Multiple sclerosis (MS) is an idiopathic demyelinating disease in which meningeal inflammation correlates with accelerated disease progression. The study of meningeal inflammation in MS has been limited because of constrained access to MS brain/spinal cord specimens and the lack of experimental models recapitulating progressive MS. Unlike induced models, a spontaneously occurring model would offer a unique opportunity to understand MS immunopathogenesis and provide a compelling framework for translational research. We propose granulomatous meningoencephalomyelitis (GME) as a natural model to study neuropathological aspects of MS. GME is an idiopathic, progressive neuroinflammatory disease of young dogs with a female bias. In the GME cases examined in this study, the meninges displayed focal and disseminated leptomeningeal enhancement on magnetic resonance imaging, which correlated with heavy leptomeningeal lymphocytic infiltration. These leptomeningeal infiltrates resembled tertiary lymphoid organs containing large B cell clusters that included few proliferating Ki67+ cells, plasma cells, follicular dendritic/reticular cells, and germinal center B cell–like cells. These B cell collections were confined in a specialized network of collagen fibers associated with the expression of the lympho-organogenic chemokines CXCL13 and CCL21. Although neuroparenchymal perivascular infiltrates contained B cells, they lacked the immune signature of aggregates in the meningeal compartment. Finally, meningeal B cell accumulation correlated significantly with cortical demyelination reflecting neuropathological similarities to MS. Hence, during chronic neuroinflammation, the meningeal microenvironment sustains B cell accumulation that is accompanied by underlying neuroparenchymal injury, indicating GME as a novel, naturally occurring model to study compartmentalized neuroinflammation and the associated pathology thought to contribute to progressive MS.

Multiple sclerosis (MS) is a neuroinflammatory disease characterized by demyelination and axonal loss that can lead to major neurologic impairment (1). MS onset is more prevalent in young adults, especially women (2). Most patients initially experience relapsing-remitting MS (RRMS) characterized by peripheral leukocyte infiltration that leads to the formation of perivascular demyelinating lesions (3). A smoldering course often emerges termed secondary progressive MS (4). About 10% of patients progress from onset without obvious relapses (referred to as primary progressive MS) (5). The progressive injury in both secondary progressive MS and primary progressive MS is thought to be mediated by chronic CNS-compartmentalized inflammatory and degenerative mechanisms. The overall MS spectrum is pathologically characterized by leukocyte infiltration associated with oligodendrocyte damage, demyelination, and axonal injury (as the pathologic consequence of relapse biology), as well as injury in the subpial cortex, intracortical gray matter, and leukocortical areas (considered major substrates for progressive MS) (6, 7). The submeningeal cortical injury has been associated with the presence of adjacent meningeal immune cell infiltrates containing T cells, myeloid cells and numerous B cells/plasma cells (8, 9). Such B cell–rich meningeal infiltrates have been shown to correlate with accelerated progressive disease (10, 11). Additionally, these leptomeningeal aggregates can display the arrangement of a tertiary lymphoid organ (TLO)–like structure (8, 9). TLOs are abnormal lymph node–like formations arising at sites of lingering inflammation, such as chronic infection, and autoimmune-related diseases including MS (12). TLOs constitute satellite lymphoid structures able to promote inflammatory processes that in MS are thought to sustain the intrathecal differentiation, maturation, and persistence of pathogenic B cells (13). As B cells can remain for years to decades within the inflamed CNS, their contribution to propagating neuroinflammation has recently gained large attention, including through their interaction with T and myeloid cells as well as their established Ab-independent functions (14).

Similar to MS, granulomatous meningoencephalomyelitis (GME) is an idiopathic spontaneous neuroinflammatory disease with female predilection occurring in dogs of young and middle age (1517). The distribution of inflammation can be disseminated, focal, or confined to the optic nerve, and the localization of inflammation correlates with presenting clinical signs (18, 19). Clinical onset can be acute, though disease progression can be delayed with immunosuppressive therapy (18, 20). Antemortem diagnosis of GME is difficult and is typically made by excluding other etiologies and by a combination of imaging and clinical pathologic diagnostics. Cerebrospinal fluid analysis of pleocytosis with >50% mononuclear cells combined with negative serology for infectious diseases, is supportive of a GME diagnosis (20). Magnetic resonance imaging (MRI) is frequently used to localize areas of inflammation, which are characterized by single or multiple hyperintense lesions with irregular borders, as well as variable enhancement of parenchyma and/or meninges following gadolinium administration (19, 20). Histologically, GME has been characterized by inflammation within the meninges and white matter of the forebrain, brainstem, and/or spinal cord (16, 21). Lesions are composed of multifocal to regionally diffuse perivascular cuffs of macrophages, lymphocytes, and plasma cells (18, 22). Inflammation is predominant in the white matter but can extend into the gray matter (23). Although leptomeningeal immune cell infiltration has been noted (24), little is known about the nature of the immune cells, and their association with neuropathological changes.

Numerous studies have demonstrated that clinical and pathological manifestations of certain neurologic diseases are similar in humans and domestic animal species (25, 26). Because of this, the potential of validating novel therapeutics in canine clinical trials prior to or in tandem with clinical trials in humans is gaining widespread recognition and participation (27, 28). Given the genetic and immunologic similarities between humans and dogs, along with advancements in analytical techniques for canine tissues, the list of therapeutics validated by these types of comparative clinical trials applied in spontaneous, natural models of human disease continues to grow (29, 30).

We hypothesize that GME recapitulates certain immunological and neuropathological processes of MS, particularly at the leptomeningeal level, and as such may serve as a useful natural model of progressive disease. The GME cases studied here displayed areas of both focal and disseminated leptomeningeal enhancement (LME) on imaging. These correlated with heavy leptomeningeal infiltration characterized by large collections of B cells displaying lympho-organogenic features, properties not found in parenchymal infiltrates. Notably, the leptomeningeal B cell infiltration correlated with areas of submeningeal cortical demyelination emulating pathological aspects of progressive MS. As a whole, these findings suggest that in GME, leptomeningeal inflammation drives pathology within the underlying neuroparenchyma and indicates GME might serve as a novel, naturally occurring model to study compartmentalized neuroinflammation and the resulting neuropathology seen in progressive MS.

The Section of Neurology of the Ryan Veterinary Hospital of the School of Veterinary Medicine, University of Pennsylvania (Penn Vet) identified potential GME cases based on presenting clinical signs, cerebrospinal fluid analysis, and diagnostic MRI. The anatomic pathology database was also searched from January 2008 through December 2018 for cases with multifocal brain disease with a diagnosis including GME. Thirteen GME cases with cortical leptomeningeal inflammatory infiltrates not contiguous with invasive inflammatory lesions of an initial 27 cases of GME were included in this study (Table I). Ten dogs had MRI performed as part of their diagnostic workup, and six of these dogs had magnetic resonance images available for review. Postcontrast LME was scored on a scale of 0 to 3, with 0 = no enhancement, 1 = mild enhancement seen in normal animals, 2 = moderate increase in enhancement, 3 = severe increase in enhancement. T1-weighted (pre- and postcontrast), T2-weighted, and fluid-attenuated inversion recovery images were evaluated. Additionally, MRI was available for review from two cases each of necrotizing meningoencephalitis (NME) and canine distemper virus (CDV). Formalin-fixed and paraffin-embedded (FFPE) blocks from 4 cases each of NME and CDV in dogs aged 3–10 y old were evaluated histologically. MRI images and histology were evaluated and compared with the GME cases. FFPE blocks sampled from the cerebrum from four neurologically normal dogs spanning the same age range (3–11 y old) of GME cases were used as comparative controls for immunohistochemistry (IHC) analysis that is described below.

Table I.

Clinical features of the GME cases studied

BreedSexAge (y)MRIMRI LME scoreClinical Presentation
Dachshund FS Several week history vomiting and inappetence. Few days low head carriage, leaning/veering, ataxia noted, then decreased responsiveness and unable to stand. 
Maltese 1-wk history of decreased energy progressing to kyphotic posture and holding head extended to right side. Ultimately progressed to seizure episodes and presented laterally recumbent. 
Mixed breed FS 10-d history of neck pain progressing to central vestibular disease. 
Portuguese water dog FS 10 2-wk duration of circling, seizures, and obtundation. 
Labrador MC NA 1-wk history of decreased appetite with neck pain and change in mentation, which progressed to circling to right with head turn and mild vestibular ataxia. 
American pit bull terrier MC Not stated 
Shih tzu FS NA Acute onset ataxia four times and lethargy. 
Eurasier MC NA Presented status epilepticus, which was progressive. 
American pit bull terrier FS NA History of inappetence, dull and altered mentation, ataxia, and right head-turn and vertical nystagmus. 
Mixed breed FS 10 History of mental dullness and circling; left-sided postural reaction deficits 
Greyhound FS UN NA History of ataxia and proprioceptive deficits (greater in thoracic limbs than pelvic limbs). 
Boxer MC NA Acute onset nonambulatory tetraparesis that progressed to mental obtundation. 
Mixed breed MC NA History of recent onset of progressive seizures. 
BreedSexAge (y)MRIMRI LME scoreClinical Presentation
Dachshund FS Several week history vomiting and inappetence. Few days low head carriage, leaning/veering, ataxia noted, then decreased responsiveness and unable to stand. 
Maltese 1-wk history of decreased energy progressing to kyphotic posture and holding head extended to right side. Ultimately progressed to seizure episodes and presented laterally recumbent. 
Mixed breed FS 10-d history of neck pain progressing to central vestibular disease. 
Portuguese water dog FS 10 2-wk duration of circling, seizures, and obtundation. 
Labrador MC NA 1-wk history of decreased appetite with neck pain and change in mentation, which progressed to circling to right with head turn and mild vestibular ataxia. 
American pit bull terrier MC Not stated 
Shih tzu FS NA Acute onset ataxia four times and lethargy. 
Eurasier MC NA Presented status epilepticus, which was progressive. 
American pit bull terrier FS NA History of inappetence, dull and altered mentation, ataxia, and right head-turn and vertical nystagmus. 
Mixed breed FS 10 History of mental dullness and circling; left-sided postural reaction deficits 
Greyhound FS UN NA History of ataxia and proprioceptive deficits (greater in thoracic limbs than pelvic limbs). 
Boxer MC NA Acute onset nonambulatory tetraparesis that progressed to mental obtundation. 
Mixed breed MC NA History of recent onset of progressive seizures. 

FS, female spayed; M, male; MC, male castrated; N, no; NA, not available; UN, unknown; Y, yes.

