throbber
Glatiramer acetate-specific T cells in the brain
`express T helper 2兾3 cytokines and
`brain-derived neurotrophic factor insitu
`
`Rina Aharoni*, Basak Kayhan*, Raya Eilam†, Michael Sela*‡, and Ruth Arnon*
`
`Departments of *Immunology and †Veterans Resources, The Weizmann Institute, Rehovot 76100, Israel
`
`involved in the therapeutic effect induced by GA on MS as well
`(5–7). Furthermore, we have recently demonstrated that GA-
`specific regulatory cells, induced in the periphery either by
`injection or by oral treatment with GA, pass the blood–brain
`barrier and accumulate in the CNS. This finding was manifested
`by their isolation from brains of actively sensitized GA-treated
`mice and by localization of GA-specific cells in the brain after
`their passive transfer to the periphery (8, 9).
`Although the presence of GA-specific Th2 cells in the CNS
`was confirmed, their ability to actually secrete Th2 cytokines and
`induce therapeutic effect in situ has not been verified. Some
`reservation concerning the capability of T cells to secrete Th2
`cytokines in the CNS was raised after finding that the CNS
`environment induces bias toward the Th1 pathway and that
`myelin basic protein-specific Th2 cells can aggravate EAE (6,
`10). It was therefore essential to elucidate whether the GA-
`specific cells accumulating in the brain actually function as
`suppressor cells in the diseased organ and secrete antiinflam-
`matory cytokines in situ.
`It was recently demonstrated that GA-specific cells of both
`mouse and human origin also secrete the potent brain-derived
`neurotrophic factor (BDNF) (3, 11), which induces axonal
`outgrowth, remyelination, regeneration, and rescue of degener-
`ating neurons (12, 13). Yet, this BDNF secretion was found only
`in peripheral T cell lines in vitro and not in the target organ.
`BDNF expression by GA-specific cells in the CNS may indicate
`that GA activity is not restricted to antiinflammatory effect but
`generates actual neuroprotection and regeneration processes as
`well. In this study, we investigated whether GA-induced T cells
`that penetrate the brain do function as regulatory cells by
`producing Th2兾3 cytokines and neurotrophic factor. We wish to
`report that, indeed, the cytokines IL-10, transforming growth
`factor ␤(TGF-␤), and BDNF are expressed by GA-induced cells
`in situ.
`
`Materials and Methods
`Mice. (SJL兾JxBALB兾c)F1 female mice, 10–16 weeks old, were
`purchased from The Jackson Laboratory.
`
`Glatiramer Acetate. GA consists of acetate salts of synthetic
`polypeptides, containing four amino acids: L-alanine, L-
`glutamate, L-lysine, and L-tyrosine (4). GA from batch no.
`242990599, with an average molecular weight of 7,300, was
`obtained from Teva Pharmaceutical Industries (Petach Tikva,
`Israel) and used throughout the study.
`
`T Cells. GA-specific cells from two T cell lines established from
`spleens of mice that had received daily injections (2 mg兾day, 10
`
`IMMUNOLOGY
`
`Abbreviations: GA, glatiramer acetate; BDNF, brain-derived neurotrophic factor; TGF-␤,
`transforming growth factor ␤; MS, multiple sclerosis; EAE, experimental autoimmune
`encephalomyelitis; GFAP, glial fibrillary acidic protein; Th, T helper.
`‡To whom correspondence should be addressed. E-mail: michael.sela@weizmann.ac.il.
