Caspase-3
0.0 ± 0.0*
0.40 ± 0.13
4.97 ± 1.77
Nuclear AIF
1.6 ± 0.40**
0.00 ± 0.00
0.00 ± 0.00
Data are given as percentage ± SEM
p values: PML vs. HSV, p = 0.0013, PML vs. CMV, p = 0.02, CMV vs. HSV, p = 0.20
** p values: PML vs. HSV, p = 0.006, PML vs. CMV, p = 0.004, CMV vs. HSV, p > 0.99
Since oligodendrocyte death in our PML cases does not seem to result from cytotoxic T cell induced activation of the caspase-cascade, we investigated caspase-independent pathways of cell death. One of these pathways is the Poly(ADP-ribose) polymerase-1-(PARP-1) dependent cell death pathway in which poly-(ADP-ribose) (PAR) and apoptosis-inducing factor (AIF) are some of the key molecules [23]. In normal cells of control brain, weak staining for PAR was seen in the nucleus of cells (Fig. 4d). In PML, enhanced PAR reactivity in uninfected cells was observed (Fig. 4e). In addition, in some of the infected oligodendrocytes, PAR not only was present in the nucleus, but also was seen to translocate to the cytosol (Fig. 4e–h). Some of these cells showed largely degenerating cytoplasms with large vacuoles (Fig. 4g). Staining for AIF in control brain revealed the granular presence of AIF restricted to the mitochondria of normal cells (Fig. 4i). In PML brain, AIF reactivity was upregulated in the mitochondria of uninfected cells (Fig. 4j–l) as well as in mitochondria of most of the infected oligodendrocytes (Fig. 4n). In part of the infected cells, however, we observed that staining of AIF was largely absent from the mitochondria and was translocated to the nucleus (Fig. 4m, o, p). Quantification showed that AIF translocation to the nucleus was detectable in 1.6% of these cells (Table 2). Confocal fluorescence triple staining for SV40, PAR and AIF confirmed the presence of infected cells with simultaneous translocation of PAR from nucleus to cytoplasm and AIF from mitochondria to nucleus (Fig. 4p).
In normal adult brain, oligodendrocyte cell bodies do not express myelin proteins such as major oligodendrocyte glycoprotein (MOG) and proteolipid protein (PLP). During analysis of our PML tissues we however noticed that some of the infected oligodendrocytes at the border of demyelinated lesions strongly express MOG (Fig. 4q) and PLP (Fig. 4r) in the cell bodies. These findings suggest that these proteins are trapped in the endoplasmic reticulum (ER), something we also have shown previously in PLP overexpressing transgenic animals with ER stress [13]. Analysis of protein disulfide isomerase (PDI) and Binding Protein/Glucose Regulated Protein 78 (BiP/GRP78), both involved in endoplasmic reticulum stress, revealed strong upregulation in such infected oligodendrocytes (Fig. 4s, t). ER stress also has been shown to induce hypoxia inducing factor-1α (HIF-1α) [8]. Previously, we already showed the presence of HIF-1α in cases of PML [1]. Here, double-staining for HIF-1α with SV40 shows that HIF-1α is strongly upregulated in some of the infected cells (Fig. 4u).

HSV encephalitis

To detect apoptosis in infected cells we performed double-labeling for caspase-3 and HSV. These stainings revealed the presence of large numbers of positive cells of which, based on their morphology, most were recognized as astrocytes (Fig. 5a). Quantification of these stainings showed that on average 0.4% of infected cells were caspase-3+ (Table 2). The presence of infected apoptotic astrocytes was confirmed by triple labeling of HSV with caspase-3 and GFAP (Fig. 5b). Many of these apoptotic astrocytes showed degeneration of the astrocytic processes (Fig. 5b–c) as also seen previously in Rasmussen encephalitis [5]. Besides these astrocytes also some infected neurons showed caspase-3 reactivity (Fig. 5d). In many of these apoptotic cells, HSV immunostaining became less strong and HSV antigens were only present in the cell body of these cells (Fig. 5e–f). HSVE brains also were stained for caspase-6. In both uninfected (Fig. 5g–h) as well as in many HSV-infected cells (Fig. 5i–j), caspase-6 was present in mitochondria. In areas with large numbers of degenerating caspase-3+ cells we also found caspase-6+ cells with apoptotic nuclei (Fig. 5k). Interestingly, such apoptotic cells showed a diffuse cytoplasmic staining for caspase-6 and loss of mitochondrial caspase-6 (Fig. 5k). Like in caspase-3 stainings, many of these caspase-6+ cells were astrocytes (Fig. 5l), although also cells with oligodendrocyte morphology with cytoplasmic staining of caspase-6 (Fig. 5m) were found.
Since it was difficult to discriminate small HSV-infected cells from normal cells, we also analyzed HSVE brains by performing quadruple labeling of HSV with PAR, AIF and nuclear DAPI stain. These stainings clearly showed that in infected cells, PAR always remained in the nucleus (Fig. 5n–p). Staining for AIF in HSV-infected cells, on the other hand, never was seen outside of the mitochondria (Table 2; Fig. 5n–p). Furthermore, part of the HSV+ cells, again showed a diminished reactivity for HSV, present only in nucleus and cell body (Fig. 5p).