Tissue samples were fixed for at least 24 h and FFPE blocks were sectioned 5-µm thick, mounted on glass slides, and stained with H&E. Six to nine brain and spinal cord sections were examined from each case, including sections from the level of the cerebral hemispheres and lateral ventricles, hippocampus and thalamus, midbrain, cerebellum, and spinal cord. For evaluation of leptomeningeal inflammatory infiltrates and demyelination, rostral cerebral sections from the level of the frontal lobe to the hippocampus at the interthalamic adhesion, dependent upon which FFPE blocks were available for each case, were sampled.

To analyze markers sensitive to formalin fixation, frozen blocks of CNS tissue were processed as previously published (31). In brief, frozen blocks of brain tissue from four GME dogs were sectioned with a Leica cryostat into 8-μm thick slices. Frozen sections were fixed in acetone 10 min at –20°C. All following incubations were done at room temperature unless noted otherwise. Sections were blocked with serum 10% from the species of the secondary Ab for 90 min. For double immunofluorescence staining, the sections were incubated with the primary Abs (Supplemental Table I) overnight at 4°C. The secondary Abs were diluted in 3% serum and incubated for 60 min. Finally, the sections were mounted in mounting media and Hoescht (1/5000) (Thermo Fisher Scientific). For negative controls, the primary Abs were omitted. Immunostaining of FFPE sections was performed as previously described (32). Slides containing 5-μm CNS sections were deparaffinized, hydrated, and placed in Dako Ag retrieval solution to be heated in a pressure cooker for 22 min and allowed to cool for 30 min. After blocking, primary and secondary Abs were added as described above for the frozen sections. Immunostained sections were imaged using an Olympus IX83 set up equipped with a motorized X, Y, Z stage and a spinning disk confocal head (X-Light V2; CrestOptics, Rome, Italy) using a Hamamatsu R2 cooled CMOS camera (Hamamatsu, Hamamatsu City, Japan) operated by the MetaMorph software (Molecular Devices, Sunnyvale, CA). Imaging processing and analysis was performed as previously described (33, 34). Paraffin embedded canine thymus and brain, and frozen tonsil and brain were sectioned and used to standardize the staining of CNS/immunological markers.

Tissue sections were processed for IHC as previously described (32, 35). In brief, cortical areas of all 13 GME cases and four unaffected dogs were evaluated with Abs to CD3 (T cells) (M7254, Ms mAb; Agilent Dako), Multiple Myeloma 1 (MUM1/IRF4) (plasma cells) (M7259, Ms mAb; Dako), CD79b (B cells) (no. 96024, Rb mAb Cell Signaling Technology), Iba1 (macrophages/microglia) (019-19741, Rb pAb; WAKO), and myelin-oligodendrocyte glycoprotein (MOG) (myelin) (CST, #96457, Rb mAb). All of these stains were performed using an LEICA Bond RXm automated IHC stain according to the manufacturer’s recommendations. Isotype control Abs were used as negative controls. Positive controls were normal canine brain tissue and lymph node. All GME and control IHC slides stained immunohistochemically were scanned at 20× using a Leica Aperio AT2 slide scanner (Leica Biosystems, Buffalo Grove, IL) and image acquisition was performed with ImageScope (Leica Biosystems, Buffalo Grove, IL). For the quantification of the MOG staining to assess demyelination, image analysis was performed using a positive pixel count algorithm using ImageScope software. A single algorithm was developed using appropriate thresholds for the staining intensity, and was applied to specific annotated regions on each slide.

Scoring of leptomeningeal and parenchymal inflammation was based on previously described methods (4). We evaluated slides of cerebral cortex stained for B cells (CD79b), T cells (CD3), and used MUM1 to label plasma cells. Leptomeningeal inflammatory infiltrates were scored using a four-tiered scale (absent = no inflammatory cells present; mild = fewer than 20 diffusely distributed inflammatory cells or leptomeningeal perivascular cuffing up to 3 layers; moderate = 20–50 diffusely distributed inflammatory cells or leptomeningeal perivascular cuffing three to seven layers; marked >50 diffusely distributed inflammatory cells or leptomeningeal perivascular cuffing greater than seven layers). Parenchymal cortical inflammation was scored using a similar scale: 0 = absent, 1 = mild (vessels with one cuff), 2 = moderate (many vessels with two cuffs), 3 = marked (scattered or many vessels with >3 cuffs).

Data were analyzed using GraphPad Prism v.8 software. Unpaired student t test was used for comparisons between number of cell populations within leptomeningeal infiltrates and for determining the differential expression of MOG in subcortical areas of controls versus GME cases. Two tailed (95%) Pearson correlation analysis was used to determine the association between the percentage of MOG expression (demyelination index) and distinct neuropathological parameters including leptomeningeal B cell accumulation.

T1W hyperintensity following gadolinium administration is indicative of a break in the CNS barriers from various underlying causes including neovascularization, inflammation, cerebral ischemia, and CNS pressure changes (36). In GME, MRI has become an important tool to diagnose the disease when combined with cerebrospinal fluid analysis and neurologic assessment (18). In our cohort, MRI of six dogs were available for review by a board-certified veterinary neurologist (C.H.V.). We found dogs with GME had mild to strong contrast enhancing, poorly delineated and ill-defined lesions involving the white and gray matter (Fig. 1), as has been previously described (37). However, we also noted the leptomeninges commonly displayed areas of focal or multifocal contrast enhancement in 6 out of 10 of the dogs examined, which has not been commonly reported in GME (Fig. 1, Table I). The brains of two dogs each with NME and CDV, imaged during the same time period, exhibited no LME (data not shown). Histological evaluation of the contrast enhancing lesions confirmed correlative inflammatory lesions in the leptomeningeal space and also in the neuroparenchyma. In general, the intensity of LME was increased in GME cases with more severe leptomeningeal inflammation; however, the limited number of cases with imaging available for enhancement scoring (6 out of 13 cases) precluded statistical analysis of this trend. This relatively consistent finding of LME led us to evaluate both the neuroparenchymal and leptomeningeal infiltrates, as well as the associated neuropathological changes in the abnormal regions detected with MRI in the GME cases.

FIGURE 1.

LME in GME. T1-weighted pre- and postcontrast transverse brain images of a GME dog. The leptomeninges show multifocal regions of contrast enhancement indicating inflammation and increased vascular permeability over the cerebrum (dashed boxes) and brainstem (dotted box).

FIGURE 1.

LME in GME. T1-weighted pre- and postcontrast transverse brain images of a GME dog. The leptomeninges show multifocal regions of contrast enhancement indicating inflammation and increased vascular permeability over the cerebrum (dashed boxes) and brainstem (dotted box).

Close modal

We examined 13 cases demonstrating the classic histologic changes characteristic of GME. Brain sections, including leptomeninges, were evaluated by two board-certified veterinary anatomic pathologists (M.E.C., M.D.S.). As is seen in GME, all cases exhibited cuffing of leptomeningeal and parenchymal blood vessels morphologically characterized by lymphocytes as well as variable numbers of aggregated and/or intermixed mononuclear cells composed of macrophages and plasma cells (Fig. 2). The leptomeningeal clusters ranged from small perivascular aggregates composed primarily of small lymphocytic cuffs (Fig. 2A) to further progressed aggregates, with moderate numbers of lymphocytes admixed with macrophages and plasma cells (Fig. 2B). In the most developed aggregates, large numbers of tightly packed lymphocytes resembling TLO-like structures expanded the leptomeninges with macrophages accumulating peripherally (Fig. 2C).

FIGURE 2.

Leptomeningeal B cell predominant inflammation in GME. Characteristic perivascular cuffing of meningeal blood vessels by lymphocytes (l) and plasma cells (pc) with peripheral aggregates of macrophages (m) range from (A) small perivascular aggregates composed primarily of small lymphocyte cuffs to (B) moderate numbers of lymphocytes admixed with macrophages to (C) large numbers of tightly packed lymphocytes resembling TLO-like structures with peripheral macrophages. High power views of representative areas are shown and include arrowheads (pc and m) and dotted lines for lymphocytic clusters (l). High power views in (a′), (a″), (b′), (b″), (c′), and (c″) of representative areas are shown and include arrowheads (pc and m) and dotted lines for lymphocytic clusters (l). (D) Immunofluorescence staining of lymphocytes in the leptomeningeal inflammatory infiltrates of GME brains composed of large numbers of CD20+ B cells (red) and small numbers of dispersed CD3+ T cells (green) with no distinct distribution of either cell population. (n = 8 and 2–4 infiltrates per animal). (E) CD3+CD8+ T cells (yellow, indicative of coexpression) made up most of the cells scattered between CD20+ B cells compared with the number of CD3+CD8 cells (green) (n = 4 and 4–6 infiltrates per animal). (F) CD20+ B cell (red) aggregation costained with Ki67 (green) highlights the low numbers of dividing B cells in leptomeningeal aggregates (n = 8 and 2–4 infiltrates per animal). Tissues were counterstained with DAPI (blue) for nuclear staining, and high-power views of each image are shown on the right. Error bars, mean ± SEM. Scale bars, 50 μm. **** p ≤ 0.0001.

FIGURE 2.

Leptomeningeal B cell predominant inflammation in GME. Characteristic perivascular cuffing of meningeal blood vessels by lymphocytes (l) and plasma cells (pc) with peripheral aggregates of macrophages (m) range from (A) small perivascular aggregates composed primarily of small lymphocyte cuffs to (B) moderate numbers of lymphocytes admixed with macrophages to (C) large numbers of tightly packed lymphocytes resembling TLO-like structures with peripheral macrophages. High power views of representative areas are shown and include arrowheads (pc and m) and dotted lines for lymphocytic clusters (l). High power views in (a′), (a″), (b′), (b″), (c′), and (c″) of representative areas are shown and include arrowheads (pc and m) and dotted lines for lymphocytic clusters (l). (D) Immunofluorescence staining of lymphocytes in the leptomeningeal inflammatory infiltrates of GME brains composed of large numbers of CD20+ B cells (red) and small numbers of dispersed CD3+ T cells (green) with no distinct distribution of either cell population. (n = 8 and 2–4 infiltrates per animal). (E) CD3+CD8+ T cells (yellow, indicative of coexpression) made up most of the cells scattered between CD20+ B cells compared with the number of CD3+CD8 cells (green) (n = 4 and 4–6 infiltrates per animal). (F) CD20+ B cell (red) aggregation costained with Ki67 (green) highlights the low numbers of dividing B cells in leptomeningeal aggregates (n = 8 and 2–4 infiltrates per animal). Tissues were counterstained with DAPI (blue) for nuclear staining, and high-power views of each image are shown on the right. Error bars, mean ± SEM. Scale bars, 50 μm. **** p ≤ 0.0001.