`
`© 2003 by The National Academy of Sciences of the USA
`
`Contributed by Michael Sela, September 25, 2003
`
`The ability of a remedy to modulate the pathological process in the
`target organ is crucial for its therapeutic activity. Glatiramer
`acetate (GA, Copaxone, Copolymer 1), a drug approved for the
`treatment of multiple sclerosis, induces regulatory T helper 2兾3
`cells that penetrate the CNS. Here we investigated whether these
`GA-specific T cells can function as suppressor cells with therapeutic
`potential in the target organ by in situ expression of T helper 2兾3
`cytokines and neurotrophic factors. GA-specific cells and their in
`situ expression were detected on the level of whole-brain tissue by
`using a two-stage double-labeling system: (i) labeling of the
`GA-specific T cells, followed by their adoptive transfer, and (ii)
`detection of the secreted factors in the brain by immunohistolog-
`ical methods. GA-specific T cells in the CNS demonstrated intense
`expression of the brain-derived neurotrophic factor and of two
`antiinflammatory cytokines, IL-10 and transforming growth factor
`␤. No expression of the inflammatory cytokine IFN-␥was observed.
`This pattern of expression was manifested in brains of normal and
`experimental autoimmune encephalomyelitis-induced mice to
`which GA-specific cells were adoptively transferred, but not in
`control mice. Furthermore, infiltration of GA-induced cells to the
`brain resulted in bystander expression of IL-10 and transforming
`growth factor ␤by resident astrocytes and microglia. The ability of
`infiltrating GA-specific cells to express antiinflammatory cytokines
`and neurotrophic factor in the organ in which the pathological
`processes occur correlates directly with the therapeutic activity
`of GA in experimental autoimmune encephalomyelitis兾multiple
`sclerosis.
`
`Neurotrophic factors and antiinflammatory cytokines have
`
`been shown to play a crucial role in the modulation of
`multiple sclerosis (MS) and its experimental model, experimen-
`tal autoimmune encephalomyelitis (EAE) (1–3). Yet, the infor-
`mation available on their expression within the CNS is limited.
`Various current therapies for MS attempt to interfere with the
`pathological processes by immune intervention and up-
`regulation of these regulatory substances. However, whereas the
`peripheral effect of these treatments has been extensively stud-
`ied, little information exists on their effect in the target organ,
`namely the brain. The study of CNS diseases is particularly
`complicated, because the accessibility of immune factors and
`regulatory cells is strictly controlled by the blood–brain barrier,
`constituting a distinct and unique microenvironment.
`Glatiramer acetate (GA, Copaxone, Copolymer 1), a drug
`approved for the treatment of MS, exerts a marked suppressive
`effect on EAE induced by various encephalitogens in several
`species (4). We previously demonstrated that the therapeutic
`effect of GA is mediated by regulatory cells that suppress disease
`and secrete high amounts of antiinflammatory T helper (Th) 2兾3
`cytokines (5). These GA-induced suppressor cells crossreact
`with the autoantigen myelin basic protein by secreting Th2兾3
`cytokines and induce bystander suppression of processes medi-
`ated by other encephalitogens (6). GA-specific Th2兾3 cells have
`been found in the periphery, in the spleens and lymph nodes of
`experimental animals, and in peripheral blood mononuclear
`cells of humans treated with GA, suggesting that these cells are
`
`www.pnas.org兾cgi兾doi兾10.1073兾pnas.2336171100
`
`PNAS 兩 November 25, 2003 兩 vol. 100 兩 no. 24 兩 14157–14162
`
`MYLAN PHARMS. INC. EXHIBIT 1084 PAGE 1
`
`

`
`Immunohistochemical analysis of BDNF expression by GA-specific cells in the brain. Activated labeled GA-specific cells were injected into the peritoneum
`Fig. 1.
`of either EAE-induced (A–C, G–J, and M–O) or normal (D–F) mice. After 7 days, the mice were perfused and brain sections (20 ␮m) were stained immunocyto-
`chemically for BDNF expression. (A–C) Area surrounding the lateral ventricle. (D–F) Choroid plexus in the lateral ventricle. (G–I) Perivascular infiltration in the
`cortex. (J) Overlap image of enlarged Hoechst-labeled, GA-specific cells and their BDNF secretion in the cortex. (M–O) Enlarged images of single GA-specific
`PK-labeled cells in the cortex. Controls demonstrate BDNF expression in corresponding brain regions of an EAE-induced mouse (K) and a normal mouse (L). (Scale
`bar: A–F, K, and L, 100 ␮m; G–I, 20 ␮m; J and M–O, 10 ␮m.)