CMV encephalitis

In CMVE, like in PML, activated caspase-3 again could be found in lymphocytes. In addition we found strong cytoplasmic upregulation of caspase-3 in infected cells (Fig. 6a). This finding was confirmed in double-labeling studies for caspase-3 and CMV where almost 5% of CMV+ cells were reactive for caspase-3 (Table 2; Fig. 6b, c). In comparison to other cells, part of the CMV-infected cells only revealed little granular caspase-6 immunoreactivity (arrow Fig. 6d). Some of the infected cells with apoptotic nuclei showed strong cytoplasmic staining (arrowhead Fig. 6d) in line with caspase-mediated apoptosis. Other infected cells however revealed intense caspase-6 immunoreactivity in a granular mitochondrial pattern (Fig. 6e). Notably, unlike in uninfected cells, caspase-6+ mitochondria in some infected cells seemed to be located close to the cell membrane (Fig. 6e). In other CMV-infected cells, caspase-6 immunoreactivity was seen both in mitochondria as well as diffusely dispersed in the cytoplasm (arrows Fig. 6e, f). To confirm that the cell death mechanism in CMVE differs from PML, we also investigated PAR and AIF expression. Immunoreactivity for PAR in infected cells was solely present in the nucleus and never found in a cytoplasmic localization (Fig. 6g, i). In addition, much like the staining for caspase-6, mitochondrial AIF showed a granular pattern mainly located at the outer side of the cytoplasm close to the cell membrane (Fig. 6h, i). AIF reactivity in CMV-infected cells was never found to translocate to the nucleus (Table 2). Altogether, the absence of translocation of PAR and AIF, the presence of activated caspase-3 and the translocation of caspase-6 from mitochondria to cytoplasm in CMV and HSV-infected cells strongly suggest that these cells, unlike cells in PML, die by caspase-mediated apoptosis.

Discussion

The introduction of new highly potent anti-inflammatory drugs, such as Natalizumab and Rituximab has led to an increasing incidence of PML in cancer and autoimmune disease patients [6]. In addition, variants of PML with co-existence of the immune reconstitution inflammatory syndrome (IRIS), also known as PML-IRIS, have in recent years been brought up [6, 35]. Previously, we and others have shown that in PML-IRIS the numbers of inflammatory T cells and plasma cells are increased about 50-fold as compared to PML [34, 36]. Analysis of PML and PML-IRIS cases showed that numbers of infected cells in PML-IRIS were decreased and direct apposition of cytotoxic T cells towards virus-infected cells was increased, suggesting that the reconstituted immune system in PML-IRIS indeed eliminates virus-infected cells more rigorous than in PML [33]. In that study we also analyzed apoptosis of infected cells in PML or PML-IRIS but did not detect any activated caspase-3 immunoreactive oligodendrocytes [33].
Since the question regarding the death of infected cells in PML remained open, we decided to analyze cell death pathways and inflammatory reactions in more detail. Like in our previous study [33], in PML we could not demonstrate a significant attack of JC-infected cells by cytotoxic T cells. This is in contrast with CMVE and HSVE where cytotoxic GrB+ T cells in apposition to virus-infected cells were more obvious. Notably, the absence of cytotoxic T cells targeting infected oligodendrocytes in PML is not because of a lack of MHC class I expression, since we have shown previously that these cells reveal strong MHC class I immunoreactivity [6]. Probably one of the reasons for a more effective elimination of virus-infected cells in CMVE compared to PML is that within lesions the ratio of GrB+ T cells per virus-infected cell seems higher in CMVE. In HSVE absolute numbers of CD3+ T cells and CD4+ T helper cells were much higher than in CMVE and PML. This may be related to the AIDS-associated compromised immune system. The relatively low number of appositions in HSVE probably again is a reflection of the unfavorable ratio between HSV-infected cells and GrB+ T cells. Overall, our results suggest that in PML the immune system is not able to contain the virus infection due to a low number of inflammatory cells relative to the number of infected oligodendrocytes.
Previously, in animal models for EAE we showed that oligodendrocytes and astrocytes undergo caspase-3-mediated apoptosis after attack by CD8+ cytotoxic T cells [16, 42]. In addition, in human Rasmussen Encephalitis we revealed that cells are attacked by cytotoxic T cells and undergo caspase-3 mediated apoptosis [5, 10]. These findings suggest that elimination of neural cells by cytotoxic T cells predominantly is achieved by induction of caspase-mediated apoptosis. Here, in CMVE and HSVE we indeed detected infected cells with caspase-3 reactivity in areas where infected cells were attacked by cytotoxic T cells. These findings are in line with findings by DeBiasi et al. who detected caspase-3 in infected cells in acute HSV encephalitis and congenital CMV encephalitis of mostly young children [19]. These authors at that time however could not demonstrate a role of the immune system as we did here. Our findings in CMVE and HSVE are in sharp contrast with PML, where we did not find any caspase-3 reactivity in the cytoplasm of JC-infected cells.
Despite being a central pathological feature, until now very few studies dealt with the question of oligodendrocyte death in PML. One paper favored apoptosis as the death inducing mechanism [41], whereas the other study argued that apoptosis is not involved in oligodendrocyte death since these cells are protected by the anti-apoptotic protein survivin [40]. Richardson-Burns et al. showed DNA fragmentation in oligodendrocytes by TUNEL. They however did not identify morphological changes in the nucleus, as usually seen in apoptotic cells. Moreover, caspase-3 in this study was observed in the nucleus rather than the cytoplasm. Although these findings are striking at first glance, there are some caveats which at that time were not considered. First, it has been reported that DNA viruses cause DNA double-strand breaks in host cells [25, 28, 44, 49] and thus it is conceivable that rather than apoptosis or necrosis, the TUNEL staining detects DNA double-strand breaks induced by the JC virus. Secondly, we and others have shown that apoptotic cells with nuclear condensation or the presence of apoptotic bodies show cytoplasmic rather than nuclear distribution of activated caspase-3 [3, 13, 14, 20, 45]. Moreover, nuclear caspase-3 has been suggested to play a role other than in apoptosis [38].
Instead of activated caspase-3, infected degenerating oligodendrocytes in PML expressed nuclear AIF and cytoplasmic PAR. In addition, in these cells we could show signs of ER stress and upregulation of hypoxia inducible factor-1α (HIF-1α). The translocation of PAR and AIF suggests that these cells died by a pathway designated as parthanatos [18, 23]. Interestingly, parthanatos is often linked to cell death induced by hypoxia [32]. Also in PML a role of hypoxic mechanisms has been suggested [30, 39]. Hypoxia might be induced by molecules such as nitric oxide or oxygen radicals produced by macrophages and microglia. In addition, hypoxic pathways may result from thrombotic occlusion of small inflamed vessels [21]. Another possibility, however, is that a virtual hypoxia is induced in JC-virus-infected cells indirectly. By taking over the host’s protein production machinery, viruses can induce endoplasmic reticulum (ER) stress [22]. More specifically, ER stress can be induced by so-called viroporins [22]. In JC-virus-infected cells, agnoprotein acts as such a viroporin [46]. ER stress by viroporins is induced by multiple mechanisms such as membrane remodeling, delayed glycoprotein trafficking and disruption of calcium homeostasis. The latter results in an increase in cytosolic calcium followed by calcium-dependent cell death signaling and finally cell death [22, 46]. ER stress therefore, can act as an intracellular inducer of hypoxic pathways as shown in intermittent hypoxia where ER stress induced HIF-1α activity [8].
In summary, our results show that, compared to CMVE and HSVE, interaction of GrB+ cytotoxic T cells with infected oligodendrocytes are much less frequent in PML lesions. Furthermore, in CMVE and HSVE, infected cells seem to die by caspase-mediated apoptotic pathways. In PML however, infected oligodendrocytes show translocation of PAR and AIF, hypoxia and ER stress which are all compatible with the parthanatos pathway of cell death. Our findings suggest that, unlike in HSVE and CMVE, in PML the immune system not adequately enough eradicates JC-infected oligodendrocytes. Instead these cells seem to die by virus-induced changes in homeostatic pathways.