Close modal

To address the specificity of the leptomeningeal neuropathology seen in GME, we also examined two spontaneous cases of neuroinflammatory diseases that occur in canine patients and were previously considered potential natural models for MS (17, 38, 39). We found that these two canine diseases, NME and CDV (n = 4 for each), demonstrated different inflammatory patterns and different associated neuropathology. NME featured malacia and loss of cortical gray and subcortical white matter with large neuroparenchyma aggregates of activated macrophages/microglia and minimal to mild perivascular cuffing by lymphocytes (Supplemental Fig. 1A–C). In CDV, there was moderate to marked perivascular cuffing by lymphocytes in the parenchyma with only mild inflammation in the meninges and myelin loss was generally confined to cerebellar white matter (Supplemental Fig. 1D–1F). Thus, the unique histopathological features detected in GME indicate robust immune cell infiltration in the perivascular neuroparenchyma and, specifically, within the leptomeningeal space.

Given the aggregation of lymphocytes within the leptomeninges (Figs 2A–C), we assessed the immune cell composition in this compartment. We found large numbers of CD20+ B cells in leptomeningeal aggregates with a distribution and spatial arrangement confirming the histopathological TLO-like phenotype (Fig. 2D). In contrast to the leptomeningeal B cell organization, the perivascular cuffs within the neuroparenchyma lacked the lymphoid-like arrangement and were composed of a smaller proportion of B cells compared with the leptomeninges (Supplemental Fig. 2A). Scattered CD3+ T cell infiltration similar to autoimmune-driven delayed-type hypersensitivity responses has been described in GME lesions (22, 24). The leptomeningeal TLO-like aggregates presented significantly lower numbers of CD3+ T cells than B cells and were distributed randomly between the B cell aggregates (Fig. 2D). Of note, within the B cell clusters studied we did not observe distinct B cell and T cell areas. CD3+ T cells also distributed sparsely within perivascular cuffs in the parenchyma (Supplemental Fig. 2B). To discriminate the type of T cell infiltrating the leptomeninges, we interrogated the presence of cytotoxic T cells. We found that most of the T cells scattered between the CD20+ B cells were CD3+CD8+, in both the leptomeningeal space (Fig. 2E) and neuroparenchyma (Supplemental Fig. 2C), indicating that CD8 T cells are the predominant T cell population over CD4 T cells in GME infiltrates. We could not directly assess the CD4+ T cell compartment as none of the Abs tested detected canine CD4. The leptomeningeal TLO-like aggregates in GME were characterized by the presence of few Ki67+ B cells (Fig. 2F), indicating a limited proliferation rate and the likely importance of a persistent peripheral pool of B cells to support these structures. In contrast, B cells accumulating perivascularly in the neuroparenchyma lacked Ki67+ expression (Supplemental Fig. 2D). These findings indicate that B cells represent an important component of GME immunopathology and that the leptomeningeal compartment is conducive to their aggregation as seen in MS.

In MS, leptomeningeal B cell–rich clusters vary from rudimentary clusters of cells to relatively organized TLO-like structures (9). In these GME cases, we found a similar gradation of leptomeningeal B cell aggregation (Figs. 2A–C). Highly organized TLOs feature segregated T and B cell zones containing germinal center (GC) B cells, follicular dendritic cell (FDC)/follicular reticular cell (FRC) networks, high endothelial venules (HEVs) and a reticular network supporting the homing and activation of T and B cells (40). GC B cells are characterized by the expression of the markers GL7 (41) and BCL6 (42). As we were unsuccessful in staining canine tissues with these markers, we relied on CD81 for the identification of GC B cell–like cells (43, 44). Leptomeningeal B cell aggregates displayed CD81 expression within TLO-like aggregates (Fig. 3A), whereas CD81 expression was sparse in between B cells within parenchymal infiltrates (Supplemental Fig. 2E). To address the involvement of FDCs and FRCs, we used CD271, a marker expressed by both cell types in secondary lymphoid organs (45, 46). The leptomeningeal TLO-like aggregates exhibited enrichment in CD271+ cells displaying a reticular morphology (Fig. 3B) as compared with parenchymal perivascular infiltrates (Supplemental Fig. 2F). Of note, some of the CD271 enrichment around parenchymal vasculature (blood–brain barrier) might reflect labeling of pericytes (47). Immune cell recruitment at sites of inflammation and their interaction with tissue-resident and stromal cells driving TLO formation promotes the release of CXCL13, CCL19, and CCL21. These chemokines promote the recruitment and spatial organization of immune cells into TLOs (12). The B cell chemoattractant CXCL13 was prominently expressed in the TLO-like leptomeningeal aggregates within the B cell clusters and meningeal blood vessels (Fig. 3C). Likewise, the T cell and dendritic cell chemoattractant CCL21 was also expressed within those aggregates and by meningeal vessels (Fig. 3D). These findings highlight the important role of the blood–meningeal barrier (BMB) in supporting immune cell aggregation in the leptomeninges.

FIGURE 3.

Lympho-organogenic features of TLO-like leptomeningeal B cell aggregates. Immunofluorescence staining of leptomeningeal inflammatory infiltrates of GME brains (images are representative of eight cases). (A) CD20+ B (red) and CD81+ (green) cells accumulate in the inflamed leptomeninges. (B) CD271+ (green) staining of cells resembling reticular cells in B cell (red) rich areas within inflamed leptomeninges infiltrates. (C) Expression of the B cell chemoattract CXCL13 (red) in TLO-like leptomeningeal aggregates with scattered CD3 T cells (green). (D) CCL21 (red), expression, and costaining with CD3 (green). (E) CD20+ B cell (blue) aggregation costained with PNAd (green; marker of HEVs) and isolectin B4 (IB4; red; marker of TLO blood vessels and myeloid cells) in leptomeninges. High power views indicate colocalization of PNAd and IsoB4 in blood vessels within these aggregates. (F) Collagen type IV (green) distribution in large, perivascular leptomeningeal TLO-like infiltrate. Tissues were counterstained with DAPI (blue) for nuclear staining, and high-power views of each image are shown on the right. Scale bars, 50 μm.

FIGURE 3.

Lympho-organogenic features of TLO-like leptomeningeal B cell aggregates. Immunofluorescence staining of leptomeningeal inflammatory infiltrates of GME brains (images are representative of eight cases). (A) CD20+ B (red) and CD81+ (green) cells accumulate in the inflamed leptomeninges. (B) CD271+ (green) staining of cells resembling reticular cells in B cell (red) rich areas within inflamed leptomeninges infiltrates. (C) Expression of the B cell chemoattract CXCL13 (red) in TLO-like leptomeningeal aggregates with scattered CD3 T cells (green). (D) CCL21 (red), expression, and costaining with CD3 (green). (E) CD20+ B cell (blue) aggregation costained with PNAd (green; marker of HEVs) and isolectin B4 (IB4; red; marker of TLO blood vessels and myeloid cells) in leptomeninges. High power views indicate colocalization of PNAd and IsoB4 in blood vessels within these aggregates. (F) Collagen type IV (green) distribution in large, perivascular leptomeningeal TLO-like infiltrate. Tissues were counterstained with DAPI (blue) for nuclear staining, and high-power views of each image are shown on the right. Scale bars, 50 μm.

Close modal

The development of HEVs is a defining feature distinguishing organized TLOs from other types of inflammatory infiltrates given their critical role in supporting naive cell migration (48). HEVs specifically express peripheral node addressin (PNAd), a molecule that serves as the ligand for l-selectin/CD62L, and normally absent in other types of blood vessels even within lymphoid organs (49, 50). In GME, PNAd expression appears to be luminal and restricted to small vessels (IsoB4+) within leptomeningeal TLO-like aggregates (Fig. 3E). These HEVs display a staining pattern consistent with the well-described plump morphology of mature HEVs (51) and were mostly localized in the periphery of the B cell aggregates as previously reported (52). The organized spatiotemporal distribution of B cells and other cellular components making up TLO-like structures depends on the discrete arrangement of extracellular matrix components (53). Analysis of collagen type IV demonstrated an organized stroma interweaving and extending in between immune cells and presenting a radiocentric pattern displaying higher matrix deposition in proximity to meningeal blood vessels (Fig. 3F). This structural conformation is similar to the bundled and aligned collagen conduits present within lymph node follicles and TLO-like structures in the periphery and in the CNS (5456). Despite the technical limitations in studying specialized populations (GC B cell–like cells and FDCs/FRCs) associated with chronic inflammatory processes, our findings indicate the formation of TLO-like aggregates in the leptomeninges of GME dogs and suggest these structures might exacerbate compartmentalized neuroinflammation.

As meningeal inflammation in patients with MS has been associated with submeningeal cortical demyelination, neuroaxonal loss, and an accelerated and progressive course of MS (4, 7, 57), we evaluated leptomeningeal immune cell infiltrates and subjacent regions of cortical gray matter. We found that the leptomeningeal inflammatory cells were distributed diffusely (several individualized cells loosely arranged) or in a focal manner (dense aggregates of cells) in all the GME cases studied (Fig. 4A). Most GME cases (n = 10) exhibited both focal and diffuse immune cell infiltration, whereas two cases demonstrated only a diffuse pattern and one case presented focal immune cell infiltration without diffusely distributed cells (Fig. 4A). Whereas the focal aggregates were composed of B cells, T cells, and plasma cells (Figs 4B–D), the diffuse infiltrates contained mainly B cells with scattered T cells and only rare plasma cells (Supplemental Figs 3A–C), mirroring the compositional immune findings shown in (Fig. 2. Within the cortical gray matter underlying these leptomeningeal inflammatory aggregates, we defined the degree of demyelination by evaluating the expression of MOG, one of the main structural components of myelin (58). We examined the level of myelin expression in subpial, intracortical, leukocortical, and subcortical white matter regions relative to sections from control dogs (Figs 5A, 5B). We detected decreased myelin expression in the subpial and leukocortical regions when considered individually (p = 0.007 and 0.008, respectively) and a notable trend in the intracortical region (p = 0.053) (Supplemental Fig. 3D). In contrast, the expression of myelin in the subcortical white matter did not differ between GME and controls (Supplemental Fig. 3D). The overall percentage of myelin staining in the combined regions of cortex underlying leptomeningeal aggregates was significantly decreased in GME relative to controls (p = 0.005) (Fig. 5C). Thus, we demonstrated submeningeal demyelination in regions of the cerebral cortex previously described as affected in MS (4, 11, 59). Notably, we found a significant correlation between the severity of B cell leptomeningeal aggregates and the extent of cortical demyelination (Fig. 5D), further reflecting the remarkable neuropathological similarities between GME and MS. We also noted that cortical blood vessels within the parenchyma were often cuffed by lymphocytes and macrophages, similar to what is seen in MS (60), though their presence did not correlate with cortical demyelination (Fig. 5E). Similar to leptomeningeal inflammation, B cells tended to predominate relative to T cells within these perivascular cuffs (Fig. 5E). Additionally, we found evidence of cortical glial nodules characterized by aggregates of activated macrophages/microglia (Fig. 5F) as described in MS (4, 61). However, the presence of these nodules were not correlated with cortical demyelination (Fig. 5F). These findings indicate that in GME, leptomeningeal B cells play a role in the underlying neuropathology seen in the cortex as it has been postulated in MS. This demonstrated relationship makes GME a compelling model replicating definitive aspects of MS neuropathology.