`
`injections) or had been fed (250 ␮g兾day, eight feedings) with
`GA, respectively, were used throughout this study. Cells were
`selected in vitro by repeating exposures to GA (50 ␮g兾ml) on
`irradiated spleen cells, followed by propagation in T cell growth
`factor medium. These cells had been characterized as Th2
`regulatory cells that can suppress EAE in vivo (5, 6).
`
`T Cell Labeling. GA-specific cells were stained either by Hoechst
`33342-blue fluorescent (Molecular Probes) or by PKH26-red
`fluorescent (Sigma), which incorporate to the cell nucleus or to
`the cell membrane, respectively, according to the manufacturer’s
`instructions. Both staining methods did not interfere essentially
`with the biological activity of the cells, as demonstrated by the
`comparison of the proliferation and cytokine secretion of la-
`beled cells to unlabeled cells and by the ability of the labeled cells
`to prevent EAE.
`
`Adoptive Transfer. Activated prelabeled GA-specific T cells (30 ⫻
`106 per mouse) were injected into the peritoneum of either
`normal or EAE-induced mice 3 days after disease induction by
`whole spinal cord homogenate (5 mg in complete Freund’s
`adjuvant) and pertussis toxin injection (0.2 ␮g per mouse).
`Brains were excised 7 days after cell
`injection because our
`previous studies indicated that this time is optimal for the
`accumulation of GA-specific cells, in contrast to nonspecific
`controls (8, 9).
`
`Immunocytochemistry. Control EAE-induced mice injected with
`GA-specific T cells and normal mice were anesthetized and per-
`fused with 2.5% paraformaldehyde. Brains were sectioned (20 ␮m)
`by cryostat or sliding microtom. Sections were preincubated in PBS
`with 20% horse serum and 0.05% Saponin (Sigma) for 1 h, then
`incubated overnight with the following primary antibodies: chicken
`
`14158 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.2336171100
`
`Aharoni et al.
`
`MYLAN PHARMS. INC. EXHIBIT 1084 PAGE 2
`
`

`
`Immunohistochemical analysis of IL-10 expression by GA-specific cells in brains of EAE-induced mice. (A–F) Perivascular infiltration in the thalamus. (G–I)
`Fig. 2.
`Enlarged images of single cells in the thalamus. (L–N) Single cells in the cortex. (O–R) Nonoverlapping distribution of IL-10-expressing, GA-labeled cells and
`additional population of cells expressing IL-10 and GFAP in tissue surrounding the lateral ventricle. Controls demonstrate IL-10 expression in corresponding brain
`regions of an EAE-induced mouse (J) and a normal mouse (K). (Scale bar: A–C, 50 ␮m; D–F, 20 ␮m; G–I and L–N, 10 ␮m; J, K, and O–R, 100 ␮m.)
`
`IMMUNOLOGY
`
`anti-BDNF (Promega), goat anti-IL-10 (Santa Cruz Biotechnolo-
`gy), rabbit anti-TGF-␤(Santa Cruz Biotechnology), rat anti-IFN-␥
`(BioSource International, Camarillo, CA), or mouse anti-glial
`fibrillary acidic protein (GFAP; Pharmingen), 1–10 ␮g兾ml, diluted
`in PBS with 2% horse serum and 0.05% saponin. The second
`antibody step was performed by labeling with Cy2- or Cy3-
`conjugated donkey anti-chicken, donkey anti-goat, donkey anti-
`rabbit, donkey anti-rat, or donkey anti-mouse (Jackson ImmunoRe-
`search), 1:250, for 20 min. Additional controls were incubated with
`secondary antibody alone. Slides of Hoechst-labeled cells were
`examined by fluorescence microscope (Nikon Eclipse E600). Slides
`
`of PKH26-labeled cells were examined by confocal microscope
`(Axiovert 100M, Zeiss). Photographs were taken by using SPOT
`software program. Images were processed with PHOTOSHOP (Ado-
`be Systems, Mountain View, CA). Most photographs presented are
`from EAE-induced mice injected with prelabeled GA-specific cells.