Acknowledgements

Open access funding provided by Medical University of Vienna. The study was funded by the Austrian Science Fund (Project P26936-B24). The authors like to acknowledge Marianne Leisser and Angela Kury for expert analytical assistance. Drs. Guillaume Martin-Blondel, Roland Liblau and Jordon Dunham are thanked for critically reading the manuscript.

Compliance with ethical standards

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee (ECS number 1206/2013) and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.
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

Literatur
1.
Zurück zum Zitat Aboul-Enein F, Rauschka H, Kornek B, Stadelmann C, Stefferl A, Bruck W, Lucchinetti C, Schmidbauer M, Jellinger K, Lassmann H (2003) Preferential loss of myelin-associated glycoprotein reflects hypoxia-like white matter damage in stroke and inflammatory brain diseases. J Neuropathol Exp Neurol 62:25–33CrossRefPubMed Aboul-Enein F, Rauschka H, Kornek B, Stadelmann C, Stefferl A, Bruck W, Lucchinetti C, Schmidbauer M, Jellinger K, Lassmann H (2003) Preferential loss of myelin-associated glycoprotein reflects hypoxia-like white matter damage in stroke and inflammatory brain diseases. J Neuropathol Exp Neurol 62:25–33CrossRefPubMed
2.
Zurück zum Zitat Astrom KE, Mancall EL, Richardson EP Jr (1958) Progressive multifocal leuko-encephalopathy; a hitherto unrecognized complication of chronic lymphatic leukaemia and Hodgkin’s disease. Brain 81:93–111CrossRefPubMed Astrom KE, Mancall EL, Richardson EP Jr (1958) Progressive multifocal leuko-encephalopathy; a hitherto unrecognized complication of chronic lymphatic leukaemia and Hodgkin’s disease. Brain 81:93–111CrossRefPubMed
3.
Zurück zum Zitat Bauer J, Bradl M, Hickley WF, Forss-Petter S, Breitschopf H, Linington C, Wekerle H, Lassmann H (1998) T-cell apoptosis in inflammatory brain lesions: destruction of T cells does not depend on antigen recognition. Am J Pathol 153:715–724CrossRefPubMedPubMedCentral Bauer J, Bradl M, Hickley WF, Forss-Petter S, Breitschopf H, Linington C, Wekerle H, Lassmann H (1998) T-cell apoptosis in inflammatory brain lesions: destruction of T cells does not depend on antigen recognition. Am J Pathol 153:715–724CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Bauer J, Bradl M, Klein M, Leisser M, Deckwerth T, Wekerle H, Lassmann H (2002) Endoplasmic reticulum stress in PLP-overexpressing transgenic rats: gray matter oligodendrocytes are more vulnerable than white matter oligodendrocytes. J Neuropathol Exp Neurol 61:12–22CrossRefPubMed Bauer J, Bradl M, Klein M, Leisser M, Deckwerth T, Wekerle H, Lassmann H (2002) Endoplasmic reticulum stress in PLP-overexpressing transgenic rats: gray matter oligodendrocytes are more vulnerable than white matter oligodendrocytes. J Neuropathol Exp Neurol 61:12–22CrossRefPubMed
5.
Zurück zum Zitat Bauer J, Eiger CE, Hans VH, Schramm J, Urbach H, Lassmann H, Bien CG (2007) Astrocytes are a specific immunological target in Rasmussen’s encephalitis. Ann Neurol 62:67–80CrossRefPubMed Bauer J, Eiger CE, Hans VH, Schramm J, Urbach H, Lassmann H, Bien CG (2007) Astrocytes are a specific immunological target in Rasmussen’s encephalitis. Ann Neurol 62:67–80CrossRefPubMed
6.
Zurück zum Zitat Bauer J, Gold R, Adams O, Lassmann H (2015) Progressive multifocal leukoencephalopathy and immune reconstitution inflammatory syndrome (IRIS). Acta Neuropathol 130:751–764CrossRefPubMed Bauer J, Gold R, Adams O, Lassmann H (2015) Progressive multifocal leukoencephalopathy and immune reconstitution inflammatory syndrome (IRIS). Acta Neuropathol 130:751–764CrossRefPubMed
8.
Zurück zum Zitat Belaidi E, Thomas A, Bourdier G, Moulin S, Lemarie E, Levy P, Pepin JL, Korichneva I, Godin-Ribuot D, Arnaud C (2016) Endoplasmic reticulum stress as a novel inducer of hypoxia inducible factor-1 activity: its role in the susceptibility to myocardial ischemia-reperfusion induced by chronic intermittent hypoxia. Int J Cardiol 210:45–53CrossRefPubMed Belaidi E, Thomas A, Bourdier G, Moulin S, Lemarie E, Levy P, Pepin JL, Korichneva I, Godin-Ribuot D, Arnaud C (2016) Endoplasmic reticulum stress as a novel inducer of hypoxia inducible factor-1 activity: its role in the susceptibility to myocardial ischemia-reperfusion induced by chronic intermittent hypoxia. Int J Cardiol 210:45–53CrossRefPubMed
9.