FIGURE 4.

Spatial distribution of meningeal inflammatory infiltrates. (A) Most cases (11 out of 13) demonstrated both diffuse and focal distribution of leptomeningeal inflammatory infiltrates. Focal infiltrates were composed primarily of (B) B cells (CD79b) with fewer (C) T cells (CD3) and (D) plasma cells (MUM1). IHC. Scale bars, 50 μm.

FIGURE 4.

Spatial distribution of meningeal inflammatory infiltrates. (A) Most cases (11 out of 13) demonstrated both diffuse and focal distribution of leptomeningeal inflammatory infiltrates. Focal infiltrates were composed primarily of (B) B cells (CD79b) with fewer (C) T cells (CD3) and (D) plasma cells (MUM1). IHC. Scale bars, 50 μm.

Close modal
FIGURE 5.

Submeningeal demyelination associated with leptomeningeal inflammatory infiltrates. (A and B) Myelin staining (MOG IHC) in specific submeningeal regions (subpial, intracortical, leukocortical) and subcortical white matter of GME cases was compared with staining in control dogs. The pia is labeled as a dotted line separating the leptomeningeal space from parenchyma. (C) Extent of MOG staining in combined regions of cortex (excluding subcortical white matter) in GME dogs (n = 13) relative to controls (n = 4). Error bars, mean ± SEM. ** p ≤ 0.01. (D) Correlation analysis between the extent of cortical myelin loss and the leptomeningeal B cell infiltration (r = –0.57; p = 0.03). (E) Blood vessels within the cortical parenchyma were also cuffed by B cells (CD79b IHC) and T cells (CD3 IHC), though the presence of these inflammatory cells did not correlate with the degree of demyelination (r = 0.27; p = 0.35). (F) Iba1 positive microglial cells arranged in nodules (arrowheads) were found scattered throughout the cortical tissue, yet their presence did not correlate with the extent of demyelination (r = 0.23; p = 0.44). IHC. Scale bars, 50 μm.

FIGURE 5.

Submeningeal demyelination associated with leptomeningeal inflammatory infiltrates. (A and B) Myelin staining (MOG IHC) in specific submeningeal regions (subpial, intracortical, leukocortical) and subcortical white matter of GME cases was compared with staining in control dogs. The pia is labeled as a dotted line separating the leptomeningeal space from parenchyma. (C) Extent of MOG staining in combined regions of cortex (excluding subcortical white matter) in GME dogs (n = 13) relative to controls (n = 4). Error bars, mean ± SEM. ** p ≤ 0.01. (D) Correlation analysis between the extent of cortical myelin loss and the leptomeningeal B cell infiltration (r = –0.57; p = 0.03). (E) Blood vessels within the cortical parenchyma were also cuffed by B cells (CD79b IHC) and T cells (CD3 IHC), though the presence of these inflammatory cells did not correlate with the degree of demyelination (r = 0.27; p = 0.35). (F) Iba1 positive microglial cells arranged in nodules (arrowheads) were found scattered throughout the cortical tissue, yet their presence did not correlate with the extent of demyelination (r = 0.23; p = 0.44). IHC. Scale bars, 50 μm.

Close modal

GME has long been considered a T cell–mediated autoimmune disease of unknown origin (22, 62). In this study we present evidence supporting the importance of B cells to the pool of leukocytes infiltrating the CNS, particularly at the leptomeningeal level. MS and its animal models, like experimental autoimmune encephalomyelitis (EAE), were similarly long considered to be T cell–mediated diseases (6). However, the ability to substantially limit new disease activity in MS using anti–B cell therapies has highlighted the important contribution of B cells to neuroinflammation (6365). In addition to the contribution of peripheral B cells to MS relapses, their long-term persistence in leptomeningeal infiltrates is considered a potential driver of progressive MS (4, 8, 9, 11, 66). In this study, postcontrast meningeal enhancement by MRI was present in GME. This pattern of enhancement corresponded with leptomeningeal B cell accumulations, which in turn correlated with submeningeal cortical demyelination and closely resembles that of progressive MS (8, 9, 11). Thus, we propose that B cell–rich meningeal immune cell aggregates may potentiate chronic inflammation in GME and contribute to submeningeal CNS injury, as has been postulated to occur in MS (14, 67, 68).

The pattern of leukocyte infiltration in the GME leptomeninges demonstrated enrichment of B cells organized at times as TLO-like structures. In MS, meningeal infiltrates rich in B cells are associated with accelerated progressive clinical disease as compared with progressive cases lacking these structures (11, 59). Attempts to recapitulate TLO-like development have been carried out using the most common model of MS, murine EAE (54, 6971). EAE exhibits great variability depending upon the mouse strain, Ag used, as well as immunization protocol, and the observed disease/neuropathology does not recapitulate important features of MS (72, 73). Additionally, although other canine neurologic diseases previously postulated to mimic MS do demonstrate meningeal inflammation (NME) and demyelination (CDV), the specific pattern of inflammation and demyelination found in GME closely mirrors the compartmentalized immunopathology seen in MS. Furthermore, GME affects both the brain and the spinal cord and prevails in young female dogs, as is seen clinically in MS. The consistent pattern of B cell–rich leptomeningeal aggregation that correlates with underlying cortical demyelination suggests that this compartmentalized inflammation exacerbates the clinical course of GME as occurs in MS. A TLO-like pattern of infiltration arises at predefined locations as a result of the structural and chemical support provided by stromal and endothelial cells, which in the CNS, are enriched within the leptomeninges (69, 7476). Correspondingly, we found that chemokines responsible for immune cell aggregation/organization within TLO-like structures were enriched in the leptomeningeal space in GME. CCL21 and the B cell chemoattractant CXCL13 might sustain the migration of lymphocytes into the leptomeninges across the BMB, a vascular bed more supportive of immune cell interactions than its parenchymal counterpart, the blood–brain barrier (7781). In addition, the presence of HEVs further supports this process and suggests that naive lymphocytes can migrate into the leptomeninges and TLO-like aggregates. As TLOs are considered focal sites that promote lymphocyte activation, Ag presentation, somatic hypermutation, and class switching of B cells (48, 82), our findings are consistent with the postulation that leptomeningeal TLO/TLO-like formations exacerbate and sustain inflammatory responses within this niche.

The overrepresentation of B cells within the GME leptomeningeal clusters and the relative abundance of CD8+ T cells over CD4+ T cells challenges the prevailing view that CD4+ T helper responses drive the pathogenesis of GME (83) and further highlights immunopathological similarities between GME and MS. As we have found in GME, CD8+ T cells also outnumber CD4+ T cells in MS lesions (8486). These similarities contrast EAE in which the immunization protocol evokes T cell responses heavily biased toward inflammatory CD4+ T cells with relatively little involvement of CD8+ T cells (73, 87). In terms of the inflammatory response, analysis of GME brains indicates high levels of IL-17 and IFN-γ expression as compared with other neuroinflammatory canine diseases (83). Given that most T cells in the GME brains are cytotoxic, we speculate that IFN-γ–producing CD8+ T cells (Tc1) and IL-17–producing CD8+ (Tc17) cells represent important drivers of disease. This immune phenotype has also been reported in MS in which Tc1 and Tc17 cells are enriched in the cerebrospinal fluid of patients and their intrathecal frequencies correlate with disability (88, 89). Moreover, Tc1 and Tc17 cells are found within MS lesions (90, 91), and aside from IFN-γ and IL-17 production, they can also display a cytotoxic phenotype (92, 93). We postulate that the increased migration of B cells and CD8+ T cells across the BMB (79, 80), the stromal meningeal microenvironment (8, 9), and the compartmentalized nature of the inflammatory CD8+ T cell–B cell response reported in MS (60), is mirrored by the leptomeningeal immune cell infiltrates present in GME. These findings suggest that CD8+ T cells migrate to and accumulate in the meningeal compartment in which B cells then stimulate their activation via cytokines and/or through Ag presentation. These interactions likely drive CD8+ T cell effector phenotypes, which in turn induce submeningeal demyelination and axonal pathology. Studies in MS have attributed these neuropathological changes to B cells, Tc1, and Tc17 cells releasing soluble mediators known to be cytotoxic for oligodendrocytes and neurons, resulting in demyelination and neuronal death (4, 9498). Future studies of GME could aim to fully characterize the cytokine composition present in cerebrospinal fluid and in serum in an effort to establish diagnostic and prognostic biomarkers for GME and MS alike.

In MS, leptomeningeal B cell/TLO-like aggregates are associated with submeningeal cortical lesions (8, 99). Our findings in this study suggest that a similar B cell driven destruction of the underlying cortex occurs in GME. The distribution of meningeal inflammatory cells (in both focal and diffuse patterns) and the associated cortical demyelination noted in the GME cases reviewed in this study, are similar to those described in MS (7, 57, 61, 100, 101). Most prior studies examining pathologic lesions of GME have concentrated on the neuroparenchymal inflammatory infiltrates with little emphasis on meningeal inflammation (62, 83). In this study we demonstrate that the extent of cortical demyelination, considered the major substrate for progressive MS (6, 7), correlates with the severity of leptomeningeal B cell aggregation. Similar to lesions in early MS (4), the subpial lesions in GME are not fully demyelinated, suggesting that GME might be modeling the more acute and/or regenerative process associated with early stages of disease progression. Although this contrasts with the fully demyelinated cortical lesions reported in progressive MS (57, 102), other features of progressive MS (especially the presence of leptomeningeal aggregates of B cells associated with these regions of demyelination) are present in GME. As subpial cortical demyelination correlates with an accelerated and progressive course of MS (4, 7, 57) and because no other MS model replicates this chronic compartmentalized immunopathology (103), GME holds a tremendous potential to advance our understanding of the mechanisms driving disease progression and to develop and test novel therapeutics. In addition to demyelination, subpial cortical neuronal loss is also seen in progressive MS (104), an aspect not evaluated in our GME cohort and to be addressed in future studies. Finally, in two cases, we observed areas in which leptomeningeal infiltrates invade/displace the underlying cortex. This might indicate a more active/acute immune–mediated inflammatory process in which GME could differ from MS or might represent earlier stages of aggressive leptomeningeal driven pathology as has been reported in acute MS (4).