`
`Results
`The Presence of Fluorescent-Labeled Adoptively Transferred GA-
`Specific T Cells in the Brain. Abundance of fluorescently prelabeled
`GA-specific cells was observed in brain sections with adoptively
`transferred GA-specific T cells in normal mice and particularly
`
`Aharoni et al.
`
`PNAS 兩 November 25, 2003 兩 vol. 100 兩 no. 24 兩 14159
`
`MYLAN PHARMS. INC. EXHIBIT 1084 PAGE 3
`
`

`
`Immunohistochemical analysis of TGF-␤expression by GA-specific cells in brains of EAE-induced mice. (A–D) Perivascular infiltration in the cortex. (G–I)
`Fig. 3.
`3D reconstruction of PK-labeled cells in the thalamus by confocal scanning microscope. (J–M) Nonoverlapping distribution of TGF-␤-expressing GA-labeled cells
`and additional population of cells expressing TGF-␤and GFAP in tissue surrounding a blood vessel. Controls demonstrate TGF-␤in corresponding brain region
`of an EAE-induced mouse (E) and a normal mouse (F) in the cortex. (Scale bar: A–C, 50 ␮m; D, 20 ␮m; E, F, and M–P, 100 ␮m; G–L, 10 ␮m.)
`
`in brains of EAE-induced mice 7 days after their injection to the
`periphery. The GA-specific cells were visible in the ventricles, in
`particular, in the choroid plexus (Fig. 1D) and in the tissue
`surrounding the ventricles (Fig. 1 A). Many clusters of cells were
`seen around blood vessels, indicating perivascular infiltration
`into the brain (Fig. 1G). Individual or clustered cells penetrated
`various regions of the brain, including the cortex, the thalamus,
`the basal ganglia, and the hippocampus. Brain sections, contain-
`ing the penetrating fluorescently labeled GA-specific T cells and
`control brain sections from EAE-induced or normal mice were
`immunohistochemically stained for the expression of the neu-
`rotrophic factor BDNF, the Th2 cytokines IL-10 and TGF-␤, and
`the Th1 cytokine IFN-␥. Images were taken from various regions
`of the thalamus and cortex, mainly from the laterodorsal thal-
`amus nucleus, the ventral posteromedial thalamic nucleus, the
`ventral posterolateral thalamic nucleus, and the somatosensory
`cortex.
`
`BDNF. As shown in Fig. 1, adoptively transferred GA-specific cells
`that had been accumulated in the brain (Fig. 1 A, D, G, and M)
`
`expressed BDNF in situ (Fig. 1 B, E, H, and N). This finding was
`verified by colocalization of the corresponding fields (Fig. 1 C,
`F, I, and O). Furthermore, staining of GA cells in the CNS by
`BDNF antibodies was also demonstrated on the single-cell level
`in Hoechst-labeled (Fig. 1J) and PK-labeled (Fig. 1 M–O) cells.
`GA-specific cells expressing BDNF were observed in various
`regions of the brain, mainly in tissue surrounding the ventricles
`(Fig. 1 A–C) and in perivascular cuffs (Fig. 1 G–I). BDNF
`immunostaining localized both in the cytoplasm and in the
`membrane of the cells (Fig. 1 J and N). In contrast to the strong
`BDNF expression in the brains containing GA-specific cells, only
`marginal BDNF staining was observed within the CNS of control
`EAE-induced (Fig. 1K) and in normal (Fig. 1L) mice into which
`no GA-specific cells were injected.
`
`IL-10. Fig. 2 illustrates that GA-specific cells in the brain (Fig. 2
`A, D, and L) demonstrated intensive staining with anti-IL-10
`antibodies (Fig. 2 B, E, and M), as verified by colocalization of
`the corresponding fields (Fig. 2 C, F, and N). This in situ IL-10
`
`14160 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.2336171100
`
`Aharoni et al.