Zurück zum Zitat Berger JR, Kaszovitz B, Post MJ, Dickinson G (1987) Progressive multifocal leukoencephalopathy associated with human immunodeficiency virus infection. A review of the literature with a report of sixteen cases. Ann Intern Med 107:78–87CrossRefPubMed Berger JR, Kaszovitz B, Post MJ, Dickinson G (1987) Progressive multifocal leukoencephalopathy associated with human immunodeficiency virus infection. A review of the literature with a report of sixteen cases. Ann Intern Med 107:78–87CrossRefPubMed
10.
Zurück zum Zitat Bien CG, Bauer J, Deckwerth TL, Wiendl H, Deckert M, Wiestler OD, Schramm J, Elger CE, Lassmann H (2002) Destruction of neurons by cytotoxic T cells: a new pathogenic mechanism in Rasmussen’s encephalitis. Ann Neurol 51:311–318CrossRefPubMed Bien CG, Bauer J, Deckwerth TL, Wiendl H, Deckert M, Wiestler OD, Schramm J, Elger CE, Lassmann H (2002) Destruction of neurons by cytotoxic T cells: a new pathogenic mechanism in Rasmussen’s encephalitis. Ann Neurol 51:311–318CrossRefPubMed
11.
Zurück zum Zitat Bien CG, Vincent A, Barnett MH, Becker AJ, Blumcke I, Graus F, Jellinger KA, Reuss DE, Ribalta T, Schlegel J et al (2012) Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain 135:1622–1638CrossRefPubMed Bien CG, Vincent A, Barnett MH, Becker AJ, Blumcke I, Graus F, Jellinger KA, Reuss DE, Ribalta T, Schlegel J et al (2012) Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain 135:1622–1638CrossRefPubMed
12.
Zurück zum Zitat Bollag B, Prins C, Snyder EL, Frisque RJ (2000) Purified JC virus T and T’ proteins differentially interact with the retinoblastoma family of tumor suppressor proteins. Virology 274:165–178CrossRefPubMed Bollag B, Prins C, Snyder EL, Frisque RJ (2000) Purified JC virus T and T’ proteins differentially interact with the retinoblastoma family of tumor suppressor proteins. Virology 274:165–178CrossRefPubMed
13.
Zurück zum Zitat Bradl M, Bauer J, Inomata T, Zielasek J, Nave KA, Toyka K, Lassmann H, Wekerle H (1999) Transgenic Lewis rats overexpressing the proteolipid protein gene: myelin degeneration and its effect on T cell-mediated experimental autoimmune encephalomyelitis. Acta Neuropathol (Berl) 97:595–606CrossRef Bradl M, Bauer J, Inomata T, Zielasek J, Nave KA, Toyka K, Lassmann H, Wekerle H (1999) Transgenic Lewis rats overexpressing the proteolipid protein gene: myelin degeneration and its effect on T cell-mediated experimental autoimmune encephalomyelitis. Acta Neuropathol (Berl) 97:595–606CrossRef
14.
Zurück zum Zitat Brecht S, Gelderblom M, Srinivasan A, Mielke K, Dityateva G, Herdegen T (2001) Caspase-3 activation and DNA fragmentation in primary hippocampal neurons following glutamate excitotoxicity. Mol Brain Res 94:25–34CrossRefPubMed Brecht S, Gelderblom M, Srinivasan A, Mielke K, Dityateva G, Herdegen T (2001) Caspase-3 activation and DNA fragmentation in primary hippocampal neurons following glutamate excitotoxicity. Mol Brain Res 94:25–34CrossRefPubMed
15.
Zurück zum Zitat Budka H, Shah KV (1983) Papovavirus antigens in paraffin sections of PML brains. Prog Clin Biol Res 105:299–309PubMed Budka H, Shah KV (1983) Papovavirus antigens in paraffin sections of PML brains. Prog Clin Biol Res 105:299–309PubMed
16.
Zurück zum Zitat Cabarrocas J, Bauer J, Piaggio E, Liblau R, Lassmann H (2003) Effective and selective immune surveillance of the brain by MHC class I-restricted cytotoxic T lymphocytes. Eur J Immunol 33:1174–1182CrossRefPubMed Cabarrocas J, Bauer J, Piaggio E, Liblau R, Lassmann H (2003) Effective and selective immune surveillance of the brain by MHC class I-restricted cytotoxic T lymphocytes. Eur J Immunol 33:1174–1182CrossRefPubMed
17.
Zurück zum Zitat Carson KR, Focosi D, Major EO, Petrini M, Richey EA, West DP, Bennett CL (2009) Monoclonal antibody-associated progressive multifocal leucoencephalopathy in patients treated with rituximab, natalizumab, and efalizumab: a review from the research on adverse drug events and reports (RADAR) project. Lancet Oncol 10:816–824CrossRefPubMed Carson KR, Focosi D, Major EO, Petrini M, Richey EA, West DP, Bennett CL (2009) Monoclonal antibody-associated progressive multifocal leucoencephalopathy in patients treated with rituximab, natalizumab, and efalizumab: a review from the research on adverse drug events and reports (RADAR) project. Lancet Oncol 10:816–824CrossRefPubMed
18.