The underlying etiologic trigger(s) of inflammation have not been identified in MS nor in GME. The CD8+ T cell representation in both diseases suggests the involvement of a viral trigger. Previous infection with EBV is one of the strongest environmental risk factors linked with MS development (105). An underlying infectious cause for GME has been sought after for decades to no avail (106108). Canid herpesvirus 1 is a well-known cause of encephalitis in young dogs, and the pathogenesis of latent infection is not well investigated (109). Furthermore, the expression of viral-induced epitopes has been described in GME, suggestive of an underlying infectious cause (110). Dysregulation of latent EBV infection may induce viral reactivation and neuropathology in MS patients (111113). EBV-transformed B cells preferentially accumulate in the leptomeninges in which they may activate CD8+ T cells to promote chronic neuroinflammation (111, 114, 115). The accumulation of B cells and CD8+ T cells in GME recapitulates elements of the leptomeningeal immune cell composition observed in MS and EBV infection and may offer a clue to an underlying etiology of GME.

Even though no single model can, by itself, recapitulate the entire spectrum of clinical and pathological features of MS, we believe GME can serve as a naturally occurring model to study the compartmentalized meningeal inflammation and resulting neuropathology seen in progressive forms of MS. Further studies aimed at elucidating the mechanisms driving progressive submeningeal injury may contribute to the development of therapeutic strategies for both GME and MS. Given that GME demonstrates B cell leptomeningeal enrichment that correlates with LME, this constitutes a significant advancement to establish biological proof of principle to target leptomeningeal inflammation. As the dog is often the large animal species used for the final safety and pharmacokinetics evaluation of novel drugs before approval for human use (116, 117), GME could offer more immediate translational benefits in the hope of halting chronic aspects of the disease and ultimately improve treatment options for GME and MS patients.

We thank the anatomic pathology and neurology residents as well as histology technicians of the Penn Vet for help in procuring and processing tissues used in this study. We also thank the clinical veterinary neurologists for contributions in the clinical diagnosis of these canine patients. We are indebted to Brian and Caris Chan for the kind gift of the confocal microscope use in this study to C.H.V. and Penn Vet. Finally, we highlight the expert input from Dr. Michael May (Biomedical Sciences, Penn Vet) regarding immune features of chronic inflammation and from Dr. Zissimos Mourelatos, Dr. MacLean Nasrallah, and Dr. Edward Lee (Perelman School of Medicine at the University of Pennsylvania) regarding feedback on the histopathological characterization of leptomeningeal immune cell infiltrates.

This work was supported by the National Institute of Health through National Institute of Neurological Disorders and Stroke Grant 5K01NS097519-03 (to J.I.A.) and National Center for Advancing Translational Sciences Grant 5UL1TR001878-04 to the Institute of Translational Medicine and Therapeutics and via the Program in Comparative Animal Biology of the University of Pennsylvania (to A.B.-O. and J.I.A.). J.I.A. held the David L. Torrey End MS Transitional Career Development Award from the Multiple Sclerosis Society of Canada.

M.E.C., G.C., M.M., M.C.M., P.F., and M.D.S. processed the samples and performed the histological and immunofluorescence experiments; M.E.C., M.D.S., and J.I.A. surveyed the School of Veterinary Medicine, University of Pennsylvania pathology bank to select the cases included in this study; M.E.C., E.G.S., C.-A.A., and J.I.A. performed neuropathological assessment and analyses; J.I.A. and M.C.M. performed microscopy analysis, with input from G.P.S.; C.H.V. performed the magnetic resonance imaging analysis of all the cases included in this study, with input from A.B.-O.; M.E.C. and J.I.A. wrote the manuscript, with input from A.B.-O. and C.H.V.; J.I.A. designed and directed the study.

The online version of this article contains supplemental material.