`
`MYLAN PHARMS. INC. EXHIBIT 1084 PAGE 4
`
`

`
`IMMUNOLOGY
`
`that had been accumulated in the brain (Fig. 4 A and D) did not
`express IFN-␥ in situ (Fig. 4 B and E). It was not possible to
`detect any IFN-␥in the brains of mice injected with GA-labeled
`cells, neither in EAE-induced (Fig. 4 A–C) nor in normal (Fig.
`4 D–F) mice. On the other hand, intense staining for IFN-␥was
`found in corresponding brain regions of EAE-induced mice that
`were not injected with GA-specific cells (Fig. 4 G and H). This
`staining was much higher than that observed in normal mice
`(Fig. 4I).
`
`Discussion
`Immunomodulating therapies for CNS pathologies have been
`studied extensively and in clinical use for several years. However,
`the understanding of the pathway by which they actually operate
`in the target organ has lagged behind. Thus, neurotrophic factors
`and antiinflammatory cytokines are known to play a crucial role
`in MS and EAE modulation (1–3, 12–15), but information on
`their expression within the CNS is limited. In the present study
`we tested whether treatment with the MS drug GA results in
`secretion of neurotrophic factor and antiinflammatory cytokines
`in situ through specific Th2兾3 cells that penetrate the brain. To
`trace the low amounts of factors secreted by a specific subset of
`T cells on the level of whole-brain tissue, we used the double-
`labeling approach in which prelabeled specific T cells were
`adoptively transferred and their cytokines兾neurotrophic factor
`expression was subsequently immunohistologically detected.
`As clearly demonstrated, GA-specific cells in the brain man-
`ifested intensive expression of the neurotrophic factor BDNF
`(Fig. 1) and of the two antiinflammatory cytokines, IL-10 (Fig.
`2) and TGF-␤(Fig. 3), but no trace of the inflammatory cytokine
`IFN-␥ (Fig. 4). This expression was clearly verified on the
`single-cell level by labeling with two different dyes, i.e., Hoechst
`and PKH26, which incorporate into different cell
`loci (the
`nucleus and the membrane, respectively), indicating that these
`findings do not reflect an artifact caused by cell staining. It has
`been claimed that, because of its ability to bind cellular DNA,
`Hoechst labeling inhibits lymphocyte activity and interacts with
`bystander cells (16). We have not observed these phenomena, as
`demonstrated by the comparison of the proliferation and cyto-
`kine secretion of labeled cells with unlabeled cells and by the
`ability of the labeled cells to prevent EAE (8). Yet, to affirm
`staining specificity, we used, in addition, a vital labeling proce-
`dure by the membrane inserting dye PKH26. Parallel results
`were obtained with both staining methods.
`BDNF, IL-10, and TGF-␤manifested cytoplasmatic as well as
`membranalic expression. Positively stained GA-specific cells
`were apparent in various regions throughout the brain, including
`the cortex, the thalamus, the basal ganglia, and the hippocampus,
`mainly in tissue surrounding the ventricles and in perivascular
`locations. They were found in brains of normal and EAE-
`induced mice after adoptive transfer of GA-specific cells. In both
`cases, staining intensity on a single-cell level looked similar. Yet,
`the total expression on the whole-tissue level was significantly
`higher in EAE-induced mice, because a great deal more GA-
`specific cells were present in the brains of those mice than in the
`brains of normal mice. The difference in cell infiltration could
`result from blood–brain barrier injury caused by the injection of
`pertussis toxin during EAE induction and from the destruction
`typical to the disease process. In any event, an extensive expres-
`sion of these modulatory substances by GA-specific cells in the
`CNS was manifested during the pathological process. T cells
`from lines induced by oral immunization or by daily injections of
`GA demonstrated similar CNS penetration and expression pat-
`terns of all the factors tested.