Zurück zum Zitat David KK, Andrabi SA, Dawson TM, Dawson VL (2009) Parthanatos, a messenger of death. Front Biosci 14:1116–1128CrossRef David KK, Andrabi SA, Dawson TM, Dawson VL (2009) Parthanatos, a messenger of death. Front Biosci 14:1116–1128CrossRef
19.
Zurück zum Zitat DeBiasi RL, Kleinschmidt-DeMasters BK, Richardson-Burns S, Tyler KL (2002) Central nervous system apoptosis in human herpes simplex virus and cytomegalovirus encephalitis. J Infect Dis 186:1547–1557CrossRefPubMed DeBiasi RL, Kleinschmidt-DeMasters BK, Richardson-Burns S, Tyler KL (2002) Central nervous system apoptosis in human herpes simplex virus and cytomegalovirus encephalitis. J Infect Dis 186:1547–1557CrossRefPubMed
20.
Zurück zum Zitat Eberhardt O, Coelln RV, Kugler S, Lindenau J, Rathke-Hartlieb S, Gerhardt E, Haid S, Isenmann S, Gravel C, Srinivasan A et al (2000) Protection by synergistic effects of adenovirus-mediated X-chromosome-linked inhibitor of apoptosis and glial cell line-derived neurotrophic factor gene transfer in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. J Neurosci 20:9126–9134PubMed Eberhardt O, Coelln RV, Kugler S, Lindenau J, Rathke-Hartlieb S, Gerhardt E, Haid S, Isenmann S, Gravel C, Srinivasan A et al (2000) Protection by synergistic effects of adenovirus-mediated X-chromosome-linked inhibitor of apoptosis and glial cell line-derived neurotrophic factor gene transfer in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. J Neurosci 20:9126–9134PubMed
21.
Zurück zum Zitat Esiri MM, Kennedy PG (1997) Viral diseases. In: Graham D, Lantos PL (eds) Greenfield’s neuropathology. Arnold, London, pp 3–63 Esiri MM, Kennedy PG (1997) Viral diseases. In: Graham D, Lantos PL (eds) Greenfield’s neuropathology. Arnold, London, pp 3–63
22.
Zurück zum Zitat Fung TS, Torres J, Liu DX (2015) The emerging roles of viroporins in ER stress response and autophagy induction during virus infection. Viruses 7:2834–2857CrossRefPubMedPubMedCentral Fung TS, Torres J, Liu DX (2015) The emerging roles of viroporins in ER stress response and autophagy induction during virus infection. Viruses 7:2834–2857CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Galluzzi L, Kepp O, Krautwald S, Kroemer G, Linkermann A (2014) Molecular mechanisms of regulated necrosis. Semin Cell Dev Biol 35:24–32CrossRefPubMed Galluzzi L, Kepp O, Krautwald S, Kroemer G, Linkermann A (2014) Molecular mechanisms of regulated necrosis. Semin Cell Dev Biol 35:24–32CrossRefPubMed
24.
Zurück zum Zitat Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, Dawson TM, Dawson VL, El-Deiry WS, Fulda S et al (2012) Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ 19:107–120CrossRefPubMed Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, Dawson TM, Dawson VL, El-Deiry WS, Fulda S et al (2012) Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ 19:107–120CrossRefPubMed
25.
Zurück zum Zitat Gaspar M, Shenk T (2006) Human cytomegalovirus inhibits a DNA damage response by mislocalizing checkpoint proteins. Proc Natl Acad Sci USA 103:2821–2826CrossRefPubMedPubMedCentral Gaspar M, Shenk T (2006) Human cytomegalovirus inhibits a DNA damage response by mislocalizing checkpoint proteins. Proc Natl Acad Sci USA 103:2821–2826CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Kappos L, Bates D, Edan G, Eraksoy M, Garcia-Merino A, Grigoriadis N, Hartung HP, Havrdova E, Hillert J, Hohlfeld R et al (2011) Natalizumab treatment for multiple sclerosis: updated recommendations for patient selection and monitoring. Lancet Neurol 10:745–758CrossRefPubMed Kappos L, Bates D, Edan G, Eraksoy M, Garcia-Merino A, Grigoriadis N, Hartung HP, Havrdova E, Hillert J, Hohlfeld R et al (2011) Natalizumab treatment for multiple sclerosis: updated recommendations for patient selection and monitoring. Lancet Neurol 10:745–758CrossRefPubMed
27.
Zurück zum Zitat Kleinschmidt-DeMasters BK, Miravalle A, Schowinsky J, Corboy J, Vollmer T (2012) Update on PML and PML-IRIS occurring in multiple sclerosis patients treated with natalizumab. J Neuropathol Exp Neurol 71:604–617CrossRefPubMed Kleinschmidt-DeMasters BK, Miravalle A, Schowinsky J, Corboy J, Vollmer T (2012) Update on PML and PML-IRIS occurring in multiple sclerosis patients treated with natalizumab. J Neuropathol Exp Neurol 71:604–617CrossRefPubMed
28.