Abbreviations used in this article

BMB

blood–meningeal barrier

CDV

canine distemper virus

EAE

experimental autoimmune encephalomyelitis

FDC

follicular dendritic cell

FFPE

formalin-fixed and paraffin-embedded

FRC

follicular reticular cell

GC

germinal center

GME

granulomatous meningoencephalomyelitis

HEV

high endothelial venule

IHC

immunohistochemistry

LME

leptomeningeal enhancement

MRI

magnetic resonance imaging

MS

multiple sclerosis

NME

necrotizing meningoencephalitis

Penn Vet

School of Veterinary Medicine, University of Pennsylvania

PNAd

peripheral node addressin

TLO

tertiary lymphoid organ

1.
Milo
R.
,
E.
Kahana
.
2010
.
Multiple sclerosis: geoepidemiology, genetics and the environment.
Autoimmun. Rev.
9
:
A387
A394
.
2.
Compston
A.
,
A.
Coles
.
2008
.
Multiple sclerosis.
Lancet
372
:
1502
1517
.
3.
Baecher-Allan
C.
,
B. J.
Kaskow
,
H. L.
Weiner
.
2018
.
Multiple Sclerosis: Mechanisms and Immunotherapy.
Neuron
97
:
742
768
.
4.
Lucchinetti
C. F.
,
B. F.
Popescu
,
R. F.
Bunyan
,
N. M.
Moll
,
S. F.
Roemer
,
H.
Lassmann
,
W.
Brück
,
J. E.
Parisi
,
B. W.
Scheithauer
,
C.
Giannini
, et al
2011
.
Inflammatory cortical demyelination in early multiple sclerosis.
N. Engl. J. Med.
365
:
2188
2197
.
5.
Weiner
H. L.
2009
.
The challenge of multiple sclerosis: how do we cure a chronic heterogeneous disease?
Ann. Neurol.
65
:
239
248
.
6.
Dendrou
C. A.
,
L.
Fugger
,
M. A.
Friese
.
2015
.
Immunopathology of multiple sclerosis.
Nat. Rev. Immunol.
15
:
545
558
.
7.
Kuhlmann
T.
,
S.
Ludwin
,
A.
Prat
,
J.
Antel
,
W.
Brück
,
H.
Lassmann
.
2016
.
An updated histological classification system for multiple sclerosis lesions.
Acta Neuropathol.
133
:
13
24
.
8.
Magliozzi
R.
,
O.
Howell
,
A.
Vora
,
B.
Serafini
,
R.
Nicholas
,
M.
Puopolo
,
R.
Reynolds
,
F.
Aloisi
.
2007
.
Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology.
Brain
130
:
1089
1104
.
9.
Serafini
B.
,
B.
Rosicarelli
,
R.
Magliozzi
,
E.
Stigliano
,
F.
Aloisi
.
2004
.
Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis.
Brain Pathol.
14
:
164
174
.
10.
Choi
S. R.
,
O. W.
Howell
,
D.
Carassiti
,
R.
Magliozzi
,
D.
Gveric
,
P. A.
Muraro
,
R.
Nicholas
,
F.
Roncaroli
,
R.
Reynolds
.
2012
.
Meningeal inflammation plays a role in the pathology of primary progressive multiple sclerosis.
Brain
135
:
2925
2937
.
11.
Howell
O. W.
,
C. A.
Reeves
,
R.
Nicholas
,
D.
Carassiti
,
B.
Radotra
,
S. M.
Gentleman
,
B.
Serafini
,
F.
Aloisi
,
F.
Roncaroli
,
R.
Magliozzi
,
R.
Reynolds
.
2011
.
Meningeal inflammation is widespread and linked to cortical pathology in multiple sclerosis.
Brain
134
:
2755
2771
.
12.
Pitzalis
C.
,
G. W.
Jones
,
M.
Bombardieri
,
S. A.
Jones
.
2014
.
Ectopic lymphoid-like structures in infection, cancer and autoimmunity.
Nat. Rev. Immunol.
14
:
447
462
.
13.
Mitsdoerffer
M.
,
A.
Peters
.
2016
.
Tertiary Lymphoid Organs in Central Nervous System Autoimmunity.
Front. Immunol.
7
:
451
.
14.
Li
R.
,
K. R.
Patterson
,
A.
Bar-Or
.
2018
.
Reassessing B cell contributions in multiple sclerosis.
Nat. Immunol.
19
:
696
707
.
15.
Granger
N.
,
P. M.
Smith
,
N. D.
Jeffery
.
2010
.
Clinical findings and treatment of non-infectious meningoencephalomyelitis in dogs: a systematic review of 457 published cases from 1962 to 2008.
Vet. J.
184
:
290
297
.
16.
Cordy
D. R.
1979
.
Canine granulomatous meningoencephalomyelitis.
Vet. Pathol.
16
:
325
333
.
17.
Vandevelde
M.
,
R.
Fatzer
,
R.
Fankhauser
.
1981
.
Immunohistological studies on primary reticulosis of the canine brain.
Vet. Pathol.
18
:
577
588
.
18.
Coates
J. R.
,
N. D.
Jeffery
.
2014
.
Perspectives on meningoencephalomyelitis of unknown origin.
Vet. Clin. North Am. Small Anim. Pract.
44
:
1157
1185
.
19.
Vitale
S.
,
K.
Foss
.
2019
.
Immune-Mediated Central Nervous System Disease-Current Knowledge and Recommendations.
Top. Companion Anim. Med.
34
:
22
29
.
20.
Lowrie
M.
,
P. M.
Smith
,
L.
Garosi
.
2013
.
Meningoencephalitis of unknown origin: investigation of prognostic factors and outcome using a standard treatment protocol.
Vet. Rec.
172
:
527
.
21.
Braund
K. G.
,
M.
Vandevelde
,
T. L.
Walker
,
R. W.
Redding
.
1978
.
Granulomatous meningoencephalomyelitis in six dogs.
J. Am. Vet. Med. Assoc.
172
:
1195
1200
.
22.
Kipar
A.
,
W.
Baumgärtner
,
C.
Vogl
,
K.
Gaedke
,
M.
Wellman
.
1998
.
Immunohistochemical characterization of inflammatory cells in brains of dogs with granulomatous meningoencephalitis.
Vet. Pathol.
35
:
43
52
.
23.
Uchida
K.
,
E.
Park
,
M.
Tsuboi
,
J. K.
Chambers
,
H.
Nakayama
.
2016
.
Pathological and immunological features of canine necrotising meningoencephalitis and granulomatous meningoencephalitis.
Vet. J.
213
:
72
77
.
24.
Suzuki
M.
,
K.
Uchida
,
M.
Morozumi
,
T.
Hasegawa
,
T.
Yanai
,
H.
Nakayama
,
S.
Tateyama
.
2003
.
A comparative pathological study on canine necrotizing meningoencephalitis and granulomatous meningoencephalomyelitis.
J. Vet. Med. Sci.
65
:
1233
1239
.
25.
Devinsky
O.
,
J. M.
Boesch
,
S.
Cerda-Gonzalez
,
B.
Coffey
,
K.
Davis
,
D.
Friedman
,
B.
Hainline
,
K.
Houpt
,
D.
Lieberman
,
P.
Perry
, et al
2018
.
A cross-species approach to disorders affecting brain and behaviour.
Nat. Rev. Neurol.
14
:
677
686
.
26.
Amin
S. B.
,
K. J.
Anderson
,
C. E.
Boudreau
,
E.
Martinez-Ledesma
,
E.
Kocakavuk
,
K. C.
Johnson
,
F. P.
Barthel
,
F. S.
Varn
,
C.
Kassab
,
X.
Ling
, et al
2020
.
Comparative molecular life history of spontaneous canine and human gliomas.
Cancer Cell
37
:
243
257.e7
.
27.
LeBlanc
A. K.
,
C. N.
Mazcko
,
C.
Khanna
.
2015
.
Defining the value of a comparative approach to cancer drug development.
Clin. Cancer Res.
22
:
2133
2138
.
28.
LeBlanc
A. K.
,
C. N.
Mazcko
.
2020
.
Improving human cancer therapy through the evaluation of pet dogs.
Nat. Rev. Cancer
20
:
727
742
.
29.
Saba
C.
,
M.
Paoloni
,
C.
Mazcko
,
W.
Kisseberth
,
J. H.
Burton
,
A.
Smith
,
H.
Wilson-Robles
,
S.
Allstadt
,
D.
Vail
,
C.
Henry
, et al
2016
.
A comparative oncology study of iniparib defines its pharmacokinetic profile and biological activity in a naturally-occurring canine cancer model.
PLoS One
11
:
e0149194
.
30.
Panjwani
M. K.
,
M. J.
Atherton
,
M. A.
MaloneyHuss
,
K. P.
Haran
,
A.
Xiong
,
M.
Gupta
,
I.
Kulikovsaya
,
S. F.
Lacey
,
N. J.
Mason
.
2019
.
Establishing a model system for evaluating CAR T cell therapy using dogs with spontaneous diffuse large B cell lymphoma.
OncoImmunology
9
:
1676615
.
31.
Alvarez
J. I.
,
O.
Saint-Laurent
,
A.
Godschalk
,
S.
Terouz
,
C.
Briels
,
S.
Larouche
,
L.
Bourbonnière
,
C.
Larochelle
,
A.
Prat
.
2015
.
Focal disturbances in the blood-brain barrier are associated with formation of neuroinflammatory lesions.
Neurobiol. Dis.
74
:
14
24
.
32.
Alvarez
J. I.
,
C. H.
Colegial
,
C. A.
Castaño
,
J.
Trujillo
,
J. M.
Teale
,
B. I.
Restrepo
.
2002
.
The human nervous tissue in proximity to granulomatous lesions induced by Taenia solium metacestodes displays an active response.
J. Neuroimmunol.
127
:
139
144
.
33.
Alvarez
J. I.
,
H.
Kébir
,
L.
Cheslow
,
M.
Charabati
,
M.
Chabarati
,
C.
Larochelle
,
A.
Prat
.
2015
.
JAML mediates monocyte and CD8 T cell migration across the brain endothelium.
Ann. Clin. Transl. Neurol.
2
:
1032
1037
.
34.
Benallegue
N.
,
R.
Kapoor
,
H.
Kebir
,
A. M.
Crockett
,
C.
Li
,
L.
Cheslow
,
M. S.
Abdel-Hakeem
,
J.
Gesualdi
,
M. C.
Miller
,
E. J.
Wherry
,
M. E.
Church
,
M. A.
Blanco
,
J. I.
Alvarez
.
2021
.
The hedgehog pathway suppresses neuropathogenesis in CD4 T cell-driven inflammation.
Brain
:
Mar
16
;
awab083
.
35.
Church
M. E.
,
S. M.
Veluvolu
,
A. C.
Durham
,
K. D.
Woolard
.
2019
.
Clinical outcomes, ultrastructure and immunohistochemical features of canine high-grade olfactory neuroblastoma.
Vet. Comp. Oncol.
17
:
578
584
.
36.
Smirniotopoulos
J. G.
,
F. M.
Murphy
,
E. J.
Rushing
,
J. H.
Rees
,
J. W.
Schroeder
.
2007
.
Patterns of contrast enhancement in the brain and meninges.
Radiographics
27
:
525
551
.
37.
Cherubini
G. B.
,
S. R.
Platt
,
T. J.
Anderson
,
C.
Rusbridge
,
V.
Lorenzo
,
P.
Mantis
,
R.
Cappello
.
2006
.
Characteristics of magnetic resonance images of granulomatous meningoencephalomyelitis in 11 dogs.
Vet. Rec.
159
:
110
115
.
38.
Cook
S. D.
,
B.
Blumberg
,
P. C.
Dowling
,
W.
Deans
,
R.
Cross
.
1987
.
Multiple sclerosis and canine distemper on Key West, Florida.
Lancet
1
:
1426
1427
.
39.
Greer
K. A.
,
A. K.
Wong
,
H.
Liu
,
T. R.
Famula
,
N. C.
Pedersen
,
A.
Ruhe
,
M.
Wallace
,
M. W.
Neff
.
2010
.
Necrotizing meningoencephalitis of Pug dogs associates with dog leukocyte antigen class II and resembles acute variant forms of multiple sclerosis.
Tissue Antigens
76
:
110
118
.
40.
Jones
G. W.
,
D. G.
Hill
,
S. A.
Jones
.
2016
.
Understanding Immune Cells in Tertiary Lymphoid Organ Development: It Is All Starting to Come Together.
Front. Immunol.
7
:
401
.
41.
Han
S.
,
B.
Zheng
,
D. G.
Schatz
,
E.
Spanopoulou
,
G.
Kelsoe
.
1996
.
Neoteny in lymphocytes: Rag1 and Rag2 expression in germinal center B cells.
Science
274
:
2094
2097
.
42.
Dent
A. L.
,
A. L.
Shaffer
,
X.
Yu
,
D.