`Intense staining with BDNF, IL-10, and TGF-␤antibodies was
`found only in brains of mice that had been injected with
`GA-specific cells. In control mice only faint staining was noted.
`It is of interest that BDNF staining in brains of EAE-induced
`
`Lack of expression of IFN-␥by GA-specific cells in the brain. Cells in the
`Fig. 4.
`cortex of an EAE-induced mouse (A–C) and a normal mouse (D–F). Expression
`of IFN-␥in corresponding brain sections of controls: an EAE-induced mouse (G
`and H) and a normal mouse (I). (Scale bar: A–F and H, 40 ␮m; G and I, 100 ␮m.)
`
`expression by GA cells was clearly demonstrated on a single-cell
`level in Hoechst-labeled (Fig. 2 G–I) and PK-labeled (Fig. 2
`L–N) cells. IL-10 was expressed both in the cytoplasm and in the
`membrane of the cells (Fig. 2 H and M).
`Clusters of GA-labeled cells expressing IL-10 were abundant
`in various regions throughout the brain. Moreover, unlabeled
`cells within and in the vicinity of these clusters also expressed
`high levels of IL-10 (Fig. 2 B, C, E, and F). These unlabeled
`IL-10-positive cells had elongated astrocyte-like morphology
`consistent with activated microglia. Triple staining of slides
`containing GA-specific cells (Fig. 2O) for IL-10 (Fig. 2P) and
`GFAP, a component of astrocyte filaments (Fig. 2Q), confirmed
`the astrocyte phenotype of these adjacent cells (Fig. 2R). Thus,
`IL-10 expression was evident not only in the labeled GA-specific
`cells that migrated to the brain but also in the surrounding
`resident CNS cells.
`The intense IL-10 secretion was observed only in brains with
`adoptively transferred GA cells, whereas control mice expressed
`low to moderate IL-10 levels. IL-10 expression in brains of
`EAE-induced mice (Fig. 2 J) was slightly higher than that
`observed in brains of normal mice (Fig. 2K). Moderate IL-10
`expression appeared to be associated with areas of cellular
`infiltrated and perivascular cuffs.
`
`TGF-␤. A similar phenomenon was observed regarding TGF-␤, as
`shown in Fig. 3. Adoptively transferred GA-specific cells in the
`brain manifested extensive TGF-␤ expression (Fig. 3 A–D and
`G–L), with strong cytoplasmatic and membranal staining of
`individual cells (Fig. 3 D and H). The staining pattern was similar
`to the pattern observed for IL-10, including the positive staining
`of surrounding elongated cells of astrocyte morphology, which
`was confirmed by triple labeling with GFAP (Fig. 3 J–M). These
`surrounding bystander resident cells displayed similar or even
`higher TGF-␤expression intensity compared with the staining of
`the GA-specific cell population (Fig. 3D).
`In contrast to the strong TGF-␤ secretion in brains with
`adoptively transferred GA cells, control mice expressed low to
`moderate TGF-␤ levels, which were higher in brains of EAE-
`induced mice (Fig. 3E) than in those of normal mice (Fig. 3F).
`
`IFN-␥. The results presented hitherto pertain to the Th2-related
`cytokines. It was of interest to test the expression pattern of a
`Th1 cytokine such as IFN-␥. As shown in Fig. 4, GA-specific cells
`
`Aharoni et al.
`
`PNAS 兩 November 25, 2003 兩 vol. 100 兩 no. 24 兩 14161
`
`MYLAN PHARMS. INC. EXHIBIT 1084 PAGE 5
`
`

`
`mice was marginal and similar to the staining of normal mice
`(Fig. 1 K and L), whereas IL-10 and TGF-␤expression in brains
`of EAE-induced mice were somewhat higher than in brains of
`normal mice (Figs. 2 J and K and 3 E and F). This finding agrees
`with other studies that demonstrated up-regulation of these
`cytokines during disease (1, 2, 14, 15). In contrast to Th2兾3
`cytokine expression, which was increased only marginally in
`brains of EAE-induced mice, a significant augmentation in the
`expression of IFN-␥ was observed in brains of EAE-induced
`mice (Fig. 4 G and H) in comparison with normal mice (Fig. 4I).