Zurück zum Zitat Kudoh A, Fujita M, Zhang L, Shirata N, Daikoku T, Sugaya Y, Isomura H, Nishiyama Y, Tsurumi T (2005) Epstein-Barr virus lytic replication elicits ATM checkpoint signal transduction while providing an S-phase-like cellular environment. J Biol Chem 280:8156–8163CrossRefPubMed Kudoh A, Fujita M, Zhang L, Shirata N, Daikoku T, Sugaya Y, Isomura H, Nishiyama Y, Tsurumi T (2005) Epstein-Barr virus lytic replication elicits ATM checkpoint signal transduction while providing an S-phase-like cellular environment. J Biol Chem 280:8156–8163CrossRefPubMed
29.
Zurück zum Zitat Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D (2005) Progressive multifocal leukoencephalopathy in a patient treated with Natalizumab. N Engl J Med 353:375–381CrossRefPubMed Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D (2005) Progressive multifocal leukoencephalopathy in a patient treated with Natalizumab. N Engl J Med 353:375–381CrossRefPubMed
30.
Zurück zum Zitat Lassmann H (2003) Hypoxia-like tissue injury as a component of multiple sclerosis lesions. J Neurol Sci 206:187–191CrossRefPubMed Lassmann H (2003) Hypoxia-like tissue injury as a component of multiple sclerosis lesions. J Neurol Sci 206:187–191CrossRefPubMed
31.
Zurück zum Zitat Lima MA, Marzocchetti A, Autissier P, Tompkins T, Chen Y, Gordon J, Clifford DB, Gandhi RT, Venna N, Berger JR et al (2007) Frequency and phenotype of JC virus-specific CD8+ T lymphocytes in the peripheral blood of patients with progressive multifocal leukoencephalopathy. J Virol 81:3361–3368CrossRefPubMedPubMedCentral Lima MA, Marzocchetti A, Autissier P, Tompkins T, Chen Y, Gordon J, Clifford DB, Gandhi RT, Venna N, Berger JR et al (2007) Frequency and phenotype of JC virus-specific CD8+ T lymphocytes in the peripheral blood of patients with progressive multifocal leukoencephalopathy. J Virol 81:3361–3368CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Liou AK, Clark RS, Henshall DC, Yin XM, Chen J (2003) To die or not to die for neurons in ischemia, traumatic brain injury and epilepsy: a review on the stress-activated signaling pathways and apoptotic pathways. Prog Neurobiol 69:103–142CrossRefPubMed Liou AK, Clark RS, Henshall DC, Yin XM, Chen J (2003) To die or not to die for neurons in ischemia, traumatic brain injury and epilepsy: a review on the stress-activated signaling pathways and apoptotic pathways. Prog Neurobiol 69:103–142CrossRefPubMed
33.
Zurück zum Zitat Martin-Blondel G, Bauer J, Cuvinciuc V, Uro-Coste E, Debard A, Massip P, Delisle MB, Lassmann H, Marchou B, Mars LT et al (2013) In situ evidence of JC virus control by CD8+ T cells in PML-IRIS during HIV infection. Neurology 81:964–970CrossRefPubMed Martin-Blondel G, Bauer J, Cuvinciuc V, Uro-Coste E, Debard A, Massip P, Delisle MB, Lassmann H, Marchou B, Mars LT et al (2013) In situ evidence of JC virus control by CD8+ T cells in PML-IRIS during HIV infection. Neurology 81:964–970CrossRefPubMed
34.
Zurück zum Zitat Martin-Blondel G, Bauer J, Uro-Coste E, Biotti D, Averseng-Peaureaux D, Fabre N, Dumas H, Bonneville F, Lassmann H, Marchou B et al (2015) Therapeutic use of CCR5 antagonists is supported by strong expression of CCR5 on CD8(+) T cells in progressive multifocal leukoencephalopathy-associated immune reconstitution inflammatory syndrome. Acta Neuropathol 129:463–465CrossRefPubMed Martin-Blondel G, Bauer J, Uro-Coste E, Biotti D, Averseng-Peaureaux D, Fabre N, Dumas H, Bonneville F, Lassmann H, Marchou B et al (2015) Therapeutic use of CCR5 antagonists is supported by strong expression of CCR5 on CD8(+) T cells in progressive multifocal leukoencephalopathy-associated immune reconstitution inflammatory syndrome. Acta Neuropathol 129:463–465CrossRefPubMed
35.
Zurück zum Zitat Martin-Blondel G, Brassat D, Bauer J, Lassmann H, Liblau RS (2016) CCR5 blockade for neuroinflammatory diseases—beyond control of HIV. Nat Rev Neurol 12:95–105CrossRefPubMed Martin-Blondel G, Brassat D, Bauer J, Lassmann H, Liblau RS (2016) CCR5 blockade for neuroinflammatory diseases—beyond control of HIV. Nat Rev Neurol 12:95–105CrossRefPubMed
36.
Zurück zum Zitat Metz I, Radue EW, Oterino A, Kumpfel T, Wiendl H, Schippling S, Kuhle J, Sahraian MA, Gray F, Jakl V et al (2012) Pathology of immune reconstitution inflammatory syndrome in multiple sclerosis with natalizumab-associated progressive multifocal leukoencephalopathy. Acta Neuropathol 123:235–245CrossRefPubMed Metz I, Radue EW, Oterino A, Kumpfel T, Wiendl H, Schippling S, Kuhle J, Sahraian MA, Gray F, Jakl V et al (2012) Pathology of immune reconstitution inflammatory syndrome in multiple sclerosis with natalizumab-associated progressive multifocal leukoencephalopathy. Acta Neuropathol 123:235–245CrossRefPubMed