Allman
,
L. M.
Staudt
.
1997
.
Control of inflammation, cytokine expression, and germinal center formation by BCL-6.
Science
276
:
589
592
.
43.
Clavarino
G.
,
N.
Delouche
,
C.
Vettier
,
D.
Laurin
,
M.
Pernollet
,
T.
Raskovalova
,
J. Y.
Cesbron
,
C.
Dumestre-Pérard
,
M. C.
Jacob
.
2016
.
Novel Strategy for Phenotypic Characterization of Human B Lymphocytes from Precursors to Effector Cells by Flow Cytometry.
PLoS One
11
:
e0162209
.
44.
Luo
R. F.
,
S.
Zhao
,
R.
Tibshirani
,
J. H.
Myklebust
,
M.
Sanyal
,
R.
Fernandez
,
D.
Gratzinger
,
R. J.
Marinelli
,
Z. S.
Lu
,
A.
Wong
, et al
2010
.
CD81 protein is expressed at high levels in normal germinal center B cells and in subtypes of human lymphomas.
Hum. Pathol.
41
:
271
280
.
45.
Steiniger
B. S.
2015
.
Human spleen microanatomy: why mice do not suffice.
Immunology
145
:
334
346
.
46.
Steiniger
B. S.
,
V.
Wilhelmi
,
A.
Seiler
,
K.
Lampp
,
V.
Stachniss
.
2014
.
Heterogeneity of stromal cells in the human splenic white pulp. Fibroblastic reticulum cells, follicular dendritic cells and a third superficial stromal cell type.
Immunology
143
:
462
477
.
47.
Beckenkamp
L. R.
,
L. E. B.
Souza
,
F. U. F.
Melo
,
C. H.
Thomé
,
D. A. R.
Magalhães
,
P. V. B.
Palma
,
D. T.
Covas
.
2018
.
Comparative characterization of CD271+ and CD271- subpopulations of CD34+ human adipose-derived stromal cells.
J. Cell. Biochem.
119
:
3873
3884
.
48.
Drayton
D. L.
,
S.
Liao
,
R. H.
Mounzer
,
N. H.
Ruddle
.
2006
.
Lymphoid organ development: from ontogeny to neogenesis.
Nat. Immunol.
7
:
344
353
.
49.
Ager
A.
,
M. J.
May
.
2015
.
Understanding high endothelial venules: Lessons for cancer immunology.
OncoImmunology
4
:
e1008791
.
50.
Streeter
P. R.
,
B. T.
Rouse
,
E. C.
Butcher
.
1988
.
Immunohistologic and functional characterization of a vascular addressin involved in lymphocyte homing into peripheral lymph nodes.
J. Cell Biol.
107
:
1853
1862
.
51.
Ager
A.
2017
.
High endothelial venules and other blood vessels: critical regulators of lymphoid organ development and function.
Front. Immunol.
8
:
45
.
52.
Luo
S.
,
R.
Zhu
,
T.
Yu
,
H.
Fan
,
Y.
Hu
,
S. K.
Mohanta
,
D.
Hu
.
2019
.
Chronic inflammation: a common promoter in tertiary lymphoid organ neogenesis.
Front. Immunol.
10
:
2938
.
53.
Mueller
S. N.
,
R. N.
Germain
.
2009
.
Stromal cell contributions to the homeostasis and functionality of the immune system.
Nat. Rev. Immunol.
9
:
618
629
.
54.
Peters
A.
,
L. A.
Pitcher
,
J. M.
Sullivan
,
M.
Mitsdoerffer
,
S. E.
Acton
,
B.
Franz
,
K.
Wucherpfennig
,
S.
Turley
,
M. C.
Carroll
,
R. A.
Sobel
, et al
2011
.
Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation.
Immunity
35
:
986
996
.
55.
Sixt
M.
,
N.
Kanazawa
,
M.
Selg
,
T.
Samson
,
G.
Roos
,
D. P.
Reinhardt
,
R.
Pabst
,
M. B.
Lutz
,
L.
Sorokin
.
2005
.
The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node.
Immunity
22
:
19
29
.
56.
Link
A.
,
D. L.
Hardie
,
S.
Favre
,
M. R.
Britschgi
,
D. H.
Adams
,
M.
Sixt
,
J. G.
Cyster
,
C. D.
Buckley
,
S. A.
Luther
.
2011
.
Association of T-zone reticular networks and conduits with ectopic lymphoid tissues in mice and humans.
Am. J. Pathol.
178
:
1662
1675
.
57.
L.
,
C. A.
Vedeler
,
H. I.
Nyland
,
B. D.
Trapp
,
S. J.
Mørk
.
2003
.
Subpial demyelination in the cerebral cortex of multiple sclerosis patients.
J. Neuropathol. Exp. Neurol.
62
:
723
732
.
58.
Johns
T. G.
,
C. C. A.
Bernard
.
1999
.
The structure and function of myelin oligodendrocyte glycoprotein.
J. Neurochem.
72
:
1
9
.
59.
Magliozzi
R.
,
O. W.
Howell
,
R.
Nicholas
,
C.
Cruciani
,
M.
Castellaro
,
C.
Romualdi
,
S.
Rossi
,
M.
Pitteri
,
M. D.
Benedetti
,
A.
Gajofatto
, et al
2018
.
Inflammatory intrathecal profiles and cortical damage in multiple sclerosis.
Ann. Neurol.
83
:
739
755
.
60.
Machado-Santos
J.
,
E.
Saji
,
A. R.
Tröscher
,
M.
Paunovic
,
R.
Liblau
,
G.
Gabriely
,
C. G.
Bien
,
J.
Bauer
,
H.
Lassmann
.
2018
.
The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8+ T lymphocytes and B cells.
Brain
141
:
2066
2082
.
61.
Kutzelnigg
A.
,
C. F.
Lucchinetti
,
C.
Stadelmann
,
W.
Brück
,
H.
Rauschka
,
M.
Bergmann
,
M.
Schmidbauer
,
J. E.
Parisi
,
H.
Lassmann
.
2005
.
Cortical demyelination and diffuse white matter injury in multiple sclerosis.
Brain
128
:
2705
2712
.
62.
Park
E. S.
,
K.
Uchida
,
H.
Nakayama
.
2012
.
Comprehensive immunohistochemical studies on canine necrotizing meningoencephalitis (NME), necrotizing leukoencephalitis (NLE), and granulomatous meningoencephalomyelitis (GME).
Vet. Pathol.
49
:
682
692
.
63.
Vincent
T.
,
P.
Saikali
,
R.
Cayrol
,
A. D.
Roth
,
A.
Bar-Or
,
A.
Prat
,
J. P.
Antel
.
2008
.
Functional consequences of neuromyelitis optica-IgG astrocyte interactions on blood-brain barrier permeability and granulocyte recruitment.
J. Immunol.
181
:
5730
5737
.
64.
Hauser
S. L.
,
E.
Waubant
,
D. L.
Arnold
,
T.
Vollmer
,
J.
Antel
,
R. J.
Fox
,
A.
Bar-Or
,
M.
Panzara
,
N.
Sarkar
,
S.
Agarwal
, et al
HERMES Trial Group
.
2008
.
B-cell depletion with rituximab in relapsing-remitting multiple sclerosis.
N. Engl. J. Med.
358
:
676
688
.
65.
Comi
G.
,
A.
Bar-Or
,
H.
Lassmann
,
A.
Uccelli
,
H. P.
Hartung
,
X.
Montalban
,
P. S.
Sørensen
,
R.
Hohlfeld
,
S. L.
Hauser
;
Expert Panel of the 27th Annual Meeting of the European Charcot Foundation
.
2021
.
Role of B cells in multiple sclerosis and related disorders.
Ann. Neurol.
89
:
13
23
.
66.
Magliozzi
R.
,
B.
Serafini
,
B.
Rosicarelli
,
G.
Chiappetta
,
C.
Veroni
,
R.
Reynolds
,
F.
Aloisi
.
2013
.
B-cell enrichment and Epstein-Barr virus infection in inflammatory cortical lesions in secondary progressive multiple sclerosis.
J. Neuropathol. Exp. Neurol.
72
:
29
41
.
67.
Krumbholz
M.
,
D.
Theil
,
F.
Steinmeyer
,
S.
Cepok
,
B.
Hemmer
,
M.
Hofbauer
,
C.
Farina
,
T.
Derfuss
,
A.
Junker
,
T.
Arzberger
, et al
2007
.
CCL19 is constitutively expressed in the CNS, up-regulated in neuroinflammation, active and also inactive multiple sclerosis lesions.
J. Neuroimmunol.
190
:
72
79
.
68.
Sabatino
J. J.
 Jr.
,
A. K.
Pröbstel
,
S. S.
Zamvil
.
2019
.
B cells in autoimmune and neurodegenerative central nervous system diseases. [Published erratum appears in 2020 Nat. Rev. Neurosci. 21: 56].
Nat. Rev. Neurosci.
20
:
728
745
.
69.
Pikor
N. B.
,
J. L.
Astarita
,
L.
Summers-Deluca
,
G.
Galicia
,
J.
Qu
,
L. A.
Ward
,
S.
Armstrong
,
C. X.
Dominguez
,
D.
Malhotra
,
B.
Heiden
, et al
2015
.
Integration of Th17- and lymphotoxin-derived signals initiates meningeal-resident stromal cell remodeling to propagate neuroinflammation.
Immunity
43
:
1160
1173
.
70.
Dang
A. K.
,
Y.
Tesfagiorgis
,
R. W.
Jain
,
H. C.
Craig
,
S. M.
Kerfoot
.
2015
.
Meningeal infiltration of the spinal cord by non-classically activated B cells is associated with chronic disease course in a spontaneous B cell-dependent model of CNS autoimmune disease.
Front. Immunol.
6
:
470
.
71.
Kuerten
S.
,
A.
Schickel
,
C.
Kerkloh
,
M. S.
Recks
,
K.
Addicks
,
N. H.
Ruddle
,
P. V.
Lehmann
.
2012
.
Tertiary lymphoid organ development coincides with determinant spreading of the myelin-specific T cell response.
Acta Neuropathol.
124
:
861
873
.
72.
Terry
R. L.
,
I.
Ifergan
,
S. D.
Miller
.
2016
.
Experimental autoimmune encephalomyelitis in mice.
Methods Mol. Biol.
1304
:
145
160
.
73.
Simmons
S. B.
,
E. R.
Pierson
,
S. Y.
Lee
,
J. M.
Goverman
.
2013
.
Modeling the heterogeneity of multiple sclerosis in animals.
Trends Immunol.
34
:
410
422
.
74.
Schläger
C.
,
H.
Körner
,
M.
Krueger
,
S.
Vidoli
,
M.
Haberl
,
D.
Mielke
,
E.
Brylla
,
T.
Issekutz
,
C.
Cabañas
,
P. J.
Nelson
, et al
2016
.
Effector T-cell trafficking between the leptomeninges and the cerebrospinal fluid.
Nature
530
:
349
353
.
75.
Russi
A. E.
,
M. A.
Brown
.
2015
.
The meninges: new therapeutic targets for multiple sclerosis.
Transl. Res.
165
:
255
269
.
76.
DeSisto
J.
,
R.
O’Rourke
,
H. E.
Jones
,
B.
Pawlikowski
,
A. D.
Malek
,
S.
Bonney
,
F.
Guimiot
,
K. L.
Jones
,
J. A.
Siegenthaler
.
2020
.
Single-cell transcriptomic analyses of the developing meninges reveal meningeal fibroblast diversity and function.
Dev. Cell.
54
:
43
59.e4
.
77.
Alvarez
J. I.
,
J. M.
Teale
.
2007
.
Evidence for differential changes of junctional complex proteins in murine neurocysticercosis dependent upon CNS vasculature.
Brain Res.
1169
:
98
111
.
78.
Bartholomäus
I.
,
N.
Kawakami
,
F.
Odoardi
,
C.
Schläger
,
D.
Miljkovic
,
J. W.
Ellwart
,
W. E.
Klinkert
,
C.
Flügel-Koch
,
T. B.
Issekutz
,
H.
Wekerle
,
A.
Flügel
.
2009
.
Effector T cell interactions with meningeal vascular structures in nascent autoimmune CNS lesions.
Nature
462
:
94
98
.
79.
Michel
L.
,
C.
Grasmuck
,
M.
Charabati
,
M. A.
Lécuyer
,
S.
Zandee
,
T.
Dhaeze
,
J. I.
Alvarez
,
R.
Li
,
S.
Larouche
,
L.
Bourbonnière
, et al
2019
.
Activated leukocyte cell adhesion molecule regulates B lymphocyte migration across central nervous system barriers.
Sci. Transl. Med.
11
:
eaaw0475
.
80.
Alvarez
J. I.
,
H.
Kebir
,
L.
Cheslow
,
S.
Terouz
,
L.
Bourbonniere
,
A.
Prat
.
2016
.
The blood meningeal barrier orchestrates the immune response in multiple sclerosis.