`This finding corroborates our previous results, indicating that
`lymphocytes isolated from brains of EAE-induced mice secrete
`high levels of IFN-␥ (8, 9). Notably, this increase in IFN-␥ was
`not manifested in brain lymphocytes from EAE-induced mice
`treated with GA (8, 9) nor in corresponding brain sections from
`EAE-induced mice that had been injected with GA-specific cells
`(Fig. 4B). In both cases, IFN-␥ levels were similar to those of
`normal mice. IFN-␥ is a principal Th1 effector cytokine, which
`by and large is believed to be involved in the pathological
`processes of EAE and MS (1). Hence, the significance of the
`down-regulation of this inflammatory cytokine in the target
`organ, induced by the MS drug GA.
`The current consensus is that activated T cells of any speci-
`ficity cross the blood–brain barrier and penetrate the CNS (18),
`but, thereafter, T cells that are not able to recognize their
`specific antigen in the CNS decline to baseline level. The ability
`of GA-induced T cells to persist in the CNS, while cells with
`different specificity were completely absent, was previously
`shown and attributed to their crossreactivity with the myelin
`antigen myelin basic protein (8, 9). The present study demon-
`strates that GA-specific cells not only accumulate in the brain but
`actually function in the diseased organ as regulatory cells by the
`secretion of Th2兾3 and neurotrophic factor. Of special interest
`is the finding that IL-10 and TGF-␤are expressed not only by the
`GA-labeled cells but also by unlabeled cells within their vicinity
`(Figs. 2 C and F and 3D). These surrounding bystander cells have
`elongated astrocyte-like morphology consistent with activated
`microglia. Staining with GFAP, a component of astrocyte
`filaments, corroborated the astrocyte nature of these cells (Figs.
`
`2 O–R and 3 J–M). The glial cells displayed similar or even higher
`IL-10 and TGF-␤ expression intensity compared with the stain-
`ing of the GA specific population. Such spreading of positive
`staining could result either from genuine IL-10 and TGF-␤
`secretion by the astrocytes that had been activated by the
`cytokines secreted from the GA-specific cells, or from the
`binding of IL-10 and TGF-␤secreted by the GA-specific cells to
`their specific receptors on these astrocytes (17, 19). In both cases,
`this Th2兾3 spreading suggests a bystander therapeutic effect of
`GA on the CNS resident cells.
`IL-10 is a potent regulatory cytokine in autoimmunity that
`inhibits Th1 cells and macrophage activation in addition to its
`effector multifunctional therapeutic reactivity (1, 2, 14). More-
`over, IL-10 can modulate glial cell responses by inhibiting MHC
`class II, NO, and chemokine expression (19). TGF-␤suppresses
`cytotoxic T cell response, production of tumor necrosis factor ␣,
`IFN-␥, and factors that contribute to myelin damage, such as
`lysosomal enzymes and nitrogen intermediates (15). The ability
`of GA-infiltrating cells to express and induce the expression of
`these potent modulating cytokines, also by bystander CNS cells,
`substantiates GA therapeutic activity. Furthermore, the potent
`neurotrophic factor BDNF, expressed by the GA-specific cells in
`situ, is a key regulator of neuronal development, which supports
`neuronal survival and regulates neurotransmitter release and
`dendritic growth (12, 13). BDNF can rescue injured or degen-
`erating neurons and induce axonal outgrowth, remyelination,
`and regeneration (20). The full-length BDNF receptor, tyrosine
`kinase receptor B, has been found in neurons in the vicinity of
`MS plaques and in reactive astrocytes in MS lesions (21). The
`BDNF secreted by the GA-infiltrating cells can therefore actu-
`ally function in the MS兾EAE target tissue. Thus, the expression
`of BDNF, IL-10, and TGF-␤ by GA-specific cells in the CNS
`directly links the therapeutic activity of GA in MS兾EAE and its
`in situ immunomodulatory effect, namely, the antiinflammatory
`bystander suppression and neuroprotection induced by GA-
`specific T cells in the brain.