37.
Zurück zum Zitat Miller JR, Barrett RE, Britton CB, Tapper ML, Bahr GS, Bruno PJ, Marquardt MD, Hays AP, McMurtry JG 3rd, Weissman JB et al (1982) Progressive multifocal leukoencephalopathy in a male homosexual with T-cell immune deficiency. N Engl J Med 307:1436–1438CrossRefPubMed Miller JR, Barrett RE, Britton CB, Tapper ML, Bahr GS, Bruno PJ, Marquardt MD, Hays AP, McMurtry JG 3rd, Weissman JB et al (1982) Progressive multifocal leukoencephalopathy in a male homosexual with T-cell immune deficiency. N Engl J Med 307:1436–1438CrossRefPubMed
38.
Zurück zum Zitat Noyan-Ashraf MH, Brandizzi F, Juurlink BH (2005) Constitutive nuclear localization of activated caspase 3 in subpopulations of the astroglial family of cells. Glia 49:588–593CrossRefPubMed Noyan-Ashraf MH, Brandizzi F, Juurlink BH (2005) Constitutive nuclear localization of activated caspase 3 in subpopulations of the astroglial family of cells. Glia 49:588–593CrossRefPubMed
39.
Zurück zum Zitat Pina-Oviedo S, Khalili K, Del Valle L (2009) Hypoxia inducible factor-1 alpha activation of the JCV promoter: role in the pathogenesis of progressive multifocal leukoencephalopathy. Acta Neuropathol 118:235–247CrossRefPubMedPubMedCentral Pina-Oviedo S, Khalili K, Del Valle L (2009) Hypoxia inducible factor-1 alpha activation of the JCV promoter: role in the pathogenesis of progressive multifocal leukoencephalopathy. Acta Neuropathol 118:235–247CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Pina-Oviedo S, Urbanska K, Radhakrishnan S, Sweet T, Reiss K, Khalili K, Del Valle L (2007) Effects of JC virus infection on anti-apoptotic protein survivin in progressive multifocal leukoencephalopathy. Am J Pathol 170:1291–1304CrossRefPubMedPubMedCentral Pina-Oviedo S, Urbanska K, Radhakrishnan S, Sweet T, Reiss K, Khalili K, Del Valle L (2007) Effects of JC virus infection on anti-apoptotic protein survivin in progressive multifocal leukoencephalopathy. Am J Pathol 170:1291–1304CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Richardson-Burns SM, Kleinschmidt-DeMasters BK, DeBiasi RL, Tyler KL (2002) Progressive multifocal leukoencephalopathy and apoptosis of infected oligodendrocytes in the central nervous system of patients with and without AIDS. Arch Neurol 59:1930–1936CrossRefPubMed Richardson-Burns SM, Kleinschmidt-DeMasters BK, DeBiasi RL, Tyler KL (2002) Progressive multifocal leukoencephalopathy and apoptosis of infected oligodendrocytes in the central nervous system of patients with and without AIDS. Arch Neurol 59:1930–1936CrossRefPubMed
42.
Zurück zum Zitat Saxena A, Bauer J, Scheik T, Zappulla J, Audebert M, Desbois S, Waisman A, Lassmann H, Liblau RS, Mars LT (2008) Cutting edge: multiple sclerosis-like lesions induced by effector CD8 T cells recognizing a sequestered antigen on oligodendrocytes. J Immunol 181:1617–1621CrossRefPubMed Saxena A, Bauer J, Scheik T, Zappulla J, Audebert M, Desbois S, Waisman A, Lassmann H, Liblau RS, Mars LT (2008) Cutting edge: multiple sclerosis-like lesions induced by effector CD8 T cells recognizing a sequestered antigen on oligodendrocytes. J Immunol 181:1617–1621CrossRefPubMed
43.
Zurück zum Zitat Schmedt N, Andersohn F, Garbe E (2012) Signals of progressive multifocal leukoencephalopathy for immunosuppressants: a disproportionality analysis of spontaneous reports within the US Adverse Event Reporting System (AERS). Pharmacoepidemiol Drug Saf 21:1216–1220CrossRefPubMed Schmedt N, Andersohn F, Garbe E (2012) Signals of progressive multifocal leukoencephalopathy for immunosuppressants: a disproportionality analysis of spontaneous reports within the US Adverse Event Reporting System (AERS). Pharmacoepidemiol Drug Saf 21:1216–1220CrossRefPubMed
44.
Zurück zum Zitat Shirata N, Kudoh A, Daikoku T, Tatsumi Y, Fujita M, Kiyono T, Sugaya Y, Isomura H, Ishizaki K, Tsurumi T (2005) Activation of ataxia telangiectasia-mutated DNA damage checkpoint signal transduction elicited by herpes simplex virus infection. J Biol Chem 280:30336–30341CrossRefPubMed Shirata N, Kudoh A, Daikoku T, Tatsumi Y, Fujita M, Kiyono T, Sugaya Y, Isomura H, Ishizaki K, Tsurumi T (2005) Activation of ataxia telangiectasia-mutated DNA damage checkpoint signal transduction elicited by herpes simplex virus infection. J Biol Chem 280:30336–30341CrossRefPubMed
45.
Zurück zum Zitat Srinivasan A, Roth KA, Sayers RO, Shindler KS, Wong AM, Fritz LC, Tomaselli KJ (1998) In situ immunodetection of activated caspase-3 in apoptotic neurons in the developing nervous system. Cell Death Differ 5:1004–1016CrossRefPubMed Srinivasan A, Roth KA, Sayers RO, Shindler KS, Wong AM, Fritz LC, Tomaselli KJ (1998) In situ immunodetection of activated caspase-3 in apoptotic neurons in the developing nervous system. Cell Death Differ 5:1004–1016CrossRefPubMed
46.
Zurück zum Zitat Suzuki T, Orba Y, Okada Y, Sunden Y, Kimura T, Tanaka S, Nagashima K, Hall WW, Sawa H (2010) The human polyoma JC virus agnoprotein acts as a viroporin. PLoS Pathog 6:e1000801CrossRefPubMedPubMedCentral Suzuki T, Orba Y, Okada Y, Sunden Y, Kimura T, Tanaka S, Nagashima K, Hall WW, Sawa H (2010) The human polyoma JC virus agnoprotein acts as a viroporin. PLoS Pathog 6:e1000801CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Wagner L, Yang OO, Garcia-Zepeda EA, Ge Y, Kalams SA, Walker BD, Pasternack MS, Luster AD (1998) Beta-chemokines are released from HIV-1-specific cytolytic T-cell granules complexed to proteoglycans. Nature 391:908–911CrossRefPubMed Wagner L, Yang OO, Garcia-Zepeda EA, Ge Y, Kalams SA, Walker BD, Pasternack MS, Luster AD (1998) Beta-chemokines are released from HIV-1-specific cytolytic T-cell granules complexed to proteoglycans. Nature 391:908–911CrossRefPubMed
48.
Zurück zum Zitat Wuthrich C, Kesari S, Kim WK, Williams K, Gelman R, Elmeric D, De Girolami U, Joseph JT, Hedley-Whyte T, Koralnik IJ (2006) Characterization of lymphocytic infiltrates in progressive multifocal leukoencephalopathy: co-localization of CD8(+) T cells with JCV-infected glial cells. J Neurovirol 12:116–128CrossRefPubMed Wuthrich C, Kesari S, Kim WK, Williams K, Gelman R, Elmeric D, De Girolami U, Joseph JT, Hedley-Whyte T, Koralnik IJ (2006) Characterization of lymphocytic infiltrates in progressive multifocal leukoencephalopathy: co-localization of CD8(+) T cells with JCV-infected glial cells. J Neurovirol 12:116–128CrossRefPubMed
49.
Zurück zum Zitat Xiao Y, Chen J, Liao Q, Wu Y, Peng C, Chen X (2013) Lytic infection of Kaposi’s sarcoma-associated herpesvirus induces DNA double-strand breaks and impairs non-homologous end joining. J Gen Virol 94:1870–1875CrossRefPubMed Xiao Y, Chen J, Liao Q, Wu Y, Peng C, Chen X (2013) Lytic infection of Kaposi’s sarcoma-associated herpesvirus induces DNA double-strand breaks and impairs non-homologous end joining. J Gen Virol 94:1870–1875CrossRefPubMed
50.
Zurück zum Zitat zu Rhein G, Chou SM (1965) Particles resembling papova viruses in human cerebral demyelinating disease. Science 148:1477–1479CrossRef zu Rhein G, Chou SM (1965) Particles resembling papova viruses in human cerebral demyelinating disease. Science 148:1477–1479CrossRef
Metadaten
Titel
Differences in T cell cytotoxicity and cell death mechanisms between progressive multifocal leukoencephalopathy, herpes simplex virus encephalitis and cytomegalovirus encephalitis
verfasst von
Susanne Laukoter
Helmut Rauschka
Anna R. Tröscher
Ulrike Köck
Etsuji Saji
Kurt Jellinger
Hans Lassmann
Jan Bauer
Publikationsdatum
05.11.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Acta Neuropathologica / Ausgabe 4/2017
Print ISSN: 0001-6322
Elektronische ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-016-1642-1

Weitere Artikel der Ausgabe 4/2017

Acta Neuropathologica 4/2017 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Stuhltransfusion könnte Fortschreiten von Parkinson-Symptomen bremsen

03.05.2024 Parkinson-Krankheit Nachrichten

Kann eine frühzeitige Stuhltransplantation das Fortschreiten von Parkinson-Symptomen verlangsamen? Die Ergebnisse einer randomisierten Phase-2-Studie scheinen dafür zu sprechen.

Frühe Tranexamsäure-Therapie nützt wenig bei Hirnblutungen

02.05.2024 Hirnblutung Nachrichten

Erhalten Personen mit einer spontanen Hirnblutung innerhalb von zwei Stunden nach Symptombeginn eine Tranexamsäure-Therapie, kann dies weder die Hämatomexpansion eindämmen noch die Mortalität senken.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders scheint das auf weibliche Kranke zuzutreffen, wie eine Studie zeigt.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Update Neurologie

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