ECTRIMS.
400
705
.
81.
Alvarez
J. I.
,
J. M.
Teale
.
2008
.
Multiple expression of matrix metalloproteinases in murine neurocysticercosis: Implications for leukocyte migration through multiple central nervous system barriers.
Brain Res.
1214
:
145
158
.
82.
Jones
G. W.
,
S. A.
Jones
.
2015
.
Ectopic lymphoid follicles: inducible centres for generating antigen-specific immune responses within tissues.
Immunology
147
:
141
151
.
83.
Park
E. S.
,
K.
Uchida
,
H.
Nakayama
.
2013
.
Th1-, Th2-, and Th17-related cytokine and chemokine receptor mRNA and protein expression in the brain tissues, T cells, and macrophages of dogs with necrotizing and granulomatous meningoencephalitis.
Vet. Pathol.
50
:
1127
1134
.
84.
Babbe
H.
,
A.
Roers
,
A.
Waisman
,
H.
Lassmann
,
N.
Goebels
,
R.
Hohlfeld
,
M.
Friese
,
R.
Schröder
,
M.
Deckert
,
S.
Schmidt
, et al
2000
.
Clonal expansions of CD8(+) T cells dominate the T cell infiltrate in active multiple sclerosis lesions as shown by micromanipulation and single cell polymerase chain reaction.
J. Exp. Med.
192
:
393
404
.
85.
Booss
J.
,
M. M.
Esiri
,
W. W.
Tourtellotte
,
D. Y.
Mason
.
1983
.
Immunohistological analysis of T lymphocyte subsets in the central nervous system in chronic progressive multiple sclerosis.
J. Neurol. Sci.
62
:
219
232
.
86.
Hauser
S. L.
,
A. K.
Bhan
,
F.
Gilles
,
M.
Kemp
,
C.
Kerr
,
H. L.
Weiner
.
1986
.
Immunohistochemical analysis of the cellular infiltrate in multiple sclerosis lesions.
Ann. Neurol.
19
:
578
587
.
87.
Segal
B. M.
2019
.
The diversity of encephalitogenic CD4+ T cells in multiple sclerosis and its animal models.
J. Clin. Med.
8
:
120
.
88.
Lolli
F.
,
H.
Martini
,
A.
Citro
,
D.
Franceschini
,
E.
Portaccio
,
M. P.
Amato
,
R.
Mechelli
,
V.
Annibali
,
J.
Sidney
,
A.
Sette
, et al
2013
.
Increased CD8+ T cell responses to apoptotic T cell-associated antigens in multiple sclerosis.
J. Neuroinflammation
10
:
94
.
89.
Huber
M.
,
S.
Heink
,
A.
Pagenstecher
,
K.
Reinhard
,
J.
Ritter
,
A.
Visekruna
,
A.
Guralnik
,
N.
Bollig
,
K.
Jeltsch
,
C.
Heinemann
, et al
2012
.
IL-17A secretion by CD8+ T cells supports Th17-mediated autoimmune encephalomyelitis.
J. Clin. Invest.
123
:
247
260
.
90.
Larochelle
C.
,
M. A.
Lecuyer
,
J. I.
Alvarez
,
M.
Charabati
,
O.
Saint-Laurent
,
S.
Ghannam
,
H.
Kebir
,
K.
Flanagan
,
T.
Yednock
,
P.
Duquette
,
N.
Arbour
,
A.
Prat
.
2015
.
MCAM CD8 T lymphocytes mediate CNS inflammation.
Ann Neurol.
91.
Tzartos
J. S.
,
M. A.
Friese
,
M. J.
Craner
,
J.
Palace
,
J.
Newcombe
,
M. M.
Esiri
,
L.
Fugger
.
2008
.
Interleukin-17 production in central nervous system-infiltrating T cells and glial cells is associated with active disease in multiple sclerosis.
Am. J. Pathol.
172
:
146
155
.
92.
Ifergan
I.
,
H.
Kebir
,
J. I.
Alvarez
,
G.
Marceau
,
M.
Bernard
,
L.
Bourbonnière
,
J.
Poirier
,
P.
Duquette
,
P. J.
Talbot
,
N.
Arbour
,
A.
Prat
.
2011
.
Central nervous system recruitment of effector memory CD8+ T lymphocytes during neuroinflammation is dependent on α4 integrin.
Brain
134
:
3560
3577
.
93.
Salou
M.
,
B.
Nicol
,
A.
Garcia
,
D. A.
Laplaud
.
2015
.
Involvement of CD8(+) T cells in multiple sclerosis.
Front. Immunol.
6
:
604
.
94.
Bitsch
A.
,
J.
Schuchardt
,
S.
Bunkowski
,
T.
Kuhlmann
,
W.
Brück
.
2000
.
Acute axonal injury in multiple sclerosis. Correlation with demyelination and inflammation.
Brain
123
:
1174
1183
.
95.
Göbel
K.
,
N.
Melzer
,
A. M.
Herrmann
,
M. K.
Schuhmann
,
S.
Bittner
,
C. W.
Ip
,
T.
Hünig
,
S. G.
Meuth
,
H.
Wiendl
.
2010
.
Collateral neuronal apoptosis in CNS gray matter during an oligodendrocyte-directed CD8(+) T cell attack.
Glia
58
:
469
480
.
96.
Lisak
R. P.
,
L.
Nedelkoska
,
J. A.
Benjamins
,
D.
Schalk
,
B.
Bealmear
,
H.
Touil
,
R.
Li
,
G.
Muirhead
,
A.
Bar-Or
.
2017
.
B cells from patients with multiple sclerosis induce cell death via apoptosis in neurons in vitro.
J. Neuroimmunol.
309
:
88
99
.
97.
Neumann
H.
,
I. M.
Medana
,
J.
Bauer
,
H.
Lassmann
.
2002
.
Cytotoxic T lymphocytes in autoimmune and degenerative CNS diseases.
Trends Neurosci.
25
:
313
319
.
98.
Schirmer
L.
,
D.
Velmeshev
,
S.
Holmqvist
,
M.
Kaufmann
,
S.
Werneburg
,
D.
Jung
,
S.
Vistnes
,
J. H.
Stockley
,
A.
Young
,
M.
Steindel
, et al
2019
.
Neuronal vulnerability and multilineage diversity in multiple sclerosis.
Nature
573
:
75
82
.
99.
Faissner
S.
,
J. R.
Plemel
,
R.
Gold
,
V. W.
Yong
.
2019
.
Progressive multiple sclerosis: from pathophysiology to therapeutic strategies.
Nat. Rev. Drug Discov.
18
:
905
922
.
100.
Lassmann
H.
2019
.
Pathogenic Mechanisms Associated With Different Clinical Courses of Multiple Sclerosis.
Front. Immunol.
9
:
3116
.
101.
Gilmore
C. P.
,
I.
Donaldson
,
L.
,
T.
Owens
,
J.
Lowe
,
N.
Evangelou
.
2009
.
Regional variations in the extent and pattern of grey matter demyelination in multiple sclerosis: a comparison between the cerebral cortex, cerebellar cortex, deep grey matter nuclei and the spinal cord.
J. Neurol. Neurosurg. Psychiatry
80
:
182
187
.
102.
Haider
L.
,
T.
Zrzavy
,
S.
Hametner
,
R.
Höftberger
,
F.
Bagnato
,
G.
Grabner
,
S.
Trattnig
,
S.
Pfeifenbring
,
W.
Brück
,
H.
Lassmann
.
2016
.
The topograpy of demyelination and neurodegeneration in the multiple sclerosis brain.
Brain
139
:
807
815
.
103.
Lassmann
H.
,
M.
Bradl
.
2016
.
Multiple sclerosis: experimental models and reality.
Acta Neuropathol.
133
:
223
244
.
104.
Bevan
R. J.
,
R.
Evans
,
L.
Griffiths
,
L. M.
Watkins
,
M. I.
Rees
,
R.
Magliozzi
,
I.
Allen
,
G.
McDonnell
,
R.
Kee
,
M.
Naughton
, et al
2018
.
Meningeal inflammation and cortical demyelination in acute multiple sclerosis.
Ann. Neurol.
84
:
829
842
.
105.
Belbasis
L.
,
V.
Bellou
,
E.
Evangelou
,
J. P.
Ioannidis
,
I.
Tzoulaki
.
2015
.
Environmental risk factors and multiple sclerosis: an umbrella review of systematic reviews and meta-analyses.
Lancet Neurol.
14
:
263
273
.
106.
Hoon-Hanks
L. L.
,
S.
McGrath
,
K. L.
Tyler
,
C.
Owen
,
M. D.
Stenglein
.
2017
.
Metagenomic investigation of idiopathic meningoencephalomyelitis in dogs.
J. Vet. Intern. Med.
32
:
324
330
.
107.
Collinet
A.
,
G.
Garcia
,
J.
Wellehan
,
A.
Childress
,
S.
Carrera-Justiz
.
2019
.
Investigation of astrovirus and bornavirus in the cerebrospinal fluid of dogs clinically diagnosed with meningoencephalitis of unknown etiology.
J. Vet. Intern. Med.
34
:
232
236
.
108.
Barber
R. M.
,
B. F.
Porter
,
Q.
Li
,
M.
May
,
M. K.
Claiborne
,
A. B.
Allison
,
E. W.
Howerth
,
A.
Butler
,
S.
Wei
,
J. M.
Levine
, et al
2012
.
Broadly reactive polymerase chain reaction for pathogen detection in canine granulomatous meningoencephalomyelitis and necrotizing meningoencephalitis.
J. Vet. Intern. Med.
26
:
962
968
.
109.
Jager
M. C.
,
E. A.
Sloma
,
M.
Shelton
,
A. D.
Miller
.
2017
.
Naturally acquired canine herpesvirus-associated meningoencephalitis.
Vet. Pathol.
54
:
820
827
.
110.
Porter
B. F.
,
A.
Ambrus
,
R. W.
Storts
.
2006
.
Immunohistochemical evaluation of mx protein expression in canine encephalitides.
Vet. Pathol.
43
:
981
987
.
111.
Serafini
B.
,
B.
Rosicarelli
,
D.
Franciotta
,
R.
Magliozzi
,
R.
Reynolds
,
P.
Cinque
,
L.
Andreoni
,
P.
Trivedi
,
M.
Salvetti
,
A.
Faggioni
,
F.
Aloisi
.
2007
.
Dysregulated Epstein-Barr virus infection in the multiple sclerosis brain.
J. Exp. Med.
204
:
2899
2912
.
112.
Serafini
B.
,
B.
Rosicarelli
,
C.
Veroni
,
G. A.
Mazzola
,
F.
Aloisi
.
2019
.
Epstein-Barr virus-specific CD8 T cells selectively infiltrate the brain in multiple sclerosis and interact locally with virus-infected cells: clue for a virus-driven immunopathological mechanism.
J. Virol.
93
:
e00980-19
.
113.
Tzartos
J. S.
,
G.
Khan
,
A.
Vossenkamper
,
M.
Cruz-Sadaba
,
S.
Lonardi
,
E.
Sefia
,
A.
Meager
,
A.
Elia
,
J. M.
Middeldorp
,
M.
Clemens
, et al
2011
.
Association of innate immune activation with latent Epstein-Barr virus in active MS lesions.
Neurology
78
:
15
23
.
114.
Veroni
C.
,
B.
Serafini
,
B.
Rosicarelli
,
C.
Fagnani
,
F.
Aloisi
.
2018
.
Transcriptional profile and Epstein-Barr virus infection status of laser-cut immune infiltrates from the brain of patients with progressive multiple sclerosis.
J. Neuroinflammation
15
:
18
.
115.
Serafini
B.
,
M.
Severa
,
S.
Columba-Cabezas
,
B.
Rosicarelli
,
C.
Veroni
,
G.
Chiappetta
,
R.
Magliozzi
,
R.
Reynolds
,
E. M.
Coccia
,
F.
Aloisi
.
2010
.
Epstein-Barr virus latent infection and BAFF expression in B cells in the multiple sclerosis brain: implications for viral persistence and intrathecal B-cell activation.
J. Neuropathol. Exp. Neurol.
69
:
677
693
.
116.
Trepanier
L. A.
2013
.
Applying pharmacokinetics to veterinary clinical practice.
Vet. Clin. North Am. Small Anim. Pract.
43
:
1013
1026
.
117.
Van den Bergh
A.
,
S.
Van Hemelryck
,
J.
Bevernage
,
A.
Van Peer
,
M.
Brewster
,
C.
Mackie
,
E.
Mannaert
.
2018
.
Preclinical bioavailability strategy for decisions on clinical drug formulation development: an in depth analysis.
Mol. Pharm.
15
:
2633
2645
.

The authors have no financial conflicts of interest.

Supplementary data