`
`This study was supported in part by grants from Teva Pharmaceutical
`Industries (Israel) and Terry and Dr. Claude Oster and by a special fund
`of the Eugene Applebaum Family Foundation (Bloomfield Hills, MI).
`
`1. Liblau, R. S., Singer, S. M. & McDevitt, H. O. (1995) Immunol. Today 16,
`34–38.
`2. Kennedy, M. K., Torrance, D. S., Picha, K. S. & Mohler, K. M. (1992)
`J. Immunol. 149, 2496–2505.
`3. Ziemssen, T., Kumpfel, T., Klinkert, W. E., Neuhaus, O. & Hohlfeld, R. (2002)
`Brain 125, 2381–2391.
`4. Teitelbaum, D., Aharoni, R., Fridkis-Hareli, M., Arnon, R. & Sela, M. (1998)
`in The Decade in Autoimmunity, ed. Shoenfeld, Y. (Elsevier, New York), pp.
`183–188.
`5. Aharoni, R., Teitelbaum, D., Sela, M. & Arnon, R. (1997) Proc. Natl. Acad. Sci.
`USA 94, 10821–10826.
`6. Aharoni, R., Teitelbaum, D., Sela, M. & Arnon, R. (1998) J. Neuroimmunol.
`91, 135–146.
`7. Duda, P. W., Schmied, M. C., Cook, S. L., Krieger, J. I. & Hafler, D. A. (2000)
`J. Clin. Invest. 105, 967–976.
`8. Aharoni, R., Teitelbaum, D., Leitner, O., Meshorer, A., Sela, M. & Arnon, R.
`(2000) Proc. Natl. Acad. Sci. USA 97, 11472–11477.
`9. Aharoni, R., Meshorer, A., Sela, M. & Arnon, R. (2002) J. Neuroimmunol. 126,
`58–68.
`
`10. Krakowski, M. L. & Owens, T. (1997) Eur. J. Immunol. 27, 2840–2847.
`11. Kipnis, J., Yoles, E., Porat, Z., Cohen, A., Mor, F., Sela, M., Cohen, I. R. &
`Schwartz, M. (2000) Proc. Natl. Acad. Sci. USA 97, 7446–7451.
`12. Thoenen, H. (1995) Science 270, 593–598.
`13. Barde, Y. A. (1997) Nature 385, 391–393.
`14. Bettelli, E., Nicholson, L. B. & Kuchroo, V. K. (2003) J. Autoimmun. 20,
`265–267.
`15. Morris, M. M., Dyson, H., Baker, D., Harbige, L. S., Fazakerley, J. K. & Amor,
`S. (1997) J. Neuroimmunol. 74, 185–197.
`16. Parish, C. R. (1999) Immunol. Cell Biol. 77, 499–512.
`17. Hulshof, S., Montagne, L., De Groot, C. J. A. & Van Der Valk, P. (2002) Glia
`38, 24–35.
`18. Flugel, A., Berkowicz, T., Ritter, T., Labeur, M., Li, Z., Ellwart, J. W., Willem,
`M., Lassmann, H. & Wekerle, H. (2001) Immunity 14, 547–560.
`19. Ledeboer, A., Breve, J. P., Poole, S., Tilders, F. J. & Van Dam, A. M. (2000)
`Glia 30, 134–142.
`20. Gravel, C., Gotz, R., Lorrain, A. & Sendtner, M. (1997) Nat. Med. 3, 765–770.
`21. Stadelmann, C., Kerschensteiner, M., Misgeld, T., Bruck, W., Hohlfeld, R. &
`Lassmann, R. (2002) Brain 125, 75–85.
`
`14162 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.2336171100
`
`Aharoni et al.
`
`MYLAN PHARMS. INC. EXHIBIT 1084 PAGE 6

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket